Download as pdf or txt
Download as pdf or txt
You are on page 1of 690

Oxford Textbook of

Vasculitis
Oxford Textbook of

Vasculitis
THIRD EDITION

Edited by
Gene V. Ball, MD
Jane Knight Lowe Professor of Medicine Emeritus,
Division of Clinical Immunology and Rheumatology,
The University of Alabama at Birmingham,
Birmingham, Alabama, USA

Barri J. Fessler, MD, MSPH


Associate Professor of Medicine,
Division of Clinical Immunology and Rheumatology,
The University of Alabama at Birmingham,
Birmingham, Alabama, USA

S. Louis Bridges, Jr., MD, PhD


Department of Medicine,
Division of Clinical Immunology and Rheumatology,
The University of Alabama at Birmingham,
Birmingham, Alabama, USA
3
Great Clarendon Street, Oxford, OX2 6DP,
United Kingdom
Oxford University Press is a department of the University of Oxford.
It furthers the University’s objective of excellence in research, scholarship,
and education by publishing worldwide. Oxford is a registered trade mark of
Oxford University Press in the UK and in certain other countries
© Oxford University Press 2014
The moral rights of the authors have been asserted
First Edition published in 2002
Second Edition published in 2008
Third Edition Published in 2014
Impression: 1
All rights reserved. No part of this publication may be reproduced, stored in
a retrieval system, or transmitted, in any form or by any means, without the
prior permission in writing of Oxford University Press, or as expressly permitted
by law, by licence or under terms agreed with the appropriate reprographics
rights organization. Enquiries concerning reproduction outside the scope of the
above should be sent to the Rights Department, Oxford University Press, at the
address above
You must not circulate this work in any other form
and you must impose this same condition on any acquirer
Published in the United States of America by Oxford University Press
198 Madison Avenue, New York, NY 10016, United States of America
British Library Cataloguing in Publication Data
Data available
Library of Congress Control Number: 2013949082
ISBN 978–0–19–965986–9
Printed in the UK by
Bell & Bain Ltd, Glasgow
Oxford University press makes no representation, express or implied, that the
drug dosages in this book are correct. Readers must therefore always check
the product information and clinical procedures with the most up-to-date
published product information and data sheets provided by the manufacturers
and the most recent codes of conduct and safety regulations. The authors and
the publishers do not accept responsibility or legal liability for any errors in the
text or for the misuse or misapplication of material in this work. Except where
otherwise stated, drug dosages and recommendations are for the non-pregnant
adult who is not breast-feeding
Links to third party websites are provided by Oxford in good faith and
for information only. Oxford disclaims any responsibility for the materials
contained in any third party website referenced in this work.
Contents

Abbreviations vii SECTION 3


Contributors xi Clinical manifestations common
to vasculitis
SECTION 1 10. Cutaneous manifestations of vasculitis 121
Introductory chapters Nicole M. Fett and Victoria P. Werth

1. Nomenclature and classification 11. Ocular manifestations of systemic


of vasculitic syndromes 3 vasculitis 131
Gene V. Ball and S. Louis Bridges, Jr Russell W. Read

2. Epidemiology of vasculitis 7 12. Cardiopulmonary manifestations


Richard A. Watts and David G.I. Scott of vasculitis 143
Stephen K. Frankel and Marvin I. Schwarz

SECTION 2 13. Vasculitic neuropathy 157


Shin J. Oh
Basic science
14. Vasculitic manifestations in the
3. Hypersensitivity 29
gastrointestinal tract 179
T. Prescott Atkinson
Gaafar Ragab
4. Biology of endothelial cells 41
15. Renal manifestations 197
Zoltán Szekanecz, György Kerekes, and Alisa E. Koch
Cees G.M. Kallenberg and Jan Willem Cohen Tervaert
5. Complement in vasculitis 53
16. Digital ischaemia and Raynaud’s
John E. Volanakis
phenomenon 209
6. Autoantibodies in vasculitis 61 K. Kwasind Huston, John H. Stone, and Fredrick M. Wigley
Jan Damoiseaux and Jan Willem Cohen Tervaert
7. Pathogenesis of vasculitis 71 SECTION 4
Gene V. Ball, S. Louis Bridges, Jr, and T. Prescott Atkinson Imaging and percutaneous
8. Animal models of vasculitis 93 interventions
Peter Heeringa
17. Angiography and percutaneous
9. Pathological features of vasculitis 101 interventions 227
Andrew Churg Souheil Saddekni and Rachel F. Oser
vi contents

18. Cross-sectional imaging in vasculitis 247 32. Eosinophilic granulomatosis with polyangiitis
Enrique A. Sabater and Anthony W. Stanson (Churg–Strauss syndrome) 433
Jeremy M. Clain and Ulrich Specks
19. PET imaging in vasculitis 267
Daniel Blockmans 33. Vasculitis in primary connective
tissue diseases 443
Laura B. Hughes
SECTION 5
34. Behçet’s syndrome: pathogenesis, clinical
Vasculitic diseases and syndromes manifestations, and treatment 467
and related disorders Emire Seyahi, Gulen Hatemi, Izzet Fresko,
20. Approach to the diagnosis of Melike Melikoglu, and Hasan Yazici
vasculitis in adult patients 275 35. Juvenile Behçet’s syndrome 491
Barri J. Fessler Gülsevim Azizlerli and Rifkiye Sarica-Kucukoglu
21. Vasculitis in infancy, childhood, 36. Vasculitis of the central nervous system 497
and adolescence 283 Rula A. Hajj-Ali, George F. Duna,
Ross E. Petty and Leonard H. Calabrese
22. Assessment of disease activity and damage 299 37. Thromboangiitis obliterans
Raashid Luqmani (Buerger’s disease) 507

23. Giant cell arteritis and polymyalgia Michael Frank and Jean-Noel Fiessinger
rheumatica 307 38. Cutaneous small-vessel vasculitis 517
Kenneth J. Warrington and Cornelia M. Weyand Anna Haemel, Lindy Fox, and M. Kari Connolly
24. Takayasu’s arteritis 319 39. IgA vasculitis (Henoch-Schönlein purpura) 527
Yasushi Kobayashi, Tomohiro Ishii, and Hideo Harigae Miguel A. González-Gay, Ricardo Blanco,
and Trinitario Pina
25. Polyarteritis nodosa 331
Loïc Guillevin and Benjamin Terrier 40. Cryoglobulinaemic vasculitis 547
Massimo Galli, Salvatore Sollima, Francesco
26. Microscopic polyangiitis 351
Saccardo, and Giuseppe Monti
Loïc Guillevin and Benjamin Terrier
41. Miscellaneous forms of vasculitis 569
27. Cutaneous polyarteritis 363 Gim Gee Teng, Sumapa Chaiamnuay,
Angelo L. Gaffo and W. Winn Chatham
28. Kawasaki’s disease 373 42. Experimental therapies for vasculitis 599
Jane C. Burns Sebastian Unizony and John H. Stone
29. Granulomatosis with polyangiitis (Wegener’s
granulomatosis): pathogenesis 385
SECTION 6
Peter Lamprecht, Konstanze Holl-Ulrich,
and Wolfgang L. Gross Mimickers of vasculitis
30. Granulomatosis with polyangiitis 43. Antiphospholipid syndrome 615
(Wegener’s granulomatosis): clinical David P. D’Cruz, Munther A. Khamashta,
and immunodiagnostic aspects 401 and Graham R.V. Hughes
Susanne Schinke, Wolfgang L. Gross, and Elena Csernok 44. Imitators of vasculitis 635
31. Granulomatosis with polyangiitis (Wegener’s Sharon A. Chung and Kenneth E. Sack
granulomatosis): treatment 417
Julia U. Holle, Bernhard Hellmich, and Wolfgang L. Gross Index 657
Abbreviations

AAV antineutrophil cytoplasmic antibody-associated CDR complementary determining region


vasculitis CGRP calcitonin gene related peptide
ABI ankle-brachial pressure index CHCC Chapel Hill Consensus Conference
ABRA Aβ-related angiitis CIDP chronic inflammatory demyelinating polyneuropathy
ACE angiotensin-converting enzyme CMAP compound muscle action potential
ACR American College of Rheumatology CML chronic myelogenous leukaemia
ADCC antibody-dependent cellular toxicity CMRI cardiac magnetic resonance imaging
ADEM acute disseminated encephalomyelitis CMV cytomegalovirus
ADMA asymmetric dimethylarginine CNAP compound nerve action potential
AECA antiendothelial cell antibodies CNS central nervous system
AGBMA antiglomerular basement membrane antibodies COX cyclo-oxygenase
AHEI acute haemorrhagic oedema of infancy CPN classis periarteritis nodosa
AIL angioimmunolymphoproliferative lesion CRP C-reactive protein
AION anterior ischaemic optic neuropathy CRVO central retinal vein occlusion
ALS amyotrophic lateral sclerosis CS Cogan’s syndrome
ANCA antineutrophil cytoplasmic antibodies CS corticosteroid
APACHE acute physiology and chronic health evaluation score CSS Churg–Strauss syndrome
APC antigen-presenting cell CSVV cutaneous small-vessel vasculitis
APS antiphospholipid syndrome CT computed tomography
APTT activated partial thromboplastin time CTA computed tomography angiography
AS ankylosing spondylitis CTD connective tissue disease
ASCA anti-Saccharomyces cerevisiae antibodies CTL cytotoxic T-lymphocyte
ATG antithymocyte globulin CUS chronic ulcerative stomatitis
AVN avascular necrosis CV cryoglobulinaemic vasculitis
AZT zidovudine CXR chest radiograph
BAFF B-cell activating factor CYC cyclophosphamide
BAL bronchoalveolar lavage DAD diffuse alveolar damage
BCS Budd–Chiari syndrome DAH diffuse alveolar haemorrhage
BD Behçet’s disease DC dendritic cell
BOOP bronchiolitis obliterans organizing pneumonia DEI disease extent index
BPI bactericidal permeability-increasing protein DIC disseminated intravascular coagulation
BRVO branch retinal vein occlusion DIF direct immunofluorescence
BS Behçet’s syndrome DIV drug induced vasculitis
BUN blood urea nitrogen DM dermatomyositis
BVAS Birmingham Vasculitis Activity Score DMARD disease modifying antirheumatic drug
CAA cerebral amyloid angiopathy DSA digital subtraction angiography
CAM cell adhesion molecule DTH delayed-type hypersensitivity
CBC complete blood count DVT deep venous thrombosis
CCA common carotid artery DWI diffusion weighted images
CD Crohn’s disease EBV Epstein–Barr virus
CDA combined damage assessment EC endothelial cell
viii abbreviations

ECM extracellular matrix IgAV IgA vasculitis


ECP eosinophil cationic protein IGF insulin-like growth factor
EDHF endothelium-derived hyperpolarizing factor IIF indirect immunofluorescence
EED erythema elevatum diutinum IL interleukin
EGF epidermal growth factor ILD interstitial lung disease
EGPA eosinophilic granulomatosis with polyangiitis IMA inferior mesenteric artery
EID endothelium-independent vasodilatation IMT intima–media thickness
ELAM-1 endothelial–leukocyte adhesion molecule-1 INR International Normalized Ratio
ELISA enzyme-linked immunoabsorbent assay ION ischaemic optic neuropathy
ELP enterocolic lymphocytic phlebitis IPN infantile polyarteritis nodosa
EM erythema multiforme IRF interferon regulatory factor
EMC essential mixed cryoglobulinaemia ISKDC International Study for Kidney Disease in Children
EMG electromyography ISV idiopathic systemic vasculitis
EPC endothelial progenitor cell ITAS Indian Takayasu’s Arteritis Score
ERK extracellular signal-regulated kinase IVC inferior vena cava
ESAM EC-selective adhesion molecule IVIg intravenous immunoglobulin
ESL E-selectin ligand JAM junctional adhesion molecule
ESR erythrocyte sedimentation rate JBS juvenile Behçet’s syndrome
FcεRI high-affinity IgE receptor JDM juvenile dermatomyositis
FDG fluoro-18-deoxyglucose JRA juvenile rheumatoid arthritis
FEIA fluorescent-enzyme immunoassay KCS keratoconjunctivitis sicca
FFS Five Factor Score KD Kawasaki’s disease
FGF fibroblast growth factor KIR killer-immunoglobulin-like receptor
FMD flow-mediated vasodilation LA lupus anticoagulant
FMF familial Mediterranean fever LAMP lysosomal-associated membrane protein
fMLP formyl-methionine leucine phenylalanine LCV leukocytoclastic vasculitis
FUO fever of unknown origin LEF leflunomide
GACNS granulomatous angiitis of the central nervous system LFA-1 lymphocyte function-associated antigen-1
GBM glomerular basement membrane LG lymphomatoid granulomatosis
GBS Guillain–Barré LMV lupus mesenteric vasculitis
GCA giant cell arteritis LON late-onset neutropenia
G-CSF granulocyte-colony stimulating factor LPS lipopolysaccharides
GM-CSF granulocyte-macrophage colony-stimulating factor LR likelihood ratio
GN glomerulonephritis LT leukotriene
GPA granulomatosis with polyangiitis LTRA leukotriene receptor antagonists
GPI glycan phosphatidylinositol LVV large-vessel vasculitis
GSE gluten-sensitive enteropathy MAbs monoclonal antibodies
GWAS genome-wide association study MAC membrane attack complex
HBV hepatitis B virus MALT mucosa-associated lymphoid tissue
HCL hairy cell leukaemia MAPK mitogen-activated protein kinase
HCV hepatitis C virus MASP MBL-associated serine protease
HeAU herpetiform aphthous ulcers MBL mannan-binding lectin
HES hypereosinophilic syndromes MC mixed cryoglobulinaemia
HIDS hyperimmunoglobulin D syndrome M-CSF macrophage-colony stimulating factor
HIV human immunodeficiency virus MCTD mixed connective tissue disease
HLA human leukocyte antigen MDS melodysplastic syndromes
HLE human leukocyte elastase MGUS monoclonal gammopathy of undetermined
HR hazard ratio significance
HRCT high-resolution computed tomography MHC major histocompatibility complex
HSP Henoch–Schönlein purpura MI myocardial infarction
HSV herpes simplex virus MiAU minor aphthous ulcers
HUV hypocomplementaemic urticarial vasculitis MIF migration inhibitory factor
HUVEC human umbilical vein endothelial cell MjAU major aphthous ulcers
IC immune complex ML mass lesion
ICAM-1 intercellular adhesion molecule-1 MMF mycophenolate mofetil
IE infective endocarditis MMP matrix metalloproteinase
IFN interferon MP methylprednisolone
IFX infliximab MPA microscopic polyangiitis
IgAN IgA nephropathy MPGN membranoproliferative glomerulonephritis
abbreviations ix

MPO myeloperoxidase RCVS reversible cerebral vasoconstriction syndrome


MRA magnetic resonance angiography RF rheumatoid factor
MRI magnetic resonance imaging RMSF Rocky Mountain spotted fever
MRV magnetic resonance venography ROI reactive oxygen intermediates
MS multiple sclerosis RP relapsing polychondritis
MSU monosodium urate RPGN rapidly progressive glomerulonephritis
MTX methotrexate RVCL retinal vasculopathy with cerebral leukodystrophy
MUP motor unit potential SAA serum amyloid A
MVV medium-vessel vasculitis SAg superantigen
NCGN necrotizing crescentic glomerulonephritis SCG spontaneous crescentic glomerulonephritis
NCV nerve conduction velocity SCLC small cell lung cancer
NET neutrophil extracellular trap SE shared epitope
NHL non-Hodgkin’s lymphoma SLE systemic lupus erythematosus
NIH National Institutes of Health SMA superior mesenteric artery
NK natural killer cell SNP single nucleotide polymorphism
NOS nitric oxide synthetase SNV systemic necrotizing vasculitis
NPV negative predictive value SOV single-organ vasculitis
NSAID non-steroidal anti-inflammatory drug SPECT single photon emission computed tomography
NSG necrotizing sarcoid granulomatosis SS Sjögren’s syndrome
NSIP non-specific interstitial pneumonia SSc systemic sclerosis
NSVN non-systemic vasculitic neuropathy SVN sensory vasculitic neuropathy
NUV normocomplementaemic urticarial vasculitis SVR sustained virological response
OR odds ratio SVV small-vessel vasculitis
PAA pulmonary artery aneurysm TA Takayasu’s arteritis
PACNS primary angiitis of the central nervous system TAB temporal artery biopsy
PAF platelet-activating factor TAO thromboangiitis obliterans
PAI pulmonary arterial involvement TCR T-cell receptor
PAN polyarteritis nodosa TCZ tocilizumab
PAR proteinase-activated receptors TEM effector memory T cell
PAT pulmonary artery thrombosis TGF transforming growth factor
PCR polymerase chain reaction TLR Toll-like receptor
PDGF platelet-derived growth factor TN naïve T cell
PECAM platelet-endothelial adhesion molecule TNF tumour necrosis factor
PET positron emission tomography TSST toxic shock syndrome toxin
PFM protracted febrile myalgia UC ulcerative colitis
PJP Pneumocystis jiroveci pneumonia UIP usual interstitial pneumonia
PM polymyositis UKGPRD UK General Practice Research Database
PMN polymorphonuclear neutrophil URI upper respiratory tract infection
PMR polymyalgia rheumatica US ultrasonography
PMR polymyalgia rheumatica UV urticarial vasculitis
PNP purine nucleoside phosphorylase UVR ultraviolet radiation
PORH postocclusive reactive hyperaemia VAP vascular adhesion protein
PPD purified protein derivative VATS video-assisted thoracic surgery
PPV positive predictive value VCAM vascular cell adhesion molecule
PRD prednisolone VDI vasculitis damage index
PRES Paediatric Rheumatology European Society VEGF vascular endothelial growth factor
PSW positive sharp wave VTE venous thromboembolic disease
PTA percutaneous transluminal angioplasty VVV variable-vessel vasculitis
PTU propylthiouracil VVV vasa vasorum vasculitis
PUK peripheral ulcerative keratitis vWF von Willebrand factor
PV paraneoplastic vasculitis VZV varicella zoster virus
PVAS paediatric vasculitis activity score WAS Wiskott–Aldrich syndrome
RA rheumatoid arthritis WASP Wiskott–Aldrich syndrome protein
RAS recurrent aphthous stomatitis WBC white blood cell
RAS renin–angiotensin system WG Wegener’s granulomatosis
RBC red blood cell XLP X-linked lymphoproliferative disease
RBV ribavirin
Contributors

T. Prescott Atkinson Jane C. Burns


University of Alabama at Birmingham, Professor of Pediatrics, Director,
Department of Pediatrics, Kawasaki Disease Research Center,
Birmingham, AL, USA University of California San Diego,
La Jolla, CA, USA
Gülsevim Azizlerli
Istanbul University, Leonard H. Calabrese
Department of Dermatology, Professor of Medicine,
Turgut Ozal Cd., Capa, Cleveland Clinic Lerner College of Medicine of Case Western
Istanbul, Turkey Reserve University,
Vice Chairman, Department of Rheumatic and Immunologic
Gene V. Ball Diseases, R. J. Fasenmyer Chair of Clinical Immunology,
University of Alabama at Birmingham, Cleveland Clinic, Cleveland, OH, USA
Department of Medicine,
Division of Clinical Immunology and Rheumatology, Sumapa Chaiamnuay
Birmingham, AL, USA Division of Clinical Immunology and Rheumatology,
Department of Medicine,
Ricardo Blanco University of Alabama at Birmingham, AL, USA
Rheumatology Division,
Hospital Universitario “Marques de Valdecilla”, W. Winn Chatham
Facultad de Medicina, University of Alabama at Birmingham,
Universidad de Cantabria, Department of Medicine,
Santander, Spain Division of Clinical Immunology and Rheumatology,
Birmingham, AL, USA
Daniel Blockmans
University Hospital Gasthuisberg, Sharon A. Chung
General Internal Medicine, University of California San Francisco,
Leuven, Belgium San Francisco, CA, USA

S. Louis Bridges, Jr Andrew Churg


University of Alabama at Birmingham, Department of Pathology,
Department of Medicine, University of British Columbia,
Division of Clinical Immunology and Rheumatology, Vancouver, Canada
Birmingham, AL, USA
xii contributors

Jeremy M. Clain Lindy Fox


Mayo Clinic, Assistant Professor of Clinical Dermatology,
Division of Pulmonary and Critical Care Medicine, Department of Dermatology,
Rochester, MN, USA University of California,
San Francisco, USA
Jan Willem Cohen Tervaert
Department of Clinical Immunology, Michael Frank
University Hospital Maastricht, Centre de Référence des Maladies Vasculaires Rares et Service de
Maastricht, The Netherlands Médecine Vasculaire/HTA,
M. Kari Connolly Hôpital Européen Georges Pompidou,
University of California San Francisco, Assistance Publique des Hôpitaux de Paris,
San Francisco, CA, USA Paris, France

Elena Csernok Stephen K. Frankel


Department of Rheumatology, Associate Professor of Medicine,
University Hospital of Schleswig-Holstein, National Jewish Health,
Campus Lübeck, and Klinikum Bad Bramstedt, Denver,
Lübeck, Germany Colorado and Division of Pulmonary Sciences and Critical Care
Medicine,
David P. D’cruz
University of Colorado at Denver,
Lupus Research Unit,
Anschutz Medical Campus,
The Rayne Institute,
Aurora, Colorado, USA
St Thomas' Hospital London,
London, UK Izzet Fresko
Jan Damoiseaux Professor of Medicine,
Laboratory of Clinical Immunology, Division of Rheumatology,
Maastricht University Medical Center, Department of Medicine,
Maastricht, The Netherlands Cerrahpasa Medical Faculty,
University of Istanbul,
George F. Duna Istanbul, Turkey
Associate Professor of Medicine, Baylor College of Medicine,
Chief of Rheumatology, Angelo L. Gaffo
St. Luke’s Episcopal Hospital, Division of Rheumatology and Clinical Immunology,
Houston, Texas, USA University of Alabama at Birmingham, AL, USA
Barri J. Fessler
Massimo Galli
Associate Professor of Medicine,
Istituto di Malattie Infettive e Tropicali,
Division of Clinical Immunology and Rheumatology,
Università di Milano,
University of Alabama at Birmingham,
Ospedale Luigi Sacco, Milan, Italy
Birmingham, Alabama, USA
Nicole M. Fett Miguel A. González-Gay
University of Pennsylvania, Rheumatology Division,
School of Medicine, Hospital Universitario Marqués de Valdecilla IFIMAV,
Perelman Center for Advanced Medicine, Santander, Spain
Philadelphia, PA USA
Wolfgang L. Gross
Jean-Noel Fiessinger University of Lübeck,
Centre de Référence des Maladies Vasculaires Rares et Service de Department of Rheumatology,
Médecine Vasculaire/HTA, Vasculitis Center UKSH and Clinical Center Bad Bramstedt,
Hôpital Européen Georges Pompidou, Lübeck, Germany
Assistance Publique des Hôpitaux de Paris,
Paris, France and Université Paris Descartes,
Paris, France
contributors xiii

Loïc Guillevin Graham R.V. Hughes


Professor of Medicine, London Lupus Centre,
Department of Internal Medicine, London Bridge Hospital,
National Reference Center for Rare Autoimmune and Systemic London, UK
Diseases,
INSERM U1060, Hôpital Cochin, Laura B. Hughes
Assistance Publique–Hôpitaux de Paris, University of Alabama at Birmingham,
University of Paris 5-René-Descartes, Paris, France Department of Medicine,
Division of Clinical Immunology and Rheumatology,
Anna Haemel Birmingham, AL, USA
Assistant Professor of Clinical Dermatology,
Department of Dermatology, K. Kwasind Huston
University of California, Clinical Assistant Professor of Medicine,
San Francisco, USA University of Missouri Kansas City School of Medicine,
Saint Luke’s Physician Partners,
Rula A. Hajj-Ali Kansas City, Missouri, USA
Assistant Professor of Medicine,
Cleveland Clinic Lerner College of Medicine of Case Western Tomohiro Ishii
Reserve University, Department of Hematology and Rheumatology,
Center for Vasculitis Care and Research, Tohoku University Graduate School of Medicine,
Cleveland Clinic, USA Sendai, Japan
Hideo Harigae Cees G.M. Kallenberg
Department of Hematology and Rheumatology, University Medical Center,
Tohoku University Graduate School of Medicine, Antonius Deusinglaan 1,
Sendai, Japan Groningen, The Netherlands
Gulen Hatemi György Kerekes
Associate Professor, Angiology and Intensive Care Unit,
Division of Rheumatology, Institute of Medicine,
Department of Medicine, University of Debrecen Medical and Health Sciences Center,
Cerrahpasa Medical Faculty, Debrecen, Hungary
University of Istanbul, Istanbul, Turkey
Munther A. Khamashta
Peter Heeringa Director, Lupus Research Unit,
Department of Pathology and Medical Biology, St Thomas’ Hospital,
University Medical Center Groningen, London, UK
Groningen, The Netherlands
Yasushi Kobayashi
Bernhard Hellmich Department of Hematology and Rheumatology,
Department of Rheumatology,
Tohoku University Graduate School of Medicine,
University Hospital of Schleswig-Holstein,
Sendai, Japan
Campus Luebeck, and Klinikum Bad Bramstedt,
Luebeck, Germany Alisa E. Koch
Professor of Rheumatology,
Julia U. Holle
University of Michigan Medical School,
Department of Rheumatology,
Department of Internal Medicine,
University Hospital of Schleswig-Holstein,
Division of Rheumatology,
Campus Luebeck, and Klinikum Bad Bramstedt,
Ann Arbor, MI, USA
Luebeck, Germany:
Konstanze Holl-Ulrich Peter Lamprecht
University of Lübeck, University of Lübeck,
Department of Pathology, Department of Rheumatology,
Reference Center for Vasculitis Diagnosis UKSH, Vasculitis Center UKSH and Clinical Center Bad Bramstedt,
Lübeck, Germany. Lübeck, Germany
xiv contributors

Raashid Luqmani Enrique A. Sabater


NIHR Musculoskeletal Biomedical Research Unit, HIMA-San Pablo Bayamon Hospital,
Nuffield Department of Orthopaedics Rheumatology and Head, Vascular and Interventional Radiology,
Musculoskeletal Sciences Botnar Research Centre, Bayamon, Puerto Rico
University of Oxford,
Francesco Saccardo
Oxford, United Kingdom
Rheumatology Unit, L.
Melike Melikoglu Sacco University Hospital of Milan,
Professor of Medicine, Milan, Italy
Division of Rheumatology,
Kenneth E. Sack
Department of Medicine, Cerrahpasa Medical Faculty,
University of California San Francisco,
University of Istanbul,
San Francisco, CA, USA
Istanbul, Turkey
Souheil Saddekni
Giuseppe Monti Director, Interventional Oncology,
Saronno Hospital, Professor of Radiology and Medicine,
Saronno, Italy Interventional Radiology Section, University of Alabama at
Shin J. Oh Birmingham,
Distinguished Professor of Neurology, Birmingham, AL, USA
Department of Neurology, Rifkiye Sarica-Kucukoglu
University of Alabama at Birmingham, Department of Dermatology,
Department of Veterans Affairs Medical Center, Istanbul Medical Faculty,
UAB Station, Birmingham, Istanbul University, Istanbul, Turkey
Birmingham, AL, USA
Susanne Schinke
Rachel F. Oser Department of Rheumatology,
University of Alabama at Birmingham, University Hospital of Schleswig-Holstein,
Department of Radiology, Campus Lübeck and Klinikum Bad Bramstedt,
Birmingham, AL, USA Lübeck, Germany
Ross E. Petty Marvin I. Schwarz
Professor, Division of Rheumatology, James C. Campbell Professor of Pulmonary Medicine,
Department of Pediatrics, Division of Pulmonary Sciences and Critical Care Medicine,
University of British Columbia, and British Columbia’s Children’s University of Colorado at Denver,
Hospital Anschutz Medical Campus,
Aurora, CO, USA
Trinitario Pina
University Hospital Marqués de Valdecilla, David G.I. Scott
Santander, Spain Department of Rheumatology,
Norfolk and Norwich Health Care,
Gaafar Ragab NHS Trust, Norwich, UK
Cairo University,
Mohandeseen, Emire Seyahi
Giza, Egypt Professor of Medicine,
Division of Rheumatology,
Russell W. Read Department of Medicine,
Professor of Ophthalmology and Pathology, Cerrahpasa Medical Faculty,
Director of Education and Residency Program Director, University of Istanbul, Istanbul, Turkey
UAB Department of Ophthalmology,
Director, Uveitis and Ocular Inflammatory Diseases Service, Salvatore Sollima
Chief of Ophthalmology, Institute of Infectious and Tropical Diseases,
Cooper Green Mercy Medical Center, University of Milan,
University of Alabama at Birmingham, L. Sacco Hospital,
Birmingham, AL, USA Milan, Italy
contributors xv

Ulrich Specks Kenneth J. Warrington


Professor of Medicine, Department of Internal Medicine,
Mayo Clinic College of Medicine, Division of Rheumatology,
Chair, Division of Pulmonary and Critical Care Medicine, Mayo Clinic and College of Medicine,
Mayo Clinic, Rochester, MN;
Rochester, USA Department of Medicine,
Division of Immunology and Rheumatology, Stanford University
Anthony W. Stanson School of Medicine,
Professor Emeritus of Radiology, Stanford, CA, USA
Mayo Clinic College of Medicine,
Mayo Clinic, Richard A. Watts
Rochester, USA Department of Rheumatology,
Ipswich Hospital NHS Trust,
John H. Stone Ipswich, Suffolk, UK
Professor of Medicine,
Harvard Medical School, Victoria P. Werth
Director, Clinical Rheumatology, University of Pennsylvania,
Massachusetts General Hospital, School of Medicine, Perelman Center for Advanced Medicine,
Boston, Massachusetts, USA Philadelphia, PA USA

Zoltán Szekanecz Cornelia M. Weyand


Department of Rheumatology, Department of Internal Medicine,
University of Debrecen Medical and Health Sciences Center, Division of Rheumatology,
Debrecen, Hungary Mayo Clinic and College of Medicine,
Rochester, MN;
Gim Gee Teng Department of Medicine,
University Medicine Cluster, Division of Immunology and Rheumatology,
Division of Rheumatology, Stanford University School of Medicine,
National University Health System, Stanford, CA, USA
Singapore; Department of Medicine,
Yong Loo Lin School of Medicine, Fredrick M. Wigley
National University of Singapore, Singapore Professor of Medicine,
Associate Director,
Benjamin Terrier Division of Rheumatology,
Department of Internal Medicine, Director, Johns Hopkins Scleroderma Center,
Université Pierre et Marie Curie, Johns Hopkins University School of Medicine,
Groupe Hospitalier Pitié-Salpêtrière, Baltimore, Maryland, USA
Paris, France
Hasan Yazici
Sebastian Unizony Professor of Medicine,
Rheumatology Unit, Division of Rheumatology,
Massachusetts General Hospital, Department of Medicine,
Boston, MA, USA Cerrahpasa Medical Faculty,
John E. Volanakis University of Istanbul, Istanbul, Turkey
Professor Emeritus of Medicine,
University of Alabama at Birmingham,
Birmingham, AL, USA
SECTION 1

Introductory chapters
CHAPTER 1

Nomenclature and
classification of
vasculitic syndromes
Gene V. Ball and S. Louis Bridges, Jr

The inadequacies of schemes for classifying vasculitis, and their that histological diagnosis is not always possible. Although the
misapplication to the diagnosis of an individual’s illness, are ACR classification criteria have been useful for establishing uni-
well recognized. The most frequently cited classification in the form criteria for inclusion of patients in research studies, they were
recent past was devised by a committee of the American College promulgated before widespread availability of reliable assays for
of Rheumatology (ACR) for the purpose of facilitating research ANCA. They did not differentiate PAN and microscopic polyangii-
(Bloch et  al. 1990; Fries et  al. 1990; Hunder et  al. 1990a). The tis (MPA); however, the specificity for granulomatosis with poly-
authors of this scheme for classifying seven distinct vasculitic angiitis (WG) and eosinophilic granulomatosis with polyangiitis
syndromes (Arend et al. 1990; Calabrese et al. 1990; Hunder et al. (Churg–Strauss syndrome (CSS)) appear to be 92% and 99.7%,
1990b; Leavitt et al. 1990; Lightfoot et al. 1990; Masi et al. 1990; respectively.
Mills et al. 1990) stated explicitly that the criteria were not meant Because of confusion concerning classification vis-à-vis diagno-
for diagnostic purposes in an individual patient even though they sis, the dictionary definitions of these two words are useful. The
have been useful in differentiating one vasculitis syndrome from Oxford English Dictionary defines classification as arranging in
another. Recognizing the widespread use of the criteria for diag- classes according to common characteristics or affinities. Diagnosis
nosis, Rao, Allen, and Pincus (1998) evaluated their effectiveness is defined as identification of a disease by careful investigation of
in diagnosing Wegener’s granulomatosis (WG) (now referred to its symptoms and history, or distinctive characterization in pre-
as granulomatosis with polyangiitis), giant cell arteritis (GCA), cise terms. The definition of diagnosis stresses the identification of
polyarteritis nodosa (PAN), and hypersensitivity vasculitis in 198 the nature and cause of a disease through evaluation of all clini-
patients who were referred to medical centres with diagnoses of cal and laboratory data. Jennette et al. (1994), in their influential
possible vasculitis. They concluded that when the criteria were proposal on the nomenclature of systemic vasculitides, observed
applied to these patients, their positive predictive values for WG, that ‘it is advantageous for everyone to use the same name for the
GCA, PAN, and hypersensitivity vasculitis ranged from only 17 to same disease, and to do this, there must be clear agreement on the
29%. Thus, there is empirical evidence that the ACR criteria should definition of the name and therefore of the disease’. This seems
not be used for diagnosis of specific syndromes in a patient who straightforward; however, while acknowledging that a diagnosis is,
does not have a confirmed diagnosis of vasculitis. This is in accord- in essence, the name of a disease, they disavowed determination of
ance with the intent of the committee: that the criteria not be used clinical criteria required to classify or diagnose individual patients.
to distinguish vasculitis from other diseases, and that the criteria Diagnostic criteria would require analysis of large numbers of
were established to insure uniformity in epidemiological and other patients with unequivocal defining features. Jennette et al. (1994)
research studies (Hunder et al. 1990a). The ACR and later defini- tabulated the names and definitions of ten of the more common
tional criteria require emphatically that vasculitis be established vasculitides. These definitions are expanded in the chapters of this
as a prerequisite for defining a specific vasculitic syndrome. For text dealing with individual diseases.
most syndromes, a histological diagnosis of vasculitis, whose clas- The ambiguities of clinical presentations, limited diagnostic
sical features are endothelial swelling, inflammatory infiltrates, and laboratory tests, and difficulty in obtaining appropriate tissue for
fibrinoid necrosis, has been required or recommended strongly but histological examination (which may not provide categorical infor-
the biopsy findings must be correlated with clinical and laboratory mation) may impede precise diagnosis/ classification at any time.
data, and with the understanding that the histological features of Furthermore, a diagnosis that is perfectly tenable at a given time,
vascular lesions may change with time (see Chapter 9). may later be inappropriate for a patient whose disease has evolved
The concept of surrogate markers of vasculitis such as anti- over time. This is particularly true of cutaneous leukocytoclastic
neutrophil cytoplasmic antibodies (ANCA) and imaging studies, angiitis, probably the most common of the vasculitides, which may
which are not features of the ACR classification criteria, recognizes be the sole manifestation of a vasculitis that later blossoms into
4 SECTION 1 introductory chapters

a systemic disease. This concept of a changing clinical condition, Table 1.1 Names for vasculitides adopted by the 2012 International
requiring the rethinking of a diagnosis, applies as well to many Chapel Hill Consensus Conference on the Nomenclature of Vasculitides
non-vasculitic diseases.
One of the first descriptions of a specific systemic vasculitis was Large-vessel vasculitis (LVV)
that of polyarteritis (periarteritis) nodosa (PAN) by Kussmaul and Takayasu’s arteritis (TAK)
Maier in 1866. Despite their detailed and careful description of the Giant cell arteritis (GCA)
gross and microscopic anatomy of a disease defined by anatomical Medium-vessel vasculitis (MVV)
characteristics, the term was used thereafter in a generic sense to Polyarteritis nodosa (PAN)
encompass all types of vasculitis. Other distinctive vasculitic syn- Kawasaki’s disease (KD)
dromes were recognized as WG in 1936, and CSS in 1951, and in
1952, Zeek proposed a classification of vasculitis, from which most, Small-vessel vasculitis (SVV)
if not all, others have descended. Her ‘necrotizing angiitis’ differ- Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV)
entiated hypersensitivity angiitis; allergic granulomatous angiitis; Microscopic polyangiitis (MPA)
rheumatic arteritis; periarteritis nodosa; and temporal arteritis. Granulomatosis with polyangiitis (Wegener’s) (GPA)
Disease characteristics that have been used for diagnosis/ classi- Eosinophilic granulomatosis with polyangiitis (Churg–Strauss) (EGPA)
fication have included: size, type and distribution of involved ves- Immune complex SVV
sels; histological appearance of the vessels and extravascular lesions; Antiglomerular basement membrane (anti-GBM) disease
clinical and laboratory features; and associations with other disor- Cryoglobulinaemic vasculitis (CV)
ders. The size of diseased vessels has been the major criterion for IgA vasculitis (Henoch–Schönlein) (IgAV)
classifying these disorders in most schemes, as size is a major deter- Hypocomplementaemic urticarial vasculitis (HUV) (anti-C1q vasculitis)
minant of their clinical symptoms. Large size signifies the aorta and Variable-vessel vasculitis (VVV)
its branches; medium signifies vessels that, while smaller than the Behçet’s disease (BD)
major branches of the aorta, contain an internal elastic membrane Cogan’s syndrome (CS)
as well as muscular media and adventitia. Small size denotes cap-
Single-organ vasculitis (SOV)
illaries, and postcapillary venules and arterioles. The value of this
Cutaneous leukocytoclastic angiitis
diagnostic classification is limited by the difficulty in acquiring tis-
Cutaneous arteritis
sue from visceral organs, as well as the variation within major syn-
Primary central nervous system vasculitis
dromes in regards to the predominant size of affected vessels. For
Isolated aortitis
example, although GCA is predominantly a disease of large arteries,
Others
smaller vessels are sometimes involved. Another example again uses
GCA; this disease, described by Hutchinson in 1890, may differ in its Vasculitis associated with systemic disease
clinical features from Takayasu’s arteritis (TA), which was described Lupus vasculitis
in 1909, yet both affect large arteries primarily, as do other, albeit, Rheumatoid vasculitis
rare syndromes. The distinctions between these two syndromes are Sarcoid vasculitis
based largely on age of onset: GCA after 50 years, and TA before Others
age 50. Any valid classification that will be used for diagnostic pur-
Vasculitis associated with probable aetiology
poses must be based on distinctive features. These include: the organ
Hepatitis C virus-associated cryoglobulinaemic vasculitis
distribution of lesions; the presence or absence of granulomatous
Hepatitis B virus-associated vasculitis
inflammation; the presence of certain infections; autoantibodies
Syphilis-associated aortitis
such as ANCA; other laboratory features; and the age of the patient.
Drug-associated immune complex vasculitis
The Chapel Hill Consensus Conference 1994 (CHCC) on the
Drug-associated ANCA-associated vasculitis
Nomenclature of Systemic Vasculitides definition of PAN made
Cancer-associated vasculitis
it accord more closely to the classic description by Kussmaul and
Others
Maier (1866) of a medium or small sized arteritis, without dis-
ease in arterioles, venules, and capillaries, and with sparing of (Reprinted with permission from Jennette J.C. et al. Arthritis and Rheumatism, 65, 1–11.
pulmonary arteries (Jennette et al. 1994). The CHCC nomencla- John Wiley and Sons. Copyright © 2013 by the American College of Rheumatology)

ture purges PAN of small-vessel disease, assigning this and glo-


merulonephritis to the syndrome of MPA, which was probably
first described (as microscopic periarteritis nodosa) in 1923 by characteristic of MPA, CSS, and WG. ANCA are found in about
Friedrich Wohlwill (Wohlwill 1998). Cupps and Fauci, in their 75% of these three disorders. In general, about 75% of patients
textbook The Vasculitides (1981), referred to a group of patients with WG are cANCA positive; about 40–75% of CSS patients are
with clinical and pathological characteristics of both classic PAN positive for either cANCA or pANCA, and pANCA is found in
and allergic angiitis, but who did not fit precisely into either cat- about 50% of MPA. Either ANCA can be found in any of these
egory. Lesions of muscular arteries, arterioles, and venules typi- three disorders, and are absent in 5–30%.
fied the syndrome, which they referred to as an ‘overlap’. This is Citing advances in understanding of vasculitis, the most recent
probably the group now referred to as ANCA-associated vasculitis Chapel Hill Consensus Conference Nomenclature of Vasculitides
which differs from classic PAN, not only in size of affected vessels, (CHCC 2012) was convened to improve nomenclature, to change
but in absence of immune complex deposits, the frequent pres- names and definitions, and add categories of vasculitis that were
ence of ANCA, and involvement of lungs and glomeruli, which are not included in the previous CHCC. The recommendations were
CHAPTER 1 nomenclature and classification of vasculitic syndromes 5

reported by a group of 28 experts representing medical specialties American College of Rheumatology 1990 criteria for the classification of
most germane to vasculitis. The authors stressed that CHCC is a vasculitis. Introduction. Arthritis and Rheumatism, 33, 1065–7.
nomenclature; therefore, it is not a classification system that speci- Hunder, G.G., Bloch, D.A., Michel, B.A., Stevens, M.B., Arend, W.P.,
Calabrese, L.H., Edworthy, S.M., Fauci, A.S., Leavitt, R.Y., and Lie, J.T.
fies what findings must be observed in a specific patient, nor is it a
(1990b). The American College of Rheumatology 1990 criteria for the
diagnostic system directing clinical management. They explained classification of giant cell arteritis. Arthritis and Rheumatism, 33, 1122–8.
their terminology by way of examples: diagnosis is the name of a Jennette, J.C., Falk, R.J., Andrassy, K., Bacon, P.A., Churg, J., Gross, W.L.,
disease, for example polyarteritis nodosa. Definition is based on Hagen, E.C., Hoffman, G.S., Hunder, G.G., and Kallenberg, C.G. (1994).
the disease processes in a patient that justifies use of the diagno- Nomenclature of systemic vasculitides. Proposal of an international con-
sis. Classification criteria are observations that classify a specific sensus conference. Arthritis and Rheumatism, 37, 187–92.
patient into a standardized category for study. Diagnostic criteria Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F.,
are observations that demonstrate or predict the presence of the Flores-Suarez, L.F., Gross, W.L., Guillevin, L., Hagen, E.C., Hoffman,
defining features of the disease in a specific patient. G.S., Jayne, D.R., Kallenberg, C.G.M., Lamprecht, P., Langford, C.A.,
Luqmani, R.A., Mahr, A.D., Matteson, E.L., Merkel, P.A., Ozen, S.,
A major feature of CHCC 2012 is the phasing out of eponyms
Pusey, C.D., Rasmussen, N., Rees, A.J., Scott, D.G.I., Specks, U., Stone,
when there was sufficient understanding of the pathophysiology J.H., Takahashi, K., and Watts, R.A. (2013). 2012 Revised International
to propose an alternative name. Major shifts away from eponyms Chapel Hill Consensus Conference Nomenclature of Vasculitides.
are granulomatosis with polyangiitis (Wegener’s granulomatosis) Arthritis and Rheumatism, 65, 1–11.
(GPA); IgA vasculitis (Henoch–Schönlein) (IgAV); eosinophilic Kussmaul, A. and Maier, R. (1866). On a previously undescribed peculiar
granulomatosis with polyangiitis (Churg–Strauss) (EGPA). The arterial disease (periarteritis nodosa), accompanied by Bright’s disease
nomenclature used in each chapter of this text follows the recom- and rapidly progressive general muscle weakness. Deutsches Archiv fur
mendations of CHCC 2012, which are embodied in Table 1.1. klinische Medicin, 1, 484. (Translated by E. Matteson.)
Leavitt, R.Y., Fauci, A.S., Bloch, D.A., Michel, B.A., Hunder, G.G., Arend,
References W.P., Calabrese, L.H., Fries, J.F., Lie, J.T., and Lightfoot, R.W. Jr (1990).
The American College of Rheumatology 1990 criteria for the classifi-
Arend, W.P., Michel, B.A., Bloch, D.A., Hunder, G.G., Calabrese, L.H., cation of Wegener’s granulomatosis. Arthritis and Rheumatism, 33,
Edworthy, S.M., Fauci, A.S., Leavitt, R.Y., Lie, J.T., and Lightfoot, R.W. 1101–7.
Jr (1990). The American College of Rheumatology 1990 criteria for Lightfoot, R.W. Jr, Michel, B.A., Bloch, D.A., Hunder, G.G., Zvaifler, N.J.,
the classification of Takayasu arteritis. Arthritis and Rheumatism, 33, McShane, D.J., Arend, W.P., Calabrese,L.H., Leavitt, R.Y., and Lie, J.T.
1129–34. (1990). The American College of Rheumatology 1990 criteria for the
Bloch, D.A., Michel, B.A., Hunder, G.G., McShane, D.J., Arend, W.P., classification of polyarteritis nodosa. Arthritis and Rheumatism, 33,
Calabrese, L.H., Edworthy, S.M., Fauci, A.S., Fries, J.F., and Leavitt, 1088–93.
R.Y. (1990). The American College of Rheumatology 1990 criteria Masi, A.T., Hunder, G.G., Lie, J.T., Michel, B.A., Bloch, D.A., Arend, W.P.,
for the classification of vasculitis. Patients and methods. Arthritis and Calabrese, L.H., Edworthy, S.M., Fauci, A.S., and Leavitt, R.Y. (1990).
Rheumatism, 33, 1068–73. The American College of Rhematology 1990 criteria for the classifica-
Calabrese, L.H., Michel, B.A., Bloch, D.A., Arend, W.P., Edworthy, S.M., Fauci, tion of Churg-Strauss syndrome (allergic granulomatosis and angiitis).
A.S., Fries, J.F., Hunder, G.G., Leavitt, R.Y., and Lie, J.T. (1990). The Arthritis and Rheumatism, 33, 1094–100.
American College of Rheumatology 1990 criteria for the classification of Mills, J.A., Michel, B.A., Bloch, D.A., Calabrese, L.H., Hunder, G.G., Arend,
hyper-sensitivity vasculitis. Arthritis and Rheumatism, 33, 1108–13. W.P., Edworthy, S.M., Fauci, A.S., Leavitt, R.Y., and Lie, J.T. (1990). The
Cupps, T.R. and Fauci, A.S. (1981). The Vasculitides:  Major Problems in American College of Rheumatology 1990 criteria for the classification
Internal Medicine, Vol. 21. W.B. Saunders, Philadelphia. of Henoch-Schönlein purpura. Arthritis and Rheumatism, 33, 1114–21.
Fries, J.F., Hunder, G.G., Bloch, D.A., Michel, B.A., Arend, W.P., Calabrese, Rao, J.K., Allen, N.B., and Pincus, T. (1998). Limitations of the 1990 American
L.H., Fauci, A.S., Leavitt, R.Y., Lie, J.T., and Lightfoot, R.W. Jr (1990). The College of Rheumatology classification criteria in the diagnosis of vascu-
American College of Rheumatology 1990 criteria for the classification of litis. Annals of Internal Medicine, 129, 345–52.
vasculitis. Summary. Arthritis and Rheumatism, 33, 1135–6. Wohlwill, F. (1998). On the only microscopically recognizable form of peri-
Hunder, G.G., Arend, W.P., Bloch, D.A., Calabrese, L.H., Fauci, A.S., Fries, arteritis nodosa. (Translated by E. Matteson with H.R. Matteson.) Mayo
J.F., Leavitt, R.Y., Lie, J.T., Lightfoot, R.W. Jr and Masi, A.T. (1990a). The Clinic Scientific Press, Rochester, Minnesota.
CHAPTER 2

Epidemiology of vasculitis
Richard A. Watts and David G.I. Scott

Introduction international consensus meeting held in 2011 to reflect changes in


our understanding of the aetiopathogenesis of vasculitis (Jennette
The vasculitides are a group of relatively uncommon conditions et al. 2013). A new tree hierarchy has been developed which rec-
whose aetiology is still poorly understood, thus impeding accurate ognizes that some conditions cannot be simply classified by vessel
epidemiological studies. They affect individuals of all ages but are size. ANCA-associated vasculitis is recognized as a specific group
predominately diseases of the extremes of age (Figure 2.1). Over the
past few years, prospective, population-based registers have been
established and results from these are now becoming available.
Table 2.1 Classification of the vasculitides
The epidemiology of rare diseases such as the vasculitides poses
considerable challenges to epidemiologists. The first is the difficulty
Dominant Idiopathic (primary) Probable aetiology
of defining a case with a lack of clear distinction between the differ-
vessel (secondary)
ent disorders. The classification of the vasculitides is based on the
size of the predominant vessels involved (Table 2.1). Until the 1990s, Large Takayasu’s arteritis Aortitis associated with
there were no validated classification criteria. This was addressed by Giant cell arteritis Tuberculosis,
the American College of Rheumatology (ACR) in 1990, which pro- Syphilis,
posed criteria for the classification of seven different vasculitides RA, AS
with sensitivities varying from 71.0 to 95.3% and specificities of
78.7 to 99.7% (Fries et al. 1990). The most sensitive and specific cri- Medium Polyarteritis nodosa Polyarteritis nodosa (HBV
teria were found in eosinophilic granulomatosis with polyangiitis associated)
(Churg–Strauss) (EGPA), giant cell arteritis (GCA), and Takayasu’s Kawasaki’s disease
arteritis (TA); hypersensitivity (leukocytoclastic) vasculitis was Small
the least well-defined condition. This development was important
ANCA Microscopic polyangiitis Drugs
because it allowed epidemiological studies to be performed using
established criteria. The ACR criteria are not perfect; the main criti- Propylthiouracila
cism is that they do not include microscopic polyangiitis (MPA) or Hydrallazinea
consider antineutrophil cytoplasmic antibodies (ANCA). Granulomatosis with
In 1994, the Chapel Hill Consensus Conference (CHCC) pro- polyangiitis (Wegener’s)
duced definitions for vasculitis (Jennette et al. 1994). They included Eosinophilic granulomatosis
for the first time MPA, but were not intended as classification or with polyangiitis
diagnostic criteria. They have recently been updated following an (Churg–Strauss)
Immune Cryoglobulinaemic vasculitis Cryoglobulinaemic vasculitis
1 complex (non-HCV) (HCV associated)
2 Anti-GBM disease Vasculitis associated with RA,
1 Kawasaki disease
4 SLE, Sjögren’s syndrome, and
2 IgA vasculitis (Henoch-SchÖnlein)
many drugsb
3 ANCA-associated vasculitis
4 Giant cell arteritis IgA vasculitis
(Henoch–Schönlein)
Incidence

Hypocomplementaemic
vasculitis
Variable Behçet’s syndrome
3 Cogan’s syndrome
a Most commonly induce MPO-ANCA
b For example sulfonamides, penicillins, thiazide diuretics, and many others.
10 20 30 40 50 60 70 80 Age RA, rheumatoid arthritis; AS, ankylosing spondylitis; SLE, systemic lupus erythematosus;
Fig. 2.1 Relative incidence of vasculitis by age. HBV, hepatitis B virus; HCV, hepatitis C virus.
8 SECTION 1 introductory chapters

of small vessel vasculitis different from immune complex-mediated Large-vessel vasculitis


vasculitis. The hierarchy now includes vasculitis affecting ves-
sels of variable size, single-organ vasculitis, together with vascu- Giant cell arteritis
litis associated either with systemic disease or specific aetiologies. Giant cell arteritis is a vasculitic disease in which the characteristic
A new nomenclature was adopted with a move away from eponyms feature is the presence of giant cells in a biopsy of a large artery.
towards names reflecting pathology or aetiopathogenesis follow- The CHCC 2012 defined GCA as ‘Arteritis, often granulomatous,
ing the introduction of the term granulomatosis with polyangii- usually affecting the aorta and/or its major branches, with a predi-
tis (Wegener’s) (GPA) for Wegener’s granulomatosis. The name lection for the branches of the carotid and vertebral arteries. Often
eosinophilic granulomatosis with polyangiitis (Churg–Strauss) involves the temporal artery. Onset usually in patients older than 50
was adopted for Churg–Strauss syndrome (EGPA), and IgA vas- and often associated with polymyalgia rheumatica.’ (Jennette et al.
culitis (IgAV) for Henoch–Schönlein purpura. Definitions were 2013). Although the ACR criteria for GCA (Hunder et al. 1990a)
developed for new categories of conditions including single-organ do not require histological evidence of arteritis, many studies only
vasculitis, vasculitis associated with specific aetiologies including report biopsy-positive cases. Clearly the biopsy rate and inten-
systemic disease (rheumatoid arthritis, systemic lupus erythemato- sity of histological search for evidence of vasculitis influence the
sus), and aetiologies such as infection (cryoglobulinaemia, hepati- reported incidence. Furthermore, many of the studies have been
tis B and C), or drugs (e.g. propylthiouracil). hospital based and retrospective. GCA is closely related to poly-
The ACR criteria and CHCC (1994) definitions are widely used myalgia rheumatica (PMR); population-based studies of PMR have
for epidemiological studies. Comparative epidemiology requires shown that 16–21% of patients have biopsy-proven GCA (Salvarani
the use of well-validated and generally accepted classification cri- et  al. 1995a; Franzen et  al. 1992). Conversely, 40–60% of GCA
teria. For some types of vasculitis, for example GCA, TA, GPA, and patients have PMR symptoms (Salvarani et al. 1995b). Many stud-
EGPA, this condition is satisfied; however, for others such as MPA ies include patients with PMR and do not clearly distinguish those
and polyarteritis nodosa (PAN) there is no consensus. This lack of with pure GCA.
consensus on case definition and distinction between the different Prospective studies from Scandinavia have reported annual inci-
types of vasculitis has hindered progress. While differences in inci- dence figures for biopsy-proven GCA of 15–35 per 100 000 indi-
dence between geographic areas may reflect a genuine difference viduals aged over 50 years; a similar rate has been reported from
due to environmental or ethnic factors, there remains the possi- Olmsted County (Minnesota, USA) and in a community-based
bility that a systematic difference in the application of the criteria/ study in the UK (Table 2.2). There is an increasing incidence with
definitions may be responsible for at least some of the variation. age, peaking in people aged 80 years or greater; very few cases occur
There is therefore an urgent need to develop a better classification in people aged less than 50 years. Most series from Northern Europe
system for the vasculitides (Watts et al. 2011). report a greater incidence in women, with a female to male ratio
The second difficulty is case capture. The vasculitides are rare and of around 2.5:1; the female excess is lower in southern European
therefore a large population is required to determine the incidence countries and Israel (Bas-Lando et al. 2007; Salvarani et al. 1991),
and prevalence, and this poses questions of feasibility. A large pop- whilst in Northwest Spain, India, and Turkey the ratio is equal
ulation increases the risk of incomplete case detection but permits (González-Gay et al. 2007; Singh et al. 2010; Pamuk et al. 2009).
a reasonable number of cases to be collected in a practicable time
Time trends
frame; whereas a smaller population requires a much longer time
Several studies have suggested an increase in the incidence of GCA
frame to collect the necessary cases, which also may not be feasible.
with time. In Olmsted County between 1950–54 and 1980–84
Statistical methods of capture–recapture analysis enable estimates
there was an increase from 6.7/100  000 to 28.5/100  000 in per-
to be made of the number of missing cases.
sons aged >50 years. The rate then stabilized and has not risen
The third difficulty is case ascertainment. Some vasculitides such
further (Salvarani et  al. 2004). A  similar increase in incidence
as the ANCA-associated vasculitides (AAV) are virtually always
has been documented in Göteborg, Sweden between 1976 and
managed in secondary care, whereas GCA is often managed solely
1995 from 16.8/100  000 to 30.1/100  000 persons aged >50 years
in primary care. Different methods of case capture are required.
(Petursdottir et  al. 1999). The estimated rate of increase was
The rarity of the conditions makes prospective case–control studies
10.9% per year. González-Gay and colleagues in northwest Spain
difficult to conduct because the population size required to achieve
reported an increasing incidence of GCA between 1981 and 1998,
statistical confidence is in excess of that readily available. Thus,
but noted that the frequency of classic manifestations decreased
much of the data on risk factors are derived from retrospective
(González-Gay et al. 1997) and that the increase was more marked
studies with inherent potential bias.
in women (González-Gay and García-Porrúa 1999). This increase
The aetiology of vasculitis is unknown but is clearly multifacto-
is independent of changing population structures in which there is
rial; among the influences on disease expression are ethnicity, genes
an increase in the elderly and very elderly (aged >80 years) popula-
(HLA and others), gender, and environment (UV light, infections,
tion (Salvarani et al. 2004; Nordborg et al. 2003). A recent study
toxins, drugs, smoking, and surgery). There are sufficient differ-
from a primary care database in the UK has reported that there is
ences, even with the limited epidemiological data available, to
a significant birth cohort effect with the incidence increasing with
suggest that not all these factors work in the same direction in the
more recent birth, suggesting the presence of risk factors to which
various vasculitic syndromes described. In this chapter, we will
individuals born more recently were exposed (Dubreuil et al. 2010).
review the available evidence and attempt to identify the key epide-
miological factors involved in the major vasculitis syndromes. We Geographical factors
have structured this around a classification system based on the size Giant cell arteritis seems to be more frequent in high latitudes.
of vessels predominately involved (Table 2.1). In the northern hemisphere there is a significant increase in both
CHAPTER 2 epidemiology of vasculitis 9

Table 2.2 Reported annual incidence rates for giant cell arteritis

Place Period Incidencea Case definition Study type Reference


Europe
Lothian, Scotland 1964–77 4.2 Biopsy Retrospective Jonasson et al. 1979
Tampere, Finland 1969–89 7.2 Biopsy Retrospective Rajala et al. 1993
Loire, France 1970–79 9.4 Biopsy/clinical Retrospective Barrier et al. 1982
Goteborg, Sweden 1973–75 16.8 Biopsy Retrospective Bengtsson and Malmvall
1981
Goteborg, Sweden 1977–86 18.3 Biopsy Retrospective Nordborg and Bengtsson
1990
Reggio Emilia, Italy 1980–88 6.9 Biopsy Retrospective Salvarani et al. 1991
Lugo, NW Spain 1981–98 10.3 Biopsy Retrospective González-Gay et al. 2001
Ribe, Denmark 1982 23.3 Biopsy Prospective Boesen and Sorensen 1987
Denmark 1982–94 20.4 Biopsy Prospective (since 1984) Elling et al. 1996
Iceland 1984–90 27.0 ACR b Retrospective Baldursson et al. 1994
W Nyland, Finland 1984–87 17.4 Biopsy/clinical Retrospective Franzen et al. 1992
W Nyland, Finland 1987–88 26.2 Biopsy/clinical Prospective Franzen et al. 1992
S Sweden 1986 33.6 Biopsy/clinical Prospective, population Noltrop and Svensson 1991
Frederiksborg, Denmark 1986–88 17.6 Biopsy/clinical Retrospective Fledelius and Nissen 1992
Aust Agder, Norway 1987–94 29.0 Biopsy/clinical Retrospective, hospital Gran and Myklebust 1997
Bodø, Norway 1992–96 27.5 Biopsy/ACR Retrospective, hospital Haugeberg et al. 2003
Ålesund, Norway 1992–96 36.7 Biopsy/ACR Retrospective, hospital Haugeberg et al. 2003
Kristiansand, S. Norway 1992–96 32.8 Biopsy/ACR Retrospective, hospital Haugeberg et al. 2003
Goteborg, Sweden 1976–95 22.2 Biopsy, clinical Retrospective Nordborg et al. 2003
Schleswig Holstein, Germany 1998–2002 3.2 CHCC Prospective, population Reinhold-Keller et al. 2005
Vilnius, Lithuania 1990–99 0.72 ACR Prospective, hospital Dadoniene et al. 2005
Israel 1960–78 0.5 Biopsy Retrospective Friedman et al. 1982
Jerusalem, Israel 1980–91 10.2 Biopsy Retrospective Sonnenblick et al. 1986
UK GPRD 1990–2001 22.0 Clinicalc Prospective Smeeth et al. 2006
Edirne, Turkey 2003–2009? 1.14 Clinical Retrospective Pamuk et al. 2009
Jerusalem 1980–2004 11.3 Biopsy + ACR Retrospective Bas-Lando et al. 2007
9.5 (Biopsy +ve)
Lugo, Spain 1981–2005 10.13 Biopsy +ve GCA Prospective González-Gay et al. 2007
UK Thin GP registry 1990–2008 Males 13/100 000 Clinical Prospective Dubreuil et al. 2010
Females 30/100 000
Americas
Olmsted County, MN 1950–99 18.8 ACRb Retrospective (to 1985), Salvarani et al. 1995a, 2004
population
Olmsted County, MN 2000–04 18.9 ACR Prospective population Kermani et al. 2010
Memphis, TN 1971–80 1.58 all Biopsy/clinical Retrospective Smith et al. 1983
2.24 white
0.36 black
Saskatoon, Canada 1998–2003 9.4 (inc European Biopsy +ve Prospective Ramstead and Patel 2007
and natives)
7.7 Europeans
16 Aboriginals
Alaska 1983–2003 1.0 Biopsy +ve Retrospective Mader et al. 2009
Australasia
Otago, New Zealand 1996–2005 12.73 Biopsy Retrospective Abdul-Rahman et al. 2011
a Incidence figures/100 000 population aged over 50 years.
b ACR (1990) criteria.
c A sample of cases from the UK GPRD register was selected for validation of the diagnosis of GCA.
10 SECTION 1 introductory chapters

incidence and prevalence with increasingly northerly latitudes. The Northern European populations and this may explain the rarity of
highest incidence rates have reported from Scandinavia and the UK GCA in Japan.
(Table 2.2). Studies from southern Europe (Italy, Spain, and France), Other genetic factors will be important in the susceptibility to
have consistently reported lower incidence rates than those from GCA, particularly those involved in the immune and inflammatory
Scandinavia. In North America, there is a lower incidence in the pathways. No large-scale, genome-wide association study (GWAS)
southern USA Caucasian population (Smith et al. 1983) than in the has yet been performed in GCA to elucidate these potential genetic
Olmsted county population and an Alaskan population. Whether associations.
these changes reflect the different genetic backgrounds of the popu- Cardiovascular risk factors
lations and/or an additional environmental factor is unknown (see
In the Olmsted county population, smoking was associated with
Sections Ethnic differences, Genetic factors, and Environmental
a statistically significant, twofold increase in biopsy-proven GCA
factors).
(Machado et al. 1989). In a prospective case–control study from
Ethnic differences France, past or present smoking in women, but not men, was
Giant cell arteritis appears to be most common in Caucasian associated with GCA with an odds ratio of 6, and heavy smok-
populations and is uncommon in non-Caucasians. The inci- ing (defined as greater than 10 pack-years) with an odds ratio of
dence is highest in Scandinavians and in populations descended 16. Previous atheromatous disease was associated with a 4.5-fold
from them. The Viking heritage of the UK might be responsible increase in women with GCA (Duhaut et  al. 1998). This was an
for the relatively high frequency of GCA seen in a study from independent effect. For men, no significant cardiovascular risk fac-
the UK General Practice Research Database (UKGPRD); inter- tors for GCA were identified. The same group of investigators also
estingly, the incidence is highest in East Anglia, an area with reported that previous pregnancy was not an additional risk fac-
marked Viking ancestry (Smeeth et  al. 2006). The Olmsted tor for GCA, and might even be protective (Duhaut et al. 1999).
county population is descended from Scandinavian migrants The reason for this is unclear but may be related to the protective
to the USA. GCA is much less common in southern European effect of pregnancy on cardiovascular disease. In Scandinavia, early
populations, which have a different genetic background (Tian menopause, low body mass index, and smoking were independent
et al. 2009). risk factors for development of GCA (Larsson et al. 2006). In the
There are few studies directly comparing different popula- UK, ischaemic manifestations have been associated with residence
tions. A  study from Tennessee (USA) reported a low incidence in areas of socioeconomic deprivation, but not with traditional car-
in African-Americans (0.36/100  000 aged >50  years) compared diovascular risk factors (Mackie et al. 2011).
with 2.24/100  000 in the white population (Smith et  al. 1983). Environmental factors
A  retrospective review from the Texas Gulf Coast reported that
The incidence of GCA shows a marked variation with latitude.
13/27 patients were black women, but that the disease was rare in
Multiple regression analysis correcting for the effect of increas-
Hispanics (González et al. 1989). No incidence figures are available
ing incidence with time suggests that there is a significant trend
from either study. In contrast, a survey from Florida did show a dif-
to increasing incidence with more northerly (higher) latitude
ference in occurrence between Hispanics and non-Hispanics (Lam
(Watts et  al. 2000b). This geographical trend is similar to that
et al. 2007). Referral bias might account for their results. Two stud-
seen in multiple sclerosis and rheumatoid arthritis in both hemi-
ies from California have suggested that GCA is much less common
spheres. Exposure to ultraviolet (UV) light has been proposed
in Asians than Caucasians (Liu et al. 2001; Pereira et al. 2011), with
as an aetiological factor for autoimmunity:  the amount of UV
an odds ratio of 0.049.
radiation (UVR) reaching the earth’s surface varies inversely with
There is a low prevalence in Japan compared with Europe
latitude; UVR down-regulates cellular immunity, and low levels
(Kobayashi et al. 2003). A study from Saskatoon (Canada) suggested
of UVR are associated with low vitamin D synthesis (Staples et al.
a higher frequency in the Aboriginal population (16/100 000) com-
2003). Hence UVR might have a protective effect on the develop-
pared with 7.7/100 000 in the Caucasian population. The numbers
ment of GCA. Further studies are required to investigate the role
of subjects was very small (two non-Caucasians) (Ramstead and
of UVR and vitamin D in the development of GCA. An alterna-
Patel 2007).
tive explanation is that there might be an infectious agent which
Genetic factors is either more common or operates more effectively at higher
There is an increased frequency of GCA among first-degree rela- latitudes.
tives and occasional familial cases, implying a possible genetic pre- The Olmsted County study identified peaks in incidence occur-
disposition to GCA (Liozon et al. 2009). ring every 7 years, a finding that would be consistent with an
In Europe, the marked difference between Northern and infectious aetiology (Salvarani et  al. 1995a). In Denmark, peak
Southern populations might reflect the different genetic make up incidences were correlated with the occurrence of epidemics of
of those populations. Detailed genetic population substructure Mycoplasma pneumoniae infection (Elling et  al. 1996). Using a
studies using genome-wide single nucleotide polymorphism (SNP) matched case–control method, Russo et al. (1995) showed a cor-
panels show that Southern European populations (Italian, Spanish, relation between infection and onset of GCA but could not iden-
Portuguese, and Greek) can be distinguished from Northern tify a specific infection. In addition to cyclic peaks, occasional
European populations (Tian et al. 2009). clusters have been reported; in Jerusalem, Sonnenblick and col-
Susceptibility to GCA has been associated with an increased leagues observed five cases in a 7-week period when the expected
incidence of HLA-DR4 and HLA-DRB1*0401 (González-Gay background rate was one case per year (Sonnenblick et al. 1986).
et al. 2003a). The HLA-DRB1*0401 and HLA-DRB1*0404 alleles Temporal clusters have been reported from general practice in
are very much less common in the Japanese population than in the UK, with six cases occurring among a population of 4400 in
CHAPTER 2 epidemiology of vasculitis 11

a 7-month period (McCreay 1986). Several cases of GCA occur- Table 2.3 Reported annual incidence rates for Takayasu’s arteritis
ring in families or in close temporal relation have been reported
and used to adduce an infectious or genetic aetiology; both sibling Place Period Incidencea Reference
and spouse pairs have been reported (Liozon et al. 2009). In con-
Europe
trast, neither the Swedish nor Spanish groups observed any cycli-
Sweden 1969–75 0.8 Waern et al. 1983
cal fluctuations (Petursdottir et al. 1999; González-Gay et al. 2001). Schleswig Holstein 1998–02 04–1.0 Reinhold-Keller
In Germany, GCA was observed to be more prevalent in urban et al. 2005
environments compared with rural areas, with a 2.25 relative risk Vilnius, Lithuania 1990–99 1.3 Dadoniene
(Reinhold-Keller et al. 2000). et al. 2005
A seasonal pattern of onset has been identified by several stud- United Kingdom 2000–05 0.8 (overall) Watts et al. 2009a
ies but the precise season of maximum occurrence has not been 0.3 (age <40 years)
consistent. The most recent nationwide, community-based study Norfolk, UK 2000–05 0.4 Watts et al. 2009a
(UKGPRD study) noted that GCA was more likely to be diagnosed Denmark 1990–09 0.4 Dreyer et al. 2011
in the warmer summer months (Smeeth et al. 2006). Turkey 2006–10 1.0 Kucukyavas et al.
The factors described above point towards an infectious aeti- 2012
ology; however, despite intensive searches no specific infectious Americas
agent has been identified. The striking feature of the epidemiol- Olmsted County, MN 1971–83 2.6 Hall et al. 1985
ogy of GCA is the age distribution. As the immune system ages,
Asia
immune competence declines and thymic T cell generation Japan 1982–84 1–2 Koide 1992
ceases (Goronzy and Weyand 2012) with consequent induction of
autoimmunity. Arabia
In summary, despite all the available epidemiological data on Kuwait 1989–94 2.2 (overall) El-Reshaid
GCA, including large studies looking at infectious causes, which 3.3 (age<40 years) et al. 1995
have been far more extensive than for most other vasculitides, no a Incidence expressed /million population
unifying hypothesis has emerged.

Takayasu’s arteritis Associations have been reported with cytokine genes such as IL-2,
Takayasu’s arteritis (TAK) was first described in Japan in 1908. Il-6, and IL-12 and the NFKBIL1 promoter region but not with
The CHCC 2012 defined TAK as ‘Arteritis, often granulomatous, autoimmunity-associated genes such as PTPN22 and PDCD1.
predominantly affecting the aorta and/or its major branches. Studies from East Asian countries and Turkey suggest that the
Onset is usually in patients younger than 50.’ (Jennette et  al. main HLA association is with HLA-B*52 (Kimura et al. 1996; Sahin
2013). Since then, it has generally been considered to be more et al. 2012).
common in Asia although it has been described world-wide. The
annual incidence is 0.5–3/million (Table 2.3). The peak age of dis- Medium-vessel vasculitis
ease onset is in the third decade and the disease is more common Kawasaki’s disease
in women. There are too few studies to reliably assess time trends,
Kawasaki’s disease (mucocutaneous lymph node syndrome) (KD)
but the available data suggest no major change (Table 2.3). TAK
is an acute vasculitis. The CHCC 2012 defined KD as ‘Arteritis
has been described in most ethnic groups including African, for
associated with the mucocutaneous lymph node syndrome and
which quite large series have been published (Vanoli et al. 2005;
predominantly affecting medium and small arteries. Coronary
Mwipatayi et al. 2005). TAK has a higher prevalence in Japan (40/
arteries are often involved. Aorta and large arteries may be
million) (Toshihiko 1996) compared with Europe (4–7/million)
involved. Usually occurs in infants and young children.’ (Jennette
(Waern et al. 1983; Watts et al. 2009a), but is slightly more com-
et al. 2013). The striking feature of the epidemiology of KD is the
mon in Turkey 15/million (Kucukyavas et  al. 2012). Five types
predominance of infants and young children. The condition was
of TAK are recognized, depending on the site of radiological
first recognized in Japan in 1967 (Kawasaki 1967). KD occurs in
involvement (Hata et al. 1996). The distribution of arterial lesions
both epidemic and endemic forms world-wide. The occurrence of
appears to vary between populations. In India and other South
epidemic KD has led to speculation that the condition is caused
Asian countries, the renal arteries are affected in the majority
by an infectious agent, but so far no specific organism has been
of patients; in South African patients, lower abdominal aorta
identified.
involvement with leg ischaemia is more common than in Japan.
Aneurysmal disease is much more common in Africa compared Geographical factors
with Japan (Mwipatayi et al. 2005). There are pronounced geographical variations in the incidence of
The aetiopathogenesis of TAK is unknown; possible environ- KD (Table  2.4). KD is most common in the Far East, especially
mental factors include mycobacterial infection because of the gran- Japan, Korea, and China, and relatively uncommon in the USA and
ulomatous nature of the disease and its more frequent occurrence Europe (Uehara and Belay 2012).
in areas endemic for TB, but no clear causal relationship has been Early studies on the epidemiology of KD came from Japan.
established. Nationwide surveys have been conducted every two year since 1970.
There is evidence for a genetic susceptibility to TAK; it has been The most recent survey covers 2007–08 (Nakamura et al. 2010). The
recorded in monozygotic twins and around 1% of Japanese patients overall incidence was 216.9/100 000 children younger than 5 years.
with TAK have an affected relative (Kobayashi and Numano 2002). The incidence is greater in boys (245.4/100 000 aged <5 years) than
12 SECTION 1 introductory chapters

Table 2.4 Reported annual incidence rates for Kawasaki’s disease rate amongst Japanese Americans is as high as that in Japan. The
occurrence amongst whites in Hawaii is similar to that seen in con-
Place Period Incidencea Reference tinental USA. These differences between ethnic groups in the same
environment, and presumably therefore exposed to the same infec-
Asia
tions, suggest that genetic factors are key in determining disease
Japan 2008 218.6 Nakamura et al. 2010
Korea 2006–08 113.1 Park et al. 2011 susceptibility.
Taiwan 2003–06 69.0 Huang et al. 2009 Genetic factors
China, Beijing 2004 55.1 Du et al. 2007
The genetics of KD have been studied using GWAS methodology
China, Shanghai 2007 53.3 Ma et al. 2010
in several populations from Japan and in Han Chinese. A  num-
China, Hong Kong 1997–2000 39.0 Ng et al. 2005
China, Sichuan 2001 9.8 Li et al. 2008 ber of susceptibility loci have been identified in T cell receptor
China, Jilin 1999–2008 11.1 Zhang et al. 2012 signalling, regulation of proinflammatory cytokines, HLA, and
India 2007 4.5b Singh et al. 2011 antibody-mediated immune responses (Onouchi et al. 2012; Lee
Thailand 2002 3.4 Durongpisitkul et al. 2006 et al. 2012; Tsai et al. 2011).
Americas Environmental factors
USA 2009 19.0 Uehara and Belay 2012 There is indirect evidence to support an infectious aetiology. KD
Hawaii 1996–2006 50.4 Holman et al. 2010 rarely occurs before the age of 6 months or after the age of 5 years,
California 1995–99 15.3 Chang et al. 2002b suggesting that early in life the infant is protected by passively
Georgia 1997–98 9.8 Gibbons et al. 2002 acquired maternal immunity, or that the immature immune system
Canada 2004–06 26.2 Lin et al. 2010
is unable to mount a response to induce KD. The low frequency
Guadelope, WI 1995–2000 25.4 Tourneux et al. 2005
after 5 years of age suggests early exposure to a common pathogen
Europe against which most children mount an appropriate and protective
UK 1998–2003 8.4 Harnden et al. 2009 immune response. In Japan, three epidemics have been observed,
UK, Birmingham 1996–99 5.5 Gardner-Medwin et al. 2002 in 1979, 1982, and 1986. Other outbreaks have been documented
Ireland 1996–2000 15.2 Lynch et al. 2003 in the USA in 1984–85, Canada in the 1980s, and Finland in 1981.
Finland 1992 7.2 Salo et al. 1993
Clustering and community-wide outbreaks have been quite widely
Denmark 1999–2004 4.9 Fischer et al. 2007
recorded.
Sweden 1990–92 6.2 Schiller et al. 1995
France 2005–06 9.0 Heuclin et al. 2009 A seasonal variation in KD has long been observed but the sea-
Italy 1981–82 14.7 Tamburlini et al. 1984 son of highest incidence varies. In Japan there is a bimodal distri-
Czech Republic 1997–99 1.6 Dolezalova et al. 2004 bution with peak incidence in winter (January) and summer (July)
and a nadir in October. This pattern is consistent throughout the
Australiasia
Japanese archipelago (Burns et al. 2005). Spring and summer pre-
Australia 1993–95 3.7 Royle et al. 1998
dominate in China (Du et al. 2007), and summer (June–July) and
New Zealand 2001–02 8 Heaton et al. 2006 winter (December–January) in Korea (Park et al. 2011). In the UK,
a Incidence rates expressed/100 000 aged <5 years USA, and Australia, KD is most common in spring (Harnden et al.
b Expressed/100 000 aged <15 years 2002; Royle et al. 1998; Chang et al. 2002a,b).
There has been an extensive, and so far fruitless, search for a sin-
gle causative organism; numerous organisms have been proposed
girls (187.0/100 000 aged <5 years). The age-specific incidence is but not proven (Burgner and Harnden 2005). Other proposed risk
greatest in children aged 6–11  months, and 88.4% of cases were factors include a history of shampooing household rugs, proxim-
aged less than 5  years and 67.7% less than 3  years. Nationwide ity to water (Davis et  al. 1995), mercury, and house dust mites.
surveys have also been conducted in Korea and have shown high The failure to identify a single organism suggests either that KD
occurrence rates: in 2006–08 the incidence was 113.1/100 000 chil- can occur as a final common pathway response to many different
dren aged less than 5 years. In Europe the rate is very much lower at organisms or that a novel infectious agent is involved.
5–15/100 000 children aged <5 years. Polyarteritis nodosa
Ethnic factors The original description of periarteritis nodosa by Kussmaul and
KD has been reported in all ethnic groups, but there are marked Maier (1866) was of a patient with inflammation and necrosis of
variations. Within the USA and UK populations the highest medium-sized arteries leading to aneurysm formation and organ
incidence is in individuals from southeast Asia. In Birmingham, infarction. The CHCC 2012 defined PAN as ‘Necrotizing arteritis
UK, the incidence in Asians (from the Indian subcontinent) was of medium or small arteries without glomerulonephritis or vascu-
14.6/100  000 aged <5  years, Black Afro-Caribbeans 5.9/100  000, litis in arterioles, capillaries, or venules; and not associated with
and Caucasians 4.6/100 000 (Gardner-Medwin et al. 2002). ANCA.’ (Jennette et  al. 2013). The dominant feature of PAN is
In the USA, incidence is highest amongst South East Asians organ infarction (gut, nerve) due to involvement of medium-sized
and Pacific Islanders, with the incidence being around 2.5 times arteries. The literature on the epidemiology of PAN and MPA
that seen in the white population. In Hawaii, the state with the has to be carefully interpreted, because many older studies used
highest proportion of South East Asians or Pacific Islanders, the the term polyarteritis nodosa as a generic term for any form of
incidence amongst Japanese Americans is 210.5/100  000, Native necrotizing vasculitis. The CHCC (1994) used the term classical
Hawaiians 86.9/100 000, and Chinese Americans 83.2/100 000. The PAN to distinguish it from MPA. The 2012 revision has dropped
CHAPTER 2 epidemiology of vasculitis 13

the term classical because MPA is now well established. PAN is Infection
typically ANCA negative whilst MPA is typically myeloperoxidase The major environmental factor associated with PAN is HBV infec-
(MPO) ANCA positive. tion; one of the highest rates for PAN yet reported comes from an
In Europe and the USA the estimated annual incidence of PAN area endemic for HBV infection. Many studies do not report HBV
ranges from 2.0 to 9.0/million (Table 2.5). Re-analysis of the data infection rates. In France, as HBV infection rates have fallen due to
from a UK study (Bath/Bristol) in the 1970s suggests that the pol- vaccination, the prevalence of PAN has also decreased (Mahr et al.
yarteritis group included patients who would now be considered 2004; Pagnoux et al. 2010).
to have MPA or EGPA (Scott et al. 1982; Scott, unpublished data).
Overall, the European and American studies of PAN do not suggest
a change over time (Table 2.5), nor does our Norwich study over
Small-vessel vasculitis—
22 years, but differences in classification make direct comparison ANCA-associated vasculitis
difficult. Use of the CHCC definitions makes PAN a much rarer dis- The classification of the small-vessel ANCA-associated vasculitides
ease; this has been observed in a number of studies (Table 2.5), the (AAV) poses a particular difficulty to the epidemiologist, as there
exception being one from Kuwait. A prevalence study from France is no uniform agreement on the application of the ACR (1990) cri-
suggests that improved public health control of hepatitis B virus teria and CHCC 1994 definitions. To address this issue, a consen-
(HBV) infection by vaccination is producing a fall in the prevalence sus approach to the use of the ACR (1990) and CHCC definitions
of PAN (Mahr et al. 2004; Pagnoux et al. 2010). has been developed and validated. This uses an algorithm to sort
patients into a single category with only a minimum of unclassifi-
Ethnic differences able patients (Watts et al. 2007; Liu et al. 2008).
The highest incidence of PAN recorded was 77/million in Alaskan
Indians (McMahon et al. 1989). The population was small (14 000) Granulomatosis with polyangiitis
and all the cases were positive for hepatitis B surface and e anti- (wegener’s granulomatosis)
gen at diagnosis. Detailed hepatitis serology is not available in Granulomatosis with polyangiitis (previously known as Wegener’s
most other studies. Whether these data reflect geographical and granulomatosis) was first described in the 1930s as a disease
ethnic differences or the high infection rate with HBV is unclear characterized by necrotizing granulomata of the upper and
as no other comparable study has been reported. A  study from lower respiratory tract, focal glomerulonephritis, and necrotiz-
Paris reported a prevalence of classical PAN of 30.7/million (Mahr ing systemic vasculitis (Godman and Churg 1954). The CHCC
et  al. 2004), similar to Sweden (31/million) (Mohammad et  al. 2012 defined GPA as ‘Necrotizing granulomatous inflamma-
2007), higher than in Germany (2–9/million) (Reinhold-Keller tion usually involving the upper and lower respiratory tract, and
et al. 2000), or Scandinavia (3.3/million using the ACR criteria) necrotizing vasculitis affecting predominantly small vessels (e.g.
(Haugeberg et al. 1998). Of the French PAN patients, 30% were capillaries, venules, arterioles, arteries, and veins). Necrotizing
HBV positive. glomerulonephritis is common.’ Jennette et al. 2013). The ACR

Table 2.5 Reported annual incidence rates for polyarteritis nodosa

Place Period Classification Incidencea Reference


Europe
Bristol/Bath, UK 1972–80 PAN 4.6 Scott et al. 1982
Norwich, UK 1988–97 ACR 8.0 Watts et al. 2000a
Kristiansand, Norway 1992–96 ACR 6.6 Haugeberg et al. 1998
Lund, Sweden 1997–2006 EMEA (ACR + CHCC) 0.9 Mohammad et al. 2009
Schleswig-Holstein, Germany 1998–2002 CHCC 0.4–2.0 Reinhold-Keller et al. 2005
Lugo, Spain 1988–97 ACR 6.6 González-Gay and García-Porrúa 1999
Vilnius, Lithuania 1990–99 ACR 7.7 Dadoniene et al. 2005
Lugo, Spain 1988–2001 CHCC 0.9 González-Gay et al. 2003b
Americas
Olmsted County, MN 1951–67 PAN 7.0 Kurland et al. 1969
Michigan, USA 1957–71 PAN 2.0 Sack et al. 1975
Alaska 1974–85 PANb 77.0 McMahon et al. 1989
Olmsted County, MN 1976–79 PAN 9.0 Kurland et al. 1984
Australia
Capital Territory, Australia 1995–99 ACR 2.3 Ormerod et al. 2008
2000–04 ACR 1.1 Ormerod et al. 2008
Arabia
Kuwait 1993–96 CHCC 16.0 El-Reshaid et al. 1997
a Incidence expressed /million population
b Hepatitis B positive
14 SECTION 1 introductory chapters

(1990) criteria for GPA have high sensitivity and specificity estimates from Norwich record a point prevalence of 146/million
(Leavitt et al. 1990). in 2008 (Watts et al. 2012) whilst in Lund the rate was 160/million
in 2003 (Mohammad et al. 2007).
Time trends
The incidence of GPA may have increased during the 1980s and Geographical factors
1990s. Andrews and colleagues (1990) in Leicester (UK) reported The annual incidence of GPA in Europe since 2000 is 5–12/million
an increase in the annual incidence of GPA from 0.7/million (Table 2.6). In Western USA and Australia the incidence is similar
(1980–1986) to 2.8/million (1987–1989) (Table  2.6). This was to that seen in Europe (Table 2.6). In Japan, GPA and PR3-ANCA
partially attributed to heightened diagnostic awareness following associated vasculitis are relatively rare compared with MPA and
the introduction of assays for ANCA in 1987. Long-term studies MPO-associated vasculitis (Table 2.6) (Fujimoto et al. 2011). In India,
conducted since the late 1990s suggest a relatively stable incidence. GPA is recognized but there are no epidemiological data (Malaviya
We did not observe any significant overall change in incidence dur- et al. 1990). In areas where TB is common, patients presenting with
ing the 22-year period of our Norwich study (Watts et al. 2012); pulmonary vasculitis are liable to be misdiagnosed with tuberculosis
however, we saw a periodicity to the occurrence of GPA with and ANCA serology is more difficult to interpret (Pradhan et al. 2004).
peaks every 7.7  years, which was not seen in MPA (Watts et  al.
2012) (Figure 2.2). The prevalence of GPA has gradually increased Ethnic factors
in Europe and the USA and has been attributed to better treat- The incidence rates are similar in most Caucasian populations
ment, leading to improved mortality (Table 2.7). The most recent (Table 2.6); the populations studied in the Western USA, Australia,

Table 2.6 Reported annual incidence of granulomatosis with polyangiitis (Wegener’s) and microscopic polyangiitis

Place Period Criteria Incidence GPAa Incidence MPAa Reference


Europe
Leicester, UK 1980–86 Fauci 0.7 0.5 Andrews et al. 1990
Leicester, UK 1987–89 Fauci 2.8 3.3 Andrews et al. 1990
Norwich, UK 1988–2010 EMEA 11.3 5.9 Watts et al. 2012
UKGPRD, UK 1990–2005 ACR 8.4 NA Watts et al. 2009b
Kristiansand, Norway 1992–96 ACR 6.6 NA Haugeberg et al. 1998
Tromso, Norway 1984–98 ACR 8.0 2.7 Koldingsnes and Nossent 2000
Lund, Sweden 1971–93 ACR 2.1 NA Westman et al. 1998
Sweden 1975–2001 ICD 0.78 nA Knight et al. 2006
South Sweden 1997–2006 EMEA 9.8 10.1 Mohammad et al. 2009
Finland 1980–85 ACR 1.9 NA Takala et al. 2008
Finland 1996–2000 ACR 9.3 NA Takala et al. 2008
Heidelberg 1984–9 MPA b NR 1.5 Andrassy et al.. 1991
Schleswig-Holstein, Germany 1998–2002 CHCC 6–12 3 Reinhold-Keller et al. 2005
Vilnius, Lithuania 1990–99 ACR 2.1 3 Dadoniene et al. 2005
Crete, Greece 1995–2003 ACR 6.6 10.2 Panagiotakis et al. 2009
Lugo, Spain 1988–97 ACR 4.8 NA González-Gay, et al. 1999
Lugo, Spain 1988–2001 CHCC 2.95 7.9 González-Gay et al. 2003b
Reggio Emilia, Italy 2004–09 ACR 3.5 NA Macchioni et al. 2011
Greenland (Inuit) 1992–2011 ACR 1 NA Faurschou et al. 2013
Faroes 1992–2011 ACR 6.4 NA Faurschou et al. 2013
Americas
Western Montana, USA 1993–2004 ACR 8.6 2.9 Zeft et al. 2005
Lima, Peru 1990–2004 CHCC 0.5 NA Sanchez et al. 2006
Australasia
South Australia 1985–2004 ACR, ICD 11.2 NA Hissaria et al. 2008
Capital Territory, Australia 1995–99 ACR 8.8 2.3 Ormerod et al. 2008
Capital Territory, Australia 2000–04 ACR 8.4 5.0 Ormerod et al. 2008
New Zealand (North Island) 1999–2003 ICD 10 M313 2.9 NA O’Donnell et al. 2007
New Zealand (South Island) 1999–2003 ICD 10 M313 7.5 NA O’Donnell et al. 2007
Japan 2005–09 EMEA 2.1 18.2 Fujimoto et al. 2011
Arabia
Kuwait 1993–96 CHCC NR 24.0 El-Reshaid et al. 1997

Fauci, Fauci et al. 1983; EMEA, European Medicines Agency Algorithm; ACR, American College of Rheumatology criteria (1990); CHCC, Chapel Hill Consensus Conference definition (1994);
ICD, International Classification of Diseases; UKGPRD, United Kingdom General Practice Research Database.
a Incidence expressed /million population
b Renal involvement only
CHAPTER 2 epidemiology of vasculitis 15

20 from China suggest that MPA is more common than GPA (Liu et al.
MPA
18 GPA 2008). In a multiethnic series from Chapel Hill in the USA, GPA was
SRV relatively uncommon in African Americans (Cao et al. 2011).
16
14 Genetic factors
12
Familial heritability is similar to that seen in rheumatoid arthritis
million population

and it has been estimated as relative risk of 1.56 (Knight et al. 2008).
10 Clusters of GPA occurring in families usually involve no more than
8 two affected members and that is most commonly in 1st degree rel-
6 atives. It is noteworthy that distant relatives are less affected, which
is suggestive of a more dominant role of environmental factors.
4 HLA associations have been reported and the strongest for the
2 AAV is with HLA DPB1*0401 (Jagiello et  al. 2004). The recently
0 completed GWAS in AAV has provided further evidence that GPA
and MPA should be viewed as separate conditions (Lyons et  al.
8 0
9 1
9 2
9 3
9 4
9 5
9 6
9 7
9 8
9 9
9 0
0 1
0 2
0 3
0 4
0 5
0 6
0 7
0 8
08 9
0
2012). The patients studied were from the UK and Northern Europe
19 8–9
19 9–9
19 0–9
19 1–9
19 2–9
19 3–9
19 4–9
19 5–9
19 6–9
19 7–9
19 8–0
20 9–0
20 0–0
20 1–0
20 2–0
20 3–0
20 4–0
20 5–0
20 6–0
20 7–0
–1
8

where the link between disease (GPA or MPA) and ANCA speci-
19

Fig. 2.2 Incidence of granulomatosis with polyangiitis (GPA), microscopic ficity is closest. PR3-ANCA disease was associated with HLA-DP,
polyangiitis (MPA), and systemic rheumatoid vasculitis (SRV) plotted as SERPINA1, and PRTN3, while MPO-ANCA disease was associated
overlapping triennial periods. with HLA-DQ. SERPINA1 encodes alpha-1 antitrypsin, a serine
protease which has PR3 as one of its substrates. PRTN3 encodes
and New Zealand are predominately white Caucasian populations proteinase 3.  Thus the immune response against the autoantigen
of North European descent. Recent studies in non-white Caucasian PR3 is a central aetiological feature of PR3-ANCA-associated vas-
populations suggest a lower incidence (Table  2.6). The French culitis. Several previous studies have suggested that the Z allele of
prevalence study suggested that GPA is less common in people SERPINA1 is associated with GPA. The GWAS found association
of non-European ancestry than MPA (Mahr et al. 2004). In New with a SNP in linkage disequilibrium with the Z allele. Haplotype
Zealand, GPA was twice as common in Europeans than Maoris or analysis suggest that the causal variant at SERPINA1/SERPINA11 is
Asians (O’Donnell et al. 2007). In Japan, GPA is also much less com- either the z allele of SERPINA1 or in close linkage disequilibrium
mon than MPA, and cANCA-associated disease is less frequent than with it. The strongest SNP associations were with ANCA specific-
pANCA-associated disease (Fujimoto et al. 2011). Large case series ity and not with the clinical syndromes. The study did not confirm

Table 2.7 Reported prevalence rates for granulomatosis with polyangiitis (Wegener’s) and microscopic polyangiitis

Place Period Classification Prevalence GPAa Prevalence MPAa Reference


Europe
Norwich, UK 31/12/2008 EMEA 146 63 Watts et al. 2012
UKGPRD, UK 1990 ACR 28.8 NA Watts et al. 2009b
UKGPRD, UK 2005 ACR 64.8 NA Watts et al. 2009b
Paris, France 2000 ACR 23.7 25.1 Mahr et al. 2004
Germany (North) 1994 CHCC 58 9 Reinhold-Keller et al. 2000
Germany (South) 1994 CHCC 42 0 Reinhold-Keller et al. 2000
Denmark 1977–2001 ICD 100 NA Eaton et al. 2007
Southern Sweden 1/1/2003 ACR/CHCC 160 94 Mohammad et al. 2007
Norway 1992–96 ACR 53.0 NA Koldingsnes and Nossent 2000
Australasia
Canterbury, New Zealand 1999–2003 ACR 131 58 Gibson et al. 2006
31/12/2003 ACR 93.5 37 Gibson et al. 2006
Capital Territory, Australia 1995–99 ACR CHCC 64.3 17.5 Ormerod et al. 2008
2000–04 ACR CHCC 95.0 39.1 Ormerod et al. 2008
Americas
New York, USA 1986–90 ACR 32.0 NA Cotch et al. 1996
USA 1986–90 ACR 26.0 NA Cotch et al. 1996
Western Montana, USA 2004 ACR 90 NA Zeft et al. 2005
a Prevalence rates expressed as/million population
EMEA, European Medicines Agency Algorithm; ACR, American College of Rheumatology criteria (1990); CHCC, Chapel Hill Consensus Conference definition; ICD, International
Classification of Diseases; UKGPRD, United Kingdom General Practice Research Database.
16 SECTION 1 introductory chapters

previously reported polymorphisms in CTL4A and PTPN22 genes. hydrocarbon exposure in male MPA and GPA patients compared
These genetic differences may explain the geographical variation in to matched blood donors and non-significantly greater exposures
occurrence of GPA as there is a well-documented global variation in in female patients with pulmonary haemorrhage (Pai et al.1998).
the occurrence of the z allele of SERPINA1 (Watts and Scott 2012). Heavy metals (mercury, lead) have been linked to GPA (Albert
et al. 2004).
Environmental factors Farming in the year prior to the onset of vasculitis was associ-
Seasonality ated with GPA with an OR of 2.7. It was not possible to distinguish
Seasonality in onset has been suggested by reports of a higher between exposures to crops, livestock, or animal species, as most
incidence rate of onset in winter (29.8%) than in summer (14.3%) individuals were exposed to more than one type, but the associa-
(Raynauld et al. 1993). This has been variable and not consistent tion appeared stronger in livestock than crops (Lane et al. 2003).
with studies in the United States (Cotch et al. 1996). Several studies Another study failed to find a significant association between GPA
have not reported seasonality (Cotch et al. 1996; Duna et al. 1998; and farming (Duna et al. 1998).
Tidman et al. 1998; Lane et al. 2003; Aries et al. 2008). Difficulty in In a case–control study of inpatient admission of 2288 cases of
establishing an accurate date of onset of the disease process, meth- GPA in Sweden, no association with any specific occupation was
odological discrepancies, and variation of triggering factors in dif- found (Knight et al. 2010).
ferent geographic areas may explain this inconsistency.
Infection Microscopic polyangiitis
The possible role of infection, both in triggering de novo disease and In 1948, Davson described patients with segmental necrotizing
relapse, has been extensively investigated for many years. No clear glomerulonephritis who also had features of PAN with extrarenal
associations have been demonstrated except for Staphylococcus aureus small and medium artery involvement (Davson et al. 1948). The
infection. Staphylococcus aureus is the organism most commonly iso- term microscopic polyarteritis was used to describe these patients
lated in cultures from the upper airways of GPA patients and nasal car- in whom the dominant feature was rapidly progressive renal failure,
riage of S. aureus has been associated with increased risk of relapse in and subsequently this was changed by the CHCC to microscopic
GPA (Stegeman et al. 1996). Our recent report of a cyclical pattern of polyangiitis to emphasize the differentiation from PAN. The CHCC
occurrence is supportive of an infectious aetiology (Watts et al. 2012). defined MPA as ‘Necrotizing vasculitis, with few or no immune
deposits, predominantly affecting small vessels (i.e. capillaries,
Ultraviolet radiation
venules, or arterioles). Necrotizing arteritis involving small and
The latitudinal variation in the occurrence of GPA in both the
medium arteries may be present. Necrotizing glomerulonephritis is
Northern and Southern hemispheres has led to the suggestion that
very common. Pulmonary capillaritis often occurs. Granulomatous
variation in exposure to ambient UV radiation might be causative
inflammation is absent.’ (Jennette et al. 2013). The literature on the
acting through vitamin D metabolism (Staples et al. 2003). An eco-
epidemiology of PAN and MPA has to be carefully interpreted,
logical study suggested that there is a protective effect of ambient
because many older studies used the term polyarteritis nodosa as a
UV radiation on the onset of GPA (Gatenby et al. 2009).
generic term for any form of necrotizing vasculitis.
Occupational exposure
Time trends
The occurrence of ENT and pulmonary disease in GPA has led to the
notion that dust exposure might be important in the pathogenesis. In Leicester (UK) the annual incidence of MPA between 1980
Some counties in New York state (USA) have a higher prevalence than and 1986 was 0.5/million and between 1987 and 1989 it was 3.3/
others but this does not appear to be linked to rural/ urban differ- million (Andrews et al. 1990) (Table 2.6). This increase followed
ences (Cotch et al. 1996). Similarly, there was no urban/ rural differ- introduction of testing for ANCA and was ascribed to possibly
ence in Germany (Reinhold-Keller et al. 2000). Duna and colleagues increased physician awareness. A recent study from Norwich UK
(1998) studied self-reported exposure to heat, fumes, and particu- reported an incidence of 5.9/million (Watts et al. 2012). A renal
lates. There was a higher incidence of exposure in GPA patients com- biopsy study in Stockholm reported a doubling of the annual inci-
pared with normal control subjects, but no difference between GPA dence of pauci-immune necrotizing and crescentic glomerulone-
patients and patients with other types of lung disease. Paradoxically, phritis from 6.0 to 12.0/million between 1986 and 1992. Most of
smoking appears to offer some protection against development of these patients would have had MPA (Pettersson et al. 1995). More
ANCA-associated small-vessel vasculitis (Haubitz et al. 2005). recent studies suggest that the incidence was greater during the
Systemic vasculitis (mainly MPA; see Section Microscopic 1990s than the 1980s (González-Gay and Garcia-Porrua 1999). In
Polyangiitis  – Environmental Factors) has been associated with our prospective Norwich study, there was no significant change
exposure to particulate silica, for example quartz, granite, sand- in incidence during the period 1988–2010 (Watts et  al. 2012).
stone, and grain dust. Metal fumes have been associated with Tidman (1998) also did not observe an increase in incidence dur-
pulmonary vasculitis in Russian brass bronze smelters, and Nuyts ing 1986–1995. During this period they noted a periodic fluctua-
reported significantly raised odds ratios (ORs) for exposure to tion with peaks every 3–4 years. We did not observe peaks in our
various metals and welding fumes (OR 2.0, 95% CI 1.1–4.6) in a study. It is possible that MPA is becoming more common but the
group of renal patients with GPA and glomerulonephritis (Nuyts number of series is small and the reported increases could repre-
et al. 1995). Our case–control study failed to find an association sent increasing physician awareness and changing ideas of diagno-
with occupational metal exposure and GPA (Lane et  al. 2003). sis and classification.
There has been conflicting evidence regarding a link between Geographical factors
occupational exposure to hydrocarbons, for example paints and In Europe there appears to be a North–South gradient in inci-
glues, and systemic vasculitis. Pai reported significantly higher dence, with MPA being more common in Southern Europe (Lugo,
CHAPTER 2 epidemiology of vasculitis 17

Spain latitude 43°N) compared with Tromsø in Northern Norway compared to healthy individuals but not disease controls (systemic
(latitude 70°N) (Watts et al. 2001). Care was taken in this study lupus erythematosus and systemic sclerosis). A causal association
to eliminate variations in classification. The period prevalence in was suggested between the high levels of silica dust experienced
New Zealand in 2000–2003, which has a similar latitude south after the 1995 great earthquake in Kobe, Japan and the subsequent
(40–44°S) to Paris in the north (49°N), is 58/million and appears increase in MPO-ANCA-associated vasculitis (Yashiro et al. 2000).
to be similar to that observed in Paris (Gibson et al. 2006; Mahr Several case–control studies have found significant associations
et al. 2004). between systemic vasculitis and silica. Hogan et al. (2001) found an
Two geographical regions with an apparently high incidence odds ratio of 4.6 (95% CI 1.8–12.1) for reported silica exposure in
of MPA and PAN are Japan and Kuwait. In the Kuwaiti national ANCA-positive patients (36 MPA, 21 GPA, eight necrotizing glo-
population, the incidence of MPA was 24/million (El-Reshaid et al. merulonephritis) compared to renal controls and Gregorini et al.
1997). In Japan, the incidence of MPA was 18.2/million, higher (1993) reported that ANCA-positive patients with rapidly progres-
than any European figure (Fujimoto et al. 2011). sive glomerulonephritis were 14 times more likely to have been
exposed to silica dust than controls. In a case–control study from
Ethnic differences
Norfolk (UK), we reported ORs of 3.0 (95% CI 1.0–8.4) with silica
In the Kuwait study the incidence figures were only calculated exposure in 75 cases of AAV (47 GPA, 12 MPA, 16 EGPA), but there
for Kuwaitis but MPA and PAN also occurred in other non-white was no significant association between MPA alone and occupational
groups. In the Miyazaki prefecture, Japan, the incidence of MPA silica exposure (Lane et al. 2003). Stratta et al. reported an odds ratio
is higher than in European Caucasian populations (Fujimoto et al. of 2.4 (P = 0.04) for silica exposure in 31 cases of biopsy-proven
2006), and MPO-ANCA associated disease is seen more frequently vasculitis compared to controls but no other significant risk factors
than PR3-ANCA associated disease. Whether this reflects ascer- including hydrocarbons, metals, and asbestos (Stratta et al. 2001).
tainment bias or a genuine difference in response to the (as yet
unknown) triggering factor(s) remains to be determined.
Eosinophilic granulomatosis with
Most of the populations studied in Europe have been predomi-
nantly white Caucasians. In Paris, France, a study from a multiethnic
polyangiitis (Churg–Strauss)
urban area reported that the prevalence of AAV was twice as high in In 1951, Churg and Strauss described the post-mortem features
individuals of European descent (104.7/million) as in non-Europeans of 13 patients who died following an illness characterized by
(52.5/million) (Mahr et al. 2004). GPA was relatively less frequent asthma, eosinophilia, fever, and granulomatous necrotizing vas-
than MPA in non-Europeans, and none of the GPA patients came culitis (Churg and Strauss 1951). The CHCC 2012 defined EGPA
from Africa. The non-European population was derived from the as ‘Eosinophil-rich and necrotizing granulomatous inflammation
Maghreb, sub-Saharan Africa, Asia, and the Caribbean, and com- often involving the respiratory tract, and necrotizing vasculitis pre-
prised 28% of the study population (1.09/million). dominantly affecting small to medium vessels, and associated with
asthma and eosinophilia. ANCA is more frequent when glomeru-
Environmental factors lonephritis is present.’ (Jennette et al. 2013). The annual incidence
MPA has been associated with a number of environmental factors of EGPA appears to be broadly similar in all populations studied
including silica, hydrocarbons, drugs, and infections. (Table 2.8), and it is much less common than GPA or MPA. The
Drugs data are too limited to draw conclusions about changes in inci-
Drug exposure has been predominately associated with dence with time. Prevalence data (Table 2.9) confirm that EGPA
MPO-ANCA-positive vasculitis. The most commonly reported are is much rarer than GPA or MPA in New Zealand (22.3/million),
propylthiouracil and hydralazine although penicillamine, minocy- Sweden 14.0/million), and France (10.7/million) (Gibson et  al.
cline, methimazole, carbimazole, thiamazole, sulfasalazine, pheny- 2006; Mohammad et al. 2007; Mahr et al. 2004).
toin, and allopurinol have also been implicated (Holder et al. 2002). Environmental factors
Hyperthyroid patients treated with propylthiouracil develop a vas- The majority of cases of EGPA are idiopathic; inhaled antigens,
culitis associated with MPO-ANCA (Choi et  al. 2000). Choi et  al. vaccination, and desensitization have been reported as trigger-
reviewed 250 MPO-positive systemic vasculitis patients and detected ing factors (Guillevin et al. 1999). Drugs, including sulfonamides,
hydralazine, propylthiouracil, allopurinol, penicillamine, or sulfasala- penicillin, anticonvulsants, and thiazides, have been associated
zine use in 18 cases within 9 months prior to the onset of disease (Choi with the syndrome. Development of EGPA has been associated
et al. 2000). Positivity for MPO-ANCA, IgM anticardiolipin antibod- with the use of leukotriene inhibitors and more recently with the
ies, and antihistone antibodies is characteristic of drug-induced vas- anti-IgE monoclonal antibody omalizumab (Wechsler et al. 2009).
culitis syndromes (Bonaci-Nikolic et  al. 2005). Skin vasculitis and In a recent study looking at the US Food and Drug Administration
arthralgias are more common than in idiopathic vasculitis, with renal Adverse Events Reporting System database, leukotriene inhibi-
lesions being less common. The condition is milder than idiopathic tor therapy was a suspect medication in most confirmed cases
vasculitis and may remit on withdrawal of the precipitating drug. of EGPA reported (Bibby et  al. 2010). The incidence of EGPA
Occupation among patients with asthma has been estimated to be 34.6/million
Case reports have described systemic vasculitis in association with person-years (Harrold et al. 2005); the occurrence of EGPA fol-
pulmonary silicosis and in individuals exposed to high levels of sil- lowing leukotriene inhibitors and omalizumab may be the result
ica, for example miners and quarrymen (Cohen Tervaert et al. 1998). of unmasking previous undiagnosed disease with reduction in the
Clinical cases of MPA, limited GPA, and MPO-ANCA-positive dis- glucocorticoid use. The epidemiological data may be confounded
ease have been reported. A study of individuals exposed to silica by the severity of the preceding asthma (Harrold et  al. 2007;
found a significantly higher number positive for MPO-ANCA Hauser et al. 2008).
18 SECTION 1 introductory chapters

Table 2.8 Reported incidence rates for eosinophilic granulomatosis with angiitis (Churg–Strauss)

Place Period Criteria Incidencea Reference


Europe
Norwich, UK 2005–09 EMEA 0.9 Fujimoto et al. 2011
Lugo, Spain 1988–97 ACR 1.1 González-Gay et al. 1999
Lugo, Spain 1988–2001 CHCC & ACR 1.3 González-Gay et al. 2003b
Crete, Greece 1995–2003 CHCC 0 Panagiotakis et al. 2009
Burgundy, France 1998–2008 ACR 1.2 Vinit et al. 2009
Schleswig-Holstein, Germany 1998–2002 CHCC 1 Reinhold-Keller et al. 2005
Vilnius, Lithuania 1990–99 ACR 1.3 Dadoniene et al. 2005
TromsØ, Norway 1988–98 ACR 0.5 Watts et al. 2001
Southern Sweden 1997–2006 EMEA 0.9 Mohammad et al. 2009
Americas
Olmsted, Minnesota 1976–79 Clinical 4.0 Kurland et al. 1984
Lima, Peru 1990–2004 CHCC 0.14 Sanchez et al. 2006
Australia
Capital Territory, Australia 1995–2004 ACR 2.3 Ormerod et al. 2008
a Incidence figures expressed /million population
EMEA, European Medicines Agency Algorithm; ACR, American College of Rheumatology criteria (1990); CHCC, Chapel Hill Consensus Conference definition; ICD, International
Classification of Diseases.

Small-vessel vasculitis—immune complex for cutaneous vasculitis; they did not consider hypersensitivity
vasculitis), because they included the presence of IgA deposits
IgA vasculitis (henoch–schönlein purpura) in the definition (Jennette et  al. 1994). The CHCC 2012 defined
Schönlein (1837) first described a childhood illness characterized HSP as ‘Vasculitis, with IgA1-dominant immune deposits, affect-
by acute purpura and arthritis. Henoch (1874) described the addi- ing small vessels (predominantly capillaries, venules, or arteri-
tional features of colicky abdominal pain and nephritis. Vasculitis oles). Often involves skin and gut, and frequently causes arthritis.
due to allergic or hypersensitivity mechanisms has been considered Glomerulonephritis indistinguishable from IgA nephropathy may
a distinct entity since the 1940s and was included by Zeek in her occur’ (Jennette et al. 2013). To reflect this the CHCC 2012 recom-
original classification (Zeek 1952). The ACR (1990) considered mended adoption of the name IgA vasculitis (IgAV).
Henoch–Schönlein purpura (HSP) and leukocytoclastic vascu- IgAV occurs predominately in children. The annual incidence
litis (LCV) (hypersensitivity vasculitis) to be separate conditions. of IgAV in children is 10–20/100  000 aged less than 17  years
The criteria for LCV were the least sensitive (71.0%) and specific (Table  2.10). The peak age of onset in most studies is 5–6 years
(78.5%) (Fries et al. 1990). There is considerable overlap between (Yang et al. 2005) with a peak onset in autumn and winter.
the ACR and CHCC (1994) criteria (Watts et al. 1998). The CHCC The incidence in adults is approximately tenfold less than in chil-
definition (1994) for HSP makes a much better distinction between dren (Table 2.10) at around 3–10/million.
HSP and cutaneous leukocytoclastic angiitis (their preferred term Ethnic factors
In Birmingham (UK), IgAV was more common in Asians from
Table 2.9 Reported prevalence rates for eosinophilic granulomatosis
the Indian subcontinent (24.0/100 000 aged <17 years) compared
with angiitis (Churg–Strauss)
with white Caucasians (17.8/100 000 aged less than <17 years), and
blacks (predominantly Afro-Caribbean) (6.2/100 000 aged less than
Place Period Criteria Prevalencea Reference
<17 years). The Chinese population in Taiwan has an annual inci-
Norwich, UK 31/12/2008 EMEA 45.7 Watts et al. 2009c dence of 12.9/100 000 children aged <17 years (Yang et al. 2005).
Southern, Sweden 01/01/2003 EMEA 14.0 Mohammad et al. Environmental factors
2007
Drug use (antibiotics, especially beta-lactams, analgesics, or
Capital Territory, 2000–04 ACR 22.3 Ormerod non-steroidal anti-inflammatory drugs (NSAIDs)), or upper res-
Australia et al. 2008 piratory tract infections shortly before development of vasculitis,
Paris, France 2000 ACR 10.7 Mahr et al. 2004 was more common in patients with LCV than in those with IgAV
(García-Porrúa and González-Gay, 1999). All patients in this study
Burgundy, France 1998–08 ACR 11.3 Vinit et al. 2009
who had drug-induced vasculitis met ACR (1990) criteria for LCV
Germany (North) 1994 CHCC 7 Reinhold-Keller or IgAV, and none fulfilled criteria for PAN or GPA.
et al. 2000
Germany (South) 1994 CHCC 2 Reinhold-Keller
et al. 2000
Vasculitis in childhood
The spectrum of vasculitis seen in children is very different from
a Prevalence rates expressed /million population
that seen in adults. In children the predominant diseases are
ACR, American College of Rheumatology (1990) criteria; EMEA, European Medicines
Agency Algorithm.
CHAPTER 2 epidemiology of vasculitis 19

Table 2.10 Reported annual incidence rates for IgA vasculitis (Henoch–Schönlein purpura)

Place Period Criteria Incidencea Reference


Children
Saudi Arabia 1981–87 Not known 6.7b Abdel-Al et al. 1990
Czech Republic 1997–99 ACR 10.2 Dolezalova et al. 2004
Taiwan 1999–2002 ACR 12.9 Yang et al. 2005
Lugo, Spain 1988–97 ACR 10.5c González-Gay and García-Porrúa, 1999
Birmingham, UK 1996–99 ACR 20.4 Gardner-Medwin et al. 2002
Belfast 1980s Clinical 13.5 Stewart et al. 1988
Scotland 1995–2007 Not known 20.3–26.7 Penny et al. 2010
The Netherlands 2004 ACR 6.1 Aalberse et al. 2007
Japan 1987–97 Nephritis 3.5 Kawasaki et al. 2010
Japan 1998–2007 Nephritis 3.6 Kawasaki et al. 2010
Adults
Schleswig-Holstein, Germany 1998–2002 CHCC 3–10 d Reinhold-Keller et al. 2005
Norfolk, UK 1990–94 ACR 13.0 Watts et al. 1998
Norfolk, UK 1990–94 CHCC 3.4 Watts et al. 1998
Vilnius, Lithuania 1990–99 ACR 3.0 Dadoniene et al. 2005
Lugo, Spain 1988–2001 Biopsy +ve 14.0 García-Porrúa and González-Gay, 1999
a for children/100 000 aged <17 years and for adults/million population
b aged less than 12 years
c aged <14 years
d includes adults and children

ACR (1990) criteria.

Kawasaki’s disease and IgAV (Figure 2.1) (see Sections Kawasaki’s multiplex (Bywaters and Scott 1963). Since then a wider spectrum of
disease and IgA vasculitis (Henoch–Schönlein purpura)). disease has been recognized to include carditis, scleritis, nodules, and
Validated classification criteria have been developed for childhood systemic disease (Scott et al. 1981). All sizes of vessel may be involved,
(Ozen et al. 2010). The large-vessel vasculitides are rare in both from aorta to capillaries. Small-vessel vasculitis can occur in isolation
adults and children. Takayasu’s arteritis occurs rarely in children as small nail edge or nail fold lesions which are considered benign
and GCA does not occur. The medium/ small-size vasculitides but which may herald or coexist with major arterial disease (Watts
associated with ANCA are all very rare in children, with incidence et al. 1995). Systemic rheumatoid vasculitis usually occurs in patients
figures of probably <1/million aged <15 years (Gardner-Medwin with long-standing, rheumatoid factor-positive, erosive RA. Males
et  al. 2003). A  recent series of GPA presenting in childhood with RA are at greater risk than females.
observed that the mean age of onset was 14.5 years; unlike adult
GPA, there was a 1:4 male to female ratio (Akikusa et al. 2005). In Time trends
Southern Alberta, the incidence of paediatric GPA increased from Systemic rheumatoid vasculitis first became widely recognized and
2.75/million to 6.39/million aged <15 years over a 15-year period reported during the 1960s, a period when glucocorticoids were
(Grisaru et al. 2010). widely used for the management of RA. This has led to the notion
that vasculitis could be caused by the inappropriate use of (by cur-
Vasculitis associated with rent standards) high doses of glucocorticoids, although this is con-
jectural. It has also been suggested that RA is becoming less severe
specific conditions and that therefore the frequency of rheumatoid vasculitis should
Vasculitis occurring in the presence of a connective tissue disease also be declining.
or infection is usually considered to be secondary (Table  2.1). In Spain, the annual incidence of biopsy-proven rheumatoid
This most typically occurs in patients with rheumatoid arthri- vasculitis during 1988–1997 was 6.4/million (González-Gay and
tis (RA), systemic lupus erythematosus (SLE), or Sjögren’s syn- García-Porrúa 1999). In our population in Norfolk, UK from
drome. There are no recognized criteria for the definition of 1988 to 1992 the incidence was 11.6/million and this fell to 3.9/
vasculitis occurring in association with SLE or Sjögren’s syn- million during 2001–2010 (Figure  2.2) (Ntatsaki et  al. 2013).
drome and hence no data on the epidemiology of these condi- A similar pattern was seen for males and females. There was no
tions. Apart from vasculitis associated with RA there are few difference in age or disease duration at onset during the study.
epidemiological data. This decline could be due to better disease control (increased use
of immunosuppressive agents such as methotrexate) or changes
Systemic rheumatoid vasculitis in smoking habits.
Vasculitis as a complication of RA was first described in 1898 in a A recent population-based US study looking at the incidence of
patient with histological evidence of vascular inflammation of the extra-articular RA reported a reduction in the 10-year cumulative
vasa nervorum. The early clinical descriptions in the 1940s and 1950s incidence of SRV from 3.6% in 1985-1994 to 0.6% in 1995–2007
were of the classical features of peripheral gangrene and mononeuritis (Myasoedova et al. 2011). The reduction of the prevalence of SRV
20 SECTION 1 introductory chapters

has also been reported in a serial cross sectional study looking at Bas-Lando, M., Breuer, G.S., Berkun, Y., Mates, M., Sonenblick, M., and
both hospitalized and ambulatory patients from the US veteran Nesher, G. (2007). The incidence of giant cell arteritis in Jerusalem
population during 1985–2006, comprising a similar duration of over a 25-year period:  annual and seasonal fluctuations. Clinical and
Experimental Rheumatology, 25 (1 Suppl. 44), S15–17.
cohort observation with our study of 22 years (Bartels et al. 2009).
Bengtsson, B-Å. and Malmvall, B.E. (1981). The epidemiology of giant cell
There has also been a decline in hospitalization in California of SRV arteritis including temporal arteritis and polymyalgia rheumatica: inci-
patients between 1980 and 2001 (Ward 2004). dences of different clinical presentations and eye complications. Arthritis
and Rheumatism, 24, 899–904.
Bibby, S., Healy, B., Steele, R., et al. (2010). Association between leukotriene
Other vasculitides receptor antagonist therapy and Churg-Strauss syndrome: an analysis of
The epidemiology of rarer conditions such as Cogan’s syndrome, the FDA AERS data- base. Thorax, 65, 132–8.
cryoglobulinaemia and anti-GBM disease has not been well studied. Boesen, P. and Sorensen, S.F. (1987). Giant cell arteritis, temporal arteritis and
polymyalgia rheumatica in a Danish county. Arthritis and Rheumatism,
30, 294–9.
Conclusions Bonaci-Nikolic, B., Nikolic, M.M., Andrejivic, S., et al. (2005). Antineutrophil
The vasculitides continue to present a challenge to the epidemiolo- cytoplasmic antibody asoociated autoimmune diseases induced by thy-
roid drugs:  comparison with idiopathic ANCA vasculitides. Arthritis
gist. There is increasing interest in these rare diseases and improve-
Research and Therapy, 7, R1072–81.
ments in classification mean that the picture is gradually unfolding. Burgner, D. and Harnden, A. (2005). Kawasaki disease: what is the epidemi-
There is clearly age tropism, suggesting that the causes of vasculitis ology telling us about the aetiology? International Journal of Infectious
in childhood are likely to be very different from those triggering Disease, 9, 185–94.
vasculitis in the elderly. Ethnic differences are beginning to emerge; Burns, J.C., Cayan, D.R., Tong, G., et al. (2005). Seasonality and temporal
this has been most clearly described in KD, but there are clues that clustering of Kawasaki syndrome. Epidemiology, 16, 220–5.
GCA, GPA, and MPA may occur with different frequencies in dif- Bywaters, E.G.L. and Scott, J.T. (1963). The natural history of vascular lesions
ferent ethnic groups. The genetic basis for this variation is being in rheumatoid arthritis. Journal of Chronic Disease, 16, 905–14.
unravelled. Slow progress is being made in understanding the Cao, Y., Schmitz, J.L., Hogan, S.L., Bunch, D., Hu, Y., Jennette, C.E., et al.
causative factors, with environmental agents such as silica or drugs (2011). DRB1*15 allele is a risk factor for PR3-ANCA disease in
African Americans. Journal of the American Society for Nephrology, 22,
being important in GPA and MPA. 1161–7.
Chang, R-K. (2002a). Hospitalizations for Kawasaki disease among children
References in the United States, 1988–97. Pediatrics, 109, e87.
Aalberse, J., Dolman, K., Ramnath, G., Pereira, R.R., and David, J.C. (2007). Chang, R-K. (2002b). Epidemiologic characteristics of children hospital-
Henoch Schönlein purpura in children: an epidemiological study among ized for Kawasaki disease in California. Pediatric Infectious Diseases,
Dutch paediatricians on incidence and diagnostic criteria. Annals of the 21,1150–5.
Rheumatic Diseases, 66, 1648–50. Choi, H.K., Merkel, P.A., Walker, A.M., and Niles, J.L. (2000). Drug induced
Abdel-Al, Y.K., Hejaz, Z., and Majeed, H.A. (1990). Henoch Schönlein antineutrophil cytoplasmic antibody positive vasculitis -prevalence
purpura in Arab children. Analysis of 52 cases. Tropical Geography among patients with high titres of myeloperoxidase antibodies. Arthritis
Medicine, 42, 52–7. and Rheumatism, 43, 405–13.
Abdul-Rahman, A.M., Molteno, A.C., and Bevin, T.H. (2011). The epidemi- Churg, J. and Strauss, L. (1951). Allergic granulomatosis, allergic angiitis and
ology of giant cell arteritis in Otago, New Zealand: a 9-year analysis. New periarteritis nodosa. American Journal of Pathology, 27, 277–301.
Zealand Medical Journal, 124, 44–52. Cohen Tervaert, J. W., Stegeman, C.A., and Kallenberg, C.G.M. (1998). Silicon
Akikusa, J., Silverman, E.D., Laxer, R.M., and Schneider, R. (2005). Clinical exposure and vasculitis. Current Opinion in Rheumatology, 10, 12–7.
features and outcome of paediatric Wegener’s granulomatosis. Arthritis Cotch, M.F., Hoffman, G.S., Yerg, D.E., et al. (1996). The epidemiology of
and Rheumatism, 52, S534. Wegener’s granulomatosis: estimates of the five year period prevalence,
Albert, D., Clarkin, C., Komoroski, J., et al. (2004). Wegener’s granulomato- annual mortality and geographic disease distribution from population
sis: possible role of environmental agents in its pathogenesis. Arthritis based data sources. Arthritis and Rheumatism, 39, 87–92.
and Rheumatism, 51, 656–64. Dadoniene, J., Kirdaite, G., Mackiewicz, Z., Rimkevicus, A., and Haugeber,
Andrassy, K., Küster, S., Waldherr, R., et al. (1991). Rapidly progressive glomeru- G. (2005). Incidence of primary systemic vasculitis in Vilnius: a univer-
lonephritis: analysis of prevalence and clinical course. Nephron, 59, 206–12. sity hospital population based study. Annals of the Rheumatic Diseases,
Andrews, M., Edmunds, M., Campbell, A., et al. (1990). Systemic vasculitis 64, 335–6.
in the 1980’s—is there an increasing incidence of Wegener’s granuloma- Davis, R. L., Waller, P. L., Mueller, B. A., et al. (1995). Kawasaki syndrome
tosis and microscopic polyarteritis? Journal Royal College of Physicians, in Washington State. Race specific incidence rates and residential
24, 284–8. proximity to water. Archives of Pediatric and Adolescent Medicine,
Areis, P.M., Herlyn, K., and Reinhold-Keller, E. (2008). No seasonal variation 149, 66–9.
in the onset of symptoms of 445 patients with Wegener’s granulomatosis. Davson, J., Ball, J., and Platt, R. (1948). The kidney in periarteritis nodosa.
Arthritis and Rheumatism, 59, 904. Quarterly Journal of Medicine, 17, 175–202.
Baldursson, O., Steinsson, K., Bornsson, L., et al. (1994). Giant cell arteritis Dolezalova, P., Telekesova, P., Nemcova, D., and Hoza, J. (2004). Incidence of
in Iceland. An epidemiologic and histopathologic analysis. Arthritis and vasculitis in children in the Czech Republic: 2-year prospective epidemi-
Rheumatism, 37, 1007–12. ology survey. Journal of Rheumatology, 31, 2295–9.
Barrier, J.H., Pion, P., Massari, R., et al. (1982). Approche epidemiologique de Dreyer, L., Faurschou, M., and Baslund, B. (2011). A population based
la maladie de Horton dans le departement de Loire-Atlantique—110 cas study of Takayasu’s arteritis in Denmark. Clinical and Experimental
en 10 ans (1970–9). Revue Medicine Interne, 1, 13–20. Rheumatology, 29 (1 Suppl. 64), S40–2.
Bartels, C., Bell, C., Rosenthal, A., Shinki, K., and Bridges, A. (2009). Decline Du, Z.D., Zhao, D., Du, J., Zhang, Y.L., Lin, Y., Lin, C., et  al. (2007).
in rheumatoid vasculitis prevalence among US veterans: a retrospective Epidemiologic picture of Kawasaki disease in Beijing from 2000 through
cross-sectional study. Arthritis and Rheumatism, 60, 2553–7. 2004. Pediatric Infectious Disease Journal, 26, 449–51.
CHAPTER 2 epidemiology of vasculitis 21

Dubreuil, M., Choi, H.K., Zhang Y., Zhu, Y. (2010). Sex, age, and birth cohort Gatenby, P.A., Lucas, R.M., Engelson, O., Ponsonby, A.L., and Clements.
effects on incidence of giant cell arteritis. Arthritis and Rheumatism, 62 M. (2009). Anti-neutrophil cytoplasmic antibody associated vasculi-
(Suppl. 10), 2199. tis: could geographic patterns be explained by ambient ultraviolet radia-
Duhaut, P., Pinede, L., Demolombe-Rague, S., et al. (1998). Giant cell arteritis tion. Arthritis and Rheumatism, 61, 1417–24.
and cardiovascular risk factors. A multi-centre, prospective case-control Gibbons, R.V., Parashar, U.D., Holman, R.C., et al. (2002). An evaluation of
study. Arthritis and Rheumatism, 41, 1960–5. hospitalizations for Kawasaki syndrome in Georgia. Archives of Pediatric
Duhaut, P., Pinede, L., Demolombe-Rague, S., et al. (1999). Giant cell arteritis Adolescent Medicine, 156, 492–6.
and polymyalgia rheumatica: are pregnancies a protective factor? A mul- Gibson, A., Stamp, L.K., Chapman, P.T., and O’Donnell, J.L. (2006). The epi-
ticentre, prospective case-control study. Rheumatology, 38, 118–23. demiology of Wegner’s granulomatosis and microscopic polyangiitis in
Duna, G.F., Cotch, M.F., Galperin, C., et  al. (1998). Wegener’s granulo- a southern hemisphere region. Rheumatology, 45, 624–8.
matosis:  role of environmental exposures. Clinical and Experimental Godman, G.C. and Churg, J. (1954). Wegener’s granulomatosis: pathology
Rheumatology, 16, 669–74. and review of the literature. Archives of Pathology, 58, 533–53.
Durongpisitkul, K., Sandtawesin, C., Khongphatthanayopthin, A., Panamonta, González, E.B., Varner, W.T., Lisse, J.R., et al. (1989). Giant cell arteritis
M., Soponammarak, S., Sittiwangkul, R., et  al. (2006). Epidemiologic in the Southern United States. Archives of Internal Medicine, 149,
study of Kawasaki disease and cases resistant to IVIg therapy in Thailand. 1561–5.
Asian Pacific Journal of Allergy Immunology, 24, 27–32. González-Gay, M.A., Amoli, M.M., Garcia-Porrua, C., and Ollier, W.E.
Eaton, W.W., Rose, N.R., Kalaydjian, A., et al. (2007) Epidemiology of auto- (2003a). Genetic markers of disease susceptibility and severity in
immune diseases in Denmark. Journal of Autoimmunity, 29, 1–9. giant cell arteritis and polymyalgia rheumatica. Seminars in Arthritis
Elling, P., Olsson, A.T., and Elling, H. (1996). Synchronous variations of the Rheumatism, 33, 38–48.
incidence of temporal arteritis and polymyalgia rheumatica in different González-Gay, M.A., Blanco, R., Sánchez-Andrade, A., et al. (1997). Giant
regions of Denmark, association with epidemics of Mycoplasma pneu- cell arteritis in Lugo, Spain: A more frequent disease with fewer classic
moniae infection. Journal of Rheumatology, 23, 112–9. features. Journal of Rheumatology, 24, 2166–70.
El-Reshaid, K., Kapoor, M., El-Reshaid, W., et al. (1997). The spectrum of González-Gay, M.A. and García-Porrúa, C. (1999). Systemic vasculitis in
renal disease associated with microscopic polyangiitis and classical adults in North Western Spain 1988–97. Clinical and epidemiological
polyarteritis nodosa in Kuwait. Nephrology Dialysis and Transplant, 12, aspects. Medicine, 78, 292–308.
1874–82. González-Gay, M., García-Porrúa, C., Guerrero, J., Rodriguez-Ledo, P.,
El-Reshaid, K., Varro, J., Al-Duwairi, Q., et al. (1995). Takayasu arteritis in Llorca, J. (2003b). The epidemiology of the primary systemic vascu-
Kuwait. Journal of Tropical Medicine and Hygiene, 98, 299–305. litides in northwest Spain: implications of the Chapel Hill Consensus
Fauci, A.S., Haynes, B.F., Katz, P., et  al. (1983). Wegener’s granulomato- Conference definitions. Arthritis and Rheumatism, 49, 388–93.
sis: prospective clinical and therapeutic experience with 85 patients for González-Gay, M., García-Porrúa, C., Rivas, M.J., Rodriguez-Ledo, P.,
21 years. Annals of Internal Medicine, 98, 76–85. and Llorca, J. (2001). Epidemiology of biopsy proven giant cell arteri-
Faurschou, M., Hlleberg, M., Obel, N., and Baslund, B. (2013). Incidence tis in northwestern Spain: trend over an 18 year period. Annals of the
of granulomatosis with polyangiitis (Wegener’s) in Greenland and the Rheumatic Diseases, 60, 367–71.
Faroe Islands:  epidemiology of an ANCA-associated vasculitis syn- González-Gay, M., Miranda-Filloy, J.A., Lopez-Diaz, J., Perez-Alvarez, R.,
drome in two ethnically distinct populations in the North Atlantic area. Gonzalez-Juanatey, C., Sanchez-Andrade, A., et  al. (2007). Giant cell
Clinical Experimental Rheumatology, 31 (Suppl. 75), S52–5. arteritis in north western Spain: a 25 year epidemiologic study. Medicine
Fischer, T.K., Holman, R.C., Yorita, K.L., Belay, E.D., Melbye, M., and Koch, (Baltimore), 86, 61–8.
A. (2007). Kawasaki syndrome in Demark. Pediatric Infectious Diseases Goronzy, J.J. and Weyand, C.M. (2012). Immune aging and autoimmunity.
Journal, 26, 411–5. Cellular and Molecular Life Sciences, 69, 1615–23.
Fledelius, H.C. and Nissen, K.R. (1992). Giant cell arteritis and visual loss. Gran, J.T. and Myklebust, G. (1997). The incidence of polymyalgia rheumatica
Acta Ophthalmologica, 70, 801–5. and temporal arteritis in the County of Aust Agder, South Norway: A pro-
Franzen, P., Sutinen, S., and Von Knorring, J. (1992). Giant cell arteritis and spective study 1987–94. Journal of Rheumatology, 24, 1739–43.
polymyalgia rheumatica in region of Finland: an epidemiologic, clinical Gregorini, G., Ferioli, A., Donato, F., et al. (1993). Association between silica
and pathologic study, 1984–1988. Journal of Rheumatology, 19, 273– 80. exposure and necrotizing crescentic glomerulonephritis with p-ANCA
Friedman, G., Friedman, B., and Benbassat, J. (1982). Epidemiology of tem- and anti-MPO antibodies: a hospital-based case control study. Advances
poral arteritis in Israel. Israel Journal of Medical Science, 18, 241–4. in Experimental Medicine and Biology, 336, 435–40.
Fries, J.F., Hunder, G.G., Bloch, D.A., et al. (1990). The American College of Grisaru, S., Yuen, G.W., Miettunen, P.M., and Hamikawa, L.A. (2010).
Rheumatology 1990 criteria for the classification of vasculitis: summary. Incidence of Wegener’s granulomatosis in children. Journal of
Arthritis and Rheumatism, 33, 1135–6. Rheumatology, 37, 440–2.
Fujimoto, S., Uezono, S., Hisanaga, S., et al. (2006). Incidence of ANCA-Associated Guillevin, L. (1999). Virus-associated vasculitides. Rheumatology, 38, 588–90.
Primary Renal Vasculitis in the Miyazaki Prefecture:  The First Guillevin, L., Cohen, P., Gayraud, M., et  al. (1999). Churg Strauss syn-
Population-Based, Retrospective, Epidemiologic Survey in Japan. Clinical drome: clinical study and long term follow up of 96 patients. Medicine
Journal of the American Society for Nephrology, 1, 1016–22. (Baltimore), 78, 26–37.
Fujimoto, S., Watts, R.A., Kobayashi, S., Suzuki, K., Jayne, D.R.W., Scott, Hall, S., Barr, W., Lie, J.T., et al. (1985). Takayasu arteritis: a study of 32 North
G.D.I, Hashimoto, H., and Nunoi, H. (2011). Comparison of the epi- American patients. Medicine, 64, 89–99.
demiology of antineutrophil cytoplasmic antibody (ANCA)-associated Harnden, A., Alves, B., and Sheikh, A. (2002). Rising incidence of Kawasaki
vasculitis between Japan and UK. Rheumatology, 50, 1916–20. disease in England: analysis of hospital admission data. British Medical
García-Porrúa, C. and González-Gay, M.A. (1999). Comparative clinical and Journal, 324, 1424–5.
epidemiological study of hyper- sensitivity vasculitis versus Henoch Harnden, A., Mayon-White, R., Perera, R., Yeates, D., Goldacre, M., and
Schönlein purpura in adults. Seminars in Arthritis and Rheumatism, 28, Burgner, D. (2009). Kawasaki disease in England:  ethnicity, dep-
404–12. rivation, and respiratory pathogens. Pediatrics Infectious Disease,
Gardner-Medwin, J.M., Dolezalova, P., Cummins, C., and Southwood, T.R. 28, 21–4.
(2002). Incidence of Henoch Schönlein purpura, Kawasaki disease, and Harrold, L.R., Andrade, S.E., Go, A.S., et  al. (2005). Incidence of Churg
rare vasculitides in children of different ethnic origins. Lancet, 360, Strauss syndrome in asthma drug users: a population based perspective.
1197–202. Journal of Rheumatology, 32, 1076–80.
22 SECTION 1 introductory chapters

Harrold, L.R., Patterson, M.K., Andrade, S.E., et  al. (2007). Asthma nephritis over a 22 year period in Fukushima prefecture, Japan. Internal
drug use and the development of Churg-Strauss syndrome (CSS). Urology Nephrology, 42, 1023–9.
Pharmacoepidemiology Drug Safety, 16, 620–6. Kermani, T.A., Schäfer, V.S., Crowson, C.S., Hnder, G.G., Gabriel, S.E.,
Hata, A., Noda, M., Moriwaka, R., and Numano, F. (1996). Angiographic Matteson, E.L., and Warrington, K.J. (2010). Increase in age at onset of
findings of Takayasu’s arteritis: a new classification. International Journal giant cell arteritis: a population based study. Annals of the Rheumatic
of Cardiology, 54 (Suppl.), S155–63. Diseases, 69, 780–1.
Haubitz, M., Waywadt, A., De Groot, K., et  al. (2005). Smoking habits in Kimura, A., Kitamura, H., Date, Y., and Numano, F. (1996). Comprehensive
patients diagnosed with ANCA associated small vessel vasculitis. Annals analysis of HLA genes in Takayasu’s arteritis in Japan. International
of the Rheumatic Diseases, 64, 1500–02. Journal of Cardiology, 54 (Suppl.), s61–9.
Haugeberg, G., Bie, R., Bendvold, A., et  al. (1998). Primary vasculitis in Knight, A., Ekbom, A., Brandt, L., et  al. (2006). Increasing incidence
a Norwegian community hospital:  a retrospective study. Clinical of Wegener’s granulomatosis in Sweden, 1975–2001. Journal of
Rheumatology, 17, 364–8. Rheumatology, 33, 2060–3.
Haugeberg, G., Irgens K.A., and Thomsen, R.S. (2003). No major differences Knight, A., Sandin, S., and Askling, J. (2008). Risks and relative risks of
in incidence of temporal arteritis in northern and western Norway Wegener’s granulomatosis among close relatives of patients with the dis-
compared with reports from southern Norway. Scandinavian Journal of ease. Arthritis and Rheumatism, 58, 302–7.
Rheumatology, 32, 318–9. Knight, A., Sandin, S., and Askling, J. (2010). Occupational risk factors for
Hauser, T., Mahr, A., Metzler, C., et  al. (2008). The leucotriene receptor Wegener’s granulomatosis. Annals of the Rheumatic Diseases, 69, 737–40.
antagonist montelu- kast and the risk of Churg-Strauss syndrome:  a Kobayashi, Y. and Numano, F. (2002). Takayasu’s arteritis. Internal Medicine,
case-crossover study. Thorax, 63, 677–82. 41, 44–6.
Heaton, P., Wilson, N., Nicholson, W., et al. (2006). Kawasaki Disease in New Kobayashi, S., Yano, T., Matsumoto, Y., et al. (2003). Clinical and epidemio-
Zealand. Journal of Paediatrics and Child Health, 42, 184–90. logical analysis of giant cell arteritis from a nationwide survey in 1998
Henoch, E.H. (1874). Uber ein eigenthumlioche Form von Purpura. Berliner in Japan: The first government supported nationwide survey. Arthritis
Klinika Wochenschrift, 11, 641–3. Rheumatism (Arthritis Care Research), 49, 594–8.
Heuclin T., Dubos F., Hue V., Godart F., Francart C., and Vincent, P. (2009). Koide, K. (1992). Takayasu arteritis in Japan. Heart Vessels, 7, 48–52.
Increased detection rate of Kawasaki disease using diagnostic algorithm, Koldingsnes, W. and Nossent, J.C. (2000). Epidemiology of Wegener’s granu-
including early use of echocardiography. Journal of Pediatrics, 155, 695–9. lomatosis in northern Norway. Arthritis and Rheumatism, 43, 2481–7.
Hissaria, P., Cai, F.Z., Ahern, M., et al. (2008). Wegener’s granulomatosis: epi- Kucukyavas, Y., Aksu, K., Solmaz, D., Can, G., Taylan, A., Sari, I., et al. (2012)
demiological and clinical features in a South Australian study. Internal Prevalence of Takayasu’s arteritis in Izmir, turkey: a multicentre study.
Medicine Journal, 38, 776–80. Annals of the Rheumatic Diseases, 71 (Suppl. 3), 231.
Hogan, S.L., Satterly, K.K., Dooley, M.A., Nachman, P.H., Jennette, J.C., Kurland, L.T., Chuang, T.Y., and Hunder, G.G. (1984). The epidemiology of
and Falk, R.J. (2001). Silica exposure in antineutrophil cytoplasmic systemic arteritis. In The epidemiology of the rheumatic diseases (eds R.C.
antibody-associated glomerulonephritis and lupus nephritis. Journal Lawrence and L.E. Shulman), pp. 196–205. Gower Publishing, New York.
American Society of Nephrology, 12, 134–42. Kurland, L.T., Hauser, W.A., Ferguson, R.H., et al. (1969). Epidemiologic fea-
Holder, S.M., Joy, M.S., and Falk, R.J. (2002). Cutaneous and systemic tures of diffuse connective tissue disorders in Rochester, Minn., 1951
manifestations of drug-induced vasculitis. Annals of Pharmacotherapy, through 1967, with special reference to systemic lupus erythematosus.
36, 130–47. Mayo Clinic Proceedings, 44, 649–63.
Holman, R.C., Christensen, K.Y., Belay, E.D., Steiner, C.A., Effler, P.V., Kussmaul, A. and Maier, R. (1866). Uber eine bisher nicht beschriebene
Miyamura, J., et al. (2010). Racial/ethnic differences in the incidence of eigenthümliche Arterienerkrankung (Periarteritis nodosa), die mit
Kawasaki syndrome among children in Hawaii. Hawaii Medical Journal, Morbus Brightü und rapid fortschreitender allgemeiner Muskellähmung
69, 194–7. einhergeht. Deutsche Archive Klinical Medizine, 1, 484–514.
Huang, W.C., Huang, L.M., Chang, I.S., Chang, L.Y., Chang, B.L., Chen P.J., Lam, B.L., Wirthlin, R.S., Gonzalez, A., Dubovy, S.R., and Feuer, W.J. (2007).
et  al. (2009). Epidemiologic features of Kawasaki disease in Taiwan Giant cell arteritis amongst Hispanic Americans. American Journal of
2003-2006. Pediatrics, 123, e401–5. Ophthalmology, 143, 161–3.
Hunder, G.G., Bloch, D.A., Michel, B.A., et al. (1990). The American College Lane, S.E., Watts, R.A., Bentham, G., Innes, N.J., and Scott, D.G. (2003). Are
of Rheumatology 1990 criteria for the classification of giant cell arteritis. environmental factors important in primary systemic vasculitis? A case
Arthritis and Rheumatism, 33, 1122–8. control study. Arthritis and Rheumatism, 48, 124–32.
Jagiello, P., Gencik, M., Arning, L., et  al. (2004). New genomic region Larsson, K., Mellström, D., Nordborg. C., Odén, A., and Nordborg, E. (2006).
for Wegener’s gran- ulomatosis as revealed by an extended associa- Early menopause, low body mass index, and smoking are independent
tion screen with 202 apoptosis related genes. Human Genetics, 114, risk factors for developing giant cell arteritis. Annals of the Rheumatic
468–77. Diseases, 65, 529–32.
Jennette, J.C., Falk, R.J., Andrassy, K., et  al. (1994). Nomenclature of sys- Leavitt, R.Y., Fauci, A.S., Bloch, D.A., et  al. (1990). The American
temic vasculitides. Proposal of an international consensus conference. College of Rheumatology criteria for the classification of Wegener’s
Arthritis and Rheumatism, 37, 187–92. Granulomatosis. Arthritis and Rheumatism, 33, 1101–7.
Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F., et al. Lee, Y.C., Kuo, H.C., Chank, J.S., Chang, L.Y., Huang, L., Chen, M.R., Liang,
(2013). 2012 Revised International Chapel Hill Consensus Conference C.D. et  al. (2012). Two new susceptibility loci for Kawasaki disease
Nomenclature of Vasculitides. Arthritis and Rheumatism, 65, 1–11. identified through genome-wide association analysis. Nature Genetics,
Jonasson, F., Cullen, J.F., and Elton, R.A. (1979). Temporal arteritis: a 14 year 44, 522–5.
epidemiological, clinical and prognostic study. Scottish Medical Journal, Li, X., Li, X., Li, H., Xu, M., and Zhou, M. (2008). Epidemiologic study of
24, 111–7. Kawasaki disease in Sichuan province of China. Journal of Tropical
Kawasaki, T. (1967). Acute febrile mucocutaneous syndrome with lymphoid Pediatrics, 52, 133–6.
involvement with specific desquamation of the fingers and toes in chil- Lin, Y.T., Manlhiot, C., Ching, J.C., Han, R.K., Nield, L.E., Dillenburg, R.,
dren: clinical observations in 50 cases. Japanese Journal of Allergology, et al. (2010). Repeated surveillance of Kawasaki disease in Ontario from
16, 178–222. 1995 to 2006. Pediatrics International, 52, 699–706.
Kawasaki, Y., Suyama, K., Yugeta, E., Katayose, M., Suzuki, S., and Nemeto, Liozon, E., Ouattara, B., Rhaiem, K., Ly, K., Bezanahary, H., Loustaud, V.,
K. (2010). The incidence of and severity of Henoch Schönlein purpura et al. (2009). Familial aggregation in giant cell arteritis and polymyalgia
CHAPTER 2 epidemiology of vasculitis 23

rheumatica: a comprehensive literature review including 4 new families. Noltrop, S. and Svensson, B. (1991). High incidence of polymyalgia rheu-
Clinical and Experimental Rheumatology, 27, S89–94. matica and giant cell arteritis in a Swedish community. Clinical and
Liu, L.J., Chen, M., Yu, F., Zhao, M.H., and Wang, H.Y. (2008). Evaluation of Experimental Rheumatology, 9, 351–5.
a new algorithm in classification of systemic vasculitis. Rheumatology, Nordborg, C., Johanssen, H., Petursdottir, V., and Nordborg, E. (2003). The
47, 708–12. epidemiology of giant cell arteritis: special reference to changes in the
Liu, N.H., LaBree, L.D., Feldon, S.E., and Rao, N.A. (2001). The epidemiol- age of the population. Rheumatology, 42, 549–52.
ogy of giant cell arteritis: a 12 year retrospective study. Ophthalmology, Nordborg, E. and Bengtsson, B.-Å. (1990). Epidemiology of biopsy-proven
108, 1145–9. giant cell arteritis (GCA). Journal of Internal Medicine, 227, 233–6.
Lyons, P.A., Rayner, T.F., Trivedi, S., Holle, J.U., Watts, R.A., Jayne, D.R.W., Ntatsaki, E., Mooney, J., Scott, D.G.I., and Watts, R.A.,(2013). Systemic rheu-
et  al. (2012). Genetically Distinct Subsets within ANCA-Associated matoid vasculitis in the era of modern immunosuppressive therapy.
Vasculitis. New England Journal of Medicine, 367, 214–23. Rheumatology, (in press).
Lynch, M., Holman, R.C., Mulligan, A., Belay, E.D., and Schonberger, L.B. Nuyts, G.D., van Vlem, E., de Vos, A., et al. (1995). Wegener’s granulomatosis
(2003). Kawasaki syndrome hospitalizations in Ireland, 1996 through is associated with exposure to silicon compounds: a case control study.
2000. Pediatric Infectious Disease Journal, 22, 959–63. Nephrology Dialysis and Transplantation, 10, 1162–5.
Ma, X.J., Yu, C.Y, Huang, M., Chen, S.B., Huang, M.R., Hunag, G.Y., et al. O’Donnell, J.L., Stevanovic, V.R., Frampton, C., et al. (2007). Wegener’s gran-
(2010). Epidemiologic features of Kawasaki disease in Shanghai ulomatosis in New Zealand: evidence for a latitude-dependent incidence
from 2003 through 2007. Chinese Medical Journal (English), gradient. Internal Medicine Journal, 37, 242–6.
123, 2629–34. Onouchi, Y., Ozaki, K., Burns, J.C., Terai, M., Hamada, H., et  al. (2012).
Macchioni, P., Catanoso, M., Chiarolanza, I., Manenti, L., Cavazza, A., Restuccia, A genome-wide association study identifies three new risk loci for
G., Germanò, G., et  al. (2011). Incidence of Wegener’s granulomatosis Kawasaki disease. Nature Genetics, 25, 517–21.
in the Reggio Emilia area of northern Italy: a 15-year population-based Ormerod, A.S. and Cook, M.C. (2008). Epidemiology of primary systemic
study. Annals of the Rheumatic Diseases, 70 (Suppl. 3), 634. vasculitis in the Australian Capital Territory and south eastern New
Machado, E.B., Gabriel, S.E., Beard, C.M., et  al. (1989). A population South Wales. Internal Medicine Journal, 38, 816–23.
based case-control study of temporal arteritis:  evidence for an asso- Ozen, S., Pistorio, A., Lusan, S.M., Bakkaloglu, A., Herlin, T., et  al.
ciation between temporal arteritis and degenerative vascular disease? (2010). Ruperto N., Dillon, M., et  al. (2006). EULAR/PRINTO/
International Journal of Epidemiology, 18, 836–41. PRES criteria for Henoch-Schönlein purpura, childhood Wegener’s
Mackie, S.L., Dasgupta, B., Hordon, L., Gough, A., Green, M., Hollywood, J., granulomatosis and childhood Takayasu arteritis: Ankara 2008. Part
et al. (2011). Ischemic manifestations in giant cell arteritis are associated II: final classification criteria. Annals of the Rheumatic Diseases, 69,
with area level socio-economic deprivation, but not cardiovascular risk 798–806.
factors. Rheumatology, 50, 2104–22. Pagnoux, C., Seror, R., Heneger, C., Mahr, A., Cohen, P., Le Guern, V., et al.
Mader, T.H., Werner, R.P., Chamberlain, D.G., and Doornbos, D. (2009). (2010). Clinical features and outcomes in 348 patients with polyarteritis
Giant cell arteritis in Alaska Natives. Canadian Journal of Ophthalmology, nodosa. Arthritis and Rheumatism, 62, 616–26.
44, 53–6. Pai, P., Bone, J.M., and Bell, G.M. (1998). Hydrocarbon exposure and
Mahr, A., Guillevin, L., Poissonnet, M., and Ayme, S. (2004). Prevalences of glomerulonephritis due to systemic vasculitis. Nephrology Dialysis
Polyarteritis nodosa, microscopic polyangiitis, Wegener’s granulomato- Transplantation, 13, 1321–3.
sis, and Churg Strauss syndrome in a French Urban multiethnic popula- Pamuk, O.N., Dönmezm S., Karahan, B., Pamuk, G.E., and Cakir, N.
tion in 2000: a capture-recapture estimate. Arthritis and Rheumatism, (2009). Giant cell arteritis and polymyalgia rheumatica in northwest-
51, 92–9. ern Turkey:  clinicalfeatures and epidemiological data. Clinical and
Malaviya, A.N., Kumar, A., Singh, Y.N., et al. (1990). Wegener’s granuloma- Experimental Rheumatology, 27, 830–3.
tosis in India: not so rare. British Journal of Rheumatology, 29, 499–500. Panagiotakis, S.H., Perysinakis, G.S., Kritikos, H., et al. (2009). The epidemi-
McCreay, R.D. (1986). ‘Epidemic’ of polymyalgia and temporal arteritis. ology of primary systemic vasculitides involving small vessels in Crete
Journal of Royal College of General Practitioners, 36, 523–4. (southern Greece): a comparison of older versus younger adult patients.
McMahon, B.J., Heyward, W.L., Templin, D.W., et al. (1989). Hepatitis B asso- Clinical and Experimental Rheumatology, 27, 409–15.
ciated polyarteritis nodosa in Alaskan eskimos: clinical and epidemio- Park, Y.W., Han, J.W., Hong, Y.M., Ma, J.S., Cha, S.H., Kwon, T.C., et  al.
logical features and long term follow up. Hepatology, 9, 97–101. (2011). Epidemiologic features of Kawasaki disease in Korea, 2006–
Mohammad, A., Jacobsson, L.T.H., Mahr, A., Sturfelt, G., and Segelmark, M. 2008. Pediatrics International, 53, 36–9.
(2007). Prevalence of Wegener’s Granulomatosis, microscopic polyangi- Penny, K., Fleming, M., Kazmierczak, D., and Thomas, A. (2010). An epi-
itis, polyarteritis nodosa and Churg Strauss syndrome. Rheumatology, demiological study of Henoch Schönlein purpura. Paediatric Nursing,
46, 1329–37. 22, 30–5.
Mohammad, A., Jacobsson, L.T.H., Westman, K.W.A., Sturfelt, G., and Pereira, L.S., Yoon, M.K., Hwang, T.N., Hong, J.E., Ray, K., Porco, T., and
Segelmark, M. (2009). Incidence and survival rates in Wegener’s McCulley, T. (2011). Giant cell arteritis in Asians: a comparative study.
Granulomatosis, microscopic polyangiitis, Churg Strauss syndrome and British Journal of Ophthalmology, 95, 214–16.
polyarteritis nodosa. Rheumatology, 48, 1560–5. Pettersson, E.E., Sundelin, B., and Heigl, Z. (1995). Incidence and outcome
Myasoedova, E., Crowson, C.S., Turesson, C., Gabriel, S.E., and Matteson, of pauci-immune necrotising glomerulonephritis in adults. Clinical
E.L. (2011). Incidence of extraarticular rheumatoid arthritis in Olmsted Nephrology, 43, 141–9.
County, Minnesota, in 1995-2007 versus 1985-1994: a population-based Petursdottir, V., Johannsson, H., Nordborg, C., and Nordborg, E. (1999). The
study. Journal of Rheumatology, 38, 983–9. epidemiology of biopsy-positive giant cell arteritis: special reference to
Mwipatayi, B.P., Jeffrey, P.C., Beningfield, S.J., et al. (2005). Takayasu arteri- cyclic fluctuations. Rheumatology, 38, 1208–12.
tis: clinical features and management: repport of 272 cases. Australian Pradhan, V.D., Badakere, S.S., Ghosh, K., and Pawar, A.R. (2004). Spectrum
New Zealand Journal of Surgery, 75, 1107. of anti-neutrophil cytoplasmic antibodies in patients with pulmonary
Nakamura, H., Yashiro, M., Uehara, R., Sadakane, A., Chihara, I., Aoyama, Y., tuberculosis overlaps with that of Wegener’s granulomatosis. Indian
et al. (2010). Epidemiologic features of Kawasaki disease in Japan: results Journal of Medical Science, 58, 283–8.
of the 2007-08 nationwide survey. Journal of Epidemiology, 20, 302–7. Rajala, S.A., Ahvenainen, J.E., Mattila, K.J., et al. (1993). Incidence and sur-
Ng, Y.M., Sung, R.Y., So, L.Y., et al. (2005). Kawasaki disease in Hong Kong. vival rate in cases of biopsy-proven temporal arteritis. Scandinavian
Hong Kong Medical Journal, 11, 331–5. Journal of Rheumatology, 22, 89–91.
24 SECTION 1 introductory chapters

Ramstead, C.L. and Patel, A.D. (2007). Giant cell arteritis in a neuro- Staples, J.A., Ponsonby, A.-L., Lim, L.L.-Y., and McMichael, A.M. (2003).
ophthalmology clinic in Saskatoon, 1988-2003. Canadian Journal of Ecologic analysis of some immune related disorders, including type
Ophthalmology, 42, 295–8. I diabetes, in Australia: Latitude, regional ultraviolet radiation, and dis-
Raynauld, J-P., Bloch, D.A., and Fries, J.F. (1993). Seasonal variation in the ease prevalence. Environmental Health Perspective, 111, 518–23.
onset of Wegener’s granulomatosis, polyarteritis nodosa and giant cell Stegeman, C.A., Cohen Tervaert, J.W., de Jong, P.E., et  al. (1996).
arteritis. Journal of Rheumatology, 20, 1524–6. Trimethoprim-ulphamethoxazole (co-trimoxazole) for the preven-
Reinhold-Keller, E., Herlyn, K., Wagner-Bastmeyer, R., and Gross, W. tion of relapses in Wegener’s granulomatosis. New England Journal of
(2005). Stable incidence of primary systemic vasculitides over five Medicine, 335, 16–20.
years:  results from the German vasculitis register. Arthritis and Stewart, M., Savage, J.M., Bell, B., et al. (1988). Long term renal prognosis
Rheumatism, 53, 93–9. of Henoch Schönlein purpura in an unselected childhood population.
Reinhold-Keller, E., Zeidler, A., Gutfleisch, J., et al. (2000). Giant cell arte- European Journal of Paediatrics, 147, 113–5.
ritis is more prevalent in urban than rural populations:  results of an Stratta, P., Messuerotti, A., Canavese, C., Coen, M., and Luccoli, L. (2001).
epidemiological study of primary systemic vasculitides in Germany. The role of metals in autoimmune vasculitis: epidemiological and patho-
Rheumatology, 39, 1396–402. genic study. Science Total Environment, 270, 179–90.
Royle, J.A., Williams, K., Elliott, E., et  al. (1998). Kawasaki disease in Takala, J.H., Kautiainen, H., Malmberg, H., et  al. (2008). Incidence
Australia, 1993–95. Archives of Diseases of Childhood, 78, 33–9. of Wegener’s granulomatosis in Finland 1981–2000. Clinical and
Russo, M.G., Waxman, J., Abdoh, A.A., et al. (1995). Correlation between Experimental Rheumatology, 26 ( Suppl. 49), S81–5.
infection and the onset of the giant cell arteritis (temporal) arteritis syn- Tamburlini, G., Strinati, R., Cadrini S., Calligaris, A., Coprivez, A., Cozzi,
drome. Arthritis and Rheumatism, 38, 374–80. M., et  al. (1984). A two year survey of mucocutaneoius lymph node
Sack, M., Cassidy, J.T., and Bole, G.G. (1975). Prognostic factors in polyarte- syndrome in north eastern Italy. Epidemiological and clinical findings.
ritis. Journal of Rheumatology, 2, 411–20. Helvetica Pediatrcis Acta, 39, 319–29.
Sahin, Z., Bicakcigil, M., Aksu, K., Kamali, S., Akar, S., Onen, F., et al. (2012). Tian, C., Kosoy, R., Nassir, R., Lee, A., Villoslada, P.L., et al. (2009). European
Takayasu’s arteritis is associated with HLA-B*52, but not with HLA-B*51 population genetic substructure: further definition of ancestry informa-
in Turkey. Arthritis Research and Therapy, 14, R27. tive marker for distinguishing among diverse European ethnic groups.
Salo, E. (1993). Kawasaki disease in Finland 1982–1992. Scandinavian Molecular Medicine, 15, 371–83.
Journal of Infectious Diseases, 25, 497–502. Tidman, M., Olander, R., Svalander, C., et al. (1998). Patients hospitalised
Salvarani, C., Crowson, C.S., O’Fallon, W.M., Hunder, G.G., and Gabriel, S.E. because of small vessel vasculitides with renal involvement in the period
(2004). Reappraisal of the epidemiology of giant cell arteritis in Olmsted 1975–95:  organ involvement, anti-neutrophil cytoplasmic antibodies
County, Minnesota, over a 50  year period. Arthritis and Rheumatism patterns, seasonal attack rates and fluctuation of annual frequencies.
(Arthritis Care Research), 51, 264–8. Journal of Internal Medicine, 244, 133–41.
Salvarani, C., Gabriel, S.E., O’Fallon, W.M., et al. (1995a). The incidence of Toshihiko, N. (1996). Current status of large and small vessel vasculitis in
giant cell arteritis in Olmstead County, Minnesota: apparent fluctuations Japan. International Journal of Cardiology, 54, S91–8.
in a cyclic pattern. Annals of Internal Medicine, 123, 192–4. Tourneux, P., Dufillot, D., Belloy, M., et  al. (2005). Kawasaki disease in
Salvarani, C., Gabriel, S.E., O’Fallon, W.M., et  al. (1995b). Epidemiology Guadeloupe. Presse Medicale, 15, 25–8.
of polymyalgia rheumatica in Olmsted County, Minnesota, 1970–91. Tsai, F.-J., Lee, Y.-C., Chang, J.-S., Huang, L.-M., Huang, F.-Y., Chiu, N.-C.,
Arthritis and Rheumatism, 38, 369–73. et  al. (2011). Identification of novel susceptibility loci for Kawasaki
Salvarani, C., Macchioni, P., Zizzi, F., et al. (1991). Epidemiologic and immu- diease in a Han Chinese popiulation by a genome-wide association
nogenetic aspects of polymyalgia rheumatica and giant cell arteritis in study. PLOS one, 6, e16853.
northern Italy. Arthritis and Rheumatism, 34, 351–6. Uehara, R. and Belay, E.D. (2012). Epidemiology of Kawasaki disease in Asia,
Sanchez, A.A., Acevedo, E.M., Sanchez, C.G., et  al. (2006). Evaluation of Europe and the United States. Journal of Epidemiology, 22, 79–85.
epidemiology of the primary systemic vasculitides in a Latin-American Vanoli, M., Daina, E., Salvarani, C., et al. (2005). Takayasu arteritis: a study of
well-defined population. Arthritis and Rheumatism, 54 (Suppl.), S757. 107 Italian patients. Arthritis and Rheumatism, 53, 100–7.
Schiller, B., Fasth, A., Bjorkheim, G., et  al. (1995). Kawasaki disease in Vinit, J., Muller, G., Bielefeld, P., Pfitzenmeyer, P., Bonniaud, P., Lorcerie, B.,
Sweden: incidence and clinical features. Acta Paediatrica, 84, 769–74. et al. (2009). Churg-Strauss syndrome: retrospective study in Burgundian
Schönlein, H. (1837). Allgemeine und Specielle Pathologie und Therapie, Vol. population in France in past 10 years. Rheumatology International, 31,
2, 3rd edn. Wurzburg, Herisau. 587–93.
Scott, D.G.I., Bacon, P.A., Elliott, P.J., et al. (1982). Systemic vasculitis in a district Waern, A.U., Andersson, P., and Hemmingsson, A. (1983). Takayasu’s arteri-
general hospital 1972–80:  clinical and laboratory features, classification tis: a hospital-region based study on occurrence, treatment and progno-
and prognosis of 80 cases. Quarterly Journal of Medicine, 203, 292–311. sis. Angiology, 34, 311–20.
Scott, D.G.I., Bacon, P.A., and Tribe, C.R. (1981). Systemic rheumatoid vas- Ward, M.M. (2004). Decreases in rates of hospitalizations for manifestations
culitis: a clinical and laboratory study of 50 cases. Medicine, 60, 288–97. of severe rheumatoid arthritis, 1983–2001. Arthritis and Rheumatism,
Singh, S., Aulakh, R., Bhalla, A.K., Suri, D., Manojkumar, R., Narula, N., 50, 1122–31.
et al. (2011). Is Kawasaki disease incidence rising in Chandrigarh, North Watts, R.A., Al-Taier, A., MacGregor, A., and Scott, D.G.I. (2009a). The inci-
India? Archives Disease Childhood, 96, 137–40. dence of Takayasu arteritis in the UK GPRD. Rheumatology, 48, 1008–11.
Singh, S., Balakrishnan, C., Mangat, G., Samant, R., Bambani, M., Kalke, S., Watts, R.A., Al-Taier, A., MacGregor, A., and Scott, D.G.I. (2009b). Prevalence
et  al. (2010). Giant cell arteritis in Mumbai. Journal of Association of and incidence of Wegener’s granulomatosis in the UK general practice
Physicians of India, 58, 372–4. research database. Arthritis and Rheumatism, 61, 1412–6.
Smeeth, L., Cook, C., and Hall, A.J. (2006). Incidence of diagnosed poly- Watts, R.A., Carruthers, D.M., and Scott, D.G. (1995). Isolated nail fold
myalgia rheumatica and temporal arteritis in the United Kingdom, vasculitis in rheumatoid arthritis. Annals of the Rheumatic Diseases,
1990-2001. Annals of the Rheumatic Diseases, 65, 1093–8. 54, 927–9.
Smith, C.A., Fidler, W.J., and Pinals, R.S. (1983). The epidemiology of giant Watts, R.A., González-Gay, M., Garcia-Porrua, C., Lane, S., Bentham, G., and
cell arteritis. Report of a ten year study in Shelby County, Tennessee. Scott, D.G.I. (2001). Geoepidemiology of systemic vasculitis. Annals of
Arthritis and Rheumatism, 26, 1214–9. the Rheumatic Diseases, 60, 170–2.
Sonnenblick, M., Nesher, G., Friedlande, Y., et al. (1986). Giant cell arteri- Watts, R.A., Jolliffe, V.A., Grattan, C.E.H., et al. (1998). Cutaneous vasculi-
tis in Jerusalem:  A  12  year epidemiological study. British Journal of tis in a defined population—clinical and epidemiological associations.
Rheumatology, 33, 938–41. Journal of Rheumatology, 25, 920–4.
CHAPTER 2 epidemiology of vasculitis 25

Watts, R.A., Lane, S.E., Bentham, G., et  al. (2000a). Epidemiology of sys- Wechsler, M., Wong, D., Miller, M.K., et al. (2009). Churg Strauss syndrome
temic vasculitis—a ten year study in the United Kingdom. Arthritis and in patients treated with omalizumab. Chest, 136, 507–18.
Rheumatism, 43, 414–19. Westman, K.W.A., Bygren, P.G., Olsson, H., et al. (1998). Relapse rate, renal
Watts, R.A., Lane, S., Bentham, G., and Scott, D.G.I. (2000b). Is giant cell survival, and cancer morbidity in patients with Wegener’s granulo-
arteritis more common in the north of Europe. Clinical Experimental matosis or microscopic polyangiitis and renal involvement. Journal of
Immunology, 120 (Suppl. 1), 63. American Society for Nephrology, 9, 842–52.
Watts, R.A., Lane S., Hanslik, T., et al. (2007). Development and validation of Yang, Y.H., Hung, C.F., Hsu, C.R., et al. (2005). A nationwide survey on epi-
a consensus methodology for the classification of the ANCA-associated demiological characteristics of childhood Henoch Schonlein purpura in
vasculitides and polyarteritis nodosa for epidemiological studies. Annals Taiwan. Rheumatology, 44, 618–22.
of the Rheumatic Diseases, 66, 222–7. Yashiro, M., Muso, E., Itoh-Ihara, T., et al. (2000). Significantly high regional
Watts, R.A., Mooney, J., Scott, D.G.I, et al. (2009c). A twenty year study of the morbidity of MPO-ANCA related angiitis and/or nephritis with respira-
epidemiology of ANCA associated vasculitis in UK. APMIS, 117 (Suppl. tory tract involvement after the 1995 great earthquake in Kobe (Japan).
127), 159. American Journal of Kidney Disease, 35, 889–95.
Watts, R.A., Mooney, J., Skinner, J., Scott, D.G.I., and MacGregor, A.J. Zeek, P.M. (1952). Periarteritis nodosa—a critical review. American Journal
(2012). The contrasting epidemiology of granulomatosis with poly- of Clinical Pathology, 22, 777–90.
angiitis (Wegener’s granulomatosis) and microscopic polyangiitis. Zeft, A., Schlesinger, M., Weiss, N., and Emery, H. (2005). Case control
Rheumatology, 51, 926–31. study of ANCA associated vasculitis in Western Montana. Arthritis and
Watts, R.A. and Scott, D.G.I. (2012). ANCA vasculitis:  to lump or split? Rheumatism, 52, S648.
Rheumatology, 51, 2115–7. Zhang, X., Zhang, Z., and Liu, S. (2012). Epidemiologic survey of
Watts, R.A., Suppiah, R., Merkel, P.A., and Luqmani, R.A. (2011). Systemic Kawasaki disease in Jilin from 1999 through 2008. Pediatric
Vasculitis—is it time to reclassify. Rheumatology, 50, 643–5. Cardiology, 33, 272–9.
SECTION 2

Basic science
CHAPTER 3

Hypersensitivity
T. Prescott Atkinson

Introduction practice of using hyperimmune animal antisera in the treatment of


such diseases as tetanus and diphtheria became more widespread
The study of hypersensitivity reactions, tissue damage occurring as (Goodall 1899; Goodall 1907; Goodall 1911; Currie 1907). At about
a result of an immunological response, is intertwined in the roots the same time, Maurice Arthus described the reaction that bears
of the science of immunology in the late nineteenth and early twen- his name:  a necrotizing, haemorrhagic local skin reaction that
tieth centuries. Many of the giants of the early days of immunology developed in rabbits repeatedly injected with horse serum subcu-
made indelible marks in the history of science through their studies taneously at 6-day intervals (Arthus 1903). Thus, within a 15-year
of hypersensitivity phenomena. The driving impetus for discovery period in the early twentieth century, hypersensitivity reactions
in the days before antibiotics was the medical treatment of infec- later recognized as involving IgE-mast cell activation, antibody–
tions in patients for whom little could be done outside immuno- complement–Fc receptor interactions, and T-cell activation had
logical modulation. In the early twentieth century it is ironic that been identified. All primarily depend upon the adaptive portion of
scientific interest in immunomodulation using vaccines and other the immune system. It would be decades before even the most basic
‘biologics’ as prevention and cure for infectious and inflammatory molecular details of the mechanisms at work in these reactions
diseases is renewed with the realization that microbial resistance became clear, and it would be fair to say that none are completely
to antibiotics is inexorably increasing. This chapter is an attempt to understood at present.
review the history of the various hypersensitivity phenomena, the To these classical immunological hypersensitivity reactions
framework for differentiating them, and the recent advances in our should be added at least one more that involves the innate arm of the
understanding of the complex mechanisms at work. immune response. Gregory Shwartzman first described the ‘phe-
The history of the discovery of hypersensitivity reactions is punc- nomenon of local tissue reactivity’ (Shwartzman 1928; Shwartzman
tuated by many famous names. As with any other area of science, few 1937). In his systematic exploration of this remarkable hypersensi-
of these researchers could claim that their work was not built upon tivity reaction Shwartzman demonstrated that a preparatory intra-
an edifice of knowledge constructed by a multitude of predecessors dermal injection of culture filtrate from a variety of Gram-negative
and contemporaries whose names are much less well known today. bacteria followed by an intravenous injection of filtrate about 24
It is noteworthy that in 1798, far before the development of the sci- hours later resulted in a thrombohaemorrhagic reaction at the site
ence of immunology, Edward Jenner published the first example of of the original injection. The phenomenon did not seem to involve
delayed hypersensitivity in a controlled setting with his description classical immunological reactivity as it occurred too rapidly and
of a local cutaneous ‘reaction of immunity’, which developed over was not transferable by serum; however, it could be blocked by pre-
24–48 hours in subjects reinnoculated with vaccinia virus (Movat treatment with antiserum to the specific bacterial agent used.
1979). Robert Koch rediscovered the phenomenon in 1890 with his A generalized form of the Shwartzman reaction resembling dis-
description of a similar reaction in tuberculous patients injected seminated intravascular coagulation (DIC) was probably observed
intradermally with tuberculin. The phenomenon of anaphylaxis prior to Shwartzman’s work by Sanarelli (1924) and later by Apitz
described by Paul Portier and Charles Richet in 1902 (Portier and (1934). This systemic thrombohaemorrhagic reaction characterized
Richet 1902), for which work Richet received the Nobel prize in by coagulopathy, microthrombi, reduced platelets, and leukocytes,
1913, provided evidence of the existence of explosive hypersensi- and often bilateral renal cortical haemorrhage, could be reliably
tivity reactions which obviously differed fundamentally from the induced by two intravenous injections of live Gram-negative bac-
delayed reactions described by Koch and Jenner. teria or culture supernatant 24 hours apart. Later workers who
The term ‘allergy’, meaning ‘other reactivity’ (as opposed to ‘nor- explored the pathogenesis of the generalized reaction included such
mergy’, the normal host response), was first suggested in 1906 by famous researchers as Lewis Thomas and Robert A. Good (Thomas
Clemens P. von Pirquet, who believed that immunity and hyper- and Good 1952). Insight (albeit as yet incomplete) into the patho-
sensitivity, host protection and tissue damage, were opposite ends genesis of the Shwartzman reaction would have to wait until the
of the spectrum of the immune response, a concept that is arguably close of the twentieth century with the explosion of knowledge
still acceptable today. Together with Bela Schick one year earlier, regarding inflammatory cytokines and the cellular elements of the
von Pirquet had described serum sickness, a peculiar illness that immune system that generate them.
developed in individuals who had been treated with horse serum as The generalized Shwartzman reaction has been shown to depend
treatment for diphtheria (Pirquet and Schick 1905). Physicians of upon the action of interferon gamma (IFN-γ), tumour necrosis fac-
the time became intimately acquainted with serum sickness as the tor (TNF-α), and IL-12 (Ozmen et al. 1994). Lipopolysaccharide
30 SECTION 2 basic science

(LPS)-induced IL-12 and IL-15 from macrophages appear to induce Table 3.1 Gell and Coombs classification of hypersensitivity reactions
IFN-γ production, which then sensitizes macrophages to the sec-
ond exposure 18–24 hours later (Fehniger et  al. 2000). In mice Type I: Anaphylactic
there is an increased susceptibility with increasing age that corre- Allergic rhinoconjunctivitis
lates with increased production of TNF-α in response to LPS, and Acute urticaria
priming with IL-12 can substitute for the priming dose of LPS (Sato Allergic asthma
et al. 2005). Furthermore, neutralization of LPS-induced TNF-α or Atopic dermatitis
IL-12-induced IFN-γ protects mice from the lethal effects of the Anaphylaxis
systemic reaction. The primary subsets of cells that generate the Type II: Cytolytic or cytotoxic
key inflammatory cytokine IFN-γ, at least in mice, appear to be Drug-induced immune cytopenias
CD3+ natural killer T (NKT) cells when low doses of LPS are used Infection-induced (e.g. Goodpasture’s syndrome)
and orthodox natural killer (NK) cells at higher doses of endotoxin Transfusion reactions
(Kim et al. 2000; Dieli et al. 2000). Sato and colleagues identified a Hyperacute graft rejection
CD8+ CD122+ T-cell subset in mice that appears to play an essen- Haemolytic disease of the newborn (Rh incompatibility)
tial role in the reaction (Sato et al. 2005). A storm of inflamma- Type III: Immune complex/Arthus reaction
tory cytokines following the second injection results in DIC and Drug-induced immune complex disease (‘serum-sickness like’)
life-threatening shock. Interestingly, activation of NKT cells with Infection-induced (e.g. acute rheumatic fever)
KRN7000, a synthetic homolog for the potent NKT cell antigen Arthus reactions (e.g. to immunizations)
αGalCer, has been reported to block the generalized Shwartzman Hypersensitivity pneumonitis
Serum sickness
reaction if administered near the time of the priming dose of LPS,
possibly by converting the cytokine response of NKT cells from Type IV: Cell-mediated
IFN-γ to IL-4 and IL-10 (Sireci et al. 2007). Drug reactions
The best-known classification scheme for hypersensitivity Delayed-type hypersensitivity (e.g. insect bites, vaccines)
reactions involving the adaptive immune system was published Contact hypersensitivity (e.g. Rhus dermatitis)
by P.G.H. Gell and R.R.A. Coombs in 1963 (Table 3.1) (Coombs Hypersensitivity pneumonitis
Foreign body reaction
and Gell 1968), and it is still a useful framework for thinking
Chronic graft rejection
about them. Although the different forms of antigen-specific
hypersensitivity disorders will be discussed within this setting,
it should be kept in mind that they offer only a general reference
point for understanding the initiation points for the complex ‘without place’) to describe a heterogeneous group of hypersensitiv-
processes of inflammation that ensue; as knowledge increases ity disorders including hay fever, allergic asthma, and atopic der-
with regard to more enigmatic elements of the immune system, matitis (or ‘flexural neurodermatitis’ as it was then known), which
for example NK cells, NKT cells, or γδ T cells, our ability to dis- had hitherto been difficult to categorize (Coca and Cooke 1923).
tinguish specific hypersensitivity disorders related to those ele- The existence of a transferable serum factor in systemic anaphylaxis
ments may improve. had been demonstrated in animals by Charles Richet and a simi-
It is likely that as knowledge expands, new types of hypersensitiv- lar factor was shown to exist in humans with local cutaneous ana-
ity disorders involving the adaptive immune system will be found phylaxis by Prausnitz and Küstner in 1921 (Prausnitz and Küstner
that do not fit this classification system. For example, the recently 1921). The nature of this transferable ‘reaginic’ hypersensitivity
described entity now designated IgG4-related disease (IgG4-RD), remained undefined for another 50 years until the discovery of IgE
a pleomorphic group of disorders affecting diverse organ systems by Kimishige Ishizaka and colleagues in 1966 (Ishizaka et al. 1966).
with the unifying features of a lymphoplasmacytic infiltrate with The genetic basis for atopy is the subject of intensive ongo-
IgG4-producing plasma cells, storiform fibrosis, obliterative phle- ing study (Binia and Kabesch 2012). A number of chromosomal
bitis, and mild-to-moderate tissue eosinophilia, often accompa- regions have been associated with allergic disease, including
nied by an elevated serum IgG4 level, may eventually turn out to most prominently 11q13 (containing the high-affinity IgE recep-
represent a type of hypersensitivity syndrome (Carruthers et  al. tor (FcεRI) beta subunit), 5q31-33 (containing the cytokine gene
2012). Uniquely among the immunoglobulins, IgG4 has the ability cluster of interleukin-3,4,5,9,13, and granulocyte-macrophage
to exchange Fab arms so that heavy chains of different specificities colony-stimulating factor), and 12q (containing IFN-γ, KIT ligand
can dissociate and recombine, producing immunoglobulins with (stem cell factor), insulin-like growth factor-1 (IGF-1), and the
different binding specificities (Davies et al. 2013). It may be that constitutive form of nitric oxide synthetase (NOS1)) (Manian
this monovalency combined with an inability to activate comple- 1997). Specific polymorphisms have been identified in a number
ment is related to the propensity for IgG4 overproduction to result of candidate genes that appear to confer susceptibility to atopy and/
in localized tissue inflammation. or allergic asthma (Barnes 1999; Blumenthal 2000; Borish 1999;
Rosenwasser 1999). These include the IL-4 receptor alpha chain
(Hershey et al. 1997; Kruse et al. 1999; Ober et al. 2000; Oiso et al.
Type I hypersensitivity 2000; Rosa-Rosa et al. 1999), the promoter region of the IL-4 gene
Disorders such as ‘summer catarrh’ or ‘hay fever’ (allergic rhinocon- (Burchard et al. 1999; Song et al. 1996), the b2 adrenergic receptor
junctivitis) had been described beginning in the early 1800s but the (Green et al. 1993; Reihsaus et al. 1993), the 5′ flanking region of the
pathogenic mechanism was unclear (Bostock 1828). In 1923, Coca CD14 gene (Baldini et al. 1999), and acidic mammalian chitinase
and Cooke first proposed the term ‘atopy’ (from the Greek meaning (Zhu et al. 2004; Bierbaum et al. 2005). A recent review of hundreds
CHAPTER 3 hypersensitivity 31

of genetic association studies revealed that eight genes had been potentially reducing the time required for initial release follow-
linked with asthma/ atopy in at least five studies each: interleukin-4 ing cellular activation (Bradding et al. 1993; Bradding et al. 1995;
(IL4), interleukin-13 (IL13), β2 adrenergic receptor (ADRB2), MacLeod et al. 1997; Wedemeyer and Galli 2000). Early activation
human leukocyte antigen DRB1 (HLA-DRB1), TNF-α (TNF), of mast cells in mice appears to be essential for protection from
lymphotoxin-alpha (LTA), FcεRI beta subunit (FCER1B), and IL-4 fast moving infections in the peritoneal cavity; mice genetically
receptor alpha subunit (IL4RA) (Hoffjan et al. 2003). With convinc- deficient in mast cells succumb to experimental peritonitis induced
ing evidence for contributions from such a wide variety of genes, by caecal ligation and puncture (Echtenacher et al. 1996; Malaviya
atopy is most likely the product of contributions from multiple et al. 1996). IL-4 is essential for the production of IgE and the induc-
genes as well as the environment. tion of Th2 responses (Brown and Hural 1997; Kuhn et al. 1991).
The aptly named Hygiene Hypothesis postulates that it is the low Mast cell activation also results in rapid de novo synthesis of
level of infection, exposure to microbial products, and parasitism in lipid-derived mediators including leukotrienes C4 and D4 (LTC4
Western society that has fostered the rise in type I hypersensitivity and LTD4), which act primarily to increase vascular permeability at
disorders (Heederik and von Mutius 2012; Schaub et al. 2006; Liu the site of mediator release, and prostaglandin D2 (PGD2), which
and Leung 2006). Under this line of reasoning, individuals with a acts to increase smooth muscle tone, to increase blood flow locally,
polygenic predisposition for atopy have a lower threshold for spe- and to stimulate mucus secretion (Boyce 2005). Platelet-activating
cific IgE formation, a trait that may be adaptive in regions where factor (PAF), another lipid-derived mediator probably also pro-
helminth infections are endemic but is detrimental in developed duced by mast cell activation, increases smooth muscle tone, pro-
nations where such parasites are less common. In atopic individu- duces increased microvascular permeability, and activates local
als, the IgE system is thus more likely to respond inappropriately to endothelial cells promoting expression of adhesion molecules
environmental antigens from pollen and foods, producing symp- (Stafforini et al. 2003).
toms in the absence of high levels of parasite-specific IgE. This Mast cell granules contain a variety of enzymes, particularly
predisposition to exuberant IgE responses is exacerbated in envi- neutral proteases (Caughey 2011; Lutzelschwab et al. 1997) as well
ronmental conditions lacking in Th1 stimulating infections and as a proteoglycan matrix, which is responsible for their distinctive
microbial compounds such as LPS. metachromatic staining property in histological sections. At least
Mast cells are of fundamental importance in type I hypersensi- five different chymases and two different tryptases are associated
tivity (Galli and Tsai 2012). Particularly abundant along mucosal with mast cell granules. The functions of mast cell-derived tryptases
and epithelial surfaces and in perivascular locations, mast cells are appear to include a kallikrein-like activity catalysing the formation
well-suited to early amplification of local inflammatory responses of bradykinin, which enhances increased vascular permeability
and appear to act as sentinels for local injury. IgE binds to the mul- produced by histamine (Imamura et al. 2004; Fiorucci and Ascoli
timeric high-affinity IgE receptor, FceRI, primarily on mast cells 2004). Activation of diverse cellular responses to mast cell-derived
and basophils, where it may remain on the cell surface for weeks or chymases and tryptases occurs partly through the activation of
months. Atopic individuals respond to antigenic stimulation, par- G-protein-coupled proteinase-activated receptors (PAR), with chy-
ticularly at low levels over mucosal borders, with Th2 type cytokine mases selectively activating PAR1 and tryptases PAR2 (Rothmeier
production, which results in specific IgE production (Biedermann and Ruf 2012; Reed and Kita 2004).
and Rocken 1999). Dosing of antigen-mediated receptor aggrega- Mast cells are capable of responding to a variety of other stim-
tion on mast cells has revealed that the EC50 for IL-4 production uli, including complement fragments C5a and C3a, and physical
is among the lowest of all the cytokines produced by mast cells stimuli such as heat, cold, and hyperosmolarity. Polyamines such
(Gonzales-Espinosa et al. 2003), occurring even below the thresh- as compound 48/80, spermine, and a variety of basic polypeptides
old for histamine release, and this may be at least part of the expla- such as substance P elicit rapid exocytosis from rodent mast cells as
nation for the tendency for Th2 responses to occur at low-level well as human skin mast cells (Ansel et al. 1993; Mousli et al. 1994;
antigen stimulation. Yano et al. 1989). Plant lectins such as concanavalin A aggregate
Experimental activation of mucosal mast cells through the cell surface glycoproteins on the cell, including the IgE receptor and
high-affinity IgE receptor with allergen challenge results in two produce robust mast cell activation. Finally, a number of cytokines,
distinct phases of inflammation in many subjects: an acute phase particularly stem cell factor, have been found to induce degranula-
marked by angioedema, pruritus, and vasodilatation and a delayed tion in mast cells (Alam et al. 1994; Galli and Costa 1995; Lukacs
phase some 6–12 hours later, which is characterized by a cellular et al. 1996; Tsai et al. 1993). The ability of mast cells to respond to
infiltrate, principally with neutrophils, eosinophils, and basophils such a wide array of stimuli highlights their importance in the early
(Charlesworth et al. 1989; White 1999). With intracellular amounts phase of the inflammatory response.
at approximately 10 pg/cell, histamine can reach millimolar con- The late phase inflammatory response that follows extensive mast
centrations locally around the mast cell when released. The bioac- cell degranulation is marked by a cellular influx rich in neutrophils
tivity of histamine accounts for virtually all the acute-phase effects and eosinophils, but basophils have also been recognized to play a
of mast cell activation while TNF- accounts for a major portion of role in allergic inflammation (Galli and Tsai 2012; Tsai et al. 2005).
delayed phase effects (Wershil et al. 1991). Like mast cells, basophils also express the high-affinity IgE recep-
A bewildering array of cytokines is synthesized by mast cells, tor at relatively high surface densities enabling their response to
including IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-8, IL-13, IL-16, allergens in the area, and they also release stored histamine as well
GM-CSF, TNF-α, TGF-β, bFGF, FGF-2, PDGF, NGF, VPF/VEGF, as newly synthesized eicosanoids and cytokines, particularly IL-4
and several C-C chemokines (Wedemeyer and Galli 2000; Stone and IL-13, in response to IgE receptor aggregation by allergen (Guo
et al. 2010). In at least some mast cell populations in humans and et al. 1994; Iliopoulos et al. 1992; Lichtenstein and Bochner 1991;
rodents TNF-α and IL-4 can be stored preformed in granules, Marone et al. 1997; Redrup et al. 1998). Mast cell elaboration of
32 SECTION 2 basic science

cytokines such as IL-5, IL-8, and TNF-α with concomitant activa- membranous nephritis has been shown to be associated with
tion of local endothelium is likely the major factor controlling the autoantibodies, often of the IgG4 subclass, to podocyte antigens,
influx of these granulocytic cell types (Bradding et al. 1993; Moller most notably the M-type phospholipase A2 receptor, suggesting an
et al. 1993; Tanimoto et al. 1992; Zhang et al. 1995). The immigrant overlap between this type of nephropathy and IgG4-related disease
cell populations called in by initial mast cell activation are thus able (IgG4-RD), a recently described disorder of essentially unknown
to potentiate the regional inflammatory response with their own pathogenesis involving overproduction of IgG4 locally in a variety
mediators. of organ systems with subsequent inflammatory sequellae (Makker
and Tramontano 2011; Carruthers et al. 2012).
Fcγ receptors on phagocytes have recently assumed an increas-
Type II hypersensitivity ingly important role in type II and type III hypersensitivity dis-
Despite extensive experimental work over the course of the entire orders (Li et  al. 2009). Using mice homozygous for a targeted
twentieth century, many of the details of the type II and type III deletion of the Fc receptor common gamma chain, Ravetch and
hypersensitivity reactions remain to be deciphered. The distinc- colleagues have shown that even prototypical type II disorders such
tion between the two is probably somewhat artificial, but there is as immune haemolytic anaemia and thrombocytopenia in mice do
some merit in discriminating between antibody reactions directed not develop in the absence of functional Fc receptors (Clynes and
against surfaces (cells or structural elements of the extracellular Ravetch 1995; Ravetch 2010). Importantly, mutational deletion of a
matrix) (type II) and those that occur initially in liquid phase (type specific Fc receptor, FcγRIIB, which down-regulates the activation
III). The production of cellular damage from binding of antibody of other Fc receptors, renders the animals more prone to the devel-
to the surfaces of cells or tissue structures followed by activation of opment of autoreactive antibodies (Ravetch 2010; Nakamura et al.
complement and Fc receptor bearing cells such as neutrophils and 2000; Suzuki et al. 1998).
macrophages is the traditional model used to explain tissue injury The production of antibodies to cell surface molecules may also
in type II hypersensitivity. Since the reaction occurs against com- result in modulation of cellular functions when the surface mol-
ponents of tissues, not surprisingly many examples involve autoim- ecule is a receptor, a transmembrane channel, or other molecule
munity. The involvement of autoantibodies against endothelial cell with a signalling function. Cell-bound antibody may either block
antigens is, at present, a plausible but controversial cause of vascu- normal receptor function as in myasthenia gravis or stimulate
litis (Youinou et al. 2005), which will be discussed elsewhere in this abnormal receptor activity as in Graves’ disease. Recent evidence
volume (see Chapter 6). indicates that chronic urticaria, formerly thought to be a form of
The development of accurate animal models of human autoim- type I hypersensitivity, is caused in about 50% of cases by autoanti-
mune diseases represents an important step in understanding the bodies against IgE or the alpha chain of the high-affinity IgE recep-
pathogenesis of these disorders and in the production of novel tor, which triggers mast cell degranulation and the typical pruritic
therapies (Table 3.2) (Cohen and Miller 1994; Bigazzi 1998; Hall rash (Greaves 2000; Kaplan and Greaves 2009).
and Cook 2011). The roles and relative importance of complement
and Fc receptors have proven particularly difficult to understand.
In animal models of antibody-mediated autoimmune disease, such Type III hypersensitivity
as glomerulonephritis induced by heterologous antibody to renal Tissue damage produced by soluble immune complexes (IC) has
tissues or specific glomerular components, the involvement of been studied extensively over the past century. IC probably form
phagocytes is generally essential to full expression of the disease routinely during the clearance of antigens derived from infec-
(Cochrane et  al. 1965; Schrijver et  al. 1990; Suzuki et  al. 1998). tious agents and under normal circumstances are cleared without
In certain cases, however, such as Heymann nephritis, which is sequellae. Under certain conditions, however, IC may occasion-
induced by immunization of rats with brush border antigens, par- ally produce devastating tissue damage. As with type II reactions,
ticularly megalin, the resultant nephritis appears to be also depend- IC disease may also be due to autoimmunity when the antigen
ent on the assembly of terminal complement components into involved is self-derived, but the source of the offending complexes
the membrane attack complex (Farquhar et al. 1995). In humans, in this disorder is frequently related to infection or drug therapy.
Much of the morbidity and mortality in the prototypical autoim-
mune disease systemic lupus erythematosus (SLE) is mediated by
Table 3.2 Animal models in type II hypersensitivity
immune complexes. In contrast to the situation in type II disor-
Heymann nephritis (Farquhar et al. 1995)
ders, IC disease is clearly implicated in at least one form of vas-
culitis, previously designated hypersensitivity vasculitis but now
Anti-Thy-1 antibody-induced glomerulonephritis (Eppel et al. 2000) termed small-vessel vasculitis (SVV) (Jennette et al. 2013; Russell
Autoimmune glomerular basement membrane disease (Nakamura et al. 2000; and Gibson 2006). This form of vasculitis is discussed in detail else-
Tang et al. 2000) where in this volume (see Chapter 5).
Experimental autoimmune myasthenia gravis (Vincent 1994) Immune complex injury to tissues may be induced by reaction of
antigen and antibody to form IC (a) within the vessel lumen where
Experimental Graves’ disease (Ludgate 2000)
they subsequently move into the vessel and tissues (serum sickness
Collagen-induced arthritis (Myers et al. 1997) reaction) or (b) within the vessel wall and outside the vessel within
Experimental bullous pemphigoid (Liu et al. 1993; Liu et al. 1995) the tissues (Arthus reaction). Different forms of the Arthus reac-
tion have been produced, and, although usually elicited in the skin,
Experimental autoimmune haemolytic anaemia (Cox and Howles 1981;
the reaction may be produced in any vascular tissue (Ranadive and
Nisitani et al. 1997; Sharon and Naor 1992)
Movat 1979). The active or direct reaction occurs when antigen is
CHAPTER 3 hypersensitivity 33

injected intradermally in a hyperimmune animal. In this situation, rabbits occurs in three phases: the first 2 days being that of equili-
IgG antibody within the extravascular space reacts with the injected bration in the extra- and intravascular compartments, the second
antigen locally. Passive reactions are produced when antibody and consisting of relatively slow catabolism over the next 8 days, and a
antigen are both injected in a naïve animal, the reverse passive reac- final rapid phase of elimination over 2–3 days as immune complex
tion occurring when antibody is injected intradermally and antigen formation rises. The size of the complexes increases with increasing
is given intravenously. antibody concentration until antibody excess occurs, and the devel-
Perhaps not surprisingly, experimental evidence demonstrates opment of vasculitis and glomerulonephritis is accompanied by a
that the most severe vascular injury occurs when the reaction significant fall in serum haemolytic complement titres (Cochrane
occurs within the vessel wall as in the reverse passive Arthus reac- and Hawkins 1968; Dixon et  al. 1958). Vascular and glomerular
tion, as opposed to administration of both antibody and antigen lesions in rabbits correlate almost entirely with the appearance of
either intravenously or intradermally (Cochrane and Weigle 1958). large circulating IC (greater than 19S). IC decline in serum and free
Mast cells appear to be important in the evolution of the direct antibody appears by day 13, approximately 2–3 days prior to resolu-
response because extravasation of Evans Blue dye from the vascu- tion of vasculitis. In the course of a reaction to a single injection,
lature is seen within 3–5 minutes after injection, and this increased segmental vasculitic lesions are produced that are typically found
permeability is suppressible by antihistamines (Hayashi et al. 1964). within coronaries, joints, and glomeruli. There is usually a trans-
This immediate increase in local vascular permeability is largely mural neutrophilic infiltrate but little necrosis or haemorrhage.
absent in the passive Arthus reaction. Deposition of immunoglobulin and complement in vessel walls,
The mechanisms involved in immune complex disease have been on the surface of the synovium, and in glomeruli is readily dem-
the focus of intensive study. It has been known for decades that neu- onstrated in fresh lesions in serum sickness. Severe glomerulone-
trophils are required for the development of Arthus lesions (Cochrane phritis in experimental animals usually develops only with chronic
et al. 1959). Phagocytosis and clearance of IC within vessel walls begins antigenaemia induced by repeated injections. The mild glomerular
within minutes of initiation (Hopken et al. 1997). More recent stud- injury in acute serum sickness is ablated neither by decomplemen-
ies using mice in which the Fc receptor common gamma chain has tation nor by induced leukopenia while the arteritis is significantly
been eliminated by targeted genetic deletion demonstrate that, as with decreased by either manipulation. Interestingly, vasoactive amines
cytotoxic antibodies in type II reactions, type III reactions require the potentiate the development of vascular lesions. Passive anaphylaxis
presence of functional Fc receptors (Clynes et al. 1999; Clynes et al. or injection of anaphylatoxins in experimental animals worsens
1998; Ravetch 2010). At least a part of this requirement appears to inflammatory lesions, and their appearance in response to these
relate to a necessary function for FcγRIII on mast cells in the direct agents is blocked by antihistamines (Ranadive and Movat 1979). In
Arthus reaction (Sylvestre and Ravetch 1996). Finally, as in type II human patients with circulating immune complexes, injection of
hypersensitivity reactions, deletion of the down-regulatory FcγRIIB histamine into uninvolved areas of skin can elicit necrotizing cuta-
receptor in mice potentiates IC-mediated tissue damage (Clynes et al. neous vasculitis (Chossegros et al. 1987; Gower et al. 1977; Jorizzo
1999). The interaction of Fc receptors on phagocytes and mast cells et al. 1983; Wolff et al. 1978). On the basis of these studies, the acti-
with IC thus forms an essential step in both the clearance of IC and vation of basophils and platelets with release of vasoactive amines
the production of the severe inflammatory lesion. in individuals with circulating IC is likely to worsen the vascular
Decomplementation of animals with cobra venom factor gener- injury during a serum sickness reaction.
ally does not abrogate immune complex-mediated inflammatory The importance of complement in clearance of soluble IC as
reactions, although the resulting tissue damage may be impaired well as those that have become deposited in vessel walls and other
(Ranadive and Movat 1979). Activation of complement and release tissues is believed to explain the pronounced tendency for indi-
of anaphylatoxins C3a and C5a have been regarded as important in viduals with genetic deficiencies in early classical pathway compo-
the activation of local mast cells in the direct reaction and basophils nents (C1, C2, C4) to develop a SLE-like disorder (Pettigrew et al.
and platelets within the vasculature as well as directing the neutro- 2009). Genetic deficiency of C1 has a nearly 100% association with
philic influx. In support of this suggestion, deletion of the receptor SLE-like symptoms, the strongest risk factor yet identified (Walport
for C5a in mice eliminates IC-mediated pulmonary inflammation et al. 1998). In addition, autoantibodies to C1q, particularly of the
and significantly reduces, but does not eliminate, the inflammatory IgG2 subclass, are strongly associated with lupus nephritis, partic-
response to the reverse passive Arthus reaction (Hopken et al. 1997). ularly in combination with the FcγRIIB R131 allele, which binds
Shushakova and colleagues found that C5a may regulate the levels of IgG2 poorly (Haseley et al. 1997). In aggregate, these observations,
activating and inhibitory Fcγ receptors on phagocytes (Shushakova both in animals and in humans, suggest that complement gener-
et  al. 2002). Recent evidence suggests that the regulation of Fcγ ally plays an augmentative but not an essential role in IC-mediated
receptor activation by the C5a receptor is further modulated by inflammation. On the other hand, deposition of complement in
the C-type lectin dectin-1, which binds to highly galactosylated immune complexes is important in targeting complexes for clear-
IgG1-containing immune complexes and by recruiting FcγRIIB acts ance by phagocytes; persistence of complexes in tissues produced
to down-regulate C5a receptor activation (Karsten et al. 2012). either by deficiencies in early classical pathway complement com-
A somewhat different mechanism of tissue injury occurs when ponents or in the phagocyte receptors for complement appears to
massive quantities of immune complexes arise within the circu- set the stage for chronic inflammation and eventual autoimmunity.
lation during the development of serum sickness. In rabbits the
condition can be readily produced by intravenous injection of 10
ml of xenogeneic serum or 0.25 g of purified foreign protein (e.g. Type IV hypersensitivity
albumin) per kg of weight (Ranadive and Movat 1979). Early stud- As noted in the introductory paragraphs, cellular hypersensitivity
ies determined that antigen (bovine serum albumin) elimination in as a clinical phenomenon was noted by Jenner and Koch. It had
34 SECTION 2 basic science

been observed that delayed-type hypersensitivity (DTH) was most Table 3.3 Animal models in type IV hypersensitivity
often seen in subjects suffering from infections, particularly chronic
infections, with mycobacteria, fungi, or viruses (Pappenheimer and Antigen-dependent
Freund 1959). The diagnostic use of DTH reactions to intrader- Murine hypersensitivity pneumonitis (Fink et al. 2005)
mal antigens derived from the organisms causing tuberculosis, Contact hypersensitivity (numerous models, e.g. Landsteiner and Chase
histoplasmosis, brucellosis, leptospirosis, and a variety of other 1939; Maguire and Chase 1967; Christensen and Haase 2012)
infections has been extensively employed in the clinic. However, Chronic graft rejection (Bedi et al. 2010)
Granulomatous hypersensitivity (Kunkel et al. 1998)
immunization of control subjects or animals with purified micro-
bial products from the same organisms or other foreign proteins Infection-induced
elicited antibody formation but usually little DTH unless the anti- Experimental leprosy (granulomatous hypersensitivity) (Crawford
gens were injected intradermally (Lipton and Freund 1953; Mote and Hardwick 2011)
and Jones 1936; Salvin 1958). The addition of microbial products, Murine leishmaniasis (Belosevic et al. 1989)
Murine listeriosis (Mielke et al. 1997; North et al. 1997)
particularly mycobacterial, as adjuvants greatly increased the DTH
response elicited with routes of administration other than intra- Autoimmunity
dermal (Pappenheimer and Freund 1959). One of the earliest and Experimental autoimmune uveitis (Merryman et al. 1987; Tarrant et al.
most efficient methods discovered involved the injection of protein 1998; Xu et al. 1997)
antigens directly into tuberculous foci such as lymph nodes (Hay Experimental autoimmune encephalomyelitis (Smeltz and Swanborg 1998)
1979). The separate nature of DTH reactions from those involving Experimental autoimmune thyroiditis (Braley-Mullen et al. 1985; Okayasu 1985)
Experimental autoimmune neuritis (Zhu et al. 1998)
antibody was convincingly demonstrated by the ready elicitation of
Experimental autoimmune myocarditis (Smith and Allen 1993)
DTH in patients with Bruton’s agammaglobulinaemia (Good et al.
Experimental autoimmune sialoadenitis (Greiner et al. 1991)
1957; Porter 1957).
Extensive experimental work was carried out to explore the clini-
cal observation of DTH. Landsteiner and colleagues did much to
shed light on the underlying mechanisms (Landsteiner and Jacobs by erythema, often several-fold higher than the baseline flow rates
1935). The dependence of DTH reactions upon cells rather than in surrounding skin (Hay et al. 1975). Increased vascular perme-
humoral factors was first demonstrated convincingly in work by ability can simultaneously be demonstrated experimentally using
Landsteiner, Chase, and Lawrence in the 1940s (Landsteiner and Evan’s blue, and this phenomenon is not affected by antihistamines,
Chase 1940; Chase 1945; Chase 1946; Lawrence 1949; Chase 1976). indicating that mast cell-derived mediators play a minimal role in
With the identification of thymus-derived lymphocytes as a distinct the evolving response (Phair et  al. 1970). Although neutrophils
lineage of lymphoid cells, it quickly became apparent that it was this may appear in variable numbers at the injection site within hours
fraction of circulating leukocytes that transferred the DTH reaction of challenge, the most characteristic cellular infiltrate is composed
(Cooper 1972; Jaffer et al. 1973; Youdim et al. 1973). Following the of lymphocytes and monocytes, typically in prominent perivenular
discovery of the Th1/Th2 T cell paradigm in mice by Mosmann and cuffs. The increased blood flow, vascular permeability, and the cel-
colleagues (Mosmann et al. 1986; Mosmann 1992), most cellular lular infiltrate create an induration in the tissues that is a useful
hypersensitivity reactions, including those involved in experimen- measure of the intensity of the reaction in experimental animals
tal animal models of organ-specific autoimmunity (some examples and can be quantitated by measuring changes in footpad or ear
are listed in Table 3.3) were subsequently found to depend upon thickness with a micrometer.
cells with a Th1 phenotype (Cher and Mosmann 1987; De Carli IL-12 produced by dendritic cells and macrophages during the
et al. 1994; Ohta et al. 1997). Although the dichotomy of functional initial stages of inflammation is a key cytokine involved in driv-
T cell subsets has not proven to be as well-defined in humans, the ing Th1 development (Trinchieri 1996). The Th1 subset of T lym-
general patterns of cytokine production remain a useful conceptual phocytes elaborates an arsenal of inflammatory cytokines, which
framework in the analysis of immune responses leading to DTH. function in amplifying the activation of phagocytes in the defence
DTH reactions differ from antibody-mediated reactions in their against intracellular pathogens such as viruses, atypical bacteria,
slow development, generally peaking after 1–2  days rather than and fungal pathogens. Among these, T cell and NK cell-derived
minutes (type I  reactions) or hours (types II–III) (Hay 1979). IFN-γ is perhaps the most crucial in further activating tissue mac-
A variety of somewhat different reactions can be elicited depending rophages and in suppression of Th2 differentiation (Mosmann et al.
on the tissue and antigen used. These include contact hypersensi- 1995). IFN-γ produced by Th1 cells forms part of a positive feed-
tivity (Christensen and Haase 2012), tuberculin-type hypersensi- back loop with IL-12 produced by phagocytes, each driving the
tivity (Black 1999), hypersensitivity pneumonitis (Fink et al. 2005), other higher (Trinchieri 1996). This feedback loop is opposed by
granulomatous hypersensitivity (Crawford and Hardwicke 2011; Th2 cytokines, particularly IL-10.
Kunkel et al. 1998), and allograft rejection (Bedi et al. 2010). The Some of the clearest evidence for the importance of the Th1
complex cascade of immunological events from the initial sensi- response in resistance to intracellular infections has come from
tization and subsequent re-exposure, leading to the final effector experimental infection of mice with Leishmania major. The infec-
response in each type, varies depending on the type of tissue and tion results in a Th1 pattern of T-cell activation, which, when
the nature of the challenge. opposed by the injection of anti-IFN-γ antibody, is converted to a
The classic reaction is that seen following the intradermal Th2 cytokine pattern with a lethal outcome (Belosevic et al. 1989).
injection of tuberculin in an individual who has been previously The importance of the IL-12–IFN-γ amplification loop in protection
infected with M. tuberculosis. Within several hours of the injection from intracellular pathogens is further illustrated by the propensity
increased blood flow develops around the injection site manifested of human patients deficient in the receptor for IFN-γ, IL-12, or the
CHAPTER 3 hypersensitivity 35

IL-12 receptor (or essential downstream signalling components, Bigazzi, P.E. (1998). Animal models of autoimmunity:  spontaneous and
e.g. STAT 1) to develop recurrent Infections with Listeria monocy- induced. In The Autoimmune Diseases (3rd edn) (ed. N.R. Rose and I.R.
togenes and atypical mycobacteria (Rosenzweig and Holland 2005; Mackay), pp. 211–44. Academic Press, St Louis, USA.
Binia, A. and Kabesch, M. (2012). Respiratory medicine—genetic base for
Cottle 2011). Well-formed granulomas are not seen in the tissues
allergy and asthma. Swiss Medical Weekly, 142, w13612.
of such patients despite abundant mycobacteria, indicating that
Black, C.A. (1999). Delayed type hypersensitivity: current theories with an
cellular activation by this cytokine pathway is an essential step in historic perspective. Dermatology Online Journal, 5,7.
containing the pathogen. Furthermore, individuals who form neu- Blumenthal, M.N. (2000). Genetics of asthma and allergy. Allergy and Asthma
tralizing autoantibodies to IFN-γ or IL-12 may also develop sus- Proceedings, 21, 55–9.
ceptibility to similar types of infections, further underscoring the Borish, L. (1999). Genetics of allergy and asthma. Annals of Allergy, Asthma,
importance of these cytokines in cellular immune defence against and Immunology, 82, 413–24.
intracellular pathogens (Browne and Holland 2010). Bostock, J. (1828). On the catarrhus aestivus or summer catarrh.
Medico-Chirurgical Transactions (London), XIV, 437–46.
Boyce, J.A. (2005). Eicosanoid mediators of mast cells: receptors, regulation
Conclusion of synthesis, and pathobiologic implications. Chemical Immunology and
Hypersensitivity reactions are exaggerated immune responses, that Allergy, 87, 59–79.
is normal responses of various arms of the immune system to anti- Bradding, P., Feather, I.H., Wilson, S., Bardin, P.G., Heusser, C.H., Holgate,
S.T., and Howarth, P.H. (1993). Immunolocalization of cytokines in the
genic stimulation that have become so exuberant as to damage the nasal mucosa of normal and perennial rhinitic subjects. The mast cell as
host they evolved to protect. In living animals they rarely occur in a source of IL-4, IL-5, and IL-6 in human allergic mucosal inflammation.
isolation, but are combined with each other during the evolution Journal of Immunology, 151, 3853–65.
of an inflammatory response. Natural infections frequently induce Bradding, P., Okayama, Y., Howarth, P.H., Church, M.K., and Holgate, S.T.
both cellular and humoral immunity, and the relative balance of (1995). Heterogeneity of human mast cells based on cytokine content.
these two arms of the specific immune response is critical in the Journal of Immunology, 155, 297–307.
defence against pathogens, which vary from obligate intracellular Braley-Mullen, H., Johnson, M., Sharp, G.C., and Kyriakos, M. (1985).
pathogens such as viruses and complex extracellular parasites such Induction of experimental autoimmune thyroiditis in mice with in vitro
activated splenic T cells. Cellular Immunology, 93, 132–43.
as helminths. The study of these phenomena has provided the sci-
Brown, M.A. and Hural, J. (1997). Functions of IL-4 and control of its expres-
ence of immunology with windows into the intricate workings of sion. Critical Reviews in Immunology, 17, 1–32.
immunity. Browne, S.K. and Holland, S.M. (2010). Immunodeficiency secondary to
anticytokine autoantibodies. Current Opinions in Allergy and Clinical
References Immunology, 10, 534–41.
Alam, R., Kumar, D., Anderson-Walters, D., and Forsythe, P. A. (1994). Burchard, E.G., Silverman, E.K., Rosenwasser, L.J., Borish, L., Yandava, C.,
Macrophage inflammatory protein-1 alpha and monocyte chemoattract- Pillari, A., Weiss, S.T., Hasday, J., Lilly, C.M., Ford, J.G., and Drazen,
ant peptide-1 elicit immediate and late cutaneous reactions and activate J.M. (1999). Association between a sequence variant in the IL-4 gene
murine mast cells in vivo. Journal of Immunology, 152, 1298–303. promoter and FEV(1) in asthma. American Journal of Respiratory and
Ansel, J.C., Brown, J.R., Payan, D.G., and Brown, M.A. (1993). Substance P Critical Care Medicine, 160, 919–22.
selectively activates TNF-alpha gene expression in murine mast cells. Carruthers, M.N., Stone, J.H., and Khosroshahi, A. (2012). The latest on
Journal of Immunology, 150, 4478–85. IgG4-RD: a rapidly emerging disease. Current Opinion in Rheumatology,
Apitz, K. (1934). Die Wirkung bakterieller Kulturfi ltrate nach Umstimmung 24, 60–9.
des gesamten Endothels beim Kaninchen. Virchows Archiv, 293, 1–33. Caughey, G.H. (2011). Mast cell proteases as protective and inflammatory
Arthus, M. (1903). Injections répétées de sérum de cheval chez le lapin. mediators. Advances in Experimental Medicine and Biology, 716, 212–34.
Comptes Rendus de la Societé de Biologie, 55, 817–20. Charlesworth, E.N., Iliopoulos, O., MacDonald, S.M., Kagey-Sobotka, A.,
Baldini, M., Lohman, I.C., Halonen, M., Erickson, R.P., Holt, P.G., and and Lichtenstein, L.M. (1989). Cells and secretagogues involved in the
Martinez, F.D. (1999). A Polymorphism* in the 5’ flanking region of the human late-phase response. International Archives of Allergy and Applied
CD14 gene is associated with circulating soluble CD14 levels and with Immunology, 88, 50–3.
total serum immunoglobulin E. American Journal of Respiratory Cell Chase, M.W. (1945). The cellular transfer of cutaneous hypersensitivity to
and Molecular Biology, 20, 976–83. tuberculin. Proceedings of the Society for Experimental Biology and
Barnes, K.C. (1999). Gene-environment and gene-gene interaction stud- Medicine, 59, 134–5.
ies in the molecular genetic analysis of asthma and atopy. Clinical and Chase, M.W. (1946). The cellular transfer of cutaneous hypersensitivity.
Experimental Allergy, 29 (Suppl. 4), 47–51. Journal of Bacteriology, 51, 643.
Bedi D.S., Riella L.V., Tullius, S.G., and Chandraker A. (2010). Animal models Chase, M.W. (1976). Developments in delayed-type hypersensitivi-
of chronic allograft injury: contributions and limitations to understand- ties: 1950-1975. Journal of Investigative Dermatology, 67,136–48.
ing the mechanism of long-term graft dysfunction. Transplantation, 90, Cher, D.J. and Mosmann, T.R. (1987). Two types of murine helper T cell
935–44. clone. II. Delayed-type hypersensitivity is mediated by TH1 clones.
Belosevic, M., Finbloom, D.S., Van Der Meide, P.H., Slayter, M.V., and Nacy, Journal of Immunology, 138, 3688–94.
C.A. (1989). Administration of monoclonal anti- IFN-γ antibodies in Chossegros, P., Wu, R., Hermier, C., Doutre, M.S., Brette, R., and Trepo, C.
vivo abrogates natural resistance of C3H/HeN mice to infection with (1987). [Satellite vasculitis of B or non-A non-B hepatitis. Diagnostic
Leishmania major. Journal of Immunology, 143, 266–74. value of a provocation test by intradermal injection of histamine].
Biedermann, T. and Rocken, M. (1999). Th1/Th2 balance in atopy. Springer Annales de Medecine Interne, 138,193–8.
Seminars in Immunopathology, 21, 295–316. Christensen, A.D. and Haase, C. (2012). Immunological mechanisms of con-
Bierbaum, S., Nickel, R., Coch, A., Lau, S., Deichmann, K.A., Wahn, U., tact hypersensitivity in mice. APMIS, 120,1–27.
Superti-Furga, A., and Heinzmann, A. (2005). Polymorphisms and haplo- Clynes, R., Dumitru, C., and Ravetch, J.V. (1998). Uncoupling of immune
types of acid mammalian chitinase are associated with bronchial asthma. complex formation and kidney damage in autoimmune glomerulone-
American Journal of Respiratory and Critical Care Medicine, 172, 1505–9. phritis. Science, 279, 1052–54.
36 SECTION 2 basic science

Clynes, R., Maizes, J.S., Guinamard, R., Ono, M., Takai, T., and Ravetch, J.V. Fink, J.N., Ortega, H.G., Reynolds, H.Y., Cormier, Y.F., Fan, L.L., Franks,
(1999). Modulation of immune complex-induced inflammation in vivo T.J., Kreiss, K., Kunkel, S., Lynch, D., Quirce, S., Rose, C., Schleimer,
by the coordinate expression of activation and inhibitory Fc receptors. R.P., Schuyler, M.R., Selman, M., Trout, D., and Yoshizawa, Y. (2005).
Journal of Experimental Medicine, 189, 179–85. Needs and opportunities for research in hypersensitivity pneumonitis.
Clynes, R. and Ravetch, J.V. (1995). Cytotoxic antibodies trigger inflamma- American Journal of Respiratory and Critical Care Medicine, 171, 792–8.
tion through Fc receptors. Immunity, 3, 21–6. Fiorucci, L. and Ascoi, F. (2004). Mast cell tryptase, a still enigmatic enzyme.
Coca, A.F. and Cooke, R.A. (1923). On the classification of the phenomena of Cellular and Molecular Life Sciences, 61, 1278–95.
hypersensitiveness. Journal of Immunology, 8, 163–75. Galli, S.J. and Costa, J.J. (1995). Mast-cell-leukocyte cytokine cascades in
Cochrane, C.G. and Hawkins, D. (1968). Studies on circulating immune allergic inflammation. Allergy, 50, 851–62.
complexes III. Factors governing the ability of circulating complexes to Galli, S.J. and Tsai, M. (2012). IgE and mast cells in allergic disease. Nature
localize in blood vessels. Journal of Experimental Medicine, 127, 137–54. Medicine, 18, 693–704.
Cochrane, C.G., Unanue, E.R., and Dixon, F.J. (1965). A role of polymorpho- Gonzales-Espinosa, C., Odom, S., Olivera, A., Hobson, J.P., Martinez, M.E.C.,
nuclear leukocytes and complement in nephrotoxic nephritis. Journal of Oliveira-dos-Santos, A., Barra, L., Spiegel, S., Penninger, J.M., and Rivera,
Experimental Medicine, 122, 99–119. J. (2003). Preferential signaling and induction of allergy-promoting
Cochrane, C.G. and Weigle, W.O. (1958). The cutaneous reaction to soluble lymphokines upon weak stimulation of the high affinity IgE receptor on
antigen-antibody complexes. A comparison with the Arthus reaction. mast cells. Journal of Experimental Medicine, 197, 1453–65.
Journal of Experimental Medicine, 108, 591–604. Good, R.A., Zak, S.J., Jensen, D.R., and Pappenheimer, A.M. (1957). Delayed
Cochrane, C.G., Weigle, W.O., and Dixon, F.J. (1959). The role of polymor- allergy and agammaglobulinemia. Journal of Clinical Investigation,
phonuclear leukocytes in the initiation and cessation of the Arthus vas- 36, 894.
culitis. Journal of Experimental Medicine, 110, 481–94. Goodall, E.W. (1899). On the value of the treatment of diphtheria by anti-
Cohen, I.R. and Miller, A. (eds) (1994). Autoimmune disease models. toxin. British Medical Journal, 1, 268–70.
Academic Press, Inc., New York. Goodall, E.W. (1907). On the supersensitation of persons by horse-serum.
Coombs, R.R.A. and Gell, P.G.H. (1968). Classification of allergic reactions Journal of Hygiene (London), 7, 607–18.
responsible for clinical hypersensitivity and disease. In Clinical aspects of Goodall, E.W. (1911). On some points connected with the serum treatment
immunology (2nd edn) (ed. R.R.A. Coombs and P.G.H. Gell), pp. 575– of diphtheria. British Medical Journal, 1, 292–6.
96. Alden Press, Oxford. Gower, R.G., Sams, W.M., Jr, Thorne, E.G., Kohler, P.F., and Claman, H.N.
Cottle, L.E. (2011). Mendelian susceptibility to mycobacterial disease. (1977). Leukocytoclastic vasculitis: sequential appearance of immuno-
Clinical Genetics, 79, 17–22. reactants and cellular changes in serial biopsies. Journal of Investigative
Cooper, M.G. (1972). Delayed-type hypersensitivity in the mouse. II. Transfer Dermatology, 69, 477–84.
by thymus-derived (T) cells. Scandinavian Journal of Immunology, 1, Greaves, M. (2000). Chronic urticaria. Journal of Allergy andClinical
237–45. Immunology, 105, 664–72.
Cox, K.O. and Howles, A. (1981). Induction and regulation of autoimmune Green, S.A., Cole, G., Jacinto, M., Innis, M., and Liggett, S.B. (1993). A poly-
hemolytic anemia in mice. Immunological Reviews, 55, 31–53. morphism of the human beta 2-adrenergic receptor within the fourth
Crawford, C.L. and Hardwicke, P.M. (2011). Experimental leprosy: a model transmembrane domain alters ligand binding and functional properties
of epithelioid cell granuloma. International Journal of Dermatology, 50, of the receptor. Journal of Biological Chemistry, 268, 23116–21.
255–61. Greiner, D.L., Angelillo, M., Wayne, A.L., Fitzgerald, K.M., Rozenski, D., and
Currie, J.R. (1907). On the supersensitation of persons suffering from diph- Cutler, L.S. (1991). Experimental autoallergic sialadenitis in the LEW
theria by repeated injections of horse serum. Journal of Hygiene (Lond), rat. III. Role of CD4+ T cells in EAS induction. Cellular Immunology,
7, 35–60. 135, 354–9.
Davies, A.M., Rispens, T., den Bleker, T.H., McDonnell, J.M., Gould, H.J., Guo, C.B., Liu, M.C., Galli, S.J., Bochner, B.S., Kagey-Sobotka, A., and
Aalberse, R.C., and Sutton, B.J. (2013). Crystal structure of the human Lichtenstein, L.M. (1994). Identification of IgE-bearing cells in the
IgG4 C(H)3 dimer reveals the role of Arg409 in the mechanism of late-phase response to antigen in the lung as basophils. American Journal
Fab-arm exchange. Molecular Immunology, 54, 1–7. of Respiratory Cell and Molecular Biology, 10, 384–90.
De Carli, M., D’Elios, M.M., Zancuoghi, G., Romagnani, S., and Del Prete, G. Hall, S.W. and Cooke, A. (2011). Autoimmunity and inflammation: murine
(1994). Human Th1 and Th2 cells: functional properties, regulation of models and translational studies. Mammalian Genome, 22, 377–89.
development and role in autoimmunity. Autoimmunity, 18, 301–8. Haseley, L.A., Wisnieski, J.J., Denburg, M.R., Michael-Grossman, A.R., Ginzler,
Dieli, F., Sireci, G., Russo, D., Taniguchi, M., Ivanyi, J., Fernandez, C., E.M., Gourley, M.F., Hoffman, J.H., Kimberly, R.P., and Salmon, J.E. (1997).
Troye-Blomberg, M., De Leo, G., and Salerno, A. (2000). Resistance of Antibodies to C1q in systemic lupus erythematosus: characteristics and
natural killer T cell-deficient mice to systemic Shwartzman reaction. relation to Fc gamma RIIA alleles. Kidney International, 52,1375–80.
Journal of Experimental Medicine, 192, 1645–51. Hay, J.B. (1979). Delayed (cellular) hypersensitivity. In Inflammation, immu-
Dixon, F.J., Vazquez, J.J., Weigle, W.O., and Cochrane, C.G. (1958). nity, and hypersensitivity (2nd ed) (ed H.Z. Movat), pp. 271–318. Harper
Pathogenesis of serum sickness. A.M.A. Archives of Pathology, 65, 18–28. and Row, New York.
Echtenacher, B., Mannel, D. N., and Hultner, L. (1996). Critical protective Hay, J.B., Johnston, M.G., Hobbs, B.B., and Movat, H.Z. (1975). The use of
role of mast cells in a model of acute septic peritonitis. Nature, 381, 75–7. radioactive microspheres to quantitate hyperemia in dermal inflam-
Eppel, G.A., Takazoe, K., Nikolic-Paterson, D.J., Lan, H.Y., Atkins, R.C., and matory sites. Proceedings of the Society for Experimental Biology and
Comper, W.D. (2000). Characteristics of albumin processing during Medicine, 150, 641–4.
renal passage in anti-Thy1 and anti-glomerular basement membrane Hayashi, H., Yoshinaga, M., Koono, M., Miyoshi, H., and Matsumura, M.
glomerulonephritis. American Journal of Kidney Diseases, 35, 418–26. (1964). Endogenous permeability factors and their inhibitors affecting
Farquhar, M.G., Saito, A., Kerjaschki, D., and Orlando, R.A. (1995). The vascular permeability in cutaneous Arthus reactions and thermal injury.
Heymann nephritis antigenic complex:  megalin (gp330) and RAP. British Journal of Experimental Pathology, 45, 419–35.
Journal of the American Society of Nephrology, 6, 35–47. Heederik, D., and von Mutius, E. (2012). Does diversity of environmental
Fehniger, T.A., Yu, H., Cooper, M.A., Suzuki, K., Shah, M.H., and Caligiuri, microbial exposure matter for the occurrence of allergy and asthma?
M.A. (2000). Cutting edge:  IL-15 costimulates the generalized Journal of Allergy andClinical Immunology, 130, 44–50.
Shwartzman reaction and innate immune IFN-γ production in vivo. Hershey, G.K., Friedrich, M.F., Esswein, L.A., Thomas, M.L., and Chatila,
Journal of Immunology, 164, 1643–47. T.A. (1997). The association of atopy with a gain-of-function mutation
CHAPTER 3 hypersensitivity 37

in the alpha subunit of the interleukin-4 receptor. New England Journal Lipton, M. M. and Freund, J. (1953). Allergic encephalomyelitis in the rat
of Medicine, 337, 1720–5. induced by the intracutaneous injection of central nervous tissue and
Hoffjan, S., Nicolae, D., and Ober, C. (2003). Association studies for asthma adjuvants. Journal of Immunology, 71, 98.
and atopic diseases: a comprehensive review of the literature. Respiratory Liu, A.H. and Leung, D.Y.M. (2006). Renaissance of the hygiene hypothesis.
Research, 4, 14–25. Journal of Allergy andClinical Immunology, 117, 163–66.
Hopken, U.E., Lu, B., Gerard, N.P., and Gerard, C. (1997). Impaired inflam- Liu, Z., Diaz, L.A., Troy, J.L., Taylor, A.F., Emery, D.J., Fairley, J.A., and
matory responses in the reverse Arthus reaction through genetic dele- Giudice, G.J. (1993). A passive transfer model of the organ-specific
tion of the C5a receptor. Journal of Experimental Medicine, 186, 749–56. autoimmune disease, bullous pemphigoid, using antibodies gener-
Iliopoulos, O., Baroody, F.M., Naclerio, R.M., Bochner, B.S., Kagey-Sobotka, ated against the hemidesmosomal antigen, BP180. Journal of Clinical
A., and Lichtenstein, L.M. (1992). Histamine-containing cells obtained Investigation, 92, 2480–8.
from the nose hours after antigen challenge have functional and pheno- Liu, Z., Giudice, G.J., Swartz, S.J., Fairley, J.A., Till, G.O., Troy, J.L., and Diaz,
typic characteristics of basophils. Journal of Immunology, 148, 2223–8. L.A. (1995). The role of complement in experimental bullous pemphig-
Imamura, T., Potempa, J., and Travis, J. (2004). Activation of the oid. Journal of Clinical Investigation, 95, 1539–44.
kallikrein-kinin system and release of new kinins through alterna- Ludgate, M. (2000). Animal models of Graves’ disease. European Journal of
tive cleavage of kininogens by microbial and human cell proteinases. Endocrinology, 142, 1–8.
Biological Chemistry, 385, 989–96. Lukacs, N.W., Kunkel, S.L., Strieter, R.M., Evanoff, H.L., Kunkel, R.G., Key,
Ishizaka, K., Ishizaka, T., and Hornbrook, M.M. (1966). Physicochemical M.L., and Taub, D.D. (1996). The role of stem cell factor (c-kit ligand)
properties of human reaginic antibody. IV. Presence of a unique immu- and inflammatory cytokines in pulmonary mast cell activation. Blood,
noglobulin as a carrier of reaginic activity. Journal of Immunology, 87, 2262–8.
97, 75–85. Lutzelschwab, C., Pejler, G., Aveskogh, M., and Hellman, L. (1997). Secretory
Jaffer, A.M., Jones, G., Kasdon, E.J., and Schlossman, S.F. (1973). Local trans- granule proteases in rat mast cells. Cloning of 10 different serine pro-
fer of delayed hypersensitivity by T lymphocytes. Journal of Immunology, teases and a carboxypeptidase A from various rat mast cell populations.
111, 1268–9. Journal of Experimental Medicine, 185,13–29.
Jennette, J.C., Falk, R.J., Bacon, P.A., et al. (2013). 2012 revised international MacLeod, J.D., Anderson, D.F., Baddeley, S.M., Holgate, S.T., McGill, J.I., and
chapel hill consensus conference nomenclature of vasculitides. Arthritis Roche, W.R. (1997). Immunolocalization of cytokines to mast cells in nor-
Rheum, 65, 1–11. mal and allergic conjunctiva. Clinical andExperimental Allergy, 27, 1328–34.
Jorizzo, J.L., Daniels, J.C., Apisarnthanarax, P., Gonzalez, E.B., and Cavallo, Maguire, H.C. Jr and Chase, M.W. (1967). Exaggerated delayed-type hyper-
T. (1983). Histamine-triggered localized vasculitis in patients with sensitivity to simple chemical allergens in the guinea pig. Journal of
seropositive rheumatoid arthritis. Journal of the American Academy of Investigative Dermatology, 49, 460–8.
Dermatology, 9, 845–51. Makker, S.P. and Tramontano, A. (2011). Idiopathic membranous nephropa-
Kaplan, A.P. and Greaves, M. (2009). Pathogenesis of chronic urticaria. thy: an autoimmune disease. Seminars in Nephrology, 31, 333–40.
Clinical andExperimental Allergy, 39, 777–87. Malaviya, R., Ikeda, T., Ross, E., and Abraham, S. N. (1996). Mast cell modu-
Karsten, C.M., Pandey, M.K., Figge, J., et al. (2012). Anti-inflammatory activ- lation of neutrophil influx and bacterial clearance at sites of infection
ity of IgG1 mediated by Fc galactosylation and association of FcγRIIB through TNF-alpha. Nature, 381, 77–80.
and dectin-1. Nature Medicine, 18, 1401–6. Manian, P. (1997). Genetics of asthma: a review. Chest, 112, 1397–408.
Kim, S., Iizuka, K., Aguila, H.L., Weissman, I.L., and Yokoyama, W.M. (2000). Marone, G., Casolaro, V., Patella, V., Florio, G., and Triggiani, M. (1997).
In vivo natural killer cell activities revealed by natural killer cell-deficient Molecular and cellular biology of mast cells and basophils. International
mice. Proceedings of the National Academy of Sciences (USA), 97, 2731–36. Archives of Allergy and Immunology, 114, 207–17.
Kruse, S., Japha, T., Tedner, M., Sparholt, S.H., Forster, J., Kuehr, J., and Merryman, C.F., Donoso, L.A., Sery, T.W., Sciutto, E., Bauer, A., and
Deichmann, K.A. (1999). The polymorphisms S503P and Q576R in the Shinohara, T. (1987). S-antigen. Adoptive transfer of experimental auto-
interleukin-4 receptor alpha gene are associated with atopy and influ- immune uveitis following immunization with a small synthetic peptide.
ence the signal transduction. Immunology, 96, 365–71. Archives of Ophthalmology, 105, 841–3.
Kuhn, R., Rajewsky, K., and Muller, W. (1991). Generation and analysis of Mielke, M.E., Peters, C., and Hahn, H. (1997). Cytokines in the induction
interleukin-4 deficient mice. Science, 254, 707–10. and expression of T-cell-mediated granuloma formation and protection
Kunkel, S.L., Lukacs, N.W., Strieter, R.M., and Chensue,S.W. (1998). Animal in the murine model of listeriosis. Immunological Reviews, 158, 79–93.
models of granulomatous inflammation. Seminars in Respiratory Moller, A., Lippert, U., Lessmann, D., Kolde, G., Hamann, K., Welker, P.,
Infections, 13, 221–8. Schadendorf, D., Rosenbach, T., Luger, T., and Czarnetzki, B.M. (1993).
Landsteiner, K. and Chase, M.W. (1939). Studies on the sensitization of ani- Human mast cells produce IL-8. Journal of Immunology, 151, 3261–6.
mals with simple chemical compounds: VI. Experiments on the sensi- Mosmann, T.R. (1992). T lymphocyte subsets, cytokines, and effector func-
tization of guinea pigs to poison ivy. Journal of Experimental Medicine, tions. Annals of the New York Academy of Sciences, 664, 89–92.
69,767–84. Mosmann, T.R., Cherwinski, H., Bond, M.W., Giedlin, M.A., and Coffman,
Landsteiner, K. and Chase, M.W. (1940). Studies on the sensitization of ani- R.L. (1986). Two types of murine helper T cell clone. I.  Definition
mals with simple compounds. VII. Skin sensitization by intraperitoneal according to profiles of lymphokine activities and secreted proteins.
injections. Journal of Experimental Medicine, 71, 237–45. Journal of Immunology, 136, 2348–57.
Landsteiner, K. and Jacobs, J. (1935). Studies on the sensitization of animals Mosmann, T.R., Sad, S., Krishnan, L., Wegmann, T.G., Guilbert, L.J., and
with simple chemical compounds. Journal of Experimental Medicine, 61, Belosevic, M. (1995). Differentiation of subsets of CD4+ and CD8+ T
643–56. cells. Ciba Foundation Symposium, 195, 42–54.
Lawrence, H.S. (1949). The cellular transfer of cutaneous hypersensitivity to Mote, J.R. and Jones, T.D. (1936). The development of foreign protein sensiti-
tuberculin in man. Proceedings of the Society for Experimental Biology zation in human beings. Journal of Immunology, 30, 149–67.
and Medicine, 71, 516–22. Mousli, M., Hugli, T.E., Landry, Y., and Bronner, C. (1994). Peptidergic
Li, X., Ptacek, T.S., Brown, E.E., and Edberg, J.C. (2009). Fcgamma recep- pathway in human skin and rat peritoneal mast cell activation.
tors: structure, function and role as genetic risk factors in SLE. Genes Immunopharmacology, 27, 1–11.
and Immunity, 10, 380–9. Movat, H.Z. (1979). The acute inflammatory reaction. In Inflammation,
Lichtenstein, L.M. and Bochner, B.S. (1991). The role of basophils in asthma. immunity, and hypersensitivity (2nd edn) (ed. H. Z. Movat), pp. 1–161.
Annals of the New York Academy of Sciences, 629, 48–61. Harper and Row, New York.
38 SECTION 2 basic science

Myers, L.K., Rosloniec, E.F., Cremer, M.A., and Kang, A.H. (1997). Reihsaus, E., Innis, M., MacIntyre, N., and Liggett, S.B. (1993). Mutations
Collagen-induced arthritis, an animal model of autoimmunity. Life in the gene encoding for the beta 2-adrenergic receptor in normal and
Sciences, 61,1861–78. asthmatic subjects. American Journal of Respiratory Cell and Molecular
Nakamura, A., Yuasa, T., Ujike, A., Ono, M., Nukiwa, T., Ravetch, J.V., Biology, 8, 334–9.
and Takai, T. (2000). Fcgamma receptor IIB-deficient mice develop Rosa-Rosa, L., Zimmermann, N., Bernstein, J.A., Rothenberg, M.E., and
Goodpasture’s syndrome upon immunization with type IV collagen: a Khurana Hershey, G.K. (1999). The R576 IL-4 receptor alpha allele cor-
novel murine model for autoimmune glomerular basement membrane relates with asthma severity. Journal of Allergy andClinical Immunology,
disease. Journal of Experimental Medicine, 191, 899–906. 104, 1008–14.
Nisitani, S., Murakami, M., and Honjo, T. (1997). Anti-red blood cell immuno- Rosenwasser, L.J. (1999). Promoter polymorphism in the candidate genes,
globulin transgenic mice. An experimental model of autoimmune hemo- IL-4, IL-9, TGF-beta1, for atopy and asthma. International Archives of
lytic anemia. Annals of the New York Academy of Sciences, 815, 246–52. Allergy and Immunology, 118, 268–70.
North, R.J., Dunn, P.L., and Conlan, J.W. (1997). Murine listeriosis as a model Rosenzweig, S.D. and Holland, S.M. (2005). Defects in the interferon-γ and
of antimicrobial defense. Immunological Reviews, 158, 27–36. interleukin-12 pathways. Immunological Reviews, 203, 38–47.
Ober, C., Leavitt, S.A., Tsalenko, A., Howard, T.D., Hoki, D.M., Daniel, R., Rothmeier, A.S. and Ruf, W. (2012). Protease-activated receptor 2 signaling
Newman, D.L., Wu, X., Parry, R., Lester, L.A., Solway, J., Blumenthal, in inflammation. Seminars in Immunopathology, 2012, 34, 133–49.
M., King, R.A., Xu, J., Meyers, D.A., Bleecker, E.R., and Cox, N.J. (2000). Russell, J.P. and Gibson, L.E. (2006). Primary cutaneous small vessel vas-
Variation in the interleukin 4-receptor alpha gene confers susceptibility culitis:  approach to diagnosis and treatment. International Journal of
to asthma and atopy in ethnically diverse populations. American Journal Dermatology, 45, 3–13.
of Human Genetics, 66, 517–26. Salvin, S.B. (1958). Occurrence of delayed hypersensitivity during the
Ohta, A., Sato, N., Yahata, T., Ohmi, Y., Santa, K., Sato, T., Tashiro, H., Habu, development of Arthus type hypersensitivity. Journal of Experimental
S., and Nishimura, T. (1997). Manipulation of Th1/Th2 balance in vivo Medicine, 107, 109–124.
by adoptive transfer of antigen-specific Th1 or Th2 cells. Journal of Sanarelli, G. (1924). De la pathogenie du cholera (neuvieme memoire). Le
Immunological Methods, 209, 85–92. Cholera Experimentale. Annal. Inst. Pasteur (Paris), 38:11–72.
Oiso, N., Fukai, K., and Ishii, M. (2000). Interleukin 4 receptor alpha chain Sato, K., Kinoshita, M., Motegi, A., Habu, Y., Takayama, E., Nonoyama, S.,
polymorphism Gln551Arg is associated with adult atopic dermatitis in Hiraide, H., and Seki, S. (2005). Critical role of the liver CD8+ CD122+
Japan. British Journal of Dermatology, 142, 1003–6. T cells in the generalized Shwartzman reaction of mice. European
Okayasu, I. (1985). Transfer of experimental autoimmune thyroiditis to nor- Journal of Immunology, 35, 593–602
mal syngeneic mice by injection of mouse thyroglobulin-sensitized T Schaub, B., Lauener, R., and von Mutius, E. (2006). The many faces of the hygiene
lymphocytes after activation with concanavalin A. Clinical Immunology hypothesis. Journal of Allergy andClinical Immunology, 117, 969–77.
andImmunopathology, 36,101–9. Schrijver, G., Bogman, M.J., Assmann, K.J., de Waal, R.M., Robben, H.C.,
Ozmen, L., Pericin, M., Hakimi, J., Chizzonite, R.A., Wysocka, M., Trinchieri, van, Gasteren, H., and Koene, R.A. (1990). Anti-GBM nephritis in
G., Gately, M., and Garotta, G. (1994). Interleukin 12, interferon γ, the mouse:  role of granulocytes in the heterologous phase. Kidney
and tumor necrosis factor α are the key cytokines of the generalized International, 38, 86–95.
Shwartzman reaction. Journal of Experimental Medicine, 180, 907–15. Sharon, R. and Naor, D. (1992). Experimental model of autoimmune hemolytic
Pappenheimer, A.M. and Freund, J. (1959). Induction of delayed hyper- anemia induced in mice with levodopa by intraperitoneal injection or oral
sensitivity to protein antigens. In Cellular and humoral aspects of the feeding. International Journal of Immunopharmacology, 14,1241–47.
hypersensitive states, (ed. H. S. Lawrence), pp. 67–88. Hoeber-Harper, Shushakova, N., Skokowa, J., Schulman, J., Bauman, U., Zwirner, J., Schmidt,
New York. R.E., and Gessner, J.E. (2002). C5a anaphylatoxin is a major regulator
Pettigrew, H.D., Teuber, S.S., and Gershwin, M.E. (2009). Clinical signifi- of activating versus inhibitory FcγRs in immune complex-induced lung
cance of complement deficiencies. Annals of the New York Academy of disease. Journal of Clinical Investigation, 110, 1823–30.
Sciences, 1173,108–23. Shwartzman, G. (1928). Studies on Bacillus typhosus toxic substances:  I.
Phair, J.P., Eisenfeld, A.J., Levine, R.J., and Kantor, F.S. (1970). Effects of Phenomenon of local skin reactivity to B.  typhosus culture filtrate.
pharmacological inhibition of histamine synthesis upon immunological Journal of Experimental Medicine,48, 247–68
reactions in guinea-pigs. Immunology, 18, 611–19. Shwartzman, G. (1937). Phenomenon of local tissue reactivity. Paul B. Hoeber,
Pirquet, C.V. and Schick, B. (1905). Die Serumkrankheit. Franz Deuticke, Inc., New York.
Leipzig (English translation by B. Schick (1951),Williams and Wilkins). Sireci, G., La Manna, M.P., Di Sano, C., Di Liberto, D., Porcelli, S.A., Kronenberg, M.,
Porter, H.M. (1957). Demonstration of delayed type reactivity in congenital Dieli, F., and Salerno, A. (2007). Pivotal advance: alpha-galactosylceramide
agammaglobulinemia. 2nd Tissue Homotransplantation Conference. induces protection against lipopolysaccharide-induced shock. Journal of
Annals of the New York Academy of Sciences, 64, 932. Leukocyte Biology, 81, 607–22.
Portier, P. and Richet, C. (1902). De l’action anaphylactique de certains ven- Smeltz, R.B. and Swanborg, R.H. (1998). Concordance and contradiction
ins. Comptes Rendus de la Societé de Biologie., 54, 170–2. concerning cytokines and chemokines in experimental demyelinating
Prausnitz, C. and Küstner, H. (1921). Studien über die Ueberempfindlichkeit. disease. Journal of Neuroscience Research, 51, 147–53.
Zentrabl Bakteriol Parasitenk, 86, 160–9. Smith, S.C. and Allen, P.M. (1993). The role of T cells in myosin-induced
Ranadive, N.S. and Movat, H.Z. (1979). Tissue injury and inflammation induced autoimmune myocarditis. Clinical Immunology andImmunopathology,
by immune complexes. In Inflammation, immunity and hypersensitivity 68, 100–6.
(2nd edn) (ed. H. Z. Movat), pp. 409–43. Harper and Row, New York. Song, Z., Casolaro, V., Chen, R., Georas, S.N., Monos, D., and Ono, S.J.
Ravetch, J. (2010). In vivo veritas:  the surprising roles of Fc receptors in (1996). Polymorphic nucleotides within the human IL-4 promoter that
immunity. Nature Immunology, 11, 183–5. mediate overexpression of the gene. Journal of Immunology, 156, 424–9.
Redrup, A.C., Howard, B.P., MacGlashan, D.W., Jr, Kagey-Sobotka, A., Stafforini, D.M., McIntyre, T.M., Zimmerman, G.A., and Prescott, S.M.
Lichtenstein, L. M., and Schroeder, J.T. (1998). Differential regulation of (2003). Platelet-activating factor, a pleiotrophic mediator of physiologi-
IL-4 and IL-13 secretion by human basophils: their relationship to histamine cal and pathological processes. Critical Reviews in Clinical Laboratory
release in mixed leukocyte cultures. Journal of Immunology, 160, 1957–64. Sciences, 40, 643–72.
Reed, C.E. and Kita, H. (2004). The role of protease activation of inflam- Stone K.D., Prussin C., Metcalfe D.D. (2010). IgE, mast cells, basophils, and
mation in allergic respiratory diseases. Journal of Allergy andClinical eosinophils. Journal of Allergy andClinical Immunology, 125 (2 Suppl.
Immunology, 114, 997–1008. 2), S73–80.
CHAPTER 3 hypersensitivity 39

Suzuki, Y., Shirato, I., Okumura, K., Ravetch, J.V., Takai, T., Tomino, Y., Wershil, B.K., Wang, Z.S., Gordon, J.R., and Galli, S.J. (1991). Recruitment
and Ra, C. (1998). Distinct contribution of Fc receptors and angioten- of neutrophils during IgE-dependent cutaneous late phase reactions
sin II-dependent pathways in anti-GBM glomerulonephritis. Kidney in the mouse is mast cell-dependent. Partial inhibition of the reaction
International, 54,1166–74. with antiserum against tumor necrosis factor-alpha. Journal of Clinical
Sylvestre, D.L. and Ravetch, J.V. (1996). A dominant role for mast cell Fc Investigation, 87, 446–53.
receptors in the Arthus reaction. Immunity, 5, 387–90. White, M. (1999). Mediators of inflammation and the inflammatory process.
Tang, W.W., Feng, L., Loskutoff, D.J., and Wilson, C.B. (2000). Glomerular Journal of Allergy andClinical Immunology, 103, S378–81.
extracellular matrix accumulation in experimental anti-GBM Ab glo- Wolff, H.H., Maciejewski, W., Scherer, R., and Braun-Falco, O. (1978).
merulonephritis. Nephron, 84, 40–8. Immunoelectronmicroscopic examination of early lesions in hista-
Tanimoto, Y., Takahashi, K., and Kimura, I. (1992). Effects of cytokines mine induced immune complex vasculitis in man. British Journal of
on human basophil chemotaxis. Clinical and Experimental Allergy, Dermatology, 99, 13–24.
22,1020–5. Xu, H., Rizzo, L.V., Silver, P.B., and Caspi, R.R. (1997). Uveitogenicity is asso-
Tarrant, T.K., Silver, P.B., Chan, C.C., Wiggert, B., and Caspi, R.R. (1998). ciated with a Th1-like lymphokine profile: cytokine-dependent modula-
Endogenous IL-12 is required for induction and expression of experi- tion of early and committed effector T cells in experimental autoimmune
mental autoimmune uveitis. Journal of Immunology, 161, 122–7. uveitis. Cellular Immunology, 178, 69–78.
Thomas, L. and Good, R.A. (1952). Studies on the generalized Shwartzman Yano, H., Wershil, B.K., Arizono, N., and Galli, S.J. (1989). Substance
reaction: I. General observations concerning the phenomenon. Journal P-induced augmentation of cutaneous vascular permeability and gran-
of Experimental Medicine, 96, 605–25. ulocyte infiltration in mice is mast cell dependent. Journal of Clinical
Trinchieri, G. (1996). Role of interleukin-12 in human Th1 response. Investigation, 84, 1276–86.
Chemical Immunology, 63, 14–29. Youdim, S., Stuntman, O., and Good, R.A. (1973). Thymus dependency of
Tsai, M., Grimbaldeston, M.A., Yu, M., Tam, S.Y., Galli, S.J. (2005). cells involved in transfer of delayed hypersensitivity to Listeria monocy-
Using mast cell knock-in mice to analyze the roles of mast cells in togenes in mice. Cellular Immunology, 8, 395–402.
allergic responses in vivo. Chemical Immunology and Allergy, 87, Youinou, P., le Dantec, C., Bendaoud, B., Renaudineau, Y., Pers, J.O., and
179–97. Jamin, C. (2005). Endothelium, a target for immune-mediated assault in
Tsai, M., Tam, S.Y., and Galli, S.J. (1993). Distinct patterns of early response connective tissue disease. Autoimmunity Reviews, 5, 222–8.
gene expression and proliferation in mouse mast cells stimulated by Zhang, Y., Ramos, B.F., Jakschik, B., Baganoff, M.P., Deppeler, C.L., Meyer,
Stem Cell Factor, Interleukin-3, or IgE and antigen. European Journal of D.M., Widomski, D.L., Fretland, D.J., and Bolanowski, M.A. (1995).
Immunology, 23, 867–72. Interleukin 8 and mast cell-generated tumor necrosis factor-alpha in
Vincent, A. (1994). Experimental autoimmune myasthenia gravis. In neutrophil recruitment. Inflammation, 19, 119–32.
Autoimmune Disease Models (ed. I.R. Cohen and A. Miller), pp. 83–106. Zhu, J., Mix, E., and Link, H. (1998). Cytokine production and the pathogen-
Academic Press, San Diego. esis of experimental autoimmune neuritis and Guillain-Barre syndrome.
Walport, M.J., Davies, K.A., and Botto, M. (1998). C1q and systemic lupus Journal of Neuroimmunology, 84, 40–52.
erythematosus. Immunobiology, 199, 265–85. Zhu, Z., Zheng, T., Homer, R.J., Kim, Y.K., Chen, N.Y., Cohn, L., Hamid, Q.,
Wedemeyer, J. and Galli, S.J. (2000). Mast cells and basophils in acquired and Elias, J.A. (2004). Acidic mammalian chitinase in asthmatic Th2
immunity. British Medical Bulletin, 56, 936–55. inflammation ad IL-13 pathway activation. Science, 304, 1678–82.
CHAPTER 4

Biology of endothelial cells
Zoltán Szekanecz, György Kerekes, and
Alisa E. Koch

Introduction and proteases and thus regulate inflammation in the surrounding


tissue. On the other hand, ECs are active responders to external
Endothelial cells (ECs) line the lumina of blood vessels and thus stimuli as they may become targets of inflammatory mediators pro-
they separate and connect the blood and the interstitial matrix of duced by leukocytes (Lum and Roebuck 2001; Szekanecz and Koch
the vessel wall. The endothelium is involved in numerous homeo- 2004; Savage 2009; Pober and Cotran 1990; Szekanecz and Koch
static mechanisms including the regulation of vascular tone, hae- 2005; Tesfamariam and DeFelice 2007; Widlansky et al. 2003; Blann
mostasis, as well as in pathological states, such as inflammation et al. 2005; Giannotti and Landmesser 2007).
(Szekanecz and Koch 2004; Szekanecz and Koch 2005; Tesfamariam ECs express a variety of CAMs that mediate leukocyte–endothe-
and DeFelice 2007; Szekanecz and Koch 2000). lial interactions (Figure  4.1). ECs are active players in leukocyte
ECs express a variety of cell surface molecules and synthesize tethering, rolling, firm adhesion, and migration (Szekanecz and
and release soluble mediators (Figure 4.1). In inflammatory states, Koch 2000; Springer 1990; Szekanecz and Koch 2004; Butcher 1991;
such as vasculitis, ECs interact with other cell types in this matrix Szekanecz et al. 1996; Buckley et al. 2005; Imhof and Aurrand-Lions
including leukocytes, fibroblasts, smooth muscle cells, and others. 2004; Tesfamariam and DeFelice 2007).
ECs are not only passive bystanders but they express numerous cell Vascular inflammation is associated with the perpetuation of new
adhesion molecules (CAMs), adhere to extracellular matrix (ECM) vessel formation from pre-existing vessels, termed angiogenesis, as
components, secrete numerous inflammatory mediators, such as well as with the impairment of capillary generation from endothe-
cytokines, nitric oxide (NO), prostaglandins, endothelin 1 (ET-1), lial progenitor cells (EPCs), termed vasculogenesis (Pakozdi et al.
2009; Szekanecz et al. 2009a; Szekanecz et al. 2010a; Koch 2000).
CAM Endothelial dysfunction has also been associated with inflam-
chemokine mation and inflammatory cytokine production (Lum and Roebuck
chemokine receptor
cytokine Monocyte/macrophage 2001; Zhang 2008; Widlansky et  al. 2003). This vascular impair-
cytokine receptor ment also precedes atherosclerosis (Kerekes et al. 2012; Szekanecz
T-cell
Smooth muscle cell and Koch 2008; Szekanecz et al. 2007; Shoenfeld et al. 2005; Kerekes
et al. 2008; Zhang 2008) (Table 4.1). Both endothelial dysfunction
and atherosclerosis have been detected in various types of vasculitis,
EXTRAVASCULAR
COMPARTMENT
such as granulomatosus with polyangiitis (de Leeuw et al. 2005) or

ACTIVATED Table 4.1 Factors and mechanisms involved in endothelial dysfunction


ENDOTHELIUM and injury underlying vasculitis inflammationa
soluble

INTRAVASCULAR chemokines Changes in endothelial VasodilationEndothelial cell contraction and


CAMs morphology retraction increased permeability (leakage)
COMPARTMENT
cytokines
Inflammatory cells Neutrophils, T- and B-cells, monocyte/
macrophages, smooth muscle cells
Fig. 4.1 Crosstalk between inflammatory cells and endothelial cells (ECs)
Autoantibodies AECA, ANCA
during vascular inflammation. Cell surface-bound adhesion molecules (CAMs)
mediate adhesion of T cells, monocyte/ macrophages, and smooth muscle Inflammatory mediators Cytokines, chemokines, prostaglandins,
cells in the extravascular compartment or in the circulation. All cells produce leukotrienes, NO, PAF, thrombin, histamine, ROI,
proinflammatory cytokines that bind to EC cytokine receptors during endothelial MMPs, endothelin 1, EDHF, ADMA
activation. ECs, as well as other cells, produce chemokines that bind to chemokine
receptors. These chemokines chemoattract leukocytes. These molecular Complement factors
interactions are involved in localized inflammation. Furthermore, there are high Cell adhesion molecules ICAM-1, VCAM-1, E-selectin
amounts of soluble CAMs, cytokines, and chemokines in the circulation which
will also trigger systemic inflammation in other compartments of the body. a See text for abbreviations
42 SECTION 2 basic science

Takayasu’s arteritis (Seyahi et  al. 2006). Endothelial dysfunc- cells, as well as soluble mediators (Gonzalez-Gay et  al. 2006;
tion, even subclinically, can be assessed by the ultrasound-based Szekanecz et al. 2007; Tesfamariam and DeFelice 2007; Varani et al.
flow-mediated vasodilation (FMD) technique (Kerekes et al. 2012; 1989) (Figure 4.1; Table 4.1).
Kerekes et al. 2008; Gonzalez-Gay et al. 2008). Leukocyte–EC interactions may themselves cause EC injury and
In this chapter, we briefly review those basic mechanisms that are thus increased vascular leakage. In brief, the key mediators in this
the most relevant for the pathogenesis of vascular inflammation. process are reactive oxygen intermediates (ROI) and matrix met-
Some aspects of EC morphology and pathophysiology, such as vas- alloproteinase (MMP) enzymes primarily produced by inflam-
cular tone, permeability, endothelial dysfunction, and injury, will matory leukocytes (Lum and Roebuck 2001; Varani et  al. 1989;
be briefly presented. The role of ECs in inflammation, leukocyte– Tesfamariam and DeFelice 2007; Szekanecz and Koch 2000; Kvietys
EC adhesion, transendothelial inflammatory cell migration, angio- and Granger 2012). The outcome of leukocyte-mediated vascu-
genesis and vasculogenesis will be discussed in more detail. Finally, lar injury highly depends on activated leukocytes. Non-activated
the role of the same mechanisms in the pathogenesis of vasculitides neutrophils extravasate without affecting endothelial permeability,
will be summarized. Although we will describe these mechanisms while transendothelial migration of activated leukocytes results
as distinct entities, there is certainly significant overlap between in vascular leakage (Tesfamariam and DeFelice 2007; Varani et al.
them and the outcome of vascular inflammation is highly depend- 1989). Neutrophil and ECs activation is associated with altered
ent on this complex inflammatory network (Figure 4.1). membrane morphology and cytoskeletal, as well as ECM reorgani-
zation and increased expression of endothelial CAMs, primarily that
Endothelial morphology of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhe-
and pathophysiology sion molecule 1 (VCAM-1), and E-selectin (ELAM-1). Abundant
CAM expression enhances neutrophil recruitment into inflam-
Changes in endothelial morphology and permeability matory sites (Pober and Cotran 1990; Szekanecz and Koch 2000;
The vascular endothelium undergoes morphological changes Szekanecz and Koch 2004; Szekanecz and Koch 2005; Tesfamariam
including vasodilatation and increased permeability (leakage) dur- and DeFelice 2007). Inhibition of neutrophil adhesion or inhibi-
ing vasculitis. Vasodilatation and vascular leakage may result from tion of the production of ROIs in animal models of inflammation
several mechanisms, including EC contraction and retraction, suppress vascular leakage and oedema formation in rats (Mulligan
leukocyte- or anti-EC antibody (AECA)-mediated vascular injury, et  al. 1991). Further details on neutrophil-mediated endothelial
and regeneration (Buckley et al. 2005; Savage 2009; Zhang 2008; injury and antineutrophil cytoplasmic antibodies (ANCA) will not
Szekanecz and Koch 2004) (Table 4.1). be presented in this chapter; we refer to other chapters in this vol-
ECs, as well as leukocytes, release vasodilatory mediators includ- ume, as well as other reviews (Kallenberg 2010; Kallenberg 2011)
ing prostacyclin (PGI2), nitric oxide (NO), and platelet-activating (Tables 4.1 and 4.2).
factor (PAF) (Brenner et  al. 1989; Szekanecz and Koch 2005; In addition to neutrophils, T-cell and B-cell mediated immune
Szekanecz and Koch 2004; Pober and Cotran 1990). Other media- responses also play a significant role in vascular injury (Figure 4.1,
tors, such as thrombin, histamine, and leukotriene C4, enhance the Table 4.1). Interactions of CD4+ and CD8+ T cells with the endothe-
synthesis of these EC-derived vasodilators (Jaffe et al. 1987; Zhang lium and the endothelial expression of MHC molecules have been
2008) (Table 4.1). reported in association with inflammation-induced vascular injury
The morphological basis for vascular leakage is EC retraction (Pober et al. 2001; Tesfamariam and DeFelice 2007). Although neu-
and contraction. These result in the formation and widening of trophils and ANCA had traditionally been the key players in the
intercellular gaps between ECs (Pober and Cotran 1990; Imhof pathogenesis of systemic vasculitis, the impairment of regulatory T
and Aurrand-Lions 2004). Factors triggering EC leakage include cells and the activation of Th17 cells have also been implicated in
histamine, serotonin, complement factors (C3a, C5a), bradykinin, vasculitis-associated vascular injury (Abdulahad et al. 2011).
leukotrienes, PAF, and AECA (Brenner et al. 1989; Joris et al. 1987; Various mediators produced by ECs themselves and other cells
Szekanecz and Koch 2004; Bodolay et al. 2004) (Table 4.1). lead to endothelial dysfunction and injury. ECs synthesize NO,
The gaps form as a result of EC contraction mediated by vas- prostaglandins, ET-1, endothelium-derived hyperpolarizing fac-
oactive agents, primarily histamine. EC retraction, associated tor (EDHF), and other substances that regulate vascular tone and
with cytoskeletal reorganization, occurs in EC monolayers upon permeability, as well as endothelial CAM expression (Giannotti
activation of proinflammatory cytokines, such as interleukin 1 and Landmesser 2007; Tesfamariam and DeFelice 2007; Widlansky
(IL-1), tumour necrosis factor α (TNF-α) or interferon γ (IFN-γ). et al. 2003; Szekanecz and Koch 2004; Szekanecz and Koch 2005;
Treatment of EC cultures with these cytokines for 4–6 hours Kvietys and Granger 2012; Feletou et  al. 2010)(Table  4.1). NO
results in cytoskeletal reorganization and EC retraction, leading exerts several functions including vasodilation, leukocyte adhe-
to sustained vascular permeability. Sustained EC retraction pri- sion, migration, angiogenesis, and vascular smooth muscle cell pro-
marily induced by cytokines is very different from the rapid and liferation (Kvietys and Granger 2012; Gunnett et al. 2005; Feletou
histamine-dependent EC contraction. EC retraction and contrac- et  al. 2010). During vascular inflammation, ECs and leukocytes
tion are examples of EC stimulation and EC activation, respectively produce high amounts of NO (Gunnett et al. 2005; Tesfamariam
(Zhang 2008; Szekanecz and Koch 2004; Szekanecz and Koch 2005; and DeFelice 2007). During endothelial injury, NO interacts with
Pober and Cotran 1990; Brett et al. 1989; Joris et al. 1987). ROI (Feletou et al. 2010; Tesfamariam and DeFelice 2007; Kvietys
and Granger 2012). ROI also induce endothelial dysfunction (Heo
Mechanisms of endothelial dysfunction and injury et al. 2012; Kvietys and Granger 2012). Asymmetric dimethylargi-
In vasculitis, accelerated atherosclerosis, and other states of vas- nine (ADMA) is a naturally produced amino acid that circulates in
cular inflammation, endothelial injury is caused by inflammatory the plasma. Increased ADMA production has been associated with
CHAPTER 4 biology of endothelial cells 43

Table 4.2 Relevant endothelial adhesion molecules in inflammationa and DeFelice 2007; Szekanecz and Koch 2000; Szekanecz and Koch
2004; Szekanecz and Koch 2005; Coll-Vinent et al. 1998).
Adhesion molecule Receptor on Ligand(s)
superfamily endothelium Endothelial regeneration after injury
Integrins β1 integrins ECM components
and prevention of damage
α4β1 integrin VCAM-1, fibronectin Vascular regeneration occurring after inflammatory injury is
linked to EPCs and vasculogenesis. In brief, differentiation and
αVβ3 integrin ECM components
activation of EPCs lead to vessel formation. (Yamahara and Itoh
Immunoglobulins ICAM-1 (ICAM-3?) β2 integrins 2009; Pakozdi et  al. 2009; Szekanecz et  al. 2010a; Szekanecz and
VCAM-1 α4β1 and α4β7 integrins Koch 2010b; Giannotti and Landmesser 2007). EPCs may be used
LFA-3 CD2 in therapy in order to stimulate vascular regeneration (Asahara
PECAM-1 (CD31) homotypic, αvβ3 integrin 2007; Isner et al. 2001; Szekanecz et al. 2010a). Elevated numbers
Selectins E-selectin ESL-1, PSGL-1, CLA
of circulating ECs and EPCs were detected in childhood vasculitis,
suggesting the perpetuation of vascular regeneration after injury
P-selectin PSGL-1
(Clarke et al. 2010). Recently, the role of adipose tissue in endothe-
Cadherins VE-cadherin homotypic lial regeneration has been proposed. Adipocyte progenitors share
Other CD44 hyaluronate numerous features with mesenchymal stem cells and they exert
endoglin TGF-β strong provasculogenic potential (Casteilla et al. 2010).
VAP-1 ? Endothelial regeneration may also occur without the formation
of new blood vessels. In this situation, regeneration is accompanied
JAMs homotypic
by increased capillary permeability due to open intercellular junc-
CD99 homotypic
tions and incomplete basement membranes (Joris et al. 1990; Joris
ESAM homotypic et al. 1987).
a See text for abbreviations Apart from naturally occurring processes leading to regenera-
tion, there have been several attempts to identify novel therapeu-
tic targets for preserving endothelium and for the prevention of
increased vascular tone, vascular inflammation and injury, stroke, endothelial injury. These molecules and synthetic compounds
cardiovascular disease, as well as inflammatory rheumatic diseases include traditionally used drugs, such as organic nitrates (ISDN
(Tesfamariam and DeFelice 2007; Kemeny-Beke et al. 2011; Zsuga and ISMN) that prevent endothelial injury by increasing NO pro-
2008; Feletou et al. 2010). ET-1 is a vasoconstrictor peptide pro- duction. Thrombomodulin is expressed on the EC surface. Loss of
duced by ECs. ET-1 activates neutrophil leukocyte adhesion and thrombomodulin would enhance vascular damage. In animal stud-
macrophage activation. ET-1 is also involved in ECM remodelling ies, overexpression of thrombomodulin achieved by gene transfer
and thus in vascular injury (Zouki et al. 1999; Tesfamariam and opened new horizons towards the protection of arterial endothe-
DeFelice 2007) (Table 4.1). lium. Other molecules not discussed in more detail include tissue
On the other hand, ECs respond to mediators primarily pro- factor, prostacyclin (PGI), adiponectin, sirtuins (Sirt-1), ligustra-
duced by inflammatory leukocytes, such as ROIs, proteases zine, and several others (Ramli et al. 2011).
and proinflammatory cytokines including, TNF-α, IL-1, and
IFN-γ (Tesfamariam and DeFelice 2007; Pober and Cotran 1990; Clinical assessment of endothelial dysfunction
Szekanecz and Koch 2000; Szekanecz and Koch 2004; Szekanecz Endothelial dysfunction is the earliest, usually reversible manifesta-
and Koch 2005; Lum and Roebuck 2001). The inflamed endothe- tion of vascular disease that precedes irreversible vascular damage.
lium is a major target of oxidative stress that also increases vas- There are a number of currently available tests that detect the func-
cular endothelial permeability, promotes leukocyte adhesion, and tional capacity of the endothelium; however, endothelial function
involves redox-regulated transcription factors such as activator is not a well-established parameter, its pathophysiological basis is
protein-1 and nuclear factor-κB (NFκB) (Lum and Roebuck 2001). partially understood, and there is no invasive, haemodinamically
Cytokines induce MHC class I antigen expression thus activating supported ‘gold standard’.
neutrophil accumulation and triggering an acute inflammatory Endothelial function can be appraised in vivo by direct or
cascade that eventually leads to vascular injury (Tesfamariam and indirect assessment of microcirculation after provocation by the
DeFelice 2007; Szekanecz and Koch 2000; Szekanecz and Koch application of acetylcholine, substance P, bradykinin, serotonin,
2004; Szekanecz and Koch 2005; Kallenberg 2010). or histamine, compounds able to induce endothelium-dependent
There is active interplay between ECs and many components of vasodilation. The degree of vasodilation depends on the bioavail-
the complement system. ECs may also secrete complement pro- ability of NO produced by the endothelial layer upon provocation
teins and activation products. EC–complement interaction is regu- (Poredos 2001; Kerekes et al. 2012). Among physical manoeuvres
lated by proinflammatory mediators. Unrestricted complement today, the induction of postocclusive reactive hyperaemia (PORH)
activation on the endothelial surface promotes endothelial injury by inflation off a pneumatic cuff around an extremity, most often
(Fischetti and Tedesco 2006) (Table 4.1). the forearm, has become the most commonly applied provocation
These mediators trigger endothelial dysfunction and vascular method due to its non-invasive nature. Suprasystolic pressure is
injury either by direct cytotoxicity, induction of an immune pro- applied, triggering vasodilation in the resistance vessels and rapid
cess, or by the stimulation of CAM expression. These mechanisms deflation of the cuff will result in intensive flow. The late, mostly
have been described in relation to systemic vasculitis (Tesfamariam NO-dependent phase, commonly detected over the brachial artery,
44 SECTION 2 basic science

mainly results from the high shear-induced secondary vasodila- by ECs, while L-selectin is mostly expressed by leukocytes (Patel
tion of the conduit arteries (Higashi et al. 2001; Kerekes et al. 2012; et al. 2002). During leukocyte transendothelial migration, selectins
Giannotti and Landmesser 2007). In clinical practice, PORH is mediate the initial tethering and rolling of leukocytes (Springer
commonly assessed as an increase in forearm blood flow (FBF), as 1990; Patel et  al. 2002; Butcher 1991). E-selectin is a marker
flow-mediated dilation of the afferent artery (FMD), or as increase for cytokine-induced endothelial activation (Patel et  al. 2002).
in skin microcirculatory flow detected by laser Doppler (Cracowski E-selectin ligand-1 (ESL-1) and P-selectin ligand-1 (PSGL-1) con-
et al. 2006; Celermajer et al. 1992; Kerekes et al. 2012; Giannotti and tain sialylated glycan motifs, such as sialyl Lewis-X (sLex) (Patel
Landmesser 2007). et al. 2002). P-selectin is constitutively present on the membrane
of endothelial Weibel–Palade bodies (Patel et al. 2002). P-selectin
is involved in the very early phases of leukocyte–endothelial adhe-
Leukocyte–endothelial adhesion sion (Butcher 1991). L-selectin serves as a lymphocyte homing
and transendothelial cell migration receptor, where it mediates the physiological recirculation of naïve
lymphocytes through specialized high-endothelial venules (HEV)
Endothelial adhesion molecules (Springer 1990; Patel et al. 2002). Selectins also play an important
Cell adhesion molecules (CAM) have been classified into integrin, role in leukocyte adhesion to endothelium during the inflamma-
selectin, immunoglobulin, and cadherin superfamilies (Agarwal tory process. There is increased expression of E-selectin on acti-
and Brenner 2006; Springer 1990; Szekanecz et  al. 1996; Muller vated ECs (Szekanecz et al. 1996).
2009) (Table 4.2). Other CAMs expressed by ECs include the CD44 antigen. It has
Integrins are αβ heterodimers. Each of the common β chains is a proteoglycan-like structure, it is a receptor for hyaluronate, and
associated with one or more α subunits (Albelda and Buck 1990; is expressed on activated ECs in inflammatory sites (Agarwal and
Springer 1990). Cell adhesion to the ECM is mostly mediated by Brenner 2006; Johnson et al. 1993; Halloran et al. 1996; Sarraj et al.
β1  and β3, while intercellular adhesion is facilitated through β1 2006; Szekanecz et al. 1996; Springer 1990). Vascular adhesion pro-
and β2 integrins (Springer 1990; Imhof and Aurrand-Lions 2004; tein 1 (VAP-1) was isolated from synovial ECs. VAP-1 is a marker of
Albelda and Buck 1990). β1 and β3 integrins are expressed on ECs, activated endothelium, as its expression is increased in inflamma-
while β2 integrins are leukocyte CAMs (Albelda and Buck 1990). tion (Salmi et al. 1993). Endoglin (CD105) has been identified as a
Among β1 integrins, the α4β1 heterodimer (very late antigen 4; receptor for transforming growth factor β1 (TGF-β1) and TGF-β3.
VLA-4) plays an important role in lymphocyte–endothelial inter- This molecule contains the RGD (arginine-glycine-aspartate)
actions. This molecule recognizes at least two alternative ligands, sequence, a motif found in a number of extracellular matrix ligands
such as vascular cell adhesion molecule 1 (VCAM-1), a member for integrins. Thus, endoglin may be involved in intercellular adhe-
of the immunoglobulin superfamily of CAMs, and the CS-1 region sion. Endoglin was detected on most inflamed and normal ECs
of fibronectin. VLA-4 is present on lymphocytes and monocytes, (Szekanecz et al. 1996; Szekanecz and Koch 2008). CD99 is a mole-
while VCAM-1, an endothelial marker, can be induced by cytokines cule of unique structure. Its function is similar to that of PECAM-1.
on a number of other cells (Szekanecz et al. 1996; Albelda and Buck Via homotypic adhesion, CD99 is involved in EC-EC adhesion at
1990; Springer 1990). endothelial junctions, as well as in leukocyte–endothelial interac-
The immunoglobulin superfamily of CAMs is a group tions (Muller 2009).
of transmembrane glycoproteins containing one or more Junctional cell adhesion molecules (JAMs) are also involved in
immunoglobulin-like motifs of 60–100 amino acids (Springer transendothelial migration of leukocytes. JAMs are concentrated at
1990; Albelda and Buck 1990; Muller 2009). VCAM-1 is constitu- EC borders. JAMs are normally engaged in haemophilic adhesion;
tively expressed on ECs; however, its expression is up-regulated by however, during inflammation, JAM-A can also bind to the LFA-1
proinflammatory cytokines (Springer 1990; Szekanecz et al. 1996). integrin. JAM-A and JAM-C have been implicated in adhesive
ICAM-1, the counter-receptor for the β2 integrins LFA-1 (αLβ2), events underlying inflammatory conditions (Rabquer et al. 2008;
Mac-1 (αMβ2), and αXβ2 is expressed on both endothelia and leu- Imhof and Aurrand-Lions 2004; Naik et  al. 2008; Muller 2009).
kocytes (Albelda and Buck 1990; Springer 1990; Szekanecz et al. EC-selective adhesion molecule (ESAM) is molecularly related to
1994; Szekanecz et al. 1996; Muller 2009). Among other ICAMs, JAMs; its cellular distribution is mostly limited to EC junctions.
ICAM-2 is constitutively expressed on endothelial and may not ESAM binds homotypically to another ESAM molecule on the
be an activation marker (Szekanecz et  al. 1994; Muller 2009). neighbouring cell (Muller 2009) (Table 4.2).
ICAM-3 is a leukocyte CAM; however, it is also present on some
ECs (Szekanecz et al. 1994). All three ICAMs bind to β2 integrins Leukocyte–endothelial adhesion
(Szekanecz et al. 1994; Szekanecz et al. 1996). Platelet-endothelial during inflammation
adhesion molecule 1 (PECAM-1; CD31) mediates homotypic Leukocyte extravasation into the synovium and into other inflam-
adhesion by binding to another PECAM-1 molecule, as well as matory sites is an active process mediated by a number of CAMs
heterotypic adhesion by recognizing the αVβ3 integrin (Szekanecz (reviewed in Jalkanen 1989; Albelda and Buck 1990; Springer 1990;
et  al. 1996; Springer 1990; Szekanecz et  al. 1995; Muller 2009). Haskard 1995; Szekanecz et al. 1996; Agarwal and Brenner 2006;
PECAM-1 is a marker of endothelial activation (Szekanecz et al. Muller 2009) (Figure 4.1). The inflammatory cascade of leukocyte
1995; Springer 1990). adhesion and migration begins with the adhesion of neutrophils
Selectins contain a lectin-like extracellular N-terminal domain, and mononuclear cells to endothelium (Ziff 1991; Butcher 1991;
an epidermal growth factor (EGF)-like motif, and two to nine moi- Szekanecz et al. 1996). Leukocytes transmigrate through the vessel
eties related to complement regulatory proteins (Albelda and Buck wall, and they then adhere to other cells in the ECM (Agarwal and
1990; Patel et al. 2002) (Table 4.1). E- and P-selectin are expressed Brenner 2006; Jalkanen 1989; Szekanecz et al. 1996; Muller 2009).
CHAPTER 4 biology of endothelial cells 45

Thus, leukocyte ingress into the inflamed synovium may be consid- Table 4.3 Relevant mediators and inhibitors of angiogenesisa
ered as pathological leukocyte ‘homing’ into the joints, which is a
temporally and sequentially well-regulated process. Mediators Inhibitors
The sequential regulation of the adhesion cascade is similar in
Chemokines CXCL1, CXCL5, CXCL7, CXCL4, CXCL9, CXCL10,
various types of inflamed tissues. The initial adhesion of leuko- CXCL8, CXCL12, CCL2, CCL21
cytes to endothelium (‘tethering’ and ‘rolling’) is mediated mainly CCL21, CCL23, CX3CL1
by selectins. This is followed by leukocyte activation, as well as
the up-regulation and conformational changes in the expression Matrix molecules Type I collagen, fibronectin, Thrombospondin, RGD
laminin, heparin, heparan sequence
of a number of CAMs, mainly leukocyte (β2) integrins, resulting
sulphate
in firm, more stable interactions. Several CAMs, including inte-
grins, PECAM-1 (CD31), CD99, and others, may be involved in Cell adhesion β1 and β3 integrins, RGD sequence (integrin
the transendothelial migration of leukocytes into the synovium; molecules E-selectin, P-selectin, ligand)
VCAM-1, endoglin,
however, some aspects of these molecular mechanisms are not fully
PECAM-1, VE-cadherin,
understood (Agarwal and Brenner 2006; Haskard 1995; Szekanecz JAMs
et al. 1996; Muller 2009).
The functional roles of CAMs, as well as the interactions of these Growth factors VEGF, bFGF, aFGF, PDGF, TGF-βb
EGF, IGF-I, HIF-1, TGF-βb
molecules with inflammatory mediators, have been investigated in
a number of experimental systems. Cell cultures of isolated ECs are Cytokines TNF-α, IL-6b, IL-15, IL-18 IL-4, IL-6b, IFN-α, IFN-γ
suitable for studies on CAM expression regulation by cytokines, Proteases MMPs, plasminogen TIMPs, plasminogen
while the relative importance of certain adhesion pathways can activators activator inhibitors
be determined by using in vitro adhesion assays. Animal models
Others Angiogenin, substance DMARDs, anti-TNF
are used to characterize all these mechanisms in vivo. Thus, leu- P, prolactin, thrombin, biologics, tocilizumab,
kocyte–endothelial adhesion mechanisms underlying vascular histamine, ROI, NO, angiostatin, endostatin,
inflammation can be studied in such experimental systems. For SAA, ET-1, COX-2, S1P inhibitors, ET-1
example, in EC cultures, ICAM-1 expression can be induced by prostaglandins, S1P antagonists, VEGF
IL-1β, TNF-α, and IFN-γ. IL-1β stimulates endothelial E-selectin targeting peptides
expression as well. Peripheral blood monocytes obtained from a See text for abbreviations.
rheumatoid arthritis (RA) patients adhere well to both resting and b Mediators with both pro- and antiangiogenic effects.
cytokine-stimulated synovial ECs (Szekanecz et al. 1996; Haskard
1995; Pober and Cotran 1990).
inflamed tissues (Taylor and Sivakumar 2005). The angiopoietin-1
(Ang1)/Tie2 complex interacts with VEGF during the stabilization
The endothelium in angiogenesis of newly formed blood vessels (Holash et  al. 1999). In contrast,
and vasculogenesis Ang2, an antagonist of Ang1, inhibits vessel maturation (Holash
et al. 1999; Shibuya 2008). Elevated VEGF and Ang2 levels were
Angiogenesis and its mediators detected in children with active vasculitis (Clarke et al. 2010).
Angiogenesis is the formation of new capillaries from pre-existing Apart from VEGF, other growth factors including fibroblast
blood vessels, while vasculogenesis is the outgrowth of vessels from growth factors (FGF-1 and -2), transforming growth factor β
endothelial progenitor cells (EPC) (Paleolog 2005; Folkman 1995; (TGF-β), connective tissue growth factor (CTGF), platelet-derived
Koch 2000; Szekanecz et al. 2009a; Szekanecz et al. 2010a; Szekanecz growth factor (PDGF), and others have been implicated in syno-
and Koch 2007b; Szekanecz and Koch 2008; Szekanecz and Koch vial angiogenesis (Szekanecz et  al. 2009b; Szekanecz and Koch
2010; Pakozdi et al. 2009; Veale and Fearon 2006). Angiogenesis 2008; Szekanecz and Koch 2007b). Placenta growth factor (PIGF),
may increase the total endothelial surface and thus may enable like VEGF, binds to the flt-1/VEGF-R1 receptor (Yoo et  al.
leukocyte extravasation into inflammatory sites (Szekanecz et al. 2009) (Table 4.3).
2009a; Szekanecz et al. 2010a; Szekanecz and Koch 2007b). The per- Proinflammatory cytokines may exert direct angiogenic
petuation of angiogenesis has been associated with inflammatory activity or may act indirectly via VEGF-dependent pathways
diseases, such as RA. The outcome of such ‘angiogenic diseases’ is (Brennan and Beech 2007; Szekanecz and Koch 2007b). Primarily
dependent on the balance or imbalance between angiogenic medi- TNF-α, IL-1, IL-6, IL-15, IL-17, IL-18, oncostatin M, monocyte
ators and angiostatic factors (Taylor and Sivakumar 2005; Veale migration inhibitory factor (MIF), granulocyte (G-CSF), and
and Fearon 2006; Szekanecz et al. 2009a; Szekanecz et al. 2010a; granulocyte-macrophage colony-stimulating factors (GM-CSF)
Szekanecz and Koch 2007b) (Table 4.3). have been implicated in inflammatory angiogenesis (Pakozdi
The hypoxia-vascular endothelial growth factor et  al. 2006; Szekanecz and Koch 2007b; Brennan and Beech
(VEGF)-angiopoietin system seems to be of outstanding impor- 2007) (Table 4.3).
tance in arthritis-associated angiogenesis (Taylor and Sivakumar Numerous chemokines and chemokine receptors have been
2005; Koch et al. 1994; Shibuya 2008). VEGF is induced by hypoxia implicated in inflammatory neovascularization. The angiogenic
and hypoxia-inducible factors 1 and 2 (HIF-1, -2) in RA (Taylor nature of most CXC chemokines has been associated with the
and Sivakumar 2005; Fearon and Veale 2007; Giatromanolaki et al. glutamyl-leucyl-arginyl (ELR) amino acid motif within their
2003; Biniecka et al. 2010; Ballara et al. 1999; Muz et al. 2009; Westra structure. The most relevant ELR-containing CXC chemokines
et al. 2010; Shibuya 2008). Significant hypoxia is characteristic of that mediate angiogenesis include CXCL1/groα, CXCL5/ENA-78,
46 SECTION 2 basic science

CXCL7/CTAP-III, and CXCL8/IL-8 (Szekanecz and Koch 2001; suppress angiogenic factors, among others, VEGF and/or Ang1/
Strieter et al. 1995; Koch et al. 2001). In contrast, the ELR-lacking Tie2 (Lainer-Carr and Brahn 2007; Szekanecz et al. 2009a; Veale
CXCL4/PF4, CXCL9/Míg, and CXCL10/IP-10 inhibit angiogen- and Fearon 2006) (Table 4.3).
esis. Some authors suggest that the effect of VEGF on ECs may be, Direct VEGF inhibitors already tried in cancer studies include
in part, mediated by CXCL10 (Szekanecz et al. 2009b). CXCL12/ antibodies to VEGF or VEGF receptors (VEGFR), soluble VEGFR
SDF-1 may play a significant role in inflammatory angiogenesis, constructs, small molecule VEGF and VEGFR inhibitors, or inhib-
vasculogenesis, and lymphoid neogenesis despite the fact that itors of VEGF and VEGFR signalling (Lainer-Carr and Brahn
this chemokine lacks the ELR motif (Petit et al. 2007). Among CC 2007; Kiselyov et  al. 2007; Shibuya 2008; Szekanecz et  al. 2009a;
chemokines, CCL2/MCP-1 may induce EC chemotaxis in vitro Szekanecz and Koch 2008; Veale and Fearon 2006). HIFs may also
and angiogenesis in vivo. CCL23 has been implicated in the migra- be targeted. For example, YC-1, a superoxide-sensitive stimulator
tion of vascular ECs and angiogenesis-associated matrix metallo- of soluble guanylyl cyclase and a HIF-1 inhibitor could potentially
proteinase (MMP) production. CX3CL1/fractalkine is involved in be used to suppress inflammatory angiogenesis (Muz et al. 2009;
both angiogenesis and atherosclerosis underlying inflammatory Lainer-Carr and Brahn 2007). As far as the Ang1/Tie2 system is
rheumatic diseases including vasculitides (Szekanecz et  al. 2011; concerned, a soluble Tie2 receptor transcript delivered via an
Szekanecz et  al. 2010b; Kasama et  al. 2010). Among chemokine adenoviral vector to mice attenuated the incidence and severity of
receptors, CXCR2 may be the most relevant one to ELR-containing arthritis (Lainer-Carr and Brahn 2007; Szekanecz et al. 2009a). An
angiogenic CXC chemokines, such as CXCL1, CXCL5 and CXCL8 Ang2-targeting peptide has been genetically fused to the anti-TNF
on ECs. CCR2–CCL2 and CCR7–CCL21 interactions are also antibody adalimumab. This novel bispecific antibody enhanced
involved in inflammatory neovascularization (Szekanecz et  al. anti-TNF efficacy (Yang et al. 2011) (Table 4.3).
2011; Szekanecz et al. 2010b) (Table 4.3). Anticytokine therapy currently used to treat autoimmune-
Several ECM components, CAMs, and proteases have been inflammatory rheumatic diseases may also influence angiogenesis.
implicated in adhesive and angiogenic processes in inflammation. For example, TNF-α blockade by infliximab reduced VEGF expres-
Type I  collagen, fibronectin, laminin, vitronectin, tenascin, and sion and vascularity within the arthritic synovium. Infliximab also
proteoglycans mediate, while thrombospondin 1 (TSP-1) inhib- reduced synovial Ang1 and Tie2 expression. The anti-IL-6 receptor
its EC adhesion and neovascularization. Most β1 and β3 integrins, antibody tocilizumab also decreased VEGF production (Brennan
E-selectin, the L-selectin ligand CD34, selectin-related glycocon- and Beech 2007; Szekanecz et al. 2009a; Shibuya 2008).
jugates including Lewisy/H and MUC18, VCAM-1, PECAM-1 Proteases play a crucial role in the regulation of angiogenesis.
(CD31), endoglin, and JAMs (JAM-A and -C) are expressed on Numerous protease inhibitors, including tissue inhibitors of metal-
the endothelial surface and promote angiogenesis. The αVβ3 inte- loproteinases (TIMPs) and plasminogen activator inhibitors (PAIs)
grin seems to be of significant importance. Focal adhesion kinases that antagonize the effects of proteases, have been tried in angi-
(FAK) are involved in αVβ3 integrin signalling. Other angiogenic ogenesis models (Szekanecz et al. 2009a; Lainer-Carr and Brahn
factors, such as chemokines may act via integrin-dependent path- 2007; Skotnicki et al. 1999).
ways. Some MMPs, ADAM (A dysintegrin and metalloproteinase), Antibiotic derivatives, including minocycline, fumagillin ana-
and ADAMTS (ADAM with thrombospondin epitope) proteases logues, deoxyspergualin, roxithromycin, and clarithromycin, also
digest the ECM, release growth factors and other angiogenic medi- inhibit the release of VEGF and other angiogenic mediators and
ators and thus promote inflammatory neovascularization (Agarwal thus neovascularization. Synthetic fumagillin derivatives, TNP-470
and Brenner 2006; Szekanecz et al. 1996; Szekanecz et al. 2009a; and PPI-2458 inhibit VEGF and VEGF-dependent angiogenesis
Naik et al. 2008; Madri and Williams 1983; Infusino and Jacobson (Lainer-Carr and Brahn 2007; Koch 2000; Veale and Fearon 2006;
2012; Folkman 1995) (Tables 4.2 and 4.3). Szekanecz et al. 2009a).
Further angiogenic factors implicated in inflammation include, Among other inhibitors of angiogenic mediators, ET-1 antago-
without mentioning further details, NO, ROI, serum amyloid nists currently used in the treatment of pulmonary hypertension
A (SAA), endothelin 1 (ET-1), members of the cyclooxygenase-2 and scleroderma may also exert antiangiogenic effects (Koch and
(COX-2)-prostaglandin E 2 network, angiogenin, angiotro- Distler 2007). The microtubule destabilizer paclitaxel (taxol), used
pin, pleiotrophin, platelet-activating factor (PAF), substance P, in cancer therapy, also acts via HIF-1α blockade. S1P in also angi-
erythropoietin, adenosine, histamine, prolactin, thrombin, and ogenic and S1P inhibitors, such as fingolimod (FTY720), are in
sphingosine-1-phosphate (S1P) (Mullan et al. 2006; Szekanecz and clinical trials in various autoimmune diseases. FTY720 also acts as
Koch 2007b; Szekanecz and Koch 2007a; Szekanecz and Koch 2008; an inhibitor of FAKs (Lainer-Carr and Brahn 2007; Infusino and
Szekanecz et al. 2009a; Kvietys and Granger 2012) (Table 4.3). Jacobson 2012; Veale and Fearon 2006; Szekanecz et  al. 2009a).
Very recently, peptides have been isolated from libraries of phages
Angiogenesis inhibition preferentially homing to inflamed joints of rats. Some of these pep-
Angiogenesis can be inhibited by either exogenously blocking the tides that bound to ECs were able to inhibit VEGF signalling and
action of angiogenic mediators or by using endogenously pro- angiogenesis (Yang et al. 2011) (Table 4.3).
duced angiostatic compounds (Kiselyov et  al. 2007; Lainer-Carr An emerging number of compounds in traditional Chinese and
and Brahn 2007; Shibuya 2008; Szekanecz et al. 2009a; Veale and Korean medicine have been implicated in angiogenesis research.
Fearon 2006). The most commonly used antirheumatic agents, Among these compounds, scopolin, a coumarin derivative found
such as rofecoxib, dexamethasone, chloroquine, sulfasalazine, in Erycibe obtusifolia Benth; celastrol, an active ingredient of
methotrexate, azathioprine, cyclophosphamide, leflunomide, tha- Tripterygium wilfordii, also known as ‘thunder god vine’; or fise-
lidomide, tacrolimus, minocycline, and anti-TNF biologics, apart tin, the active ingredient of Rhus verniciflua Stokes, may also have
from other important anti-inflammatory effects, non-specifically angiostatic effects (Szekanecz et al. 2009a).
CHAPTER 4 biology of endothelial cells 47

Apart from angiogenesis inhibition, endogenously produced in the pathogenesis of vascular injury underlying autoimmune
angiostatic compounds also block the action of angiogenic media- vasculitis.
tors, such as VEGF, HIFs, or the αVβ3 integrin, and thus they indi-
rectly suppress neovascularization. Some of these compounds have Endothelial injury and dysfunction in vasculitis
already been tried to target tumour- or inflammation-associated Various infectious agents, irradiation, and drugs (e.g. anti-
neovascularization. Angiostatic cytokines include interferon-α biotics, hydralazine, interferons, monoclonal antibody bio-
(IFN-α), IFN-γ, IL-4, IL-12, IL-13, and leukaemia inhibitory fac- logics, colony-stimulating factors) can induce inflammatory
tor (LIF). These cytokines inhibit the release of VEGF and other vascular damage. The most relevant mechanisms involved in
angiogenic mediators. Angiostatic CXC chemokines that lack the vascular injury underlying vasculitis are shear stress, immune
ELR motif include CXCL4 (PF4), CXCL9 (Mig), and CXCL10 complexes, and autoantibodies, such as AECA (Tesfamariam and
(IP-10) (Szekanecz et al. 2009b). CCL21 (SLC) is an angiostatic CC DeFelice 2007).
chemokine that inhibits tumour progression. Angiostatin, a fragment Agents causing prolonged vasodilation and increase in blood flow
of plasminogen, and endostatin, a fragment of type XIII collagen, will induce shear stress. Increased shear stress results in the release
and their derivatives block αVβ3 integrin-dependent angiogenesis. of shear-responsive mediators, such as NO, other NO-dependent
Type IV collagen derivatives, including arresten, canstatin, and tum- mediators, chemokines, cytokines, and growth factors. Most vaso-
statin, also inhibit neovascularization. 2-methoxyestradiol (2-ME), active drugs are able to stimulate blood flow and thus shear stress,
a natural metabolite of oestrogen, inhibits angiogenesis by disrupt- eventually leading to the development of vasculitis (Tesfamariam
ing microtubules and by suppressing HIF-1α activity (Szekanecz and DeFelice 2007; Resnick et  al. 2003; Lu and Kassab 2011).
et al. 1998; Brennan and Beech 2007; Lainer-Carr and Brahn 2007; Flow-mediated shear stress also induces ROI production and thus
Szekanecz et al. 2009b; Veale and Fearon 2006) (Table 4.3). leads to endothelial dysfunction (Heo et al. 2012).
Foreign antigens activate the complement system leading
Vasculogenesis in inflammatory conditions to the formation of immune complexes. Both circulating and
EPCs are haematopoietic stem cells expressing, among other anti- tissue-bound immune complexes can trigger the development of
gens, CD34, CD133, type 2 VEGF receptor (VEGFR-2 or Flk-1), inflammatory lesions and damage the endothelium (Tesfamariam
and the CXCR4 chemokine receptor. During vasculogenesis, EPCs and DeFelice 2007; Sibelius et  al. 1998; Chen and Kallenberg
differentiate into mature ECs. Vasculogenesis is involved in tissue 2010). Immune complex-mediated endothelial injury, also impli-
development, vascular repair, and atherosclerosis, as well as inflam- cated in vasculitis, involves neutrophils, macrophages, T cells, and
mation (Szekanecz and Koch 2010; Pakozdi et al. 2009; Szekanecz mast cells. The process is associated with plasma extravasation,
et al. 2010a; Peichev et al. 2000; Paleolog 2005; Freedman and Isner increased permeability, the recruitment of leukocytes, and haem-
2002; Isner et al. 2001). orrhagic tissue necrosis (Sibelius et al. 1998; Chen and Kallenberg
Several groups have described defective vasculogenesis related 2010). Complement disrupts EC membrane integrity leading to
to impaired EPC numbers and functions in RA and scleroderma endothelial detachment (Fischetti and Tedesco 2006; Sibelius
(Szekanecz and Koch 2008; Pakozdi et al. 2009; Grisar et al. 2007; et al. 1998; Tesfamariam and DeFelice 2007; Chen and Kallenberg
Herbrig et al. 2006; Kuwana et al. 2004; Allanore et al. 2007; Jodon 2010). Circulating immune complexes activate leukocytes via their
de Villeroche et al. 2011). Impaired vasculogenesis has been asso- Fc receptors and also lead to increased vascular permeability and
ciated with increased cardiovascular morbidity and mortality in widening of endothelial junctions followed by immune-mediated
these disease states (Szekanecz and Koch 2008; Freedman and tissue injury (Tesfamariam and DeFelice 2007). Iimpaired regula-
Isner 2001; Kuwana et al. 2004). Effective control of inflammation tory T-cell function and proinflammatory cytokines, such as IL-17
using corticosteroids and anti-TNF agents may stimulate EPCs and are involved in vasculitis-associated endothelial injury (Abdulahad
thus may restore defective vasculogenesis (Ablin et al. 2006; Grisar et al. 2011) (Table 4.1).
et al. 2007). In addition, the induction of vasculogenesis may be AECAs have been detected in several autoimmune and
beneficial for patients with cardiovascular disease (Freedman and inflammatory diseases (Westphal et  al. 1994; Bodolay et  al.
Isner 2001) and the stimulation of EPCs and vasculogenesis may 2004; Tesfamariam and DeFelice 2007; Kollmeier et  al. 1998).
also suppress premature atherosclerosis in RA (Szekanecz and The AECA family is a heterogenous group of antibodies
Koch 2008). directed against various endothelial antigenic membrane pro-
Corticosteroids and anti-TNF agents may stimulate EPCs and teins (Kollmeier et al. 1998). AECAs may upregulate endothelial
thus restore impaired vasculogenesis in inflammatory conditions, CAM expression, and stimulate the release of chemokines and
such as RA. The CXCL12 (SDF-1) inhibitor bicyclam (AMD3100) cytokines, thus recruiting leukocytes and subsequently causing
mobilized EPCs in mice. The suppression of systemic inflammation cell apoptosis, necrosis, and endothelial injury (Kollmeier et al.
and disease activity by any means would improve vasculogenesis 1998; Drouet et al. 2003). AECAs act via complement-dependent,
(Pakozdi et al. 2009; Szekanecz et al. 2010a). as well as antibody-dependent cytotoxicity (Kollmeier et  al.
1998). In autoimmune diseases, AECAs have been implicated in
vascular injury and AECA has also been correlated with some
Endothelial activation and injury, biomarkers of clinical activity in these disorders (Westphal
adhesion, angiogenesis, and et al. 1994; Bodolay et al. 2004; Kollmeier et al. 1998). AECAs
have been detected more frequently in the sera of patients with
vasculogenesis in vasculitis rheumatoid vasculitis in comparison to RA without vasculitis
We will now briefly summarize data indicating that most of the (Westphal et al. 1994; van der Zee et al. 1991). Circulating cyto-
complex mechanisms described in this chapter are also involved toxic AECAs that reacted with ECs were described in a variety
48 SECTION 2 basic science

of vasculitides, such as in Kawasaki disease and granulomato- Abnormal angiogenesis and defective
sis with polyangiitis (Savage 2009; Savage and Williams 2007). vasculogenesis in vasculitides
In systemic vasculitis, AECAs may trigger endothelial CAM
VEGF and other angiogenic factors, including chemokines and
expression, induce calcium flux, stimulate proinflammatory
cytokines, have also been implicated in the pathogenesis of sys-
cytokine and chemokine release, activate intracellular signal-
temic vasculitides. In vasculitis, angiogenesis is also aggravated
ling cascades, such as JNK, and induce EC apoptosis (Karasawa
in response to hypoxia (Mitchell and Cestari 2009; Maruotti et al.
et  al. 2011; Baguley and Hughes 1988; Savage and Williams
2008). Elevated VEGF and Ang1 levels were detected in some
2007; Holmen et al. 2007; Jamin et al. 2005; Savage 2009). Thus,
vasculitides and Behçet’s disease (Kamoun et al. 2008; Mitchell
AECAs may be involved in the evolution in vasculitis-associated
and Cestari 2009; Choe et al. 2010). Among MMPs involved in
endothelial injury and these autoantibodies may be markers of
inflammatory angiogenesis, serum levels of MMP-3 have been
endothelial damage (Table 4.1).
associated with disease activity, as well as with C-reactive pro-
There have been numerous studies assessing endothelial function
tein (CRP) in ANCA-associated vasculitides (Monach et  al.
in various rheumatic diseases, such as RA, lupus, or scleroderma
2012) (Table 4.3).
(Kerekes et al. 2008; Gonzalez-Gay et al. 2004; Gonzalez-Juanatey
Impaired vasculogenesis based on EPC dysfunction has been
et  al. 2003; Vaudo et  al. 2004; Szucs et  al. 2007; Shoenfeld et  al.
reported in various types of vasculitis with kidney involvement.
2005). Endothelial dysfunction indicated by impaired FMD has
Patients with active disease had decreased numbers of progenitor
been reported in various types of primary systemic vasculitides,
cell colony-forming units, which was associated with high expres-
including polyarteritis nodosa, granulomatosis with polyangiitis,
sion of VAP-1 on kidney ECs. VAP-1 is a marker of inflamma-
eosinophilic granulomatosis with polyangiitis, and Kawasaki’s syn-
tory ECs. Thus, endothelial progenitor cell dysfunction is highly
drome, as well as in vasculitides secondary to RA or lupus (Raza
dependent on activated, inflammatory EC-mediated vascular dam-
et al. 2000; Bacon 2005; Bijl 2003; Filer et al. 2003; Espinola-Zavaleta
age (Choe et al. 2010; Holmen et al. 2005; Maruotti et al. 2008). Low
et  al. 2009; Buckley et  al. 2005). Anti-TNF agents may restore
levels of circulating EPCs and antivasculogenic capacity of serum
endothelial dysfunction in vasculitis. This also supports the role of
have been described in thromboangiitis obliterans (Hewing et al.
TNF-α in the initiation and progression of endothelial dysfunction.
2012). There has been a limited number of trials on therapeutic
TNF-α-mediated endothelial dysfunction may also be involved in
angiogenesis in systemic vasculitis patients (Kawanaka et al. 2009).
the progression of vasculitis. Thus, early treatment of vasculitis may
be able to reverse endothelial dysfunction (Raza et al. 2000; Buckley
et al. 2005; Bacon 2005). Summary
Adhesion molecules in vasculitis ECs are active participants in vascular inflammation. Vascular tone
and permeability are altered under inflammatory conditions. The
It has been shown that CAMs play a role in leukocyte–EC interac- ingress of inflammatory cells and the release of several mediators
tions underlying vasculitides associated with other autoimmune will result in endothelial injury. Leukocyte adhesion to ECs and
syndromes (Pankhurst et  al. 2009). ANCA may play an impor- leukocyte transendothelial migration are mediated by numerous
tant role in the regulation of CAM expression as it may induce CAMs. Early endothelial dysfunction may be detected by ultra-
β2 integrin expression on neutrophils leading to the perpetuation sound based techniques, such as FMD. Abundant production of
of neutrophil influx in ANCA-associated vasculitis (Calderwood angiogenic mediators but impaired vasculogenesis have also been
et  al. 2005). ICAM-1 and VCAM-1 have been associated with associated with inflammatory diseases. Most of these pathological
the development of vasculitis in Sjögren’s syndrome (Turkcapar events have been implicated in the pathogenesis of vasculitides.
et  al. 2005). VCAM-1 is involved in the pathogenesis of mixed
cryoglobulinaemic vasculitis (Kaplanski et  al. 2005). Regarding
soluble CAMs, there is an increased production of circulating References
ICAM, VCAM-1, and E-selectin in ANCA-associated vasculitis Abdulahad, W.H., Lamprecht, P., and Kallenberg, C.G. (2011). T-helper cells
(Schneeweis et  al. 2010; Monach et  al. 2012; Visvanathan et  al. as new players in ANCA-associated vasculitides. Arthritis Research and
2011). In rheumatoid vasculitis, increased concentrations of Therapy, 13, 236.
Ablin, J.N., Boguslavski, V., Aloush, V., Elkayam, O., Paran, D., Caspi, D.,
serum soluble ICAM-1 and ICAM-3 were found in comparison to
and George, J. (2006). Effect of anti-TNFalpha treatment on circulat-
RA without vasculitis. Furthermore, when comparing the active ing endothelial progenitor cells (EPCs) in rheumatoid arthritis. Life
and inactive phase of rheumatoid vasculitis, soluble ICAM-3 lev- Sciences, 79, 2364–9.
els were significantly higher in the active disease (Voskuyl et al. Agarwal, S.K., and Brenner, M.B. (2006). Role of adhesion molecules in syno-
1995). Serum levels of various soluble endothelial CAMs, includ- vial inflammation. Current Opinion in Rheumatology, 18, 268–76.
ing VCAM-1 and ICAM-1, have been associated with disease Albelda, S.M. and Buck, C.A. (1990). Integrins and other cell adhesion mol-
activity in systemic vasculitis (Monach et  al. 2012; Schneeweis ecules. FASEB Journal, 4, 2868–80.
et  al. 2010). Serum soluble ICAM-1 levels were higher close to Allanore, Y., Batteux, F., Avouac, J., Assous, N., Weill, B., and Kahan, A.
relapse in infliximab-treated vasculitis patients (Visvanathan et al. (2007). Levels of circulating endothelial progenitor cells in systemic scle-
rosis. Clinical and Experimental Rheumatology, 25, 60–6.
2011). Increased production of soluble ICAM-1 has been associ-
Asahara, T. (2007). Cell therapy and gene therapy using endothelial pro-
ated with more intensive vascular changes using nailfold capil-
genitor cells for vascular regeneration. Handbook of Experimental
laroscopy in rheumatoid vasculitis (Witkowska et al. 2003). These Pharmacology, 180, 181–94.
data suggest that CAM expression and soluble CAM production Bacon, P.A. (2005). Endothelial cell dysfunction in systemic vasculi-
may be useful markers of vascular inflammation in patients with tis:  new developments and therapeutic prospects. Current Opinion in
vasculitis (Figure 4.1, Table 4.2). Rheumatology, 17, 49–55.
CHAPTER 4 biology of endothelial cells 49

Baguley, E. and Hughes, G.R. (1988). Lytic IgG anti-endothelial cell antibod- Detection of antiendothelial cell antibodies by an enzyme-linked
ies in vasculitis. Lancet, 2, 907. immunosorbent assay using antigens from cell lysate: minimal interfer-
Ballara, S.C., Miotla, J.M., and Paleolog, E.M. (1999). New vessels, new ence with antinuclear antibodies and rheumatoid factors. Clinical and
approaches: angiogenesis as a therapeutic target in musculoskeletal dis- Diagnostic Laboratory Immunology, 10, 934–9.
orders. International Journal of Experimental Pathology, 80, 235–50. Espinola-Zavaleta, N., Soto-Lopez, M.E., Carreon-Torres, E., Gamboa, R.,
Bijl, M. (2003). Endothelial activation, endothelial dysfunction and pre- Mejia, A.M., Marquez-Velasco, R., Bojalil, R., Huesca-Gomez, C., Reyes,
mature atherosclerosis in systemic autoimmune diseases. Netherlands P.A., and Perez-Mendez, O. (2009). Altered flow-mediated vasodilata-
Journal of Medicine, 61, 273–7. tion, low paraoxonase-1 activity, and abnormal high-density lipopro-
Biniecka, M., Kennedy, A., Fearon, U., Ng, C.T., Veale, D.J., and O’Sullivan, tein subclass distribution in Takayasu’s arteritis. Circulation Journal,
J.N. (2010). Oxidative damage in synovial tissue is associated with in 73, 760–6.
vivo hypoxic status in the arthritic joint. Annals of the Rheumatic Fearon, U. and Veale, D.J. (2007). Angiogenesis in arthritis: methodological
Diseases, 69, 1172–8. and analytical details. Methods in Molecular Medicine, 135, 343–57.
Blann, A.D., Woywodt, A., Bertolini, F., Bull, T. M., Buyon, J. P., Clancy, R.M., Feletou, M., Kohler, R., and Vanhoutte, P.M. (2010). Endothelium-derived
Haubitz, M., Hebbel, R.P., Lip, G.Y., Mancuso, P., Sampol, J., Solovey, A., vasoactive factors and hypertension: possible roles in pathogenesis and
and Dignat-George, F. (2005). Circulating endothelial cells. Biomarker as treatment targets. Current Hypertension Reports, 12, 267–75.
of vascular disease. Thrombosis and Haemostasis, 93, 228–35. Filer, A.D., Gardner-Medwin, J.M., Thambyrajah, J., Raza, K., Carruthers,
Bodolay, E., Csipo, I., Gal, I., Sipka, S., Gyimesi, E., Szekanecz, Z., and D.M., Stevens, R.J., Liu, L., Lowe, S.E., Townend, J.N., and Bacon, P.A.
Szegedi, G. (2004). Anti-endothelial cell antibodies in mixed connec- (2003). Diffuse endothelial dysfunction is common to ANCA associated
tive tissue disease: frequency and association with clinical symptoms. systemic vasculitis and polyarteritis nodosa. Annals of the Rheumatic
Clinical and Experimental Rheumatology, 22, 409–15. Diseases, 62, 162–7.
Brennan, F., and Beech, J. (2007). Update on cytokines in rheumatoid arthri- Fischetti, F. and Tedesco, F. (2006). Cross-talk between the complement sys-
tis. Current Opinion in Rheumatology, 19, 296–301. tem and endothelial cells in physiologic conditions and in vascular dis-
Brenner, B.M., Troy, J.L., and Ballermann, B.J. (1989). Endothelium-dependent eases. Autoimmunity, 39, 417–28.
vascular responses. Mediators and mechanisms. Journal of Clinical Folkman, J. (1995). Angiogenesis in cancer, vascular, rheumatoid and other
Investigation, 84, 1373–8. disease. Nature Medicine, 1, 27–31.
Brett, J., Gerlach, H., Nawroth, P., Steinberg, S., Godman, G., and Stern, Freedman, S.B. and Isner, J.M. (2001). Therapeutic angiogenesis for ischemic
D. (1989). Tumor necrosis factor/cachectin increases permeability of cardiovascular disease. Journal of Molecular and Cellular Cardiology, 33,
endothelial cell monolayers by a mechanism involving regulatory G 379–93.
proteins. Journal of Experimental Medicine, 169, 1977–91. Freedman, S.B. and Isner, J.M. (2002). Therapeutic angiogenesis for coronary
Buckley, C.D., Rainger, G.E., Nash, G.B., and Raza, K. (2005). Endothelial artery disease. Annals of Internal Medicine, 136, 54–71.
cells, fibroblasts and vasculitis. Rheumatology (Oxford), 44, 860–3. Giannotti, G. and Landmesser, U. (2007). Endothelial dysfunction as an early
Butcher, E.C. (1991). Leukocyte-endothelial cell recognition: three (or more) sign of atherosclerosis. Herz, 32, 568–72.
steps to specificity and diversity. Cell, 67, 1033–6. Giatromanolaki, A., Sivridis, E., Maltezos, E., Athanassou, N., Papazoglou, D.,
Calderwood, J.W., Williams, J.M., Morgan, M.D., Nash, G.B., and Savage, C.O. Gatter, K. C., Harris, A.L., and Koukourakis, M.I. (2003). Upregulated
(2005). ANCA induces beta2 integrin and CXC chemokine-dependent hypoxia inducible factor-1alpha and -2alpha pathway in rheumatoid
neutrophil-endothelial cell interactions that mimic those of highly arthritis and osteoarthritis. Arthritis Research and Therapy, 5, R193–201.
cytokine-activated endothelium. Journal of Leukocyte Biology, 77, 33–43. Gonzalez-Gay, M.A., Gonzalez-Juanatey, C., and Martin, J. (2006).
Casteilla, L., Planat-Benard, V., Cousin, B., Laharrague, P., and Bourin, Inflammation and endothelial dysfunction in rheumatoid arthritis.
P. (2010). Vascular and endothelial regeneration. Current Stem Cell Clinical and Experimental Rheumatology, 24, 115–7.
Research and Therapy, 5, 141–4. Gonzalez-Gay, M.A., Gonzalez-Juanatey, C., and Ollier, W.E. (2004).
Celermajer, D.S., Sorensen, K.E., Gooch, V.M., Spiegelhalter, D.J., Miller, O.I., Endothelial dysfunction in rheumatoid arthritis:  influence of
Sullivan, I.D., Lloyd, J.K., and Deanfield, J.E. (1992). Non-invasive detec- HLA-DRB1 alleles. Autoimmunity Reviews, 3, 301–4.
tion of endothelial dysfunction in children and adults at risk of athero- Gonzalez-Gay, M.A., Gonzalez-Juanatey, C., Vazquez-Rodriguez, T.R.,
sclerosis. Lancet, 340, 1111–5. Martin, J., and Llorca, J. (2008). Endothelial dysfunction, carotid
Chen, M.  and Kallenberg, C.G. (2010). ANCA-associated vasculitides— intima-media thickness, and accelerated atherosclerosis in rheumatoid
advances in pathogenesis and treatment. Nature Reviews Rheumatology, arthritis. Seminars in Arthritis and Rheumatism, 38, 67–70.
6, 653–64. Gonzalez-Juanatey, C., Testa, A., Garcia-Castelo, A., Garcia-Porrua, C.,
Choe, J.Y., Park, S.H., and Kim, S.K. (2010). Serum angiopoietin-1 level is Llorca, J., Vidan, J., Hajeer, A.H., Ollier, W.E., Mattey, D.L., and
increased in patients with Behcet’s disease. Joint Bone Spine, 77, 340–4. Gonzalez-Gay, M.A. (2003). HLA-DRB1 status affects endothelial func-
Clarke, L.A., Hong, Y., Eleftheriou, D., Shah, V., Arrigoni, F., Klein, N.J., and tion in treated patients with rheumatoid arthritis. American Journal of
Brogan, P.A. (2010). Endothelial injury and repair in systemic vasculitis Medicine, 114, 647–52.
of the young. Arthritis and Rheumatism, 62, 1770–80. Grisar, J., Aletaha, D., Steiner, C.W., Kapral, T., Steiner, S., Saemann, M.,
Coll-Vinent, B., Cebrian, M., Cid, M.C., Font, C., Esparza, J., Juan, M., Yague, Schwarzinger, I., Buranyi, B., Steiner, G., and Smolen, J.S. (2007).
J., Urbano-Marquez, A., and Grau, J.M. (1998). Dynamic pattern of Endothelial progenitor cells in active rheumatoid arthritis:  effects
endothelial cell adhesion molecule expression in muscle and perineu- of tumour necrosis factor and glucocorticoid therapy. Annals of the
ral vessels from patients with classic polyarteritis nodosa. Arthritis and Rheumatic Diseases, 66, 1284–8.
Rheumatism, 41, 435–44. Gunnett, C.A., Lund, D.D., Mcdowell, A.K., Faraci, F.M., and Heistad, D.D.
Cracowski, J.L., Minson, C.T., Salvat-Melis, M., and Halliwill, J.R. (2006). (2005). Mechanisms of inducible nitric oxide synthase-mediated vas-
Methodological issues in the assessment of skin microvascular endothe- cular dysfunction. Arteriosclerosis Thrombosis and Vascular Biology, 25,
lial function in humans. Trends in Pharmacological Sciences, 27, 503–8. 1617–22.
De Leeuw, K., Sanders, J.S., Stegeman, C., Smit, A., Kallenberg, C.G., and Bijl, Halloran, M.M., Szekanecz, Z., Barquin, N., Haines, G.K., and Koch, A.E.
M. (2005). Accelerated atherosclerosis in patients with Wegener’s granu- (1996). Cellular adhesion molecules in rat adjuvant arthritis. Arthritis
lomatosis. Annals of the Rheumatic Diseases, 64, 753–9. and Rheumatism, 39, 810–9.
Drouet, C., Nissou, M.F., Ponard, D., Arvieux, J., Dumestre-Perard, C., Haskard, D.O. (1995). Cell adhesion molecules in rheumatoid arthritis.
Gaudin, P., Imbert, B., Massot, C., and Sarrot-Reynauld, F. (2003). Current Opinion in Rheumatology, 7, 229–34.
50 SECTION 2 basic science

Heo, K.S., Fujiwara, K., and Abe, J. (2012). Disturbed-flow-mediated vascu- Kamoun, M., Houman, M.H., Hamzaoui, A., and Hamzaoui, K. (2008).
lar reactive oxygen species induce endothelial dysfunction. Circulation Vascular endothelial growth factor gene polymorphisms and serum lev-
Journal, 75, 2722–30. els in Behcet’s disease. Tissue Antigens, 72, 581–7.
Herbrig, K., Haensel, S., Oelschlaegel, U., Pistrosch, F., Foerster, S., and Kaplanski, G., Maisonobe, T., Marin, V., Gres, S., Robitail, S., Farnarier, C.,
Passauer, J. (2006). Endothelial dysfunction in patients with rheumatoid Harle, J.R., Piette, J.C., and Cacoub, P. (2005). Vascular cell adhesion
arthritis is associated with a reduced number and impaired function of molecule-1 (VCAM-1). plays a central role in the pathogenesis of severe
endothelial progenitor cells. Annals of the Rheumatic Diseases, 65, 157–63. forms of vasculitis due to hepatitis C-associated mixed cryoglobuline-
Hewing, B., Stangl, V., Stangl, K., Enke-Melzer, K., Baumann, G., and Ludwig, mia. Journal of Hepatology, 42, 334–40.
A. (2012). Circulating angiogenic factors in patients with thromboangii- Karasawa, R., Yudoh, K., Ozaki, S., and Kato, T. (2011). Anti-endothelial cell
tis obliterans. PLoS ONE, 7, e34717. antibodies (AECA) in patients with systemic vasculitis:  our research
Higashi, Y., Sasaki, S., Nakagawa, K., Matsuura, H., Kajiyama, G., and using proteomics. Expert Opinion on Biological Therapy, 11, 77–87.
Oshima, T. (2001). A noninvasive measurement of reactive hyperemia Kasama, T., Wakabayashi, K., Sato, M., Takahashi, R., and Isozaki, T. (2010).
that can be used to assess resistance artery endothelial function in Relevance of the CX3CL1/fractalkine-CX3CR1 pathway in vasculitis
humans. American Journal of Cardiology, 87, 121–5, A9. and vasculopathy. Translational Research, 155, 20–6.
Holash, J., Maisonpierre, P.C., Compton, D., Boland, P., Alexander, C.R., Kawanaka, H., Takagi, G., Miyamoto, M., Tara, S., Takagi, I., Takano, H.,
Zagzag, D., Yancopoulos, G.D., and Wiegand, S.J. (1999). Vessel coop- Yasutake, M., Tabata, Y., and Mizuyno, K. (2009). Therapeutic angio-
tion, regression, and growth in tumors mediated by angiopoietins and genesis by controlled-release fibroblast growth factor in a patient with
VEGF. Science, 284, 1994–8. Churg-Strauss syndrome complicated by an intractable ischemic leg
Holmen, C., Elsheikh, E., Christensson, M., Liu, J., Johansson, A.S., Qureshi, ulcer. American Journal of Medicine Science, 338, 341–2.
A.R., Jalkanen, S., and Sumitran-Holgersson, S. (2007). Anti endothe- Kemeny-Beke, A., Gesztelyi, R., Bodnar, N., Zsuga, J., Kerekes, G., Zsuga,
lial cell autoantibodies selectively activate SAPK/JNK signalling in M., Biri, B., Keki, S., Szodoray, P., Berta, A., Szekanecz, Z., and Szanto, S.
Wegener’s granulomatosis. Journal of the American Society of Nephrology, (2011). Increased production of asymmetric dimethylarginine (ADMA)
18, 2497–508. in ankylosing spondylitis: association with other clinical and laboratory
Holmen, C., Elsheikh, E., Stenvinkel, P., Qureshi, A.R., Pettersson, E., parameters. Joint Bone Spine, 78, 184–7.
Jalkanen, S., and Sumitran-Holgersson, S. (2005). Circulating inflamma- Kerekes, G., Soltesz, P., Nurmohamed, M.T., Gonzalez-Gay, M.A., Turiel, M.,
tory endothelial cells contribute to endothelial progenitor cell dysfunc- Vegh, E., Shoenfeld, Y., McInnes, I., and Szekanecz, Z. (2012). Validated
tion in patients with vasculitis and kidney involvement. Journal of the methods for assessment of subclinical atherosclerosis in rheumatology.
American Society of Nephrology, 16, 3110–20. Nature Reviews Rheumatology, 8, 224–34.
Imhof, B.A. and Aurrand-Lions, M. (2004). Adhesion mechanisms regulat- Kerekes, G., Szekanecz, Z., Der, H., Sandor, Z., Lakos, G., Muszbek, L., Csipo,
ing the migration of monocytes. Nature Reviews Immunology, 4, 432–44. I., Sipka, S., Seres, I., Paragh, G., Kappelmayer, J., Szomjak, E., Veres,
Infusino, G.A. and Jacobson, J.R. (2012). Endothelial FAK as a therapeutic K., Szegedi, G., Shoenfeld, Y., and Soltesz, P. (2008). Endothelial dys-
target in disease. Microvascular Research, 83, 89–96. function and atherosclerosis in rheumatoid arthritis: a multiparametric
Isner, J.M., Kalka, C., Kawamoto, A., and Asahara, T. (2001). Bone marrow analysis using imaging techniques and laboratory markers of inflamma-
as a source of endothelial cells for natural and iatrogenic vascular repair. tion and autoimmunity. Journal of Rheumatology, 35, 398–406.
Annals of the New York Academy of Sciences, 953, 75–84. Kiselyov, A., Balakin, K.V., and Tkachenko, S.E. (2007). VEGF/VEGFR
Jaffe, E.A., Grulich, J., Weksler, B.B., Hampel, G., and Watanabe, K. (1987). signalling as a target for inhibiting angiogenesis. Expert Opinion on
Correlation between thrombin-induced prostacyclin production Investigational Drugs, 16, 83–107.
and inositol trisphosphate and cytosolic free calcium levels in cul- Koch, A.E. (2000). The role of angiogenesis in rheumatoid arthri-
tured human endothelial cells. Journal of Biological Chemistry, 262, tis:  recent developments. Annals of the Rheumatic Diseases, 59
8557–65. (Suppl. 1), i65–71.
Jalkanen, S. (1989). Leukocyte-endothelial cell interaction and the control Koch, A.E. and Distler, O. (2007). Vasculopathy and disordered angiogenesis
of leukocyte migration into inflamed synovium. Springer Seminars in in selected rheumatic diseases: rheumatoid arthritis and systemic scle-
Immunopathology, 11, 187–98. rosis. Arthritis Research and Therapy, 9 (Suppl 2), S3.
Jamin, C., Dugue, C., Alard, J.E., Jousse, S., Saraux, A., Guillevin, L., Piette, Koch, A.E., Harlow, L.A., Haines, G.K., Amento, E.P., Unemori, E.N., Wong,
J.C., and Youinou, P. (2005). Induction of endothelial cell apopto- W.L., Pope, R. M., and Ferrara, N. (1994). Vascular endothelial growth
sis by the binding of anti-endothelial cell antibodies to Hsp60 in factor. A  cytokine modulating endothelial function in rheumatoid
vasculitis-associated systemic autoimmune diseases. Arthritis and arthritis. Journal of Immunology, 152, 4149–56.
Rheumatism, 52, 4028–38. Koch, A.E., Volin, M.V., Woods, J.M., Kunkel, S.L., Connors, M.A., Harlow,
Jodon De Villeroche, V., Avouac, J., Ponceau, A., Ruiz, B., Kahan, A., Boileau, L.A., Woodruff, D.C., Burdick, M.D., and Strieter, R.M. (2001).
C., Uzan, G., and Allanore, Y. (2011). Enhanced late-outgrowth circulat- Regulation of angiogenesis by the C-X-C chemokines interleukin-8
ing endothelial progenitor cell levels in rheumatoid arthritis and correla- and epithelial neutrophil activating peptide 78 in the rheumatoid joint.
tion with disease activity. Arthritis Research and Therapy, 12, R27. Arthritis and Rheumatism, 44, 31–40.
Johnson, B.A., Haines, G.K., Harlow, L.A., and Koch, A.E. (1993). Adhesion Kollmeier, A., Kreditor, M., George, J., Levy, Y., and Shoenfeld, Y. (1998).
molecule expression in human synovial tissue. Arthritis and Rheumatism, Functional properties of antiendothelial cell antibodies. Clinical Reviews
36, 137–46. in Allergy and Immunology, 16, 297–302.
Joris, I., Cuenoud, H.F., Doern, G.V., Underwood, J.M., and Majno, G. Kuwana, M., Okazaki, Y., Yasuoka, H., Kawakami, Y., and Ikeda, Y. (2004).
(1990). Capillary leakage in inflammation. A study by vascular labeling. Defective vasculogenesis in systemic sclerosis. Lancet, 364, 603–10.
American Journal of Pathology, 137, 1353–63. Kvietys, P.R. and Granger, D.N. (2012). Role of reactive oxygen and nitrogen
Joris, I., Majno, G., Corey, E.J., and Lewis, R.A. (1987). The mechanism of species in the vascular responses to inflammation. Free Radical Biology
vascular leakage induced by leukotriene E4. Endothelial contraction. and Medicine, 52, 556–92.
American Journal of Pathology, 126, 19–24. Lainer-Carr, D.  and Brahn, E. (2007). Angiogenesis inhibition as a thera-
Kallenberg, C.G. (2010). Pathogenesis of ANCA-associated vasculitides. peutic approach for inflammatory synovitis. Nature Clinical Practice
Annals of the Rheumatic Diseases, 70 (Suppl. 1), i59–63. Rheumatology, 3, 434–42.
Kallenberg, C.G. (2011). Pathogenesis of ANCA-associated vasculitis, an Lu, D. and Kassab, G.S. (2011). Role of shear stress and stretch in vascular
update. Clinical Reviews in Allergy and Immunology, 41, 224–31. mechanobiology. Journal of the Royal Society Interface, 8, 1379–85.
CHAPTER 4 biology of endothelial cells 51

Lum, H. and Roebuck, K.A. (2001). Oxidant stress and endothelial cell dys- Ramli, J., Calderonartero, P., Block, R.C., and Mousa, S.A. (2011). Novel
function. American Journal of Physiology Cell Physiology, 280, C719–41. therapeutic targets for preserving a healthy endothelium: strategies for
Madri, J.A. and Williams, S.K. (1983). Capillary endothelial cell cultures: pheno- reducing the risk of vascular and cardiovascular disease. Cardiology
typic modulation by matrix components. Journal of Cell Biology, 97, 153–65. Journal, 18, 352–63.
Maruotti, N., Cantatore, F.P., Nico, B., Vacca, A., and Ribatti, D. (2008). Raza, K., Thambyrajah, J., Townend, J.N., Exley, A.R., Hortas, C., Filer, A.,
Angiogenesis in vasculitides. Clinical and Experimental Rheumatology, Carruthers, D.M., and Bacon, P.A. (2000). Suppression of inflamma-
26, 476–83. tion in primary systemic vasculitis restores vascular endothelial func-
Mitchell, E.B. and Cestari, D.M. (2009). Giant cell arteritis and angiogen- tion: lessons for atherosclerotic disease? Circulation, 102, 1470–2.
esis: a review. Seminars in Ophthalmology, 24, 190–3. Resnick, N., Yahav, H., Shay-Salit, A., Shushy, M., Schubert, S., Zilberman,
Monach, P.A., Tomasson, G., Specks, U., Stone, J.H., Cuthbertson, D., L.C., and Wofovitz, E. (2003). Fluid shear stress and the vascular
Krischer, J., Ding, L., Fervenza, F.C., Fessler, B.J., Hoffman, G.S., Ikle, endothelium:  for better and for worse. Progress in Biophysics and
D., Kallenberg, C.G., Langford, C.A., Mueller, M., Seo, P., St Clair, E.W., Molecular Biology, 81, 177–99.
Spiera, R., Tchao, N., Ytterberg, S.R., Gu, Y.Z., Snyder, R.D., and Merkel, Salmi, M., Kalimo, K., and Jalkanen, S. (1993). Induction and function
P.A. (2012). Circulating markers of vascular injury and angiogenesis in of vascular adhesion protein-1 at sites of inflammation. Journal of
antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis and Experimental Medicine, 178, 2255–60.
Rheumatism, 63, 3988–97. Sarraj, B., Ludanyi, K., Glant, T.T., Finnegan, A., and Mikecz, K. (2006).
Mullan, R.H., Bresnihan, B., Golden-Mason, L., Markham, T., O’hara, R., Expression of CD44 and L-selectin in the innate immune system is required
Fitzgerald, O., Veale, D.J., and Fearon, U. (2006). Acute-phase serum amy- for severe joint inflammation in the proteoglycan-induced murine model
loid A stimulation of angiogenesis, leukocyte recruitment, and matrix of rheumatoid arthritis. Journal of Immunology, 177, 1932–40.
degradation in rheumatoid arthritis through an NF-kappaB-dependent Savage, C.O. (2009). Vascular biology and vasculitis. APMIS Supplementum,
signal transduction pathway. Arthritis and Rheumatism, 54, 105–14. 127, 37–40.
Muller, W.A. (2009). Mechanisms of transendothelial migration of leuko- Savage, C.O. and Williams, J.M. (2007). Anti endothelial cell antibodies in
cytes. Circulation Research, 105, 223–30. vasculitis. Journal of the American Society of Nephrology, 18, 2424–6.
Mulligan, M.S., Varani, J., Dame, M.K., Lane, C.L., Smith, C.W., Anderson, Schneeweis, C., Rafalowicz, M., Feist, E., Buttgereit, F., Rudolph, P.E.,
D.C., and Ward, P. A. (1991). Role of endothelial-leukocyte adhesion Burmester, G.R., and Egerer, K. (2010). Increased levels of BLyS and
molecule 1 (ELAM-1). in neutrophil-mediated lung injury in rats. sVCAM-1 in anti-neutrophil cytoplasmatic antibody (ANCA)-associated
Journal of Clinical Investigation, 88, 1396–406. vasculitides (AAV). Clinical and Experimental Rheumatology, 28, 62–6.
Muz, B., Khan, M.N., Kiriakidis, S., and Paleolog, E.M. (2009). Hypoxia. The Seyahi, E., Ugurlu, S., Cumali, R., Balci, H., Seyahi, N., Yurdakul, S., and
role of hypoxia and HIF-dependent signalling events in rheumatoid Yazici, H. (2006). Atherosclerosis in Takayasu arteritis. Annals of the
arthritis. Arthritis Research and Therapy, 11, 201. Rheumatic Diseases, 65, 1202–7.
Naik, T.U., Naik, M.U., and Naik, U.P. (2008). Junctional adhesion molecules Shibuya, M. (2008). Vascular endothelial growth factor-dependent and
in angiogenesis. Frontiers in Bioscience, 13, 258–62. -independent regulation of angiogenesis. BMB Reports, 41, 278–86.
Pakozdi, A., Amin, M.A., Haas, C.S., Martinez, R.J., Haines, G.K., 3rd, Santos, Shoenfeld, Y., Gerli, R., Doria, A., Matsuura, E., Cerinic, M.M., Ronda, N.,
L.L., Morand, E.F., David, J.R., and Koch, A.E. (2006). Macrophage migra- Jara, L.J., Abu-Shakra, M., Meroni, P.L., and Sherer, Y. (2005). Accelerated
tion inhibitory factor: a mediator of matrix metalloproteinase-2 produc- atherosclerosis in autoimmune rheumatic diseases. Circulation, 112,
tion in rheumatoid arthritis. Arthritis Research and Therapy, 8, R132. 3337–47.
Pakozdi, A., Besenyei, T., Paragh, G., Koch, A.E., and Szekanecz, Z. (2009). Sibelius, U., Hattar, K., Schenkel, A., Noll, T., Csernok, E., Gross, W.L., Mayet,
Endothelial progenitor cells in arthritis-associated vasculogenesis and W.J., Piper, H.M., Seeger, W., and Grimminger, F. (1998). Wegener’s
atherosclerosis. Joint Bone Spine, 76, 581–3. granulomatosis:  anti-proteinase 3 antibodies are potent inductors
Paleolog, E. (2005). It’s all in the blood: circulating endothelial progenitor of human endothelial cell signaling and leakage response. Journal of
cells link synovial vascularity with cardiovascular mortality in rheuma- Experimental Medicine, 187, 497–503.
toid arthritis? Arthritis Research and Therapy, 7, 270–2. Skotnicki, J.S., Zask, A., Nelson, F.C., Albright, J.D., and Levin, J.I. (1999).
Pankhurst, T., Savage, C.O., and Little, M.A. (2009). Review arti- Design and synthetic considerations of matrix metalloproteinase inhibi-
cle: Leukocyte-endothelial dysregulation in systemic small vessel vascu- tors. Annals of the New York Academy of Sciences, 878, 61–72.
litis. Nephrology (Carlton), 14, 3–10. Springer, T.A. (1990). Adhesion receptors of the immune system. Nature,
Patel, K.D., Cuvelier, S.L., and Wiehler, S. (2002). Selectins: critical mediators 346, 425–34.
of leukocyte recruitment. Seminars in Immunology, 14, 73–81. Strieter, R.M., Polverini, P.J., Kunkel, S.L., Arenberg, D.A., Burdick, M.D.,
Peichev, M., Naiyer, A.J., Pereira, D., Zhu, Z., Lane, W. J., Williams, M., Oz, Kasper, J., Dzuiba, J., Van Damme, J., Walz, A., Marriott, D. et al. (1995).
M.C., Hicklin, D.J., Witte, L., Moore, M.A., and Rafii, S. (2000). Expression The functional role of the ELR motif in CXC chemokine-mediated angi-
of VEGFR-2 and AC133 by circulating human CD34(+). cells identifies a ogenesis. Journal of Biological Chemistry, 270, 27348–57.
population of functional endothelial precursors. Blood, 95, 952–8. Szekanecz, Z., Besenyei, T., Paragh, G., and Koch, A.E. (2009a). Angiogenesis
Petit, I., Jin, D., and Rafii, S. (2007). The SDF-1-CXCR4 signaling pathway: a in rheumatoid arthritis. Autoimmunity, 42, 563–73.
molecular hub modulating neo-angiogenesis. Trends in Immunology, 28, Szekanecz, Z., Besenyei, T., Szentpetery, A., and Koch, A.E. (2010a).
299–307. Angiogenesis and vasculogenesis in rheumatoid arthritis. Current
Pober, J.S. and Cotran, R.S. (1990). Cytokines and endothelial cell biology. Opinion in Rheumatology, 22, 299–306.
Physiological Reviews, 70, 427–51. Szekanecz, Z., Haines, G K., Harlow, L.A., Shah, M.R., Fong, T.W., Fu, R., Lin,
Pober, J.S., Kluger, M.S., and Schechner, J.S. (2001). Human endothelial cell S.J., and Koch, A.E. (1995). Increased synovial expression of the adhe-
presentation of antigen and the homing of memory/effector T cells to sion molecules CD66a, CD66b, and CD31 in rheumatoid and osteoar-
skin. Annals of the New York Academy of Sciences, 941, 12–25. thritis. Clinical Immunology and Immunopathology, 76, 180–6.
Poredos, P. (2001). Endothelial dysfunction in the pathogenesis of atheroscle- Szekanecz, Z., Haines, G.K., Lin, T.R., Harlow, L.A., Goerdt, S., Rayan, G.,
rosis. Clinical and Applied Thrombosis Hemostasis, 7, 276–80. and Koch, A.E. (1994). Differential distribution of intercellular adhesion
Rabquer, B.J., Pakozdi, A., Michel, J.E., Gujar, B.S., Haines, G.K., 3rd, Imhof, molecules (ICAM-1, ICAM-2, and ICAM-3). and the MS-1 antigen in
B.A., and Koch, A.E. (2008). Junctional adhesion molecule C mediates normal and diseased human synovia. Their possible pathogenetic and
leukocyte adhesion to rheumatoid arthritis synovium. Arthritis and clinical significance in rheumatoid arthritis. Arthritis and Rheumatism,
Rheumatism, 58, 3020–9. 37, 221–31.
52 SECTION 2 basic science

Szekanecz, Z., Kerekes, G., Der, H., Sandor, Z., Szabo, Z., Vegvari, A., Synergistic interaction of oxygen products and proteases. American
Simkovics, E., Soos, L., Szentpetery, A., Besenyei, T., Szucs, G., Szanto, S., Journal of Pathology, 135, 435–8.
Tamasi, L., Szegedi, G., Shoenfeld, Y., and Soltesz, P. (2007). Accelerated Vaudo, G., Marchesi, S., Gerli, R., Allegrucci, R., Giordano, A., Siepi, D., Pirro,
atherosclerosis in rheumatoid arthritis. Annals of the New York Academy M., Shoenfeld, Y., Schillaci, G., and Mannarino, E. (2004). Endothelial
of Sciences, 1108, 349–58. dysfunction in young patients with rheumatoid arthritis and low disease
Szekanecz, Z.  and Koch, A.E. (2000). Cell-cell interactions in synovitis. activity. Annals of the Rheumatic Diseases, 63, 31–5.
Endothelial cells and immune cell migration. Arthritis Research, 2, 368–73. Veale, D.J. and Fearon, U. (2006). Inhibition of angiogenic pathways in rheu-
Szekanecz, Z. and Koch, A.E. (2001). Chemokines and angiogenesis. Current matoid arthritis: potential for therapeutic targeting. Best Practice and
Opinion in Rheumatology, 13, 202–8. Research Clinical Rheumatology, 20, 941–7.
Szekanecz, Z.  and Koch, A.E. (2004). Vascular endothelium and immune Visvanathan, S., Rahman, M.U., Hoffman, G.S., Xu, S., Garcia-Martinez, A.,
responses: implications for inflammation and angiogenesis. Rheumatic Segarra, M., Lozano, E., Espigol-Frigole, G., Hernandez-Rodriguez, J.,
Disease Clinics of North America, 30, 97–114. and Cid, M.C. (2011). Tissue and serum markers of inflammation dur-
Szekanecz, Z. and Koch, A.E. (2005). Endothelial cells in inflammation and ing the follow-up of patients with giant-cell arteritis—a prospective lon-
angiogenesis. Current Drug Targets Inflammation and Allergy, 4, 319–23. gitudinal study. Rheumatology (Oxford), 50, 2061–70.
Szekanecz, Z. and Koch, A.E. (2007a). Macrophages and their products in Voskuyl, A.E., Martin, S., Melchers, L., Zwinderman, A.H., Weichselbraun, I.,
rheumatoid arthritis. Current Opinion in Rheumatology, 19, 289–95. and Breedveld, F.C. (1995). Levels of circulating intercellular adhesion
Szekanecz, Z. and Koch, A.E. (2007b). Mechanisms of Disease: angiogenesis in molecule-1 and -3 but not circulating endothelial leucocyte adhesion
inflammatory diseases. Nature Clinical Practice Rheumatology, 3, 635–43. molecule are increased in patients with rheumatoid vasculitis. British
Szekanecz, Z. and Koch, A.E. (2008). Vascular involvement in rheumatic dis- Journal of Rheumatology, 34, 311–5.
eases: ‘vascular rheumatology’. Arthritis Research and Therapy, 10, 224. Westphal, J.R., Boerbooms, A.M., Schalwijk, C.J., Kwast, H., De Weijert, M.,
Szekanecz, Z. and Koch, A.E. (2010). Vasculogenesis in rheumatoid arthritis. Jacobs, C., Vierwinden, G., Ruiter, D.J., Van De Putte, L.B., and De Waal,
Arthritis Research and Therapy, 12, 110. R.M. (1994). Anti-endothelial cell antibodies in sera of patients with
Szekanecz, Z., Koch, A.E., Kunkel, S.L., and Strieter, R.M. (1998). Cytokines autoimmune diseases: comparison between ELISA and FACS analysis.
in rheumatoid arthritis. Potential targets for pharmacological interven- Clinical and Experimental Immunology, 96, 444–9.
tion. Drugs Aging, 12, 377–90. Westra, J., Molema, G., and Kallenberg, C.G. (2010). Hypoxia-Inducible
Szekanecz, Z., Koch, A.E., and Tak, P.P. (2011). Chemokine and chemokine Factor-1 as Regulator of Angiogenesis in Rheumatoid Arthritis—
receptor blockade in arthritis, a prototype of immune-mediated inflam- Therapeutic Implications. Current Medicinal Chemistry, 172, 54–63.
matory diseases. Netherlands Journal of Medicine, 69, 356–66. Widlansky, M.E., Gokce, N., Keaney, J.F. Jr, and Vita, J.A. (2003). The clinical
Szekanecz, Z., Pakozdi, A., Szentpetery, A., Besenyei, T., and Koch, A.E. implications of endothelial dysfunction. Journal of the American College
(2009b). Chemokines and angiogenesis in rheumatoid arthritis. of Cardiology, 42, 1149–60.
Frontiers in Bioscience (Elite Ed), 1, 44–51. Witkowska, A.M., Kuryliszyn-Moskal, A., Borawska, M.H., Hukalowicz, K.,
Szekanecz, Z., Szegedi, G., and Koch, A.E. (1996). Cellular adhesion mole- and Markiewicz, R. (2003). A study on soluble intercellular adhesion
cules in rheumatoid arthritis: regulation by cytokines and possible clini- molecule-1 and selenium in patients with rheumatoid arthritis compli-
cal importance. Journal of Investigative Medicine, 44, 124–35. cated by vasculitis. Clinical Rheumatology, 22, 414–9.
Szekanecz, Z., Vegvari, A., Szabo, Z., and Koch, A.E. (2010b). Chemokines Yamahara, K.  and Itoh, H. (2009). Potential use of endothelial progeni-
and chemokine receptors in arthritis. Frontiers in Bioscience (Schol Ed), tor cells for regeneration of the vasculature. Therapeutic Advances in
2, 153–67. Cardiovascular Diseases, 3, 17–27.
Szucs, G., Timar, O., Szekanecz, Z., Der, H., Kerekes, G., Szamosi, S., Yang, Y.H., Rajaiah, R., Ruoslahti, E., and Moudgil, K.D. (2011). Peptides
Shoenfeld, Y., Szegedi, G., and Soltesz, P. (2007). Endothelial dysfunc- targeting inflamed synovial vasculature attenuate autoimmune arthritis.
tion precedes atherosclerosis in systemic sclerosis—relevance for pre- Proceedings of the National Academy of Sciences U S A, 108, 12857–62.
vention of vascular complications. Rheumatology (Oxford), 46, 759–62. Yoo, S.A., Yoon, H.J., Kim, H.S., Chae, C.B., De Falco, S., Cho, C.S., and Kim,
Taylor, P.C., and Sivakumar, B. (2005). Hypoxia and angiogenesis in rheuma- W.U. (2009). Role of placenta growth factor and its receptor flt-1 in
toid arthritis. Current Opinion in Rheumatology, 17, 293–8. rheumatoid inflammation: a link between angiogenesis and inflamma-
tion. Arthritis and Rheumatism, 60, 345–54.
Tesfamariam, B., and Defelice, A.F. (2007). Endothelial injury in the initiation
and progression of vascular disorders. Vascul Pharmacol, 46, 229–37. Zhang, C. (2008). The role of inflammatory cytokines in endothelial dysfunc-
tion. Basic Research in Cardiology, 103, 398–406.
Turkcapar, N., Sak, S.D., Saatci, M., Duman, M., and Olmez, U. (2005).
Vasculitis and expression of vascular cell adhesion molecule-1, intercel- Ziff, M. (1991). Role of the endothelium in chronic inflammatory synovitis.
lular adhesion molecule-1, and E-selectin in salivary glands of patients Arthritis and Rheumatism, 34, 1345–52.
with Sjogren’s syndrome. Journal of Rheumatology, 32, 1063–70. Zouki, C., Baron, C., Fournier, A., and Filep, J.G. (1999). Endothelin-1
Van Der Zee, J.M., Heurkens, A. H., Van Der Voort, E.A., Daha, M.R., and enhances neutrophil adhesion to human coronary artery endothe-
Breedveld, F.C. (1991). Characterization of anti-endothelial antibodies lial cells: role of ET(A) receptors and platelet-activating factor. British
in patients with rheumatoid arthritis complicated by vasculitis. Clinical Journal of Pharmacology, 127, 969–79.
and Experimental Rheumatology, 9, 589–94. Zsuga, J. (2008). [Asymmetric dimethil-arginine (ADMA) as a link between
Varani, J., Ginsburg, I., Schuger, L., Gibbs, D.F., Bromberg, J., Johnson, K.J., insulin resistance and atherosclerosis]. Ideggyógyászati Szemle, 61,
Ryan, U.S., and Ward, P.A. (1989). Endothelial cell killing by neutrophils. 183–92.
CHAPTER 5

Complement in vasculitis
John E. Volanakis

Complement and complement to form the C4b2a complex, which is a C3 convertase (Figure 5.1).
In the lectin pathway, recognition of pathogens is carried out
activation by mannan-binding lectin (MBL) or the serum ficolins L and H
The human complement system consists of more than 35 plasma (Holmskov 2003; Matsushita and Fujita 2001). All three proteins
and membrane-bound proteins, including recognition molecules, are pattern-recognition lectins composed of collagen and globular
proteases, cellular receptors, and regulatory proteins. Together they lectin domains. They are oligomers of homotrimeric subunits in
constitute a major recognition and effector system in host defence which the collagen regions form a triple helix. In MBL the recogni-
and act in a self-controlled fashion (Volanakis 1998; Walport tion domain is a C-type lectin, while in the ficolins, it is a fibrinogen
2001). The system aims at eliminating invading pathogens from domain. MBL has binding specificity for mannose, glucose, fucose,
blood and tissues and also participates in regulation of adaptive N-acetylmannosamine, and N-acetylglucosamine. The ficolins
immune responses. Activation of complement is necessary for have specificity for N-acetylglucosamine. Their lectin specificity
expression of biological function and is initiated through three endows these proteins with the ability to bind to a wide variety of
independent recognition mechanisms, the classical, the lectin, and Gram-positive and Gram-negative bacteria. In blood all three pro-
the alternative pathways. The three pathways merge at C3 cleavage, teins circulate as complexes with homodimers of MBL-associated
which is catalysed by two distinct but structurally similar and cata- serine proteases (MASP)-1, -2, or, -3. Binding of the lectin to a
lytically identical proteases, termed C3 convertases (Figure  5.1). bacterial cell surface leads to activation of MASP, through a cur-
Although complement activation is primarily beneficial to the host, rently unknown mechanism. Activated MASP-2 can then act like
complement-mediated inflammatory responses can lead to tissue C1s, activating sequentially C4 and C2 to form the C4b2a, C3 con-
damage under conditions of excessive or inappropriate activation vertase. The function of MASP-1 and -3 is currently controversial.
(Volanakis 2005). It has become clear that the physiological spec- The alternative pathway is activated by a variety of cellular sur-
trum of complement by far exceeds host defence and inflammation, faces, including those of certain bacteria, viruses, fungi, and para-
and that the complement network is highly intertwined with a vari- sites (Fearon and Austen 1980; Pangburn and Müller-Eberhard
ety of bodily systems. Biologically active products of complement 1984). Antibodies, particularly in the form of large, insoluble com-
are involved in the development and differentiation of B and T plexes with antigen, can also activate this pathway. Assembly of the
lymphocytes and also have been implicated in non-immunological C3 convertase is initiated by the covalent attachment of freshly gen-
functions associated with developmental biology, CNS develop- erated, metastable C3b to the activator surface. A supply of metasta-
ment and neurodegeneration, tissue regeneration, and haemat- ble C3b is always available in blood through a mechanism termed
opoiesis (Mastellos et al. 2005, Haynes et al. 2013). However, in this
chapter we focus on the proinflammatory function of complement Classical Lectin Alternative
because of its relevance to vasculitis. C1q MBL, ficolins H and L
The three complement activation pathways recognize a wide C1r
range of pathogens and necrotic or apoptotic cells of the host and C1s MASP2 D C3b
initiate the assembly of the complement convertases by using dif-
ferent proteins and different though similar mechanisms; how- C4 C2 P B
ever, all three pathways result in the production of the same
biological activities. In the classical pathway, recognition of
activators is through the C1q component of the C1 complex. C1 C3 convertase
is a large protein complex comprising one molecule of C1q and C3 C3b + C3a
a Ca2+-dependent tetramer C1s-C1r-C1r-C1s, consisting of two
serine proteases, C1r and C1s, in their zymogen form (Gaboriaud + C3b
et al. 2004). C1q recognizes antigen-bound IgG and IgM antibod-
ies, ligand-bound C-reactive protein, and certain bacteria and C5 convertase
apoptotic cells. Binding of C1q to a target cell or protein complex C5 C5b + C5a
elicits a signal that triggers self-activation of C1r, which in turn
activates C1s. Activated C1s then cleaves sequentially C4 and C2 Fig. 5.1 Complement activation pathways.
54 SECTION 2 basic science

C3 tick-over. The chemical nature of the activator surface allows of C5 by a C5 convertase generates two fragments:  C5a, a small
factor B to form a complex with activator-bound-C3b. Factor B peptide, and C5b, a large two-polypeptide chain fragment. C5b can
bound to C3b is then cleaved by factor D into two fragments, the initiate the assembly of a large protein–protein complex, termed
largest of which, Bb, is a serine protease and remains attached to membrane attack complex (MAC), by interacting sequentially with
C3b. The resulting C3bBb complex is the C3 convertase of the alter- single molecules of C6, C7, and C8, and with six to 18 molecules of
native pathway. This complex has a short half-life, but it is stabilized C9 (Podack and Tschopp 1984; Esser 1994). During these sequen-
by the binding of properdin (P) and is usually termed the amplifica- tial interactions, hydrophobic regions of the participating proteins
tion C3 convertase because it generates many C3b fragments and, become exposed on the surface of the complex. Assembly of the
thus, additional C3 convertase complexes. MAC on the surface of a biological membrane favours interactions
of these hydrophobic regions with the fatty acid chains of mem-
Biologically active complement brane phospholipids (Figure 5.2). The complex becomes gradually
inserted into the lipid bilayer and eventually forms a transmem-
activation products brane channel, which can elicit various cellular functions (Rus et al.
A C3 convertase assembled on the surface of a complement acti- 2001) or lead to apoptosis or killing of susceptible cells.
vator can catalyse the cleavage of numerous C3 molecules into The acute inflammatory response is the principal means of host
the small peptide C3a and the large fragment C3b. A portion of defence against infection by pyogenic bacteria. Complement can
the resulting metastable C3b binds covalently to the surface of elicit all aspects of acute inflammation and frequently is activated not
the activator around the C3-convertase site. Such C3b molecules only during infections but also in inflammatory diseases of immune
have one of several functions. They can initiate the assembly of or autoimmune aetiology. Three products of complement activa-
additional C3-convertase, they can be recognized by complement tion, C3a, C5a, and the MAC, elicit vascular changes characteris-
receptor 1 (CR1; CD35) or complement receptor CRIg (Helmy tic of inflammation. The two small peptides, C3a and C5a, termed
et al. 2006), or can serve as precursors for smaller C3 fragments anaphylatoxins, interact with their respective receptors, C3aR
(iC3b, C3dg, C3d), produced by factor I cleavage and recognized (Ames et al. 1996) and C5aR (CD88) (Gerard and Gerard 1991) to
by complement receptors 2 (CR2; CD21), 3 (CR3; CD11b/CD18), elicit multiple functions. Both receptors have a structure typical of
or 4 (CR4; CD11c/CD18). Binding of C3b or its fragments to these seven transmembrane domain, G-protein coupled, rhodopsin fam-
complement receptors triggers important biological functions. For ily receptors and signal through Gai and Ga16. A second receptor
example, binding of immune complex-bound C3b to CR1 on red for C5a, C5L2 (gpr77) (Ohno et al. 2000) is structurally similar to
blood cells provides the main mechanism for removal of immune C5aR but it does not appear to couple to G proteins and its ability to
complexes from the circulation (Davies and Walport 1998); CRIg transduce signals is disputed. C3aR is expressed on cells of myeloid
is the main phagocytosis-promoting complement receptor. It is origin such as neutrophils, basophils, eosinophils, mast cells, mono-
expressed exclusively on tissue-resident macrophages, for exam- cytes/macrophages, dendritic cells (DC), and microglia (Ward 2009).
ple Kupffer cells. Binding of pathogen-bound C3b to CRIg on Additionally, T cells and some non-myeloid cells express C3aR.
such cells leads to phagocytosis of the bacteria. Binding of iC3b C5aR is expressed on a wider variety of cell types. It is most abun-
fragments to CR3 enhances phagocytosis of bacteria and other dantly expressed on neutrophils, eosinophils and basophils, mono-
particles; binding of C3dg or C3d to CR2 on B cells contributes cytes/macrophages, mast cells, and DCs (Ward 2009). C5aR is also
to B-cell activation and maturation (Fearon and Locksey 1996; expressed on lymphoid cells, including T cells (Lalli et al. 2008; Sacks
Carroll 2004). 2010). In addition to immune cells, a body of evidence has accumu-
Metastable C3b, generated by the action of a C3 convertase, lated that C5aR is expressed in a broad range of non-immune tissue
can also bind to the non-catalytic subunit, C4b or C3b, of the cells (reviewed in Monk et al. 2007; Klos et al. 2009). These include
convertase, giving rise to a trimolecular complex, C4b2a3b or endothelial cells, neurons, astrocytes, and microglia. Engagement of
(C3b)2Bb, respectively (Takata et al. 1987; Kinoshita et al. 1988). C3aR or C5aR by their respective anaphylatoxin triggers a variety of
Both of these trimolecular complexes are C5 convertases express- functions that depend on the cells expressing them. Best studied are
ing proteolytic activity restricted to the cleavage of C5. Cleavage acute inflammatory reactions, including activation and chemotactic

C6 C7 C8 C9 C9n

MAC

C5b C5b6 C5b67 C5b678 C5b-9 C5b-9n

Fig. 5.2 Sequential non-enzymatic protein–protein interactions among the terminal complement proteins
initiated by C5b and leading to the formation of the membrane attack complex (MAC).
CHAPTER 5 complement in vasculitis 55

responses of phagocytic cells. However, the known repertoire of ‘homologous restriction’ is not, however, absolute and considerable
anaphylatoxin-elicited functions has been expanded to include differ- overlap exists among different species (Morgan et al. 2005).
entiation of naïve CD4+ T cells to T effector and Th17 cells (Strainic The importance of the regulatory proteins in protecting self tis-
et al. 2008), but also non-immunological functions associated with sues from complement activation is demonstrated by the clinical
tissue development and regeneration (Monk et al. 2007; Klos et al. syndromes associated with mutations of their genes. Heterozygous
2009, Haynes et al. 2013). The function of C5L2 is at present con- C1 INH deficiency is associated with hereditary angioneurotic
troversial. There are numerous conflicting reports suggesting that oedema (Davis 2005) and factor I deficiency with susceptibility to
C5L2 is a ‘default receptor’ that attenuates C5a-dependent bio- infection, vasculitis, and glomerulonephritis (Genel et  al. 2005).
logical responses by competing with C5aR for binding of C5a and Mutations of the factor H, MCP, or factor I  genes predispose to
C5a-desArg (C5a missing the carboxyl terminal arginine, which is membranoproliferative glomerulonephritis and also to atypical
necessary for some but not all functions of C5a). On the other hand, haemolytic uremic syndrome, a thrombotic microangiopathy char-
there are other reports suggesting that C5L2 plays an active, positive acterized by haemolytic anaemia, thrombocytopenia, and acute
role in inflammatory responses (Chen et al. 2007). renal failure (Dragon-Durey and Fremeaux-Bacchi 2005). Finally,
somatic mutation and subsequent clonal expansion of the pig-A
gene, a component of the GPI-N-acetylglucosaminyltransferase,
Complement regulation results in absence from cell membranes of DAF and CD59, as well
Several mechanisms ensure that the self-destructing potential of as all other GPI-linked non-complement proteins. The defect is
complement activation products is normally kept under control associated with paroxysmal nocturnal haemoglobinuria, charac-
(Liszewski et  al. 1996). Complement convertases have a short terized by haemolysis, venous thrombosis, and cytopenia (Takeda
half-life; therefore production of active products is limited and pro- et al. 1993).
portional to the magnitude and persistence of the activating insult. While the combined action of the regulatory proteins adequately
In addition, three serum proteins, C4b-binding protein (C4bp), protects host tissues from bystander complement activation dam-
and factors H and I, regulate the convertases. C4bp dissociates the age, they are unable to fully protect tissues that become targets of
classical and lectin pathway convertase and inhibits its assembly. complement activation as a result of attachment of tissue-specific
It also serves as cofactor for cleavage/inactivation of C4b by factor autoantibodies or of deposition of immune complexes, or because
I, a serine protease. Factor H has a similar function in the alter- of changes in the surface characteristics of the tissue. It is not sur-
native pathway, dissociating the C3bBb convertase, inhibiting its prising that the vascular endothelium, which is in constant contact
formation, and serving as factor I cofactor in cleavage of C3b. As with complement in the blood, is particularly susceptible to com-
mentioned, cleavage of C3b by factor I produces fragments (iC3b, plement attack.
C3dg) with distinct biological activities, which, however, unlike
C3b, cannot form a C3 convertase. At the level of the initiating Complement-induced endothelial
enzymes of the classical and lectin pathways, control is exercised
by C1 inhibitor (C1 INH), a serpin (serine protease inhibitor) that damage
inhibits activated C1r, C1s, and MASPs (Davis et  al. 2010). The Activation of complement directed against the vascular endothe-
action of anaphylatoxins is regulated by anaphylatoxin inactivator, lium leads to profound changes in its structure and function.
a serum carboxypeptidase N (Plummer and Hurwitz 1978); finally, Endothelial damage is mediated mainly by the action of C5a and
the serum proteins vitronectin (S-protein) and clusterin inhibit the the MAC (Figure  5.3). C5a has both direct and indirect effects,
formation of the MAC, thereby precluding its insertion into lipid
bilayers (Morgan 1999).
C5 convertase
Four cell-associated proteins, decay-accelerating factor (DAF, C5 C5b + C5a
CD55) (Lukacik et  al. 2004), membrane-cofactor protein (MCP,
CD46) (Liszewski et  al. 2005), CR1 (Weisman et  al. 1990), and C6
CD59 (Huang et al. 2005), play a major role in protecting host tis- C7 neutrophil activation
sues from the damaging potential of complement activation. DAF, C8 endothelial cell retraction
MCP, and CD59 are widely distributed in human tissues, while the C9 activation of proteases
P-selectin expression
distribution of CR1 is limited to blood cells, follicular dendritic
cells, and glomerular podocytes. DAF inhibits the formation and C5b-9n
enhances the dissociation of both assembled C3 convertases, MCP
membrane attack complex (MAC)
serves as factor I  cofactor for C4b and C3b cleavage, while CR1
expresses all four of these functions. CD59 inhibits incorporation
of C9 into the C5b678 complex and prevents C9 polymerization,
thus preventing formation of the MAC. cell lysis/apoptosis
DAF and CD59 are glycan phosphatidylinositol (GPI)-linked endothelial cell activation
α secretion
membrane proteins. GPI serves as a membrane anchor for many    
additional cell surface proteins and its biosynthesis is initiated by 
   

    

an enzyme complex called GPI-N-acetylglucosaminyltransferase.     
DAF, MCP, and CD59 exhibit species-specificity, effectively pro-
tecting tissues from homologous complement activation, but not Fig. 5.3 Biological activities of C5a and MAC that play a role in the pathogenesis
from complement of other species (Atkinson et  al. 1991). Such of endothelial injury.
56 SECTION 2 basic science

the latter mediated mainly by neutrophils. Neutrophils activated with consequent activation of complement and neutrophils. A vio-
by C5a produce reactive oxygen species and release elastase and lent acute inflammatory reaction ensues, with destruction of the
cathepsin G, which may disrupt the intercellular junctions and vascular wall followed by oedema, haemorrhage, and necrosis.
impair the barrier function of the endothelium (Owen et al. 1995). The Arthus reaction is relevant to immune complex vasculitis,
Elastase cleaves proteoglycan on the endothelial surface, causing as exemplified by leukocytoclastic (hypersensitivity) vasculitis
the release of heparin sulphate and leading to loss of the antico- and perhaps also by the vasculitis of systemic lupus erythemato-
agulant, anti-inflammatory, and barrier function of the endothe- sus (SLE). Participation of complement in the pathogenesis of the
lium (Key et al. 1992). Direct effects of C5a are mediated through Arthus reaction has being questioned on the basis of experiments
its receptor, which is expressed on endothelial cells (Haviland demonstrating development of the lesion in mice deficient in C3 or
et  al. 1995). Engagement of C5aR by its ligand leads to multiple C4, whereas grossly diminished reactions were observed in animals
effects, including a sustained pertussis toxin-sensitive cytoskeletal deficient in IgG receptors (FcγR) (Sylvestre et al. 1996). Subsequent
response, resulting in cell retraction and increased paracellular per- experiments, however, indicated that complement was indeed
meability (Albrecht et al. 2004). It also leads to transient expres- necessary for development of Arthus reactions. Specifically, it was
sion of P-selectin by fusion of Weibel–Palade bodies containing shown that animals deficient in C5aR failed to develop the reaction
P-selectin, as well as von Willebrand factor and coagulation factor (Hopken et al. 1997; Baumann et al. 2001). These divergent results
V (Foreman et al. 1994). C5a also causes activation of serine and were reconciled by the observation that C5a is a major inducer of
cysteine proteases secreted by endothelial cells (Ihrcke and Platt the genes of activating, but not inhibitory, FcγRs (Shushakova et al.
1996). P-selectin causes adhesion of neutrophils and facilitates 2002). Thus, it appears that both complement and FcγRs are neces-
their infiltration into tissues (Geng et al. 1990), while activated pro- sary for a fully developed Arthus reaction.
teases cleave core proteoglycan, resulting in the release of heparin Xenotransplantation has also been used extensively to study
sulphate. In vitro stimulation of endothelial cells with C5a results in complement participation in acute vascular injury. An organ trans-
induction of a number of genes encoding cell adhesion molecules, planted into a recipient of another species is subject to hypera-
including E-selectin, ICAM-1, and V-CAM, and of cytokines and cute rejection, a severe reaction that destroys the xenograft within
their receptors, such as IL-6/Il-6R and IL-18R (Albrecht et al. 2004). minutes to a few hours and is characterized by local ischaemia,
Insertion of sublytic numbers of the MAC into the endothelial endothelial swelling, interstitial haemorrhage, oedema, and intra-
cell membrane also has multiple effects (Acosta et al. 2004). One vascular coagulation (Platt 2000). Because of the possibility that
of the most significant structural effects is the disruption of inter- it may represent a broader set of vascular diseases, there has been
cellular junctions, leading to formation of gaps and disruption of intense interest in understanding how hyperacute rejection arises
the barrier function of the endothelium (Saadi and Platt 1995). (Saadi et al. 2004). There is considerable evidence that the rejection
Another important consequence of MAC insertion is the induction is initiated by natural IgM antibodies against endothelial cells that
of the IL-1α gene and the secretion of this cytokine, which acts in activate the classical complement pathway. In primates, the main
an autocrine fashion to mediate expression of E-selectin, plasmino- xenoreactive antibodies against endothelial cells of a xenograft are
gen activator inhibitor-1, and tissue factor (Saadi et al. 1995; Acosta directed against Galα1-3Gal, a disaccharide expressed by lower
et  al. 1996). Additional cytokines and growth factors, including mammals but not primates (Lin et al. 1997). Hyperacute rejection
monocyte chemotactic protein-1 (Acosta et al. 2004), basic fibro- also occurs in allotransplantation when the recipient is presensi-
blast growth factor, and platelet-derived growth factor (Benzaquen tized to donor MHC antigens. The dominant role of complement
et al. 1994) are secreted following insertion of sublytic MAC. The in the development of the reaction has been studied extensively by
formation of MAC in endothelial cell membranes also initiates several methods, including depleting the recipient’s complement
secretion of the endothelial storage pool of von Willebrand factor using cobra venom factor (Leventhal et al. 1993), using comple-
(Hattori et al. 1989) and promotes release of plasma membrane vesi- ment inhibitors such as soluble CR1 (Pruitt et al. 1994), or using
cles that bind coagulation factor Va and expose a catalytic surface animals genetically deficient in complement proteins (Brauer et al.
for assembly of prothrombinase (Hamilton et al. 1990). In addition, 1995). Attempts at transplanting pig organs into primates empha-
MAC induces rearrangement of membrane phospholipids, leading sized the role of the cell-membrane-associated regulatory proteins
to increased presence in the outer leaflet of the bilayer of phosphati- in protecting endothelial cells from complement damage. The pig
dyl serine, which may promote plasma clotting by increasing tissue xenografts were subject to hyperacute rejection in part because
factor activity (Sims and Wiedmer 1995). Combined, the pleiotropic of incompatibility between pig DAF, MCP, and CD59 with pri-
effects of complement activation products lead to structural damage mate complement (Dalmasso et al. 1991) or alternatively because
of the vascular endothelium associated with inflammatory, prolif- of inadequate numbers of these molecules on pig endothelium
erative, and thrombotic reactions, commonly seen in vasculitis. (Morgan et al. 2005). To overcome this problem, pigs transgenic
for human DAF and/or CD59 were generated (Pascher et al. 1997;
Animal models of complement-induced Storck et al. 1997; Byrne et al. 1997). Organs from these transgenic
pigs transplanted into non-human primates survive much longer
vascular damage than organs from non-transgenic animals.
The study of the in vivo damaging effects of complement activation Animal models of ischaemia/reperfusion (I/R) injury have been
on the vasculature has been made possible by the use of animal mod- used extensively to investigate the role of complement in the vascular
els. The oldest such model is the Arthus reaction (Arthus 1903), in injury associated with this syndrome. I/R injury is an acute inflam-
which antibodies in blood encounter their intradermally injected matory response at the level of the vascular endothelium occur-
cognitive antigen at the level of the vascular wall. Antibodies are ring after an ischaemic event and subsequent restoration of blood
in large excess, so large insoluble immune complexes are formed flow. I/R injury is a major participant in many clinical conditions,
CHAPTER 5 complement in vasculitis 57

including myocardial infarction, stroke, intestinal ischaemia, cardi- Ames, R.S., Li, Y., Sarau, H.M., et al. (1996). Molecular cloning and charac-
ovascular surgery, and trauma (Hart et al. 2004). Evidence derived terization of the human anaphylatoxin C3a receptor. Journal of Biological
mainly from experimental animal models has indicated that com- Chemistry, 271, 20231–4.
plement activation plays a major role in the pathogenesis of I/R Arthus, M. (1903). Injections repetees de serum de cheval chez le lapin.
Comptes Rendus des Seances de la Societe de Biologie et de ses Filiales,
injury of the myocardial, skeletal, cerebral, renal, and intestinal tis- 55, 817–25.
sues (Weisman et  al. 1990:  Weiser et  al. 1996; Zhao et  al. 2002). Atkinson, J.P., Oglesby, T.J., White, D., Adams, E.A., and Liszewski, M.K.
Activation of the classical pathway is initiated by natural antibodies (1991). Separation of self from non-self in the complement system: a
reacting with endothelial cells altered by oxidative stress (Weiser role for membrane cofactor protein and decay accelerating factor.
et al. 1996; Zhang et al. 2004). Studies using animals deficient in Clinical Experimental Immunology, 86 (Suppl. 1), 27–30
complement proteins have demonstrated that the lectin pathway Baumann, U., Chouchakova, N., Gewecke, B., et al. (2001). Distinct tissue
also plays a dominant role in complement activation in I/R injury site-specific requirements of mast cells and complement components
(Walsh et al. 2005; Hart et al. 2005), while the alternative pathway C3/C5a receptor in IgG immune complex-induced injury of skin and
lung. Journal of Immunology, 167, 1022–7.
is necessary for full development of the injury (Stahl et al. 2003).
Benzaquen, L.R., Nicholson-Weller, A., Halperin, J.A. (1994). Terminal
complement proteins C5b-9 release basic fibroblast growth factor
and platelet-derived growth factor from endothelial cells. Journal of
Complement in clinical vasculitis Experimental Medicine, 179, 985–92.
Complement has been implicated in the pathogenesis of many vas- Brauer, R.B., Baldwin, W.M. 3rd, Ibrahim, S., Sanfilipo, F. (1995). The con-
culitic syndromes in man, although supporting evidence has not tribution of terminal complement components to acute and hyperacute
always been robust or conclusive. It is, however, generally accepted allograft rejection in the rat. Transplantation, 59, 288–93.
that complement plays a significant role in the pathogenesis of the Byrne, G.W., McCurry, K.R., Martin, M.J., McClellan, S.M., Platt, J.L.,
vasculitides associated with antineutrophil cytoplasmic autoanti- and Logan, J.S. (1997). Transgenic pigs expressing human CD59
and decay-accelerating factor produce an intrinsic barrier to
bodies (ANCA), which are characterized by necrotizing inflamma-
complement-mediated damage. Transplantation, 63, 149–53.
tion of small vessels (Wilde et al. 2011; Kallenberg 2011). Pathogenic Carroll, M.C. (2004). The complement system in regulation of adaptive
ANCA are directed against proteinase 3 or myeloperoxidase and immunity. Nature Immunology, 5, 981–6.
the associated vasculitides comprise granulomatosis with polyangi- Chen, N.-J., Mirtsos, C., Suh, D., et al. (2007). C5L2 is critical for the biologi-
itis (formerly known as Wegener’s granulomatosis), microscopic cal activities of the anaphylatoxins C5a and C3a. Nature, 446, 203–7
polyangiitis, idiopathic necrotizing crescentic glomerulonephri- Claudy, A. (1998). Pathogenesis of leukocytoclastic vasculitis. European
tis, and eosinophilic granulomatosis with polyangiitis (formerly Journal of Dermatology, 8, 75–9.
known as Churg–Strauss syndrome). It has been proposed that Dalmasso, A.P., Vercellotti, G.M., Platt, J.L., and Bach, F.H. (1991). Inhibition
factors released by ANCA-activated neutrophils activate the alter- of complement-mediated endothelial cell cytotoxicity by decay acceler-
ating factor: potential for prevention of xenograft hyperacute rejection.
native pathway of complement, resulting in enhanced recruitment
Transplantation, 52, 530–3.
and activation of neutrophils (Xiao et  al. 2007). Available data
Danning, C.L., Illei, G.G., and Boumpas, D.T. (1998). Vasculitis associated
indicate that the pathogenic effects of complement are mediated with primary rheumatologic diseases. Current Opinion in Rheumatology,
by the anaphylatoxin C5a produced during cleavage/activation of 10, 58–65.
C5. Binding of C5a to the C5a receptor (C5aR) expressed on neu- Davies, K.A. and Walport, M.J. (1998). Processing and clearance of immune
trophils up-regulates the membrane expression of ANCA antigens complexes by complement and the role of complement in immune
and primes neutrophils for a subsequent ANCA-induced respira- complex diseases. In The Human Complement System in Health and
tory burst (Schreiber et al. 2009; van Timmeren et al. 2009). Disease (ed. J.E. Volanakis and M.M. Frank), pp. 423–53. Marcel Dekker,
Complement has also been reported to participate in the New York.
Davis, A.E. 3rd (2005). The pathophysiology of hereditary angioedema.
pathogenesis of the vasculitis associated with cryoglobulinaemia
Clinical Immunology, 114, 3–9.
(Sansonno and Dammacco 2005), Henoch–Schönlein purpura
Davis, A.E. 3rd, Lu, F., and Mejia, P. (2010). C1 inhibitor, a multi-functional
(Kawana et  al. 1990), diabetes (Acosta et  al. 2000), rheumatoid serine protease inhibitor. Thrombosis and Haemostasis, 104, 886–93.
arthritis (Danning et al. 1998), and leukocytoclastic (hypersensitiv- Dragon-Durey, M.-A. and Fremeaux-Bacchi, V. (2005). Atypical haemo-
ity) vasculitis (Claudy 1998). lytic uraemic syndrome and mutations in complement regulator genes.
Springer Seminars in Immunopathology, 27, 359–74.
References Esser, A.F. (1994). The membrane attack complex of complement. Assembly,
Acosta, J.A., Benzaquen, L.R., Goldstein, D.J., Tosteson, M.T., and Halperin, structure, and cytotoxic activity. Toxicology, 87, 229–47.
J.A. (1996). The transient pore formed by homologous terminal com- Fearon, D.T. and Austen, K.F. (1980). The alternative pathway of comple-
plement complexes functions as a bidirectional route for the trans- ment:  a system for host defense of microbial infection. New England
port of autocrine and paracrine signals across human cell membranes. Journal of Medicine, 303, 259–63.
Molecular Medicine, 2, 755–65. Fearon, D.T. and Locksley, R.M. (1996). The instructive role of innate immu-
Acosta, J., Hetinga, J., Fluckiger, R., et al. (2000). Molecular basis for a link nity in the acquired immune response. Science, 272, 50–4.
between complement and the vascular complications of diabetes. Foreman, K.E., Vaporciyan, A.A., Bonish, B.K., et al. (1994). C5a-induced
Proceedings of the National Academy of Sciences USA, 97, 5450–5. expression of P-selectin in endothelial cells. Journal of Clinical
Acosta, J., Qin, X., and Halperin, J. (2004). Complement and complement Investigation, 94, 1147–55.
regulatory proteins as potencial molecular targets for vascular diseases. Gaboriaud, C., Thielens, N.M., Gregory, L.A., Rossi, V., Fontecilla-Camps, J.C.,
Current Pharmaceutical Design, 10, 203–11. and Arlaud, G.J. (2004). Structure and activation of the C1 complex of
Albrecht, E.A., Chinnaiyan, A.M., Varambally, S., et al. (2004). C5a-induced complement: unraveling the puzzle. Trends in Immunology, 25, 368–73.
gene expression in human umbilical vein endothelial cells. American Genel, F., Sjoholm, A.G., Skattum, L., and Truedsson, L. (2005). Complement
Journal of Pathology, 164, 849–59. factor I  deficieny associated with recurrent infections, vasculitis and
58 SECTION 2 basic science

immune complex glomerulonephritis. Scandinavian Journal of Infectious Lin, S.S., Kooyman, D.L., Daniels, L.J., et al. (1997). The role of natural anti-Gal
Diseases, 37, 615–18. alpha 1-3Gal antibodies in hyperacute rejection of pig-to-baboon car-
Geng, J.G., Bevilacqua, M.P., Moore, K.L., et  al. (1990). Rapid neutrophil diac xenotransplants. Transplant Immunology, 5, 212–18.
adhesion to activated endothelium mediated by GMP-140. Nature, 343, Liszewski, M., Farries, T., Lublin, D., Rooney, I., and Atkinson, J. (1996).
757–60. Control of the complement system. Advances in Immunology, 61,
Gerard, N.P. and Gerard, C. (1991). The chemotactic receptor for human C5a 201–83.
anaphylatoxin. Nature, 349, 614–17. Liszewski, M.K., Kemper, C., Price, J.D., and Atkinson, J.P. (2005). Emerging
Hamilton, K.K., Hattori, R., Esmon, C.T., and Sims, P.J. (1990). Complement roles and new functions of CD46. Springer Seminars in Immunopathology,
proteins C5b-9 induce vesiculation of the endothelial plasma membrane 27, 345–58.
and expose catalytic surface for assembly of the prothrombinase enzyme Lukacik, P., Roversi, P., White, J., et al. (2004). Complement regulation at the
complex. Journal of Biological Chemistry, 265, 3809–14. molecular level: The structure of decay-accelerating factor. Proceedings
Hart, M.L., Ceonzo, K.A., Shaffer, L.A., et  al. (2005). Gastrointestinal of the National Academy of Sciences USA, 101, 1279–84.
ischemia-reperfusion injury is lectin complement pathway dependent Mastellos, D., Germenis, A.E., and Lambris, J.D. (2005). Complement:  an
without involving C1q. Journal of Immunology, 174, 6373–80. inflammatory pathway fulfilling multiple roles at the interface of innate
Hart, M.L., Walsh, M.C., and Stahl, G.L. (2004). Initiation of complement immunity and development. Current Drug Targets Inflammation and
activation following oxidative stress. In vitro and in vivo observations. Allergy, 4, 125–7.
Molecular Immunology, 41, 165–71. Matsushita, M. and Fujita, T. (2001). Ficolins and the lectin complement
Hattori, R., Hamilton, K.K., McEver, R.P., and Sims, P.J. (1989). Complement pathway. Immunological Reviews, 180, 78–85.
proteins C5b-9 induce secretion of high molecular weight multim- Monk, P.N., Scola, A.-M., Madala, P., and Fairlie, D.P. (2007). Function,
ers of endothelial von Willebrand factor and translocation of granule structure and therapeutic potential of complement C5a receptors.
membrane protein GMP-140 to the cell surface. Journal of Biological British Journal of Pharmacology, 152, 429–48.
Chemistry, 264, 9053–60. Morgan, B.P. (1999). Regulation of the complement membrane attack path-
Haviland, D.L., McCoy, R.L., Whitehead, W.T., et al. (1995). Cellular expres- way. Critical Reviews in Immunology, 19, 173–98.
sion of the C5a anaphylatoxin receptor (C5aR): demonstration of C5aR Morgan, B.P., van den Berg, C.W., and Harris, C.L. (2005). ‘Homologous
on nonmyeloid cells of the liver and lung. Journal of Immunology, 154, restriction’ in complement lysis: roles of membrane complement regula-
1861–9. tors. Xenotransplantation, 12, 258–65.
Haynes, T., Luz-Madrigal, A., Reis, E.S., et al. (2013). Complement anaphyla- Ohno, M., Hirata, T., Enomoto, M., Araki, T., et al. (2000). A putative che-
toxin C3a is a potent inducer of embryonic chick retina regeneration. moattractant receptor, C5L2, is expressed in granulocyte and immature
Nature Communications 4:2312. dendritic cells, but not in mature dendritic cells. Molecular Immunology,
Helmy, K.Y., Katschke, K.J. Jr, Gorgani, N.N. et  al. (2006). CRIg:  A  mac- 37, 407–12.
rophage complement receptor required for phagocytosis of circulating Owen, C.A., Campbell, M.A., Sannes, P.L., Boukedes, S.S., and Campbell, E.J.
pathogens. Cell, 124, 915–27. (1995). Cell surface bound elastase and cathepsin G on human neutro-
Holmskov, U., Thiel, S., and Jensenius, J.C. (2003). Collectins and phils: a novel non-oxidative mechanism by which neutrophils focus and
Ficolins: Humoral lectins of the innate immune defense. Annual Review preserve catalytic activity of serine proteinases. Journal of Cell Biology,
of Immunology, 21, 547–78. 131, 775–89.
Hopken, U.E., Lu, B., Gerard, N.P., and Gerard, C. (1997). Impaired inflam- Pangburn, M.K. and Müller-Eberhard, H.J. (1984). The alternative pathway
matory responses in the reverse Arthus reaction through genetic dele- of complement. Springer Seminars in Immunopathology, 7, 163–92.
tion of the C5a receptor. Journal of Experimental Medicine 186, 749–56. Pascher, A., Poehlein, C., Storck, M., et al. (1997). Immunological observa-
Huang, Y., Smith, C.A., Song, H., Morgan, B.P., Abagyan, R., and Tomlinson, tions after xenogeneic liver perfusions using donor pigs transgenic for
S. (2005). Insights into human CD59 complement binding interface human decay-accelerating factor. Transplantation, 64, 384–91.
toward engineering new therapeutics. Journal of Biological Chemistry, Platt, J.L. (2000). Immunobiology of xenotransplantation. Transplantation
280, 34073–9. International, 13 (Suppl. 1), S7–10.
Ihrcke, N.S. and Platt, J.L. (1996). Shedding of heparin sulfate proteoglycan Plummer, T.H. Jr and Hurwitz, N.Y. (1978). Human plasma carboxypepti-
by stimulated endotheliasl cells: evidence for proteolysis of cell surface dase N. Journal of Biological Chemistry, 253, 3907–12.
molecules, Journal of Cell Physiology, 168, 625–37. Podack, E.R. and Tschopp, J. (1984). Membrane attack by complement.
Kallenberg, C.G.M. (2011). Pathogenesis of ANCA-associated vasculitides. Molecular Immunology, 21, 589–603.
Annals of Rheumatic Diseases, 70 (Suppl. 1), i59–63. Pruitt, S.K., Kirk, A.D., Bolinger, R.R., et al. (1994). The effect of soluble com-
Kawana, S., Shen, G.H., Kobayashi, Y., and Nishiyama, S. (1990). Membrane plement receptor type 1 on hyperacute rejection of porcine xenografts.
attack complex of complement in Henoch-Schoenlein purpura skin and Transplantation, 57, 363–70.
nephritis. Archives of Dermatological Research, 282, 183–7. Rus, H.G., Niculescu, F.I., and Shin, M.L. (2001). Role of the C5b-9 com-
Key, N.S., Platt, J.L., and Vercellotti, G.M. (1992). Vascular endothelial cell plement complex in cell cycle and apoptosis. Immunological Reviews,
proteoglycans are susceptible to cleavage by neutrophils. Arteriosclerosis 180, 49–55.
and Thrombosis, 12, 836–42. Saadi, S., Holzkhecht, R.A., Patte, C.P., Stern, D.M., and Platt, J.L. (1995).
Kinoshita, T., Takata, Y., Kozono, H., Takeda, J., Hong, K.S., and Inoue, K. Complement-mediated regulation of tissue factor activity in endothe-
(1988). C5 convertase of the alternative complement pathway: covalent lium. Journal of Experimental Medicine, 182, 1807–14.
linkage between two C3b molecules within the trimolecular complex Saadi, S. and Platt, J.L. (1995). Transient perturbation of endothelial integ-
enzyme. Journal of Immunology, 141, 3895–901. rity induced by antibodies and complement. Journal of Experimental
Klos, A., Tenner, A.J., Johswich, K.O., et al. (2009). The role of anaphylatoxins Medicine, 181, 21–31.
in health and diseade. Molecular Immunology, 46, 2753–66. Saadi, S., Takahashi, T., Holzknecht, R.A., and Platt, J.L. (2004). Pathways to
Lalli, P.N., Strainic, M.G., Yang, M., et al. (2008). Locally produced C5a binds acute humoral rejection. American Journal of Pathology, 164, 1073–80.
to T cell-expressed C5aR to enhance effector cell expansion by limiting Sacks SH (2010). Complement fragments C3a and C5a: the salt and pepper
antigen-induced apoptosis. Blood, 112, 1759–66 of the immune response. European Journal of Immunology, 40, 668–70.
Leventhal, J.R., Dalmasso, A.P., Cromwell, J.W., et al. (1993). Prolongation of Sansonno, D. and Dammacco, F. (2005). Hepatitis C virus, cryoglobuli-
cardiac xenograft survival by depletion of complement. Transplantation, naemia, and vasculitis:  immune complex relations. Lancet Infectious
55, 857–66. Diseases, 5, 227–36.
CHAPTER 5 complement in vasculitis 59

Schreiber, A., Xiao, H., Jennette, J.C., et al. (2009). C5a receptor mediates Volanakis, J.E. (2005). The molecular biology of the complement system.
neutrophil activation and ANCA-induced glomerulonephritis. Journal In Arthritis and allied conditions—a textbook of rheumatology (ed. W.J.
of the American Society of Nephrology, 20, 289–98. Koopman and L.W. Moreland) (15th edn), pp. 490–503. Williams &
Shushakova, N., Skokowa, J., Schulman, J., et al. (2002). C5a anaphylatoxin Wilkins Co., Maryland.
is a major regulator of activating versus inhibitory FcγRs in immune Walport, M.J. (2001). Complement. New England Journal of Medicine, 344,
complex-induced lung disease. Journal of Clinical Investigation, 110, 1058–66; 1140–4.
1823–30. Walsh, M.C., Bourcier, T., Takahashi, T., et al. (2005). Mannose-binding lec-
Sims, P.J. and Wiedmer, T. (1995). Induction of cellular procoagulant activ- tin is a regulator of inflammation that accompanies myocardial ischemia
ity by the membrane attack complex of complement. Seminars in Cell and reperfusion injury. Journal of Immunology, 175, 541–6.
Biology, 6, 275–82. Ward, P.A. (2009). Functions of C5a receptors. Journal of Molecular Medicine
Stahl, G.L., Xu, Y., Hao, L., et al. (2003). Role for the alternative complement (Berlin), 87, 375–8.
pathway in ischemia/reperfusion injury. American Journal of Pathology, Weiser, M.R., Williams, J.P., Moore, F.D. Jr, Kobzik, I., Ma, M., Hechtman,
162, 449–55. H.B., and Carroll, M.C. (1996). Reperfusion injury of ischemic skeletal
Storck, M., Abendroth, D., Prestel, R., et al. (1997). Morphology of hDAF muscle is mediated by natural antibody and complement. Journal of
(CD55) transgenic pig kidneys following ex-vivo hemoperfusion with Experimental Medicine, 183, 2343–8.
human blood. Transplantation, 63, 304–10. Weisman, H.F., Bartow, T., Leppo, M.K., et  al. (1990). Soluble human com-
Strainic, M.G., Liu, J., Huang, D., et al. (2008). Locally produced complement plement receptor type 1:  In vivo inhibitor of complement suppress-
fragments C5a and C3a provide both costimulatory and survival signals ing post-ischemic myocardial inflammation and necrosis. Science, 249,
to naive CD4+ T cells. Immunity, 28, 425–35. 146–51.
Sylvestre, D., Clynes, R., Ma, M., Warren, H., Carroll, M., and Ravetch, J. Wilde, B., van Paassen, P., Witzke, O., and Tervaert, J.W. (2011). New patho-
(1996). Immunoglobulin G-mediated inflammatory responses develop physiological insights ajnd treatment of ANCA-associated vasculitis.
normally in complement-deficient mice. Journal of Experimental Kidney International, 79, 599–612.
Medicine, 184, 2385–92. Xiao, H., Schreiber, A., Heeringa, P., et al. (2007). Alternative complement
Takata, Y., Kinoshita, T., Kozono, H., et al. (1987). Covalent association of pathway in the pathogenesis of disease mediated by anti-neutrophil
C3b with C4b within C5 convertase of the classical complement path- cytoplasmic autoantibodies. American Journal of Pathology,
way. Journal of Experimental Medicine, 165, 1494–507. 170, 52–64.
Takeda, J., Miyata, T., Kawagoe, K., et al. (1993). Deficiency of the GPI anchor Zhang, M., Austen, W,G. Jr, Chiu, I., et al. (2004). Identification of a specific
caused by a somatic mutation of the PIG-A gene in paroxysmal noctur- self-reactive IgM antibody that initiates intestinal ischemia/ reperfu-
nal hemoglobinuria. Cell, 73, 703–11. sion injury. Proceedings of the National Academy of Sciences USA, 101,
van Timmeren, M.M., Chen, M., and Heeringa, P. (2009). Review arti- 3886–91.
cle: pathogenic role of complement activation in anti-neutrophil cyto- Zhao, H., Montalto, M.C., Pfeiffer, K.J., Hao, L., and Stahl, G.L. (2002).
plasmic autoantibody-associated vasculitis. Nephrology, 14, 16–25. Murine model of gastrointestinal ischemia associated with
Volanakis, J.E. (1998). Overview of the complement system. In The human complement-dependent injury. Journal of Applied Physiology, 93,
complement system in health and disease (ed. J.E. Volanakis and M.M. 338–45.
Frank), pp. 9–32. Marcel Dekker, New York.
CHAPTER 6

Autoantibodies in vasculitis
Jan Damoiseaux and Jan Willem Cohen Tervaert

Introduction Tervaert et  al. 1989a; Falk and Jennette 1989). Myeloperoxidase
(MPO) appeared as the antigen that was most often responsible for
The so-called primary vasculitides comprise a wide array of dis- the pANCA pattern in sera of patients with SVV (Cohen Tervaert
eases, which are classified according the size of the blood vessels et al. 1989a; Falk and Jennette 1988; Falk and Jennette 1989).
that are primarily affected (Jennette et al. 2013). Because the blood Because ANCA have been shown to be closely associated with
vessels are the target of the chronic inflammatory response, the SVV, in animal studies have proven to have pathogenic potential
first searches for autoantibodies focused on structural compo- (Xiao et al. 2002; Little et al. 2012), and relate to the clinical phe-
nents of these blood vessels and in particular on endothelial cells. notype of these conditions, ANCA is the main topic of this chapter.
These antiendothelial antibodies (AECA) have been detected in Although PR3 and MPO are considered the main target antigens,
both small- and large-vessel vasculitis (reviewed by Belizna and some other antigen specificities will be discussed in terms of poten-
Cohen Tervaert 1997). Their presence, however, is not restricted to tial relevance to SVV.
the vasculitides, but is also observed in connective tissue diseases
and other inflammatory diseases. Moreover, methods used for the Terminology for antineutrophil
detection of AECA are very heterogeneous and the antigens rec-
ognized remain ill defined. Using novel methods for antigen iden- cytoplasmic antibodies
tification, such as proteomics, the antigenic repertoire recognized Although originally described as neutrophil-specific autoantibod-
by AECA in vasculitis is beginning to be elucidated (Régent et al. ies (NSA), granulocyte-specific antinuclear antibodies (GS-ANA),
2011). For reasons mentioned above in this paragraph, AECA do and anticytoplasmic antibodies (ACPA), the term ANCA was even-
not play a role in differential diagnosis. tually coined in 1989 by Wiik and Van der Woude (Wiik and Van
The observation that antineutrophil cytoplasmic antibodies der Woude 1990). Next, because distinct patterns can be described
(ANCA) are associated with a subset of the primary vasculitides, for ANCA as detected by IIF on ethanol-fixed neutrophils
that is the small-vessel vasculitides (SVV), including granulomatosis (Figure 6.1a), the nomenclature for these patterns has been defined
with polyangiitis (GPA; formerly called Wegener’s granulomatosis), in the international consensus statement on testing and reporting
microscopic polyangiitis (MPA), eosinophilic granulomatosis with of ANCA (Savige et  al. 1999). Basically, four patterns have been
polyangiitis (EGPA; formerly called Churg–Strauss syndrome), and defined: first, the classical (c)ANCA is characterized by a granu-
renal-limited necrotizing crescentic glomerulonephritis (NCGN), lar, cytoplasmic fluorescence with central or interlobular accentua-
has contributed enormously to the implementation of the use of tion (Figure 6.1b); second, a diffuse flat cytoplasmic fluorescence
these autoantibodies in the diagnosis and follow-up of patients with without interlobular accentuation may be referred to as atypical
these diseases. ANCA were initially detected by an indirect immu- cANCA; third, the perinuclear (p)ANCA is characterized by peri-
nofluorescence technique (IIF) in patients with leukopenia and nuclear staining, with or without nuclear extension (Figure 6.1c);
various inflammatory disorders such as rheumatoid arthritis, ulcer- and fourth, if a combination of cytoplasmic and perinuclear stain-
ative colitis, and chronic hepatitis (Calabresi et al. 1959; Calabresi ing occurs due to the presence of multiple specificities, this is called
et  al. 1961; reviewed in Cohen Tervaert and Damoiseaux 2009). atypical ANCA. In clinical practice, however, it appears that the
The relationship of autoantibodies to cytoplasmic constituents of atypical cANCA pattern is not distinguished by many clinical labo-
granulocytes with vasculitis was first reported in a few patients with ratories. Furthermore, reading of the pANCA pattern may be ham-
glomerulonephritis (Davies et al. 1982). Next, similar autoantibod- pered by the presence of interfering antinuclear antibodies (ANA).
ies were detected by IIF on ethanol-fixed neutrophils in patients This interference may be solved by using formalin-fixed neutro-
with GPA. These antibodies revealed a characteristic cytoplasmic phils as a substrate for IIF, in addition to the ethanol-fixed neutro-
fluorescence pattern referred to as cANCA (Van der Woude et al. phils. The perinuclear staining pattern on ethanol-fixed neutrophils
1985). The antigen responsible for this cANCA pattern was identi- actually is an artefact, because formalin-fixation reveals a cytoplas-
fied as a 29-kDa serine protease from myeloid azurophilic granules, mic staining pattern similar to a classical cANCA on ethanol-fixed
that is proteinase 3 (PR3) (Goldschmeding et al. 1989; Niles et al. neutrophils. Finally, in the literature the term xANCA, or atypi-
1989; Jenne et al. 1990). Subsequently, it appeared that in vasculitis cal pANCA, is often reported and refers to an ANCA detectable in
patients an alternative fluorescence pattern could also be observed. various non-SVV autoimmune diseases such as ulcerative colitis,
Instead of cytoplasmic staining of the ethanol-fixed neutrophils, a autoimmune hepatitis, and primary sclerosing cholangitis (Terjung
perinuclear fluorescence pattern (pANCA) was revealed (Cohen et al. 2001). Typically, this atypical pANCA is characterized by a
62 SECTION 2 basic science

(a) Indirect Immunofluorescence addition to MPO and PR3, cathepsin G, catalase, α-enolase, elastase,
lysozyme, lactoferrin, azurocidin, and BPI. More recently, the
lysosome-associated membrane protein (LAMP)-2 has been added
+ to the list of ANCA targets (Kain et al. 1995). In this chapter the
neutrophil test serum specificities described are considered to be relevant for AAV, that is
(ethanol fixed)
PR3, MPO, elastase, BPI, and LAMP-2. In the course of the disease,
due to epitope spreading and subsequent antigen spreading, ANCA
to the other target antigens may be revealed (Talor et al. 2007).

+ Proteinase 3 (PR3)
cell-bound anti-human
antibodies immunoglobulin PR3-ANCA were originally described reacting with a 29-kDa
serine protease in azurophilic granules isolated from the alpha
(b) cANCA (c) pANCA fraction of neutrophils, and from supernatants of degranu-
lated neutrophils. This proteolytic enzyme was identified as PR3
(Goldschmeding et al. 1989; Niles et al. 1989; Jenne et al. 1990). The
gene encoding the weak cationic PR3 is located on chromosome
19 (Sturrock et al. 1992), close to the gene encoding elastase and
azurocidin (Jenne 1994), two neutral serine protease homologues
located in the azurophilic granules. Cloning of cDNA for PR3 was
accomplished in 1990 (Campanelli et  al. 1990). The PR3 gene is
Fig. 6.1 Antineutrophil cytoplasmic antibodies (ANCA) detected by indirect
mainly expressed during the early myelocyte maturation stage of
immunofluorescence (IIF). Slides of ethanol-fixed neutrophils were incubated bone marrow development (Sturrock et  al. 1992; Müller-Bérat
with patient serum and visualized by FITC-labelled anti-human immunoglobulin et al. 1994). Enzymatically active PR3, derived after cleavage of its
G (a). Fluorescent microscopy may reveal distinct patterns, such as cANCA (b) or N-terminal activation dipeptide, consists of 228 amino acids which
pANCA (c). Figure 6.1a is adapted from: Damoiseaux, J., Austen, J., and Cohen form a highly folded protein with four disulphide bridges, keeping
Tervaert, J.W. (2011). ANCA diagnostics in clinical practice: new developments. its three-dimensional structure intact (Fujinaga et al. 1996). Most
In: Advances in the diagnosis and treatment of vasculitis. Ed. Amezcua-Guerra, L.M. of the biological functions ascribed to PR3 are dependent on its
Intech, Croatia, pp. 3–18.
proteolytic activity. The primary function of the neutrophil-derived
serine proteases is thought to be the degradation of extracellular
broad inhomogeneous rim-like staining of the nuclear periphery, matrix proteins at the site of inflammation (reviewed by Radice
often with multiple nuclear foci. The target antigens of atypical and Sinico 2005). In addition, the proteolytic activity of PR3 may
pANCA have not been unequivocally identified, but are rather also modulate the activity of platelets, endothelial cells, and inflam-
nuclear than cytoplasmic antigens and are formalin sensitive. In matory mediators. The enzymatic activity of PR3 is physiologically
general, it is thought that these atypical pANCA are not directed inhibited by α1-antitrypsin.
to the antigens relevant for SVV, that is MPO and PR3, but there The ANCA reactivity depends on the native conformation of the
are some studies that report reactivity to PR3 in a subset of patients PR3 molecule (Bini et al. 1992; Specks et al. 1996). This emphasizes
with ulcerative colitis (Elzouki et al. 1999; Xu et al. 2008). the importance of the way the antigen is presented in PR3-ANCA
The international consensus statement has defined the nomen- detection assays. Most human PR3-ANCA seem to target epitopes
clature for the antigen specificity of ANCA (Savige et  al. 1999). close to the catalytic site where four out of five linear epitopes have
These autoantibodies are to be described as MPO-ANCA and been described (Griffith et al. 2001). Characterization of epitopes
PR3-ANCA for the common specificities in SVV or, for instance, using mouse monoclonal antibodies indicated the presence of at
as bactericidal/ permeability-increasing protein (BPI)-ANCA and least four different epitopes (Van der Geld et  al. 1999). The rel-
elastase-ANCA for the miscellaneous specificities. Importantly, evance of epitope mapping, however, is highly dependent on the
terms such as antiproteinase 3 antibodies should be restricted to technology used. In a more recent publication, it was calculated
antibodies that have been produced for research purposes, for that B-cell epitopes almost exclusively consist of conformational
example monoclonal antibodies. epitopes instead of linear epitopes (Sivalingam and Shepherd
Finally, because ANCA augment clinical diagnosis of necrotiz- 2012). Due to problems with expression and purification proce-
ing SVV and rational decision making in the clinic (Franssen et al. dures, application of recombinant PR3 in clinical practice has been
2000; Choi et  al. 2001; Cohen Tervaert and Damoiseaux 2012), limited (Jenne and Kuhl 2006; Damoiseaux et al. 2009a). Because
these conditions are now referred to as ANCA-associated vascu- most PR3-ANCA react with the enzymatically active PR3 after
litis (AAV). Since the establishment of animal models for AAV cleavage of its N-terminal activation dipeptide (Sun et al. 1998),
(Xiao et al. 2002; Little et al. 2012), these diseases are referred to as recombinant PR3 requires correct post-translational processing to
ANCA-induced vasculitis or ANCA vasculitis. obtain the proper conformation.

Myeloperoxidase (MPO)
ANCA Targets in small-vessel vasculitides In the late 1980s, several authors described the presence of ANCA
Because ANCA are primarily directed against the contents of the directed to MPO in sera from patients with SVV (Cohen Tervaert
azurophilic granules, the recognized antigens consist of a whole array et al. 1989a; Falk and Jennette 1988; Falk and Jennette 1989). The
of enzymes, which are often catalytic. These constituents include, in highly cationic MPO is encoded on chromosome 17 (17q23.1) next
CHAPTER 6 autoantibodies in vasculitis 63

to the peroxidase genes eosinophil peroxidase and lactoperoxidase Kain et al. 2008). LAMP-2 (CD107b) is a heavily glycosylated type
(Chang et al. 1987). MPO transcripts are primarily detected in the 1 membrane protein of 95–120 kDa integrated in the membranes
myeloblasts and promyelocytes, whereas in later stages of granu- of MPO- and PR3-containing vesicles. LAMP-2 shuttles between
lopoiesis transcription subsides (Bjerregaard et  al. 2003; Cowland lysosomes and the cell surface and is involved in lysosomal enzyme
and Borregaard 1999). MPO, which has a molecular weight of about targeting, autophagy, and lysosomal biogenesis (reviewed by
140 kDa, is a homodimer consisting of two heavy and two light Eskelinen et al. 2003).
chains (reviewed by Malle et al. 2007). The enzymatic activity of MPO
catalyses the peroxidation of chloride into hypochlorite, important in Technical aspects of ANCA detection by IIF
intracellular killing of phagocytosed micro-organisms and in inacti-
vation of protease inhibitors. The latter contributes, like PR3, to the The method of choice for IIF demonstration of ANCA was agreed
degradation of extracellular matrix at the site of inflammation. upon at the First International ANCA Workshop (Wiik 1989) and
The antibody reactivity has been shown to depend on the native re-established in the international consensus statement on test-
conformation of MPO (Falk et al. 1992). The MPO molecule is quite ing and reporting of ANCA (Savige et  al. 1999). This method
unstable due to cleavage at a light-sensitive site located at the Met409 employed washed, isolated buffy coat cells free of platelets, smeared
position of the heavy chain (Taylor et al. 1992), and this property or cytocentrifuged onto glass slides, followed by ethanol fixation,
has great importance for preservation of the molecule as an antigen serum and conjugate incubation, and evaluation with an incident
for MPO-ANCA detection (Hagen et al. 1996). Although not yet light-illuminated fluorescence microscope (Figure 6.1). The use of
fully characterized, mapping of the molecule suggests a restricted whole buffy coat cells as substrate for ANCA detection allows dis-
number of conformational epitopes reactive with MPO-ANCA. crimination between neutrophil-specific antibodies and antibod-
Recombinant MPO seems to function just as well, or even better ies, such as ANA, that also recognize other leukocyte subsets. It is
than native MPO, as antigen for detection of MPO-ANCA (Short noteworthy that most laboratories nowadays use commercial slides
et al. 1995; Boomsma et al. 2001). which, in general, lack mononuclear leukocytes. The conjugate
should be specific for human IgG because anti-IgM and anti-IgA
Elastase conjugates may result in false-positive results. The method has been
Although elastase-ANCA are not prevalent in patients with AAV detailed in many recent publications and is available at <http://
(Cohen Tervaert et al. 1993), they may play a role in drug-induced www.vasculitis.org>. It is generally accepted that patient serum
vasculitis (Merkel 2001). In addition, elastase-ANCA can discrimi- should be diluted 1 : 20 for optimal detection, but one has to keep
nate between destructive lesions of the nasal septum due to GPA in mind that ANCA detection not only depends on the serum dilu-
or cocaine abuse (Trimarchi et al. 2001). Due to the adulteration of tion, but also on the quality of the substrate, the conjugate, and the
cocaine with levamisole, cutaneous vasculopathy may be observed fluorescence microscope.
in relation to elastase-ANCA (Pearson et al. 2012). Because elastase A draw-back of the IIF technique for ANCA detection is the need
is a member of the same chymotrypsin superfamily of serine pro- for experienced technicians to read the patterns on the slides. To
teases as PR3, these two catalytic enzymes have many structural and better distinguish the clinically relevant reactivities and to identify
functional characteristics in common (reviewed by Korkmaz et al. the presence of interfering ANA, slides with formalin-fixed neu-
2010). The gene, located on chromosome 19 (19p13.3), is primar- trophils and/or standard substrates for ANA (Hep-2 cells) can be
ily expressed at the promyelocytic stage of neutrophil maturation. run in parallel. It should be stressed, though, that in the case of a
The mature elastase protein consists of 218 amino acids, revealing positive IIF reaction, subsequent antigen specificity testing is man-
a protein of 29–33 kDa. datory (Savige et al. 1999). The biochip method permits combining
multiple substrates, even including antigen-specific substrates, in a
Bactericidal/permeability-increasing protein (BPI) single test incubation and thereby strongly facilitates optimal pat-
BPI-ANCA gives rise to a finely granular and often weak cyto- tern recognition (Damoiseaux et al. 2009b). The advent of micro-
plasmic staining pattern on ethanol-fixed neutrophils, most prob- scope devices that have integrated software for pattern recognition
ably because the molecule is anchored to the inside of azurophilic might further overcome the problem of the subjective reading of
granules. Because the cytoplasmic staining pattern resembles that the slides (Damoiseaux et al. 2012).
of PR3-ANCA, distinction of both antigen specificities may be Titration of IIF-positive samples is indicated if an interfering
clinically relevant. BPI-ANCA have been identified in different ANA or other cytoplasmic fluorescence (e.g. antibodies against
diseases associated with Gram-negative bacteria, such as inflam- Jo-1, mitochondria, or ribosomal nucleoprotein) is present in addi-
matory bowel diseases and primary sclerosing cholangitis (Stoffel tion to the ANCA, because one pattern may disappear before the
et al. 1996), and are frequently present in patients with cystic fibro- other (Savige et al. 1999). Higher titres, however, are also associ-
sis infected with Pseudomonas aeruginosa (Zhao et al. 1996). BPI ated with increased clinical relevance (Vermeersch et  al. 2009).
is a 55-kDa cationic protein present in the azurophilic granules of As for pattern recognition, automated image analyses can also be
neutrophils (reviewed by Holweg et al. 2011). BPI binds with high applied for quantification of ANCA levels in sera of AAV patients
affinity to lipopolysaccharides (LPS), enabling the efficient killing (Boomsma et al. 2003).
of Gram-negative bacteria and the neutralization of endotoxin.
Human lysosome-associated membrane Methods used to determine
protein 2 (hLAMP-2) ANCA specificity
Autoantibodies reactive with hLAMP-2 have been described in Antigen specificity of ANCA is generally determined by
patients with active phases of necrotizing SVV (Kain et al. 1995; enzyme-linked immunosorbent assays (ELISA). In most cases, a
64 SECTION 2 basic science

direct, non-competitive ELISA is the method of choice for detec- antigens. In these assays, the first coating of the solid phase does
tion of antigen-specific antibodies (Figure  6.2a) (Damoiseaux not consist of the antigen but of a monoclonal antibody specific
and Cohen Tervaert 2005). In this assay the antigen is solid-phase for the respective antigen. Next, the antigen preparation, either
bound to the microtitre plates. Because the antigen is directly as a crude neutrophil extract (Cohen Tervaert et al. 1990) or as
coated to the solid phase, this type of ELISA is referred to as a direct purified antigen, is added (Westman et al.1998; Boomsma et al.
ELISA. After coating of the antigen, diluted patient serum is incu- 2001). The antigen will be ‘captured’ by the bound monoclonal
bated to enable binding of ANCA, which is visualized by incuba- antibody and this will better consolidate the conformation of the
tion with an anti-human IgG reagent conjugated with an enzyme protein, enabling a better recognition by the ANCA potentially
and subsequently with the respective enzyme substrate. Because present in the patient serum. Detection of bound autoantibodies
the rate of colour formation is a function of the amount of autoan- is visualized by incubation with an anti-human IgG conjugate
tibody present in the serum sample, the ELISA is considered a true and, subsequently, the substrate. In theory, the capture ELISA
quantitative assay when a reference standard is available. This hap- shares better sensitivity, due to the better conformation of the
pens to be the case for both PR3- and MPO-ANCA (IUIS-CDC antigen, with better specificity, because of the additional purifi-
reference preparations) and these standards permit reporting of cation of the antigen within the assay. This happens to be the case
the results in international units. During the last decade, multiple for PR3-ANCA, but for MPO-ANCA only an increase in spec-
variants of the direct ELISA have become available. These assays ificity, and not in sensitivity, is observed (Csernok et  al. 2004;
may differ in terms of the carrier, for instance a membrane in Boomsma et al. 2001).
dot-blot assays (Rutgers et al. 2004) and line-immunoassays (LIA) The third-generation tests for ANCA detection comprise the
or beads in multiplex flow cytometry (Nifli et al. 2006; Damoiseaux recently developed anchor ELISAs (Figure 6.2c). In these assays,
et al. 2007; Kaul et al. 2009), or by replacement of the enzyme by the purified antigen is coupled to a linker peptide and this linker
a fluorochrome, as in fluorescent-enzyme immunoassays (FEIA) peptide is used to bind the antigen to the solid phase. Analogous
(Damoiseaux et al. 2005), as well as the Luminex-based technology to the capture ELISA, this also enables better maintenance of the
(Luminex, Austin, TX, USA) (Nifli et al. 2006; Damoiseaux et al. three-dimensional structure of the antigen, eventually resulting
2007; Kaul et  al. 2009). More recently, assays based on chemilu- in a better sensitivity. Indeed, a substantial increase in sensitivity
minescence technology have become available. Some of the novel for PR3-ANCA has been described, in particular in patients with
assays enable simultaneous detection of PR3- and MPO-ANCA. limited disease manifestations (Hellmich et al. 2007; Roggenbuck
On the other hand, the membrane-based assays only reveal qualita- et al. 2009; Damoiseaux et al. 2009a). These anchor ELISAs were
tive results, while the other assays share the possibility of obtaining first developed for PR3-ANCA only, but are now also avail-
quantitative results. able for MPO-ANCA. For the latter assay, no published data are
Second-generation tests for ANCA detection are so-called cap- available yet.
ture ELISAs (Figure 6.2b). Although these tests are referred to as
second generation, they were already available in the early days Use of ANCA results for prognosis
when MPO and PR3 were identified as the most important target
and diagnosis
The main reason to order an ANCA test is for diagnosing GPA,
(a) Direct ELISA
EGPA, MPA, or NCGN. In order to support one of these diagno-
ses, the presence of either MPO-ANCA or PR3-ANCA positivity
should be determined. Furthermore, ANCA testing is sometimes
asked in patients clinically suspected of a cocaine-induced midline
antigen-coated test anti-human destructive disease, chronic carriage of Pseudomonas, and/or in
carrier serum IgG patients with possible primary sclerosing cholangitis or ulcerative
(b) Capture ELISA colitis (reviewed in Cohen Tervaert and Damoiseaux 2012).
In virtually all patients with NCGN with or without an accom-
panying small-vessel vasculitis, MPO-ANCA or PR3-ANCA can
be detected. Some patients with NCGN, however, test negative for
anti-ANCA ANCA test anti-human MPO-ANCA and PR3-ANCA. ANCA negativity in these latter
carrier antigen serum IgG
patients, however, may be the consequence of suboptimal ANCA
(c) Anchor ELISA tests. Indeed, some of these cases are negative in the direct ANCA
ELISA tests but positive in the capture or anchor ELISA.
In GPA with renal involvement, nearly all patients in Europe and
the USA have PR3-ANCA. However, the geographic area is impor-
antigen linked test anti-human
to carrier serum IgG tant. In China, 60% of GPA patients have MPO-ANCA, while 38%
have PR3-ANCA (Chen et  al. 2005). In patients with early sys-
Fig. 6.2 PR3- and MPO-ANCA detection by antigen-specific assays. The antigen temic or limited GPA, most patients test positive for ANCA, either
is either coated directly onto the carrier (a), or via a capturing monoclonal PR3-ANCA or MPO-ANCA. In patients with locoregional GPA,
antibody (b), or via a peptide linker (c). Incubation with the patient serum and
however, ANCA is clearly not detected in all patients (reviewed in
the final detection reagent is similar in these assays. Adapted from: Damoiseaux,
J., Austen, J., and Cohen Tervaert, J.W. (2011). ANCA diagnostics in clinical Cohen Tervaert and Damoiseaux 2012). These locoregional GPA
practice: new developments. In: Advances in the diagnosis and treatment of patients may have either PR3-ANCA, elastase-ANCA, or ANCA of
vasculitis. Ed. Amezcua-Guerra, L.M. Intech, Croatia, pp. 3–18. other specificities. At least one-third of these patients, however, do
CHAPTER 6 autoantibodies in vasculitis 65

test completely negative, both by IIF and in antigen-specific tests in Elastase-ANCA and hLAMP-2-ANCA
which different antigens are used. These patients may suffer from
chronic ulcerative mucosal lesions in the nose, subglottic trachea Elastase-ANCA as detected by capture ELISA is only very infre-
stenosis, or pseudotumour of the orbit. Finally, patients with mul- quently found in patients suspected of systemic vasculitis.
tiple necrotic lung nodules may be ANCA-negative. In these latter Elastase-ANCA, however, is a specific finding for systemic vascu-
cases, however, an extremely careful search for other diagnosis such litis because elastase-ANCA is hardly ever found in other condi-
as tuberculosis or malignancy should be carried out. tions (Cohen Tervaert et al. 1993). Elastase-ANCA points to the
In patients with EGPA, only approximately 40% test positive for occurrence of drug-induced vasculitis. In drug-induced vasculitis
MPO-ANCA, whereas the remaining patients are ANCA-negative not only elastase-ANCA can be found but often also MPO-ANCA,
(reviewed in Cohen Tervaert and Damoiseaux 2012). Patients PR3-ANCA, and/or ANCA directed against other specificities
who test positive for MPO-ANCA more often have glomerulo- (reviewed in Cohen Tervaert and Damoiseaux 2012). Furthermore,
nephritis, whereas cardiac involvement is more often found in elastase-ANCA is specifically found in cocaine-induced mid-
MPO-ANCA-negative cases (Dennert et  al. 2010). Occasionally, line destructive disease (Trimarchi et al. 2001). Cocaine not only
patients with severe adult-onset asthma, nasal polyposis, and induces chronic midline disease but can also be associated with a
eosinophilia without clear manifestations of systemic vasculitis test systemic form of necrotizing vasculitis in which either PR3-ANCA,
positive for MPO-ANCA, suggesting that these cases have an early MPO-ANCA, and/or the combination of PR3-ANCA and
phase of EGPA. MPO-ANCA can be found (Cohen Tervaert and Stegeman 2004;
When patients suffer from systemic vasculitis affecting small McGrath et al. 2011).
to medium-sized blood vessels that cannot be classified as ANCA directed against hLAMP-2 can be found in patients with
either GPA, EGPA, IgAV (HSP), cryoglobulin-associated vas- NCGN (Kain et  al. 1995; Kain et  al. 2008). Both patients with
culitis, and/or a secondary form of vasculitis, patients must be MPO-ANCA and patients with PR3-ANCA-associated NCGN test
classified per exclusionem as suffering from MPA. Most of these positive for hLAMP-2-ANCA. Importantly, also patients that have
patients test positive for MPO-ANCA or PR3-ANCA. Within NCGN without the presence of either MPO-ANCA or PR3-ANCA
the group of patients with MPA, patients who test positive for sometimes test positive for hLAMP-2 ANCA.
PR3-ANCA, generally, have more necrotizing lesions in the res-
piratory tract compared to patients with MPO-ANCA, whereas Monitoring ANCA levels during
patients with MPO-ANCA have more often either nasal polypo-
sis or chronic bronchial obstruction (reviewed in Cohen Tervaert follow-up
and Damoiseaux 2012). Furthermore, eosinophilia is more often Several studies have demonstrated that the persistence of ANCA is
found in patients with MPO-ANCA-associated MPA. These find- related to the occurrence of relapses in the future (Stegeman et al.
ings suggest that ANCA serotype should be incorporated when 1994). The positive likelihood ratio, however, for persistence of
patients are classified. ANCA during remission for subsequent disease relapse is only 1.97
The performance of diagnostic tests is not only expressed by the (95% CI 1.43–2.70) (Tomasson et al. 2012). Importantly, however,
sensitivity of the test for the disease but also by its specificity. The in more than 80% of patients with AAV relapses occur while ANCA
specificity of the combination pANCA/MPO-ANCA or cANCA/ tests are or have become positive again.
PR3-ANCA is excellent, that is close to 99% (Hagen et al. 1998; Choi The usefulness of measuring ANCA titres to predict disease
et al. 2001). When antigen-specific tests are done without IIF tests, activities and/or to guide therapy is very controversial. Absolute
then the specificities of individual antigen-specific tests are quite titres correlate only weakly with disease severity. Rising titres
variable. Recent studies, however, suggest that the antigen-specific of PR3-ANCA occurring during clinical remission may pre-
ANCA tests of the second or third generation have a specificity for dict relapses in patients with GPA (Cohen Tervaert et al. 1989b;
AAV which is comparable to that of the combination of the IIF test Boomsma et  al. 2000). Noteworthy in these studies, the relation
with an antigen-specific test of the first generation (Damoiseaux between ANCA rises and relapses occurred in patients that were
et al. 2009a; Cohen Tervaert and Damoiseaux 2012). treated with cyclophosphamide therapy. The close relation between
Next to characteristics as sensitivity and specificity, the clinical ANCA rises and relapses, however, could not be found in other
usefulness of ANCA is best expressed when likelihood ratios (LRs) cohorts in which patients were treated with other forms of therapy
are calculated. The LR provides an estimation of the change in pre- such as methotrexate (MTX) (reviewed in Cohen Tervaert and
test probability to post-test probability of a disease given the test Damoiseaux 2012). Recently, a close correlation between ANCA
result. LRs > 10 or < 0.1 indicate large, clinically significant differ- and relapses was also found during therapy with B-cell depletion
ences. LRs between 5 and 10 and between 0.1 and 0.2 indicate mod- using rituximab that was similar to observations during ther-
est differences, while LRs between 2 and 5 and between 0.2 and 0.5 apy with cyclophosphamide (Cartin-Ceba et  al. 2012). Also, for
indicate small differences (American College of Rheumatology ad MPO-ANCA it has been demonstrated that relapses, generally,
hoc committee on immunological testing guidelines 2002; Bossuyt occur after an ANCA rise. Large prospective studies with respect to
2009). The LRs for a negative test result of either the IIF test or MPO-ANCA are, however, lacking.
antigen-specific tests are found to be between 0.1 and 0.2, sug- In a meta-analysis performed on nine out of 41 articles in which
gesting a substantial but modest difference in pretest to post-test the relationship between ANCA and relapses was studied, it was
probability (Vermeersch et al. 2009). The LRs for the diagnosis of concluded that the positive likelihood ratio for a rise of ANCA
AAV (positive test results) increases with antibody levels, while (either cANCA/PR3-ANCA or pANCA/MPO-ANCA) during
LRs of antigen-specific tests are much higher than for the IIF test remission on a subsequent relapse of disease was 2.84 (95% CI
(Vermeersch et al. 2009). 1.65–4.90) (Tomasson et al. 2012). In addition, it was found that
66 SECTION 2 basic science

the absence of an ANCA rise does not exclude disease reactivation results in the production of reactive oxygen species and the release
because the negative likelihood ratio of an ANCA rise for a sub- of lytic enzymes. This process is both dependent on the interaction
sequent relapse was only 0.49 (95% CI 0.27–0.87). Most of these of the F(ab′)2 fragments and the Fc part of ANCA. Furthermore,
studies are based on ANCA detection as performed using either this process occurs in vivo only when neutrophils are bound to a
the IIF technique or a first-generation ELISA technique. The pre- surface, for example endothelial cells, but not when the neutrophils
dictive value of an ANCA rise as detected by tests using the second are in suspension. Alternatively, ANCA may interact with soluble
or third generation of ANCA tests are slightly better for predicting ANCA antigens resulting in their inactivation or inhibition of their
an ensuing relapse (Damoiseaux et al. 2009a). However, it is clear clearance (reviewed in Reumaux et al. 2004).
that more studies in this respect are needed. Furthermore, addi- Finally, proof of a pathogenic role comes from in vivo experi-
tional prospective studies regarding epitopes, IgG subclasses, avid- mental studies. Both in mice and rats, immunization with either
ity, affinity, and/or sialylation of ANCA are needed (reviewed in murine MPO (mouse model) or human MPO (rat model) results
Cohen Tervaert and Damoiseaux 2012). in the generation of MPO-ANCA. In both species, MPO-ANCA
induces mild vasculitis, a process that is aggravated by additional
When should ANCA testing be factors such as endotoxin (‘second hit’) (Heeringa et  al. 2005;
Huugen et al. 2005).
ordered in the clinic? Also, immunization with species-specific PR3 in mice or rats
ANCA testing should be ordered when either characteristic mani- results in the generation of PR3-ANCA. This PR3-ANCA, how-
festations (e.g. pseudotumour of the orbit) are present or when ever, does not induce vasculitis, even when systemically injected
a combination of a suggestive symptom (e.g. chronic nasal san- with endotoxin (Kallenberg 2011). In contrast, when human
guineous discharge) is present in combination with non-specific PR3-ANCA in combination with endotoxin is injected into mice
symptoms such as fatigue, malaise, anorexia, weight loss, and/or with a humanized immune system a severe pulmonary–renal vas-
arthralgias. A positive PR3-ANCA or MPO-ANCA will in these culitis develops (Little et al. 2012). In addition, transfer of rabbit
cases influence plans for evaluation of organ involvement and of hLAMP-2 ANCA into rats results in renal vasculitis, suggesting
possible biopsy sites. When, in patients with characteristic clinical that not only MPO-ANCA and PR3-ANCA but also hLAMP-2
findings suggestive of AAV without other obvious causes for these ANCA is a pathogenic antibody in AAV (Kain et al. 2008).
findings, ANCA tests are ordered using strict ordering guidelines, AAV is a disease with putative complex environmental and
only very few cases of AAV are missed (see Table 1 in Mandl et al. genetic influences. Environmental factors include silica (mainly
2002). Furthermore, such a gated policy diminishes the amount of in MPO-ANCA-associated vasculitis) and Staphylococcus
requested samples by up to 67%, resulting in enormous savings in aureus (mainly in PR3-ANCA-associated vasculitis) (De Lind
laboratory costs (Robinson and Steele 2009). To avoid unnecessary van Wijngaarden et  al. 2008). Genetic factors are HLA-DP and
delay in making a diagnosis in AAV, however, clinicians should the genes encoding α(1)-antitrypsin (SERPINA1) and protein-
have the opportunity to order ANCA testing in cases that do not ase 3 (PRTN3) in PR3-ANCA-associated AAV and HLA-DQ in
fulfil strict guidelines. MPO-ANCA-associated vasculitis (Lyons et al. 2012). Thus, ANCA
and ANCA antigens have a central role in determining disease pheno-
Pathophysiological role type. Furthermore, ANCA specificity independently predicts relapse
and outcome among patients with AAV. Therefore, future classifica-
of ANCA in AAV tion and diagnostic systems should incorporate ANCA specificity,
Clinical observations that ANCA persistence or ANCA rises are for example PR3-ANCA-positive MPA and MPO-ANCA-positive
related to an increased risk of disease reactivation suggest that MPA (Cohen Tervaert and Damoiseaux 2012).
ANCA might be pathogenic. In addition, treatment with rituxi-
mab, a B-lymphocyte-depleting monoclonal antibody is effective
for induction of remission (Jones et al. 2010; Stone et al. 2010), sug- Conclusions
gesting that B lymphocytes, including those producing ANCA, play ANCA are extremely useful markers for the diagnosis of primary
a major role in AAV. Importantly, a neonate born from a mother small-vessel vasculitides, collectively referred to as AAV. At present,
with MPO-ANCA-associated vasculitis has been described who there is international consensus that ANCA should be detected
developed transient vasculitis probably due to transplacental trans- by a combination of IIF and antigen-specific assays, that is detec-
fer of MPO-ANCA (Bansal and Tobin 2004). tion of PR3- and MPO-ANCA. ANCA as detected by second and/
Further proof of a pathogenic role of ANCA comes from in or third-generation ANCA-tests may possibly replace the need to
vitro experiments. ANCA antigens may be released from neutro- perform IIF as a screening assay. For the classification of patients,
phils during activation of these cells; for instance during infec- ANCA serotype, that is PR3-ANCA or MPO-ANCA, should be
tion. Soluble ANCA antigens may bind to innocent bystander cells incorporated when patients are classified according to their clinical
such as other neutrophils or endothelial cells and subsequently subtype, that is GPA, MPA, or EGPA. The detection of ANCA to
an interaction with ANCA may result in activation of these cells. monitor disease activity is still a controversial issue. Most monitor-
Otherwise, ANCA antigens may be up-regulated on the cell sur- ing studies are based on ANCA detection as performed using either
face of neutrophils and monocytes during priming of these cells the IIF technique or an ELISA technique of the first generation.
by cytokines, such as tumour necrosis factor (TNF)-α, which are The predictive value of an ANCA rise, as detected by tests using the
released during inflammation (Muller Kobold et al. 1999). ANCA second or third generation of ANCA tests, is probably better for
antigens are subsequently captured within the neutrophil extra- predicting an ensuing relapse. However, more studies using these
cellular traps (Kessenbrock et  al. 2009). Interaction with ANCA tests are needed.
CHAPTER 6 autoantibodies in vasculitis 67

References Cohen Tervaert, J.W., Goldschmeding, R., Elema, J.D., et  al. (1990).
Autoantibodies against myeloid lysosomal enzymes in crescentic glo-
American College of Rheumatology Ad Hoc Committee on Immunologic merulonephritis. Kidney International, 37, 799–806.
Testing Guidelines (2002). Guidelines for immunologic labora- Cohen Tervaert, J.W., Huitema, M., Van der Giessen, M., et  al. (1989a).
tory testing in the rheumatic diseases:  an introduction. Arthritis and Wegener’s granulomatosis and anti-cytoplasmic antibodies:  the
Rheumatism, 27, 429–33. Groningen experience. Acta Pathologica Microbiologica et Immunologica
Bansal, P.J. and Tobin, M.C. (2004). Neonatal microscopic polyangiitis sec- Scandinavica, 97 (S6), 36.
ondary to transfer of maternal myeloperoxidase–antineutrophil cyto- Cohen Tervaert, J.W., Mulder, L., Stegeman, C.A., et al. (1993). Occurrence of
plasmic antibody resulting in neonatal pulmonary hemorrhage and autoantibodies to human leucocyte elastase in Wegener’s granulomato-
renal involvement. Annals of Allergy Asthma and Immunology, 93, sis and other inflammatory disorders. Annals of the Rheumatic Diseases,
398–401. 52, 115–20.
Belizna, C. and Cohen Tervaert, J.W. (1997). Specificity, pathogenicity, and Cohen Tervaert, J.W. and Stegeman, C.A. (2004). A difficult diagnosis.
clinical value of antiendothelial cell antibodies. Seminars in Arthritis and Lancet, 364, 1313–14.
Rheumatism, 27, 98–109. Cohen Tervaert, J.W., Van der Woude, F.J., Fauci, A.S., et  al. (1989b).
Bini, P., Gabay, J.E., Teitel, A., et  al. (1992). Antineutrophil cytoplasmic Association between active Wegener’s granulomatosis and anticytoplas-
autoantibodies in Wegener’s granulomatosis recognize conformational mic antibodies. Archives of Internal Medicine, 149, 2461–5.
epitope(s) on proteinase 3. Journal of Immunology, 149, 1409–15. Cowland, J.B. and Borregaard, N. (1999). The individual regulation of gran-
Bjerregaard, M.D., Jurlander, J., Klausen, P., et al. (2003). The in vivo profile ule protein mRNA levels during neutorphil maturation explains the
of transcription factors during neutrophil differentiation in human bone heterogeneity of neutrophil granules. Journal of Leukocyte Biology, 66,
marrow. Blood, 101, 4322–32. 989–95.
Boomsma, M.M., Damoiseaux, J.G., Stegeman, C.A., et  al. (2003). Image Csernok, E., Holle, J., Hellmich, B., et al. (2004). Evaluation of capture ELISA
analysis: a novel approach for the quantification of antineutrophil cyto- for detection of antineutrophil cytoplasmic antibodies directed against
plasmic antibody levels in patients with Wegener’s granulomatosis. proteinase 3 in Wegener’s granulomatosis:  first results from a multi-
Journal of Immunological Methods, 274, 27–35. center study. Rheumatology, 43, 174–80.
Boomsma, M.M., Stegeman, C.A., Oost-Kort, W.W., et al. (2001). Native and Damoiseaux, J. and Cohen Tervaert, J.W. (2005). Tests for autoantibodies.
recombinant proteins to analyze auto-antibodies to myeloperoxidase in In: Encyclopedic reference of immunotoxicology (ed. H. Vohr), pp. 68–72.
pauci-immune glomerulonephritis. Journal of Immunological Methods, Springer-Verlag, Heidelberg.
254, 47–58. Damoiseaux, J., Dähnrich, C., Rosemann, A., et  al. (2009a). A novel
Boomsma, M.M., Stegeman, C.A., Van der Leij, M.J., et al. (2000). Prediction enzyme-linked immunosorbent assay using a mixture of human native
of relapses in Wegener’s granulomatosis by measurement of antineu- and recombinant proteinase-3 significantly improves the diagnostic
trophil cytoplasmic antibody levels: a prospective study. Arthritis and potential for antineutrophil cytoplasmic antibody-associated vasculitis.
Rheumatism, 43, 2025–33. Annals of the Rheumatic Diseases, 68, 228–33.
Bossuyt, X. (2009). Clinical performance characteristics of a laboratory test. Damoiseaux, J., Mallet, K., Vaessen, M., et  al. (2012). Automatic read-
A  practical approach in the autoimmune laboratory. Autoimmunity ing of ANCA-slides: evaluation of the AKLIDES system. Clinical and
Reviews, 8, 543–8. Developmental Immunology, 2012, 762874.
Calabresi, P., Edwards, E.A., and Schilling, R.F. (1959). Fluorescent anti- Damoiseaux, J.G., Slot, M.C., Vaessen, M., et  al. (2005). Evaluation of a
globulin studies in leukopenic and related disorders. Journal of Clinical new fluorescent-enzyme immuno-assay for diagnosis and follow-up
Investigation, 38, 2091–100. of ANCA-associated vasculitis. Journal of Clinical Immunology,
Calabresi, P., Thayer, W.R., and Spiro, H.M. (1961). Demonstration of cir- 25, 202–8.
culating antinuclear globulins in ulcerative colitis. Journal of Clinical Damoiseaux, J., Steller, U., Buschtez, M., et  al. (2009b). EUROPLUSTM
Investigation, 40, 2126–33. ANCA BIOCHIP mosaic:  PR3 and MPO antigen microdots improve
Campanelli, D., Melchior, M., Fu, Y., et al. (1990). Cloning of cDNA for pro- the laboratory diagnostics of ANCA-associated vasculitis. Journal of
teinase 3:  a serine protease, antibiotic, and autoantigen from human Immunological Methods, 348, 67–73.
neutrophils. Journal of Experimental Medicine, 172, 1709–15. Damoiseaux, J., Vaessen, M., Knapen, Y., et  al. (2007). Evaluation of the
Cartin-Ceba, R., Golbin, J.M., Keogh, K.A., et  al. (2012). Rituximab for FIDIS vasculitis multiplex immunoassay for diagnosis and follow-up
remission induction and maintenance in refractory granulomatosis with of ANCA-associated vasculitis and Goodpasture’s disease. Annals of the
polyangiitis (Wegener’s): Ten-year experience at a single center. Arthritis New York Academy of Sciences, 1109, 454–63.
and Rheumatism, 64, 3770–8. Davies, D.J., Moran, J.E., Niall, J.F., et al. (1982). Segmental necrotising glo-
Chang, K.S., Schroeder, W., Siciliano, M.J., et al. (1987). The localization of merulonephritis with antineutrophil antibody: possible arbovirus aeti-
the human myeloperoxidase gene is in close proximity to the transloca- ology? British Medical Journal, 285, 606.
tion breakpoint in acute promyelocytic leukemia. Leukemia, 1, 458–62. De Lind van Wijngaarden, R.A., Van Rijn, L., Hagen, E.C., et  al. (2008).
Chen, M., Yu, F., Zhang, Y., et al. (2005). Characteristics of Chinese patients Hypotheses on the etiology of antineutrophil cytoplasmic autoanti-
with Wegener’s granulomatosis with anti-myeloperoxidase autoantibod- body associated vasculitis: the cause is hidden, but the result is known.
ies. Kidney International, 68, 2225–9. Clinical Journal of the American Society of Nephrology, 3, 237–52.
Choi, H.K., Liu, S., Merkel, P.A., et  al. (2001). Diagnostic performance Dennert, R.M., Van Paassen, P., Schalla, S., et al. (2010). Cardiac involvement
of antineutrophil cytoplasmic antibody tests for idiopathic vascu- in Churg-Strauss syndrome. Arthritis and Rheumatism, 62, 627–34.
litides: meta-analysis with a focus on antimyeloperoxidase antibodies. Elzouki, A.N., Eriksson, S., Löfberg, R., et  al. (1999). The prevalence and
Journal of Rheumatology, 28, 1584–90. clinical significance of alpha 1-antitrypsin deficiency (PiZ) and ANCA
Cohen Tervaert, J.W. and Damoiseaux, J. (2009). Fifty years of antineutrophil specificities (proteinase 3, BPI) in patients with ulcerative colitis.
cytoplasmic antibodies (ANCA) testing: do we need to revise the inter- Inflammatory Bowel Disease, 5, 246–52.
national consensus statement on testing and reporting on ANCA? Acta Eskelinen, E.L., Tanaka, Y., and Saftig, P. (2003). At the acidic edge: emerging
Pathologica, Microbiologica et Immunologica Scandinavica, 117, S55–9. functions for lysosomal membrane proteins. Trends in Cell Biology, 13,
Cohen Tervaert, J.W. and Damoiseaux, J. (2012). Antineutrophil cytoplas- 137–45.
mic autoantibodies:  how are they detected and what is their use for Falk, R.J., Becker, R., Terrell, R., et al. (1992). Anti-myeloperoxidase autoan-
diagnosis, classification and follow-up? Clinical Reviews in Allergy and tibodies react with native but not denatured myeloperoxidase. Clinical
Immunology, 43, 211–19. and Experimental Immunology, 89, 274–8.
68 SECTION 2 basic science

Falk, R.J. and Jennette, J.C. (1988). Anti-neutrophil cytoplasmic autoantibod- neutrophil granulocytes and a related membrane protein in glomerular
ies with specificity for myeloperoxidase in patients with systemic vascu- endothelial cells. Journal of Experimental Medicine, 181, 585–97.
litis and idiopathic necrotizing and crescentic glomerulonephritis. New Kallenberg, C.G. (2011). Pathogenesis of ANCA-associated vasculitides.
England Journal of Medicine, 318, 1651–7. Annals of Rheumatic Disease, 70 (Suppl. 1), i59–63.
Falk, R.J. and Jennette, J.C. (1989). Immunofluorescence and ELISA Kaul, R., Johnson, K., Scholz, H., et al. (2009). Performance of the BioPlexTM
determination of ANCA with description of a sub-class with 2200 autoimmune vasculitis kit. Autoimmunity Reviews, 8, 224–7.
anti-myeloperoxidase activity. Acta Pathologica Microbiologica et Kessenbrock, K., Krumbholz, M., Schönemarck, U., et  al. (2009). Netting
Immunologica Scandinavica, 97 (S6), 45. neutrophils in autoimmune small-vessel vasculitis. Nature Medicine,
Franssen, C.F., Stegeman, C.A., Kallenberg, C.G., et al. (2000). Antiproteinase 15, 623–5.
3- and antimyeloperoxidase-associated vasculitis. Kidney International, Korkmaz, B., Horwitz, M.S., Jenne, D.E., et al. (2010). Neutrophil elastase,
57, 2195–206. proteinase 3, and cathepsin G as therapeutic targets in human diseases.
Fujinaga, M., Cgernaia, M., Halenbeck, R., et al. (1996). The crystal structure Pharmacological Reviews, 62, 726–59.
of PR3, a neutrophil serine proteinase antigen of Wegener’s granuloma- Little, M.A., Al-Ani, B., Ren, S., et al. (2012). Anti-proteinase 3 anti-neutrophil
tosis antibodies. Journal of Molecular Biology, 261, 267–78. cytoplasm autoantibodies recapitulate systemic vasculitis in mice with a
Goldschmeding, R., Van der Schoot, C.E., Ten Bokkel Huinink, K., et  al. humanized immune system. PLoS One, 7, e28626.
(1989). Wegener’s granulomatosis autoantibodies identify a novel Lyons, P.A., Rayner, T.F., Trivedi, S., et al. (2012). Genetically distinct subsets
diisopropylfluorophosphate-binding protein in the lysosomes of normal within ANCA-associated vasculitis. New England Journal of Medicine,
human neutrophils. Journal of Clinical Investigation, 84, 1577–87. 367, 214–23.
Griffith, M.E., Coulthart, S., Pemberton, A., et al. (2001). Anti-neutrophil Malle, E., Furtmüller, P.G., Sattler, W., et al. (2007). Myeloperoxidase: a target
cytoplasmic antibodies (ANCA) from patients with systemic vasculitis for new drug development? British Journal of Pharmacology, 152, 838–54.
recognize restricted epitopes of proteinase 3 involving the catalytic site. Mandl, L.A., Solomon, D.H., Smith, E.L., et al. (2002). Using antineutrophil
Clinical and Experimental Immunology, 123, 170–7. cytoplasmic antibody testing to diagnose vasculitis: can test-ordering
Hagen, E.C., Andrassy, K., Csernok, E., et  al. (1996). Development and guidelines improve diagnostic accuracy? Archives of Internal Medicine,
standardization of solid phase assays for the detection of anti-neutrophil 162, 1509–14.
cytoplasmic antibodies (ANCA). A report on the second phase of an McGrath, M.M., Isakova, T., Rennke, H.G., et  al. (2011). Contaminated
international cooperative study on the standarization of ANCA assays. cocaine and antineutrophil cytoplasmic antibody-associated disease.
Journal of Immunological Methods, 196, 1–15. Clinical Journal of the American Society of Nephrology, 6, 2799–805.
Hagen, E.C., Daha, M.R., Hermans, J., et al. (1998). Diagnostic value of stand- Merkel, P. (2001). Drug-induced vasculitis. Rheumatic Disease Clinics of
ardized assays for anti-neutrophil cytoplasmic antibodies in idiopathic North America, 27, 849–62.
systemic vasculitis. EC/BCR Project for ANCA Assay Standardization. Müller-Bérat, N., Minowada, J., Tsuji-Takayama, K., et al. (1994). The phy-
Kidney International, 53, 743–53. logeny of proteinase 3/myeloblastin, the autoantigen in Wegener’s
Heeringa, P., Huugen, D., and Cohen Tervaert, J.W. (2005). Anti-neutrophil granulomatosis, and myeloperoxidase as shown by immunohistochemi-
cytoplasmic autoantibodies and leukocyte-endothelial interactions:  a cal studies on human leukemic cell lines. Clinical Immunology and
sticky connection? Trends in Immunology, 26, 561–64. Immunopathology, 70, 51–9.
Hellmich, B., Csernok, E., Fredenhagen, G., et  al. (2007). A novel high Muller Kobold, A.C., Van der Geld, Y.M., Limburg, P.C., et  al. (1999).
sensitivity ELISA for detection of anti-neutrophil cytoplasm antibod- Pathophysiology of ANCA-associated glomerulonephritis. Nephrology
ies against proteinase-3. Clinical and Experimental Rheumatology, 25 Dialysis and Transplantation, 14, 1366–75.
(S44), S1–5. Nifli, A.-P., Notas, G., Mamoulaki, M., et al. (2006). Comparison of a mul-
Holweg, A., Schnare, M., and Gessner, A. (2011). The bactericidal/ tiplex, bead-based fluorescent assay and immunofluorescence method
permeability-increasing protein (BPI) in the innate defence of the lower for the detection of ANA and ANCA autoantibodies in human serum.
airways. Biochemical Society Transactions, 39, 1045–50. Journal of Immunological Methods, 311, 189–97.
Huugen, D., Xiao, H., Van Esch, A., et  al. (2005). Aggravation of Niles, J.L., McCluskey, R.T., Ahmad, M.F., et al. (1989). Wegener’s granulo-
anti-myeloperoxidase antibody-induced glomerulonephritis by bacte- matosis autoantigen is a novel neutrophil serine proteinase. Blood, 74,
rial lipopolysaccharide: role of tumor necrosis factor-alpha. American 1888–93.
Journal of Pathology, 167, 47–58. Pearson, T., Bremmer, M., Cohen, J., et  al. (2012). Vasculopathy related
Jenne, D.E. (1994). Structure of azurocidin, proteinase 3, and neutrophil to cocaine adulterated with levamisole:  a review of the literature.
elastase genes. Implications for inflammation and vasculitis. American Dermatology Online Journal, 18, 1.
Journal of Respiratory and Critical Care Medicine, 150, S147–54. Radice, A. and Sinico, R.A. (2005). Antineutrophil cytoplasmic antibodies.
Jenne, D.E. and Kuhl, A. (2006). Production and applications of recombi- Autoimmunity, 38, 93–103.
nant proteinase 3, Wegeners’s autoantigen: problems and perspectives. Régent, A., Dib, H., Ly, K.H., et al. (2011). Identification of target antigens
Clinical Nephrology, 66, 153–9. of anti-endothelial cell and anti-vascular smooth muscle cell antibod-
Jenne, D.E., Tschopp, J., Ludemann, J., et al. (1990). Wegener’s autoantigen ies in patients with giant cell arteritis: a proteomic approach. Arthritis
decoded. Nature, 346, 520. Research and Therapy, 13, R107.
Jennette, J.C., Falk, R.J., Bacon, P., et al. (2013). Revised international Chapel Reumaux, D., Duthilleul, P., and Roos, D. (2004). Pathogenesis of diseases
Hill consensus conference nomenclature of vasculitides. Arthritis and associated with antineutrophil cytoplasm autoantibodies. Human
Rheumatism, 65, 1–11. Immunology, 65, 1–12.
Jones, R.B., Cohen Tervaert, J.W., Hauser, T., et al. (2010). Rituximab versus Robinson, P.C. and Steele, R.H. (2009). Appropriateness of antineutrophil
cyclophosphamide in ANCA-associated renal vasculitis. New England cytoplasmic antibody testing in a tertiary hospital. Journal of Clinical
Journal of Medicine, 363, 211–20. Pathology, 62, 743–5.
Kain, R., Exner, M., Brandes, R., et  al. (2008). Molecular mimicry in Roggenbuck, D., Buettner, T., Hoffmann, L., et al. (2009). High-sensitivity
pauci-immune focal necrotizing glomerulonephritis. Nature Medicine, detection of autoantibodies against proteinase-3 by a novel
14, 1088–96. third-generation enzyme-linked immunosorbent assay. Annals of the
Kain, R., Matsui, K., Exner, M., et al. (1995). A novel class of autoantigens New York Academy of Sciences, 1173, 41–6.
of anti-neutrophil cytoplasmic antibodies in necrotizing and crescentic Rutgers, A., Damoiseaux, J.G. Roozendaal, C., et  al. (2004). ANCA-GBM
glomerulonephritis: the lysosomal membrane glycoprotein h-lamp-2 in dot-blot: evaluation of an assay in the differential diagnosis of patients
CHAPTER 6 autoantibodies in vasculitis 69

presenting with rapidly progressive glomerulonephritis. Journal of Terjung, B., Worman, H.J., Herzog, V., et al. (2001). Differentiation of anti-
Clinical Immunology, 24, 435–40. neutrophil nuclear antibodies in inflammatory bowel and autoimmune
Savige, J., Gillis, D., Benson, E., et al. (1999). International consensus state- liver diseases from antineutrophil cytoplasmic antibodies (p-ANCA)
ment on testing and reporting of antineutrophil cytoplasmic antibodies using immunofluorescence microscopy. Clinical and Experimental
(ANCA). American Journal of Clinical Pathology, 111, 507–13. Immunology, 126, 37–46.
Short, A.K., Lockwood, C.M., Bollen, A., et al. (1995). Neutrophil native and Tomasson, G., Grayson, P.C., Mahr, A.D., et al. (2012). Value of ANCA meas-
recombinant myeloperoxidase as antigens in ANCA positive systemic urements during remission to predict a relapse of ANCA-associated
vasculitis. Clinical and Experimental Immunology, 102, 106–11. vasculitis—a meta-analysis. Rheumatology (Oxford), 51, 100–9.
Sivalingam, G.N. and Shepherd, A.J. (2012). An analysis of B-cell epitope dis- Trimarchi, M., Gregorini, G., Facchetti, F., et  al. (2001). Cocaine-induced
continuity. Molecular Immunology, 51, 304–9. midline destructive lesions: clinical, radiographic, histopathologic, and
Specks, U., Fass, D.N., Fautsch, M.P., et al. (1996). Recombinant human pro- serologic features and their differentiation from Wegener granulomato-
teinase 3, the Wegener’s autoantigen, expressed in HMC-1 cells is enzy- sis. Medicine, 80, 391–404.
matically active and recognized by C-ANCA. Federation of European Van der Geld, Y.M., Limburg, P.C., and Kallenberg, C.G.M. (1999).
Biochemical Societies Letters, 390, 265–70. Characterization of monoclonal antibodies to proteinase 3 (PR3) as
Stegeman, C.A., Cohen Tervaert, J.W., Sluiter, W.J., et al. (1994). Association candidate tools for epitope mapping of human anti-PR3 autoantibodies.
of chronic nasal carriage of Staphylococcus aureus and higher relapse Clinical and Experimental Immunology, 118, 487–96.
rates in Wegener granulomatosis. Annals of Internal Medicine, Van der Woude, F.J., Rasmussen, N., Lobatto, S., et al. (1985). Autoantibodies
120, 12–17. against neutrophils and monocytes: tool for diagnosis and marker of dis-
Stoffel, M.P., Csernok, E., Herzberg, C., et al. (1996). Anti-neutrophil cyto- ease activity in Wegener’s granulomatosis. Lancet, 1, 425–9.
plasmic antibodies (ANCA) directed against bactericidal/permeability Vermeersch, P., Blockmans, D., and Bossuyt, X. (2009). Use of likelihood
increasing protein (BPI): a new seromarker for inflammatory bowel dis- ratios can improve the clinical usefulness of enzyme immunoassays for
ease and associated disorders. Clinical and Experimental Immunology, the diagnosis of small-vessel vasculitis. Clinical Chemistry, 55, 1886–8.
104, 54–9. Westman, K.W., Selga, D., Bygren, P.G., et al. (1998). Clinical evaluation of a
Stone, J.H., Merkel, P.A., Spiera, R., et al. (2010). Rituximab versus cyclo- capture ELISA for detection of proteinase-3 antineutrophil cytoplasmic
phosphamide for ANCA-associated vasculitis. New England Journal of antibody. Kidney International, 53, 1230–6.
Medicine, 363, 221–32. Wiik, A. (1989). Delineation of a standard procedure for indirect immu-
Sturrock, A.B., Franklin, K.F., Rao, G., et al. (1992). Structure, chromosomal nofluorescence detection of ANCA. Acta Pathologica et Microbiologica
assignment, and expression of the gene for proteinase 3, the Wegener’s Scandinavica, 97 (S6), 12–13.
granulomatosis autoantigen. Journal of Biological Chemistry, 267, Wiik, A. and van der Woude, F.J. (1990). The new ACPA/ANCA nomencla-
21193–9. ture. Netherlands Journal of Medicine, 36, 107–8.
Sun, J., Fass, D.N., Viss, M.A., et  al. (1998). A proportion of proteinase 3 Xiao, H., Heeringa, P., Hu, P., et  al. (2002). Antineutrophil cytoplasmic
(PR3)-specific anti-neutrophil cytoplasmic antibodies (ANCA) only autoantibodies specific for myeloperoxidase cause glomerulonephritis
react with PR3 after cleavage of its N-terminal activation dipeptide. and vasculitis in mice. Journal of Clinical Investigation, 110, 955–63.
Clinical and Experimental Immunology, 114, 320–6. Xu, J., Yang, C.H., Chen, X.Y., et  al. (2008). A subset of ulcerative colitis
Talor, M.V., Stone, J.H., Stebbing, J., et  al. (2007). Antibodies to selected with positive proteinase-3 antineutrophil cytoplasmic antibody. World
minor target antigens in patients with anti-neutrophil cytoplasmic anti- Journal of Gastroenterology, 14, 7012–15.
bodies (ANCA). Clinical and Experimental Immunology, 150, 42–8. Zhao, M.H., Jayne, D.R.W., Ardiles, L.G., et al. (1996). Autoantibodies against
Taylor, K., Pohl, J., and Kinkade, J.M. (1992). Unique autolytic cleavage of bactericidal/permeability-increasing protein in patients with cystic
human myeloperoxidase. Journal of Biological Chemistry, 267, 21193–9. fibrosis. Quarterly Journal of Medicine, 89, 259–65.
CHAPTER 7

Pathogenesis of vasculitis
Gene V. Ball, S. Louis Bridges, Jr, and
T. Prescott Atkinson

The pathogenesis of vasculitis is complex, and our understand- disease (KD); Chapter 29 granulomatosis with polyangiitis (GPA)
ing of it is an evolving process, as is true of every type of human (Wegener’s granulomatosis); Chapter  32 eosinophilic granulo-
disease. Gross and microscopic pathological findings in various matosis with polyangiitis (EGPA) (Churg–Strauss syndrome);
forms of vasculitis, which provide the basis for many of the cur- Chapter  33 vasculitis of primary connective tissue diseases;
rent hypotheses on their causes and mechanisms, are discussed in Chapter  34 Behçet’s disease (BD); Chapter  36 CNS vasculitis;
Chapter 9. Pathological findings and clinical responses of patients Chapter 37 thromboangiitis obliterans (TAO); Chapter 38 cuta-
with vasculitis to anti-inflammatory and immunosuppressive neous small-vessel vasculitis (SVV); and Chapter 40 cryoglob-
therapy support the concept that immunological mechanisms ulinaemic vasculitis.1
play an active role in the pathogenesis of vasculitic diseases. These
and their participants include: (1)  pathogenic immune complex Immune complexes and
formation and deposition in vessel walls; (2) autoantibodies such
as antineutrophil cytoplasmic antibodies (ANCA) and antien- complement in vasculitis
dothelial cell antibodies (AECA); (3)  cellular and molecular Deposition of immune complexes in blood vessels has long been
immune responses involving cytokines and adhesion molecules; implicated as an initiating event in some types of vascular inflam-
(4)  granuloma formation; and (5)  damaged or altered endothe- mation. Because of the relatively high incidence of this type of vas-
lial cell function due to infectious organisms, tumours, or toxins. culitis and ready accessibility of one of the principal affected organ
Immune complexes appear to initiate the inflammation of cutane- systems, the skin, the pathogenesis is better understood (Carter
ous small-vessel (leukocytoclastic) vasculitis related to infections 1973; Cochrane and Koffler 1973; Cream 1971; Theofilopoulos
and medications, of IgA vasculitis (IgAV) (Henoch–Schönlein and Dixon 1980). Because of the widespread use of hyperimmune
purpura), polyarteritis nodosa (PAN), and of some cryoglobuli- xenogeneic antisera for the treatment of such diseases as tetanus
naemias. ANCAs are probably major factors in pathogenesis of and diphtheria, physicians and immunologists in the early twen-
so-called pauci-immune vasculitis, and pathological responses of tieth century were quite familiar with the clinical and experimen-
T cells may be involved in others. Rapid expansion of our knowl- tal aspects of acute immune complex disease and with anaphylaxis
edge of the molecules that regulate interactions among cells of induced by a second course of such treatments (Goodall 1907;
the immune system and endothelial cells promises greater insight Goodall 1899; Goodall 1911; Currie 1907). About one in three
into the pathogenesis of vessel damage, and potential sites for patients receiving antisera for the first time developed the syn-
drug intervention. The underpinnings of applied clinical research drome that came to be known as serum sickness, usually a rela-
on vasculitis are described in detail in separate chapters focused tively benign side effect compared to the lethal diseases that were
on hypersensitivity (Chapter  3); the biology of endothelial cells being treated. Development of the syndrome depended on host fac-
(Chapter 4); complement (Chapter 5); and autoantibodies, includ- tors, the timing and dose of the antisera, as well as particular lots
ing ANCA (Chapter 6). Animal models pertinent to vasculitis are of antisera.
discussed in Chapter 8. In the acute serum sickness model, a single injection of xeno-
In this chapter we focus on general pathogenic mechanisms geneic serum is followed in 10–14  days by arteritis, glomerulo-
as they apply to the vasculitides. The discussion is organized as nephritis, and endocarditis. The vasculitic lesions, which appear
follows: (1) immune complexes and complement in vasculitis; when circulating antigen–antibody complexes are formed in slight
(2) ANCA; (3) AECA; (4) drug-induced vasculitis; (5) genetic antigen excess, contain antigen, immunoglobulin, and complement
influences; (6) infectious agents; (7) tumour cell-mediated vas- (Dixon et al. 1958; Carter 1973; Theofilopoulos and Dixon 1980).
cular damage; and (8)  cell-mediated immune responses and
granuloma formation. Chapters dealing with specific vasculitis 1 The designation of different vasculitis syndromes in this chapter will follow
diseases and syndromes, which include discussions germane the recent Chapel Hill Consensus Conference Nomenclature (Jennette,
to pathogenesis, are:  Chapter  23 giant cell arteritis (GCA); J. C., Falk, R. J., Bacon, P. A. et al. (2013). 2012 Revised international Chapel
Chapter 24 Takayasu’s arteritis (TAK); Chapters 25 and 26 PAN Hill Consensus Conference nomenclature of vasculitides. Arthritis and
and microscopic polyangiitis (MPA); Chapter  28 Kawasaki’s Rheumatism, 65(1):1–11).
72 SECTION 2 basic science

After immune complexes deposit, the vasculitis of acute serum pursuit in most cases of vasculitis because the inciting antigen is
sickness resembles PAN histologically with segmental infiltration seldom known, and it is not usually possible to elute sufficient
of arterial walls by neutrophils and mononuclear cells, intimal pro- material from vasculitic lesions to isolate and quantify specific anti-
liferation, and fibrinoid necrosis. bodies (McCluskey and Fienberg 1983). Failure to detect immuno-
Vasculitis is generally absent from animal models of chronic globulin in vasculitic lesions argues against their pathogenic role
serum sickness. There is a variable host response to intravenous only when early lesions are examined because neutrophils have
injections of foreign protein given daily for several weeks. The been shown to degrade immune complexes within 24–48 hours
majority of animals rapidly clear antigen, others acquire immu- after their deposition (Cochrane et al. 1959).
nological tolerance, and 10–20% form immune complexes that For years, the presumed pathogenesis of cutaneous SVV (also
are slowly removed from the circulation, resulting in chronic glo- referred to as leukocytoclastic vasculitis, see Chapter 38) has been
merulonephritis (Christian and Sergent 1976; Wilson and Dixon that circulating immune complexes are deposited in the small
1971). An explanation of the difference between acute and chronic blood vessels of the skin, with inflammation occurring after com-
serum sickness with respect to the development of arteritis has not plement activation. The ability of immune complexes to trigger the
been forthcoming. Despite the lack of vasculitis in animal models influx of polymorphonuclear cells and to induce the production of
of chronic serum sickness, current evidence implicates immune tumour necrosis factor-alpha (TNF), perforin, and Fas ligand has
complex deposition as the primary pathogenic mechanism in vas- been shown ex vivo (Nishimura et al. 2004). Activated neutrophils
culitis associated with chronic infections such as hepatitis B and release collagenase and elastase that, in conjunction with free oxy-
hepatitis C-associated mixed cryoglobulinaemia, which primarily gen radicals, result in necrosis of vessels (Sindrilaru et al. 2007).
affect small- or medium-sized vessels (Hoffman 1997; Belizna et al. Peptide products of proteolysis of vessel wall constituents, such as
2009), and in IgAV as discussed below in this Section. proline-glycine-proline (PGP) released from collagen, may induce
A likely explanation for the absence of vasculitis is that other further neutrophil chemotaxis (O’Reilly et  al. 2009). Cytokines
factors contribute to the quantity and quality of the host immune from T cells and endothelial cells are involved, and various anti-
response, thus accounting for the inconsistent expression of vas- bodies such as those reacting with endothelial cells may contrib-
culitis in animal and human immune complex disease. Cytokines ute to the complex phenomena of inflammation (Claudy 1998).
and adhesion molecules, as well as reactive oxygen species, proteo- Immunoglobulins and complement are seen on direct immunoflu-
lytic enzymes, and other mediators expressed by neutrophils and orescence of vasculitic skin lesions. Circulating immune complexes
endothelial cells, are undoubtedly necessary for generation of an may be detected but serum complement levels are not usually
inflammatory response in immune vasculitis. The mere presence of decreased (Kammer et al. 1980; Sunderkotter et al. 2001).
circulating immune complexes is not sufficient to produce vasculi- Intradermal injection of histamine and epinephrine into
tis, as indicated by the paucity of vasculitis in animals injected with uninvolved skin of patients with cutaneous vasculitis repro-
preformed immune complexes (McCluskey and Fienberg 1983). duces the histopathological findings of spontaneous lesions.
Cochrane and colleagues showed that the release of vasoactive Although deposition of immunoreactants can be demonstrated in
amines from platelets is necessary for tissue deposition of immune normal-appearing skin of patients with leukocytoclastic vasculitis,
complexes in experimental acute serum sickness (Cochrane 1971). increased deposition of immunoglobulin and complement follows
Pretreatment with antihistamines or depletion of platelets clearly the injection of histamine in a few hours, but neither is detected
suppressed the deposition of immune complexes and prevented after 24 hours (Braverman and Yen 1975; Gower et al. 1977). Using
vasculitis. Physical properties of the immune complexes are also direct immunofluorescence, Grunwald and colleagues were able
important as only those with a sedimentation coefficient greater to demonstrate immunoreactants in skin biopsies of vasculitis of
than 19S are deposited in vessel walls (Cochrane 1971). Other spe- various causes and of early, mature, and healing stages. Biopsies
cific factors that influence their proinflammatory nature include the from 92% of patients were positive, in increasing concentration,
ability of the immune complex to activate complement, the anti- for fibrinogen, albumin, IgG, IgM, IgA, and C4 (Grunwald et al.
gen–antibody combining ratio, and structural and haemodynamic 1997). Gower and colleagues also demonstrated that the deposition
differences between various blood vessels (Haynes 1992). The ten- of immune complexes and complement precedes the inflammatory
dency for immune complexes to deposit at vessel branching sites, infiltrate (Gower et al. 1977).
heart valves, and dependent areas is partly explained by changes The ANCA-associated vasculitides (MPA, GPA, and EGPA)
in hydrodynamic forces in those regions. Areas of the vasculature have generally been characterized as ‘pauci-immune,’ meaning that
with disturbed blood flow exhibiting low and reciprocating shear immunoglobulin and complement do not seem to play a prominent
stress, and a high shear stress gradient, exhibit changes in endothe- role in their pathogenesis as they are not usually found in AAV his-
lial cell morphology and orientation, increased cell turnover, and topathology (Jennette et al. 2013). Further, GPA and EGPA exhibit
gene expression towards a proinflammatory, procoagulant, and granuloma formation, which suggests an underlying cellular patho-
proapoptotic pattern (Sheikh et al. 2003; Chiu and Chien 2011). genic mechanism; however, more recently the complement system
Detection of immunoglobulin and complement in human vascu- has been implicated as a fundamental player in the development
litic lesions by immunofluorescent or immunochemical techniques of these disorders (Wilde et al. 2011). Activation of complement
provides circumstantial evidence of immune complex mediation of by ANCA and other antibodies such as antiendothelial antibod-
disease. These proteins are most readily detected in cutaneous vas- ies generates anaphylatoxins C3a and C5a, which act as potent
culitis, but inconsistently in systemic vasculitis. Definitive support chemotactic agents that intensify neutrophil activation (Savage
for a pathogenic role for immune complexes requires the detection 2011; Schreiber et al. 2009; Yuan et al. 2012). Alternate pathway
of a relevant antigen and specific antibody simultaneously in the complement activation can be detected in renal biopsies of patients
circulation and in sites of vascular injury. This has been a fruitless with MPO-AAV (Xing et  al. 2009). Furthermore, evidence from
CHAPTER 7 pathogenesis of vasculitis 73

an animal model of MPO-ANCA-induced glomerulonephritis and (Cornacoff et al. 1983). CR1 is also a potent inhibitor of the comple-
vasculitis also indicates that this activation process involves the ment cascade, serving as a cofactor for Factor I, the serine protease
alternative pathway of complement rather than the classical path- that cleaves and inactivates C3b (Fearon 1984; Medof et al. 1982).
way as it was abrogated in complement factor B-deficient mice but In systemic lupus erythematosus (SLE) patients, relative deficien-
not in C4-deficient animals (Xiao et al. 2007). cies of CR1 have been noted on erythrocytes and there are reduced
IgAV is a relatively common, usually transient form of vasculi- expression levels of other complement regulatory molecules on
tis that mainly affects children. It is manifested by raised purpuric red cells and leukocytes (Alegretti et al. 2012). The mechanism for
lesions in skin, which on biopsy exhibit deposition of IgA, primar- these decreased levels has not been formally demonstrated to date
ily IgA1, in dermal capillaries and postcapillary venules, along with but is probably related to their consumption during the process of
angioedema, arthritis, GI involvement, and in about 40% of cases, controlling complement activation by immune complexes on the
glomerulonephritis (Lau et al. 2010; Kawasaki 2011). IgA immune cell surface.
complexes do not activate the classical complement pathway and The frequent conjunction of genetic deficiencies of complement
are ineffective in fixing C3 (Schifferli et al. 1986); detectable clas- and immune complex diseases implies an important pathogenic
sical pathway activation in IgAV (low C1q, C3 and C4) occurs interaction between the two (Genel et al. 2005) (see Chapter 5).
only transiently in a minority of patients (Garcia-Fuentes et  al. Deficiencies of the early classical pathway components C1q, C1r,
1978; Lin et  al. 2012). The membrane-attack complex, C5b-9, is C1s, C4, and C2 are strongly associated with immune complex
a component of IgA and C3 deposits in vessel walls of the skin, disease; vasculitis generally occurs in the context of SLE and
in capillary walls, and in mesangium of glomeruli of patients with other complex autoimmune disorders that probably develop due
IgAV nephritis (Kawana and Nishiyama 1992; Rauterberg et  al. to failure of efficient clearance of self-antigens and apoptotic cells
1987). Interestingly, analysis of kidney biopsies from a group of 31 (Lewis and Botto 2006; Grammatikos and Tsokos 2012; Lipsker
patients with IgAV nephritis revealed that classical pathway acti- and Hauptmann 2010; Arason et al. 2010). Complete deficiencies
vation apparently occurred primarily in the patients with mixed of some complement control proteins, particularly those associ-
IgA1/IgA2 mesangial deposits (16 patients) while the complement ated with regulation of assembly of the membrane attack complex
components in the glomeruli of patients with only IgA1 mesangial and degradation of C3b, are also clearly associated with mani-
deposits (15 patients) suggested activation through the alternative festations of vasculitis, particularly glomerulonephritis (Genel
and lectin pathways (Hisano et al. 2005). et al. 2005; Fervenza et al. 2012; Pettigrew et al. 2009; Zipfel and
Some patients with IgAV continue to experience exacerbations Skerka 2009).
of glomerulonephritis with respiratory infections and without the Thus, experimental and clinical evidence affirms the critical
other features of IgAV. This has led some to propose that IgAV is role of complement in processing immune complexes and apop-
one end of a spectrum, with IgA nephropathy (IgAN) on the oppo- totic cells so that they are targeted for elimination without caus-
site end. IgA1 appears to play a pivotal role in IgAV and IgAN with ing inflammation or overwhelming peripheral tolerance. There
increased serum levels of IgA1, IgA1, and IgG containing immune are several plausible reasons for failure of the normal processes
complexes, and IgA rheumatoid factors. First described in IgAN for eliminating immune complexes that could lead to the develop-
almost two decades ago and then also observed in patients with ment of vasculitis and other manifestations of immune complex
IgAV, terminal galactosylation of O-linked oligosaccharides in the disease:  (1)  depletion or deficiency of complement components;
hinge region of IgA1 is deficient and the aberrant glycosylation pat- (2) failure of various antibody classes within immune complexes to
tern may be familial (Allen et al. 1995; Saulsbury 1999; Feehally bind complement; (3) depletion or blockade of CR1; and (4) impair-
and Allen 1999; Kiryluk et al. 2011). The second stage in the devel- ment of tissue macrophage function (Haynes 1992). There is a com-
opment of disease appears to occur through the development of plex interaction between complement components and activation
autoantibodies against the under-glycosylated regions of IgA, per- products, and endothelial cells that stimulates their proinflamma-
haps stimulated by microbial oligosaccharides during the course tory status (Fischetti and Tedesco 2006). Acosta et al. (2004) have
of an infection (Lai 2012). Progression of IgAN has been found to reviewed complement and complement regulatory proteins, focus-
correlate with autoantibodies to galactose-deficient IgA1 (Kiryluk ing on blood vessels and vasculitis diseases (Acosta et  al. 2004).
et  al. 2011). Recent observations suggest that galactosylation of Ghebrehiwet and colleagues (Ghebrehiwet et al. 2006) have pro-
IgG1 has an important ability to impair complement activation posed a model of inflammation in which factors such as immune
through dectin-1-mediated recruitment of FcγRIIB, suggesting complexes, viruses, and bacteria convert the vascular endothelium
that this type of glycosylation may be of general importance in the into a procoagulant and proinflammatory surface and enhance
regulation of inflammation (Karsten et al. 2012). expression of cell-surface molecules including gC1qR, a compo-
The C3b component of immune complexes binds to a specific nent of the receptor for the globular heads of C1q. This might lead
receptor, complement receptor type 1 (CR1, CD35). This binding to high-affinity C1q binding and cell production of proinflamma-
prevents immune complex interaction with other structures such as tory factors and generation of bradykinin and possibly coagulation.
vascular endothelium. As much as 90–95% of the CR1 in humans
is located on the surface of erythrocytes (Schifferli et al. 1986), but Antineutrophil cytoplasmic
it is also found on polymorphonuclear leukocytes, macrophages,
B lymphocytes, some T lymphocytes, dendritic cells in germinal antibodies (ANCA)
centres, and glomerular podocytes. Activation of the classical com- Since the discovery in the early 1980s of IgG antibodies against cyto-
plement pathway results in the deposition of C3b in immune com- plasmic proteins expressed by neutrophils in patients with systemic
plexes, which then bind to CR1 on red blood cells and are then vasculitis (Davies et al. 1982; Hall et al. 1984; van der Woude 1985;
removed from the circulation by spleen and liver macrophages van der Woude et al. 1985), a wealth of literature has developed on
74 SECTION 2 basic science

the role of ANCA in systemic vasculitis (Savage 2011; Kallenberg mediators of acute inflammation and to undergo more com-
2011; Danila and Bridges 2008; Wilde et  al. 2011). Jennette and plex processes such as extrusion of neutrophil extracellular traps
Falk, who first described pANCA in patients with necrotizing glo- (NETs) and release of microparticles (Jennette et al. 2011; Jennette
merulonephritis and microscopic polyangiitis, and who identified et al. 2006) (Figure 7.1). NETs contain the key antigens MPO and
the antigen as myeloperoxidase (MPO), have summarized their PR3 that may somehow then trigger an autoantibody response
views of the role of ANCA in pathogenesis, as well as the evidence (Kessenbrock et al. 2009). Likewise, neutrophil-derived micropar-
supporting their pathogenicity (Jennette and Falk 1998; Jennette ticles contain adhesion molecules as well as both MPO and PR3
et  al. 2011). Early supportive evidence noted by these and other and can be found in increased levels in the circulation of patients
groups included the frequent occurrence of ANCA in the sera of with ANCA vasculitis (Hong et al. 2012). Figure 7.1 illustrates a
patients with vasculitis and necrotizing crescentic glomerulone- hypothetical sequence of pathogenic events that could result in
phritis, the pauci-immune nature of the vasculitic lesion, and the ANCA-mediated vascular inflammation.
response of these diseases to immunosuppressive drugs. They also Antigenic mimicry, similarities of microbial proteins to human
documented the induction of ANCA and vasculitis by medications gene products expressed in neutrophils, is a potential mechanism
such as propylthiouracil and hydralazine and the disappearance of for the development of ANCA for which there is increasing evi-
the vascular inflammation after cessation of the offending medica- dence. In 1994, a hypothetical mechanism, called the Theory of
tion. Newer studies have implicated the dysregulation of PR3 and Autoantigen Complementarity by its proponents (Wilde et  al.
MPO expression by epigenetic mechanisms in neutrophils, impor- 2011; Pendergraft et  al. 2005; Pendergraft et  al. 2004), was pub-
tant roles for neutrophil extracellular traps (NETs), and micropar- lished in which anti-idiotypic antibodies produced against a pol-
ticles in the pathogenesis of ANCA vasculitis (Jennette et al. 2011). ypeptide encoded by the complementary gene sequence for PR3
Thus, there is considerable evidence supporting a role for ANCA in (cPR3) were put forth as a mechanism for the induction of ANCA.
pathogenesis of vasculitic diseases. Current recommendations for While this mechanism has not been validated by other groups to
ANCA testing, methodologies, and clinical utility in patients with date (Tadema et al. 2011), it is of interest that Staphylococcus aureus
suspected or known vasculitic syndromes are discussed in detail in has gene sequences homologous to the cPR3 sequence, bringing up
Chapter 6. the possibility that staphylococcal antigens bearing close antigenic
similarity to the cPR3 protein could be involved in AAV pathogen-
Antigenic specificity and diagnostic utility esis (Wilde et al. 2011). Associations of onset and flares of AAV
ANCA have been thought to be specific for cytoplasmic antigens have been reported by Tervaert and colleagues to be associated with
located in primary granules of neutrophils and lysosomes of mono- increased carriage of Staphylococcus aureus (Kallenberg et al. 1996;
cytes; however, Witko-Sarsat and colleagues identified proteinase Stegeman et al. 1996; Stegeman et al. 1994).
3 (PR3), a major target antigen, in plasma membrane secretory The association between anti-lysosomal-associated membrane
vesicles (Witko-Sarsat et  al. 1999). By indirect immunofluores- protein 2 (LAMP-2) antibodies and vasculitis is not new, dating
cence (IIF), ANCA are identified as: cytoplasmic (cANCA), with back to work done in 1995, in which these antibodies were discov-
coarse granular staining of cytoplasm; perinuclear (pANCA), with ered in 16 of 17 patients with pauci-immune necrotizing crescen-
staining of the nucleus and perinuclear area; and atypical ANCA. tic glomerulonephritis (Kain et al. 1995), but it is still contentious.
The antigens responsible for cytoplasmic IIF are usually PR3 and LAMP-2 is a glycosylated protein lining neutrophil granules and
those detected as perinuclear are often MPO. Both proteins are cell surface; it is a major constituent of the lysosomal membranes,
now known to be also expressed on the plasma membrane of neu- is involved in HLA class  II antigen presentation, and is a ligand
trophils and surface expression is up-regulated by inflammatory for E-selectin (Flint and Savage 2012). These connect anti-LAMP-2
cytokines such as TNF-α (Wilde et al. 2011). Antibodies to other antibodies to clearance of intracellular pathogens and the immune
neutrophil constituents such as elastase, cathepsin G, lysozyme, and response, thus supporting the argument that these antibodies
lactoferrin produce pANCA (or atypical pattern); but the ANCA contribute to the pathogenicity of ANCA. In later work the same
of diagnostic significance for vasculitis are those directed against group (Kain et al. 2008) immunized rats with anti-LAMP-2 IgG,
PR3 and MPO. The antigen epitopes of MPO-ANCA have been precipitating a crescentic glomerulonephritis. They further showed
analysed (Erdbrugger et al. 2006). These investigators have found that these antibodies activated human neutrophils as efficiently as
that MPO-ANCA do not target a single epitope, but rather a small anti-PR3 autoantibodies, providing evidence that molecular mim-
number of regions of MPO, primarily in the carboxy-terminus of icry is a potential initiating factor in anti-LAMP-2 antibody forma-
the heavy chain. tion. Homology to LAMP-2 is shared with the bacterial adhesion
The process by which MPO and PR3 induce the production of protein FimH, which is expressed on a variety of Gram-negative
ANCA is of fundamental importance in the pathogenesis of ANCA bacteria. The work of Kain et al. could not be replicated in work by
vasculitis. Van Rossum and co-workers have shown that patients another group; only 21% of 103 patients with ANCA-positive glo-
with ANCA-positive vasculitis have increased constitutive expres- merulonephritis had anti-LAMP-2 antibodies, and these antibod-
sion of PR3 on resting neutrophils compared to healthy controls ies were also detected in 16% of 104 control subjects with urinary
(van Rossum et  al. 2003). Additionally, priming of neutrophils tract infection (Roth et al. 2012). However, in a follow-up study to
from ANCA-positive patients (and also from SLE patients, but their earlier work, Kain and colleagues at three university medical
not rheumatoid arthritis (RA) patients) produced significantly centres in Europe found antibodies to LAMP-2 in 89%, 91%, and
increased membrane PR3 expression compared to normal con- 80% of untreated ANCA-positive (MPO or PR3) vasculitis sera.
trols (van Rossum et  al. 2007). As reviewed by Jennette et  al., The LAMP-2 antibodies disappeared rapidly with treatment. The
experimental studies from many laboratories have shown that authors concluded that with robust assays, the antibodies can be
both PR3-ANCA and MPO-ANCA activate neutrophils to release detected in most Europeans with ANCA-positive vasculitis and
CHAPTER 7 pathogenesis of vasculitis 75

(a) ANCA
ANCA antigen
Cytokine
Cytokine receptor

Fc receptor
Adhesion molecule
Adhesion molecule receptor

(c)
Neutrophils

(b) Activated neutrophils

Endothelial cells

Elastic membrane  Disruption of elastic membrane


 Apoptosis
 Necrosis

Fig. 7.1 Events in the pathogenesis of ANCA small-vessel vasculitis that have been observed in vivo (a) Cytokines or other priming factors induce neutrophils to
express more ANCA antigens at the cell surface, where they are available for binding to ANCA. (b) ANCA binds to ANCA antigens, which activates neutrophils by both
neutrophil Fc receptor engagement through the Fc portion of ANCA and antibody-specific ligation of ANCA antigens on the neutrophil surface. (c) Neutrophils that
have been activated by ANCA interact with endothelial cells via adhesion molecules and release toxic factors that cause apoptosis and necrosis. They also produce
neutrophil extracellular traps (NETs) and microparticles containing ANCA antigens that may feed back to initiate and amplify ANCA production. Modified with
permission from J Am Soc Nephrol, 17, Jennette, J.C., Xiao, H., and Falk, R.J. Pathogenesis of vascular inflammation by anti-neutrophil cytoplasmic antibodies. 1235–42.
© American Society of Nephrology, 2006.

that large-scale studies must be done to determine the significance of 70 patients with tuberculosis; in 7% of 30 with interstitial lung
of these findings (Kain et al. 2012). disease; and in 4% of 100 normal controls (Pradhan et al. 2004). By
It is important to keep in mind that positive tests for ANCA are ELISA analysis, 47.6% of the ANCAs were anti-MPO; 28.6% were
found in disorders other than vasculitis. Cocaine use, especially anti-PR3; and 19.1% were antilactoferrin. It should be noted that in
drug that has been adulterated with the antihelminth drug levami- this study, the prevalence of antinuclear antibodies and antihistone
sole, produces a GPA-like presentation complete with cutaneous antibodies was unusually high (24.3% and 21.4%, respectively),
vasculitis, nasal septal perforations, and ANCA, often positive to suggesting possible drug induction of these antibodies and ANCA.
both PR3 and MPO, as well as antibodies to other neutrophil pro- Similarly, a study from Mexico showed that 44% of 45 patients with
teins such as elastase (Neynaber et al. 2008; Rongioletti et al. 1999; tuberculosis were ANCA positive; 40% were PR3-ANCA positive
Graf et al. 2011; McGrath et al. 2011; Pearson et al. 2012). ANCA by ELISA testing (Flores-Suarez et  al. 2003). The ANCA results
directed against bactericidal/ permeability-increasing protein were not related to stage of disease, co-morbidities, or known
(BPI) have been described in patients with inflammatory bowel ANCA-inducing pharmacotherapy.
disease, cystic fibrosis, and rheumatic diseases, and ANCA reacting
with azurocidin have been found in 18–25% of sera from patients Pathogenicity of ANCA
with GPA, cystic fibrosis, and chronic active hepatitis (Cooper There is compelling clinical and experimental evidence that
et al. 2000). pANCA was detected, using a commercial antibody MPO-ANCA are pathogenic in vasculitis, and laboratory evidence
panel, in 70% of 54 children and young adults with ulcerative colitis that PR3-ANCA can amplify locally produced inflammation.
and in 18% of 81 children and young adults with Crohn’s disease The clinical evidence is represented by transplacental transfer of
(Zholudev et al. 2004). A study from India detected ANCA in 30% MPO-ANCA from a mother to a 33-week gestational age neonate
76 SECTION 2 basic science

whose cord blood contained IgG MPO levels identical to that of the and other effector functions leading to tissue damage (Kelley et al.
mother’s serum, and which resulted in neonatal pulmonary haem- 2010; Ben-Smith et al. 2001; Ralston et al. 1997). Aggregation of
orrhage and renal disease. The neonate was treated with high-dose both the MPO or PR3 ligands and the FcR through the Fc portion
glucocorticoids and exchange transfusion, and became asymp- of ANCA appear to be important for efficient neutrophil activation
tomatic (Bansal and Tobin 2004). A similar case involving trans- by ANCA (Kettritz 2012).
placental MPO-ANCA with resulting pulmonary–renal disease in If ANCA are pathogenic, then targeted B cell therapy would be
the neonate was reported by Schlieben and colleagues (Schlieben expected to produce amelioration of disease; conversely, if they are
et al. 2005). an epiphenomenon, then elimination of pathogenic B-cell clones
In contrast, no clear reports exist of similar disease induced by would not be expected to affect patient outcomes. Guerry and
transfer of PR3-ANCA to a neonate from a mother with AAV. colleagues recently published treatment recommendations with
Alfhaily and colleagues reported the development of GPA at 30 regard to targeted B-cell therapy based on a review of 43 studies,
weeks’ gestation in a pregnant woman who was treated with cor- which included a preponderance of small uncontrolled series but
ticosteroids, IVIG, and azathioprine and then placed on mainte- did include two randomized controlled studies (Guerry et al. 2012).
nance with steroids and azathioprine for the remainder of the The evidence demonstrates unequivocal efficacy of targeted B-cell
pregnancy, with subsequent delivery at 37 weeks of a healthy elimination with rituximab (anti-CD20) in AAV. B-cell elimination
PR3-ANCA-positive infant (Alfhaily et  al. 2009). Devakumar in newly diagnosed patients with AAV or patients with relapsing
reviewed 42 pregnancies in 33 GPA patients (Devakumar et  al. disease is effective and at least comparable to treatment with cyclo-
2010). Most of the infants (16 of 19) born to mothers who were phosphamide. Thus, the aggregate of clinical data strongly supports
newly diagnosed or who experienced a flare during the second the hypothesis that antibodies, very probably ANCA, play an essen-
or third trimester did not show any signs of illness, although the tial role in the pathogenesis of AAV.
mothers were given treatment, usually corticosteroids and azathio- Mouse models of AAV have been developed for MPO-ANCA
prine. Although the ANCA specificity is not stated in this review, (Xiao et al. 2002; Schreiber et al. 2006) but evidence of PR3-ANCA
11 of the 16 were positive or strongly positive, one mother was pathogenicity has been elusive, primarily because ANCA epitopes
negative and five results were not available. Of the three infants on human PR3 are not shared by the murine homologue (Wiesner
who did not survive, one died when the pregnancy was termi- et al. 2005). Experiments by Little and colleagues generated chi-
nated at 18 weeks, one was stillborn at 25 weeks to a mother who meric mice by engrafting human haematopoietic stem cells into
had received several pulses of cyclophosphamide before she was irradiated mice. The mice were then given human IgG from patients
known to be pregnant (Lima et al. 1995), and the third died with with PR3-ANCA-positive lung and kidney vasculitis. Other mice
his mother 4 weeks after the mother was diagnosed with fulminant were treated with IgG from normal human controls or patients
GPA at 21 weeks’ gestation. Therefore, in contrast to two convinc- with non-vasculitic renal disease. Thirty-nine per cent of mice
ing reports of MPO-induced pulmonary–renal disease following given PR3-ANCA IgG developed haematuria and pauci-immune
passive transplacental transfer to human neonates, there seems glomerulonephritis and punctate bleeding on the surface of the
to be a paucity of similar data with PR3-ANCA. It is possible that lungs (Little et al. 2012).
PR3-ANCA-induced vasculitis induced by transplacental transfer In contrast to the other systemic vasculitides, there is little, if any,
of the pathogenic antibodies is more sensitive to anti-inflammatory evidence for a direct pathogenic role of ANCA in EGPA (Hellmich
medications, such as corticosteroids, than MPO-ANCA. Also, et al. 2005). Only about 40% of EGPA patients are ANCA positive,
because ANCA typically decrease during treatment and almost all with about 90% of those positive for pANCA, mostly against MPO,
the mothers presenting with new-onset disease or relapse received which again argues against an essential pathogenic role for ANCA
anti-inflammatory therapy, the actual serum level of ANCA in the in this form of vasculitis (Szczeklik et al. 2013). Pagnoux and col-
neonate would have been of interest but unfortunately was almost leagues have suggested that ANCA are probably more important
never measured. in the vasculitic manifestations of EGPA, for example glomerulo-
Ex vivo and in vivo evidence demonstrate that ANCA can nephritis, while eosinophil tissue infiltration and associated cyto-
directly bind to PR3 and MPO antigens on neutrophils, produc- toxicity are responsible for non-vasculitis manifestations such as
ing activation. PR3 is present on the membrane of unstimulated cardiomyopathy (Pagnoux et al. 2007; Pagnoux and Guillevin 2010;
neutrophils from AAV patients and increases after priming of the Oka et al. 2011).
cells by a variety of inflammatory mediators, including TNF, IL-1,
IL-6, IL-18, N-formyl-Met-Leu-Phe (fMLP), and complement C5a Interaction of ANCA-activated neutrophils
(Kettritz 2012). Van Rossum et al. have shown PR3-ANCA in undi- with endothelial cells
luted serum or plasma from PR3-ANCA-positive GPA patients Chapter 4 contains a full discussion of endothelial activation, and
bind to TNF-primed and PMA-stimulated PMN that express adhesion and transmigration of neutrophils in vasculitis. Cohen
PR3 on their membrane (Van Rossum et al. 2005). Double stain- Tervaert and colleagues have argued that ANCA are a primary
ing for PR3 and IgG demonstrated that IgG in plasma or serum pathogenic factor in renal vasculitis, mainly through their action
from PR3-ANCA-positive patients only bound to those PMN that in augmenting leukocyte–endothelial interactions (Heeringa et al.
expressed PR3 and not to PMN that lacked PR3 membrane expres- 2005; van Paassen et al. 2007). Using isolated human neutrophils,
sion. After binding, the Fc portion of ANCA ligates the low-affinity Radford et  al. found that ANCA treatment promoted the trans-
IgG receptors FcγRIIA (CD32B) and FcγRIIIB (CD16B) on the migration of neutrophils through the endothelium (Radford et al.
neutrophil membrane, a cross-linking process that leads to activa- 2001). Little et al. used a rat model of ANCA-associated experimen-
tion with subsequent oxidative burst, granule secretion, expression tal autoimmune vasculitis, induced by immunization with human
of adhesion molecules, inflammatory cytokine gene transcription MPO, to directly analyse the effect of ANCAs on leukocyte–venular
CHAPTER 7 pathogenesis of vasculitis 77

wall interactions in vivo by intravital microscopy (Little et al. 2005). by the neutrophil serine proteases proteinase 3 and elastase (Yang
These rats developed anti-MPO antibodies directed against rat et al. 1996). Harper et al. reported that PMN from patients with
leukocytes, showed pathological evidence of small-vessel vasculi- active vasculitis became apoptotic at a greater rate than those of
tis, and had enhanced leukocyte adhesion and transmigration in controls and that opsonization of apoptotic PMN with ANCA
response to the chemokine Gro-alpha (CXCL1 (CXC ligand 1)). enhanced recognition and phagocytosis by macrophages, leading
In other studies from the same laboratory it was shown that poly- to increased secretion of IL-1 and IL-8, thus possibly contributing
clonal ANCA from patients, and chimeric PR3-ANCA, induced to chronic inflammation (Harper et al. 2001).
the release of microparticles from primed neutrophils (Hong et al. IFN-activated neutrophils are able to release TNF-related
2012). The microparticles expressed PR3 and MPO and bound apoptosis-inducing ligand (TRAIL/APO2 ligand), a molecule
endothelial cells by a CD18-dependent mechanism, increasing involved in apoptosis of tumour cells and virus-infected cells, as
endothelial adhesion molecules, production of endothelial reactive well as immunoregulatory functions on activated T lymphocytes.
oxygen intermediates, and release of IL-6 and IL-8. The microparti- The majority of the newly synthesized TRAIL is retained intracel-
cles also promoted the generation of thrombin. Neutrophil-derived lularly, can be rapidly mobilized to the cell surface, and secreted
microparticles thereby likely play an important role in the patho- following exposure to a variety of proinflammatory mediators
genesis of AAV. such as TNF, LPS, formyl-methionyl-leucyl-phenylalanine (fMLP),
Wikman et al. studied eight patients with PR3-ANCA-positive CXC chemokine ligand 8/IL-8, insoluble immune complexes, and
acute vasculitis, examining PMN expression of adhesion molecules, heat shock protein Gp96 (Cassatella et al. 2006). This represents a
Fc receptors, and PR3 at study entry and at 1, 3, 6, and 9 months novel mechanism by which neutrophils may be involved in immu-
(Wikman et  al. 2005). Additional markers of inflammation and nomodulation relevant to ANCA-associated vasculitis.
endothelial activation (IL-8 and soluble VCAM-1) were analysed Although apoptosis is an important process in ANCA-associated
at the same times. The main finding was an activated neutrophil vasculitis, proliferation is a prominent feature in the glomerulone-
adhesion phenotype at diagnosis and after 1 month, with normal- phritis of the necrotizing small-vessel vasculitides. Kettritz et  al.
ized expression of adhesion molecules at 3–9 months. This may be examined apoptosis, apoptosis-regulating proteins, and prolifera-
germane to endothelial damage seen in vasculitis. tion in skin biopsies from patients with leukocytoclastic vasculitis
and renal biopsies from patients with ANCA-associated vasculi-
Release of toxic factors that cause apoptosis and necrosis tis, ANCA-negative crescentic glomerulonephritis, and a variety
In vitro studies have demonstrated that ANCA IgG augment sev- of other renal lesions (Kettritz et al. 2006). There was a relatively
eral neutrophil functions, inducing degranulation (Falk et al. 1990), low rate of apoptosis and a relatively high rate of proliferation
the release of superoxide (Radford et al. 1999), production of neu- (assessed by Ki-67 expression) in renal biopsy tissue of patients
trophil NETs (Sangaletti et al. 2012; Kessenbrock et al. 2009), and with ANCA-positive necrotizing crescentic glomerulonephritis.
microparticle release, both from neutrophils (Hong et al. 2012) and
endothelial cells (Erdbruegger et  al. 2008). Cytokines released
after ANCA activation of neutrophils include IL-1β (Brooks Role of antiendothelial cell antibodies
et  al. 1996)  and IL-8 (Cockwell et  al. 1999). In addition, ANCA A role for AECA in the pathogenesis of vasculitis has been inferred
induce neutrophils to firmly adhere to endothelial cells, leading from their presence in GPA, MPA, and KD, as well as in vasculitis
to increased migration across the endothelial barrier (Radford associated with SLE and RA. Despite the data presented below in this
et al. 2001). Section, it still remains unclear whether AECA are contributors to
Von Willebrand factor (vWF), a glycoprotein involved in arterial the pathogenesis of vasculitis or are a result of vasculitis-associated
thrombus formation, is secreted into the circulation by endothelial inflammation (Eichhorn et al. 1996; Belizna et al. 2006; Cid et al.
cells (Vischer 2006). Lu et al. have reported an analysis of the fac- 2004; Guilpain and Mouthon 2008). One finding that argues
tors that control the neutrophil respiratory burst and endothelial against the importance of AECA in vasculitis is their presence in
release of vWF during neutrophil–endothelial cell interactions (Lu healthy individuals (Servettaz et al. 2008a; Servettaz et al. 2008b).
et al. 2006). Paradoxically, endothelial cells were shown to inhibit They are also being reported in cardiovascular disease states not
superoxide generation by ANCA-activated neutrophils. The release thought to be primarily inflammatory in nature. For example,
of vWF was sensitive to serine protease, but not NADPH oxidase Papadopoulos et al. found that healthy subjects with blood pressure
inhibition, suggesting that serine proteases play a more important in the high end of normal have significantly higher levels of both
role than reactive oxygen species in mediating endothelial injury IgG and IgM AECA than healthy subjects with normal blood pres-
during ANCA-associated systemic vasculitis. sure (Papadopoulos et al. 2006; Papadopoulos et al. 2008). Other
It is uncertain what factors prevent degradative enzymes and findings suggest that these antibodies may be increased in types of
oxygen radicals released from neutrophils adherent to the lumi- vascular inflammation not directly associated with autoimmunity
nal side of the vessel from being washed away or neutralized by such as atherosclerosis and infection (Elsheikh et al. 2010).
serum protease inhibitors. Work by Sindralaru et  al. (Sindrilaru AECA are currently still controversial in the pathogenesis of
et al. 2007) suggests that immune complexes induce tight interac- TAK (Arnaud et al. 2011). High titres of these antibodies have been
tions between the neutrophils and endothelial cells that shield the found in the sera of 18 of 19 patients with TAK (Eichhorn et al.
cytotoxic products from the blood stream. 1996). Salojin and colleagues have also found increased levels of
Reactive oxygen species resulting from degranulation and res- AECA in patients with vasculitis of large- and medium-sized arter-
piratory burst from neutrophils are important in triggering apop- ies (Salojin et al. 1996). Park et al. found that IgM AECA and IgG
tosis (Jacobson 1996)  involving the CD95 (Fas/APO-1) pathway AECA were more prevalent in patients with TAK than in controls
(Kasahara et al. 1997). Apoptosis of endothelial cells can be induced and that IgM AECA titres correlated well with the disease activity
78 SECTION 2 basic science

of TAK (Park et al. 2006), corroborating the findings of Brasile et al. of three AECA-positive systemic vasculitis patients was found
who detected AECA in 86% of 21 patients with systemic necrotiz- to up-regulate the expression of adhesion molecules (ICAM-l,
ing vasculitis and found that they correlated with disease activity VCAM-l, E-selectin) in vitro. In contrast, AECA-negative samples
(Brasile et al. 1989). Wang et al. (2010) also found a significantly had no effect (Carvalho et  al. 1999). AECA-mediated leukocyte
higher prevalence of AECA in TAK (Wang et al. 2011). Other inves- adhesion to endothelium is thought to be mediated, in part, by
tigators have reported AECA in 19% of 27 patients with GPA and in increasing the release of endothelium-derived IL-l. The analysis of
2% of 43 patients with microscopic polyangiitis (Varagunam et al. a panel of monoclonal AECA generated from a patient with TAK
1993). Holmen et al. reported that GPA is significantly associated by Blank et  al. supports the concept that AECA directly stimu-
with non-cytotoxic AECA that selectively bind surface antigens late EC. Again, there was evidence of adhesion of monocytes and
on unstimulated nasal, kidney, and lung endothelial cells (Holmen up-regulation of adhesion molecule expression, perhaps mediated
et al. 2004). by NF-κB activation (Blank et al. 1999). Finally, sera from aortic
The identity of the antigens inducing AECAs remains unknown, AECA-positive patients with TAK induces expression of E-selectin,
but they may be important in the initiation of GPA. Brasile et al. and VCAM1, suggesting that AECAs may cause vascular dysfunc-
reported preferential binding of the AECA to splenic and inferior tion through a variety of mechanisms (Chauhan et al. 2006).
mesenteric endothelium in comparison with aortic endothelium, Finally, AECA may act in the production of endothelial cell
findings that are compatible with visceral arterial involvement injury by triggering apoptosis (Tobon et  al. 2009; Guilpain and
in vasculitis (Brasile et al. 1989). Yu et al. analysed sera from 11 Mouthon). Tripathy and colleagues found that 36% of TA patients
patients with PTU-induced ANCA-positive vasculitis during active (24 of 66) had elevated levels of anti-annexin V antibodies, which
and quiescent phases, sera from ten patients with PTU-induced have been shown to induce apoptosis in endothelial cells, com-
ANCA but no evidence of vasculitis, and sera from 30 healthy con- pared to only 6% of controls (3 of 50) (P <0.001) (Tripathy et al.
trols (Yu et al. 2005). Soluble proteins from HUVECs were used 2003). The same group later found anti-aortic endothelial cell anti-
as antigens in an immunoblotting assay to determine the presence bodies in a large majority of TA patients (30 of 35, 86%) but only
of AECA. Ten of the 11 patients with active PTU-induced ANCA two of 21 age and sex-matched controls (9%) (P <0.001) (Chauhan
vasculitis were serum IgG-AECA-positive and six protein bands et  al.). They demonstrated that sera from all TA patients that
of endothelial antigens could be detected. In the quiescent phase, were positive for anti-aortic endothelial cell antibodies induced
seven of the ten positive sera were negative. None of the other sub- cellular activation and cell death as measured by adhesion mol-
jects tested were AECA positive. Chauhan et al. sought to delineate ecule expression (e-selectin and vascular cell adhesion molecule-1
the precise role of antigenic targets of AECA and the pathogenic (VCAM-1)), cytokine production (IL-4, IL-6, IL-8), and apoptosis
mechanism of antibodies directed against aortic AECAs in TAK in cultured endothelial cells as measured by the terminal deoxynu-
(Chauhan et  al. 2006). After isolating aortic AECAs using a cel- cleotidyl transferase dUTP nick end labelling (TUNEL) method.
lular ELISA, their antigenic targets were detected by immunoblot- None of the antibody-negative control sera induced significant
ting. Aortic AECAs were detected in 86% of patients with TAK and apoptosis and the other measures of cellular activation were also
in 9% of controls. Sera obtained from aortic AECA-positive TAK significantly lower.
patients recognized a total of nine antigens ranging in size from 18
to 200 kD. The most common antigen was a 60 to 65-kD triplet. The
precise molecular composition of the antigens binding to AECA
Drug-induced vasculitis
remains to be determined. The causes of most vasculitis are unknown; however, there has been
AECA can induce endothelial cell injury and lysis via clear evidence for many years that they can be caused by drugs.
complement-mediated cytotoxicity or antibody-dependent cel- While drug-induced vasculitis is most often cutaneous, it can be
lular cytotoxicity (ADCC). Using sera from patients with KD systemic and life-threatening. Among the earliest reported cases of
and systemic vasculitis, AECA have been shown to participate in serious drug induced vasculitis (DIV) were those due to metham-
complement-mediated cytotoxicity of cultured human umbilical phetamine abuse, with visceral and intracranial lesions, and a his-
vein endothelial cells (HUVEC) (Brasile et al. 1989; Leung et al. tological picture indistinguishable at times from classic PAN (Weiss
1986a; Leung et  al. 1986b), an observation not confirmed by all et  al. 1970; Margolis and Newton 1971). Drug-related vascular
investigators (Savage et  al. 1991). ADCC is a process by which inflammation can result from pharmacological or immunological
specific antibody binds to a target cell and engages a natural killer mechanisms. Pharmacological toxicity can be predicted in animal
cell via its Fc receptor, resulting in lysis of the cell. Its role in sys- toxicology studies and thus usually avoided but animal studies have
temic vasculitis is unclear, but ADCC has been demonstrated in not predicted immunologically mediated DIV in humans (Kerns
vitro against endothelial cells (del Papa et al. 1992). These activi- et  al. 2005). Drug-related hypersensitivity reactions have been
ties of AECA may account for observed elevations of vWF and explained by the hapten concept in which a small antigen cova-
factor VIII-related antigen in the serum of patients with vasculitis. lently modifies an endogenous protein. More recent studies have
Holmen et al. found that IFN-γ and TNF were cytotoxic to nasal shown that non-covalent drug presentation can lead to the activa-
and lung endothelial cells, that cell lysis was increased by the addi- tion of drug-specific T cells. In some instances, a drug-induced
tion of serum from patients with systemic vasculitis, and that GPA hypersensitivity may occur within hours of even first exposure to
serum caused agglutination of cytokine-stimulated nasal endothe- the drug, suggesting the reaction may be mediated by existing,
lial cells (Holmen et al. 2004). preactivated T cells that display cross-reactivity for the drug. This
Another mechanism by which AECA may contribute to the implies that drugs may bypass the checkpoints for immune activa-
pathogenesis of systemic vasculitis is through increasing leu- tion by the classical antigen processing mechanisms. Gerber and
kocyte adhesion to endothelial cells. Purified IgG from each Pichler propose this to explain the idiosyncratic nature of some
CHAPTER 7 pathogenesis of vasculitis 79

drug hypersensitivity reactions (Gerber and Pichler 2006). Choi adalimumab, and rituximab, which affect immunological media-
et al. looked at 30 patients with vasculitis and the highest titres of tors of inflammation, are themselves occasionally associated with
anti-MPO found in their laboratory over a 5-year period. Ten of vasculitis (Macdonald et  al. 2004; Mohan et  al. 2004; Srivastava
the 30 had taken hydralazine and three had taken propythiouracil. et al. 2005; Haraoui and Keystone 2006; Duffy et al. 2006; Orpin
Others of the 30 patients had been exposed to penicillamine, allopu- et  al. 2006). The risk is slight, and the vasculitis has most often
rinol, and sulfasalazine. The investigators recommended that clini- been cutaneous although there are rare reports of systemic dis-
cians should examine the histories of patients with MPO-positive ease. Bosentan, which is used in some patients with scleroderma,
vasculitis for exposure to these drugs (Choi et al. 2000). has also been reported to cause leukocytoclastic vasculitis (Gasser
The differentiation of idiopathic and drug-induced vasculitis et al. 2004).
(DIV) may be difficult, with few clues other than those from a care- A contentious association is that between asthma drugs and
ful chronological history of drug intake. A study of 16 patients with EGPA. Independent of its treatment, asthma has been recognized as
DIV and 47 with idiopathic vasculitis found a significant differ- a significant risk factor for EGPA since the latter’s identification as
ence in the mean eosinophil counts obtained on histopathological a distinctive entity. Since 1998, there have been numerous anecdo-
examination of biopsied tissue (mean eosinophil ratio of 5.20 in the tal reports of EGPA appearing after the introduction of leukotriene
former versus 1.05), suggesting that tissue eosinophilia can be a rel- receptor antagonists (LTRA) into the asthma treatment regimen. It
ative indicator of drug-induced disease (Bahrami et al. 2006). The has been postulated that with improvement of asthma following the
importance of recognizing DIV was underscored by the clinical administration of LTRA, EGPA is unmasked as glucocorticoids are
evidence of systemic vasculitis in 13% of the drug-induced group. decreased or withdrawn (Wechsler et al. 1999; Keogh and Specks
Recognition of an offending drug and its withdrawal usually 2003). There are contrasting reports of the appearance of EGPA in
leads to recovery; however, a literature review in 2002 reported that patients who have been treated with LTRA and low-dose or no glu-
death was the result in 10% of all published cases, predominantly in cocorticoids (Kobayashi et al. 2003; Oberndorfer et al. 2004; Conen
patients with multiple organ involvement. Furthermore, the interval et al. 2004; DuMouchel et al. 2004). Whether treatment of asthma
between first exposure and appearance of symptoms was highly var- with LTRAs can induce EGPA or not remains an unanswered ques-
iable, from hours to years, sometimes confounding recognition of tion, but recent epidemiological data strengthen an association.
the condition as drug induced. The drugs most frequently incrimi- Bibby and colleagues reviewed the FDA Adverse Event Reporting
nated were propylthiouracil, hydralazine, colony-stimulating fac- system database and found that 163 of 181 cases of suspected
tors, allopurinol, cefaclor, minocycline, D-penicillamine, phenytoin, drug-induced EGPA included LTRA as a suspect medication (Bibby
isotretinoin, and methotrexate (ten Holder et  al. 2002). Another et al. 2010). In most cases treated with an LTRA, EGPA could not be
recent review listed 100 drugs associated with vasculitis, with the explained by corticosteroid withdrawal or pre-existing EGPA.
caveat that the level of evidence for each causing vasculitis varied The association of antithyroid drugs (carbimazole, methimazole,
greatly, and the authors advised the readers to investigate agents propylthiouracil) and vasculitis, although uncommon, has been
individually when making clinical decisions. The drugs were subdi- accepted since its recognition in 1950 (McCormick 1950), and in
vided into the categories shown in Box 7.1 (Merkel 2001). 1993 Dolman et al. reported an association of antineutrophil cyto-
Postmarketing surveillance of drugs will often uncover adverse plasmic autoantibodies and propylthiouracil (PTU) related vas-
reactions not evident in premarketing controlled studies, as has culitis (Dolman et al. 1993). In a subsequent study of 56 patients
happened with some of the newer drugs used to treat rheumatic with Graves’ disease who were given PTU, anti-myeloperoxidase
disorders. Paradoxically, leflunomide, infliximab, etanercept, antibodies (MPO-ANCA) appeared in 21; of these, 12 had no
symptoms, but nine complained of myalgia or arthralgia. The
proportion of patients positive for MPO-ANCA increased with
Box 7.1 Drugs associated with vasculitis prolongation of PTU treatment (Sera et al. 2000). Several studies
have noted the appearance of MPO-ANCA following PTU treat-
Antimicrobials ment without development of vasculitis (Noh et  al. 2001; Wada
Vaccines et al. 2002). Bonaci-Nikolic and colleagues compared the clinical
Interferons and laboratory characteristics of 56 patients with idiopathic sys-
Antithyroid agents temic vasculitis (ISV) and 16 who became ANCA-positive during
Anticonvulsants/ antiarrhythmics treatment with PTU or methimazole (Bonaci-Nikolic et al. 2005).
Diuretics Kidney disease was present in 75% of the former and 18% of the
Other cardiovascular agents antithyroid drug-treated patients; skin lesions were noted in 62.5%
Anticoagulants/ thrombolytics of those taking the drugs, and 25% of ISV patients. MPO-ANCA,
Antineoplastic/ antimetabolites ANA, anticardiolipin antibodies, cryoglobulins, and low serum
Hematopoietic growth factors C4 were more common in the patients treated with antithyroid
NSAIDs drugs. Four of these 16 had drug-induced ANCA vasculitis and 12
Leukotriene inhibitors had lupus-like illness. A similar investigation in China examined
Psychoactive agents sera from 216 patients with hyperthyroidism and found ANCA in
Sympathomimetics 22.6% of PTU-treated patients versus 2.9% of untreated patients.
Miscellaneous Of the 216 samples, 33 were positive for ANCA and antineutro-
phil antibodies by indirect immunofluorescence. By ELISA testing
Adapted from Rheum Dis Clin North Am, 27, Merkel, P.A, Drug-induced
of 22 ANCA positive sera, antibodies were found against lactofer-
vasculitis, 849–62. Copyright (2001), with permission from Elsevier.
rin, elastase, MPO, and three against PR3 (Gao et al. 2004). It thus
80 SECTION 2 basic science

appears that PTU-induced ANCA may be due to polyclonal activa- 30 were specific for MPO. There was no overall significant differ-
tion of B cells. ence in genotype distribution or allele frequencies between patients
Vanek and Samuels found reports of 42 cases of antithyroid and controls, or between patients with nephritis and controls.
drug-related ANCA-positive vasculitis since 1992, noting that There was a trend towards increased homozygosity for the NA1
kidneys, skin, and the musculoskeletal systems were most often allele in patients with vasculitis, and this was significant in patients
involved, that the mean duration of treatment was 35  months who were MPO-ANCA positive. The investigators concluded that
‘prior to presentation’, and they described a patient with central FcγRIIIB receptor polymorphism is not a major factor predispos-
nervous system vasculitis with resolution of symptoms after with- ing to development of ANCA-positive vasculitis or nephritis (Tse
drawal of PTU (Vanek and Samuels 2005). ANCA have also been et al. 2000).
found in children with Graves’ disease who were treated with PTU; Another polymorphism that has been studied in this regard is
MPO-ANCA was present in 6.7% prior to and in 64% after treat- the SNP at amino acid residue 131 of FcγRIIA that encodes argi-
ment, but no child had vasculitis (Sato et al. 2000). nine (the R131 allele) or histidine (the H131 allele) (Allison 2012;
There are rare reports of ANCA-positive DIV in patients taking Kelley et al. 2011). These alleles differ in their ability to bind human
other drugs such as: hydralazine, allopurinol, penicillamine, sul- IgG2 and IgG3. Neutrophils from individuals homozygous for the
fasalazine, minocycline (one of whom had cutaneous polyarteri- FcγRIIARIIA-H131 allele bind more efficiently to IgG3 than do
tis), proton pump inhibitors, and cefotaxime (Pelletier et al. 2003; those from individuals with the FcγRIIARIIA-R131 allele and are
Jacobs-Kosmin et al. 2006; Feriozzi et al. 2000). As mentioned pre- the only human FcγR that bind IgG2. There is a similar variant at
viously, patients using cocaine adulterated with the antihelminth amino acid reside 158 of FcγRIIIA that encodes either valine (V158
drug levamisole have been increasingly noted to develop a GPA-like allele) or phenylalanine (F158 allele). In a study of 91 patients with
vasculitis that often includes nasal septal perforations, and ANCA GPA and 154 controls, patients homozygous for both the R131
to both PR3 and MPO (Pearson et al. 2012). allele of FcγRIIA and the F158 allele of FcγRIIIA were more sus-
It is obvious that almost any drug might cause vasculitis. Illicit ceptible to disease relapse (Dijstelbloem et al. 1999).
drugs, preservatives (Moneret-Vautrin et al. 1986), and drug addi- Tse and colleagues performed genotyping of 107 Caucasian
tives (Lowry et  al. 1994)  may also cause vasculitis syndromes. patients with ANCA-positive vasculitis (of whom 89 had renal dis-
Thus, a meticulous history of drug consumption should be an ease) and 100 ethnically matched controls (Tse et al. 1999). Of the
important part of the evaluation of any patient with vasculitis, patients with ANCA-positive systemic vasculitis, 75 had ANCA
either cutaneous or systemic, although there are many pitfalls in with specificity for proteinase 3 and 32 with specificity for myelop-
definitively determining that a particular drug is the aetiological eroxidase. As was the case with FcγRIIIB (Tse et al. 2000), there
agent in vasculitis. was no skewing in FcγRIIA allotypes in patients versus controls.
No significant increase of the FcγRIIA-H131 allotype was found
among patients, regardless of ANCA specificity, and no asso-
Genetic influences ciation between the FcγRIIA genotype and nephritis was found.
The frequent conjunction of genetic deficiencies of complement Other investigators have found no association between FcγRIIA
components and immune complex diseases implies an impor- genotypes and susceptibility to leukocytoclastic vasculitis (Groger
tant pathogenic interaction (see Chapter 5). Furthermore, as the et al. 1999).
molecular pathways important in the pathogenesis of vasculitis are Another gene relevant to vasculitis is that encoding the protease
unravelled, genetic variants in the proteins and other molecules inhibitor (Pi) alpha-1-antitrypsin (α1-AT) (SERPINA1), which
involved will be sought. Such studies have grown dramatically since is considered to be the major physiological inhibitor of PR3 and
the completion of the Human Genome Project and the rapidly elastase through its large circulating excess (Korkmaz et al. 2009).
expanding databases of single nucleotide polymorphisms (SNPs), The serum concentration of α1-AT is genetically determined by the
including those that tag particular haplotypes. For example, Oztas polymorphic Pi genes (Donato et al. 2012). In one of the earliest
et al. reported that serum IL-18 (and TNF) levels are increased in reported associations of α1-AT deficiency and vasculitis, Brandrup
BD (Oztas et al. 2005). Lee et al. subsequently sought an associa- and Ostergaard described a 2-year-old child with α1-AT defi-
tion between susceptibility to the BD and polymorphisms in the ciency of the PiZZ type and cutaneous vasculitis (Brandrup and
promoter region of the IL-18 gene (Lee et al. 2006). A full review of Ostergaard 1978). Mazodier et al. reported eight patients with sys-
reports of associations between vasculitides and polymorphisms in temic necrotizing vasculitis (six with microscopic polyangiitis, one
candidate genes is beyond the scope of this text, but selected exam- with GPA, and one with IgAV) and severe α1-AT deficiency and
ples are discussed below in this Section. reviewed six previously reported cases (Mazodier et al. 1996). In a
Because of the physiological role of Fc receptors in processing study of the potential association between ANCA-positive vasculi-
immune complexes, the genes encoding these proteins have been tis and α1-AT alleles in 198 ANCA-positive vasculitis patients and
extensively analysed for polymorphisms important in the patho- 2310 controls, cANCA patients showed an increased frequency of
genesis of vasculitis. It has been postulated that FcγR alleles are a the Z allele (0.055 versus 0.018 in controls), conferring a relative
genetic risk factor for disease expression in GPA (Tse et al. 2000), risk of about 3 (Griffith et al. 1996). In an analysis of 18 PiZ-positive
and these investigators explored the importance of FcγRIIIB poly- and 81 PiZ-negative PR3-ANCA patients, PiZ-positive patients
morphism as a risk factor for development of ANCA-associated had more disseminated disease and a slightly higher mortal-
systemic vasculitis/ nephritis. Receptor genotyping was deter- ity rate (Segelmark et al. 1995). Subsequently, a number of stud-
mined by allele-specific polymerase chain reaction on 101 patients ies have strengthened the argument for a pathogenic relationship
with ANCA-positive vasculitis, of whom 84 had nephritis, and 100 between α1-AT deficiency and anti-PR3-positive systemic vasculi-
matched controls. ANCA from 71 of the patients were PR3 positive; tis, including a genome-wide association study (Lyons et al. 2012;
CHAPTER 7 pathogenesis of vasculitis 81

Morris et al. 2011; Mahr et al. 2010). Patients with ANCA-positive In the 1970s, persistent hepatitis B infection was linked to some
systemic vasculitis may have a genetically determined protease/ cases of PAN and cryoglobulinaemic vasculitis (Gocke et al. 1970;
antiprotease imbalance (in the case of patients with genotypes asso- Levo et  al. 1977). Hepatitis B surface antigen (HBsAg)-antibody
ciated with α1-AT deficiency) or an acquired decrease in formation complexes were found in the circulation in early studies (Gocke
of PR3/α1-AT complexes by anti-PR3 ANCA. et al. 1970; Trepo et al. 1974), and deposits of HBsAg, immuno-
Audrain et  al. (2001) tested 191 persons with homozygous globulin, and complement were found in lesions of muscular arter-
(PiZZ) α1-AT deficiency for ANCA activity and compared them ies (Gocke et  al. 1970; Michalak 1978), dermal vessels (Gower
to 272 PiMM matched control subjects (Audrain et  al. 2001). et al. 1978), glomeruli (Combes et al. 1971), and vasa nervorum
The incidence of antibodies directed against PMN alpha granules (Tsukada et al. 1983). Hypocomplementaemia has been noted to
and human leukocyte elastase, but not PR3, MPO, lactoferrin, or sometimes accompany vasculitis associated with hepatitis B infec-
bactericidal/permeability increasing protein, was increased in tion, and in some cases cryoglobulins have been shown to con-
the PiZZ compared to the PiMM group. The authors concluded tain HBsAg and particles resembling the virus (Levo et al. 1977).
that ANCA not directed against MPO and PR3 may be found Substantial evidence therefore correlates the immunopathological
in PiZZ patients, who do not develop systemic vasculitis. Thus, events of hepatitis B-associated vasculitis with experimental serum
these authors hypothesize that α1-AT deficiency is not sufficient sickness.
to induce ANCA-positive vasculitis, but may act as a second hit In subsequent decades, HBV-associated PAN has become
to amplifying the immune response. Concomitant α1-AT defi- the most convincingly documented immunologically mediated
ciency and vasculitis may have treatment ramifications since puri- infection-associated systemic vasculitis (Cacoub and Terrier 2009).
fied preparations of human α1-AT are available to treat deficient Combined with symptoms and angiographic evidence, HBV posi-
patients. In what remains the best documented report of efficacy tivity is the single most important feature in confirming the diag-
of α1-AT replacement in a patient with α1-AT deficiency and vas- nosis of PAN by both the Chapel Hill 1990 criteria and the French
culitis, a 49-year-old man with α1-AT deficiency and refractory Vasculitis Study Group criteria (Henegar et al. 2008). In a series of
cutaneous vasculitis responded to treatment with α1-AT and expe- 949 vasculitis patients reported by Henegar and colleagues, 108 of
rienced continued relapses and relief of symptoms by further infu- 262 patients with PAN (41.2%) had HBV-associated PAN (Henegar
sions (Dowd et al. 1995). In addition, treatments decreasing plasma et al. 2008). A larger series of 348 PAN patients reported from the
α1-AT (such as plasmapheresis without plasma replacement) may French Vasculitis Study Group Database included 123 cases of
be deleterious in systemic vasculitis. In aggregate, these data sug- HBV-associated vasculitis (35.3%) (Pagnoux et al. 2010). Hurlburt
gest that physicians should consider α1-AT deficiency in patients et al. reported a series of 15 Alaskan Natives with HBV-associated
with systemic vasculitis. PAN in what used to be one of the highest areas of acute HBV
Finally, an SNP in the gene encoding an intracellular tyrosine infection in the world (Hurlburt et al. 2007). All of the cases were
phosphatase (PTPN22) has been found to be important in suscep- diagnosed between 1974 and 1989, and record review demon-
tibility to rheumatoid arthritis (Begovich et al. 2004) and subse- strated that in most cases the vasculitis could be shown to have
quently in other diseases, including type 1 diabetes, systemic lupus developed within months of acute infection. During the 1980s, an
erythematosus, and Hashimoto’s thyroiditis. Jagiello et al. examined aggressive campaign of HBV vaccination was carried out among
this PTPN22 SNP in 199 patients with GPA and in 399 healthy indi- Alaskan Natives and accompanying a decrease in incidence of HBV
viduals (Jagiello et al. 2005). They found that the PTPN22 620W infection from 200 cases/100 000 to less than 1:100 000 was a vir-
allele frequency was significantly increased in ANCA-positive GPA tually complete disappearance of PAN from the population—no
patients compared with healthy controls, particularly in those with cases have been reported since 1989, again arguing that it is not in
generalized GPA. The 620W allele appears to influence thresholds chronically infected individuals in whom this complication arises
for T-cell receptor signalling (Gregersen et  al. 2006), invoking a but those with relatively acute or subacute infection. In addition
variety of potential disease models involving central and peripheral to PAN, a scattering of reports of other forms of HBV-associated
T-lymphocyte tolerance. The precise mechanisms for these associa- vasculitis can be found in the literature but a convincing relation-
tions, however, remain unclear. ship is less certain due to the lower incidence. These include EGPA
(Domiciano et al. 2010), TAK (Ohta et al. 2003; Etgen et al. 2003),
IgAV (Ergin et al. 2005; Maggiore et al. 1984), and SVV (Surmali
Infectious agents as triggers of vasculitis Onay et al. 2007; Filer et al. 2001; Federman and Kirsner 1999).
Infection may trigger vasculitis by a variety of mechanisms, includ- The causative relationship between hepatitis C and cryoglo-
ing direct infection of the vascular wall or by indirect effects, for bulinaemic vasculitis is well-established (Agnello et  al. 1992;
example by molecular mimicry with cross-reactive microbial and Cacoub et  al. 2002; Ferri and Mascia 2006)  (see Chapter  41).
endothelial epitopes resulting in humoral and/or cellular damage to Cryoglobulinaemic vasculitis occurs in less than 5% of HCV
the vasculature, by cytokine-mediated mechanisms, or by immune infected patients while cryoglobulins can be detected in up to half
complex deposition (Belizna et al. 2009). It is likely that many infec- of such patients. Cryoglobulinaemia is associated with HCV in up
tious agents or foreign antigens that induce an immune response to 86% of cases, often also representing co-infection with HBV.
might result in vasculitis. There are numerous case reports in the HCV-associated cryoglobulinaemic vasculitis is typically a small-
literature with a variety of viral, bacterial, fungal, and even para- and medium-vessel leukocytoclastic vasculitis associated with
sitic pathogens suggesting an association with vasculitis. Chronic systemic symptoms such as fatigue and arthralgias. In a series of
infections such as those due to hepatitis B virus (HBV), hepatitis C 231 patients Ferri and Mascia found an overall prevalence of 92%
virus (HCV), and syphilis are particularly well-described (Jennette HCV positivity and 42% HBV positivity (Ferri and Mascia 2006).
et al. 2013). Consistent with the pathological mechanism of immune complex
82 SECTION 2 basic science

formation, evidence of classical pathway complement activation clear that primary infection or reactivation of these types of viruses,
was present with decreased serum C4 levels in the large majority particularly with VZV in the central nervous system and with HSV
of patients. or CMV in the retina, especially in immunosuppressed individuals,
Although HCV-associated vasculitis is predominantly an can occasionally induce vasculitis (Geng et al. 2011; Tranos et al.
immune complex disease related to cryoglobulins (Cacoub and 2008; Samant et al. 2007; Bertelmann et al. 2005; Sanjay et al. 2011;
Saadoun 2008; Saadoun et  al. 2011), PAN comprises about 20% Gutierrez and Ortiz 2011; Amlie-Lefond and Jubelt 2009; Lidar
of HCV-associated vasculitis and tends to be more acute and et al. 2009; Tresch et al. 2009; Patel et al. 2006; Pagnoux et al. 2006;
severe than HCV-negative PAN, but exhibits a higher rate of com- Cunningham et al. 1996; Sado et al. 1994; Gilden et al. 1996).
plete remission with antiviral and immunosuppressive therapy The human retrovirus HTLV-1 has been shown to infect human
than HCV-associated vasculitis due to cryoglobulins (Saadoun endothelial cells (Hoshino 2012), which may explain the CNS vas-
et  al. 2011). In contrast to HBV-associated PAN, however, culitis occasionally associated with the progressive spastic para-
HCV-associated PAN tends to occur long after the acute phase plegia seen with HTLV-1 infections (Vernant et al. 1994). Human
of infection (mean time of PAN diagnosis 282 months after HCV immunodeficiency virus (HIV) appears to directly cause vasculi-
infection vs. 7.2 months after infection for HBV-associated PAN). tis; however, other causes of vasculitis, including infections (HBV,
Evidence to support a pathogenic role for infectious organisms HCV and, as noted above in this Section, VZV, HSV, or CMV) and
in vasculitis includes case reports of concomitant vasculitis and medications, may be identified in persons with AIDS (Patel et al.
infection. Indirect support for a potential role for an infectious 2011; Gajera and Kais 2009; Melica et  al. 2009; Guillevin 2008;
agent in ANCA-associated vasculitis is the finding that a peptide Gisselbrecht et  al. 1998). Other infectious agents that have been
translated from the antisense DNA strand of PR3 and homologous implicated in vasculitis syndromes are discussed elsewhere (see
to several microbial peptides may be involved in induction of PR3 Chapter 41) and include Mycobacterium tuberculosis (Allins et al.
ANCA (Pendergraft et al. 2004). In equine viral arteritis, evidence 1999; Blanco Garcia et al. 1999); Bartonella henselae (Cozzani et al.
supports viral infection of the vascular endothelium (Estes and 2012); dengue fever (Ishikawa et  al. 1999); brucellosis (Nagore
Cheville 1970). A more convincing example of an arteritis with- et al. 1999; Perez et al. 1999); and Salmonella sp. (Soravia-Dunand
out immunological mediation has come from experimental cen- et al. 1999).
tral nervous system arteritis in turkeys infected with Mycoplasma Bacterial superantigens (SAg) are exotoxins produced by a
gallisepticum (Thomas et al. 1966). Inoculation of a large dose of small group of bacteria, primarily Staphylococcus aureus and
organisms resulted in arteritic lesions within 24 hours, implicating Streptococcus pyogenes, that produce oligoclonal activation of
a direct toxic effect of mycoplasma, or their products, on vascular T cells through an antigen-independent mechanism (Xu and
endothelium. Numerous exotic parasites and microbes also cause McCormick 2012). Evidence continues to accumulate support-
animal, and less often human, vasculitis. ing a pathogenic role for SAg in vasculitis, most notably in KD,
In humans, invasion by infectious organisms resulting in direct the most common cause of acquired heart disease in children in
vascular injury is seen with bacteria, mycobacteria, spirochetes, Western countries. SAg could theoretically act by activating auto-
and rickettsia. Tertiary syphilis, in particular, has long been associ- reactive T cells that participate in vessel damage, or B cells that
ated with infectious vasculitis, primarily manifesting as an aortitis produce autoantibodies such as ANCA or AECA. Multiple studies
and occurring in about 10% of patients with longstanding infec- have now demonstrated a skewing of T-cell Vbeta gene usage in
tion (Lindsay 1997). The disease manifests as asymptomatic aortitis patients with KD, suggesting that SAg could be involved in KD
(seen in up to 50% of patients with untreated syphilis of greater pathogenesis (Abe et al. 1993; Brogan et al. 2008; Curtis et al. 1995;
than 10 years’ duration), aortic regurgitation, coronary ostial ste- Reichardt et al. 2002). Similar skewing was noted in T cells isolated
nosis, and aortic aneurysm. Since the infection seeds haematog- from myocardium and coronaries of a patient dying of acute KD
enously primarily through the vasa vasorum, the ascending aorta (Leung et al. 1995) and from the small intestine of patients with KD
and the arch are predominantly affected because this portion of the (Yamashiro et al. 1996). In other studies, toxin-producing bacteria
vessel is enriched in these small arteries. The presence of the organ- were isolated in cultures from 13 of 16 KD patients but from only
ism can be demonstrated in such lesions by PCR (O’Regan et al. one of 15 controls (Leung et al. 1993), and SAg genes were found
2002). Acute bacterial aortitis may be due to a variety of pathogenic in increased prevalence in the stool of patients with KD compared
bacteria, most commonly with Salmonella sp. and Staphylococcus with normal controls (Suenaga et  al. 2009). Other studies have
aureus, and occurs most commonly in older individuals in asso- failed to identify the same pattern of skewing in the TCR Vbeta
ciation with atherosclerotic plaques and pre-existing aneurysms gene utilization (Nomura et al. 1998; Mancia et al. 1998) and, in
(Lindsay 1997). addition, serological evidence has provided only mixed support
Among the viruses, varicella zoster virus (VZV), herpes sim- for a significant contribution of superantigens in KD (Morita et al.
plex virus (HSV), and cytomegalovirus (CMV) appear to cause 1997; Matsubara et al. 2006).
human vasculitis (see Chapter  41). There is conflicting evidence Cases of vasculitis have been associated with a plethora of para-
on the presence of HSV DNA within tissues affected by vasculi- sitic diseases, including toxoplasmosis, trichinosis, strongyloidiasis,
tis. Although occasional studies have claimed to find an associa- ascariasis, sarcocystosis, amoebiasis, leishmaniasis, and toxocaria-
tion of well-characterized vasculitis syndromes with herpesviruses, sis. Unusual parasitic infections have been anecdotally reported in
including GCA (Powers et al. 2005) and KD (Shingadia et al. 2002; conjunction with vasculitis. For example, a 50-year-old Chilean
Toyabe et al. 2002), substantiation of such claims in other laborato- man had systemic vasculitis and was found to have Fasciola hepat-
ries has so far failed to materialize and contrary findings have been ica infection. He was treated with triclabendazole and all symptoms
published (Cankovic and Zarbo 2006; Alvarez-Lafuente et al. 2005; and systemic manifestations resolved within weeks (Llanos et al.
Rodriguez-Pla et al. 2004; Cooper et al. 2008). Nevertheless, it is 2006). In another report, an infection of a 39-year-old woman with
CHAPTER 7 pathogenesis of vasculitis 83

the free-living saprophagous nematode Halicephalobus deletrix, a After stimulation, T cells release cytokines chemotactic for mono-
cause of disseminated infections in horses, resulted in encephalo- cytes. Macrophages derived from infiltrating monocytes are then
myelitis that was diagnosed by biopsy as a granulomatous vasculitis capable of releasing lysosomal enzymes that damage endothelial
(Ondrejka et al. 2010). The patient succumbed to the infection fol- cells (Fauci et al. 1978). Experimental evidence for this comes from
lowing treatment with prednisone and cyclophosphamide. mice that were injected with syngeneic T cells sensitized in vitro to
cultured vascular smooth muscle cells (Hart et al. 1985). Twenty
Tumour Cell-Mediated Vascular Damage percent of the mice developed granulomatous vasculitis of the pul-
monary arterioles.
Malignancies associated with vasculitis are uncommon but not In human vasculitis, cell-mediated immunity has been studied
rare, and the practitioner would do well to consider malignancy primarily in GPA. Activated T cells, particularly the CD4+ and CD8+
with certain types of vasculitis, especially with cutaneous vascu- subtypes, have been found in biopsies from the upper and lower res-
litis arising in persons over 50 years of age (Fortin 1996; Buggiani piratory tracts and kidneys (Gephardt et al. 1983; Rasmussen et al.
et al. 2010). Some studies have estimated the incidence of concur- 1988; ten Berge et al. 1985). The expanded populations show high
rent malignancies to be as high as 2–5% of all vasculitides (Park expression of activation markers HLA-DR and CD25, independent
and Ranganathan 2011). The malignancies are most often haema- of activity of disease, and there are high levels of intracellular and
tological in origin and, as with infection-associated vasculitis, dif- secreted IFN-γ and IL-2 (Giscombe et al. 1998). Patients with active
ferent pathogenic processes are probably involved (see Chapter 41 GPA had higher percentages of CD38+ (activated) B cells than did
for detailed discussion). In addition, patients with myelodys- those in remission, or control subjects; however, activated T cells
plastic syndromes seem particularly prone to develop vasculitis persisted during remission of the vasculitis (Popa et  al. 1999).
(Oostvogels et al. 2012). Potential mechanisms include: (1) direct Earlier studies found significant elevations of soluble IL-2 recep-
effects of tumour cells on vascular walls (such as hairy cells in hairy tor (sIL-2R) levels in active generalized GPA (Schmitt et al. 1992).
cell leukaemia) (Klima and Waddell 1984; Gabriel et  al. 1986); PBMC from patients with GPA proliferate in vitro on exposure to
(2)  tumour release or tumour-induced release of cytokines that PR3, suggesting the presence of antigen-primed memory T cells
promote destruction of vascular structures; and (3) formation of (Kallenberg et al. 1991). A sustained response to monoclonal anti-
immune complexes of tumour associated antigen/ antibodies; and bodies directed against CD4+ T cells (Campath 1-H and anti-CD4)
(4) antibodies cross-reactive between tumour cells and endothe- has also been observed in four patients with life-threatening sys-
lial cells (Sanchez-Guerrero et al. 1990; Kurzrock and Cohen 1993; temic vasculitis that had been unresponsive to conventional immu-
Park and Ranganathan 2011). It is assumed that a tumour antigen nosuppressive agents (Lockwood et al. 1993).
activates cellular immunity, or participates in immune complex In addition to T cells, macrophages play an important role in
formation. Fain and colleagues analysed 60 patients with paraneo- vasculitis, and Weyand and colleagues have studied mechanisms of
plastic vasculitis (Fain et al. 2007). The mean age of the patients the contribution of these cells. They used differential display poly-
was 62.4 ± 12.9 years (SD) and the male : female ratio was 2.53. merase chain reaction to identify genes differentially expressed in
Of these, 63.1% had haematological malignancies or premalignant inflamed and unaffected temporal artery specimens (Rittner et al.
conditions with 32.3% having myelodysplastic syndrome and 29.2% 1999). Multinucleated giant cells and CD68+ macrophages charac-
lymphoid malignancies. The types of vasculitis included: cutaneous teristic of those that aggregate in the media–intima junction had
SVV (45%), PAN (36.7%), GPA (6.7%), MPA (5%), and IgAV (5%). increased expression of mitochondrial products and were able to
There is a generally low prevalence of vasculitis among the leu- synthesize metalloproteinase 2. Products of lipid peroxidation were
kaemias (about 2%). PAN is particularly associated with hairy found on nearby smooth muscle cells suggesting the action of reac-
cell leukaemia, having been first described in 1979 (Elkon et  al. tive oxygen species.
1979; Klima and Waddell 1984; Krol et al. 1983; Thorwarth et al. In summary, we have discussed in detail the pathogenic mecha-
1983; Le Pogamp et al. 1982; Goedert et al. 1981; Pope et al. 1980; nisms thought to be most relevant to the vasculitides, a complex set
Vankalakunti et  al. 2007). Hasler and colleagues reported 42 of disorders. The full roles and relationships between these mecha-
patients with vasculitis associated with hairy cell leukaemia (Hasler nisms are yet to be fully elucidated, but progress is being achieved
et al. 1995). Of these, 17 (40.5%) had PAN, 21 (50%) had cutane- as our understanding of the pathogenesis of vasculitis continues to
ous SVV, and four (9.5%) had vessel wall infiltration by hairy cells. evolve.
Immune complexes were found in three of four patients tested.
Three patients, of 12 tested, were HBsAg positive. Vasculitis most References
often occurred after splenectomy.
Abe, J., Kotzin, B.L., Meissner, C., et al. (1993). Characterization of T cell
repertoire changes in acute Kawasaki disease. Journal of Experimental
Cell-mediated immune responses and Medicine, 177, 791–6.
Acosta, J., Qin, X., and Halperin, J. (2004). Complement and complement
granuloma formation in vasculitis regulatory proteins as potential molecular targets for vascular diseases.
The occurrence of granulomas in GPA and EGPA is indicative of Current Pharmaceutical Design, 10, 203–11.
activation of lymphocytes and macrophages. It has been hypoth- Agnello, V., Chung, R.T., and Kaplan, L.M. (1992). A role for hepatitis C virus
infection in type II cryoglobulinemia. New England Journal of Medicine,
esized that CD4+ T cells are triggered by antigens in the circula-
327, 1490–5.
tion or within vascular walls. Despite the involvement of T cells in
Alegretti, A.P., Schneider, L., Piccoli, A.K., et al. (2012). Diminished expres-
vasculitis, there is no consistent evidence of association between sion of complement regulatory proteins on peripheral blood cells from
genes in the major histocompatibility complex and vasculitic syn- systemic lupus erythematosus patients. Clinical and Developmental
dromes, with the exception of Behçet’s syndrome (see Chapter 29). Immunology, 2012, 725684.
84 SECTION 2 basic science

Alfhaily, F., Watts, R., and Leather, A. (2009). Wegener’s granulomato- Bonaci-Nikolic, B., Nikolic, M.M., Andrejevic, S., Zoric, S., and Bukilica, M.
sis occurring de novo during pregnancy. Clinical and Experimental (2005). Antineutrophil cytoplasmic antibody (ANCA)-associated auto-
Rheumatology, 27, S86–8. immune diseases induced by antithyroid drugs: comparison with idio-
Allen, A.C., Harper, S.J., and Feehally, J. (1995). Galactosylation of N- and pathic ANCA vasculitides. Arthritis Research and Therapy, 7, R1072–81.
O-linked carbohydrate moieties of IgA1 and IgG in IgA nephropathy. Brandrup, F. and Ostergaard, P.A. (1978). alpha1-Antitrypsin deficiency
Clinical and Experimental Immunology, 100, 470–4. associated with persistent cutaneous vasculitis. Occurrence in a child
Allins, A.D., Wagner, W.H., Cossman, D.V., Gold, R.N., and Hiatt, J.R. with liver disease. Archives of Dermatology, 114, 921–4.
(1999). Tuberculous infection of the descending thoracic and abdomi- Brasile, L., Kremer, J.M., Clarke, J.L., and Cerilli, J. (1989). Identification of
nal aorta: case report and literature review. Annals of Vascular Surgery, an autoantibody to vascular endothelial cell-specific antigens in patients
13, 439–44. with systemic vasculitis. American Journal of Medicine, 87, 74–80.
Allison, S.J. (2012). Vasculitis:  Renal involvement in granulomatosis with Braverman, I.M. and Yen, A. (1975). Demonstration of immune com-
polyangiitis-role of ANCA isotype and Fc receptor genotype. Nature plexes in spontaneous and histamine-induced lesions and in normal
Reviews Nephrology, 8, 63. skin of patients with leukocytoclastic angitis. Journal of Investigative
Alvarez-Lafuente, R., Fernandez-Gutierrez, B., Jover, J.A., et  al. (2005). Dermatology, 64, 105–12.
Human parvovirus B19, varicella zoster virus, and human herpes virus Brogan, P.A., Shah, V., Clarke, L.A., Dillon, M.J., and Klein, N. (2008). T cell
6 in temporal artery biopsy specimens of patients with giant cell arte- activation profiles in Kawasaki syndrome. Clinical and Experimental
ritis:  analysis with quantitative real time polymerase chain reaction. Immunology, 151, 267–74.
Annals of the Rheumatic Diseases, 64, 780–2. Brooks, C.J., King, W.J., Radford, D.J., Adu, D., McGrath, M., and Savage, C.O.
Amlie-Lefond, C. and Jubelt, B. (2009). Neurologic manifestations of vari- (1996). IL-1 beta production by human polymorphonuclear leucocytes
cella zoster virus infections. Current Neurology and Neuroscience stimulated by anti-neutrophil cytoplasmic autoantibodies: relevance to
Reports, 9, 430–4. systemic vasculitis. Clinical and Experimental Immunology, 106, 273–9.
Arason, G.J., Jorgensen, G.H., and Ludviksson, B.R. (2010). Primary Buggiani, G., Krysenka, A., Grazzini, M., Vasku, V., Hercogova, J., and Lotti,
immunodeficiency and autoimmunity:  lessons from human diseases. T. (2010). Paraneoplastic vasculitis and paraneoplastic vascular syn-
Scandinavian Journal of Immunology, 71, 317–28. dromes. Dermatologic Therapy, 23, 597–605.
Arnaud, L., Haroche, J., Mathian, A., Gorochov, G., and Amoura, Z. (2011). Cacoub, P., Costedoat-Chalumeau, N., Lidove, O., and Alric, L. (2002).
Pathogenesis of Takayasu’s arteritis:  a 2011 update. Autoimmunity Cryoglobulinemia vasculitis. Current Opinion in Rheumatology,
Reviews, 11, 61–7. 14, 29–35.
Audrain, M.A., Sesboue, R., Baranger, T.A., et  al. (2001). Analysis of Cacoub, P. and Saadoun, D. (2008). Hepatitis C virus infection induced vas-
anti-neutrophil cytoplasmic antibodies (ANCA): frequency and specific- culitis. Clinical Reviews in Allergy and Immunology, 35, 30–9.
ity in a sample of 191 homozygous (PiZZ) alpha1-antitrypsin-deficient Cacoub, P. and Terrier, B. (2009). Hepatitis B-related autoimmune manifesta-
subjects. Nephrology Dialysis Transplantation, 16, 39–44. tions. Rheumatic Diseases Clinics of North America, 35, 125–37.
Bahrami, S., Malone, J.C., Webb, K.G., and Callen, J.P. (2006). Tissue eosino- Cankovic, M. and Zarbo, R.J. (2006). Failure to detect human herpes simplex
philia as an indicator of drug-induced cutaneous small-vessel vasculitis. virus, cytomegalovirus, and Epstein-Barr virus viral genomes in giant
Archives of Dermatology, 142, 155–61. cell arteritis biopsy specimens by real-time quantitative polymerase
Bansal, P.J. and Tobin, M.C. (2004). Neonatal microscopic polyangiitis sec- chain reaction. Cardiovascular Pathology, 15, 280–6.
ondary to transfer of maternal myeloperoxidase-antineutrophil cytoplas- Carter, P.M. (1973). Immune complex disease. Annals of the Rheumatic
mic antibody resulting in neonatal pulmonary hemorrhage and renal Diseases, 32, 265–71.
involvement. Annals of Allergy Asthma andImmunology, 93, 398–401. Carvalho, D., Savage, C.O., Isenberg, D., and Pearson, J.D. (1999). IgG
Begovich, A.B., Carlton, V.E., Honigberg, L.A., et  al. (2004). A missense anti-endothelial cell autoantibodies from patients with systemic lupus
single-nucleotide polymorphism in a gene encoding a protein tyros- erythematosus or systemic vasculitis stimulate the release of two
ine phosphatase (PTPN22) is associated with rheumatoid arthritis. endothelial cell-derived mediators, which enhance adhesion molecule
American Journal of Human Genetics, 75, 330–7. expression and leukocyte adhesion in an autocrine manner. Arthritis
Belizna, C., Duijvestijn, A., Hamidou, M., and Tervaert, J.W. (2006). and Rheumatism, 42, 631–40.
Antiendothelial cell antibodies in vasculitis and connective tissue dis- Cassatella, M.A., Huber, V., Calzetti, F., et al. (2006). Interferon-activated neu-
ease. Annals of the Rheumatic Diseases, 65, 1545–50. trophils store a TNF-related apoptosis-inducing ligand (TRAIL/Apo-2
Belizna, C.C., Hamidou, M.A., Levesque, H., Guillevin, L., and Shoenfeld, ligand) intracellular pool that is readily mobilizable following exposure
Y. (2009). Infection and vasculitis. Rheumatology (Oxford), 48, 475–82. to proinflammatory mediators. Journal of Leukocyte Biology, 79, 123–32.
Ben-Smith, A., Dove, S.K., Martin, A., Wakelam, M.J., and Savage, C.O. Chauhan, S.K., Tripathy, N.K., and Nityanand, S. (2006). Antigenic targets
(2001). Antineutrophil cytoplasm autoantibodies from patients with and pathogenicity of anti-aortic endothelial cell antibodies in Takayasu
systemic vasculitis activate neutrophils through distinct signaling cas- arteritis. Arthritis and Rheumatism, 54, 2326–33.
cades: comparison with conventional Fcgamma receptor ligation. Blood, Chiu, J.J. and Chien, S. (2011). Effects of disturbed flow on vascular endothe-
98, 1448–55. lium:  pathophysiological basis and clinical perspectives. Physiological
Bertelmann, E., Liekfeld, A., Pleyer, U., and Hartmann, C. (2005). Reviews, 91, 327–87.
Cytomegalovirus retinitis in Wegener’s granulomatosis: case report and Choi, H.K., Merkel, P.A., Walker, A.M., and Niles, J.L. (2000). Drug-associated
review of the literature. Acta Ophthalmoligica Scandinavica, 83, 258–61. antineutrophil cytoplasmic antibody-positive vasculitis:  prevalence
Bibby, S., Healy, B., Steele, R., Kumareswaran, K., Nelson, H., and Beasley, among patients with high titers of antimyeloperoxidase antibodies.
R. (2010). Association between leukotriene receptor antagonist therapy Arthritis and Rheumatism, 43, 405–13.
and Churg-Strauss syndrome: an analysis of the FDA AERS database. Christian, C.L. and Sergent, J.S. (1976). Vasculitis syndromes: clinical and
Thorax, 65, 132–8. experimental models. American Journal of Medicine, 61, 385–92.
Blanco Garcia, F.J., Sanchez Blas, M., and Freire Gonzalez, M. (1999). Cid, M.C., Segarra, M., Garcia-Martinez, A., and Hernandez-Rodriguez,
Histopathologic features of cerebral vasculitis associated with mycobac- J. (2004). Endothelial cells, antineutrophil cytoplasmic antibod-
terium tuberculosis. Arthritis and Rheumatism, 42, 383. ies, and cytokines in the pathogenesis of systemic vasculitis. Current
Blank, M., Krause, I., Goldkorn, T., et al. (1999). Monoclonal anti-endothelial Rheumatology Reports, 6, 184–94.
cell antibodies from a patient with Takayasu arteritis activate endothelial Claudy, A. (1998). Pathogenesis of leukocytoclastic vasculitis. European
cells from large vessels. Arthritis and Rheumatism, 42, 1421–32. Journal of Dermatology, 8, 75–9.
CHAPTER 7 pathogenesis of vasculitis 85

Cochrane, C.G. (1971). Mechanisms involved in the deposition of immune Domiciano, D.S., Shinjo, S.K., and Levy-Neto, M. (2010). Churg-strauss
complexes in tissues. Journal of Experimental Medicine, 134, 75s–89s. syndrome and active chronic hepatitis B virus infection: coincidence or
Cochrane, C.G. and Koffler, D. (1973). Immune complex disease in experi- association? Clinics (Sao Paulo), 65, 335–6.
mental animals and man. Advances in Immunology, 16, 185–264. Donato, L.J., Jenkins, S.M., Smith, C., Katzmann, J.A., and Snyder, M.R.
Cochrane, C.G., Weigle, W.O., and Dixon, F.J. (1959). The role of polymor- (2012). Reference and Interpretive Ranges for alpha1-Antitrypsin
phonuclear leukocytes in the initiation and cessation of the Arthus vas- Quantitation by Phenotype in Adult and Pediatric Populations.
culitis. Journal of Experimental Medicine, 110, 481–94. American Journal of Clinical Pathology, 138, 398–405.
Cockwell, P., Brooks, C.J., Adu, D., and Savage, C.O. (1999). Dowd, S.K., Rodgers, G.C., and Callen, J.P. (1995). Effective treatment with
Interleukin-8:  A  pathogenetic role in antineutrophil cytoplasmic alpha 1-protease inhibitor of chronic cutaneous vasculitis associated
autoantibody-associated glomerulonephritis. Kidney International, 55, with alpha 1-antitrypsin deficiency. Journal of the American Academy of
852–63. Dermatology, 33, 913–6.
Combes, B., Shorey, J., Barrera, A., et al. (1971). Glomerulonephritis with Duffy, T.N., Genta, M., Moll, S., Martin, P.Y., and Gabay, C. (2006). Henoch
deposition of Australia antigen-antibody complexes in glomerular base- Schonlein purpura following etanercept treatment of rheumatoid arthri-
ment membrane. Lancet, 2, 234–7. tis. Clinical and Experimental Rheumatology, 24, S106.
Conen, D., Leuppi, J., Bubendorf, L., Ronsdorf, A., Tamm, M., and Hauser, DuMouchel, W., Smith, E.T., Beasley, R., et al. (2004). Association of asthma
T. (2004). Montelukast and Churg-Strauss syndrome. Swiss Medical therapy and Churg-Strauss syndrome: an analysis of postmarketing sur-
Weekly, 134, 377–80. veillance data. Clinical Therapeutics, 26, 1092–104.
Cooper, R.J., D’Arcy, S., Kirby, M., et al. (2008). Infection and temporal arte- Eichhorn, J., Sima, D., Thiele, B., et al. (1996). Anti-endothelial cell antibod-
ritis: a PCR-based study to detect pathogens in temporal artery biopsy ies in Takayasu arteritis. Circulation, 94, 2396–401.
specimens. Journal of Medical Virology, 80, 501–5. Elkon, K.B., Hughes, G.R., Catovsky, D., et al. (1979). Hairy-cell leukaemia
Cooper, T., Savige, J., Nassis, L., et al. (2000). Clinical associations and charac- with polyarteritis nodosa. Lancet, 2, 280–2.
terisation of antineutrophil cytoplasmic antibodies directed against bac- Elsheikh, E., Sylven, C., and Henareh, L. (2010). Anti-endothelial cell anti-
tericidal/permeability-increasing protein and azurocidin. Rheumatology bodies are increased in patients with previous myocardial infarction.
International, 19, 129–36. Scandinavian Cardiovascular Journal, 44, 255–9.
Cornacoff, J.B., Hebert, L.A., Smead, W.L., VanAman, M.E., Birmingham, D.J., Erdbruegger, U., Grossheim, M., Hertel, B., et al. (2008). Diagnostic role of
and Waxman, F.J. (1983). Primate erythrocyte-immune complex-clearing endothelial microparticles in vasculitis. Rheumatology (Oxford), 47,
mechanism. Journal of Clinical Investigation, 71, 236–47. 1820–5.
Cozzani, E., Cinotti, E., Ameri, P., Sofia, A., Murialdo, G., and Parodi, A. Erdbrugger, U., Hellmark, T., Bunch, D.O., et al. (2006). Mapping of myelop-
(2012). Onset of cutaneous vasculitis and exacerbation of IgA nephrop- eroxidase epitopes recognized by MPO-ANCA using human-mouse
athy after Bartonella henselae infection. Clinical and Experimental MPO chimers. Kidney International, 69, 1799–805.
Dermatology, 37, 238–40. Ergin, S., Sanli Erdogan, B., Turgut, H., Evliyaoglu, D., and Yalcin, A.N.
Cream, J.J. (1971). Immune complex diseases. British Journal of Dermatology, (2005). Relapsing Henoch-Schonlein purpura in an adult patient associ-
85, 189–91. ated with hepatitis B virus infection. Journal of Dermatology, 32, 839–42.
Cunningham, E.T. Jr, Short, G.A., Irvine, A.R., Duker, J.S., and Margolis, T.P. Estes, P.C. and Cheville, N.F. (1970). The ultrastructure of vascular lesions in
(1996). Acquired immunodeficiency syndrome—associated herpes sim- equine viral arteritis. American Journal of Pathology, 58, 235–53.
plex virus retinitis. Clinical description and use of a polymerase chain Etgen, T., Winbeck, K., Conrad, B., and Sander, D. (2003). Hemiballism with
reaction—based assay as a diagnostic tool. Archives of Ophthalmology, insular infarction as first manifestation of Takayasu’s arteritis in associa-
114, 834–40. tion with chronic hepatitis B. Journal of Neurology, 250, 226–9.
Currie, J.R. (1907). On the Supersensitation of Persons suffering from Fain, O., Hamidou, M., Cacoub, P., et al. (2007). Vasculitides associated with
Diphtheria by Repeated Injections of Horse Serum. Journal of Hygiene malignancies: analysis of sixty patients. Arthritis and Rheumatism, 57,
(London), 7, 35–60. 1473–80.
Curtis, N., Zheng, R., Lamb, J.R., and Levin, M. (1995). Evidence for a supe- Falk, R.J., Terrell, R.S., Charles, L.A., and Jennette, J.C. (1990). Anti-neutrophil
rantigen mediated process in Kawasaki disease. Archives of Disease in cytoplasmic autoantibodies induce neutrophils to degranulate and pro-
Childhood, 72, 308–11. duce oxygen radicals in vitro. Proceedings of the National Academy of
Danila, M.I. and Bridges, S.L. Jr (2008). Update on pathogenic mechanisms Sciences U S A, 87, 4115–9.
of systemic necrotizing vasculitis. Current Rheumatology Reports, Fauci, A.S., Haynes, B., and Katz, P. (1978). The spectrum of vasculitis: clini-
10, 430–5. cal, pathologic, immunologic and therapeutic considerations. Annals of
Davies, D.J., Moran, J.E., Niall, J.F., and Ryan, G.B. (1982). Segmental necro- Internal Medicine, 89, 660–76.
tising glomerulonephritis with antineutrophil antibody: possible arbo- Fearon, D.T. (1984). Cellular receptors for the third component of comple-
virus aetiology? British Medical Journal (Clinical Research Ed.), 285, 606. ment. Immunology Today, 5, 105–10.
del Papa, N., Meroni, P.L., Barcellini, W., et al. (1992). Antibodies to endothe- Federman, D.G. and Kirsner, R.S. (1999). Leukocytoclastic vasculitis, hepati-
lial cells in primary vasculitides mediate in vitro endothelial cytotoxicity tis B, and the risk of endoscopy. Cutis, 63, 86–7.
in the presence of normal peripheral blood mononuclear cells. Clinical Feehally, J. and Allen, A.C. (1999). Structural features of IgA molecules which
Immunology and Immunopathology, 63, 267–74. contribute to IgA nephropathy. Journal of Nephrology, 12, 59–65.
Devakumar, V.N., Castelino, M., Chow, S.C., and Teh, L.S. (2010). Wegener’s Feriozzi, S., Muda, A.O., Gomes, V., Montanaro, M., Faraggiana, T., and
granulomatosis in pregnancy: a case report and review of the medical Ancarani, E. (2000). Cephotaxime-associated allergic interstitial nephri-
literature. BMJ Case Reports, 2010 pii, bcr09.2009.2296. tis and MPO-ANCA positive vasculitis. Renal Failure, 22, 245–51.
Dijstelbloem, H.M., Scheepers, R.H., Oost, W.W., et  al. (1999). Fcgamma Ferri, C. and Mascia, M.T. (2006). Cryoglobulinemic vasculitis. Current
receptor polymorphisms in Wegener’s granulomatosis: risk factors for Opinion in Rheumatology, 18, 54–63.
disease relapse. Arthritis and Rheumatism, 42, 1823–7. Fervenza, F.C., Smith, R.J., and Sethi, S. (2012). Association of a novel com-
Dixon, F.J., Vazquez, J.J., Weigle, W.O., and Cochrane, C.G. (1958). plement factor h mutation with severe crescentic and necrotizing glo-
Pathogenesis of serum sickness. AMA Archives of Pathology, 65, 18–28. merulonephritis. American Journal of Kidney Diseases, 60, 126–32.
Dolman, K.M., Gans, R.O., Vervaat, T.J., et al. (1993). Vasculitis and anti- Filer, A., Hughes, A., Kane, K., Mutimer, D., and Jobanputra, P. (2001).
neutrophil cytoplasmic autoantibodies associated with propylthiouracil Successful treatment of hepatitis B-associated vasculitis using lamivu-
therapy. Lancet, 342, 651–2. dine as the sole therapeutic agent. Rheumatology (Oxford), 40, 1064–5.
86 SECTION 2 basic science

Fischetti, F. and Tedesco, F. (2006). Cross-talk between the complement sys- Goodall, E.W. (1907). On the supersensitation of persons by horse-serum.
tem and endothelial cells in physiologic conditions and in vascular dis- Journal of Hygiene (London), 7, 607–18.
eases. Autoimmunity, 39, 417–28. Goodall, E.W. (1911). On some points connected with the serum treatment
Flint, S.M. and Savage, C.O. (2012). Anti-LAMP-2 autoantibodies in of diphtheria. British Medical Journal, 1, 292–6.
ANCA-associated pauci-immune glomerulonephritis. Journal of the Gower, R.G., Sams, W.M. Jr, Thorne, E.G., Kohler, P.F., and Claman, H.N.
American Society of Nephrology, 23, 378–80. (1977). Leukocytoclastic vasculitis: sequential appearance of immuno-
Flores-Suarez, L.F., Cabiedes, J., Villa, A.R., van der Woude, F.J., and reactants and cellular changes in serial biopsies. Journal of Investigative
Alcocer-Varela, J. (2003). Prevalence of antineutrophil cytoplasmic Dermatology, 69, 477–84.
autoantibodies in patients with tuberculosis. Rheumatology (Oxford), Gower, R.G., Sausker, W.F., Kohler, P.F., Thorne, G.E., and McIntosh, R.M.
42, 223–9. (1978). Small vessel vasculitis caused by hepatitis B virus immune com-
Fortin, P.R. (1996). Vasculitides associated with malignancy. Current Opinion plexes. Small vessel vasculitis and HBsAG. Journal of Allergy and Clinical
in Rheumatology, 8, 30–3. Immunology, 62, 222–8.
Gabriel, S.E., Conn, D.L., Phyliky, R.L., Pittelkow, M.R., and Scott, R.E. Graf, J., Lynch, K., Yeh, C.L., et al. (2011). Purpura, cutaneous necrosis, and anti-
(1986). Vasculitis in hairy cell leukemia: review of literature and con- neutrophil cytoplasmic antibodies associated with levamisole-adulterated
sideration of possible pathogenic mechanisms. Journal of Rheumatology, cocaine. Arthritis and Rheumatism, 63, 3998–4001.
13, 1167–72. Grammatikos, A.P. and Tsokos, G.C. (2012). Immunodeficiency and autoim-
Gajera, A. and Kais, S. (2009). HIV polyarteritis nodosa-like vasculitis pre- munity: lessons from systemic lupus erythematosus. Trends in Molecular
senting as chronic abdominal pain. Clinical Rheumatology, 28, 869–72. Medicine, 18, 101–8.
Gao, Y., Zhao, M.H., Guo, X.H., Xin, G., Gao, Y., and Wang, H.Y. (2004). The Gregersen, P.K., Lee, H.S., Batliwalla, F., and Begovich, A.B. (2006).
prevalence and target antigens of antithyroid drugs induced antineutro- PTPN22: setting thresholds for autoimmunity. Seminars in Immunology,
phil cytoplasmic antibodies (ANCA) in Chinese patients with hyperthy- 18, 214–23.
roidism. Endocrine Research, 30, 205–13. Griffith, M.E., Lovegrove, J.U., Gaskin, G., Whitehouse, D.B., and Pusey,
Garcia-Fuentes, M., Martin, A., Chantler, C., and Williams, D.G. (1978). C.D. (1996). C-antineutrophil cytoplasmic antibody positivity in vas-
Serum complement components in Henoch-Schonlein purpura. culitis patients is associated with the Z allele of alpha-1-antitrypsin,
Archives of Disease in Childhood, 53, 417–9. and P-antineutrophil cytoplasmic antibody positivity with the S allele.
Gasser, S., Kuhn, M., and Speich, R. (2004). Severe necrotising leucocytoclas- Nephrology Dialysis Transplantation, 11, 438–43.
tic vasculitis in a patient taking bosentan. BMJ, 329, 430. Groger, M., Fischer, G.F., Wolff, K., and Petzelbauer, P. (1999). Immune
Genel, F., Sjoholm, A.G., Skattum, L., and Truedsson, L. (2005). Complement complexes from vasculitis patients bind to endothelial Fc receptors
factor I deficiency associated with recurrent infections, vasculitis and independent of the allelic polymorphism of FcgammaRIIa. Journal of
immune complex glomerulonephritis. Scandinavian Journal of Infectious Investigative Dermatology, 113, 56–60.
Diseases, 37, 615–18. Grunwald, M.H., Avinoach, I., Amichai, B., and Halevy, S. (1997).
Geng, S., Ye, J.J., Zhao, J.L., Li, T.S., and Han, Y. (2011). Cytomegalovirus Leukocytoclastic vasculitis—correlation between different histologic
retinitis associated with acquired immunodeficiency syndrome. Chinese stages and direct immunofluorescence results. International Journal of
Medical Journal (English), 124, 1134–8. Dermatology, 36, 349–52.
Gephardt, G.N., Ahmad, M., and Tubbs, R.R. (1983). Pulmonary vasculitis Guerry, M.J., Brogan, P., Bruce, I.N., et al. (2012). Recommendations for the
(Wegener’s granulomatosis). Immunohistochemical study of T and B use of rituximab in anti-neutrophil cytoplasm antibody-associated vas-
cell markers. American Journal of Medicine, 74, 700–4. culitis. Rheumatology (Oxford), 51, 634–43.
Gerber, B.O. and Pichler, W.J. (2006). Noncovalent interactions of drugs with Guillevin, L. (2008). Vasculitides in the context of HIV infection. AIDS, 22
immune receptors may mediate drug-induced hypersensitivity reac- (Suppl. 3), S27–33.
tions. AAPS Journal, 8, E160–5. Guilpain, P. and Mouthon, L. (2008). Antiendothelial cells autoantibodies in
Ghebrehiwet, B., CebadaMora, C., Tantral, L., Jesty, J., and Peerschke, E.I. vasculitis-associated systemic diseases. Clinical Reviews in Allergy and
(2006). gC1qR/p33 serves as a molecular bridge between the comple- Immunology, 35, 59–65.
ment and contact activation systems and is an important catalyst in Gutierrez, J. and Ortiz, G. (2011). HIV/AIDS patients with HIV vasculopathy
inflammation. Advances in Experimental Medicine and Biology, 586, and VZV vasculitis: a case series. Clinical Neuroradiology, 21, 145–51.
95–105. Hall, J.B., Wadham, B.M., Wood, C.J., Ashton, V., and Adam, W.R. (1984).
Gilden, D.H., Kleinschmidt-DeMasters, B.K., Wellish, M., Hedley-Whyte, Vasculitis and glomerulonephritis: a subgroup with an antineutrophil
E.T., Rentier, B., and Mahalingam, R. (1996). Varicella zoster virus, a cytoplasmic antibody. Australian and New Zealand Journal of Medicine,
cause of waxing and waning vasculitis:  the New England Journal of 14, 277–8.
Medicine case 5-1995 revisited. Neurology, 47, 1441–6. Haraoui, B. and Keystone, E. (2006). Musculoskeletal manifestations and
Giscombe, R., Nityanand, S., Lewin, N., Grunewald, J., and Lefvert, A.K. autoimmune diseases related to new biologic agents. Current Opinion in
(1998). Expanded T cell populations in patients with Wegener’s granu- Rheumatology, 18, 96–100.
lomatosis: characteristics and correlates with disease activity. Journal of Harper, L., Cockwell, P., Adu, D., and Savage, C.O. (2001). Neutrophil priming
Clinical Immunology, 18, 404–13. and apoptosis in anti-neutrophil cytoplasmic autoantibody-associated
Gisselbrecht, M., Cohen, P., Lortholary, O., et al. (1998). Human immunode- vasculitis. Kidney International, 59, 1729–38.
ficiency virus-related vasculitis. Clinical presentation of and therapeu- Hart, M.N., Tassell, S.K., Sadewasser, K.L., Schelper, R.L., and Moore, S.A. (1985).
tic approach to eight cases. Annales de Médecine Interne (Paris), 149, Autoimmune vasculitis resulting from in vitro immunization of lympho-
398–405. cytes to smooth muscle. American Journal of Pathology, 119, 448–55.
Gocke, D.J., Hsu, K., Morgan, C., Bombardieri, S., Lockshin, M., and Hasler, P., Kistler, H., and Gerber, H. (1995). Vasculitides in hairy cell leuke-
Christian, C.L. (1970). Association between polyarteritis and Australia mia. Seminars in Arthritis and Rheumatism, 25, 134–42.
antigen. Lancet, 2, 1149–53. Haynes, B.F. (1992). Vasculitis:  pathogenic mechanisms of vessel damage.
Goedert, J.J., Neefe, J.R., Smith, F.S., Stahl, N.I., Jaffe, E.S., and Fauci, A.S. In Inflammation:  basic principles and clinical correlates, 2nd edn, pp.
(1981). Polyarteritis nodosa, hairy cell leukemia and splenosis. American 921–41 (ed. J.I. Gallin and I.M.S.R. Goldstein). Raven Press, New York.
Journal of Medicine, 71, 323–6. Heeringa, P., Huugen, D., and Tervaert, J.W. (2005). Anti-neutrophil cyto-
Goodall, E.W. (1899). On the value of the treatment of diphtheria by anti- plasmic autoantibodies and leukocyte-endothelial interactions: a sticky
toxin. British Medical Journal, 1, 268–70. connection? Trends in Immunology, 26, 561–4.
CHAPTER 7 pathogenesis of vasculitis 87

Hellmich, B., Csernok, E., and Gross, W.L. (2005). Proinflammatory Kammer, G.M., Soter, N.A., and Schur, P.H. (1980). Circulating immune
cytokines and autoimmunity in Churg-Strauss syndrome. Annals of the complexes in patients with necrotizing vasculitis. Clinical Immunology
New York Academy of Sciences, 1051, 121–31. and Immunopathology, 15, 658–72.
Henegar, C., Pagnoux, C., Puechal, X., et al. (2008). A paradigm of diagnostic Karsten, C.M., Pandey, M.K., Figge, J., et  al. (2012). Anti-inflammatory
criteria for polyarteritis nodosa: analysis of a series of 949 patients with activity of IgG1 mediated by Fc galactosylation and association of
vasculitides. Arthritis and Rheumatism, 58, 1528–38. FcgammaRIIB and dectin-1. Nature Medicine,18, 1401–6.
Hisano, S., Matsushita, M., Fujita, T., and Iwasaki, H. (2005). Activation of Kasahara, Y., Iwai, K., Yachie, A., et al. (1997). Involvement of reactive oxygen
the lectin complement pathway in Henoch-Schonlein purpura nephri- intermediates in spontaneous and CD95 (Fas/APO-1)-mediated apop-
tis. American Journal of Kidney Diseases, 45, 295–302. tosis of neutrophils. Blood, 89, 1748–53.
Hoffman, G.S. (1997). Vasculitic syndromes. Current Opinion in Kawana, S. and Nishiyama, S. (1992). Serum SC5b-9 (terminal complement
Rheumatology, 9, 1–2. complex) level, a sensitive indicator of disease activity in patients with
Holmen, C., Christensson, M., Pettersson, E., et al. (2004). Wegener’s granu- Henoch-Schonlein purpura. Dermatology, 184, 171–6.
lomatosis is associated with organ-specific antiendothelial cell antibod- Kawasaki, Y. (2011). The pathogenesis and treatment of pediatric
ies. Kidney International, 66, 1049–60. Henoch-Schonlein purpura nephritis. Clinical and Experimental
Hong, Y., Eleftheriou, D., Hussain, A.A., et  al. (2012). Anti-neutrophil Nephrology, 15, 648–57.
cytoplasmic antibodies stimulate release of neutrophil microparticles. Kelley, J.M., Edberg, J.C., and Kimberly, R.P. (2010). Wegener’s granuloma-
Journal of the American Society of Nephrology, 23, 49–62. tosis:  a model of auto-antibodies in mucosal autoimmunity. Clinical
Hoshino, H. (2012). Cellular factors involved in HTLV-1 entry and patho- Immunology, 134, 104–12.
genicity. Frontiers in Microbiology, 3, 222. Kelley, J.M., Monach, P.A., Ji, C., et al. (2011). IgA and IgG antineutrophil
Hurlburt, K.J., McMahon, B.J., Simonetti, J.P., et  al. (2007). Hepatitis cytoplasmic antibody engagement of Fc receptor genetic variants influ-
B-associated vasculitis in Alaska Natives:  viral genotype, clinical and ences granulomatosis with polyangiitis. Proceedings of the National
serologic outcome. Liver International, 27, 627–32. Academy of Sciences U S A, 108, 20736–41.
Ishikawa, H., Okada, S., Katayama, I., et al. (1999). A Japanese case of dengue Keogh, K.A. and Specks, U. (2003). Churg-Strauss syndrome: clinical presen-
fever with lymphocytic vasculitis: diagnosis by polymerase chain reac- tation, antineutrophil cytoplasmic antibodies, and leukotriene receptor
tion. Journal of Dermatology, 26, 29–32. antagonists. American Journal of Medicine, 115, 284–90.
Jacobs-Kosmin, D., Derk, C.T., and Sandorfi, N. (2006). Pantoprazole and Kerns, W., Schwartz, L., Blanchard, K., et al. (2005). Drug-induced vascular
perinuclear antineutrophil cytoplasmic antibody-associated vasculitis. injury—a quest for biomarkers. Toxicology and Applied Pharmacology,
Journal of Rheumatology, 33, 629–32. 203, 62–87.
Jacobson, M.D. (1996). Reactive oxygen species and programmed cell death. Kessenbrock, K., Krumbholz, M., Schonermarck, U., et al. (2009). Netting neu-
Trends in Biochemical Sciences, 21, 83–6. trophils in autoimmune small-vessel vasculitis. Nature Medicine, 15, 623–5.
Jagiello, P., Aries, P., Arning, L., et al. (2005). The PTPN22 620W allele is a risk fac- Kettritz, R. (2012). How anti-neutrophil cytoplasmic autoantibodies activate
tor for Wegener’s granulomatosis. Arthritis and Rheumatism, 52, 4039–43. neutrophils. Clinical and Experimental Immunology, 169, 220–8.
Jennette, J.C. and Falk, R.J. (1998). Pathogenesis of the vascular and glo- Kettritz, R., Wilke, S., von Vietinghoff, S., Luft, F., and Schneider, W. (2006).
merular damage in ANCA-positive vasculitis. Nephrology Dialysis Apoptosis, proliferation and inflammatory infiltration in ANCA-positive
Transplantation, 13 (Suppl. 1), 16–20. glomerulonephritis. Clinical Nephrology, 65, 309–16.
Jennette, J.C., Falk, R.J., Bacon, P.A., et  al. (2013). 2012 Revised interna- Kiryluk, K., Moldoveanu, Z., Sanders, J.T., et  al. (2011). Aberrant glyco-
tional Chapel Hill Consensus Conference nomenclature of vasculitides. sylation of IgA1 is inherited in both pediatric IgA nephropathy and
Arthritis and Rheumatism, 65, 1–11. Henoch-Schonlein purpura nephritis. Kidney International, 80, 79–87.
Jennette, J.C., Falk, R.J., and Gasim, A.H. (2011). Pathogenesis of antineutro- Klima, M. and Waddell, C.C. (1984). Hairy cell leukemia associated with
phil cytoplasmic autoantibody vasculitis. Current Opinion in Nephrology focal vascular damage. Human Pathology, 15, 657–9.
and Hypertension, 20, 263–70. Kobayashi, S., Ishizuka, S., Tamura, N., Takaya, M., Kaneda, K., and
Jennette, J.C., Xiao, H., and Falk, R.J. (2006). Pathogenesis of vascular Hashimoto, H. (2003). Churg-Strauss syndrome (CSS) in a patient
inflammation by anti-neutrophil cytoplasmic antibodies. Journal of the receiving pranlukast. Clinical Rheumatology, 22, 491–2.
American Society of Nephrology, 17, 1235–42. Korkmaz, B., Jaillet, J., Jourdan, M.L., Gauthier, A., Gauthier, F., and Attucci,
Kain, R., Exner, M., Brandes, R., et  al. (2008). Molecular mimicry in S. (2009). Catalytic activity and inhibition of wegener antigen proteinase
pauci-immune focal necrotizing glomerulonephritis. Nature Medicine, 3 on the cell surface of human polymorphonuclear neutrophils. Journal
14, 1088–96. of Biological Chemistry, 284, 19896–902.
Kain, R., Matsui, K., Exner, M., et al. (1995). A novel class of autoantigens Krol, T., Robinson, J., Bekeris, L., and Messmore, H. (1983). Hairy cell leuke-
of anti-neutrophil cytoplasmic antibodies in necrotizing and crescentic mia and a fatal periarteritis nodosa-like syndrome. Archives of Pathology
glomerulonephritis: the lysosomal membrane glycoprotein h-lamp-2 in and Laboratory Medicine, 107, 583–5.
neutrophil granulocytes and a related membrane protein in glomerular Kurzrock, R. and Cohen, P.R. (1993). Vasculitis and cancer. Clinical
endothelial cells. Journal of Experimental Medicine, 181, 585–97. Dermatology, 11, 175–87.
Kain, R., Tadema, H., McKinney, E.F., et  al. (2012). High Prevalence Lai, K.N. (2012). Pathogenesis of IgA nephropathy. Nature Reviews
of Autoantibodies to hLAMP-2 in Anti-Neutrophil Cytoplasmic Nephrology, 8, 275–83.
Antibody-Associated Vasculitis. Journal of the American Society of Lau, K.K., Suzuki, H., Novak, J., and Wyatt, R.J. (2010). Pathogenesis of
Nephrology, 23, 556–66. Henoch-Schonlein purpura nephritis. Pediatric Nephrology, 25, 19–26.
Kallenberg, C.G. (2011). Pathogenesis of ANCA-associated vasculitides. Lee, Y.J., Kang, S.W., Park, J.J., et al. (2006). Interleukin-18 promoter poly-
Annals of the Rheumatic Diseases, 70 (Suppl. 1), i59–63. morphisms in patients with Behcet’s disease. Human Immunology,
Kallenberg, C.G., Stegeman, C.A., and Tervaert, J.W. (1996). Staphylococcus 67, 812–8.
aureus, trimethoprim-sulfamethoxazole, and Wegener’s granulomatosis. Le Pogamp, P., Ghandour, C., and Le Prise, P.Y. (1982). Hairy cell leukemia
Sarcoidosis Vasculitis Diffuse Lung Diseases, 13, 253–5. and polyarteritis nodosa. Journal of Rheumatology, 9, 441–2.
Kallenberg, C.G., Tervaert, J.W., van der Woude, F.J., Goldschmeding, R., von Leung, D.Y., Collins, T., Lapierre, L.A., Geha, R.S., and Pober, J.S. (1986a).
dem Borne, A.E., and Weening, J.J. (1991). Autoimmunity to lysosomal Immunoglobulin M antibodies present in the acute phase of Kawasaki
enzymes: new clues to vasculitis and glomerulonephritis? Immunology syndrome lyse cultured vascular endothelial cells stimulated by gamma
Today, 12, 61–4. interferon. Journal of Clinical Investigation, 77, 1428–35.
88 SECTION 2 basic science

Leung, D.Y., Geha, R.S., Newburger, J.W., et al. (1986b). Two monokines, Streptococcus pyogenes in Kawasaki disease. Clinical and Experimental
interleukin 1 and tumor necrosis factor, render cultured vascular Immunology, 143, 427–34.
endothelial cells susceptible to lysis by antibodies circulating dur- Mazodier, P., Elzouki, A.N., Segelmark, M., and Eriksson, S. (1996). Systemic
ing Kawasaki syndrome. Journal of Experimental Medicine, 164, necrotizing vasculitides in severe alpha1-antitrypsin deficiency. QJM,
1958–72. 89, 599–611.
Leung, D.Y., Giorno, R.C., Kazemi, L.V., Flynn, P.A., and Busse, J.B. (1995). McCluskey, R.T. and Fienberg, R. (1983). Vasculitis in primary vasculitides,
Evidence for superantigen involvement in cardiovascular injury due to granulomatoses, and connective tissue diseases. Human Pathology, 14,
Kawasaki syndrome. Journal of Immunology, 155, 5018–21. 305–15.
Leung, D.Y., Meissner, H.C., Fulton, D.R., Murray, D.L., Kotzin, B.L., McCormick, R.V. (1950). Periarteritis occurring during propylthiouracil
and Schlievert, P.M. (1993). Toxic shock syndrome toxin-secreting therapy. Journal of the American Medical Association, 144, 1453–4.
Staphylococcus aureus in Kawasaki syndrome. Lancet, 342, 1385–8. McGrath, M.M., Isakova, T., Rennke, H.G., Mottola, A.M., Laliberte, K.A.,
Levo, Y., Gorevic, P.D., Kassab, H.J., Zucker-Franklin, D., and Franklin, E.C. and Niles, J.L. (2011). Contaminated cocaine and antineutrophil cyto-
(1977). Association between hepatitis B virus and essential mixed cryo- plasmic antibody-associated disease. Clinical Journal of the American
globulinemia. New England Journal of Medicine, 296, 1501–4. Society of Nephrology, 6, 2799–805.
Lewis, M.J. and Botto, M. (2006). Complement deficiencies in humans and Medof, M.E., Iida, K., Mold, C., and Nussenzweig, V. (1982). Unique role of
animals: links to autoimmunity. Autoimmunity, 39, 367–78. the complement receptor CR1 in the degradation of C3b associated with
Lidar, M., Lipschitz, N., Langevitz, P., and Shoenfeld, Y. (2009). The infectious immune complexes. Journal of Experimental Medicine, 156, 1739–54.
etiology of vasculitis. Autoimmunity, 42, 432–8. Melica, G., Brugieres, P., Lascaux, A.S., Levy, Y., and Lelievre, J.D. (2009).
Lima, F., Buchanan, N., Froes, L., Kerslake, S., Khamashta, M.A., and Hughes, Primary vasculitis of the central nervous system in patients infected
G.R. (1995). Pregnancy in granulomatous vasculitis. Annals of the with HIV-1 in the HAART era. Journal of Medical Virology, 81, 578–81.
Rheumatic Diseases, 54, 604–6. Merkel, P.A. (2001). Drug-induced vasculitis. Rheumatic Diseases Clinics of
Lin, Q., Min, Y., Li, Y., et al. (2012). Henoch-Schonlein purpura with hypoc- North America, 27, 849–62.
omplementemia. Pediatric Nephrology, 27, 801–6. Michalak, T. (1978). Immune complexes of hepatitis B surface antigen in the
Lindsay, J. Jr (1997). Diagnosis and treatment of diseases of the aorta. Current pathogenesis of periarteritis nodosa. A study of seven necropsy cases.
Problems in Cardiology, 22, 485–542. American Journal of Pathology, 90, 619–32.
Lipsker, D. and Hauptmann, G. (2010). Cutaneous manifestations of comple- Mohan, N., Edwards, E.T., Cupps, T.R., et al. (2004). Leukocytoclastic vascu-
ment deficiencies. Lupus, 19, 1096–106. litis associated with tumor necrosis factor-alpha blocking agents. Journal
Little, M.A., Al-Ani, B., Ren, S., et al. (2012). Anti-proteinase 3 anti-neutrophil of Rheumatology, 31, 1955–8.
cytoplasm autoantibodies recapitulate systemic vasculitis in mice with a Moneret-Vautrin, D.A., Faure, G., and Bene, M.C. (1986). Chewing-gum pre-
humanized immune system. PLoS One, 7, e28626. servative induced toxidermic vasculitis. Allergy, 41, 546–8.
Little, M.A., Smyth, C.L., Yadav, R., et  al. (2005). Antineutrophil cyto- Morita, A., Imada, Y., Igarashi, H., and Yutsudo, T. (1997). Serologic evidence
plasm antibodies directed against myeloperoxidase augment that streptococcal superantigens are not involved in the pathogenesis of
leukocyte-microvascular interactions in vivo. Blood, 106, 2050–8. Kawasaki disease. Microbiology and Immunology, 41, 895–900.
Llanos, C., Soto, L., Sabugo, F., et al. (2006). Systemic vasculitis associated Morris, H., Morgan, M.D., Wood, A.M., et al. (2011). ANCA-associated vas-
with Fasciola hepatica infection. Scandinavian Journal of Rheumatology, culitis is linked to carriage of the Z allele of alpha antitrypsin and its
35, 143–6. polymers. Annals of the Rheumatic Diseases, 70, 1851–6.
Lockwood, C.M., Thiru, S., Isaacs, J.D., Hale, G., and Waldmann, H. (1993). Nagore, E., Sanchez-Motilla, J.M., Navarro, V., Febrer, M.I., and Aliaga, A.
Long-term remission of intractable systemic vasculitis with monoclonal (1999). Leukocytoclastic vasculitis as a cutaneous manifestation of sys-
antibody therapy. Lancet, 341, 1620–2. temic infection caused by Brucella melitensis. Cutis, 63, 25–7.
Lowry, M.D., Hudson, C.F., and Callen, J.P. (1994). Leukocytoclastic vas- Neynaber, S., Mistry-Burchardi, N., Rust, C., et al. (2008). PR3-ANCA-positive
culitis caused by drug additives. Journal of the American Academy of necrotizing multi-organ vasculitis following cocaine abuse. Acta
Dermatology, 30, 854–5. Dermato-Venereologica, 88, 594–6.
Lu, X., Garfield, A., Rainger, G.E., Savage, C.O., and Nash, G.B. (2006). Nishimura, M., Ishikawa, Y., and Satake, M. (2004). Activation of polymor-
Mediation of endothelial cell damage by serine proteases, but not super- phonuclear neutrophils by immune complex:  possible involvement
oxide, released from antineutrophil cytoplasmic antibody-stimulated in development of transfusion-related acute lung injury. Transfusion
neutrophils. Arthritis and Rheumatism, 54, 1619–28. Medicine, 14, 359–67.
Lyons, P.A., Rayner, T.F., Trivedi, S., et al. (2012). Genetically distinct subsets Noh, J.Y., Asari, T., Hamada, N., et al. (2001). Frequency of appearance of
within ANCA-Associated Vasculitis. New England Journal of Medicine, myeloperoxidase-antineutrophil cytoplasmic antibody (MPO-ANCA)
367, 214–23. in Graves’ disease patients treated with propylthiouracil and the rela-
Macdonald, J., Zhong, T., Lazarescu, A., Gan, B.S., and Harth, M. tionship between MPO-ANCA and clinical manifestations. Clinical
(2004). Vasculitis associated with the use of leflunomide. Journal of Endocrinology (Oxford), 54, 651–4.
Rheumatology, 31, 2076–8. Nomura, Y., Masuda, K., Shinkoda, Y., et  al. (1998). Twenty-five types of
Maggiore, G., Martini, A., Grifeo, S., De Giacomo, C., and Scotta, M.S. (1984). T-cell receptor Vbeta family repertoire in patients with Kawasaki syn-
Hepatitis B virus infection and Schonlein-Henoch purpura. American drome. European Journal of Pediatrics, 157, 981–6.
Journal of Diseases of Childhood, 138, 681–2. O’Regan, A.W., Castro, C., Lukehart, S.A., Kasznica, J.M., Rice, P.A., and
Mahr, A.D., Edberg, J.C., Stone, J.H., et  al. (2010). Alpha-antitrypsin Joyce-Brady, M.F. (2002). Barking up the wrong tree? Use of polymerase
deficiency-related alleles Z and S and the risk of Wegener’s granuloma- chain reaction to diagnose syphilitic aortitis. Thorax, 57, 917–8.
tosis. Arthritis and Rheumatism, 62, 3760–7. O’Reilly, P.J., Hardison, M.T., Jackson, P.L., et al. (2009). Neutrophils contain
Mancia, L., Wahlstrom, J., Schiller, B., et al. (1998). Characterization of the prolyl endopeptidase and generate the chemotactic peptide, PGP, from
T-cell receptor V-beta repertoire in Kawasaki disease. Scandinavian collagen. Journal of Neuroimmunology, 217, 51–4.
Journal of Immunology, 48, 443–9. Oberndorfer, S., Beate, U., Sabine, U., et al. (2004). Churg Strauss syndrome
Margolis, M.T., and Newton, T.H. (1971). Methamphetamine (‘speed’) arteri- during treatment of bronchial asthma with a leucotriene receptor antag-
tis. Neuroradiology, 2, 179–82. onist presenting with polyneuropathy. Neurologia, 19, 134–8.
Matsubara, K., Fukaya, T., Miwa, K., et al. (2006). Development of serum Ohta, Y., Ohya, Y., Fujii, K., et al. (2003). Inflammatory diseases associated
IgM antibodies against superantigens of Staphylococcus aureus and with Takayasu’s arteritis. Angiology, 54, 339–44.
CHAPTER 7 pathogenesis of vasculitis 89

Oka, N., Kawasaki, T., Matsui, M., Shigematsu, K., Unuma, T., and Sugiyama, Pettigrew, H.D., Teuber, S.S., and Gershwin, M.E. (2009). Clinical signifi-
H. (2011). Two subtypes of Churg-Strauss syndrome with neuropa- cance of complement deficiencies. Annals of the New York Academy of
thy:  the roles of eosinophils and ANCA. Modern Rheumatology, Sciences, 1173, 108–23.
21, 290–5. Popa, E.R., Stegeman, C.A., Bos, N.A., Kallenberg, C.G., and Tervaert, J.W.
Ondrejka, S.L., Procop, G.W., Lai, K.K., and Prayson, R.A. (2010). Fatal (1999). Differential B- and T-cell activation in Wegener’s granulomato-
parasitic meningoencephalomyelitis caused by Halicephalobus delet- sis. Journal of Allergy and Clinical Immunology, 103, 885–94.
rix: a case report and review of the literature. Archives of Pathology and Pope, A., Lazarchick, J., Hoyer, L., and Weinstein, A. (1980). Hairy cell leuke-
Laboratory Medicine, 134, 625–9. mia and vasculitis. Journal of Rheumatology, 7, 895–9.
Oostvogels, R., Petersen, E.J., Chauffaille, M.L., and Abrahams, A.C. (2012). Powers, J.F., Bedri, S., Hussein, S., Salomon, R.N., and Tischler, A.S. (2005).
Systemic vasculitis in myelodysplastic syndromes. Netherlands Journal High prevalence of herpes simplex virus DNA in temporal arteritis
of Medicine, 70, 63–8. biopsy specimens. American Journal of Clinical Pathology, 123, 261–4.
Orpin, S.D., Majmudar, V.B., Soon, C., Azam, N.A., and Salim, A. (2006). Pradhan, V.D., Badakere, S.S., Ghosh, K., and Pawar, A.R. (2004). Spectrum
Adalimumab causing vasculitis. British Journal of Dermatology, of anti-neutrophil cytoplasmic antibodies in patients with pulmonary
154, 998–9. tuberculosis overlaps with that of Wegener’s granulomatosis. Indian
Oztas, M.O., Onder, M., Gurer, M.A., Bukan, N., and Sancak, B. (2005). Serum Journal Medicine Science, 58, 283–8.
interleukin 18 and tumour necrosis factor-alpha levels are increased in Radford, D.J., Lord, J.M., and Savage, C.O. (1999). The activation of the neu-
Behcet’s disease. Clinical and Experimental Dermatology, 30, 61–3. trophil respiratory burst by anti-neutrophil cytoplasm autoantibody
Pagnoux, C., Cohen, P., and Guillevin, L. (2006). Vasculitides secondary to (ANCA) from patients with systemic vasculitis requires tyrosine kinases
infections. Clinical and Experimental Rheumatology, 24, S71–81. and protein kinase C activation. Clinical and Experimental Immunology,
Pagnoux, C. and Guillevin, L. (2010). Churg-Strauss syndrome: evidence for 118, 171–9.
disease subtypes? Current Opinion in Rheumatology, 22, 21–8. Radford, D.J., Luu, N.T., Hewins, P., Nash, G.B., and Savage, C.O. (2001).
Pagnoux, C., Guilpain, P., and Guillevin, L. (2007). Churg-Strauss syndrome. Antineutrophil cytoplasmic antibodies stabilize adhesion and promote
Current Opinion in Rheumatology, 19, 25–32. migration of flowing neutrophils on endothelial cells. Arthritis and
Pagnoux, C., Seror, R., Henegar, C., et al. (2010). Clinical features and out- Rheumatism, 44, 2851–61.
comes in 348 patients with polyarteritis nodosa: a systematic retrospec- Ralston, D.R., Marsh, C.B., Lowe, M.P., and Wewers, M.D. (1997).
tive study of patients diagnosed between 1963 and 2005 and entered into Antineutrophil cytoplasmic antibodies induce monocyte IL-8 release.
the French Vasculitis Study Group Database. Arthritis and Rheumatism, Role of surface proteinase-3, alpha1-antitrypsin, and Fcgamma recep-
62, 616–26. tors. Journal of Clinical Investigation, 100, 1416–24.
Papadopoulos, D.P., Makris, T.K., Krespi, P., et al. (2006). Antiendothelial cell Rasmussen, N., Petersen, J., Ralfkiaer, E., Avnstom, S., and Wiik, A.
antibody levels in healthy normotensives with high normal blood pres- (1988). Spontaneous and induced immunoglobulin synthesis and
sure. Clinical and Experimental Hypertension, 28, 663–7. anti-neutrophil cytoplasm antibodies in Wegener’s granulomato-
Papadopoulos, D.P., Makris, T.K., Papazachou, U., Daskalaki, M., Sanidas, sis:  relation to leukocyte subpopulations in blood and active lesions.
E., and Votteas, V.E. (2008). Antiendothelial cell antibody levels in Rheumatology International, 8, 153–8.
patients with masked hypertension. International Journal of Cardiology, Rauterberg, E.W., Lieberknecht, H.M., Wingen, A.M., and Ritz, E.
130, 405–8. (1987). Complement membrane attack (MAC) in idiopathic
Park, H.J. and Ranganathan, P. (2011). Neoplastic and paraneoplastic vascu- IgA-glomerulonephritis. Kidney International, 31, 820–9.
litis, vasculopathy, and hypercoagulability. Rheumatic Diseases Clinics of Reichardt, P., Lehmann, I., Sierig, G., and Borte, M. (2002). Analysis of T-cell
North America, 37, 593–606. receptor V-beta 2 in peripheral blood lymphocytes as a diagnostic
Park, M.C., Park, Y.B., Jung, S.Y., Lee, K.H., and Lee, S.K. (2006). marker for Kawasaki disease. Infection, 30, 360–4.
Anti-endothelial cell antibodies and antiphospholipid antibodies in Rittner, H.L., Kaiser, M., Brack, A., Szweda, L.I., Goronzy, J.J., and Weyand,
Takayasu’s arteritis: correlations of their titers and isotype distributions C.M. (1999). Tissue-destructive macrophages in giant cell arteritis.
with disease activity. Clinical and Experimental Rheumatology, 24, S10–6. Circulation Research, 84, 1050–8.
Patel, A.K., Patel, K.K., Shah, S.D., and Desai, J. (2006). Immune reconsti- Rodriguez-Pla, A., Bosch-Gil, J.A., Echevarria-Mayo, J.E., et al. (2004). No
tution syndrome presenting with cerebral varicella zoster vasculitis in detection of parvovirus B19 or herpesvirus DNA in giant cell arteritis.
HIV-1-infected patient: a case report. Journal International Association Journal Clinical Virology, 31, 11–5.
of Physicians in AIDS Care (Chicago), 5, 157–60. Rongioletti, F., Ghio, L., Ginevri, F., et  al. (1999). Purpura of the ears:  a
Patel, N., Khan, T., and Espinoza, L.R. (2011). HIV infection and clinical spec- distinctive vasculopathy with circulating autoantibodies complicat-
trum of associated vasculitides. Current Rheumatology Reports, 13, 506–12. ing long-term treatment with levamisole in children. British Journal of
Pearson, T., Bremmer, M., Cohen, J., and Driscoll, M. (2012). Vasculopathy Dermatology, 140, 948–51.
related to cocaine adulterated with levamisole: A review of the literature. Roth, A.J., Brown, M.C., Smith, R.N., et al. (2012). Anti-LAMP-2 antibodies
Dermatology Online Journal, 18, 1. are not prevalent in patients with antineutrophil cytoplasmic autoanti-
Pelletier, F., Puzenat, E., Blanc, D., Faivre, B., Humbert, P., and Aubin, F. body glomerulonephritis. Journal of the American Society of Nephrology,
(2003). Minocycline-induced cutaneous polyarteritis nodosa with anti- 23, 545–55.
neutrophil cytoplasmic antibodies. European Journal of Dermatology, Saadoun, D., Terrier, B., Semoun, O., et al. (2011). Hepatitis C virus-associated
13, 396–8. polyarteritis nodosa. Arthritis Care and Research (Hoboken), 63, 427–35.
Pendergraft, W.F. 3rd, Pressler, B.M., Jennette, J.C., Falk, R.J., and Preston, Sado, K., Kimura, T., Hotta, Y., et al. (1994). Acute retinal necrosis syndrome
G.A. (2005). Autoantigen complementarity: a new theory implicating associated with herpes simplex keratitis. Retina, 14, 260–3.
complementary proteins as initiators of autoimmune disease. Journal Salojin, K.V., Le Tonqueze, M., Nassovov, E.L., et al. (1996). Anti-endothelial
Molecular Medicine (Berlin), 83, 12–25. cell antibodies in patients with various forms of vasculitis. Clinical and
Pendergraft, W.F. 3rd, Preston, G.A., Shah, R.R., et al. (2004). Autoimmunity Experimental Rheumatology, 14, 163–9.
is triggered by cPR-3(105-201), a protein complementary to human Samant, J.S., Namgoong, S.H., Parveen, T., and Katner, H.P. (2007).
autoantigen proteinase-3. Nature Medicine, 10, 72–9. Cytomegalovirus vasculitis and mucormycosis coinfection in late-stage
Perez, C., Hernandez, R., Murie, M., Vives, R., and Guarch, R. (1999). HIV/AIDS. American Journal of Medical Science, 333, 122–4.
Relapsing leucocytoclastic vasculitis as the initial manifestation of acute Sanchez-Guerrero, J., Gutierrez-Urena, S., Vidaller, A., Reyes, E., Iglesias, A.,
brucellosis. British Journal of Dermatology, 140, 1177–8. and Alarcon-Segovia, D. (1990). Vasculitis as a paraneoplastic syndrome.
90 SECTION 2 basic science

Report of 11 cases and review of the literature. Journal of Rheumatology, Srivastava, M.D., Alexander, F., and Tuthill, R.J. (2005). Immunology of
17, 1458–62. cutaneous vasculitis associated with both etanercept and infliximab.
Sangaletti, S., Tripodo, C., Chiodoni, C., et al. (2012). Neutrophil extracellu- Scandinavian Journal of Immunology, 61, 329–36.
lar traps mediate transfer of cytoplasmic neutrophil antigens to myeloid Stegeman, C.A., Tervaert, J.W., de Jong, P.E., and Kallenberg, C.G. (1996).
dendritic cells towards ANCA induction and associated autoimmunity. Trimethoprim-sulfamethoxazole (co-trimoxazole) for the prevention of
Blood, 120, 3007–18. relapses of Wegener’s granulomatosis. Dutch Co-Trimoxazole Wegener
Sanjay, S., Huang, P., and Lavanya, R. (2011). Herpes zoster ophthalmicus. Study Group. New England Journal of Medicine, 335, 16–20.
Current Treatment Options in Neurology, 13, 79–91. Stegeman, C.A., Tervaert, J.W., Sluiter, W.J., Manson, W.L., de Jong, P.E.,
Sato, H., Hattori, M., Fujieda, M., et al. (2000). High prevalence of antineu- and Kallenberg, C.G. (1994). Association of chronic nasal carriage of
trophil cytoplasmic antibody positivity in childhood onset Graves’ dis- Staphylococcus aureus and higher relapse rates in Wegener granuloma-
ease treated with propylthiouracil. Journal of Clinical Endocrinology and tosis. Annals of Internal Medicine, 120, 12–7.
Metabolism, 85, 4270–3. Suenaga, T., Suzuki, H., Shibuta, S., Takeuchi, T., and Yoshikawa, N. (2009).
Saulsbury, F.T. (1999). Henoch-Schonlein purpura in children. Report of 100 Detection of multiple superantigen genes in stools of patients with
patients and review of the literature. Medicine (Baltimore), 78, 395–409. Kawasaki disease. Journal Pediatrics, 155, 266–70.
Savage, C.O. (2011). Pathogenesis of anti-neutrophil cytoplasmic autoan- Sunderkotter, C., Seeliger, S., Schonlau, F., et al. (2001). Different pathways
tibody (ANCA)-associated vasculitis. Clinical and Experimental leading to cutaneous leukocytoclastic vasculitis in mice. Experimental
Immunology, 164 (Suppl. 1), 23–6. Dermatology, 10, 391–404.
Savage, C.O., Pottinger, B.E., Gaskin, G., Lockwood, C.M., Pusey, C.D., and Surmali Onay, O., Baskin, E., Ozcay, F., Melek, E., Canan, O., and Bilezikci, B.
Pearson, J.D. (1991). Vascular damage in Wegener’s granulomatosis and (2007). Successful treatment of hepatitis B-associated leukocytoclastic
microscopic polyarteritis:  presence of anti-endothelial cell antibodies vasculitis with lamivudine treatment in a child patient. Rheumatology
and their relation to anti-neutrophil cytoplasm antibodies. Clinical and International, 27, 869–72.
Experimental Immunology, 85, 14–9. Szczeklik, W., Jakiela, B., Adamek, D., and Musial, J. (2013). Cutting edge
Schifferli, J.A., Ng, Y.C., and Peters, D.K. (1986). The role of complement issues in the Churg-Strauss syndrome. Clinical Reviews in Allergy and
and its receptor in the elimination of immune complexes. New England Immunology,44, 39–50.
Journal of Medicine, 315, 488–95. Tadema, H., Kallenberg, C.G., Stegeman, C.A., and Heeringa, P. (2011).
Schlieben, D.J., Korbet, S.M., Kimura, R.E., Schwartz, M.M., and Lewis, E.J. Reactivity against complementary proteinase-3 is not increased in
(2005). Pulmonary-renal syndrome in a newborn with placental trans- patients with PR3-ANCA-associated vasculitis. PLoS One, 6, e17972.
mission of ANCAs. American Journal of Kidney Diseases, 45, 758–61. ten Berge, I.J., Wilmink, J.M., Meyer, C.J., et al. (1985). Clinical and immu-
Schmitt, W.H., Heesen, C., Csernok, E., Rautmann, A., and Gross, W.L. nological follow-up of patients with severe renal disease in Wegener’s
(1992). Elevated serum levels of soluble interleukin-2 receptor in granulomatosis. American Journal of Nephrology, 5, 21–9.
patients with Wegener’s granulomatosis. Association with disease activ- ten Holder, S.M., Joy, M.S., and Falk, R.J. (2002). Cutaneous and systemic
ity. Arthritis and Rheumatism, 35, 1088–96. manifestations of drug-induced vasculitis. Annals of Pharmacotherapy,
Schreiber, A., Xiao, H., Falk, R.J., and Jennette, J.C. (2006). Bone 36, 130–47.
marrow-derived cells are sufficient and necessary targets to mediate glo- Theofilopoulos, A.N. and Dixon, F.J. (1980). Immune complexes in human
merulonephritis and vasculitis induced by anti-myeloperoxidase anti- diseases: a review. American Journal of Pathology, 100, 529–94.
bodies. Journal of the American Society of Nephrology, 17, 3355–64. Thomas, L., Davidson, M., and McCluskey, R.T. (1966). Studies of PPLO
Schreiber, A., Xiao, H., Jennette, J.C., Schneider, W., Luft, F.C., and Kettritz, R. infection. I.  The production of cerebral polyarteritis by Mycoplasma
(2009). C5a receptor mediates neutrophil activation and ANCA-induced gallisepticum in turkeys; the neurotoxic property of the Mycoplasma.
glomerulonephritis. Journal of the American Society of Nephrology, 20, Journal of Experimental Medicine, 123, 897–912.
289–98. Thorwarth, W.T. Jr, Jaques, P.F., and Orringer, E.P. (1983). Polyarteritis
Segelmark, M., Elzouki, A.N., Wieslander, J., and Eriksson, S. (1995). nodosa in hairy cell leukemia. Journal of the Canadian Association of
The PiZ gene of alpha 1-antitrypsin as a determinant of outcome in Radiologists, 34, 151–2.
PR3-ANCA-positive vasculitis. Kidney International, 48, 844–50. Tobon, G.J., Alard, J.E., Youinou, P., and Jamin, C. (2009). Are autoantibodies
Sera, N., Ashizawa, K., Ando, T., et al. (2000). Treatment with propylthioura- triggering endothelial cell apoptosis really pathogenic? Autoimmunity
cil is associated with appearance of antineutrophil cytoplasmic antibod- Reviews, 8, 605–10.
ies in some patients with Graves’ disease. Thyroid, 10, 595–9. Toyabe, S., Harada, W., Suzuki, H., Hirokawa, T., and Uchiyama, M. (2002).
Servettaz, A., Guilpain, P., Camoin, L., et al. (2008a). Identification of target Large vessel arteritis associated with human herpesvirus 6 infections.
antigens of antiendothelial cell antibodies in healthy individuals: A pro- Clinical Rheumatology, 21, 528–32.
teomic approach. Proteomics, 8, 1000–8. Tranos, P.G., Georgalas, I., Founti, P., and Ladas, I. (2008). Cytomegalovirus
Servettaz, A., Guilpain, P., Tamas, N., Kaveri, S.V., Camoin, L., and Mouthon, retinitis presenting as vasculitis in a patient with Wegener’s granuloma-
L. (2008b). Natural anti-endothelial cell antibodies. Autoimmunity tosis. Clinical Ophthalmology, 2, 961–3.
Reviews, 7, 426–30. Tresch, S., Trueb, R.M., Kamarachev, J., French, L.E., and Hofbauer, G.F.
Sheikh, S., Rainger, G.E., Gale, Z., Rahman, M., and Nash, G.B. (2003). Exposure (2009). Disseminated herpes zoster mimicking rheumatoid vascu-
to fluid shear stress modulates the ability of endothelial cells to recruit neu- litis in a rheumatoid arthritis patient on etanercept. Dermatology,
trophils in response to tumor necrosis factor-alpha: a basis for local varia- 219, 347–9.
tions in vascular sensitivity to inflammation. Blood, 102, 2828–34. Tripathy, N.K., Sinha, N., and Nityanand, S. (2003). Anti-annexin V antibod-
Shingadia, D., Bose, A., and Booy, R. (2002). Could a herpesvirus be the ies in Takayasu’s arteritis: prevalence and relationship with disease activ-
cause of Kawasaki disease? Lancet Infectious Diseases, 2, 310–3. ity. Clinical and Experimental Immunology, 134, 360–4.
Sindrilaru, A., Seeliger, S., Ehrchen, J.M., et  al. (2007). Site of blood ves- Tse, W.Y., Abadeh, S., Jefferis, R., Savage, C.O., and Adu, D. (2000).
sel damage and relevance of CD18 in a murine model of immune Neutrophil FcgammaRIIIb allelic polymorphism in anti-neutrophil
complex-mediated vasculitis. Journal of Investigative Dermatology, 127, cytoplasmic antibody (ANCA)-positive systemic vasculitis. Clinical and
447–54. Experimental Immunology, 119, 574–7.
Soravia-Dunand, V.A., Loo, V.G., and Salit, I.E. (1999). Aortitis due to Tse, W.Y., Abadeh, S., McTiernan, A., Jefferis, R., Savage, C.O., and Adu, D.
Salmonella: report of 10 cases and comprehensive review of the litera- (1999). No association between neutrophil FcgammaRIIa allelic poly-
ture. Clinical Infectious Diseases, 29, 862–8. morphism and anti-neutrophil cytoplasmic antibody (ANCA)-positive
CHAPTER 7 pathogenesis of vasculitis 91

systemic vasculitis. Clinical and Experimental Immunology, 117, Weiss, S.R., Raskind, R., Morganstern, N.L., Pytlyk, P.J., and Baiz, T.C. (1970).
198–205. Intracerebral and subarachnoid hemorrhage following use of metham-
Tsukada, N., Koh, C.S., Owa, M., and Yanagisawa, N. (1983). Chronic neu- phetamine (‘speed’). International Surgery, 53, 123–7.
ropathy associated with immune complexes of hepatitis B virus. Journal Wiesner, O., Litwiller, R.D., Hummel, A.M., et  al. (2005). Differences
of the Neurological Sciences, 61, 193–210. between human proteinase 3 and neutrophil elastase and their murine
van der Woude, F.J. (1985). Anticytoplasmic antibodies in Wegener’s granu- homologues are relevant for murine model experiments. FEBS Letters,
lomatosis. Lancet, 2, 48. 579, 5305–12.
van der Woude, F.J., Rasmussen, N., Lobatto, S., et al. (1985). Autoantibodies Wikman, A., Lundahl, J., and Jacobson, S.H. (2005). Neutrophil activation in
against neutrophils and monocytes: tool for diagnosis and marker of dis- anti-proteinase 3-positive vasculitis—a prospective study. Scandinavian
ease activity in Wegener’s granulomatosis. Lancet, 1, 425–9. Journal of Immunology, 62, 539–45.
van Paassen, P., Tervaert, J.W., and Heeringa, P. (2007). Mechanisms of vascu- Wilde, B., van Paassen, P., Witzke, O., and Tervaert, J.W. (2011). New patho-
litis: how pauci-immune is ANCA-associated renal vasculitis? Nephron physiological insights and treatment of ANCA-associated vasculitis.
Experimental Nephrology, 105, e10–6. Kidney International, 79, 599–612.
van Rossum, A.P., Huitema, M.G., Stegeman, C.A., et al. (2007). Standardised Wilson, C.B. and Dixon, F.J. (1971). Quantitation of acute and chronic serum
assessment of membrane proteinase 3 expression. Analysis in sickness in the rabbit. Journal of Experimental Medicine, 134, 7–18.
ANCA-associated vasculitis and controls. Annals of the Rheumatic Witko-Sarsat, V., Cramer, E.M., Hieblot, C., et al. (1999). Presence of pro-
Diseases, 66, 1350–5. teinase 3 in secretory vesicles: evidence of a novel, highly mobilizable
van Rossum, A.P., Limburg, P.C., and Kallenberg, C.G. (2003). Membrane intracellular pool distinct from azurophil granules. Blood, 94, 2487–96.
proteinase 3 expression on resting neutrophils as a pathogenic fac- Xiao, H., Heeringa, P., Hu, P., et  al. (2002). Antineutrophil cytoplasmic
tor in PR3-ANCA-associated vasculitis. Clinical and Experimental autoantibodies specific for myeloperoxidase cause glomerulonephritis
Rheumatology, 21, S64–8. and vasculitis in mice. Journal of Clinical Investigation, 110, 955–63.
Van Rossum, A.P., van der Geld, Y.M., Limburg, P.C., and Kallenberg, C.G. Xiao, H., Schreiber, A., Heeringa, P., Falk, R.J., and Jennette, J.C. (2007).
(2005). Human anti-neutrophil cytoplasm autoantibodies to proteinase Alternative complement pathway in the pathogenesis of disease medi-
3 (PR3-ANCA) bind to neutrophils. Kidney International, 68, 537–41. ated by anti-neutrophil cytoplasmic autoantibodies. American Journal
Vanek, C. and Samuels, M.H. (2005). Central nervous system vasculitis of Pathology, 170, 52–64.
caused by propylthiouracil therapy: a case report and literature review. Xing, G.Q., Chen, M., Liu, G., et al. (2009). Complement activation is involved
Thyroid, 15, 80–4. in renal damage in human antineutrophil cytoplasmic autoantibody
Vankalakunti, M., Joshi, K., Jain, S., Nada, R., Radotra, B.D., and Varma, S. associated pauci-immune vasculitis. Journal of Clinical Immunology, 29,
(2007). Polyarteritis nodosa in hairy cell leukaemia: an autopsy report. 282–91.
Journal of Clinical Pathology, 60, 1181–2. Xu, S.X. and McCormick, J.K. (2012). Staphylococcal superantigens in coloni-
Varagunam, M., Nwosu, A.C., Adu, D., et  al. (1993). Little evidence for zation and disease. Frontiers in Cellular and Infection Microbiology, 2, 52.
anti-endothelial-cell antibodies in microscopic polyarteritis and Yamashiro, Y., Nagata, S., Oguchi, S., and Shimizu, T. (1996). Selective
Wegener’s granulomatosis. Advances in Experimental Medicine and increase of V beta 2+ T cells in the small intestinal mucosa in Kawasaki
Biology, 336, 419–22. disease. Pediatric Research, 39, 264–6.
Vernant, J.C., Smadja, D., Deforge-Lasseur, C., et al. (1994). [Vasculitis and Yang, J.J., Kettritz, R., Falk, R.J., Jennette, J.C., and Gaido, M.L. (1996).
neurologic manifestations related to HTLV-1]. Presse Médicale, 23, Apoptosis of endothelial cells induced by the neutrophil serine proteases
1421–5. proteinase 3 and elastase. American Journal of Pathology, 149, 1617–26.
Vischer, U.M. (2006). von Willebrand factor, endothelial dysfunction, and Yu, F., Zhao, M.H., Zhang, Y.K., Zhang, Y., and Wang, H.Y. (2005).
cardiovascular disease. Journal of Thrombosis and Haemostasis, 4, Anti-endothelial cell antibodies (AECA) in patients with propylthioura-
1186–93. cil (PTU)-induced ANCA positive vasculitis are associated with disease
Wada, N., Mukai, M., Kohno, M., Notoya, A., Ito, T., and Yoshioka, N. (2002). activity. Clinical and Experimental Immunology, 139, 569–74.
Prevalence of serum anti-myeloperoxidase antineutrophil cytoplasmic Yuan, J., Gou, S.J., Huang, J., Hao, J., Chen, M., and Zhao, M.H. (2012).
antibodies (MPO-ANCA) in patients with Graves’ disease treated with C5a and its receptors in human anti-neutrophil cytoplasmic antibody
propylthiouracil and thiamazole. Endocrinology Journal, 49, 329–34. (ANCA)-associated vasculitis. Arthritis Research and Therapy, 14, R140.
Wang, H., Ma, J., Wu, Q., Luo, X., Chen, Z., and Kou, L. (2011). Circulating Zholudev, A., Zurakowski, D., Young, W., Leichtner, A., and Bousvaros, A.
B lymphocytes producing autoantibodies to endothelial cells play a role (2004). Serologic testing with ANCA, ASCA, and anti-OmpC in chil-
in the pathogenesis of Takayasu arteritis. Journal of Vascular Surgery, 53, dren and young adults with Crohn’s disease and ulcerative colitis: diag-
174–80. nostic value and correlation with disease phenotype. American Journal
Wechsler, M.E., Pauwels, R., and Drazen, J.M. (1999). Leukotriene modifiers of Gastroenterology, 99, 2235–41.
and Churg-Strauss syndrome: adverse effect or response to corticoster- Zipfel, P.F. and Skerka, C. (2009). Complement regulators and inhibitory pro-
oid withdrawal? Drug Safety, 21, 241–51. teins. Nature Reviews Immunology, 9, 729–40.
CHAPTER 8

Animal models of vasculitis


Peter Heeringa

Introduction 1985). These mice spontaneously develop autoimmune manifesta-


tions that resemble human SLE, including enlargement of lymphoid
Vasculitis is defined as inflammation of blood vessels associated organs, increased IgG levels, circulating antinuclear autoanti-
with necrosis of the vessel wall. Vasculitis can affect any type of ves- bodies, immune complex glomerulonephritis, and necrotizing
sel in any organ and may lead to occlusion of the vascular lumen, vasculitis (Figure  8.1). The necrotizing vasculitis seen in MRL/
causing organ failure. In various inflammatory disorders vasculitis MpJ-Faslpr mice can affect small and medium-sized arteries, arte-
is an important histopathological feature, of which the cause can rioles, veins, and venules and is characterized by fibrinoid necrosis
be either primary or secondary. Secondary vasculitis occurs as a typically accompanied by infiltration of neutrophils, with or with-
manifestation of another underlying disease, such as drug hyper- out nuclear fragmentation (leukocytoclasis). Necrotizing vasculi-
sensitivity or response to infection. For the majority of primary tis in MRL/MpJ-Faslpr mice has been reported to occur in many
vasculitides, the aetiopathogenesis is largely unknown but in gen- organs, including salivary glands, lung, liver, kidney, and skin but
eral these disorders are considered to be complex, multifactorial the aetiopathogenesis of vasculitis development in MRL/MpJ-Faslpr
diseases involving genetic susceptibility, excessive or aberrant is not well understood (Berden et al. 1983; Alexander et al. 1985;
immune responses, and environmental factors. To study the com- Moyer et al. 1987). The presence of autoantibodies of various spe-
plex interplay between these factors and intrinsic tissue cells in the cificities has been detected in these mice, including antibodies to
development of vasculitis, animal models are required. The main nuclear antigens (double-stranded DNA, single-stranded DNA,
goal for developing such animal models is to elucidate the immu- and histones) anti-neutrophil cytoplasmic autoantibodies (ANCA)
nological, genetic, and environmental mechanisms that may con- and anti-endothelial cell antibodies (AECA) (Theofilopoulos and
tribute to vasculitis development. For that reason, it is essential that Dixon 1985; Harper et al. 1998; Dimitriu-Bona et al. 1995). This
the clinical and pathological manifestations in such models mimic suggests that autoantibody-mediated mechanisms, including
those of the human disease closely. immune complex deposition, contribute to vasculitis development
In this chapter, an overview of generally accepted and frequently and organ injury. However, MRL/MpJ-Faslpr mice that lack circu-
used animal models of vasculitis, including both spontaneous lating antibodies still develop glomerulonephritis and necrotizing
models as well as induced models, is provided with an emphasis on vasculitis indicating that (auto)antibody-independent mechanisms
models developed for primary vasculitides. are important as well (Chan et al. 1999).
Because of the spontaneous occurrence of vasculitis, MRL/
Spontaneous models of vasculitis MpJ-Faslpr mice have proven to be useful for identifying factors
that determine susceptibility for vasculitis development. To this
Vasculitis in inbred mouse strains of systemic end, MRL/MpJ-Faslpr have been crossed with various other inbred
autoimmunity strains of mice. One of these strains was established by Kinjoh
There are many reports of spontaneously occurring vasculitis in and colleagues, who aimed to develop a mouse strain that spon-
various inbred mouse strains. The majority of these strains develop taneously develops a high incidence of necrotizing crescentic glo-
a form of systemic autoimmune disease mimicking human systemic merulonephritis (Kinjoh et al. 1993). This strain of mice, termed
lupus erythematosus (SLE) and have been employed to study the spontaneous crescentic glomerulonephritis/Kinjoh (SCG/Kj)
cellular and genetic factors that contribute to disease development mice, was generated by crossing BXSB and MRL/MpJ-Faslpr mice
in SLE (Andrews et  al. 1978; Pathak and Mohan 2011). SLE is a followed by selective breeding of F1 hybrid mice whose parents
systemic autoimmune disease characterized by autoantibody for- had the highest frequency of glomerular crescent formation. In
mation to nuclear antigens, in particular to double-stranded DNA, the SCG/Kj strain, the incidence of crescentic glomerulonephritis
immune-complex-mediated glomerulonephritis and vasculitis in is as high as 58% in females and 34% in males. In addition, SCG/
various organs. Spontaneous inbred models of systemic vasculitis Kj mice develop necrotizing vasculitis of small and medium-sized
resembling SLE include the MRL/MpJ-Faslpr, NZB/NZW F1, and the arteries and arterioles in many organs, including the spleen, ovary,
NZW/BXSB F1 strains of mice, and these will be briefly discussed. heart, and stomach. The SCG/Kj strain has been proposed as a
spontaneous model for ANCA-associated vasculitis (AAV) because
Vasculitis in MRL/MpJ-Faslpr mice the development of crescentic glomerulonephritis and vasculitis
The best characterized and frequently used animal model of SLE associates with the presence of high-titre circulating autoantibod-
is the MRL/MpJ-Faslpr mouse strain (Theofilopoulos and Dixon ies that react with myeloperoxidase, a major autoantigen in AAV.
94 SECTION 2 basic science

(a) (b)

Fig. 8.1 Necrotizing glomerulonephritis (a) and vasculitis (b) in the kidney of a MRL/MpJ-Faslpr mouse. (PAS stain, ×200)

However, circulating immune complexes and antibodies directed NZB/NZW F1 and NZW/BXSB F1 models emphasize the impor-
against single-stranded DNA and double-stranded DNA are tance of genetic backgrounds in the development of vasculitis but
detected in SCG/Kj mice as well and substantial immune complex due to the low frequency of vasculitic manifestations these mice are
deposits are observed in glomerular lesions (Neumann et al. 2003). not commonly used as models of vasculitis.
These features are clearly different from human AAV, in which vas-
cular lesions are characterized by a paucity of immune deposits, Vasculitis in interleukin-1 receptor
indicating that SCG/Kj mice are not a representative model for antagonist deficient mice
human AAV. An interesting vasculitic phenotype has been reported in mice
Genetic studies in the MRL/MpJ-Faslpr mouse strain have deficient in the IL-1 receptor antagonist (IL-1ra). IL-1ra is a struc-
revealed that the background genetic makeup of the MRL strain is tural homologue of IL-1α and β but displays higher affinity for the
essential for development of vasculitis and glomerulonephritis. In IL-1 receptor and therefore inhibits the proinflammatory effects
general, such genetic studies are performed by crossing the MRL/ of IL-1α and β (Dinarello 2011). In genetically engineered mice
MpJ-Faslpr strain with a non-diseased strain followed by chromo- that lack the IL-1ra gene (l11m) development of a lethal form of
somal mapping of the progeny for polymorphic markers that are vasculitis in the aorta and branches thereof has been reported.
linked to the disease phenotype. From these studies, at least six Pathologically, the lesions were characterized by accumulation
chromosomal loci have been reported that confer susceptibility for of monocytes/ macrophages and IFN-γ-producing CD4+ T cells
vasculitis (Wang et al. 1997; Nose et al. 1998; Yamada et al. 2003; Qu resembling Takayasu’s disease and giant cell arteritis (Shepherd
et al. 2000; Hamano et al. 2006; Zhang et al. 2006). The contention and Nicklin 2005). Interestingly, IL-1ra-deficient mice back-
derived from these studies is that development of systemic vascu- crossed onto different background strains display different phe-
litis in the MRL/MpJ-Faslpr mouse strain is not a monogenic event notypes (Horai et  al. 2000; Shepherd et  al. 2004; Nicklin et  al.
but is the result of the combined effect of multiple genes. Another 2000; Matsuki et al. 2005). On a BALB/c mice background a high
interesting observation from these genetic studies is that vasculitis incidence of vasculitis is observed, whereas in IL-1ra knockout
and glomerulonephritis appear to depend on different background C57Bl6 mice no evidence for arteritis was found suggesting the
genes. McH5/lpr is a congenic strain of mice derived from crosses involvement of additional genes in arteritis development (Matsuki
between MRL/MpJ-Faslpr mice and autoimmune-resistant C3H/lpr et al. 2005).
mice (Nose et al. 1996). These mice develop severe granulomatous
arteritis, in particular in the kidney, but no glomerulonephritis,
indicating a genetic dissociation between vasculitis and glomeru-
Induced models of vasculitis
lonephritis development (Nose et al. 1996). Animal models of immune complex vasculitis
For many years, the formation of antigen–antibody complexes at
Vasculitis in NZB/NZW F1 and the site of the vessel wall has been considered the primary patho-
NZW/BXSB F1 Mice logical mechanism in all forms of vasculitis. This concept appeared
NZB/NZW F1 or B/W mice, generated by crossing the New to be confirmed by observations in a number of vasculitic condi-
Zealand Black (NZB) and New Zealand White (NZW) mouse tions and animal models showing immune complex deposits in
strains, are a well-established model of SLE and these mice con- vascular lesions. Based on these observations, a multistep model
sistently develop glomerulonephritis (Theofilopoulos and Dixon for vasculitis development was proposed. In this model, antigen
1985). The frequency of vasculitis reported in these mice is low, exposure and subsequent antibody formation leads to circulating
although it increases with age (Burnet and Holmes 1965; Hicks antigen–antibody complexes which, under specific circumstances,
1966; Staszak and Harbeck 1985). Similarly, BXSB mice develop a deposit in blood vessel walls causing a cascade of inflammatory
severe lupus-like syndrome with glomerulonephritis but no overt reactions, culminating in vessel wall damage and necrosis (Fauci
vasculitis (Dixon et  al. 1978; Berden et  al. 1983). When crossed et al. 1978). Although operative in various forms of vasculitis, par-
with NZW mice or MRL/MpJ-Faslpr the frequency and severity of ticularly secondary vasculitis, the concept is not generally applica-
vasculitis increases, especially upon ageing. The observed vasculitic ble because many of the primary forms of vasculitis lack substantial
manifestations are diverse, including arteritis in various organs. The immune complex deposits in the lesions.
CHAPTER 8 animal models of vasculitis 95

In animal models, the role of antigen–antibody complex forma- which is characterized by an immune response against structural
tion in vasculitis development has been studied extensively. In the components of the glomerular basement membrane (Lerner et al.
acute serum sickness model in rabbits, a single intravenous dose of 1967; Lahmer and Heemann 2012). This immune response results
heterologous serum albumin induces necrotizing arteritis, glomer- in autoantibodies that react with the non-collagenous domain of
ulonephritis, and arthritis that develop after 10 to 14 days (Dixon the α3 chain of collagen type IV and deposit in glomerular and
et al. 1958). These lesions develop when complexes of serum albu- alveolar capillaries, triggering vasculitis (Wieslander et al. 1984).
min, antibody, and complement are formed in the circulation and To investigate the pathogenic mechanisms involved in anti-GBM
deposit in the vessel wall of arteries and glomeruli (Dixon et  al. disease, extensive studies have been performed in mice and rats
1958). Chronic serum sickness is induced by daily administration employing heterologous anti-GBM antibodies raised in another
of heterologous protein and results in chronic immune complex species, for example rabbit and sheep. The injection of heterolo-
glomerulonephritis and leukocytoclastic small-vessel vasculitis gous anti-GBM antibodies induces glomerulonephritis and in this
(Brentjens et  al. 1975). One of the important conclusions from model two phases of injury can be distinguished. The first phase
studies in these serum sickness models is that the extent and sever- is caused by the direct effect of anti-GBM antibody binding to the
ity of vasculitic manifestations greatly depend on the physical and GBM, which causes acute glomerular inflammation (Assmann
immunological characteristics of the complexes that are being et al. 1985; Schrijver et al. 1990). The second phase develops gradu-
formed. Factors such as antigen size, affinity, and isotype of the ally as a result of an immune response directed against the injected
antibody, and binding of complement factors determine whether heterologous anti-GBM antibodies (Unanue and Dixon 1965). This
these complexes deposit in vessels or are cleared from the circu- autologous response induces antibodies against the heterologous
lation (Cochrane and Hawkins 1968). Moreover, the presence of antibodies and can be accelerated by preimmunization with immu-
circulating immune complexes alone is not sufficient for vasculi- noglobulins of the same species in which the anti-GBM antibodies
tis development but this also depends on specific microcirculatory were generated. The antibodies directed against the heterologous
circumstances. Factors such as low flow velocities in capillaries anti-GBM antibodies form immune complex deposits along the
and postcapillary venules, and increased vascular permeability basement membrane and therefore this model represents a form
induced by vasoactive amines produced by platelets and mast cells of immune complex disease. The passive and accelerated models
all promote tissue deposition of immune complexes (Henson and of heterologous anti-GBM antibody-mediated glomerulonephritis
Cochrane 1971; McCluskey and Fienberg 1983). This could explain have been extensively used to dissect the role of various inflam-
why immune complex vasculitis preferentially develops in capillar- matory pathways in disease induction. Besides models based on
ies and postcapillary venules. antibody transfer, mouse and rat models of anti-GBM disease have
Once deposited in the vessel wall, immune complexes trigger a been developed that are induced by direct immunization with anti-
chain of inflammatory effector mechanisms (Jancar and Sanchez genic material from the GBM. Such models are characterized by
Crespo 2005). In the acute phase, inflammation is driven by glomerulonephritis and variable involvement of the lungs. Studies
antibody-dependent complement activation leading to the forma- in these models have clearly shown that besides anti-GBM autoan-
tion of chemotactic factors such as C3a and C5a. Both C3A and C5a tibodies, T cells specific for GBM antigens contribute to disease
are powerful chemoattractants for leukocytes, in particular neutro- development and, in some cases, are even sufficient to induce glo-
phils, and initiate their recruitment, infiltration, and activation. The merulonephritis (Wu et al. 2002; Wu et al. 2003).
acute phase is further characterized by endothelial necrosis due to
formation of the complement membrane attack complex (comple- Animal models of vasculitis induced
ment C5b-9 complexes) and the release of proteolytic enzymes and by infectious agents
oxygen radical species from activated neutrophils. At the same For the majority of primary vasculitides the causative factor is
time, the integrity and antithrombotic capacity of the endothe- unknown but in many cases a link with infections is suspected.
lium is impaired, causing deposition of plasma proteins and fibrin. This is also true for Kawasaki’s disease, in which clinical and epi-
Employing animal models, the inflammatory mechanisms that demiological evidence strongly suggest that the disease is triggered
contribute to immune complex-mediated vascular injury have been by an infectious agent although the responsible pathogen remains
extensively studied. In many cases these studies have taken advan- obscure (Burns and Glode 2004). However, in animal models bac-
tage of the availability of mice deficient in specific components pro- terial and fungal components have been shown to induce vasculitis
posed to be crucial for the induced inflammatory reaction. Such that resembles Kawasaki’s syndrome. In mice, multiple peritoneal
studies have shown that complement activation, coagulation fac- injections of a cell wall extract of Candida albicans in susceptible
tors, neutrophils, and Fcγ-receptor engagement on leukocytes all strains causes severe vasculitis in the coronary arteries and aortic
contribute to immune complex-mediated vasculitis (Sylvestre and root (Murata 1979). Pathologically, the vasculitic lesions are char-
Ravetch 1994; Mayadas et al. 2009). Importantly, these studies have acterized by granulomatous inflammation and destruction of the
also revealed that the endothelial cells themselves are active par- internal elastic lamina and media but are devoid of fibrinoid necro-
ticipants in immune complex-mediated inflammatory reactions sis (Takahashi et al. 2004). A second model of Kawasaki’s disease
through temporal expression of adhesion molecules and the secre- is based on a single intraperitoneal injection of a cell wall extract
tion of cytokines and chemokines (Ley et al. 2007). from Lactobacillus casei (LCCWE) (Lehman et  al. 1985; Schulte
et  al. 2009). Histopathologically, this causes proximal coronary
Animal models of anti-glomerular arteritis similar to what is observed in human biopsies. Thus, both
basement membrane disease models described above in this paragraph recapitulate histopatho-
A specific form of immune complex disease is anti-glomerular logical aspects of the human disease. Moreover, in both models, the
basement membrane (GBM) disease, or Goodpasture’s syndrome, efficacy of immunomodulatory treatments similar to those used in
96 SECTION 2 basic science

Kawasaki’s disease in children has been demonstrated. Because of MPO-deficient mice were injected into C57Bl6 mice. The antibody
these features, these are accepted models for dissecting pathogenic transfer recipient mice developed pauci-immune glomerulone-
mechanisms and testing of experimental therapies for Kawasaki’s phritis, mimicking the human disease more closely (Figure  8.2).
disease. The passive transfer model now set the stage for further investiga-
tions into effector mechanisms contributing to vascular injury and
Animal models of vasculitis associated with anti- for testing of experimental therapies. Subsequent studies using this
neutrophil cytoplasmic autoantibodies (ANCA) passive antibody transfer model demonstrated that neutrophils are
Anti-neutrophil cytoplasmic autoantibody-associated vasculitides the principal effector cells early on in the disease process and that
(AAV), including granulomatosis with polyangiitis and micro- disease severity can be augmented by adding lipopolysaccharide
scopic polyangiitis, are severe systemic autoimmune disorders (Xiao et al. 2005; Huugen et al. 2005). In addition, disease induc-
affecting small blood vessels leading to organ injury and failure tion and progression in this model is critically dependent on acti-
(Chen and Kallenberg 2010). AAV can affect any organ in the body vation of the alternative, but not classical, pathway of complement
although disease manifestations most frequently occur in the upper activation (Xiao et al. 2007). This was unexpected given the lack of
airways, lungs, and kidneys. In AAV, the autoimmune response is clinical evidence of complement activation in AAV but has led to
directed against neutrophil and monocyte lysosomal enzymes, in the development of clinical trial protocols of complement inhibi-
particular myeloperoxidase (MPO) and proteinase 3 (PR3). This tion in AAV.
autoimmune response is considered to be causally related to the In a modification of the anti-MPO IgG transfer model, mMPO-
development of vasculitis via both cellular and humoral immune immunized MPO-deficient mice are irradiated and transplanted
effector mechanisms. In vitro ANCA are able to activate primed with bone marrow (BM) from wild-type mice (Schreiber et al. 2006).
neutrophils, inducing the generation of reactive oxygen species and Because in the irradiated mice anti-MPO antibody production is
release of lytic enzymes. This effect is proposed to be a key event preserved, reconstitution with MPO-expressing BM cells triggers
in AAV pathogenesis because when it occurs at the site of the ves- the development of crescentic glomerulonephritis. This model has
sel wall, inflammation and vascular injury will ensue (Jennette proven particularly useful for more detailed analysis of the role of
et al. 2006). Over the past two decades, various animal models of leukocyte function in anti-MPO-mediated glomerulonephritis via
ANCA-mediated vasculitis have been proposed, primarily based transplantation of bone marrow from transgene or gene-deficient
on the premise that ANCA-mediated neutrophil activation is the mice (Heeringa and Little 2011; Coughlan et al. 2012).
central pathogenic mechanism causing vascular injury. Using vari- A different strategy for modelling MPO autoimmunity that
ous approaches, several rat models of MPO-ANCA-mediated vas- has been proposed involves immunization of C57BL/6 mice with
culitis were developed during the 1990s (Heeringa and Little 2011; either murine or human MPO in adjuvant (Ruth et al. 2006). This
Coughlan et  al. 2012). These models involved inducing autoim- approach was found to generate a cellular and humoral anti-MPO
munity in rats with mercuric chloride (a polyclonal B-cell stimula- response but in itself does not induce vasculitis. However, when
tor) and direct infusion of ANCA antigens into the kidney of MPO followed by a challenge with a subnephritogenic dose of heter-
preimmunized rats (Mathieson et al. 1992; Brouwer et al. 1993). ologous anti-GBM antibodies mice develop necrotizing crescen-
Furthermore, it was demonstrated that in MPO immunized rats, tic glomerulonephritis. This model has been found to depend on
the anti-MPO directed immune response severely aggravated mild T-cell-mediated immunity because mice lacking the ability to pro-
glomerular injury, induced by administration of subnephritogenic duce circulating antibody still develop disease while CD4+ T-cell
amounts of anti-glomerular basement membrane (anti-GBM) anti- depletion inhibits disease development (Ruth et al. 2006). Recent
bodies (Heeringa et al. 1996). Overall, these models provided evi- studies in this model have identified an immunodominant MPO
dence for the ability of anti-MPO antibodies to exacerbate renal T-cell epitope that initiates cell-mediated glomerular injury (Ooi
injury. However, in some models, the anti-MPO response was part et  al. 2012). From these studies it is proposed that tissue injury
of a broad polyclonal antibody profile, whereas in other models in MPO-ANCA-associated vasculitis is a two-step process that
the development of glomerulonephritis relied upon the formation requires local release of MPO by activated neutrophils, conceiv-
of immune complexes in the kidney. Because vasculitic lesions in ably induced by MPO-specific autoantibodies, which is recognized
AAV are characterized by a paucity of immune deposits in fully by MPO-specific autoreactive CD4+ T cells inducing delayed-type
developed lesions, these models therefore did not accurately model hypersensitivity-like vascular injury.
the pathology of MPO-ANCA-associated small-vessel vasculitis in Besides the mouse models described above in this section, a rat
humans. model of systemic anti-MPO-associated vasculitis has been devel-
A major breakthrough in the field came in 2002 when Xiao and oped (Little et al. 2005). Immunization with purified human MPO
colleagues reported the development of an acute passive transfer in complete Freund’s adjuvant of autoimmune-prone Wistar Kyoto
mouse model employing purified antibodies or splenocytes derived (WKY) rats leads to high-titre anti-human MPO-ANCA that
from MPO-deficient mice immunized with purified murine MPO cross-react with rat MPO. After several weeks, the induction of the
(mMPO) (Xiao et  al. 2002). Injection of mMPO-positive sple- anti-MPO immune response is accompanied by the development
nocytes into immune-deficient recipient mice (RAG 2-deficient of haematuria, albuminuria, and small-vessel vasculitis in the kid-
mice) caused, over the course of 6–13  days, severe necrotizing neys and lungs. Histopathologicaly, the rat model resembles to a
glomerulonephritis and, in some recipients, pulmonary capillari- large extent human ANCA vasculitis. Similar to the pauci-immune
tis. Interpretation of these experiments was hampered by the fact nature of the glomerular capillary lesions observed in human
that the splenocyte transfer recipients had substantial glomerular ANCA-associated glomerulonephritis, only limited deposits of
immune deposits. Therefore, a second set of experiments was per- immunoglobulins and complement factors are detected in the
formed in which purified antibodies taken from mMPO-immunized lesions.
CHAPTER 8 animal models of vasculitis 97

Mpo –/– transfer C57BI6


mMPO/CFA +/– LPS

Day 1 Day 7
(a) (c)

(b) (d)

Fig. 8.2 Histopathology in the MPO-ANCA mouse model induced by intravenous injection of purified anti-mouse MPO IgG and lipopolysaccharide. (a) Acute
glomerular infiltration of neutrophils (day 1, immunohistochemistry) and (b) glomerular capillary necrosis (H&E stain, magnification ×200). (c, d) Glomerular crescent
formation (day 7, PAS stain, magnification ×200 (C), ×400 (D)).

1 Mpo –/– C57BI6

mMPO/CFA transfer of anti-MPO IgG


+/– LPS
4
Irradiation
huMPO/CFA
2 Mpo –/– Mpo –/– Pauci-immune
crescentic
glomerulonephritis WKY rats
mMPO/CFA MPO+/+
BM Tx

3 C57BI6 C57BI6

mMPO/CFA Injection of
sheep anti-mouse GBM IgG

Fig. 8.3 Overview of animal models of MPO-ANCA vasculitis.


1. Mouse model induced by systemic injection of purified anti-mouse MPO IgG derived from mouse MPO-immunized MPO-deficient mice. Administration of the anti-MPO antibodies in
C57Bl6 mice induces crescentic glomerulonephritis. Disease severity can be enhanced by co-administration of LPS. (Xiao et al. 2002, Huugen et al. 2005)
2. Mouse model induced by transplantation of MPO-positive bone marrow in irradiated mMPO-immunized MPO-deficient mice resulting in crescentic glomerulonephritis. (Schreiber et al. 2006
3. Mouse model based on immunization of C57Bl6 mice with mouse MPO. This induces a cellular and humoral anti-MPO immune response. Development of glomerulonephritis is triggered by
administering a subnephritogenic dose of heterologous anti-GBM antibodies. (Ruth et al. 2006)
4. Rat model induced by immunization of Wistar Kyoto (WKY) rats with human MPO. The elicited immune response against human MPO cross-reacts with rat MPO and induces crescentic
glomerulonephritis after 4 to 8 weeks. (Little et al. 2005)
CFA, complete Freund’s adjuvant; GBM, glomerular basement membrane; BM Tx, bone marrow transplantation; mMPO, mouse myeloperoxidase; huMPO, human myeloperoxidase.
98 SECTION 2 basic science

In summary, employing a variety of approaches, mouse and interactions between genetic, immunological, and environmental
rat models of MPO-ANCA vasculitis have been developed, sub- factors. Consequently, various animal models have been identified
stantiating the pathogenic potential of MPO-ANCA (Figure 8.3). and developed in an attempt to recapitulate specific features of the
Yet these models also have their limitations (Heeringa and Little various vasculitic disorders. One should realize, however, that none
2011). Essentially, all models can not be regarded as true autoim- of these models exactly mirrors the human situation due to species
mune models as these are reliant on active immunization proce- specific physiological and genetic differences and the sometimes
dures. Also, in the anti-MPO IgG transfer mouse model, there is artificial way in which disease is induced. These models therefore
no requirement for breaking tolerance. Despite these limitations, inevitably have their limitations and extrapolating results from
these models have proven valuable for investigations into effector animal studies directly to the human situation is not possible. The
mechanisms that contribute to ANCA disease pathogenesis. real value of animal model experimentation is that it contributes to
generating mechanistic concepts of disease processes through the
Animal models of PR3-ANCA vasculitis identification of genetic, inflammatory and environmental factors
In contrast to the models of MPO-ANCA vasculitis, development that participate in vasculitis development. This will lead to a better
of a convincing animal model of PR3-ANCA has proven much understanding of disease processes and generate hypotheses that
more difficult. In one study, immunization of autoimmune prone can be tested in humans.
non-obese diabetic mice (NOD) with recombinant mouse PR3
(rmPR3) resulted in high-titre anti-PR3 antibodies but no disease References
(Primo et  al. 2010). Adoptive transfer of splenocytes from these Alexander, E.L., Moyer, C., Travlos, G.S., Roths, J.B., and Murphy, E.D.
immunized NOD mice into immune-deficient NOD mice (NOD/ (1985). Two histopathologic types of inflammatory vascular disease in
SCID) induced vasculitis and severe necrotizing glomerulonephri- MRL/Mp autoimmune mice. Model for human vasculitis in connective
tis whereas a similar transfer into C57Bl/into immune-deficient tissue disease. Arthritis and Rheumatism, 28, 1146–55.
C57Bl6 (C57bl6 Rag1−/−) mice did not cause disease. This sug- Andrews, B.S., Eisenberg, R.A., Theofilopoulos, A.N., Izui, S., Wilson, C.B.,
gests that host factors are crucial for disease development as well. McConahey, P.J., Murphy, E.D., Roths, J.B., and Dixon, F.J. (1978).
Spontaneous murine lupus-like syndromes. Clinical and immuno-
However, as mentioned earlier, an important drawback of the
pathological manifestations in several strains. Journal of Experimental
adoptive splenocyte approach is that it causes extensive immune Medicine, 148, 1198–215.
complexes deposits in the lesions and therefore does not resemble Assmann, K.J., Tangelder, M.M., Lange, W.P., Schrijver, G., and Koene,
human pauci-immune vasculitis. R.A. (1985). Anti-GBM nephritis in the mouse: severe proteinuria in
One of the problems encountered in attempting to model the heterologous phase. Virchows Archiv. A, Pathological Anatomy and
PR3-ANCA vasculitis relates to differences between species with Histopathology, 406, 285–99.
respect to expression and physicochemical properties of PR3. To Berden, J.H., Hang, L., McConahey, P.J., and Dixon, F.J. (1983). Analysis of
circumvent species differences, strategies have been employed to vascular lesions in murine SLE. I. Association with serologic abnormali-
ties. Journal of Immunology (Baltimore, Md.: 1950), 130, 1699–705.
develop humanized experimental model systems based on engraft-
Brehm, M.A., Shultz, L.D., and Greiner, D.L. (2010). Humanized mouse
ing immunodeficient mice with a human immune system. These
models to study human diseases. Current Opinion in Endocrinology
models use severe immune deficient NOD.Cg-Prkdcscid Il2rgtm1Wjl/ Diabetes and Obesity, 17, 120–5.
SzJ mice that lack B, T, and NK cells permitting engraftment of cells Brentjens, J.R., O’Connell, D.W., Albini, B., and Andres, G.A. (1975).
of human origin (Brehm et al. 2010). Studies in such humanized Experimental chronic serum sickness in rabbits that received daily
mice have demonstrated that injection of human anti-PR3 IgG multiple and high doses of antigen:  a systemic disease. Annals of the
together with lipopolysaccharide (LPS) induces vascular injury in New York Academy of Sciences, 254, 603–13.
kidneys and lungs (Little et al. 2012). Chimeric mice were gener- Brouwer, E., Huitema, M.G., Klok, P.A., de Weerd, H., Tervaert, J.W., Weening,
ated by injecting human haematopoietic stem cells into irradiated J.J., and Kallenberg, C.G. (1993). Antimyeloperoxidase-associated pro-
liferative glomerulonephritis: an animal model. Journal of Experimental
NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ followed by administration of
Medicine, 177, 905–14.
LPS to enhance neutrophil recruitment and activation, and puri-
Burnet, F.M. and Holmes, M.C. (1965). The natural history of the NZB/NZW
fied human anti-PR3 IgG. After 6  days, 39% of anti-PR3 treated F1 hybrid mouse: a laboratory model of systemic lupus erythematosus.
mice had haematuria, which was absent in chimeric mice treated Australasian Annals of Medicine, 14, 185–91.
with normal human IgG. Macroscopically, petechiae on the surface Burns, J.C. and Glode, M.P. (2004). Kawasaki syndrome. Lancet, 364, 533–44.
of lungs of anti-PR3 treated animals were observed with histologi- Chan, O.T., Hannum, L.G., Haberman, A.M., Madaio, M.P., and Shlomchik,
cal evidence of vasculitis and haemorrhage. In addition, anti-PR3 M.J. (1999). A novel mouse with B cells but lacking serum antibody
treated mice developed a mild form of pauci-immune proliferative reveals an antibody-independent role for B cells in murine lupus. Journal
glomerulonephritis, with infiltration of human and mouse leuko- of Experimental Medicine, 189, 1639–48.
cytes. Overall, these observations in this humanized mouse model Chen, M.  and Kallenberg, C.G. (2010). ANCA-associated vasculitides—
advances in pathogenesis and treatment. Nature Reviews Rheumatology,
support the pathogenic potential of PR3-ANCA and are consist- 6, 653–64.
ent with the contention that PR3-ANCA-mediated neutrophil Cochrane, C.G. and Hawkins, D. (1968). Studies on circulating immune
activation is a key mechanism of vasculitis development in ANCA complexes. 3. Factors governing the ability of circulating complexes to
disease. localize in blood vessels. Journal of Experimental Medicine, 127, 137–54.
Coughlan, A.M., Freeley, S.J., and Robson, M.G. (2012). Animal models of
Conclusion anti-neutrophil cytoplasmic antibody-associated vasculitis. Clinical and
Experimental Immunology, 169, 229–37.
The vasculitides are a heterogeneous group of disorders with diverse Dimitriu-Bona, A., Matic, M., Ding, W., Yang, C.P., and Fillit, H. (1995).
clinicopathological characteristics that are the result of complex Cytotoxicity to endothelial cells by sera from aged MRL/lpr/lpr mice is
CHAPTER 8 animal models of vasculitis 99

associated with autoimmunity to cell surface heparan sulfate. Clinical Ley, K., Laudanna, C., Cybulsky, M.I., and Nourshargh, S. (2007). Getting
Immunology and Immunopathology, 76, 234–40. to the site of inflammation:  the leukocyte adhesion cascade updated.
Dinarello, C.A. (2011). Interleukin-1 in the pathogenesis and treatment of Nature Reviews Immunology, 7, 678–89.
inflammatory diseases. Blood, 117, 3720–32. Little, M.A., Al-Ani, B., Ren, S., Al-Nuaimi, H., Leite, M. Jr, Alpers, C.E.,
Dixon, F.J., Andrews, B.S., Eisenberg, R.A., McConahey, P.J., Theofilopoulos, Savage, C.O., and Duffield, J.S. (2012). Anti-proteinase 3 anti-neutrophil
A.N., and Wilson, C.B. (1978). Etiology and pathogenesis of a spontane- cytoplasm autoantibodies recapitulate systemic vasculitis in mice with a
ous lupus-like syndrome in mice. Arthritis and Rheumatism, 21, S64–7. humanized immune system. PloS One, 7,e28626.
Dixon, F.J., Vazquez, J.J., Weigle, W.O., and Cochrane, C.G. (1958). Little, M.A., Smyth, C.L., Yadav, R., Ambrose, L., Cook, H.T., Nourshargh, S.,
Pathogenesis of serum sickness. AMA Archives of Pathology, 65, 18–28. and Pusey, C.D. (2005). Antineutrophil cytoplasm antibodies directed
Fauci, A.S., Haynes, B., and Katz, P. (1978). The spectrum of vasculitis: clini- against myeloperoxidase augment leukocyte-microvascular interactions
cal, pathologic, immunologic and therapeutic considerations. Annals of in vivo. Blood, 106, 2050–8.
Internal Medicine, 89, 660–76. Mathieson, P.W., Thiru, S., and Oliveira, D.B. (1992). Mercuric
Hamano, Y., Tsukamoto, K., Abe, M., Sun, G.D., Zhang, D., Fujii, H., Matsuoka, chloride-treated brown Norway rats develop widespread tissue injury
S., Tanaka, M., Ishida-Okawara, A., Tachikawa, H., Nishimura, H., including necrotizing vasculitis. Laboratory Investigation, 67, 121–9.
Tokunaka, K., Hirose, S., and Suzuki, K. (2006). Genetic dissection Matsuki, T., Isoda, K., Horai, R., Nakajima, A., Aizawa, Y., Suzuki, K., Ohsuzu,
of vasculitis, myeloperoxidase-specific antineutrophil cytoplasmic F., and Iwakura, Y. (2005). Involvement of tumor necrosis factor-alpha
autoantibody production, and related traits in spontaneous crescentic in the development of T cell-dependent aortitis in interleukin-1 receptor
glomerulonephritis-forming/Kinjoh mice. Journal of Immunology, 176, antagonist-deficient mice. Circulation, 112, 1323–31.
3662–73. Mayadas, T.N., Tsokos, G.C., and Tsuboi, N. (2009). Mechanisms of immune
Harper, J.M., Thiru, S., Lockwood, C.M., and Cooke, A. (1998). complex-mediated neutrophil recruitment and tissue injury. Circulation,
Myeloperoxidase autoantibodies distinguish vasculitis mediated by 120, 2012–24.
anti-neutrophil cytoplasm antibodies from immune complex disease McCluskey, R.T. and Fienberg, R. (1983). Vasculitis in primary vasculitides,
in MRL/Mp-lpr/lpr mice: a spontaneous model for human microscopic granulomatoses, and connective tissue diseases. Human Pathology, 14,
angiitis. European Journal of Immunology, 28, 2217–26. 305–15.
Heeringa, P., Brouwer, E., Klok, P.A., Huitema, M.G., Van Den Born, J., Moyer, C.F., Strandberg, J.D., and Reinisch, C.L. (1987). Systemic
Weening, J.J., and Kallenberg, C.G. (1996). Autoantibodies to myeloper- mononuclear-cell vasculitis in MRL/Mp-lpr/lpr mice. A histologic and
oxidase aggravate mild anti-glomerular-basement-membrane-mediated immunocytochemical analysis. American Journal of Pathology, 127,
glomerular injury in the rat. American Journal of Pathology, 149, 229–42.
1695–706. Murata, H. (1979). Experimental candida-induced arteritis in mice. Relation
Heeringa, P. and Little, M.A. (2011). In vivo approaches to investigate to arteritis in the mucocutaneous lymph node syndrome. Microbiology
ANCA-associated vasculitis: lessons and limitations. Arthritis Research and Immunology, 23, 825–31.
and Therapy, 13, 204. Neumann, I., Birck, R., Newman, M., Schnulle, P., Kriz, W., Nemoto, K., Yard,
Henson, P.M. and Cochrane, C.G. (1971). Acute immune complex disease in B., Waldherr, R., and van der Woude, F.J. (2003). SCG/Kinjoh mice: a
rabbits. The role of complement and of a leukocyte-dependent release model of ANCA-associated crescentic glomerulonephritis with immune
of vasoactive amines from platelets. Journal of Experimental Medicine, deposits. Kidney International, 64, 140–8.
133, 554–71. Nicklin, M.J., Hughes, D.E., Barton, J.L., Ure, J.M., and Duff, G.W. (2000).
Hicks, J.D. (1966). Vascular changes in the kidneys of NZB mice and F1 Arterial inflammation in mice lacking the interleukin 1 receptor antago-
NZBxNZW hybrids. Journal of Pathology and Bacteriology, 91, 479–86. nist gene. Journal of Experimental Medicine, 191, 303–12.
Horai, R., Saijo, S., Tanioka, H., Nakae, S., Sudo, K., Okahara, A., Ikuse, T., Nose, M., Nishimura, M., Ito, M.R., Toh, J., Shibata, T., and Sugisaki, T. (1996).
Asano, M., and Iwakura, Y. (2000). Development of chronic inflamma- Arteritis in a novel congenic strain of mice derived from MRL/Lpr lupus
tory arthropathy resembling rheumatoid arthritis in interleukin 1 receptor mice: genetic dissociation from glomerulonephritis and limited autoan-
antagonist-deficient mice. Journal of Experimental Medicine, 191, 313–20. tibody production. American Journal of Pathology, 149, 1763–9.
Huugen, D., Xiao, H., van Esch, A., Falk, R.J., Peutz-Kootstra, C.J., Buurman, Nose, M., Terada, M., Nishihara, M., Kamogawa, J., Miyazaki, T., Mori,
W.A., Tervaert, J.W., Jennette, J.C., and Heeringa, P. (2005). Aggravation S., Nishimura, M., Wang, Y., Kamoto, T., and Hiai, H. (1998).
of anti-myeloperoxidase antibody-induced glomerulonephritis by bac- Vasculitis-susceptible genes in mice with a deficit in Fas-mediated apop-
terial lipopolysaccharide: role of tumor necrosis factor-alpha. American tosis. International journal of cardiology, 66 (Suppl. 1), S37–41; S43.
Journal of Pathology, 167, 47–58. Ooi, J.D., Chang, J., Hickey, M.J., Borza, D.B., Fugger, L., Holdsworth, S.R.,
Jancar, S. and Sanchez Crespo, M. (2005). Immune complex-mediated tissue and Kitching, A.R. (2012). The immunodominant myeloperoxidase
injury: a multistep paradigm. Trends in Immunology, 26, 48–55. T-cell epitope induces local cell-mediated injury in antimyeloperoxidase
Jennette, J.C., Xiao, H., and Falk, R.J. (2006). Pathogenesis of Vascular glomerulonephritis. Proceedings of the National Academy of Sciences of
Inflammation by Anti-Neutrophil Cytoplasmic Antibodies. Journal of the United States of America, 109, E2615–24.
the American Society of Nephrology, 17, 1235–42. Pathak, S. and Mohan, C. (2011). Cellular and molecular pathogenesis of
Kinjoh, K., Kyogoku, M., and Good, R.A. (1993). Genetic selection for cres- systemic lupus erythematosus:  lessons from animal models. Arthritis
cent formation yields mouse strain with rapidly progressive glomerulo- Research and Therapy, 13, 241.
nephritis and small vessel vasculitis. Proceedings of the National Academy Primo, V.C., Marusic, S., Franklin, C.C., Goldmann, W.H., Achaval, C.G.,
of Sciences of the United States of America, 90, 3413–7. Smith, R.N., Arnaout, M.A., and Nikolic, B. (2010). Anti-PR3 immune
Lahmer, T. and Heemann, U. (2012). Anti-glomerular basement membrane responses induce segmental and necrotizing glomerulonephritis.
antibody disease: A rare autoimmune disorder affecting the kidney and Clinical and Experimental Immunology, 159, 327–37.
the lung. Autoimmunity reviews, 12, 169–73. Qu, W.M., Miyazaki, T., Terada, M., Lu, L.M., Nishihara, M., Yamada, A.,
Lehman, T.J., Walker, S.M., Mahnovski, V., and McCurdy, D. (1985). Mori, S., Nakamura, Y., Ogasawara, H., Yazawa, C., Nakatsuru, S., and
Coronary arteritis in mice following the systemic injection of group Nose, M. (2000). Genetic dissection of vasculitis in MRL/lpr lupus
B Lactobacillus casei cell walls in aqueous suspension. Arthritis and mice: a novel susceptibility locus involving the CD72c allele. European
Rheumatism, 28, 652–9. Journal of Immunology, 30, 2027–37.
Lerner, R.A., Glassock, R.J., and Dixon, F.J. (1967). The role of anti-glomerular Ruth, A.J., Kitching, A.R., Kwan, R.Y., Odobasic, D., Ooi, J.D., Timoshanko,
basement membrane antibody in the pathogenesis of human glomerulo- J.R., Hickey, M.J., and Holdsworth, S.R. (2006). Anti-neutrophil cyto-
nephritis. Journal of Experimental Medicine, 126, 989–1004. plasmic antibodies and effector CD4+ cells play nonredundant roles
100 SECTION 2 basic science

in anti-myeloperoxidase crescentic glomerulonephritis. Journal of the Wang, Y., Nose, M., Kamoto, T., Nishimura, M., and Hiai, H. (1997). Host
American Society of Nephrology, 17, 1940–9. modifier genes affect mouse autoimmunity induced by the lpr gene.
Schreiber, A., Xiao, H., Falk, R.J., and Jennette, J.C. (2006). Bone American Journal of Pathology, 151, 1791–8.
marrow-derived cells are sufficient and necessary targets to mediate glo- Wieslander, J., Barr, J.F., Butkowski, R.J., Edwards, S.J., Bygren, P., Heinegard,
merulonephritis and vasculitis induced by anti-myeloperoxidase anti- D., and Hudson, B.G. (1984). Goodpasture antigen of the glomerular
bodies. Journal of the American Society of Nephrology, 17, 3355–64. basement membrane: localization to noncollagenous regions of type IV
Schrijver, G., Bogman, M.J., Assmann, K.J., de Waal, R.M., Robben, H.C., collagen. Proceedings of the National Academy of Sciences of the United
van Gasteren, H., and Koene, R.A. (1990). Anti-GBM nephritis in States of America, 81, 3838–42.
the mouse:  role of granulocytes in the heterologous phase. Kidney Wu, J., Borillo, J., Glass, W.F., Hicks, J., Ou, C.N., and Lou, Y.H. (2003). T-cell
International, 38, 86–95. epitope of alpha3 chain of type IV collagen induces severe glomerulone-
Schulte, D.J., Yilmaz, A., Shimada, K., Fishbein, M.C., Lowe, E.L., Chen, S., phritis. Kidney International, 64, 1292–301.
Wong, M., Doherty, T.M., Lehman, T., Crother, T.R., Sorrentino, R., Wu, J., Hicks, J., Borillo, J., Glass, W.F. 2nd, and Lou, Y.H. (2002). CD4(+)
and Arditi, M. (2009). Involvement of innate and adaptive immunity T cells specific to a glomerular basement membrane antigen mediate
in a murine model of coronary arteritis mimicking Kawasaki disease. glomerulonephritis. Journal of Clinical Investigation, 109, 517–24.
Journal of Immunology (Baltimore, Md.: 1950), 183, 5311–8. Xiao, H., Heeringa, P., Hu, P., Liu, Z., Zhao, M., Aratani, Y., Maeda, N., Falk,
Shepherd, J., Little, M.C., and Nicklin, M.J. (2004). Psoriasis-like cutaneous R.J., and Jennette, J.C. (2002). Antineutrophil cytoplasmic autoantibod-
inflammation in mice lacking interleukin-1 receptor antagonist. Journal ies specific for myeloperoxidase cause glomerulonephritis and vasculitis
of Investigative Dermatology, 122, 665–9. in mice. Journal of Clinical Investigation, 110, 955–63.
Shepherd, J. and Nicklin, M.J. (2005). Elastic-vessel arteritis in interleukin-1 Xiao, H., Heeringa, P., Liu, Z., Huugen, D., Hu, P., Maeda, N., Falk, R.J., and
receptor antagonist-deficient mice involves effector Th1 cells and Jennette, J.C. (2005). The role of neutrophils in the induction of glo-
requires interleukin-1 receptor. Circulation, 111, 3135–40. merulonephritis by anti-myeloperoxidase antibodies. American Journal
Staszak, C. and Harbeck, R.J. (1985). Mononuclear-cell pulmonary vasculitis of Pathology, 167, 39–45.
in NZB/W mice. I. Histopathologic evaluation of spontaneously occur- Xiao, H., Schreiber, A., Heeringa, P., Falk, R.J., and Jennette, J.C. (2007).
ring pulmonary infiltrates. American Journal of Pathology, 120, 99–105. Alternative complement pathway in the pathogenesis of disease medi-
Sylvestre, D.L. and Ravetch, J.V. (1994). Fc receptors initiate the Arthus reac- ated by anti-neutrophil cytoplasmic autoantibodies. American Journal
tion: redefining the inflammatory cascade. Science, 265, 1095–8. of Pathology, 170, 52–64.
Takahashi, K., Oharaseki, T., Wakayama, M., Yokouchi, Y., Naoe, S., and Yamada, A., Miyazaki, T., Lu, L.M., Ono, M., Ito, M.R., Terada, M., Mori, S.,
Murata, H. (2004). Histopathological features of murine systemic vascu- Hata, K., Nozaki, Y., Nakatsuru, S., Nakamura, Y., Onji, M., and Nose, M.
litis caused by Candida albicans extract—an animal model of Kawasaki (2003). Genetic basis of tissue specificity of vasculitis in MRL/lpr mice.
disease. Inflammation Research, 53, 72–7. Arthritis and Rheumatism, 48, 1445–51.
Theofilopoulos, A.N. and Dixon, F.J. (1985). Murine models of systemic Zhang, M.C., Misu, N., Furukawa, H., Watanabe, Y., Terada, M., Komori,
lupus erythematosus. Advances in Immunology, 37, 269–390. H., Miyazaki, T., Nose, M., and Ono, M. (2006). An epistatic effect of
Unanue, E.R. and Dixon, F.J. (1965). Experimental glomerulonephritis. the female specific loci on the development of autoimmune vasculitis
Vi. the autologous phase of nephrotoxic serum nephritis. Journal of and antinuclear autoantibody in murine lupus. Annals of the Rheumatic
Experimental Medicine, 121, 715–25. Diseases, 65, 495–500.
CHAPTER 9

Pathological features
of vasculitis
Andrew Churg

Introduction and classification New names have been proposed for some types of vasculitis by
the 2012 Chapel Hill Consensus Conference (Jennette et al. 2013).
This chapter presents a brief overview and illustrations of the path- Whether all these proposals will be adopted is uncertain, so this
ological features of vasculitis. Pathological examination remains chapter uses traditional terminology but indicates the new pro-
the gold standard for the diagnosis of vasculitis, but arriving at a posed terminology as well.
definitive diagnosis on the basis of a biopsy (or autopsy) specimen
alone is not always straightforward. For one thing, only a limited
number of reaction patterns are seen in the vessels, but these identi-
Large-vessel vasculitis
cal reaction patterns can be found in a wide variety of conditions, The large-vessel vasculitides are defined as those that primar-
some of which require vastly different treatments and have consid- ily involve the aorta and its major branches, particularly the
erably different prognoses (Jennette and Falk 1997). Interpretation branches to the head and neck and to the upper and lower limbs
of biopsies for a diagnosis of vasculitis thus requires careful correla- (Jennette et al. 1994; Jennette and Falk 1997; Jennette and Falk
tion with clinical information and a variety of laboratory data, such 2000b; Lie 1989), but also the coronary and pulmonary arteries.
as antineutrophil cytoplasmic antibodies (ANCA) status, to arrive The primary entities included in this category are Takayasu’s arte-
at a precise diagnosis. ritis (TA) and giant cell arteritis (GCA) (Jennette et al. 2013), both
An additional problem is the wide range of pathological findings of which typically show a giant cell or granulomatous appearance.
that can be seen in some forms of vasculitis, some of which do not Both entities are relatively indolent processes that often have a
look vasculitic at all. Indeed, for purposes of pathological diagno- long time course and often progress to vascular fibrosis or fibrotic
sis, it is crucial to recognize a broad spectrum of morphological obliteration, manifested clinically as reduced pulses (TA is fre-
changes; insisting on a very narrow set of pathological findings quently referred to as ‘pulseless disease’), bruits, and claudication.
(particularly insisting on seeing ‘classic’ lesions) before making a It should be noted that other types of vasculitis can involve the
diagnosis of vasculitis leads to missed cases (Churg 2005). aorta and large vessels and have a granulomatous appearance (see
Sampling, both geographic and temporal, presents another prob- below in this Section), and that a granulomatous reaction may be
lem. In some forms of vasculitis, such as giant cell arteritis (GCA), seen in forms of vasculitis that involve medium or small vessels
lesions are typically focal. They may be missed completely because
of bad luck in the area biopsied, and may also be missed if the
specimen is not sectioned completely. Similarly, the pathologist Table 9.1 Diseases that can show a giant cell or granulomatous pattern
must be aware that vascular lesions change their appearance over of vasculitis
time; necrotizing arteritis with fibrinoid necrosis is readily diag-
nosed, but it is easy to mistake old sclerosed arteritic lesions for In large vessels
non-specific thrombosis. Takayasu’s arteritis (early-onset chronic aortoarteritis)
Lastly, as is true of many areas of pathology, understanding and Giant cell (temporal) arteritis (late-onset chronic aortoarteritis)
diagnosing vasculitis is greatly facilitated by reference to a defined Vasculitis associated with rheumatoid arthritis (uncommon)
classification scheme. A  variety of classification schemes have Vasculitis associated with lupus (uncommon)
been proposed, some of them are contradictory, and application In medium and small vessels
of different schemes sometimes places given patients into differ- Polyarteritis nodosa (PAN) (uncommon)
ent diagnostic categories, including categories that may require Kawasaki’s disease (uncommon)
different treatment. The discussion in this chapter follows the gen- Vasculitis associated with rheumatoid arthritis (uncommon)
eral scheme elucidated by the Chapel Hill Consensus Conference Vasculitis associated with tuberculous and fungal infections (common)
(Jennette et  al. 1994)  (see Table  1.1) with subsequent modifica- Sarcoidosis (common)
Buerger’s disease (common)
tions as noted (Jennette and Falk 2000a; Jennette and Falk 2000b;
Idiopathic granulomatous vasculitis (by definition)
Jennette et al. 2013), and as discussed in Chapter 1. This scheme
Granulomatous vasculitis of the CNS (by definition)
classifies vasculitis primarily by the size of vessel involved.
102 SECTION 2 basic science

(a) (b)

1.0 mm
100 µm

Fig. 9.1 Takayasu’s arteritis (early-onset chronic aortoarteritis). Low-power (a) and higher-power (b) micrographs of the aorta showing a somewhat thickened wall and
collections of inflammatory cells and characteristic giant cells. Courtesy of William D. Edwards, MD, Mayo Clinic, Rochester, Minnesota.

(Table 9.1). Thus, the presence of giant cells by themselves is not that are calcified and are a form of foreign body reaction to the
diagnostic of GCA or TA. calcium (Petursdottir et  al. 1996). Infarct-like necrosis has been
The clinical features of GCA and TA are discussed in detail in reported (Heggtveit et al. 1963). The inflammatory process occa-
Chapters 23 and 24. Because the morphology is essentially identi- sionally extends into surrounding tissue such as retroperitoneal fat,
cal, separation is somewhat arbitrarily based on: age (greater than mediastinal fat, and the vena cava (Kinare 1975).
50 years of age for GCA, less than 50 years of age for TA); sex (pre- The inflammatory reaction slowly destroys the elastic laminae
dominantly female in TA); the frequent association of polymyalgia of the vessel wall, and replaces the elastic tissue with fibrous tis-
rheumatica with GCA; and to some extent the sites of involvement. sue. The inflammatory infiltrate may largely or totally disappear,
TA tends to involve the extremities and abdominal aorta, while although if the disease recurs, both chronic inflammatory cells and
GCA commonly involves the temporal artery and other extrac- giant cells reappear. The vessel wall in the chronic phase is typi-
ranial branches of the carotid, with only about 10 to 15% of cases cally thickened (Figures 9.2 and 9.3) and the lumen, particularly
having clinically overt disease outside this region (Lie 1991a). in limb vessels, coronary arteries, and pulmonary arteries, may be
In the acute phase, TA appears as a patchy inflammatory infiltrate severely compromised (Figure 9.2). The severity of wall thickening
that is predominantly lymphocytes and plasma cells, sometimes appears to be proportional to the duration of disease (Lie 1991a).
with an admixture of eosinophils and histiocytes (Figure 9.1). The Thromboses may further occlude the lumen. Large-vessel ectasias/
number of giant cells is variable; in some cases they occur singly, aneurysms and dissections also occur. Hypertensive and ischaemic
and in others form poorly defined granulomas in the vessel wall changes are typically seen in the kidney secondary to renal artery
(Lie 1991a); in some otherwise typical cases giant cells may not be occlusion.
found, so their absence is not prima face evidence against the diag- GCA, frequently called temporal arteritis (see Chapter  23), is
nosis. It has been claimed that the giant cells only occur in vessels morphologically similar to TA (Figures 9.4, 9.5, 9.6, and 9.7), but

Fig. 9.2 Takayasu’s arteritis (early-onset chronic aortoarteritis). Gross photograph


of aortic arch branches showing marked lumenal narrowing and thickening of Fig. 9.3 Takayasu’s arteritis (early-onset chronic aortoarteritis). In this example of
the arterial wall. Courtesy of William D. Edwards, MD, Mayo Clinic, Rochester, long-standing burnt out disease in the aorta, most of the elastica has disappeared
Minnesota. and been replaced by fibrous tissue.
CHAPTER 9 pathological features of vasculitis 103

(a)

(b)

Fig. 9.4 Giant cell arteritis (late-onset chronic aortoarteritis). This figure shows
the proper way to process temporal artery specimens for histology; the arterial
segment is cut into numerous short pieces and all are embedded in one or more
tissue blocks and then sectioned at multiple levels.

most cases involve the extracranial branches of the carotid artery,


with the temporal, vertebral, ophthalmic, and posterior ciliary
involved alone or in some combination in about 75% of cases
(Wilkinson and Russell 1972). Clinically evident involvement of
the aorta and large branches (Figure 9.7) is less common, although
clinically unapparent disease in these vessels can be demonstrated Fig. 9.5 Giant cell arteritis (late-onset chronic aortoarteritis). Low (a) and
on angiography in some patients with otherwise typical temporal higher-power (b) views of a temporal artery showing extensive inflammation
arteritis. Conversely, about 25% of patients with disease clinically involving largely the outer portion of the vessel wall; in (b) giant cells and
located outside the head have radiographic evidence of temporal inflammatory cells are seen to disrupt the muscularis.
arteritis (Lie 1995).
Arteritis of the temporal arteries has been studied in great detail
and there are numerous morphological variations. Giant cells are
not always present, or may only be seen in small side branches, and
occasionally only the temporal vein and not the artery is involved
(Lie 1996a). Fibrinoid necrosis of the outer zone of the intima adja-
cent to the internal elastica has been reported by Lie (1996a) in 25%
of a very large series of cases; however, its presence should raise
the issue of whether the lesion is question is really GCA or another
form of systemic vasculitis, as other systemic vasculitides can
involve the temporal artery (Lie 1996a; Walz LeBlanc et al. 1994).
In some instances the question arises of whether a fibrotic,
pauci-inflammatory lesion is old (healed) GCA, or merely arte-
riosclerosis. Both can largely or completely obstruct the lumen.
A useful rule of thumb is that arteriosclerosis produces minor loss
or reduplication of elastica; however, if there is disorganization
and loss of long elastic segments (Figure 9.6), this is more likely
to be the residue of GCA (Ashton-Key and Gallagher 1991; Lie
1996a). Fig. 9.6 Giant cell arteritis (late-onset chronic aortoarteritis). Elastic stain of one
Because the temporal artery is amenable to biopsy, many pathol- of the vessel segments illustrated in Fig. 9.4. Note loss of elastic over most of the
ogy specimens derive from this site. The reported sensitivity of circumference of the vessel, a characteristic finding in many forms of vasculitis.
104 SECTION 2 basic science

(a) (b)

(c)

Fig. 9.7 Giant cell arteritis (late-onset chronic aortoarteritis) involving a brachial artery. (a) This very-low-power view of a resected segment trisected shows complete luminal
occlusion. Medium (b) and high-power (c) views show inflammatory involvement of the arterial wall with giant cells, and fibrous obliteration (old thrombosis) of the lumen.

temporal artery biopsy in the literature varies from 65 to 97% Medium-vessel vasculitis
(Jacobs and Allen 1991) and bilateral biopsy is reported in some
studies to considerably increase the yield (Ponge et  al. 1988). Medium-vessel vasculitides, such as polyarteritis nodosa (PAN)
Because the lesions are segmental, with skip areas, temporal artery and Kawasaki’s disease (KD) (mucocutaneous lymph node syn-
and other putative GCA specimens need to be cut into short (1 to drome), involve the main visceral arteries and their branches.
2 mm) lengths, embedded completely, and then sectioned at multi- They may involve smaller arteries as well; however, by definition,
ple levels to ensure that a diagnostic area is not missed (Nordborg they do not involve arterioles, capillaries, or venules (Jennette
and Nordborg 1995; Lie 1996a) (Figure 9.4). It is not uncommon to and Falk 1997; Jennette and Falk 2000b; Jennette et  al. 2013).
receive biopsies after glucocorticoid therapy has been started, but Medium-vessel vasculitides typically start with a necrotizing
Achkar et al. (1994) showed that even 14 days of steroid treatment phase which often affects the vessel in an asymmetric fashion
does not affect the positive diagnosis rate in temporal artery speci- (Figure 9.8). In many instances the destructive process is manifest
mens, although the morphological appearances may be altered. as so-called fibrinoid necrosis of the vessel wall, that is replace-
Vasculitis involving the aorta and its branches with a patho- ment of the wall by granular pink material that resembles fibrin
logical pattern of GCA has been reported to occur occasionally in on haematoxylin and eosin stain (Figure 9.8c), usually accompa-
patients with rheumatoid arthritis (RA) and systemic lupus erythe- nied by an infiltrate of polymorphonuclear leukocytes. However,
matosus (SLE) (Lie 1989; Bahlas et al. 1998; Hall et al. 1983; Miller fibrinoid necrosis is not always present and the inflammatory
and Laeszewski 2011). Other types of vasculitic and primarily infiltrate can be predominantly chronic inflammatory cells. The
non-vasculitic disease can show a granulomatous vasculitic pattern vasculitic process is segmental and may be focal. In fact it is
in medium and small vessels, as listed in Table 9.1. characteristic to see acute lesions, healed or healing lesions, and
CHAPTER 9 pathological features of vasculitis 105

(a) (b)

(c) (d)

Fig. 9.8 Polyarteritis nodosa involving relatively large intrarenal arteries. (a) Note the marked variation in degree of involvement in nearby vessels (probably all branches
of the same artery). The branch 1 shows minimal damage, while the smaller branch 2 demonstrates loss of elastic over half of the perimeter. Branch 3 shows nearly
complete destruction of the wall. Elastic stain. Medium (b) and higher-power (c) views of the vessel in (a). Note the characteristic focal necrosis (arrows) which in (c) has
the appearance of so-called fibrinoid. (d) Old healed lesion showing marked lumenal narrowing and loss of elastica in the same kidney. Loss of elastica indicates that this
lesion did not result from bland thrombosis. Elastin stain.

totally uninvolved vessels in the same tissue section (Figure 9.8a). KD can occasionally show a granulomatous arteritis (Lie 1989;
This is the classic microscopic picture of vasculitis but its lack Landing and Larson 1987).
of diagnostic specificity for medium-vessel vasculitis must be The older literature is confusing because of the failure to separate
stressed, because an identical picture can be seen in many forms PAN and MPA, and it is easy to find statements that polyarteri-
of small-vessel vasculitis. tis causes a necrotizing glomerulonephritis. By current definitions
Inflammatory destruction of the wall can result in arterial such cases are labelled MPA, a form of small-vessel vasculitis.
rupture, thrombosis, and aneurysm formation. With time the Polyarteritis causes renal disease by occlusion of large vessels
inflammatory infiltrate becomes composed entirely of chronic (Figure 9.8), leading to ischaemia or infarction.
inflammatory cells and then disappears, and the wall progres- Although KD is morphologically similar to PAN (Figure 9.9), the
sively scars, in some instances leading to fibrotic obliteration of clinical setting is quite different, because it affects largely children
the lumen (Figure  9.8d). At this stage there can be significant under 5 years, and starts as an acute febrile illness associated with
morphological overlap with the changes seen in large-vessel vas- mucosal changes, desquamation in the extremities, and enlarged
culitis, and sometimes also difficulty in determining whether a lymph nodes (see Chapter 28). Small aneurysms are characteristic
vessel is merely thrombosed or the previous site of vasculitis. of both PAN and KD, but in the latter they commonly affect the
Elastic stains are very useful; loss of large portions of elastic lami- coronary arteries (Figure 9.9), although other sites are involved as
nae favour a diagnosis of vasculitis (Figure 9.8d), because ordi- well (Landing and Larson 1987; Naoe et al. 1991). Veins appear to be
nary bland thrombi do not destroy the elastica. Both PAN and involved fairly frequently in KD, and much less commonly in PAN.
106 SECTION 2 basic science

(a) (b)

Fig. 9.9 Kawasaki disease. (a) Gross photograph of heart of a child at autopsy showing aneurysmally dilated coronary artery with intralumenal thrombus. (b) Necrotizing
arteritis involving a coronary artery. Courtesy of Glenn P. Taylor, MD, Division of Pathology, Hospital for Sick Children, Toronto, Ontario.

Small-vessel vasculitides syndrome (leukocytoclastic vasculitis and glomerulonephritis),


typical of cryoglobulinaemia and Henoch–Schönlein purpura (HSP,
Small-vessel vasculitides by definition involve arterioles, capillaries, now known as IgA vasculitis, IgAV), or the pulmonary–renal syn-
and venules. Cutaneous leukocytoclastic vasculitis, various forms of drome (glomerulonephritis with pulmonary haemorrhage) most
glomerulonephritis, and pulmonary capillaritis with haemorrhage commonly seen in Wegener’s granulomatosis (now called granulo-
are characteristic findings in many different forms of small-vessel matosis with polyangiitis, GPA, see below), MPA, SLE, and occa-
vasculitis, and they are not seen in medium and large-vessel vascu- sionally in Churg–Strauss syndrome (now known as eosinophilic
litides. These findings often go together to produce the dermal–renal granulomatosis with polyangiitis, EGPA). In the past, the term

(a) (b)

Fig. 9.10 Microscopic polyangiitis. Low (a) and high-power (b) views of necrotizing vasculitis involving a small artery in a muscle biopsy. The arterial wall is completely
replaced by fibrinoid necrosis and an acute inflammatory infiltrate is present. This appearance can be seen in any form of small-vessel vasculitis and also in medium-vessel
vasculitis (polyarteritis nodosa, Kawasaki’s disease) when it involves small arteries.
CHAPTER 9 pathological features of vasculitis 107

hypersensitivity vasculitis, originally coined by Zeek et al. (1948), complexes, whereas IgAV, cryoglobulinaemia, and vasculitis asso-
has been used for some forms of small-vessel vasculitis, but this ciated with collagen vascular disease are ANCA-negative (at least
term is not recommended because of its lack of specificity. negative for myeloperoxidase and anti-proteinase 3 ANCA) and
Most forms of small-vessel vasculitis can also involve medium or are associated with immune complex deposition. Drugs can pro-
small arteries and produce necrotizing lesions (Figure 9.10) identi- duce a picture of small-vessel vasculitis, most commonly cutaneous
cal to those seen in PAN, so that biopsies showing only this pattern leukocytoclastic vasculitis, and may be associated with ANCA or
are difficult to classify without further information. By the definition immune complex formation or both (see below).
given above, the vasculitides associated with connective tissue dis- Cutaneous leukocytoclastic vasculitis involves primarily post-
eases, notably SLE and RA, are designated as forms of small-vessel capillary venules in the upper dermis. The venules show endothelial
vasculitis, but they can involve much larger vessels, including the swelling and, occasionally, actual fibrinoid necrosis (Figure 9.11).
aorta (see below). This is true also of Behçet’s syndrome (BS), where Most show an infiltrate of polymorphonuclear leukocytes, occa-
in fact large-vessel vasculitis is common (see below). Aneurysms sionally accompanied by eosinophils, and the neutrophils typically
have been viewed as pathognomonic of PAN, and are also common undergo karyorrhexis, so that nuclear fragments and extravasated
in KD; however, aneurysms can be seen in small-vessel vasculitides red cells are found in the dermis around the affected vessels. In
(Jennette and Falk 2000b; Jennette et al. 2001; Lie 1989) and do not EGPA, the infiltrate can be mostly or entirely composed of eosin-
allow separation of medium and small-vessel diseases. ophils. In some instances the infiltrate is composed of lympho-
Information on ANCA status and the presence of immune com- cytes and plasma cells and there is minimal or no karyorrhexis.
plexes or circulating antibodies is extremely helpful in subclassify- It is unclear whether this appearance represents a late or healing
ing small-vessel vasculitis (see Chapter 6). GPA, EGPA, and MPA stage of the form with neutrophils, or has a different pathogenesis
are typically ANCA-positive and do not show deposition of immune (Swerlick and Lawley 1991).

(a) (b)

(c)

Fig. 9.11 Cutaneous leukocytoclastic vasculitis. Low-power (a) and high-power (b) views showing a perivascular inflammatory infiltrate with extensive nuclear
karyorrhexis and red cell extravasation, along with infiltration of the wall of a small venule by neutrophils. (c) Fibrinoid necrosis of a small dermal vessel. Leukocytoclastic
vasculitis may be seen in any type of small-vessel vasculitis; this example is a drug reaction caused by trimethoprim/ sulfamethoxazole.
108 SECTION 2 basic science

Table 9.2 Diagnostic separation scheme for small-vessel vasculitides

Pauci-immune (generally Immune complex positive


ANCA positive)
Wegener’s granulomatosis Collagen-vascular diseases:
(granulomatosis with polyangiitis): immune complexes visible by light
Granulomatous vasculitis with or electron microscopy
neutrophils
Churg–Strauss syndrome (eosinophilic Henoch–Schönlein purpura (IgA
granulomatosis with polyangiitis): vasculitis): complexes contain IgA
Asthma, eosinophilia, eosinophilic
vasculitis, sometimes granulomas with
eosinophilic necrosis
Microscopic polyangiitis: no granulomas, Cryoglobulinaemic
no asthma or eosinophils vasculitis: circulating cryoglobulins
Some drug-induced vasculitis: usually Some drug-induced vasculitis
myeloperoxidase ANCA or atypical
Fig. 9.12 Pulmonary capillaritis. Characteristic pattern of neutrophils in alveolar walls pANCA positive (other antigens)
and evidence of acute and chronic haemorrhage (haemosiderin-laden macrophages)
in airspaces. This example is from a patient with rheumatoid arthritis and hemoptysis.
MPA (Lauque et al. 2000), although survival is much better in
Pulmonary capillaritis consists of an infiltrate of neutrophils that patients with minor or subclinical hemoptysis (Cordier and Cottin
remains largely confined to the interstitium of the alveolar walls 2011). For this reason, cases with clinical massive hemoptysis
(Figure  9.12), although there may be spill-over into the alveoli. need to be reported by the pathologist in an emergent fashion and
Capillaritis is usually accompanied by recent and old haemor- treated immediately. Necrotizing and crescentic (Figure 9.13) glo-
rhage, the latter manifest as haemosiderin-laden macrophages in merulonephritis is characteristic of WG, MPASS, and, occasionally,
the air spaces (Figure  9.12). Although the process is referred to CSS (Falk et al. 2000) and may accompany pulmonary capillaritis.
as capillaritis, involvement of small arterioles and venules is fre-
quently seen (Myers and Katzenstein 1986). Foci of granulation tis- Specific forms of small-vessel vasculitis
sue (bronchiolitis obliterans organizing pneumonia (BOOP)) are Specific forms of small-vessel vasculitis are discussed in this sec-
sometimes present and represent a response to haemorrhage. tion; Table 9.2 provides a scheme for separating the small-vessel
Capillaritis can be very subtle, but one useful aid in histological vasculitides by morphology and laboratory findings.
diagnosis is the presence of fibrin tufts attached to alveolar walls; ANCA-positive vasculitis
careful examination, perhaps aided by elastic stains or stains for Granulomatosis with polyangiitis
basement membrane, will reveal that the underlying alveolar wall The classic formulation of GPA involved a triad of upper airway lesions,
is disrupted or actually necrotic. necrotizing granulomatous vasculitis in the lung, and necrotizing glo-
Many cases of pulmonary capillaritis are manifest clinically by merulonephritis (Godman and Churg 1954). Carrington and Liebow
massive hemoptysis. The presence of capillaritis in patients with (1966) subsequently described what they labelled ‘limited Wegener’s
massive hemoptysis is associated with a mortality of up to 50% in
patients with SLE (Zamora et al. 1997) and 31% in patients with

Fig. 9.14 Wegener’s granulomatosis (granulomatosis with polyangiitis). Characteristic


pattern of ‘geographic necrosis’ (irregular necrosis); the surrounding palisade of
Fig. 9.13 Crescentic glomerulonephritis from a case of microscopic polyangiitis. epithelioid histiocytes, that is the ‘granulomatosis’, cannot be seen at this low power.
CHAPTER 9 pathological features of vasculitis 109

giant cells and chronic inflammatory cells (Figure 9.16); how-


ever, sarcoid-type hard granulomas are not a feature of GPA and
their presence indicates an alternate diagnosis. Vasculitis is not
always obvious, particularly in lung biopsies, but this does not
change the diagnosis. Capillaritis with haemorrhage is common
(Figure 9.17) and may be the sole manifestation of GPA in the
lung (Travis et al. 1987).
Early lesions of GPA in the lung often show no vasculitis at all;
rather there is necrosis of collagen with small neutrophilic abscesses
(Figure  9.18). Over time these become necrotic and develop the
characteristic granulomatous response. This pattern is also seen in
some examples of GPA involving the orbit. Outside the lung, the
granulomatous response is also much less frequent and the typical
vasculitis shows neutrophils and fibrinoid necrosis (Figure 9.19),
although, again, a wide range of patterns is seen. Infarcts are com-
mon when fairly large vessels are involved. In the upper respiratory
tract, vasculitis is relatively uncommon, or at least hard to demon-
Fig. 9.15 Wegener’s granulomatosis (granulomatosis with polyangiitis). Small
strate on biopsy (Devaney et al. 1990), and most lesions appear as
artery in lung showing necrotizing vasculitis. non-specific ulcers, sometimes containing giant cells (Figure 9.20).
A  positive proteinase 3 or myeloperoxidase ANCA is extremely
helpful in making a diagnosis in this setting.
granulomatosis,’ in which other organs, particularly the skin, were
involved. It is now recognized that GPA can present in virtually any
organ, and that the pattern of organ involvement frequently changes
over time. It is, in fact, relatively uncommon to see the classic triad of
(a)
involvement at the time of initial presentation. Thus a broader and
more encompassing view of organ involvement by GPA must be kept
in mind. Active granulomatosis with polyangiitis is almost always
cANCA (proteinase 3) positive (Finkielman et al. 2007).
The vascular lesions in GPA are variable. In the lung the clas-
sic finding is basophilic necrosis with a surrounding palisade
of epithelioid histiocytes, that is, granulomatosis, producing
so-called geographic necrosis (Figure  9.14). This process may
involve vessels or the parenchyma itself; however, the vascu-
litis in the lung may simply have giant cells with no necrosis,
or fibrinoid necrosis (Figure 9.15), or sometimes just an infil-
trate of chronic inflammatory cells. The vasculitis and necrosis
are superimposed on a parenchymal background of individual

(b)

Fig. 9.17 Wegener’s granulomatosis (granulomatosis with polyangiitis). (a) Gross


Fig. 9.16 Wegener’s granulomatosis (granulomatosis with polyangiitis). Typical photograph showing extensive haemorrhage and an irregular upper lobe
inflammatory background seen in the lung, and composed of acute and chronic necrotizing mass lesion. (b) Microscopic pattern of capillaritis with haemorrhage, a
inflammatory cells and individual giant cells, but not hard granulomas. frequent finding in the lung in Wegener’s granulomatosis.
110 SECTION 2 basic science

(a)

Fig. 9.18 Wegener’s granulomatosis (granulomatosis with polyangiitis). Early


lesion consisting of a small neutrophilic abscess. In this example a palisade of (b)
epithelioid histiocytes is beginning to form; eventually such lesions end up as large
or small areas of geographic necrosis.

Eosinophilic granulomatosis with polyangiitis


(formerly known as churg−strauss syndrome)
Patients with eosinophilic granulomatosis with polyangiitis
(EGPA) have asthma or allergic rhinitis and, usually, blood eosino-
philia. The original cases (Churg and Strauss 1951) all had a com-
bination of eosinophilic tissue infiltration, necrotizing vasculitis,
and granulomas with eosinophilic material in the centres. In prac-
tice, this definition has proven to be much too restrictive, and few
cases, at least on biopsy, show all three features (Lanham et al. 1984;
Churg 2001).
A more realistic view, similar to that for GPA, is that EGPA is a vas-
culitis with variable manifestations. In practice, EGPA can be divided
into early or prevasculitic cases, cases with overt vasculitis, and cases Fig. 9.20 Wegener’s granulomatosis (granulomatosis with polyangiitis). Biopsy of
a nasal ulcer. (a) Most of the lesion is non-specific inflammation; a few giant cells
with healed vasculitis (Churg 2001; Churg 2005). These distinctions
are seen in the high power view (b). This appearance by itself is not diagnostic;
however, if there is a positive cANCA, then these findings support a diagnosis of
Wegener’s granulomatosis.

are of prognostic and treatment value, because early-phase disease


responds well to glucocorticoids, but vasculitic-phase disease often
requires addition of a cytotoxic agent and has associated mortality.
EGPA may be positive for pANCA (antimyeloperoxidase antibodies)
By definition, in early-phase disease, vasculitis cannot be dem-
onstrated clinically or on biopsy. Biopsy specimens typically
show only an eosinophilic tissue infiltrate. In the lung this is
identical to chronic eosinophilic pneumonia (Figure 9.21), but
may also manifest as eosinophilic gastroenteritis or eosinophilic
lymphadenitis.
In the vasculitic phase, the most characteristic lesion is a
necrotizing vasculitis with fibrinoid necrosis that by itself cannot
be distinguished from other small- and medium-sized vasculitides
except for the background eosinophil infiltrate in the surround-
ing tissue (Figure  9.22). The vasculitis need not be necrotizing;
Fig. 9.19 Wegener’s granulomatosis (granulomatosis with polyangiitis). sometimes it simply consists of eosinophilic infiltration of vessels
Necrotizing vasculitis with fibrinoid necrosis involving a mesenteric artery. without necrosis (Figure 9.23), and it may also be granulomatous
CHAPTER 9 pathological features of vasculitis 111

(Figure  9.24). Eosinophilic capillaritis can be seen in the lung


(Figure  9.25) but it is relatively uncommon (about 3% of cases
in the series of Guillevin et  al. (1999a)), and eosinophilic leu-
kocytoclastic vasculitis can be present in the skin (Figure 9.26).
Eosinophilic infiltration of parenchymal tissues without vasculitis,
for example eosinophilic myocarditis, is common (Figure  9.27).
Some cases show granulomas, typically formed on vessels, and
containing eosinophilic necrotic centres (Figure 9.28), but the pro-
portion of cases with granulomas in modern material is relatively
small. In the postvasculitic phase the microscopic appearance is
that of a burnt out vasculitis, typically with vascular occlusion and
loss of elastica.
Microscopic polyangiitis
MPA is, by definition, an exclusionary diagnosis for a patient with
a positive ANCA (either antiproteinase 3 or antimyeloperoxidase)
and small-vessel vasculitis with no history of asthma/ allergic rhi-
nitis, no eosinophilia, and no granulomatous disease on biopsy.
Fig. 9.21 Churg–Strauss syndrome (eosinophilic granulomatosis with MPA again shows, in its most characteristic form, disseminated
polyangiitis). Eosinophilic pneumonia-like area without vasculitis from a case of necrotizing vasculitis with fibrinoid necrosis (Figure 9.10), but may
early-phase (prevasculitic phase) disease. In this example there is eosinophilic
necrosis, but this is a common finding in eosinophilic pneumonia of any cause
and is not diagnostic of Churg–Strauss syndrome.

(a) (b)

(c)

Fig. 9.22 Churg–Strauss syndrome (eosinophilic granulomatosis with polyangiitis). Various appearances of necrotizing vasculitis in heart (a), nerve (b), and mesentery
(c). Only the eosinophilic inflammatory infiltrate allows morphological separation from other forms of small-vessel vasculitis.
112 SECTION 2 basic science

Fig. 9.23 Churg–Strauss syndrome (eosinophilic granulomatosis with Fig. 9.26 Churg–Strauss syndrome (eosinophilic granulomatosis with
polyangiitis). Non-necrotizing eosinophilic vasculitis in the lung. polyangiitis). Leukocytoclastic vasculitis in a patient with palpable purpura. Note
the mixture of eosinophils and neutrophils, commonly seen in leukocytoclastic
vasculitis in Churg–Strauss syndrome.

Fig. 9.24 Churg–Strauss syndrome (eosinophilic granulomatosis with


polyangiitis). Granulomatous eosinophilic vasculitis.
Fig. 9.27 Churg–Strauss syndrome (eosinophilic granulomatosis with
polyangiitis). Eosinophilic myocarditis.

Fig. 9.25 Churg–Strauss syndrome (eosinophilic granulomatosis with


polyangiitis). Eosinophilic capillaritis in the lung. In Churg–Strauss syndrome the Fig. 9.28 Churg–Strauss syndrome (eosinophilic granulomatosis with polyangiitis).
infiltrates in capillaritis may be only eosinophils or a mixture of eosinophils and Necrotizing granuloma with an eosinophilic centre (so-called Churg–Strauss
neutrophils. granuloma). Such lesions are relatively uncommon in modern pathology specimens.
CHAPTER 9 pathological features of vasculitis 113

also be manifest as necrotizing and crescentic glomerulonephritis (a)


(seen in 90% or more of cases) (Jennette et al. 2001) (Figure 9.13),
leukocytoclastic vasculitis in the skin, and capillaritis in the lung
(Figure 9.29). MPA is the most common cause of the pulmonary–
renal syndrome (Jennette et al. 2001).
Although most cases of MPA appear as relatively acute disease, in
some instances the course is much more indolent (Guillevin et  al.
1999b) and patients may report months to years of arthalgias or epi-
sodes of minor haemoptysis (Savage et al. 1985). Such patients some-
times develop interstitial pulmonary fibrosis secondary to chronic
haemorrhage (Eschun et al. 2003; Nada et al. 1990; Burns 1998; Gaudin
et al. 1995; Homma et al. 2004; Bhanji and Karim 2010) (Figure 9.30).
Vasculitis associated with immune complex deposition
Vasculitis in connective tissue diseases
Vasculitis is reported to occur in 5 to 15% of all patients with RA
(Danning et al. 1998) and is typically seen in patients with long-
standing, severe disease and high titres of rheumatoid factor. (b)
Cutaneous leukocytoclastic vasculitis is common, but necrotiz-
ing vasculitis of larger vessels (Figure 9.31), including coronary or
mesenteric arteries, and a form of GCA involving the aorta have
all been reported (Bahlas et al. 1998; Hall et al. 1983); however,
some cases may represent the coexistence of GCA in a patient
with RA (Hall et  al. 1983; Bahlas et  al. 1998). The medium and
small-vessel vasculitis in RA is occasionally granulomatous (Lie
1989). Capillaritis may be seen in the lung (Figure 9.12), although
it is relatively rare.
Vasculitis is reported in about one-third of patients with SLE
(Drenkard et  al. 1997). Cutaneous leukocytoclastic vasculitis
is by far the most common presentation. Involvement of larger
vessels in patterns similar to those in RA occurs but is much
less frequent. More often SLE is associated with deposition of
fibrinoid-like material, that is, immune complexes, in vessels, but
without an inflammatory response (Grishman and Spiera 1991).
Fig. 9.30 Microscopic polyangiitis. Interstitial fibrosis secondary to chronic
Pulmonary capillaritis with haemorrhage (Figure 9.32), as noted pulmonary haemorrhage in a patient with microscopic polyangiitis. (a) Low-power
above, carries a high mortality. Deposition of immune complexes view showing diffuse interstitial fibrosis and masses of haemosiderin-laden
in the lung can be shown by immunofluorescence or electron macrophages in the airspaces. (b) High-power view showing haemosiderin,
microscopy. indicative of old haemorrhage, in the interstitium, and encrustation of the vessel
elastica by iron/calcium salts, a common finding in chronic haemorrhage.

Fig. 9.31 Rheumatoid vasculitis. Necrotizing vasculitis involving an artery in


the peripancreatic fat. By itself this morphology could be seen in any form
Fig. 9.29 Microscopic polyangiitis. Capillaritis in lung. This is the most common of small-vessel or medium-vessel vasculitis and further clinical/ laboratory
pattern of lung involvement in microscopic polyangiitis. information is required for diagnosis.
114 SECTION 2 basic science

(a)
less common and the same is true of pulmonary capillaritis (1.6% of
patients in the large series of cases reported by Nadrous et al. 2004).
Cryoglobulinaemia
Cryoglobulinaemia is most often seen in patients with hepatitis C
virus infection, and is characterized by the presence of circulating
cryoglobulins that deposit in tissues. Cryoglobulinaemic vasculitis
typically involves the kidney and skin (as leukocytoclastic vascu-
litis) but may involve many other organs including the peripheral
nervous system and the lung (Figure 9.33). Eosinophilic deposits
of cryoglobulins can be seen in vessel walls and appear to evoke a
neutrophil response; the deposits are most often IgM. In the kidney
the typical lesion is glomerulonephritis with mesangial prolifera-
tion and cryoglobulin deposition. Larger vessels are occasionally
involved (Cacoub et al. 2002).
Drug-induced vasculitis
Drugs are a very common cause of small-vessel vasculitis, and the
typical morphological appearance is that of cutaneous leukocyto-
(b) clastic vasculitis (Figure 9.11). Indeed it has been suggested that
more than 95% of vasculitic skin lesions are drug-induced (Jennette
and Falk 1997). Nevertheless, a wide variety of other organs as well
as larger vessels may be involved (ten Holder et al. 2002). There is
no morphological specificity to drug-induced vasculitis, thus a his-
tory/ timing of drug use is crucial to the diagnosis.
Drug-induced vasculitis may be associated with lupus-like serol-
ogy (ANA, anti-dsDNA antibody), positive ANCA, or both (Choi
et al. 2000). Of interest, the titre of myeloperoxidase ANCA is often
extremely high in drug-associated vasculitis, much higher than
in typical ANCA-positive vasculitides, and ANCA with unusual
specificities including elastase and lactoferrin may also be found
(Choi et al. 2000). Detailed listings of drugs known to induce vas-
culitis have been published (Doyle and Cuellar 2003; ten Holder
et al. 2002).
Behçet’s syndrome
Fig. 9.32 Pulmonary capillaritis with haemorrhage in lupus. (a) Medium power BS was initially defined by the presence of oral and genital ulcers
view showing acute haemorrhage (red blood cells) and haemosiderin-laden and uveitis. In fact, BS is a systemic disease associated with:  a
macrophages; the latter are a useful finding that indicate the presence of real
(as opposed to surgical) haemorrhage. (b) High-power view of another area
showing typical capillaritis. This biopsy also demonstrated immune complex
deposits by immunofluorescence and electron microscopy.

Vasculitis in various sizes of vessels may also be seen in Sjögren’s


syndrome, systemic sclerosis, and relapsing polychondritis (see
Chapter  33). Because the morphology is relatively non-specific,
serological testing is crucial to arriving at the correct diagnosis. It
should be noted that patients with autoimmune disease may have a
positive fluorescent ANCA, but by ELISA the ANCA are atypical;
that is, directed against an antigen other than myeloperoxidase or
proteinase 3. Atypical ANCA against lactoferrin are probably the
most common (see Chapter 6).
Iga vasculitis (formerly known as henoch−schönlein purpura)
IgA vasculitis (IgAV) is a small-vessel vasculitis characterized by
deposition of IgA. The vasculitis is typically a combination of glo-
merulonephritis, which may take the form of mesangial prolifera-
tion or crescentic disease, and cutaneous leukocytoclastic vasculitis Fig. 9.33 Cryoglobulinaemia. Largely non-destructive vasculitis involving two
(White 1991). IgA can be demonstrated by immunofluorescence small vessels in the lung; there is capillaritis (arrows) as well. This combination of
examination. Larger-vessel necrotizing arteritis also occurs but is sizes of vessels involved is sometimes seen in small-vessel vasculitis.
CHAPTER 9 pathological features of vasculitis 115

variety of skin lesions; arthritis; gastrointestinal abnormalities Pathologically, Buerger’s disease affects medium and small-sized
including changes that can resemble those of chronic inflamma- arteries and veins in the extremities (characteristically the posterior
tory bowel disease; CNS involvement; and pulmonary and cardiac and anterior tibial, radial, ulnar, plantar, palmar, and digital vessels),
disease. Vascular abnormalities typically appear late in the course, and, rarely, visceral or cerebral sites (Lie 1988a; Lie 1989; Lie 1998).
frequently 5 to 10 years after initial diagnosis (see Chapter 34). The lesions start as acute inflammatory infiltrates in the vessel walls
BS is included among small-vessel vasculitides because it can and this process rapidly leads to the formation of morphologically
involve arterioles and venules and produce leukocytoclastic vas- unusual thrombi that are very cellular and may contain small collec-
culitis; however, there is a remarkably wide spectrum of vascular tions of neutrophils resembling microabscesses (Figure 9.35). Over
disease in BS (Koc et al. 1992). Both large arteries and veins are fre- time the neutrophils are replaced, in both the vessel wall and the
quently involved, and venous thrombosis is, overall, the most com- thrombi, by chronic inflammatory cells, and sometimes individual
mon vascular abnormality. In the large series reported by Koc et al. giant cells or granulomas are found in the thrombi (Figure 9.35).
(1992), significant venous thromboses were seen in 22% of cases The process is focal and segmental, and is different from most forms
and arterial disease in 12%. Some patients have antiphospholipid of vasculitis in that there is no necrosis of the vessel walls, so that
antibodies, although there is dispute about whether such antibodies the underlying structure remains intact (Figure 9.35). The thrombi
are the cause of the thromboses (Fessler 1997). eventually undergo recanalization, and specific recognition of very
BS may be associated with aortitis or inflammation of the pul- old lesions as Buerger’s disease versus thrombosis on some other
monary arteries, and with necrotizing medium-sized vessel dis- basis is problematic; however, purely arteriosclerotic vessels usually
ease (Lie 1988b; Lie 1989; Lakhanpal et al. 1985). Aneurysms are do not become thrombosed (Lie 1988a).
frequent sequelae in the aorta, and pulmonary artery aneurysms
with thromboses or rupture are the most common form of pulmo- Infectious vasculitis
nary disease (Uzun 2005) (Figure 9.34). Microscopically there is an Many types of infection can be associated with vasculitis (reviewed
inflammatory infiltrate, most often of chronic inflammatory cells, in Lie 1991b; Lie 1996b; Oyoo and Espinoza 2005). Morphologically
with destruction of the elastica. The pulmonary changes are identi- a variety of processes are seen, including direct or haematog-
cal to those found in Hughes–Stovin syndrome (pulmonary artery enous spread of the organism into the vessel wall, leading in some
aneurysms and systemic thromboses), and some authors believe instances to inflammatory destruction, often accompanied by dila-
that the latter is in fact a form of BS (Erkan et al. 2004; Slavin and tation and spread into surrounding tissue (so-called mycotic aneu-
DeGroot 1981; Durieux et al. 1981). rysm); over-running of vessels by an infection-evoked inflammatory
response; and granulomatous reactions, typical of mycobacterial and
Other forms of vasculitis fungal diseases. In these settings the diagnosis is usually obvious
from the accompanying pathological processes. In some instances,
Thromboangiitis obliterans (buerger’s disease) indirect effects produce the morphological picture of true fibrinoid
Thromboangiitis obliterans (TAO) is a form of non-necrotizing necrosis or leukocytoclastic vasculitis, for example in hepatitis B
vasculitis that affects primarily the upper and lower extremities infections where the immune response to the virus leads to immune
(see Chapter  37). The disease is seen in cigarette smokers, pre- complex formation, and in hepatitis C infection resulting in cryo-
dominantly young male smokers, and is characterized clinically by globulinaemia. HIV infection has been reported to produce virtu-
intermittent claudication of the arms and legs, migratory throm- ally every pattern of vasculitis described in this chapter (reviewed
bophlebitis, and, often, Raynaud’s phenomenon. In severe cases, in Patel et al. 2011), although some of these patients may really have
Buerger’s disease leads to ulcers and gangrene requiring amputa- had the idiopathic form of the vasculitis in question and incidental
tion of digits, hands, feet, or distal extremities. HIV positivity.

Apparently isolated vasculitis


Vasculitis is occasionally seen in small arteries, arterioles, and ven-
ules in pathology specimens removed for some other reason. The
most common sites are the appendix, gallbladder, female genital
tract, breast, testis, and bladder. The process usually appears as a
necrotizing lesion with fibrinoid necrosis (Figure 9.36), but occa-
sionally is granulomatous. How often these findings represent a true
systemic vasculitis as opposed to a localized inflammatory process
of no significance is disputed. Lie (1989) reviewed the literature and
concluded that about one-third of cases were actually part of sys-
temic disease. Hernández-Rodriguez et al. (2012a) summarized the
literature on patients presenting with apparently isolated testicular
vasculitis and found that 51% of 72 patients really had isolated vas-
culitis and 49% systemic vasculitis, and that most of the patients
with systemic vasculitis had constitutional/musculoskeletal symp-
toms at the time of presentation. However, Ganesan et al. (2000)
found evidence of systemic vasculitis in only four of 46 patients
Fig. 9.34 Behçet’s syndrome. Low power view of an aneurysmally dilated and with isolated vasculitis in the female genital tract and Brimo et al.
thrombosed pulmonary artery branch. Note the extreme thinning of the wall in (2011) reported systemic vasculitis in four of 19 cases presenting as
some areas (arrow); such vessels are prone to rupture. testicular vasculitis. Hernández-Rodriguez and Hoffman (2012b)
116 SECTION 2 basic science

(a) (b)

(c)

Fig. 9.35 Thromboangiitis obliterans (Buerger’s disease). Vasculitis involving a branch of the tibial artery. (a) Low-power view showing a cellular thrombus and
inflammatory cells in the vessel wall. (b) Elastic stain shows that, as opposed to most forms of vasculitis, the vessel wall is not destroyed in Buerger’s disease.
(c) High-power view of the centre of the thrombus shown in (a). Note the marked cellularity, presence of giant cells (arrows), and collections of neutrophils (circled).
These morphological features are typical of Buerger’s disease and are not seen in ordinary thrombi.

propose that isolated vasculitis should not be diagnosed as such


until 6 months have passed with no evidence of systemic vasculitis.
I believe that, absent evidence of systemic disease, it is probably
best for the pathologist to be very cautious about the significance
of isolated vasculitis, and to suggest workup for systemic disease if
clinically suspicious, rather than to make an outright pathological
diagnosis of vasculitis.

Selection of sites for biopsy


Selection of the proper site to biopsy greatly improves the diagnos-
tic yield in vasculitis. The older notion that blind biopsies should
be performed from muscle or nerve, regardless of symptoms,
has clearly proven to be wrong (see Chapter 20). Lie (1989) cites
overall true-positive rates of 35% and false-negative rates of 65%
in cases of PAN for such biopsies. A general rule of thumb is to
biopsy clinically affected sites such as kidney, skin, lung, and areas
Fig. 9.36 Isolated vasculitis. Necrotizing vasculitis in a small periappendiceal vessel of muscle tenderness, or to biopsy electrophysiologically affected
in a case of acute appendicitis. In the majority of cases, this type of isolated vasculitis muscle or nerve. The problems of sampling and focal lesions have
associated with inflammatory reactions is not indicative of systemic vasculitis. been discussed above for temporal artery biopsies, and the same
CHAPTER 9 pathological features of vasculitis 117

considerations hold true for other types of vasculitis; the specimen Devaney, K.O., Travis, W.D., Hoffman, G., Leavitt, R., Lebovics, R., and Fauci,
should be submitted for processing so as to maximize the amount A.S. (1990). Interpretation of head and neck biopsies in Wegener’s gran-
of tissue examined and multiple levels obtained. Note that in the ulomatosis. American Journal of Surgical Pathology, 14, 555–64.
Doyle, M.K. and Cuellar, M.L. (2003). Drug-induced vasculitis. Expert
lung, however, large sections appear, empirically, much more useful
Opinion in Drug Safety, 2, 401–9.
for detecting capillaritis than are small sections.
Drenkard, C., Villa, A.R., Reyes, E., Abello, M., and Alarcon-Segovia, D.
(1997). Vasculitis in systemic lupus erythematosus. Lupus, 6, 235–42.
Mimics of vasculitis Durieux, P., Bletry, O., Huchon, G., Wechsler, B., Chretien, J., and Godeau, P.
(1981). Multiple pulmonary arterial aneurysms in Behçet’s disease and
A number of other pathological processes can mimic vasculitis Hughes-Stovin syndrome. American Journal of Medicine, 71, 736–41.
microscopically (see Chapter 44). Most notably these include: pul- Erkan, D., Yazici, Y., Sanders, A., Trost, D., and Yazici, H. (2004). Is
monary hypertension and malignant systemic hypertension, both Hughes-Stovin syndrome Behçet’s disease? Clinical and Experimental
of which can produce fibrinoid necrosis in small arteries; any type Rheumatology, 22 (Suppl. 34), S64–8.
of inflammatory reaction which may overrun vessels; and some Eschun, G.M., Mink, S.N., and Sharma, S. (2003). Pulmonary interstitial
forms of lymphoma, particularly peripheral T-cell lymphomas and fibrosis as a presenting manifestation in perinuclear antineutrophilic
cytoplasmic antibody microscopic polyangiitis. Chest, 123, 297–301.
lymphomatoid granulomatosis, where the malignant cells infiltrate
Espinosa, G., Cervera, R., Font, J., and Asherson, R.A. (2002). The lung in
vascular walls. The antiphospholipid antibody syndrome is com- the antiphospholipid syndrome. Annals of the Rheumatic Diseases,
monly associated with thromboses but sometimes also associated 61, 195–8.
with a morphological picture of vasculitis such as pulmonary capil- Falk, R.J., Nachman, P.H., Hogan, S.L., and Jennette, J.C. (2000). ANCA glo-
laritis (Espinosa et al. 2002). merulonephritis and vasculitis: a Chapel Hill perspective. Seminars in
Nephrology, 20, 233–23.
References Fessler, B.J. (1997). Thrombotic syndromes and autoimmune diseases.
Rheumatic Disease Clinics of North America, 23, 461–79.
Achkar, A.A., Lie, J.T., Hunder, G.G., O’Fallon, W.M., and Gabriel, S.E.
(1994). How does previous corticosteroid treatment affect the biopsy Finkielman, J.D., Lee, A.S., Hummel, A.M., Viss, M.A., Jacob, G.L.,
findings in giant cell (temporal) arteritis? Annals of Internal Medicine, Homburger, H.A., Peikert, T., Hoffman, G.S., Merkel, P.A., Spiera, R.,
120, 987–92. St Clair, E.W., Davis, J.C. Jr, McCune, W.J., Tibbs, A.K., Ytterberg, S.R.,
Stone, J.H., and Specks, U. (2007). ANCA are detectable in nearly all
Ashton-Key, M. and Gallagher, P.J. (1991). Surgical pathology of cranial arte-
patients with active severe Wegener’s granulomatosis. American Journal
ritis and polymyalgia rheumatica. Baillieres Clinical Rheumatology, 5,
of Medicine, 120, 643–9.
387–404.
Ganesan, R., Ferryman, S.R., Meier, L., and Rollason, T.P. (2000). Vasculitis of
Bahlas, S., Ramos-Remus, C., and Davis, P. (1998). Clinical outcome of 149
the female genital tract with clinicopathologic correlation: a study of 46
patients with polymyalgia rheumatica and giant cell arteritis. Journal of
cases with follow-up. International Journal of Gynecological Pathology,
Rheumatology, 25, 99–104.
19, 258–65.
Bhanji, A. and, Karim, M. (2010). Pulmonary fibrosis-an uncommon mani-
Gaudin, P.B., Askin, F.B., Falk, R.J., and Jennette, J.C. (1995). The patho-
festation of anti-myeloperoxidase-positive systemic vasculitis? NDT
logic spectrum of pulmonary lesions in patients with anti-neutrophil
Plus, 3, 351–3.
cytoplasmic autoantibodies specific for anti-proteinase 3 and
Brimo, F., Lachapelle, J., and Epstein, J.I. (2011). Testicular vasculitis: a series
anti-myeloperoxidase. American Journal of Clinical Pathology, 104, 7–16.
of 19 cases. Urology, 77, 1043–8
Godman, G.C. and Churg, J. (1954). Wegener’s granulomatosis: pathology
Burns, A. (1998). Pulmonary vasculitis. Thorax, 53, 220–7.
and review of the literature. Archives of Pathology, 58, 533–53.
Cacoub, P., Costedoat-Chalumeau, N., Lidove, O., and Alric, L. (2002).
Grishman, E. and Spiera, A. (1991). Vasculitis in connective tissue diseases,
Cryoglobulinemia vasculitis. Current Opinion in Rheumatology,
including hypocomplementemic vasculitis. In Systemic vasculitides (ed.
14, 29–35.
A. Churg and J. Churg), pp. 273–92. Igaku-Shoin Medical Publishers,
Carrington, C.B. and Liebow, A. (1966). Limited forms of angiitis and New York.
granulomatosis of Wegener’s type. American Journal of Medicine, 41,
Guillevin, L., Cohen, P., Gayraud, M., Lhote, F., Jarrousse, B., and Casassus,
497–527.
P. (1999a). Churg-Strauss syndrome. Clinical study and long-term
Choi, H.K., Merkel, P.A., Walker, A.M., and Niles, J.L. (2000). Drug-associated follow-up of 96 patients. Medicine (Baltimore), 78, 26–37.
antineutrophil cytoplasmic antibody-positive vasculitis:  prevalence
Guillevin, L., Durand-Gasselin, B., Cevallos, R., Gayraud, M., Lhote, F.,
among patients with high titers of antimyeloperoxidase antibodies.
Callard, P., Amouroux, J., Casassus, P., and Jarrousse, B. (1999b).
Arthritis and Rheumatism, 43, 405–13.
Microscopic polyangiitis: clinical and laboratory findings in eighty-five
Churg, A. (2001). Recent advances in the diagnosis of Churg-Strauss syn- patients. Arthritis and Rheumatism, 42, 421–30.
drome. Modern Pathology, 14, 1284–93.
Hall, S., Ginsburg, W.W., Vollertsen, R.S., and Hunder, G.G. (1983). The
Churg, A. (2005). Churg-Strauss syndrome, microscopic polyangiitis, and coexistence of rheumatoid arthritis and giant cell arteritis. Journal of
other forms of pulmonary vasculitis. In Thurlbeck’s pathology of the lung, Rheumatology, 10, 995–7.
3rd edn, (ed. A. Churg, J. Myers, H. Tazelaar, and J.L. Wright), pp. 391–
Heggtveit, H.A., Hennigar, G.R., and Morrione, T.G. (1963). Panaortitis.
412. Thieme Medical Publishers, New York.
American Journal of Pathology, 42, 151–72.
Churg, J. and Churg, A. (1989). Primary and secondary vasculitis. Modern
Hernández-Rodríguez, J. and Hoffman, G.S. (2012b). Updating single-organ
Pathology, 2, 144–60.
vasculitis. Current Opinion in Rheumatology, 24, 38–45.
Churg, J. and Strauss, L. (1951). Allergic granulomatosis, allergic angiitis, and
Hernández-Rodríguez, J., Tan, C.D., Koening, C.L., Khasnis, A., Rodríguez,
periarteritis nodosa. American Journal of Pathology, 27, 277–301.
E.R., and Hoffman, G.S. (2012a). Testicular vasculitis: findings differen-
Cordier, J.F. and Cottin, V. (2011). Alveolar hemorrhage in vasculitis: pri- tiating isolated disease from systemic disease in 72 patients. Medicine
mary and secondary. Seminars in Respiratory and Critical Care Medicine, (Baltimore), 91, 75–85.
32, 310–21.
Homma, S., Matsushita, H., and Nakata, K. (2004). Pulmonary fibrosis in
Danning, C.L., Illei, G.G., and Boumpas, D.T. (1998). Vasculitis associated myeloperoxidase antineutrophil cytoplasmic antibody-associated vas-
with primary rheumatologic diseases. Current Opinion in Rheumatology, culitides. Respirology, 9, 190–6.
10, 58–65.
118 SECTION 2 basic science

Jacobs, M.R. and Allen, N.B. (1991). Giant cell arteritis. In Systemic vascu- Lie, J.T. (1998). Visceral intestinal Buerger’s disease. International Journal of
litides (ed. A. Churg and J. Churg), pp. 143–55. Igaku-Shoin Medical Cardiology, 66 (Suppl. 1), S249–56.
Publishers, New York. Miller, D.V. and Maleszewski, J.J. (2011). The pathology of large-vessel vascu-
Jennette, J.C. and Falk, R.J. (1997). Small-vessel vasculitis. New England litides. Clinical and Experimental Rheumatology, 29 (1 Suppl. 64), S92–8.
Journal of Medicine, 337, 1512–23. Myers, J. and Katzenstein, A.L.A. (1986). Microangiitis in lupus-induced pul-
Jennette, J.C. and Falk, R.J. (2000a). Do vasculitis categorization systems monary hemorrhage. American Journal of Clinical Pathology, 85, 552–6.
really matter? Current Rheumatology Reports, 2, 430–8. Nada, A.K., Torres, V.E., Ryu, J.H., Lie, J.T., and Holley, K.E. (1990). Pulmonary
Jennette, J.C. and Falk, R.J. (2000b). Overview of the nomenclature and fibrosis as an unusual clinical manifestation of a pulmonary-renal vascu-
diagnostic categorization of vasculitis. Wiener Kliniche Wochenschrift, litis in elderly patients. Mayo Clinic Proceedings, 65, 847–56.
112, 650–5. Nadrous, H.F., Yu, A.C., Specks, U., and Ryu, J.H. (2004). Pulmonary involve-
Jennette, J.C., Falk, R.J., Andrassy, K., Bacon, P.A., Churg, J., Gross, W.L., et al. ment in Henoch-Schonlein purpura. Mayo Clinic Proceedings, 79, 1151–7.
(1994). Nomenclature of systemic vasculitides. Proposal of an interna- Naoe, S., Takahashi, K., Masuda, H., and Tanaka, N. (1991). Kawasaki dis-
tional consensus conference. Arthritis and Rheumatism, 37, 187–92. ease. With particular emphasis on arterial lesions. Acta Pathologica
Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F., Japonica, 41, 785–97.
Flores-Suarez, L.F., Gross, W.L., Guillevin, L., Hagen, E.C., Hoffman, Nordborg, E. and Nordborg, C. (1995). The influence of sectional interval on
G.S., Jayne, D.R., Kallenberg, C.G.M., Lamprecht, P., Langford, C.A., the reliability of temporal arterial biopsies in polymyalgia rheumatica.
Luqmani, R.A., Mahr, A.D., Matteson, E.L., Merkel, P.A., Ozen, S., Pusey, Clinical Rheumatology, 14, 330–4.
C.D., Rasmussen, N., Rees, A.J., Scott, D.G.I., Specks, U., Stone, J.H., Oyoo, O. and Espinoza, L.R. (2005). Infection-related Vasculitis. Current
Takahashi, K., and Watts, R.A. (2013). Revised International Chapel Hill Rheumatology Reports, 7, 281–7.
Consensus Conference Nomenclature of the Vasculitides. Arthritis and Patel, N., Patel, N., Khan, T., Patel, N., and Espinoza, L.R. (2011). HIV
Rheumatism, 65, 1–11. infection and clinical spectrum of associated vasculitides. Current
Jennette, J.C., Thomas, D.B., and Falk, R.J. (2001). Microscopic polyangii- Rheumatology Reports, 13, 506–12.
tis (microscopic polyarteritis). Seminars in Diagnostic Pathology, Petursdottir, V., Nordborg, E., and Nordborg, C. (1996). Atrophy of the aortic
18, 3–13. media in giant cell arteritis. APMIS, 104, 191–8.
Kinare, S.G. (1975). Nonspecific aortitis (Takayasu’s disease). Autopsy study Ponge, T., Barrier, J.H., Grolleau, J.Y., Ponge, A., Vlasak, A.M., and Cottin,
of 35 cases. Pathologia et Microbiologia (Basel), 43, 134–9. S. (1988). The efficacy of selective unilateral temporal artery biopsy
Koc, Y., Gullu, I., Akpek, G., Akpolat, T., Kansu, E., Kiraz, S., Batman, F., versus bilateral biopsies for diagnosis of giant cell arteritis. Journal of
Kansu, T., Balkanci, F., and Akkaya, S. (1992). Vascular involvement in Rheumatology, 15, 997–1000.
Behçet’s disease. Journal of Rheumatology, 19, 402–10. Savage, C.O., Winearls, C.G., Evans, D.J., Rees, A.J., and Lockwood, C.M.
Lakhanpal, S., Tani, K., Lie, J.T., Katoh, K., Ishigatsubo, Y., and Ohokubo, T. (1985). Microscopic polyarteritis: presentation, pathology and progno-
(1985). Pathologic features of Behçet’s syndrome: a review of Japanese sis. Quarterly Journal of Medicine, 56, 467–83.
autopsy registry data. Human Pathology, 16, 790–5. Slavin, R.E. and de Groot, W.J. (1981). Pathology of the lung in Behçet’s
Landing, B.H. and Larson, E.J. (1987). Pathological features of Kawasaki disease. Case report and review of the literature. American Journal of
disease (mucocutaneous lymph node syndrome). American Journal of Surgical Pathology, 5, 779–88.
Cardiovascular Pathology, 1, 218–29. Swerlick, R.A. and Lawley, T.J. (1991). Small-vessel vasculitis and cutane-
Lanham, J.G., Elkon, K.B., Pusey, C.D., and Hughes, G.R. (1984). Systemic ous vasculitis. In Systemic vasculitides (ed. A. Churg and J. Churg), pp.
vasculitis with asthma and eosinophilia:  a clinical approach to the 193–201. Igaku-Shoin Medical Publishers, New York.
Churg-Strauss syndrome. Medicine (Baltimore), 63, 65–81. ten Holder, S.M., Joy, M.S., and Falk, R.J. (2002). Cutaneous and systemic man-
Lauque, D., Cadranel, J., Lazor, R., Pourrat, J., Ronco, P., Guillevin, L., and Cordier, ifestations of drug-induced vasculitis. Annals of Pharmacotherapeutics,
J.F. (2000). Microscopic polyangiitis with alveolar hemorrhage. A study of 36, 130–47.
29 cases and review of the literature. Medicine (Baltimore), 79, 222–33. Travis, W.D., Carpenter, H.A., and Lie, J.T. (1987). Diffuse pulmonary hem-
Lie, J.T. (1988a). Thromboangiitis obliterans (Buerger’s disease) revisited. orrhage. An uncommon manifestation of Wegener’s granulomatosis.
Pathology Annual, 23, 257–91. American Journal of Surgical Pathology, 11, 702–8.
Lie, J.T. (1988b). Cardiac and pulmonary manifestations of Behcet syndrome. Uzun, O., Akpolat, T., and Erkan, L. (2005). Pulmonary vasculitis in Behçet’s
Pathology Research and Practice, 183, 347–55. disease: a cumulative analysis. Chest, 127, 2243–53.
Lie, J.T. (1989). Systemic and isolated vasculitis. A rational approach to clas- Walz LeBlanc, B.A., Keystone, E.C., Feltis, J.T., Geddie, W.R., and Lie, J.T.
sification and pathologic diagnosis. Pathology Annual, 24, 25–114. (1994). Polyarteritis nodosa clinically masquerading as temporal arteri-
Lie, J.T. (1991a). Takayasu’s arteritis. In Systemic vasculitides (ed. A. tis with lymphadenopathy. Journal of Rheumatology, 21, 949–52.
Churg and J. Churg), pp. 159–80. Igaku-Shoin Medical Publishers, White, R.H.R. (1991). Henoch-Schonlein purpura. In Systemic vascu-
New York. litides (ed. A. Churg and J. Churg), pp. 203–18. Igaku-Shoin Medical
Lie, J.T. (1991b). Infection-related vasculitis. In Systemic vasculitides (ed. Publishers, New York.
A. Churg and J. Churg), pp. 243–56. Igaku-Shoin Medical Publishers, Wilkinson, I.M. and Russell, R.W. (1972). Arteries of the head and neck in
New York. giant cell arteritis. A pathological study to show the pattern of arterial
Lie, J.T. (1995). Aortic and extracranial large vessel giant cell arteritis:  a involvement. Archives of Neurology, 27, 378–91.
review of 72 cases with histopathologic documentation. Seminars in Zamora, M.R., Warner, M.L., Tuder, R., and Schwarz, M.I. (1997). Diffuse alve-
Arthritis and Rheumatology, 24, 422–31. olar hemorrhage and systemic lupus erythematosus. Clinical presentation,
Lie, J.T. (1996a). Temporal artery biopsy diagnosis of giant cell arteritis: lessons histology, survival, and outcome. Medicine (Baltimore), 76, 192–202.
from 1109 biopsies. Anatomic Pathology (Chicago, Ill. annual), 1, 69–97. Zeek, P.M., Smith, C.C., and Weeter, J.C. (1948). Studies on periarteritis nodu-
Lie, J.T. (1996b). Vasculitis associated with infectious agents. Current Opinion osa: the differentiation between the vascular lesions of periarteritis nodosa
in Rheumatology, 8, 26–9. and of hypersensitivity. American Journal of Pathology, 24, 889–917.
SECTION 3

Clinical manifestations
common to vasculitis
CHAPTER 10

Cutaneous manifestations
of vasculitis
Nicole M. Fett and Victoria P. Werth

Introduction Description of the eight most


Vasculitides, conditions manifesting with inflammation of blood common cutaneous morphologies
vessels, are a heterogeneous class of diseases ranging from benign, seen in vasculitis
self-limited inflammation of cutaneous vessels to life-threatening
multiorgan vessel inflammation. Despite this broad disease spec- Urticarial papules are pink raised lesions less than 1 cm in diameter.
trum, the cutaneous manifestations of vasculitis are limited to eight As the name implies, on first inspection they can be clinically indis-
classic presentations: urticarial papules, macular purpura, palpable tinguishable from urticaria (hives); however, each individual lesion
purpura, livedo reticularis, retiform purpura, subcutaneous nod- persists for longer than 24 hours and lesions resolve with hyperpig-
ules, necrosis, and ulcers. mentation (Figure 10.1). Macular purpura are flat, non-blanchable
To date, there are no reliable, validated, universally utilized diag- red to violaceous lesions caused by extravasation of red blood cells
nostic or classification criteria for vasculitis (Luqmani et al. 2011). from vessels damaged by inflammation (Figure  10.2). Macular
Currently, international efforts are underway to develop diagnos- purpura is non-blanchable because the red blood cells are out-
tic and classification criteria for vasculitis (Luqmani et al. 2011). side of vessel walls (blanchable lesions are caused by vasodilation
The Chapel Hill Consensus definitions provide a framework for where pressure is able to push red blood cells within the dilated
the division of vasculitis into accepted clinical subsets (Jennette vessels deeper into the vasculature). Palpable purpura are raised,
et al. 2013). A major strength of the Chapel Hill Consensus defini- non-blanchable red to violaceous lesions caused by a combination
tions is use of the size of the affected vessels to define clinical sub- of extravasated red blood cells and inflammation (Figure  10.3).
sets. The cutaneous manifestations of vasculitis are a reflection of Livedo reticularis is the visualization of the cutaneous vascular
the size of the involved vessel. Large-vessel vasculitides, giant cell arcades caused by red blood cell sludging in these small vessels.
arteritis and Takayasu’s arteritis, rarely have associated cutaneous In the setting of vasculitis, this sludging is most frequently caused
findings because these vessels are not found in the skin or sub- by inflammation of the medium vessels, which results in impaired
cutaneous tissues. Pure medium-vessel vasculitides, such as pol- blood flow to the more distal small vessels (Figure 10.4). Retiform
yarteritis nodosa, cutaneous polyarteritis nodosa, and Kawasaki’s purpura is necrosis of the skin overlying the cutaneous arcades due
disease, affect vessels with muscular walls that are found in the
deep dermis and subcutaneous tissues. Cutaneous features of
medium-vessel vasculitides include livedo reticularis, retiform
purpura, nodules, ulcers, and infarcts/ necrosis. Vasculitides that
involve both medium and small vessels include granulomatosis
with polyangiitis (GPA, previously Wegener’s granulomatosis),
eosinophilic granulomatous with polyangiitis (EGPA, previously
Churg–Strauss syndrome), microscopic polyangiitis, and cryoglo-
bulinaemic vasculitis. These vasculitides may present with features
of medium-vessel involvement and features classic for small-vessel
involvement (urticarial papules, macular purpura, and palpa-
ble purpura). Pure small-vessel vasculitides include cutaneous
small-vessel vasculitis (previously referred to as hypersensitivity
vasculitis, cutaneous leukocytoclastic angiitis, hypersensitivity
angiitis, and leukocytoclastic vasculitis), IgA vasculitis (previously
referred to as Henoch–Schönlein purpura (HSP)), erythema eleva-
tum diutinum (EED), and urticarial vasculitis. Pure small-vessel
vasculitides most commonly present with urticarial papules, mac- Fig. 10.1 Urticarial vasculitis presents with urticarial papules and plaques with
ular purpura, and palpable purpura. findings of leukocytoclastic vasculitis on histopathology.
122 SECTION 3 clinical manifestations common to vasculitis

Fig. 10.2 Macular purpura: erythematous and violaceous macules and patches.

to anoxia from hypoperfusion (Figure 10.4). In the setting of vascu-


litis, this hypoperfusion is most frequently caused by inflammation Fig. 10.4 Retiform (net-like) erythematous patch consistent with livedo reticularis.
and destruction of the medium vessels, again resulting in impaired
blood flow to the more distal small vessels. Subcutaneous nodules
are raised lesions, by convention greater than 1  cm in size, with
normal overlying epidermis (Figure 10.5). In the setting of vasculi-
tis, subcutaneous nodules are due to medium-vessel inflammation.
Necrosis (death of tissue resulting in black flat lesions) (Figure 10.6)
and ulcers (absence of epidermis and/or dermis) (Figure 10.7) may

Fig. 10.5 Retiform purpura.

Fig. 10.3 Palpable purpura: erythematous and violaceous papules. Palpable purpura


often presents on the lower legs and koebnerizes as seen here in the sock line. Fig. 10.6 Tender subcutaneous nodule.
CHAPTER 10 cutaneous manifestations of vasculitis 123

and a neutrophil-predominant perivascular inflammatory infiltrate


(the features necessary to make the diagnosis on histopathology)
(Carlson 2010). Lesions that are greater than 24 hours old begin
to have lymphocytes and macrophages replacing the neutrophils,
making diagnosis more difficult (Carlson 2010).
Direct immunofluorescence (DIF) is also an important aspect of
the work-up of cutaneous vasculitis. DIF is the only way to differen-
tiate cutaneous small-vessel vasculitis (CSVV) from IgA vasculitis.
In adults this distinction is very important, as adults who develop
IgA vasculitis are at high risk of renal manifestations that may have
a prolonged and relapsing course (Hung et al. 2009). Ideally, the
lesion biopsied for DIF should be less than 24 hours old (Carlson
2010). Direct immunofluorescence employs fluorescently labelled
antibodies directed against the pathogenic antibodies bound to ves-
sel walls. The pathogenic antibodies in antibody-mediated vasculitis
recruit inflammatory cells to the vessel wall, causing destruction of
the vessel wall and with it the pathogenic antibodies. False-negative
DIF results occur when older lesions are biopsied because the
pathogenic antibodies have already been destroyed by inflamma-
tory cells, and therefore the fluorescently labelled antibodies no
longer have a binding target. The DIF in CSVV will be positive for
complement binding (C3) and a mix of IgM, IgA, and IgG whereas
IgA vasculitis will be positive for IgA binding. Cryoglobulinaemic
vasculitis will reveal predominantly IgM staining.
Finally, the choice of biopsy technique should be guided by the
clinical exam. Physical exam findings consistent with small-vessel
involvement (urticarial papules, macular purpura, and palpable pur-
pura) should be biopsied with a mid-sized punch biopsy (4 to 6 mm)
to ensure sampling of the blood vessels into the mid-dermis. Physical
Fig. 10.7 Cutaneous ulceration. exam findings consistent with medium-vessel involvement (livedo
reticularis, retiform purpura, and subcutaneous nodules) should be
occur in the setting of small-vessel and/or medium-vessel vasculitis biopsied with a large punch biopsy (8 to 10 mm) or an incisional
and are due to hypoperfusion of the skin. biopsy to ensure inclusion of a medium vessel from the subcutaneous
The remainder of this chapter will be devoted to discussing the tissues. A histological diagnosis of a medium-vessel vasculitis cannot
most common cutaneous features seen in specific subtypes of vas- be made unless a medium vessel is included in the tissue submitted.
culitis. The epidemiology, pathogenesis, disease course, manage-
ment, and prognosis of each of these subtypes of vasculitis are Cutaneous small-vessel vasculitis
discussed in detail in subsequent chapters within this text. We refer
the reader to those chapters for more in-depth discussions of each Cutaneous small-vessel vasculitis (CSVV) is caused by
condition. antibody-mediated inflammation of the small vessels (capillaries,
venules, arterioles) in the skin. The diagnosis of CSVV requires
exclusion of predominant IgA vascular immunoglobulin deposits
Biopsies and direct immunofluorescence (which is consistent with IgA vasculitis) and exclusion of visceral
All cutaneous lesions worrisome for vasculitis should be biopsied disease (which is consistent with systemic vasculitis) (Chen and
to rule out vasculitis mimickers. Histopathological diagnosis of Carlson 2008). CSVV presents on the lower extremities as urti-
small-vessel vasculitis requires at least two of the three following carial papules, macular purpura, and palpable purpura (Blanco
criteria: an angiocentric and/or angioinvasive inflammatory infil- et al. 1998; Martinez-Taboada et al. 1997; Sais et al. 1998). Rarely,
trate, disruption of the vessel wall with inflammatory infiltrate or CSVV results in superficial ulcerations and haemorrhagic bullae
fibrin deposition (Carlson 2010). Red blood cell extravasation, on the distal legs and feet (Blanco et al. 1998; Martinez-Taboada
nuclear dust (leukocytoclasia), and endothelial cell swelling are also et  al. 1997; Sais et  al. 1998). CSVV often koebnerizes (develop-
commonly seen in vasculitis, but are not a part of the diagnostic ment of lesions at sites of traumatized uninvolved skin) in areas of
criteria. Histopathological diagnosis of medium-vessel vasculitis pressure (Figure 10.8). Most commonly, CSVV occurs as a single,
requires infiltration of the muscular wall with inflammatory cells acute self-limited reaction to a medication or infection and resolves
and fibrinoid necrosis (Carlson 2010). Because inflammatory cells with hyperpigmentation within 1 month (Chen and Carlson 2008).
do not normally migrate through the walls of medium vessels, see- Patients that develop CSVV as a reaction to a connective tissue dis-
ing inflammatory cells within the wall is always indicative of vascu- ease or a malignancy may develop chronic relapsing disease (Chen
litis. The timing of the biopsy is also important. Ideally, a lesion that and Carlson 2008).
is approximately 24 hours old should be chosen. A this time point On biopsy, CSVV presents as a small-vessel neutrophilic
there will be fibrin in the vessel wall, neutrophils in the vessel wall, vasculitis affecting the superficial dermal vessels, and direct
124 SECTION 3 clinical manifestations common to vasculitis

upper extremities, trunk, and face, but these areas of involvement


are less common than the lower extremities (Garcia-Porrua et al.
2002). Haemorrhagic bullae, ulcerations, and dermal necrosis with
scarring on the lower extremities have also been reported (Gedalia
2004). These manifestations are extremely rare in children and
more commonly seen in adults. Approximately 2 to 13% of males
with IgA vasculitis will present with acute pain and swelling of the
scrotum (‘acute scrotum’) (Hara et al. 2004; Trapani et al. 2005). The
palpable purpura usually resolves over an interval of 2 to 4 weeks
and may leave postinflammatory hyperpigmentation (brown mac-
ules). Greater than 90% of children will have one episode of vascu-
litis and no recurrences, whereas adults are more likely to have a
chronic relapsing and remitting course of both their skin and renal
involvement (Garcia-Porrua et al. 2002; Hung et al. 2009).
The histopathological examination findings of IgA vasculitis are
identical to CSVV; however, DIF reveals IgA deposits in dermal
capillaries and postcapillary venules in IgA vasculitis, thus differen-
tiating the two diseases (Gedalia 2004).

Erythema elevatum diutinum


Erythema elevatum diutinum (EED) is a reactive small-vessel vas-
culitis limited to the skin that has been reported in association with
rheumatic fever, rheumatoid arthritis, myelodysplastic syndrome,
relapsing polychondritis, monoclonal light chain disease (predomi-
nantly IgA), recurrent bacterial infections, HIV, cryoglobulinaemia,
Crohn’s disease and granulomatosis with polyangiitis (GPA, previ-
ously Wegener’s granulomatosis) (Yiannias et al. 1992; Chowdhury
et al. 2002; Wahl et al. 2005). Clinically, patients present with per-
sistent brown-red to purple papules, nodules, and plaques predom-
inantly on the extensor surfaces of the extremities and rarely on the
Fig. 10.8 Palpable purpura and cutaneous ulceration. buttocks, face, torso, and genitals (Yiannias et al. 1992; Chowdhury
et  al. 2002; Wahl et  al. 2005). Occasionally, bullous lesions and
ulcers can develop (Yiannias et al. 1992; Chowdhury et al. 2002;
immunofluorescence (DIF) reveals complement and immunoglob-
Wahl et al. 2005).
ulins within vessel walls (Chen and Carlson 2008).
Histopathologically, the early stage of the disease reveals a
neutrophil-rich leukocytoclastic vasculitis (Yiannias et  al. 1992).
IgA vasculitis (previously Biopsies of older lesions reveal a paucity of inflammatory cells, but
Henoch–Schönlein purpura) significant amounts of granulation tissue and fibrosis (Yiannias
et al. 1992). DIF studies have revealed perivascular deposits of IgA,
IgA vasculitis, previously referred to as Henoch–Schönlein purpura
IgG, IgM, fibrin, and complement (Chowdhury et al. 2002).
(HSP), anaphylactoid purpura or IgA immune complex vasculitis,
is recognized as the tetrad of palpable purpura, gastrointestinal vas-
culitis, arthritis, and renal manifestations (Henoch 1974). IgA vas- Urticarial vasculitis
culitis is a small-vessel vasculitis due to deposition of IgA immune Urticarial vasculitis is a small-vessel immune-complex-mediated
complexes in the skin, gastrointestinal system, and renal mesan- vasculitis. Patients with urticarial vasculitis present with urticarial
gium (Saulsbury 2007). lesions (pink papules and plaques). Urticarial vasculitis is differ-
One hundred per cent of patients with IgA vasculitis have cuta- entiated from urticaria by the behaviour of the cutaneous lesions
neous manifestations (Hung et al. 2009; Gedalia 2004; Trapani et al. and the histopathological findings. Urticaria are pruritic, transient
2005). The most common presentation is symmetrical urticarial (each lesion usually lasts less than 24 hours) lesions that resolve
papules on the lower extremities and buttocks that evolve into without sequelae. Lesions of urticarial vasculitis are more com-
palpable purpura within 1 to 2 days (Gedalia 2004). Petechiae and monly described as being painful or associated with a burning sen-
ecchymoses of the lower extremities are also common cutaneous sation, last longer than 24 hours, and heal with hyperpigmentation
features, reported to occur in 100% and 79% of patients, respec- (Chen and Carlson 2008; Brown and Carter 2007; Davis and Brewer
tively (Trapani et  al. 2005). Approximately 50% of patients will 2004). On histopathological exam urticaria reveal varying amounts
present with concomitant swelling of the ankles and feet, or more of dermal oedema, whereas urticarial vasculitis show changes of
rarely the hands and eyes (Trapani et  al. 2005). Adults with IgA leukocytoclastic vasculitis (Chen and Carlson 2008).
vasculitis have been reported to present with a higher frequency of Urticarial vasculitis is further subdivided into normocomple-
lower extremity oedema than children with IgA vasculitis (Hung mentaemic urticarial vasculitis, hypocomplementaemic urticar-
et al. 2009). Palpable purpura has also been reported to involve the ial vasculitis (anti-C1q vasculitis), and hypocomplementaemic
CHAPTER 10 cutaneous manifestations of vasculitis 125

urticarial vasculitis syndrome (Davis and Brewer 2004; Fiorentino patients presenting with skin manifestations (Monti et  al. 1995;
2003) with varying degrees of systemic involvement and chronicity. Cacoub and Saadoun 2008; Dupin et  al. 1995; Mironiuc et  al.
2008). Palpable purpura of the lower extremities is the most com-
mon cutaneous sign of CV (Monti et al. 1995; Cacoub and Saadoun
Acute haemorrhagic oedema of 2008; Dupin et al. 1995; Mironiuc et al. 2008), but it may also extend
infancy (finkelstein’s disease) to the trunk and upper extremities (Monti et al. 1995; Cacoub and
Acute haemorrhagic oedema of infancy (AHEI) is a benign Saadoun 2008; Dupin et al. 1995; Mironiuc et al. 2008). CV may
self-limited cutaneous small-vessel vasculitis that occurs in chil- also present with macular purpura, livedo reticularis, subcutane-
dren less than 2  years of age. AHEI has also been referred to as ous nodules, ulcers, and necrosis, usually on the lower extremi-
Finkelstein’s disease, medallion-like purpura, infantile postin- ties (Monti et  al. 1995; Cacoub and Saadoun 2008; Dupin et  al.
fectious iris-like purpura and oedema, cockade purpura, acute 1995). Ulcers most commonly occur in the supramalleolar areas
benign cutaneous leukocytoclastic vasculitis of young children, (Sansonno et al. 2007). Less-frequent cutaneous manifestations of
Seidlmayer’s disease, and Henoch–Schönlein syndrome of early CV include PAN-like lesions, livedo racemosa, splinter haemor-
childhood (Fiore et al. 2011; Dongre et al. 2012). Children most rhages, palmar erythema, and digital necrosis (Chen and Carlson
typically present with annular or targetoid palpable purpura and 2008; Mironiuc et  al. 2008). CV occurs most frequently in cold
ecchymoses on their cheeks, ears, and upper extremities. The weather, lasts for 1–2 weeks, and usually leaves behind hyperpig-
targetoid purpura and ecchymoses can be as large as 5  cm in mented plaques (Sansonno et al. 2007). Raynaud’s phenomenon is
diameter and are often tender to the touch (Emerich et al. 2011). sometimes the first manifestation at diagnosis and may affect the
Children also present with oedema of the scalp, face, hands, and distal part of the limbs, ears, and nose (Sansonno et al. 2007).
feet and occasionally the genital region (Emerich et al. 2011; Fiore Histopathologically, CV presents as a small-vessel neutrophilic
et al. 2011; Dongre et al. 2012). vasculitis of the superficial dermal and subcutaneous vessels
Usually the trunk is not involved. Less than 10% of children will (Cacoub and Saadoun 2008; Chen and Carlson 2008). In a minority
present with mucous membrane lesions (petechiae), vesicles, or of cases the medium vessels are also involved, resulting in a neutro-
bullae (Fiore et al. 2011). Despite the alarming presentation of the philic muscular-vessel vasculitis (Chen and Carlson 2008). DIF will
skin findings, the children are systemically well and recover with- reveal vascular immunoglobulins, predominantly IgM, and com-
out sequelae. Approximately 50% of children will present with an plement deposits (Chen and Carlson 2008).
accompanying low-grade fever (Fiore et  al. 2011). Spontaneous
resolution of the cutaneous findings and oedema occurs within 1 to Polyarteritis nodosa
3 weeks (Emerich et al. 2011; Fiore et al. 2011).
Histopathological exam reveals a small-vessel leukocytoclas- Polyarteritis nodosa (PAN) is a systemic, multiorgan, medium-vessel
tic vasculitis affecting the dermal vessels with fibroid necrosis, vasculitis (Morgan and Schwartz 2010). Large case series reveal
extravasation of red blood cells, and interstitial oedema (Dongre that 44 to 50% of patients with PAN have cutaneous manifestations
et al. 2012; Emerich et al. 2011). DIF results are equivalent to that (Kluger et al. 2008; Pagnoux et al. 2010), most commonly palpa-
of CSVV: deposition of complement 3 (C3), fibrinogen, IgM, IgA, ble purpura (19 to 22%), subcutaneous nodules (17 to 15%), livedo
and IgG (Emerich et al. 2011). reticularis (17%), and ulcers and necrosis (4%) (Kluger et al. 2008;
Pagnoux et al 2010). Subcutaneous nodules are usually 0.5 to 2 cm
in diameter and follow the course of superficial arteries along the
Cryoglobulinaemic vasculitis anterior low leg, knee, and dorsum of the foot (Fiorentino 2003).
Cryoglobulins are circulating immunoglobulins that complex with Rarely, digital infarction has been reported (Fiorentino 2003).
proteins or other immunoglobulins and precipitate upon cold Cutaneous manifestations occur most commonly on the lower
exposure. Three types of cryoglobulins are described: type I is com- extremities (Kluger et al. 2008; Pagnoux et al 2010). Approximately
posed of monoclonal IgM; type II is composed of monoclonal IgM one-third of patients will have cutaneous findings on disease pres-
and polyclonal IgG; and type III is composed of polyclonal IgM and entation and the vast majority will have vasculitis on skin biopsy
polyclonal IgG (Brouet et al. 1974). Types II and III are referred to (Kluger et  al. 2008). Patients with skin manifestation are more
as mixed cryoglobulinaemias because of the presence of both IgG likely to have arthralgias and ocular involvement than patients
and IgM. Type I cryoglobulinaemia occurs in the setting of hae- without skin manifestations (Kluger et al. 2008). Histopathological
matological malignancies and result in vascular obstruction rather assessment requires visualizing a medium vessel. Medium ves-
than frank vasculitis. Types II and III deposit within vessel walls, sels have muscular walls and are located in the deep dermis and
activate the complement cascade and recruit additional mediators subcutaneous tissues, therefore large punch biopsies (8 to 10 mm)
of inflammation resulting in cryoglobulinaemic vasculitis (Agnello or incisional biopsies are recommended to ensure inclusion of a
et al. 1992). medium vessel.
Approximately 40 to 60% of patients with hepatitis C virus On histopathology, PAN presents as a segmental neutrophilic
(HCV) produce circulating mixed cryoglobulins; however, overt vasculitis affecting medium-sized and small-sized arteries with
cryoglobulinaemic vasculitis (CV) develops in only 5 to 10% of muscular walls at branch points. This segmental involvement leads
these patients (Cacoub and Saadoun 2008; Mironiuc et  al. 2008; to the subcutaneous nodular lesions seen in PAN. Active vasculitis
Ramos-Casals et al. 2009). Less than 5% of all CV are HCV negative lesions are frequently associated with chronic reparative changes
(Sansonno et al. 2007). that show endarteritis obliterans. Aneurysms can form in PAN
CV causes inflammation of small and medium vessels. Skin is when the arterial wall is sufficiently weakened by the vasculitis
the most commonly involved organ, with reports of 70 to 90% of (Carlson and Chen 2007).
126 SECTION 3 clinical manifestations common to vasculitis

Cutaneous polyarteritis nodosa Kluger et al. 2008). The most frequent cutaneous manifestation (up
to 50%) is palpable purpura on the legs, followed by livedo reticu-
All patients with cutaneous polyarteritis nodosa (cPAN) have cuta- laris and nodules (Agard et al. 2003; Ahn et al. 2011; Villiger and
neous findings. Virtually 100% of these patients will present with Guillevin 2010).
painful subcutaneous nodules on their lower extremities (Ishiguro Non-descript macular erythema in the setting of livedo reticula-
and Kawashima 2010; Carlson and Chen 2007; Chan et al. 2009; ris has also been described (Seishima et al. 2004; Kawakami et al.
Chen 1989; Diaz-Perez et  al. 2007; Diaz-Perez and Winkelmann 2007; Ahn et al. 2011). Patients with skin manifestations are more
1974; Macarenco et al. 2013; Morgan and Schwartz 2010). These likely to have arthralgias, ocular involvement, and mononeuri-
nodules are generally the first manifestation of disease recognized tis multiplex than patients without skin manifestations (Kluger
by the patient. The nodules may occur singly or in crops of up et al. 2008).
to 100 and are commonly seen in various stages of development Histological examination shows a leukocytoclastic vasculitis; DIF
(Diaz-Perez et al. 2007). The nodules resolve with postinflamma- is usually negative (Dion et al. 2010; Nagai et al. 2009). Incidental
tory hyperpigmentation (brown macules) and/or diffuse areas of panniculitis can also be seen (Kawakami et al. 2007).
livedo reticularis (Diaz-Perez et al. 2007). Areas of residual livedo
reticularis are frequently involved in successive exacerbations,
even after prolonged periods of remission (Diaz-Perez et al. 2007; Granulomatosis with polyangiitis
Ishiguro and Kawashima 2010). The upper extremities, trunk, Granulomatosis with polyangiitis (GPA, previously Wegener’s
head and neck, and buttocks also can be involved (Diaz-Perez et al. granulomatosis), is a systemic ANCA-associated vasculitis with
2007; Ishiguro and Kawashima 2010; Chan et al. 2009; Morgan and frequent mucocutaneous involvement.
Schwartz 2010). Ulceration is also a frequent complication of cuta- Cutaneous signs of vasculitis are reported to be present at the time
neous PAN (Diaz-Perez et al. 2007). Ulcers occur from local tissue of diagnosis in 13 to 23% of patients diagnosed with GPA (Daoud
ischaemia due to arterial inflammation in areas with poor collat- et al. 1994; Hoffman et al. 1992; Frances et al. 1994), with upwards of
eral circulation (Diaz-Perez et al. 2007; Ishiguro and Kawashima 50% of patients developing cutaneous manifestations at some point
2010). Ulcers do not alter the benign prognosis of cutaneous PAN in their disease course (Hoffman et al. 1992). The most common
(Diaz-Perez et  al. 2007). Palpable purpura has been reported to cutaneous sign is palpable purpura on the lower extremities occur-
occur in 25 to 45% of patients with cPAN (Macarenco et al. 2013). ring in 47 to 74% of patients with cutaneous manifestations (Patten
Rarely, distal extremity gangrene and digital gangrene has been and Tomecki 1993; Daoud et  al. 1994; Frances et al. 1994; Chen
reported in patients with cPAN (Williams et al. 2012; Choi et al. and Carlson 2008; Hoffman et al. 1992). Signs of medium-vessel
2006; Stussi et al. 2001; Kumar et al. 1995; Chen 1989) and is more inflammation, subcutaneous nodules, ulcers, and digital infarcts,
likely to occur in children under the age of 10 years (Kumar et al. comprise 31% of reported cutaneous manifestations and occur
1995). The risk of progression from cPAN to PAN is low. In a case most commonly on the lower extremities (Chen and Carlson
series of 20 patients with cPAN followed for 16 years, two patients 2008). Patients with GPA may also present with flesh-coloured
developed systemic PAN (Chen 1989). papules on their extensor surfaces (previously referred to as rheu-
cPAN is a medium-vessel vasculitis requiring a large punch matoid papules or Churg–Strauss granulomas, and now referred
biopsy (8 to 10  mm) or an incisional biopsy to ensure sampling to as palisaded and granulomatous neutrophilic dermatosis), urti-
of a medium vessel from the deep dermis or subcutaneous tissues. caria, pyoderma-gangrenosum-like ulcers (often on the head and
Histologically, cPAN presents as a neutrophilic medium-sized vas- neck), and small (4 to 8 mm), punched out cutaneous ulcers (also
culitis usually at the dermal–subcutis junction where arteries bifur- common on the head and neck) (Patten and Tomecki 1993; Daoud
cate (Carlson and Chen 2007). cPAN is a segmental vasculitis and et al. 1994; Frances et al. 1994; Chen and Carlson 2008; Hoffman
therefore serial sections may be necessary to visualize the vasculitis et al. 1992). Cutaneous manifestations have been associated with a
(Carlson and Chen 2007). Older lesions of cPAN exhibit vessel wall higher frequency of articular and renal involvement and correlated
fibrosis, loss of the internal elastic lamina, and neovascularization with active systemic vasculitis (Frances et al. 1994).
of the adventitia (Carlson and Chen 2007). Incidental panniculitis Saddle nose deformity is a very rare complication in early dis-
is not an uncommon finding (Diaz-Perez and Winkelmann 1974; ease, reported to occur in only 2% of patients; however, up to 30%
Chen 1989). of patients with GPA will develop a saddle nose deformity later in
the course of their disease (Holle et al. 2010).
Microscopic polyangiitis Ocular manifestations visible on physical exam include scle-
Microscopic polyangiitis (MPA) is an ANCA-associated systemic ritis, episcleritis, and space-occupying orbital lesions. Scleritis,
vasculitis that commonly presents with cutaneous signs. Cutaneous episcleritis, and ulcerative keratitis are estimated to occur in 50 to
lesions are estimated to occur in 40 to 53% of patients with MPA 60% of patients with GPA (Aletaha et al. 2011; Pakrou et al. 2006;
(Dion et al. 2010; Nagai et al. 2009; Kawakami et al. 2007; Villiger Hoffman et al. 1992). Orbital space-occupying lesions are the pre-
and Guillevin 2010; Agard et al. 2003; Ahn et al. 2011; Kluger et al. senting sign of GPA in 8 to 16% of patients (Pakrou et al. 2006;
2008; Seishima et  al. 2004), and 14 to 33% will have cutaneous Hoffman et al. 1992). These space occupying lesions in the orbit
manifestations as their presenting sign (Agard et al. 2003; Kluger may lead to proptosis, pain, eyelid oedema, and limitation of ocular
et al. 2008). MPA affects venules, capillaries, arterioles, and small movements leading to diplopia, optic nerve ischaemia, and blind-
arteries (Kawakami et al. 2007), resulting in cutaneous manifesta- ness (Aletaha et al. 2011; Pakrou et al. 2006; Tarabishy et al. 2010).
tions of palpable purpura, nodules, livedo reticularis, and ulcers. Proptosis due to orbital involvement occurs in approximately 15
Bullae, acrocyanosis, and urticarial lesions are exceedingly rare to 20% of patients with GPA (Tarabishy et  al. 2010). One-third
(Kawakami et al. 2007; Villiger and Guillevin 2010; Ahn et al. 2011; of patients with chronic orbital involvement develop orbital
CHAPTER 10 cutaneous manifestations of vasculitis 127

contraction (enophthalmos) which may lead to optic nerve disease are reported to be present in about one-third of EGPA patients
and chronic orbital pain (Tarabishy et  al. 2010). Orbital fistulas (Dunogue et  al. 2011). The occurrence of these extensor surface
and infectious orbital abscess are additional complications of GPA granulomas was initially thought to be pathognomonic of EGPA,
orbital involvement (Tarabishy et  al. 2010). Eyelid involvement but it is now recognized that they occur in other vasculitides, auto-
may include dacryoadenitis, dacrocystitis, ptosis, lid granuloma, immune disorders, haematological and infectious processes, and in
chalazion, florid xanthalasma, and trichiasis (Tarabishy et al. 2010). patients without a recognized underlying disease.
Eyelid ulceration and fistula formation may also occur (Tarabishy Digital necrosis has also been reported (Waseda et al. 2011; Otani
et al. 2010). et al. 2003; Abbas et al. 2000; Inui et al. 2001).
Additionally, nasopharyngeal and oral manifestations are com- Skin biopsies in EGPA most commonly reveal leukocytoclastic
mon in patients with GPA. It is estimated that 10 to 62% of patients vasculitis with an accompanying eosinophilic infiltrate, eosinophil
with GPA develop nasopharyngeal or oral manifestations during infiltration without vasculitis, and extravascular granuloma (Bosco
the course of their disease (Tarabishy et al. 2010; Manchanda et al. et al. 2011; Dunogue et al. 2011; Davis et al. 1997).
2003; de Souza et al. 2010). Oral ulcers and gingivitis occur in 10
to 24% of patients (Tarabishy et al. 2010; Manchanda et al. 2003; Giant cell arteritis
de Souza et al. 2010). Gingival hyperplasia (‘strawberry gingival’)
Giant cell arteritis (GCA), also referred to as temporal arteritis, is a
may be the presenting sign in up to 7% of cases (Manchanda et al.
large-vessel vasculitis with rare cutaneous manifestations. The most
2003). Mucosal disease includes ulcers, gingival hyperplasia, pete-
common cutaneous manifestation is scalp tenderness, reported to
chiae, oroantral fistulas, osteonecrosis of the palate with ulceration,
occur in 23 to 52% of patients with GCA (Borchers and Gershwin
and labial mucosal nodules (Tarabishy et al. 2010; Manchanda et al.
2012). Accompanying retiform purpura on the frontal and temporal
2003; Patten and Tomecki 1993).
scalp has also been a reported as an early sign of GCA (Uffelmann
Careful examination of the skin, oro- and nasopharyngeal tis-
et al. 2012). Rarely, patients with GCA will develop scalp necro-
sues, and eyes may suggest the diagnosis of GPA. Skin biopsy rarely
sis, which can be bilateral (Currey 1997). Scalp necrosis uncom-
makes the diagnosis of GPA. The most common histological find-
monly occurs as a side effect of temporal artery biopsy; however,
ing in GPA is leukocytoclastic vasculitis, reported to occur in 31 to
the majority of cases developed scalp necrosis prior to temporal
80% of submitted biopsies (Daoud et al. 1994; Barksdale et al. 1995;
artery biopsy (Currey 1997). Scalp necrosis is often preceded by
Hu et al. 1977; Comfere et al. 2007). In contrast, granulomatous
tenderness of the scalp or a headache (Currey 1997; Baum et al.
vasculitis, the sine qua non of GPA, was seen in 0 to 11% of skin
1982). Between 1946 and 2007, 78 cases of scalp necrosis were pub-
biopsies (Barksdale et al. 1995; Daoud et al. 1994; Hu et al. 1977).
lished (Tsianakas et al. 2009; Baum et al. 1982). Patients with scalp
Therefore, the absence of granulomatous vasculitis on skin biopsy
necrosis are also likely to develop vision loss and tongue necro-
does not rule out GPA. Other commonly seen histological diagno-
sis (Tsianakas et al. 2009), which is a rare manifestation of GCA
ses in GPA are chronic inflammation, granulomatous inflamma-
(Currey 1997). It is postulated that such patients present later in
tion, non-specific ulceration, and erythema nodosum (Daoud et al.
their course of disease, or experienced diagnostic delay leading to
1994; Barksdale et al. 1995; Hu et al. 1977; Comfere et al. 2007).
more tissue damage (Tsianakas et  al. 2009). Additional reported
Histopathological evaluation of gingival changes in GPA reveals
skin manifestations of GCA include urticaria, erythema, vesicles,
pseudoepitheliomatous hyperplasia, microabscess formation, and
bullae, crusting, palpable purpura, livedo reticularis, ecchymosis,
multinucleated giant cells (Knight et al. 2000). This combination
and alopecia in the perfusion plane of the temporal artery.
of findings is not common in other conditions that affect the gums
and therefore may help to make the diagnosis of GPA (Knight et al.
2000; Marzano et al. 2010). Takayasu’s arteritis
Takayasu’s arteritis (TA) is a rare, chronic, recurrent vasculi-
Eosinophilic granulomatosis tis involving the aorta and great vessels. It is estimated that 8 to
28% of patients with TA will develop cutaneous manifestations
with polyangiitis (Pascual-Lopez et  al. 2004). The cutaneous manifestations most
Eosinophilic granulomatosis with polyangiitis (EGPA, previously commonly described are tender subcutaneous nodules on the ante-
Churg–Strauss syndrome) presents with cutaneous manifestations rior shins (erythema nodosum-like lesions), ulcerating tender sub-
in approximately 14% of patients (Bosco et  al. 2011). However, cutaneous nodules on the shins (erythema induratum-like lesions),
cutaneous lesions eventually occur during the disease course in ulcers (pyoderma gangrenosum-like lesions), and livedo reticula-
40 to 81% of EGPA patients (Abril 2011; Rashtak and Pittelkow ris on the lower extremities (Pascual-Lopez et al. 2004; Perniciaro
2008; Baldini et al. 2010; Bosco et al. 2011; Dunogue et al. 2011; et  al. 1987; Frances et  al. 1990). Pyoderma gangrenosum-like
Davis et  al. 1997; Guillevin et  al. 1999). Palpable purpura of the ulcers are more common in Asian populations with TA (Perniciaro
extremities is the most common manifestation, affecting half of the et al. 1987). Histopathologically these subcutaneous nodules and
patients with skin involvement (Abril et al. 2008; Baldini et al. 2010; ulcers comprise deep dermal and subcutaneous necrotizing ves-
Bosco et al. 2011; Dunogue et al. 2011; Davis et al. 1997; Guillevin sels, occasionally with a surrounding septal or lobular pannicu-
et  al. 1999). Signs of medium-vessel involvement, subcutaneous litis (Pascual-Lopez et  al. 2004; Perniciaro et  al. 1987; Frances
nodules, livedo reticularis, ulcers, and cutaneous infarcts are also et al. 1990; Carlson and Chen 2007). Erythematous to violaceous
reported with regularity (Abril et al. 2008; Baldini et al. 2010; Bosco papules located on the extensor surfaces, previously referred to
et al. 2011; Dunogue et al. 2011; Davis et al. 1997; Guillevin et al. ‘Churg–Strauss granulomas’, have also been reported to occur in
1999). Erythematous to violaceous papules located on the exten- patients with TA, revealing palisading granulomas on histopatho-
sor surfaces (previously referred to as Churg–Strauss granulomas) logical exam (Taieb et al. 1987; Carlson and Chen 2007).
128 SECTION 3 clinical manifestations common to vasculitis

Kawasaki’s disease Conclusion


Kawasaki’s disease (KD) is a medium-vessel vasculitis that most The cutaneous manifestations of vasculitis are limited to eight main
commonly occurs in children; however, 81 examples of adult KD presentations: urticarial papules, macular pupura, palpable purpura,
have been described in the literature (Gomard-Mennesson et  al. livedo reticularis, retiform purpura, subcutaneous nodules, necro-
2010). Close to 100% of patients with KD will present in the acute sis, and ulcers. Livedo reticularis, retiform purpura, and subcutane-
phase of disease with mucocutaneous findings, which are included ous nodules are more commonly seen in vasculitides that involve
in the diagnostic criteria for this vasculitis. The diagnostic criteria medium vessels. However, the most common cutaneous mani-
are as follows: presence of more than 5 days of fever with at least four festation of small- and medium-vessel vasculitis is palpable pur-
of the five following characteristic signs: polymorphous rash, cer- pura. Therefore, patients presenting with palpable purpura should
vical lymphadenopathy, non-exudative conjunctivitis, oral fissures undergo evaluation for systemic involvement and ANCA positivity.
or ‘strawberry tongue’, and peripheral cutaneous changes such as Skin biopsies are helpful to confirm the diagnosis of vasculitis; how-
oedema, erythema, and desquamation (Yuan et al. 2012). The acute ever, additional work up is required to further classify the vasculitis.
phase lasts 7 to 14 days and is characterized by fever. The subacute
phase lasts from the end of the fever to approximately day 25 when
References
inflammatory markers subside, followed by a chronic phase which Abbas, M.A., Arun, C.S., Gorsuch, A.N., and Henderson, E.B. (2000). A case
of digital vasculitis. Postgraduate Medical Journal, 76, 715, 723.
starts with subsidence of such markers. The term ‘incomplete’ KD
Abril, A. (2011). Churg-Strauss syndrome: an update. Current Rheumatology
is used for children with fever lasting at least 5 days, and at least two
Reports, 13, 489–95.
of the clinical criteria for KD, with no other explanation for the ill- Agard, C., Mouthon, L., Mahr, A., and Guillevin, L. (2003). Microscopic pol-
ness, and laboratory findings consistent with severe inflammation yangiitis and polyarteritis nodosa: how and when do they start? Arthritis
(Gomard-Mennesson et al. 2010). and Rheumatism, 49, 709–15.
The acute phase is characterized by fever, conjunctival injection, Agnello, V., Chung, R.T., and Kaplan, L.M. (1992). A role for hepatitis C virus
oropharyngeal changes, swelling and erythema of distal extremi- infection in type II cryoglobulinemia. New England Journal of Medicine,
ties, rash, and lymphadenopathy (Leung et  al. 1998; Yuan et  al. 327, 1490–5.
2012). Over 90% of children will develop an exanthem in the first Ahn, J.K., Hwang, J.W., Lee, J., Jeon, C.H., Cha, H.S., and Koh, E.M. (2011).
days of the acute phase (Leung et al. 1998). This usually involves Clinical features and outcome of microscopic polyangiitis under a
new consensus algorithm of ANCA-associated vasculitides in Korea.
the trunk and proximal extremities, with sparing of the face and
Rheumatology International, 32, 2979–86.
distal extremities, but it may be generalized. Involvement of the Aletaha, M., Tavakoli, M., Kanavi, M.R., Hashemlou, A., and Roghaei, S.
perineum occurs in 67% of children; perineal desquamation occur- (2011). Bilateral orbital mass lesions: a presentation of Wegener’s granu-
ring prior to desquamation of the palms and sole may help to make lomatosis. Journal of Ophthalmic and Vision Research, 6, 215–8.
the diagnosis (Leung et al. 1998). Over 90% of children develop Baldini, C., Talarico, R., Della Rossa, A., and Bombardieri, S. (2010). Clinical
conjunctival injection during the acute phase and close to 100% manifestations and treatment of Churg-Strauss syndrome. Rheumatic
will develop oral mucosal changes including red, dry, cracked lips, Disease Clinics of North America, 36, 527–43.
strawberry tongue (hypertrophy of the papillae and hyperaemia) Barksdale, S.K., Hallahan, C.W., Kerr, G.S., Fauci, A.S., Stern, J.B., and Travis,
(Leung et al. 1998). Oedema of hands and feet is also an almost uni- W.D. (1995). Cutaneous pathology in Wegener’s granulomatosis. A clin-
icopathologic study of 75 biopsies in 46 patients. American Journal of
versal sign that occurs within the first few days of the acute phase.
Surgical Pathology, 19, 161–72.
During the subacute phase (when the fever subsides) the tips of Baum, E.W., Sams, W.M., Jr, and Payne, R.R. (1982). Giant cell arteritis: a
the fingers and toes begin to desquamate, with the desquamation systemic disease with rare cutaneous manifestations. Journal of the
spreading proximally (Leung et al. 1998). Peripheral gangrene is American Academy of Dermatology, 6, 1081–8.
a rare manifestation of KD, having been reported to date in 17 Blanco, R., Martinez-Taboada, V.M., Rodriguez-Valverde, V., and
patients with KD as their primary diagnosis (Gomez-Moyano et al. Garcia-Fuentes, M. (1998). Cutaneous vasculitis in children and adults.
2011; Falcini et al. 2006). Associated diseases and etiologic factors in 303 patients. Medicine
KD also occurs in adults. Cervical adenopathy is more common (Baltimore), 77, 403–18.
in adults (93% of adults vs. 15% of children)(Wolff et al. 2007) but Borchers, A.T. and Gershwin, M.E. (2012). Giant cell arteritis:  a review
of classification, pathophysiology, geoepidemiology and treatment.
the mucocutaneous features are essentially identical and all adults
Autoimmunity Reviews, 11, A544–54.
develop conjunctival congestion, exanthem, and enanthem (straw- Bosco, L., Peroni, A., Schena, D., Colato, C., and Girolomoni, G. (2011).
berry tongue, dry fissured lips, oropharyngeal oedema) (Seve et al. Cutaneous manifestations of Churg-Strauss syndrome:  report of two
2005). Adults have also presented with palm and sole erythema cases and review of the literature. Clinical Rheumatology, 30, 573–80
and oedema and progression to desquamation (Seve et al. 2005). Brouet, J.C., Clauvel, J.P., Danon, F., Klein, M., and Seligmann, M. (1974).
As with children, incomplete KD has also been noted in adults. Biologic and clinical significance of cryoglobulins. A report of 86 cases.
Review of the 81 adult cases of KD reported in the literature as of American Journal of Medicine, 57, 775–88.
2010 suggest that the algorithm proposed to diagnose incomplete Brown, N.A. and Carter, J.D. (2007). Urticarial vasculitis. Current
KD in children is also useful to diagnose incomplete KD in adults Rheumatology Reports, 9, 312–9.
Cacoub, P. and Saadoun, D. (2008). Hepatitis C virus infection induced vas-
(Gomard-Mennesson et al. 2010).
culitis. Clinical Reviews in Allergy and Immunology, 35, 30–9.
Skin biopsies of KD are non-specific. Erythema multiforme-like
Carlson, J.A. (2010). The histological assessment of cutaneous vasculitis.
interface dermatitis with marked papillary dermal oedema, pso- Histopathology, 56, 3–23.
riasiform dermatitis, and subcorneal pustular dermatosis are all Carlson, J.A. and Chen, K.R. (2007). Cutaneous vasculitis update:  neutro-
patterns that have been reported to occur in KD (Carlson and philic muscular vessel and eosinophilic, granulomatous, and lymphocytic
Chen 2007). vasculitis syndromes. American Journal of Dermatopathology, 29, 32–43.
CHAPTER 10 cutaneous manifestations of vasculitis 129

Chan, P.T., Ishiko, A., Wada, N., Yamamoto, N., and Amagai, M. (2009). Frances, C., Boisnic, S., Bletry, O., Dallot, A., Thomas, D., Kieffer, E., and
Inflammatory plaque with peripheral nodules:  a new specific finding Godeau, P. (1990). Cutaneous manifestations of Takayasu arteritis.
of cutaneous polyarteritis nodosa. Journal of the American Academy of A retrospective study of 80 cases. Dermatologica, 181, 266–72.
Dermatology, 60, 320–5. Frances, C., Du, L.T., Piette, J.C., Saada, V., Boisnic, S., Wechsler, B., Bletry,
Chen, K.R. (1989). Cutaneous polyarteritis nodosa: a clinical and histopatho- O., and Godeau, P. (1994). Wegener’s granulomatosis. Dermatological
logical study of 20 cases. Journal of Dermatology, 16, 429–42. manifestations in 75 cases with clinicopathologic correlation. Archives of
Chen, K.R. and Carlson, J.A. (2008). Clinical approach to cutaneous vasculi- Dermatology, 130, 861–7.
tis. American Journal of Clinical Dermatology, 9, 71–92. Garcia-Porrua, C., Calvino, M.C., Llorca, J., Couselo, J.M., and Gonzalez-Gay,
Choi, S.W., Lew, S., Cho, S.D., Cha, H.J., Eum, E.A., Jung, H.C., and Park, M.A. (2002). Henoch-Schonlein purpura in children and adults: clini-
J.H. (2006). Cutaneous polyarteritis nodosa presented with digital gan- cal differences in a defined population. Seminars in Arthritis and
grene: a case report. Journal of Korean Medical Science, 21, 371–3. Rheumatism, 32, 149–56.
Chowdhury, M.M., Inaloz, H.S., Motley, R.J., and Knight, A.G. (2002). Gedalia, A. (2004). Henoch-Schonlein purpura. Current Rheumatology
Erythema elevatum diutinum and IgA paraproteinaemia: ‘a preclinical Reports, 6, 195–202.
iceberg’. International Journal of Dermatology, 41, 368–70. Gomard-Mennesson, E., Landron, C., Dauphin, C., Epaulard, O., Petit, C.,
Comfere, N.I., Macaron, N.C., and Gibson, L.E. (2007). Cutaneous mani- Green, L., Roblot, P., Lusson, J.R., Broussolle, C., and Seve, P. (2010).
festations of Wegener’s granulomatosis: a clinicopathologic study of 17 Kawasaki disease in adults: report of 10 cases. Medicine (Baltimore), 89,
patients and correlation to antineutrophil cytoplasmic antibody status. 149–58.
Journal of Cutaneous Pathology, 34, 739–47. Gomez-Moyano, E., Vera Casano, A., Camacho, J., Sanz Trelles, A., and
Currey, J. (1997). Scalp necrosis in giant cell arteritis and review of the litera- Crespo-Erchiga, V. (2011). Kawasaki disease complicated by cutaneous
ture. British Journal of Rheumatology, 36, 814–6. vasculitis and peripheral gangrene. Journal of the American Academy of
Daoud, M.S., Gibson, L.E., Deremee, R.A., Specks, U., El-Azhary, R.A., and Dermatology, 64, e74–5.
Su, W.P. (1994). Cutaneous Wegener’s granulomatosis:  clinical, histo- Guillevin, L., Cohen, P., Gayraud, M., Lhote, F., Jarrousse, B., and Casassus,
pathologic, and immunopathologic features of thirty patients. Journal of P. (1999). Churg-Strauss syndrome. Clinical study and long-term
the American Academy of Dermatology, 31, 605–12. follow-up of 96 patients. Medicine (Baltimore), 78, 26–37.
Davis, M.D. and Brewer, J.D. (2004). Urticarial vasculitis and hypocom- Hara, Y., Tajiri, T., Matsuura, K., and Hasegawa, A. (2004). Acute scrotum
plementemic urticarial vasculitis syndrome. Immunology and Allergy caused by Henoch-Schonlein purpura. International Journal of Urology,
Clinics of North America, 24, 183–213, vi. 11, 578–80.
Davis, M.D., Daoud, M.S., Mcevoy, M.T., and Su, W.P. (1997). Cutaneous Henoch, E.H. (1974). About a peculiar form of purpura, by Eduard H. H.
manifestations of Churg-Strauss syndrome: a clinicopathologic corre- Henoch, Berlin. American Journal of Diseases of Children, 128, 78–9.
lation. Journal of the American Academy of Dermatology, 37, 199–203. Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., Hallahan, C.W., Lebovics, R.S.,
De Souza, F.H., Radu Halpern, A.S., Valente Barbas, C.S., and Shinjo, S.K. Travis, W.D., Rottem, M., and Fauci, A.S. (1992). Wegener granulo-
(2010). Wegener’s granulomatosis: experience from a Brazilian tertiary matosis: an analysis of 158 patients. Annals of Internal Medicine, 116,
center. Clinical Rheumatology, 29, 855–60. 488–98.
Diaz-Perez, J.L., De Lagran, Z.M., Diaz-Ramon, J.L., and Winkelmann, R.K. Holle, J.U., Gross, W.L., Holl-Ulrich, K., Ambrosch, P., Noelle, B., Both, M.,
(2007). Cutaneous polyarteritis nodosa. Seminars in Cutaneous Medicine Csernok, E., Moosig, F., Schinke, S., and Reinhold-Keller, E. (2010).
and Surgery, 26, 77–86. Prospective long-term follow-up of patients with localised Wegener’s
Diaz-Perez, J.L. and Winkelmann, R.K. (1974). Cutaneous periarteritis granulomatosis: does it occur as persistent disease stage? Annals of the
nodosa. Archives of Dermatology, 110, 407–14. Rheumatic Diseases, 69, 1934–9.
Dion, J., Bachmeyer, C., Moguelet, P., Lescure, F.X., and Pagnoux, C. (2010). Hu, C.H., O’Loughlin, S., and Winkelmann, R.K. (1977). Cutaneous mani-
Livedo reticularis and erythematous macules of the forearms indicat- festations of Wegener granulomatosis. Archives of Dermatology, 113,
ing cutaneous microscopic polyangiitis. American Journal of Medicine, 175–82.
123, e5–6. Hung, S.P., Yang, Y.H., Lin, Y.T., Wang, L.C., Lee, J.H., and Chiang, B.L.
Dongre, A., Adhe, V., Kothari, D., Kardekar, S., and Khopkar, U. (2012). Acute (2009). Clinical manifestations and outcomes of Henoch-Schonlein pur-
hemorrhagic edema of infancy: A report of two cases. Indian Journal of pura:  comparison between adults and children. Pediatric Neonatology,
Dermatology, Venereology and Leprology, 78, 121. 50, 162–8.
Dunogue, B., Pagnoux, C., and Guillevin, L. (2011). Churg-strauss syn- Inui, S., Itami, S., Iwai, C., and Yoshikawa, K. (2001). Churg-Strauss syndrome
drome:  clinical symptoms, complementary investigations, prognosis involving medium-sized arteries. Journal of Dermatology, 28, 161–4.
and outcome, and treatment. Seminars in Respiratory and Critical Care Ishiguro, N. and Kawashima, M. (2010). Cutaneous polyarteritis nodosa: a
Medicine, 32, 298–309. report of 16 cases with clinical and histopathological analysis and a
Dupin, N., Chosidow, O., Lunel, F., Cacoub, P., Musset, L., Cresta, P., Frangeul, review of the published work. Journal of Dermatology, 37, 85–93.
L., Piette, J. C., Godeau, P., and Opolon, P. (1995). Essential mixed cryo- Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F.,
globulinemia. A  comparative study of dermatologic manifestations Flores-Suarez, L.F., Gross, W.L., Guillevin, L., Hagen, E.C., et  al.
in patients infected or noninfected with hepatitis C virus. Archives of (2013). 2012 revised International Chapel Hill Consensus Conference
Dermatology, 131, 1124–7. Nomenclature of Vasculitides. Arthritis and Rheumatism, 65, 1–11.
Emerich, P.S., Prebianchi, P.A., Motta, L.L., Lucas, E.A., and Ferreira, L.M. Kawakami, T., Kawanabe, T., Saito, C., Kannari, M., Mizoguchi, M.,
(2011). Acute hemorrhagic edema of infancy:  report of three cases. Nagafuchi, H., Okazaki, T., Ozaki, S., Kimura, K., and Soma, Y. (2007).
Anais Brasileiros de Dermatologia, 86, 1181–4. Clinical and histopathologic features of 8 patients with microscopic pol-
Falcini, F., Ricci, L., Poggi, G.M., Simonini, G., Calabri, G.B., and De Martino, yangiitis including two with a slowly progressive clinical course. Journal
M. (2006). Severe cutaneous manifestations in a child with refractory of the American Academy of Dermatology, 57, 840–8.
Kawasaki disease. Rheumatology (Oxford), 45, 1444–5. Kluger, N., Pagnoux, C., Guillevin, L., and Frances, C. (2008). Comparison
Fiore, E., Rizzi, M., Simonetti, G.D., Garzoni, L., Bianchetti, M.G., and of cutaneous manifestations in systemic polyarteritis nodosa and micro-
Bettinelli, A. (2011). Acute hemorrhagic edema of young children:  a scopic polyangiitis. British Journal of Dermatology, 159, 615–20.
concise narrative review. European Journal of Pediatrics, 170, 1507–11. Knight, J.M., Hayduk, M.J., Summerlin, D.J., and Mirowski, G.W. (2000).
Fiorentino, D.F. (2003). Cutaneous vasculitis. Journal of the American ‘Strawberry’ gingival hyperplasia:  a pathognomonic mucocutaneous
Academy of Dermatology, 48, 311–40. finding in Wegener granulomatosis. Archives of Dermatology, 136, 171–3.
130 SECTION 3 clinical manifestations common to vasculitis

Kumar, L., Thapa, B.R., Sarkar, B., and Walia, B.N. (1995). Benign cutaneous autoimmune diseases in patients with hepatitis C virus infection: char-
polyarteritis nodosa in children below 10 years of age—a clinical experi- acterization of 1020 cases (The HISPAMEC Registry). Journal of
ence. Annals of the Rheumatic Diseases, 54, 134–6. Rheumatology, 36, 1442–8.
Leung, D.Y., Schlievert, P.M., and Meissner, H.C. (1998). The immu- Rashtak, S. and Pittelkow, M.R. (2008). Skin involvement in systemic autoim-
nopathogenesis and management of Kawasaki syndrome. Arthritis and mune diseases. Current Directions in Autoimmunity, 10, 344–58.
Rheumatism, 41, 1538–47. Sais, G., Vidaller, A., Jucgla, A., Servitje, O., Condom, E., and Peyri, J. (1998).
Luqmani, R.A., Suppiah, R., Grayson, P.C., Merkel, P.A., and Watts, R. Prognostic factors in leukocytoclastic vasculitis:  a clinicopathologic
(2011). Nomenclature and classification of vasculitis—update on the study of 160 patients. Archives of Dermatology, 134, 309–15.
ACR/EULAR diagnosis and classification of vasculitis study (DCVAS). Sansonno, D., Carbone, A., De Re, V., and Dammacco, F. (2007). Hepatitis
Clinical and Experimental Immunology, 164 (Suppl. 1), 11–3. C virus infection, cryoglobulinaemia, and beyond. Rheumatology
Macarenco, R.S., Galan, A., Simoni, P.M., Macarenco, A.C., Tintle, S.J., (Oxford), 46, 572–8.
Rose, R., Halasz, C.L., and Carlson, J.A. (2013). Cutaneous lympho- Saulsbury, F.T. (2007). Clinical update: Henoch-Schonlein purpura. Lancet,
cytic thrombophilic (macular) arteritis: a distinct entity or an indolent 369, 976–8.
(reparative) stage of cutaneous polyarteritis nodosa? Report of 2 cases Seishima, M., Oyama, Z., and Oda, M. (2004). Skin eruptions associated with
of cutaneous arteritis and review of the literature. American Journal of microscopic polyangiitis. European Journal of Dermatology, 14, 255–8.
Dermatopathology, 35, 213–9. Seve, P., Stankovic, K., Smail, A., Durand, D.V., Marchand, G., and Broussolle,
Manchanda, Y., Tejasvi, T., Handa, R., and Ramam, M. (2003). Strawberry C. (2005). Adult Kawasaki disease:  report of two cases and literature
gingiva: a distinctive sign in Wegener’s granulomatosis. Journal of the review. Seminars in Arthritis and Rheumatism, 34, 785–92.
American Academy of Dermatology, 49, 335–7. Stussi, G., Schneider, E., Trueb, R.M., and Seebach, J.D. (2001). Acral necrosis
Martinez-Taboada, V.M., Blanco, R., Garcia-Fuentes, M., and of the fingers as initial manifestation of cutaneous polyarteritis nodosa—
Rodriguez-Valverde, V. (1997). Clinical features and outcome of 95 patients a case report. Angiology, 52, 63–7.
with hypersensitivity vasculitis. American Journal of Medicine, 102, 186–91. Taieb, A., Dufillot, D., Pellegrin-Carloz, B., Calabet, A., Clementy, J., Guillard,
Marzano, A.V., Fanoni, D., and Berti, E. (2010). Oral and cutaneous find- J.M., and Maleville, J. (1987). Postgranulomatous anetoderma associ-
ings are valuable diagnostic aids in Wegener’s granulomatosis. European ated with Takayasu’s arteritis in a child. Archives of Dermatology, 123,
Journal of Internal Medicine, 21, 49. 796–800.
Mironiuc, A., Comes, L., Constantinescu, I., Mironiuc, C., and Bontea, D. Tarabishy, A.B., Schulte, M., Papaliodis, G.N., and Hoffman, G.S (2010).
(2008). Cryoglobulinemic vasculitis with multiple digital necrosis in Wegener’s granulomatosis:  clinical manifestations, differential diag-
viral hepatitis. Romanian Journal of Internal Medicine, 46, 91–5. nosis, and management of ocular and systemic disease. Survey of
Monti, G., Saccardo, F., Pioltelli, P., and Rinaldi, G. (1995). The natural history Ophthalmology, 55, 429–44.
of cryoglobulinemia: symptoms at onset and during follow-up. A report Trapani, S., Micheli, A., Grisolia, F., Resti, M., Chiappini, E., Falcini, F., and
by the Italian Group for the Study of Cryoglobulinemias (GISC). Clinical De Martino, M. (2005). Henoch Schonlein purpura in childhood: epi-
and Experimental Rheumatology, 13 (Suppl. 13), S129–33. demiological and clinical analysis of 150 cases over a 5-year period and
Morgan, A.J. and Schwartz, R.A. (2010). Cutaneous polyarteritis nodosa: a review of literature. Seminars in Arthritis and Rheumatism, 35, 143–53.
comprehensive review. International Journal of Dermatology, 49, 750–6. Tsianakas, A., Ehrchen, J.M., Presser, D., Fischer, T., Kruse-Loesler, B., Luger,
Nagai, Y., Hasegawa, M., Igarashi, N., Tanaka, S., Yamanaka, M., and Ishikawa, T.A., and Sunderkoetter, C. (2009). Scalp necrosis in giant cell arteritis: case
O. (2009). Cutaneous manifestations and histological features of micro- report and review of the relevance of this cutaneous sign of large-vessel vas-
scopic polyangiitis. European Journal of Dermatology, 19, 57–60. culitis. Journal of the American Academy of Dermatology, 61, 701–6.
Otani, Y., Anzai, S., Shibuya, H., Fujiwara, S., Takayasu, S., Asada, Y., Terashi, Uffelmann, N.C., Kozel, J., Chaudhry, S.B., and Vidal, C.I. (2012). Reticulated
H., Takuma, M., and Yokoyama, S. (2003). Churg-Strauss syndrome lesion as an early diagnostic sign for giant cell arteritis. Dermatology
(CSS) manifested as necrosis of fingers and toes and liver infarction. Online Journal, 18, 6.
Journal of Dermatology, 30, 810–5. Villiger, P.M. and Guillevin, L. (2010). Microscopic polyangiitis:  Clinical
Pagnoux, C., Seror, R., Henegar, C., Mahr, A., Cohen, P., Le Guern, V., presentation. Autoimmunity Reviews, 9, 812–9.
Bienvenu, B., Mouthon, L., and Guillevin, L. (2010). Clinical features Wahl, C.E., Bouldin, M.B., and Gibson, L.E. (2005). Erythema elevatum diuti-
and outcomes in 348 patients with polyarteritis nodosa:  a systematic num: clinical, histopathologic, and immunohistochemical characteristics
retrospective study of patients diagnosed between 1963 and 2005 and of six patients. American Journal of Dermatopathology, 27, 397–400.
entered into the French Vasculitis Study Group Database. Arthritis and Waseda, K., Tanimoto, Y., Hasegawa, K., Miyahara, N., Nojima, D., Ikeda,
Rheumatism, 62, 616–26. G., Kanehiro, A., Okada, C., Kimata, Y., and Tanimoto, M. (2011).
Pakrou, N., Selva, D., and Leibovitch, I. (2006). Wegener’s granulomato- Churg-Strauss syndrome with necrosis of toe tips. Acta Medicinae
sis: ophthalmic manifestations and management. Seminars in Arthritis Okayama, 65, 215–8.
and Rheumatism, 35, 284–92. Williams, V.L., Guirola, R., Flemming, K., Modi, G.M., Rosales, C., and
Pascual-Lopez, M., Hernandez-Nunez, A., Aragues-Montanes, M., Dauden, Deguzman, M.M. (2012). Distal extremity necrosis as a manifestation
E., Fraga, J., and Garcia-Diez, A. (2004). Takayasu’s disease with cutane- of cutaneous polyarteritis nodosa: case report and review of the acute
ous involvement. Dermatology, 208, 10–5. management of a pediatric patient. Pediatric Dermatology, 29, 473–8.
Patten, S.F. and Tomecki, K.J. (1993). Wegener’s granulomatosis:  cutane- Wolff, A.E., Hansen, K.E., and Zakowski, L. (2007). Acute Kawasaki dis-
ous and oral mucosal disease. Journal of the American Academy of ease: not just for kids. Journal of General Internal Medicine, 22, 681–4.
Dermatology, 28, 710–8. Yiannias, J.A., El-Azhary, R.A., and Gibson, L.E. (1992). Erythema elevatum
Perniciaro, C.V., Winkelmann, R.K., and Hunder, G.G. (1987). Cutaneous diutinum: a clinical and histopathologic study of 13 patients. Journal of
manifestations of Takayasu’s arteritis. A clinicopathologic correlation. the American Academy of Dermatology, 26, 38–44.
Journal of the American Academy of Dermatology, 17, 998–1005. Yuan, K., Park, J.K., Qubti, M.A., and Haque, U.J. (2012). Recurrent Kawasaki
Ramos-Casals, M., Munoz, S., Medina, F., Jara, L.J., Rosas, J., Calvo-Alen, disease with strawberry tongue and skin desquamation in a young adult.
J., Brito-Zeron, P., Forns, X., and Sanchez-Tapias, J.M. (2009). Systemic Journal of Clinical Rheumatology, 18, 96–8.
CHAPTER 11

Ocular manifestations
of systemic vasculitis
Russell W. Read

Introduction this extensive blood supply, vasculitides affecting all vessel sizes
may involve the eye.
A feature of all vasculitides is inflammation of vessel walls at There are at least two perspectives on the ocular manifestations
some point during the course of disease (Jennette et  al. 2013). of vasculitis: that of the ophthalmologist who diagnoses an ocular
The eyeball (Figure 11.1) and surrounding structures are richly condition and must consider whether it may be a manifestation of
vascularized, supplied by branches from the internal and external a systemic vasculitis; and the non-ophthalmologist who diagnoses
carotids. The orbit is supplied by branches of the meningolacrimal (or suspects) a systemic vasculitis and seeks to determine if the eye
artery, the palpebral artery, and the ophthalmic artery, which then is involved. Because the eye is the only location in the human body
exit the orbit and anastomose in the periocular skin, providing in which vasculature can be directly visualized in the natural state,
its blood supply. Two vascular systems supply the globe, each a the ophthalmologist’s contribution in this scenario can be both
branch of the ophthalmic artery, itself the first branch of the inter- vision preserving (because of the potential for profound visual loss
nal carotid. The central retinal artery penetrates the optic nerve from vasculitis) and a diagnostic and prognostic aid to the refer-
sheath to run within the optic nerve, emerging through the optic ring physician by confirming or refuting the presence and, perhaps
cup into the intraocular space, then branching to supply the inner more importantly, the nature of the vasculitis. Thus most patients
retina. Short posterior ciliary arteries penetrate the posterior with systemic vasculitides (and certainly all with ocular symptoms)
sclera to supply the choroid, nourishing the outer retina, while should have an eye examination. This chapter discusses possible
long posterior ciliary arteries also penetrate the sclera, running ocular manifestations, organized by ocular anatomy, followed by a
in the suprachoroidal space to the anterior segment, supplying section organized by systemic vasculitides. The reader will want to
the iris and ciliary body. Branches from the muscular artery sup- refer back to ocular manifestations when reading about the various
plying the extraocular muscles continue forward to also supply diseases for a better understanding of the ocular processes identi-
the anterior segment. The retrobulbar optic nerve is supplied by fied in association with the systemic conditions.
branches from the central retinal artery as well as pial arteries
from the optic nerve sheath (Cioffi et al. 2003; Netter 1989). Given
Ocular tissues subject
to systemic vasculitis
Sclera
A variety of ocular conditions may be caused by or associated with
systemic vasculitis, and in this section we will therefore consider
the anatomy of the structures that can be involved.
Choroid
Conjunctiva
Iris Lens
The conjunctiva is a mucous membrane originating at the limbus
Cornea
Optic nerve and extending posteriorly as the bulbar conjunctiva, reflecting at
the fornices to travel anteriorly as the palpebral conjunctiva, cov-
ering the posterior surface of the upper and lower eyelid, ending
Retina
at the eyelid margin where it transitions into keratinized eyelid
skin. Vasculitis may manifest in the conjunctiva in several forms.
Ciliary Ulcerations of the conjunctiva have been reported in Behçet’s
body
disease. Similar to the oral ulcers, these tend to be painful. They
may be located on the bulbar or palpebral conjunctiva and usually
respond to local corticosteroid therapy (Zamir et al. 2003). Swelling
Fig. 11.1 Schematic cross section of eye, revealing the uveal tract (in brown), the and mass lesions of the conjunctiva have been reported as the ini-
target of uveitis. tial manifestation of eosinophilic granulomatosis with polyangiitis
132 SECTION 3 clinical manifestations common to vasculitis

inflammatory disease, resulting in scleritis and episcleritis. Scleritis


is a severe, potentially blinding condition associated with underly-
ing systemic disease in up to 50% of cases, of which half are due to
rheumatoid arthritis. Smith and colleagues reviewed the literature
on scleritis and episcleritis and found that after rheumatoid arthri-
tis, GPA was identified in approximately 4 to 8% of cases, relapsing
polychondritis in 2 to 6%, systemic lupus erythematosus in 1 to
4%, inflammatory bowel disease in 2 to 4%, seronegative spondy-
loarthropathies in 0.3 to 4%, and polyarteritis nodosa in 0.4 to 1%
(Smith et al. 2007).
Scleritis manifests as dilatation of the deep and superficial epis-
cleral vessels with underlying scleral oedema (Figure  11.3). This
vascular dilation gives a violaceous hue to the sclera that is best
appreciated in natural light. Scleritis is typically painful, often
severely so, to the point of awakening one from sleep. The pain
Fig. 11.2 Diffuse anterior scleritis with adjacent associated corneal oedema in a may be described as boring, dull, and achy, but not usually sharp.
patient with granulomatosis with polyangiitis (Wegener’s granulomatosis).
Movement of the eye causes pain, as does palpation. Scleritis may
affect any portion of the eye and is classified anatomically as: ante-
(EGPA, Churg–Strauss), with histology confirming the presence of rior diffuse, anterior nodular, or anterior necrotizing; posterior; and
necrotizing eosinophilic granulomas (Ameli et al. 2011; Margolis scleromalacia perforans, the latter a subset typically seen in patients
et al. 2007). The most common manifestation of systemic autoim- with advanced rheumatoid arthritis. Scleromalacia perforans man-
mune disease affecting the conjunctiva is keratoconjunctivitis sicca ifests as necrotizing disease without otherwise identifiable clinical
(KCS), or dry eye syndrome (Ausayakhun et al. 2002). This may signs of inflammation, such as vascular dilation. Necrotizing scle-
manifest as redness and mild pain and can be confused with more ritis is characterized by areas of avascular sclera with loss of scleral
serious surface conditions, such as peripheral ulcerative kerati- tissue and potential exposure of the underlying uveal tissue (cho-
tis or scleritis. At its most severe, KCS may itself result in serious roid and ciliary body). Diffuse anterior scleritis is the most com-
ocular complications such as corneal opacity, neovascularization, mon form (60%) followed by nodular anterior (21%), necrotizing
and keratinization of the cornea. In addition to treatment of the (12%), and posterior scleritis (7%) (Jabs et al. 2000a). Necrotizing
underlying systemic condition, local therapy with ocular lubri- scleritis is more likely to occur in the setting of a systemic vasculi-
cants, punctal occlusion, and topical anti-inflammatories such as tis (Jabs et al. 2000a; Watson and Hayreh 1976) and patients with
ciclosporin are helpful. necrotizing scleritis tend to be older, to have more ocular complica-
tions, and are more likely to require systemic therapy than those
Cornea with other anatomical categories (Jabs et al. 2000a; Sainz de la Maza
The cornea is also affected by KCS and peripheral ulcerative kera- et al. 2012). Treatment of scleritis varies based on the presence or
titis (PUK), which is more severe than KCS. It may be a sign of absence of an associated systemic condition also requiring treat-
systemic autoimmune disease, most commonly rheumatoid arthri- ment, and the severity of the ocular disease. Scleritis in the setting of
tis, and the systemic vasculitides. These include granulomatosis systemic vasculitis is treated by addressing the systemic condition
with polyangiitis (GPA, formerly Wegener’s granulomatosis), sys- with corticosteroids, antimetabolites, biologics (TNF inhibitors
temic lupus erythematosus, and polyarteritis nodosa (Yagci 2012). and rituximab primarily), and alkylating agents as needed. Scleritis
PUK may occur in isolation or in association with scleritis in up to not associated with systemic disease, or in the setting of otherwise
35% of patients; necrotizing scleritis is the most common associ- well-controlled systemic disease, may be able to be treated locally
ated form, and posterior the least (Sainz de la Maza et al. 2012). (Sohn et al. 2011; Albini et al. 2005; Zamir et al. 2002).
PUK may or may not be painful. The peripheral corneal epithelium
ulcerates, followed by stromal loss and thinning, which may pro-
gress to perforation. KCS worsens this clinical picture. Treatment
of PUK is systemic, along with local structural support measures in
the case of extreme thinning and perforation. In contrast to thin-
ning, the peripheral cornea may show oedema in areas adjacent to
scleritis (Figure 11.2). This may develop into ulceration and thin-
ning, so careful observation during treatment is necessary.

Sclera and episclera


The sclera is composed of collagen and forms the eye’s outer wall,
or ‘white of the eye’. Itself avascular, the sclera receives its blood
supply from the deep episcleral vessels that lie on its surface.
Superficial to the sclera is the episclera, containing the superficial
and deep episcleral vessels, peripheral nerves, and lymphatics in
a loose matrix of connective tissue. The conjunctiva lies on top Fig. 11.3 Diffuse anterior scleritis in a patient with granulomatosis with polyangiitis,
of the episclera. Both the sclera and episclera may be affected by revealing area of violaceous hue and scleral oedema of upper bulbar sclera.
CHAPTER 11 ocular manifestations of systemic vasculitis 133

In contrast, episcleritis is mild, superficial, and visually


non-threatening. The eye is red from dilatation of the superficial
episcleral vessels, but the deep vessels are uninvolved and pain is
mild, if present. Episcleritis is associated with a systemic vasculitic
or connective tissue disease in approximately 15% of cases (Sainz
de la Maza et al. 2012). It is probably best treated by observation
alone but if treatment is initiated, topical non-steroidals, topical
ciclosporin, or low-potency, low-penetrance topical corticosteroids
such as fluorometholone 0.25% are preferred.

Uveal tract
The uvea is a pigmented vascular tissue within the eye, composed,
from anterior to posterior, of the iris, ciliary body, and choroid. It
makes up the middle layer of the posterior segment, sandwiched
between the sclera externally and the retina internally (internal
to which is the vitreous cavity) and is a component of the barrier
between the anterior and posterior segments. Because the hall-
mark of intraocular inflammation is the presence of white blood
cells (WBC) within the eye, and the entry point for such cells is
the vasculature, the presence of intraocular WBCs is interpreted
to be a result of uveal inflammation, thus uveitis. The incidence Fig. 11.4 Retinal vasculitis with yellowish perivascular sheathing and area of
of uveitis is estimated to be between 17.4 and 340.9 cases per intraretinal haemorrhage in a patient with Toxoplasma gondii-associated retinitis
and retinal vasculitis.
100 000 person-years, with a prevalence of 69 to 1231 cases per
100 000 persons (Darrell et al. 1962; Suhler et al. 2008; Gritz and
Wong 2004; Reeves et al. 2006). While these studies included vas- uveitis patients had retinal vasculitis as a component of their dis-
culitis in their diagnostic code search, specifics on the incidence ease but that only 1.4% of patients with a systemic vasculitis had
and prevalence of systemic vasculitis as an associated diagnosis retinal vasculitis as a manifestation. Thirty per cent of Behçet’s
were not provided. Rodriguez and colleagues did provide infor- disease patients had retinal vasculitis (Rosenbaum et al. 2012).
mation on associated causes in their Northeastern United States Rodriguez and co-workers found that the most frequent diag-
tertiary referral centre. Of 1237 patients, approximately 8% had noses in which retinal vasculitis was a clinical feature included
or were strongly suspected of having a systemic vasculitis, with sarcoidosis (17.3%), SLE (15.3%), Behçet’s disease (11.7%), bird-
4.8% having systemic lupus erythematosus, 2.5% Behçet’s disease, shot retinochoroidopathy (9.1%), multiple sclerosis (3.3%), and
0.3% GPA, 0.2% relapsing polychondritis, and 0.2% giant cell arte- toxoplasmosis (2.8%) (Rodriguez et al. 1996). Occlusive retinal
ritis (Rodriguez et al. 1996). Uveitis has been reported rarely in changes lead to retinal haemorrhages (Figure 11.5a and b) and
Takayasu’s arteritis. It is treated based on the location and severity ischaemia, the latter resulting in increased levels of vascular
of the inflammation. Anterior disease may be adequately treated endothelial growth factor (VEGF) production by the retina and
with topical corticosteroids alone. Posterior segment disease neovascularization of the retina and anterior segment, potentially
requires periocular, intraocular, or systemic therapy. An escalat- leading to tractional retinal detachment, vitreous haemorrhage,
ing approach that begins with oral corticosteroids and progresses and neovascular glaucoma. Ischaemia also results in damage to
to antimetabolites, calcineurin inhibitors, biologics, and alkylating retinal function with a loss of visual function. As with other ocu-
agents (not necessarily in that order) is typically used (Jabs et al. lar manifestations of systemic disease, retinal vasculitis is treated
2000b). by targeting the underlying disorder. Local laser photocoagula-
tion and intravitreal VEGF inhibitor injection are indicated for
Retinal vasculature ischaemic complications.
In 2005, Jabs and colleagues reported the results of the first inter- Regardless of terminology, retinal vasculitis may cause signifi-
national workshop on the standardization of uveitis nomenclature cant visual loss, though Ku and co-workers examined outcomes of
(Jabs et al. 2005). The topic of retinal vasculitis was discussed, but patients with retinal vasculitis and found a decline in LogMAR vis-
no definitive defining characteristics or diagnostic criteria were ual acuity of 0.01 units per year, equivalent to a decrease of 0.1 lines
agreed upon. What was agreed was that ‘retinal vasculitis’ from of vision on the Snellen chart. Males declined more than females
an ophthalmology perspective is a descriptive term, encompass- (Ku et al. 2012).
ing clinical scenarios in which intraocular inflammation (WBCs
in the eye) and perivascular exudate, intraretinal haemorrhage, Orbital disease
and nerve fibre layer infarcts are present (Figure 11.4). There is Orbital inflammation may involve any or all of the contained struc-
no consideration of underlying aetiology, which may include tures, including fat, peripheral nerves, extraocular muscle, and lacri-
infectious and non-infectious conditions. There is rarely his- mal gland. Typical clinical signs include erythema and oedema of the
tological confirmation of vasculitic changes because biopsy of lids and conjunctiva, restriction of extraocular motility, and prop-
intraocular structures is rare. Thus the terminology used by oph- tosis. GPA is the most common systemic vasculitis causing orbital
thalmologists and rheumatologists or other internists may dif- inflammation, but it occurs in only 15 to 20% of patients (reviewed
fer significantly. Rosenbaum and colleagues found that 15% of in Perumal et  al. 2012). Orbital congestion with optic nerve
134 SECTION 3 clinical manifestations common to vasculitis

compression is the most serious vision-threatening aspect of orbital the extraocular muscles (Ghanchi and Dutton 1997). Bilateral visual
inflammation. If systemic anti-inflammatory therapy is not effective involvement occurs in approximately one-third of patients with GCA
in reducing inflammation, orbital decompression may be necessary. (Hayreh et al. 1998b) with 5% retaining permanent bilateral visual
loss (Gonzalez-Gay et al. 2000). Permanent visual loss is more fre-
Neuro-ophthalmic involvement quent in patients with transient visual loss, transient diplopia, and
The most common systemic vasculitis with neuro-ophthalmic jaw claudication (Gonzalez-Gay et  al. 2000). A  lower erythrocyte
manifestations is giant cell arteritis (GCA), causing an ischaemic sedimentation rate and absence of systemic symptoms is associated
optic neuropathy (ION). One of the few true ophthalmic emergen- with an increased risk of ischaemic complications (Cid et al. 1998).
cies, GCA ION must be recognized quickly in order to reduce visual Definitive diagnosis of GCA has always been based on tempo-
loss in the affected eye, and to protect the fellow eye. Constitutional ral artery biopsy with demonstration of granulomatous inflamma-
symptoms of fatigue, fever, and weight loss are often accompanied tion of the artery wall and disruption of the mural elastic lamina.
by scalp tenderness and jaw claudication (Hayreh 1998). Suspicion Traditionally, biopsy is carried out on the side of involvement (or
of GCA should prompt an urgent erythrocyte sedimentation rate, side of most profound effect if bilateral). Biopsy size and choice of
initiation of high-dose corticosteroids, and temporal artery biopsy initial bilateral versus unilateral biopsy has been evaluated. Breuer
(Borchers and Gershwin 2012). and colleagues evaluated the association between biopsy length and
diagnostic sensitivity. They found that the rate of positive biopsies
was 19% with a biopsy length of 5 mm or less as compared to up
Systemic vasculitis and to 79% with biopsy lengths between 6 and 20 mm. Biopsies greater
its ocular manifestations than 20 mm increased the rate to 89%. Of positive biopsies, only
Nomenclature 3% were 5 mm or less (Breuer et al. 2009a). The same group found
that bilateral biopsies increased the diagnostic sensitivity as much
The nomenclature of systemic vasculitis was established by consen- as 13% over unilateral biopsies (Breuer et al. 2009b). In contrast,
sus conference in 1992 (Jennette et al. 1994), with a recent revision three other groups found the probability of obtaining a positive
based on the 2012 Revised International Chapel Hill Consensus result following a contralateral negative biopsy was 1 to 5% (Boyev
Conference Nomenclature of Vasculitides (Jennette et al. 2013). The et  al. 1999; Danesh-Meyer et  al. 2000; Pless et  al. 2000). Recent
organization of the following sections follows the 2012 revision. work has focused on alternate non-invasive diagnostic techniques.
High-resolution colour Doppler ultrasound of the temporal artery
Ocular manifestations of the primary showing the so-called halo sign of concentric hypoechogenic vessel
systemic vasculitides wall thickening was highly specific with good sensitivity (Schmidt
et  al. 1997; Arida et  al. 2010). Finally, high-resolution MRI was
Large-vessel vasculitis found to be comparable to colour-coded duplex ultrasound in the
Large-vessel vasculitis (LVV) predominately affects the aorta, its diagnosis of GCA (Bley et al. 2008); however, the exact role of these
major branches, and the corresponding veins, though vessels of non-invasive techniques has yet to be fully determined.
other sizes may be affected as well (Jennette et al. 2013).
Takayasu’s arteritis
Giant cell arteritis Takayasu’s arteritis (TA) is an autoimmune granulomatous arte-
Giant cell arteritis (GCA) is a systemic necrotizing vasculitis with ritis affecting the aorta and its branches, predominately in young
a predilection for large- and medium-sized vessels, especially the women (Panja and Mondal 2004).
cranial arteries branching off the carotid. GCA occurs most com- Ocular manifestations
monly in Caucasian women over 50 years of age (Hayreh et al. 1998b;
Perry and co-workers conducted a large cross-sectional study over
Gonzalez-Gay et al. 2010), though other racial groups may be affected
16 months in a tertiary care teaching hospital in India (Peter et al.
more than once appreciated (Gonzalez et al. 1989). Approximately one
2011). Patients were recruited from the departments of cardiology
in five GCA patients will present with ocular only disease while half
or rheumatology, thus avoiding bias in an ophthalmology only study.
will exhibit ocular and extraocular symptoms (Hayreh et al. 1998a).
Of 61 patients, mean age was 32 years (92% had onset of disease at
Ocular manifestations less than 40 years of age); 77% were female, consistent with historical
As described previously, GCA is an ophthalmic emergency, with a literature. Decreased vision was present in 30%, headache in 41%,
potential for rapid progression to profound and permanent vision and limb claudication in 61%. No patients reported ocular pain.
loss in one or both eyes without prompt treatment (Tovilla-Canales Takayasu’s retinopathy was found in 13% of eyes (15% of patients)
1998). Ocular symptoms include visual loss (of varying levels) in 98% and hypertensive retinopathy in one of six patients. Anterior ischae-
with amaurosis fugax in 31%, diplopia in 6%, and eye pain in 8% mic optic neuropathy was present in 3% and ocular ischaemic syn-
(Hayreh et al. 1998b). Interestingly, headache, myalgias, and fever drome in 7%. Ocular ischaemic syndrome was more likely to occur
were significantly more likely to occur in patients without ocular with advancing age and increasing duration of disease (Peter et al.
involvement (Hayreh et al. 1998b; Gonzalez-Gay et al. 2000). The 2011). Signs of TA retinopathy include generalized vasodilation with
most common ocular manifestation of GCA is arteritic anterior capillary microaneurysms, and as the disease advances, arteriovenous
ischaemic optic neuropathy in 80% of patients, manifesting as pallid anastomosis and capillary dropout, followed by complications such
optic nerve swelling. Other vascular occlusive events include central as cataract, neovascular glaucoma, vitreous haemorrhage, and optic
retinal artery occlusion (14%), cilioretinal artery occlusion (22%), atrophy. Fluorescein angiography reveals a prolonged arm-to-retina
and posterior ischaemic optic neuropathy (7%)(Hayreh et al. 1998b). circulation time. Anterior ischaemic optic neuropathy is associated
Ocular motility abnormalities may occur as a result of ischaemia of with a worse prognosis (Karam et al. 1999).
CHAPTER 11 ocular manifestations of systemic vasculitis 135

Medium-vessel vasculitis disc oedema, and superficial punctate keratitis have also been
reported (Ohno et al. 1982; Jacob et al. 1982; Birnbaum et al. 2012).
Medium vessels are typified by those of the visceral arteries and
Dacryocystitis has been associated with KD, appearing 6 months
veins and their initial branches, though considerable overlap exists
following resolution of acute disease (Mauriello et  al. 1986).
in the size of vessel affected by the various subtypes of vasculitis
Post-mortem examination of a 4 month old with KD revealed vas-
(Jennette et al. 2013).
culitis with thrombosis of a branch of the ophthalmic artery and
Polyarteritis nodosa bilateral inner retinal ischaemia (Font et al. 1983).
Polyarteritis nodosa (PAN) is a rare necrotizing vasculitis of
medium or small arteries without glomerulonephritis or vasculitis
Small-vessel vasculitis
of arterioles, capillaries, or venules. It is not associated with anti- The vasculature affected in small-vessel vasculitis (SVV) is gener-
neutrophil cytoplasmic antibodies (ANCA) (Jennette et al. 2013). ally intraparenchymal and includes arteries, arterioles, capillaries,
venules, and veins (Jennette et al. 2013). SVV is further divided into
Ocular manifestations
ANCA-associated vasculitis (AAV) and immune complex SVV.
PAN may affect any portion of the ocular tissues, with manifesta-
tions including conjunctival necrosis, scleritis, peripheral ulcerative ANCA-associated vasculitis
keratitis, non-granulomatous uveitis, retinal vasculitis, pseudotu- Granulomatosis with polyangiitis
mour of the orbit, and central retinal artery occlusion (Akova et al. Granulomatosis with polyangiitis (GPA), previously Wegener’s
1993). Ocular involvement is present in up to 20% of patients with granulomatosis (Falk et al. 2011a; Falk et al. 2011b; Falk et al. 2011c)
PAN (Pagnoux et al. 2010; Perez et al. 2004). is a necrotizing granulomatous vasculitis of small to medium ves-
Scleritis due to PAN may be diffuse, nodular, or necrotizing. sels primarily, with involvement of the upper and lower respiratory
PUK may occur concurrently with the scleritis, and progress tract. Involvement of the kidney is common, as is the orbit. Both
both circumferentially and centrally, creating an undercut edge granulomatous and non-granulomatous extravascular inflamma-
similar to Mooren’s ulcer; however, in contrast to Mooren’s ulcer, tion are seen (Jennette et al. 2013). The annual incidence of GPA is
PUK in PAN frequently has associated scleritis (Messmer and 8.5 per million (Watts et al. 1995).
Foster 1999). Ocular Manifestations. GPA can manifest in any ocular or perio-
The most common ocular findings in polyarteritis nodosa are cular tissue. GPA occurs in more than one phenotype, with a lack
choroidal and retinal vasculitis. Choroidal vasculitis may not be of renal involvement characterized as limited disease (Lamprecht
apparent clinically, but may manifest as Elschnig’s spots (yellow and Gross 2004). Despite the existence of limited disease, it is
spots in the choroid) scattered throughout the posterior pole as a rare for GPA to manifest solely in one organ without an eventu-
result of choroidal ischaemia (Perez et al. 2004; Morgan et al. 1986). ally wider presence. Of 494 patients in a French GPA database, 16
These lesions are not specific for PAN, but do portend a poor prog- (3.2%) had single organ involvement, four of which were isolated
nosis (Klien 1968). Subhyaloid haemorrhage, intraretinal haemor- to the eye. Of those four, all had orbital pseudotumour with exoph-
rhage, oedema, exudates, nerve fibre layer infarcts, retinal vascular thalmia and one additionally had necrotizing scleritis (Pagnoux
irregularity and occlusion (particularly of the central retinal artery), et al. 2011). Data from a large cohort of patients treated at a GPA
and exudative retinal detachment have all been described (Akova focused clinic at the National Institutes of Health showed that 15%
1993; HSU 2001). Diffuse bilateral panuveitis (with anterior cham- of patients had evidence of ocular involvement during early-stage
ber and vitreous cells associated with retinal vasculitis) has also disease. Scleritis and conjunctivitis were the most common mani-
been described in PAN (Morgan et al. 1986). festations at that point, though orbital disease with proptosis was
The orbit may be involved, resulting in an orbital pseudotumour- considered the most diagnostically useful manifestation. Over
like picture, with proptosis. Cranial nerve palsies are the most the course of the disease, 52% of patients eventually had ocular
common neuro-ophthalmic manifestations (Golnik 2004) but oth- manifestations (Hoffman et al. 1992). Involvement of the orbit is
ers, including hemianopia, nystagmus, amaurosis fugax, diplopia, common and may occur primarily or as a consequence of adja-
and Horner’s syndrome, have been reported (Ford and Siekert cent sinus involvement (Haynes et al. 1977). Orbital inflammation
1965). Central nervous system involvement is not common but produces proptosis with eye pain and potential visual loss due to
may be devastating, including ischaemic and haemorrhagic stroke optic nerve compression. In addition to scleritis (Figures 11.2 and
(Guillevin et  al. 1997). Neuroimaging studies may show small 11.3) and orbital inflammation, dacryocystitis, retinal, and cor-
multifocal infarcts, although their appearance is non-specific neal involvement may be present (Hoffman et al. 1992). Uveitis is
(Golnik 2004). uncommon.
Kawasaki’s disease Conjunctival involvement may mimic that of mucous membrane
pemphigoid, with development of cicatrization with loss of the con-
Kawasaki’s disease (KD) is a systemic vasculitis of medium-sized
junctival fornices and symblepharon formation. Entropion and trichi-
vessels, especially coronary arteries, occurring primarily in chil-
asis may result with progressive damage to the ocular surface (Perez
dren under 5 years of age. The clinical manifestations include fever,
et al. 2004). Corneal involvement in the form of peripheral ulcera-
skin rash, conjunctival injection, and cervical lymphadenopathy
tive keratitis is frequently associated with scleritis, often necrotizing
(Iannetti et al. 2012; Jennette et al. 2013).
(Kubal and Perez 2010). PUK in GPA is distinguished from that of
Ocular manifestations Mooren’s ulcer by the lack of associated scleritis in the latter and lack
A conjunctivitis without purulent discharge is the most common of an overhanging edge in the former. Local therapy alone is inef-
ocular manifestation of KD, though its presence is not universal. fective and systemic immunosuppression is needed, though local
Acute anterior uveitis (always bilateral), vitreous opacities, optic adjunctive therapy in the form of tectonic support surgery and local
136 SECTION 3 clinical manifestations common to vasculitis

inhibition of collagenase with medroxyprogesterone may be of ben- to basement membrane in glomerular and pulmonary alveolar
efit (Kubal and Perez 2010). Involvement of the inner eye in the form capillaries (Jennette et  al. 2013). Retinal vasculitis with macular
of retinal vasculitis may also occur, but is rare (Haynes et al. 1977; oedema (Kohler 1988) and scleritis (Riono et al. 1999) have both
Harman and Margo 1998; Rosenbaum et al. 2012). When present, been reported in association with Goodpasture’s syndrome, as
GPA-associated retinal vasculitis presents with nerve fibre layer anti-GBM is referred to when both renal and pulmonary involve-
infarcts and retinal haemorrhage (Thorne and Jabs 2001). ment are present.
GPA is associated with the presence of ANCA, discussed else- Cryoglobulinaemic vasculitis
where in this volume. Consistent with evidence of the role of
Cryoglobulins are circulating immunoglobulins that precipi-
ANCA in disease pathogenesis, patients in whom ANCA levels do
tate at temperatures below normal body temperature (37°C).
not return to normal during treatment may be at greater risk for
Cryoglobulinaemia is the presence of these cryoglobulins in serum
ocular relapse following treatment withdrawal (Power et al. 1995).
and cryoglobulinaemic vasculitis (CV) is vasculitis with cryoglobu-
As mentioned above in this section, treatment for the ocular mani-
lin immune deposits that affects small vessels, predominantly capil-
festations of GPA is systemic, with adjunctive local therapy.
laries, venules, and arterioles (Jennette et al. 2013).
Eosinophilic granulomatosis with polyangiitis
Ocular Manifestations. Deposition of monoclonal IgG-kappa
Eosinophilic granulomatosis with polyangiitis (EGPA, Churg–
immunoglobulin along the corneal Bowman’s layer has been
Strauss syndrome), is an eosinophil-rich necrotizing granuloma-
reported. Superficial keratectomy was successful in restoring vision
tous inflammation involving the respiratory tract with necrotizing
(Kremer et al. 1989). Excimer laser phototherapeutic keratectomy
vasculitis affecting predominantly small to medium vessels. Asthma
has also been reported to be successful (Kremer and Blumenthal
and nasal polyps are common and eosinophilia is a prominent fea-
1997). As with the other systemic vasculitides, scleritis and PUK
ture. Extravascular inflammation is common (Jennette et al. 2013).
have been reported in CV (Kedhar et al. 2007). Anterior segment
EGPA occurs in 2.4 individuals per million per year (Watts et al.
ischaemia with resultant iris neovascularization was reported in two
1995). ANCA positivity appears to be strongly correlated with the
patients with CV (Telander et al. 2006). Posterior segment manifes-
presence of renal involvement (Sinico et al. 2006).
tations have been reported in essential mixed cryoglobulinaemia,
Ocular Manifestations. EGPA is a rare cause of ocular disease including Purtscher-like retinopathy, retinal vasculitis, and serous
(Rosenbaum et  al. 2012). When present, multiple findings have retinal and retinal pigment epithelium detachment mimicking cen-
been reported, including orbital inflammatory disease, conjuncti- tral serous choroidopathy (Cohen et al. 1996; Myers et al. 2001).
val involvement, episcleritis, PUK, uveitis, scleritis, retinal artery
IgA vasculitis
occlusion, ischaemic optic neuropathy, multifocal choroidal ischae-
mia, and cranial nerve palsy. In addition to renal disease, ANCA IgA Vasculitis (IgAV) (Henoch–Schönlein) is a vasculitis charac-
positivity also appears to be associated with more severe orbital dis- terized by IgA1-dominant immune deposits involving the small
ease and a greater risk of visual loss (Takanashi et al. 2001). vessels, predominantly capillaries, venules, and arterioles. IgAV
often involves skin and gastrointestinal tract, and frequently causes
Microscopic polyangiitis arthritis (Jennette et  al. 2013). It most commonly affects chil-
Microscopic polyangiitis (MPA) is a necrotizing vasculitis of small dren and is the most common vasculitis of childhood (Saulsbury
vessels with possible involvement of small to medium arteries. 2001)  though it does occur in approximately 1.2 per 1  million
Glomerulonephritis is commonly present and pulmonary capillary adults (Watts et al. 1995).
involvement often occurs. Extravascular inflammation is absent Ocular Manifestations. Central nervous system involvement,
(Jennette et al. 2013). under which visual abnormalities have been categorized, have
Ocular Manifestations. MPA can affect all ocular and orbital tis- been reported in up to 8% of children with IgAV (Iannetti et al.
sues, though rarely. In a cohort of 85 patients with MPA from a 2012), though involvement of the eye is rare (Saulsbury 2001).
French vasculitis database, only one had ocular manifestations and Episcleritis, anterior uveitis, and keratitis have been reported
no details of the nature of the involvement were provided (Guillevin (Yamabe et al. 1988).
et al. 1999). The most common form of ocular involvement appears Hypocomplementaemic urticarial vasculitis
to be PUK with features that resemble Mooren’s ulcer (overhang-
Hypocomplementaemic Urticarial Vasculitis (HUV) (Anti-C1q
ing ulcerated edge) but for the presence of adjacent scleritis in
vasculitis) is a vasculitis of small vessels (capillaries, venules, and
MPA (Messmer and Foster 1999). Ocular disease may be the initial
arterioles) occurring concurrent with urticaria, hypocomple-
manifestation of MPA (Messmer and Foster 1999). A patient with
mentaemia, and the presence of anti-C1q antibodies (Jennette
hypopyon anterior uveitis and nerve fibre layer infarcts (thus prob-
et al. 2013).
ably representative of retinal vasculitis) has been reported in MPA
(Mihara et al. 2005), as has a patient with periocular skin lesions Ocular Manifestations. In two non-ophthalmic series of patients
(Caster et al. 1996). As with the other systemic vasculitides, treat- with HUV, 15 to 21% of patients were reported to have ocular
ment of ocular disease should be targeted to systemic disease with manifestations (Davis et al. 1998; Sanchez et al. 1982). Including
local adjunctive therapy as needed. case reports from the ophthalmic literature, conjunctivitis, uvei-
tis, episcleritis, and scleritis have all been reported in association
Immune complex small-vessel vasculitis with HUV and as with other systemic vasculitides, require systemic
Anti-glomerular basement membrane disease immunosuppression for control (Corwin and Baum 1982; Davis
Anti-glomerular basement membrane (anti-GBM) disease results et al. 1998; Ozcakar et al. 2013; Sanchez et al. 1982; Thorne et al.
from the formation of immune complexes of autoantibodies bound 2002; Wisnieski 2000; Wisnieski et al. 1995).
CHAPTER 11 ocular manifestations of systemic vasculitis 137

Variable-vessel vasculitis data differently than that from the US study, a survey of a large
population of patients seen at a single centre in Istanbul, Turkey
Behçet’s disease
over 18  years gives an interesting perspective on the disease
Behçet’s disease (BD) is a idiopathic inflammatory disorder the (Tugal-Tutkun et al. 2004). Of 880 patients 11% had anterior uvei-
original description of which included hypopyon anterior uveitis tis and 89% involvement of posterior segment, almost exactly the
as part of the triad of manifestations, oral and genital ulcerations same as the US study. Visual acuity at presentation was legally blind
being the other two. The underlying pathology in BD is an oblit- or worse in 42% of patients, a greater proportion than in the US
erative necrotizing vasculitis involving both arteries and veins. BD population, and 29% had what was described as irreversible vision
affects individuals with genetic origins along the ancient ‘Silk Road’, loss at presentation. An additional 15% developed new vision loss
stretching from Japan to the Eastern Mediterranean, most com- over the course of follow-up (Tugal-Tutkun et al. 2004). Extensive
monly. There appears to be a geographic effect, with individuals retinal vascular disease, with occlusions, is not uncommon.
from Western countries less commonly and less severely affected
(Kacmaz et al. 2008). Cogan’s syndrome
Ocular manifestations Cogan’s syndrome (CS) is a vasculitis targeting small, medium, or
Ocular involvement is present in approximately 70% of patients large arteries (including the aorta), with possible aortic and mitral
with BD (Verity et  al. 2003). Ocular manifestations may include valvulitis. CS is characterized by ocular inflammatory lesions, clas-
the classically described hypopyon anterior uveitis, retinal vascu- sically interstitial keratitis, wherein the normally clear and avascu-
litis with or without retinal infiltrates/retinitis (Figure  11.5a and lar corneal stroma is infiltrated by blood vessels from the limbus. In
b), optic neuropathy, scleritis, vitritis, and conjunctival ulcera- the ‘atypical’ form, uveitis, episcleritis, and scleritis may be present
tion (Kacmaz et al. 2008; Zamir et al. 2003). Recent data from the rather than interstitial keratitis. All forms present in association
Systemic Immunosuppressive Therapy For Eye Diseases Cohort with inner ear disease (Pagnini et al. 2012; Jennette et al. 2013).
Study Group (SITE Study) give a clear indication of the nature and
extent of BD in US patients (Kacmaz et al. 2008). Of 168 patients Single-organ vasculitis
with BD, 11% had anterior uveitis while 85% had uveitis involving Single-organ vasculitis is, as the name implies, a vasculitis of any
the posterior segment. Other forms of involvement, including scle- size artery or vein affecting a single organ, without evidence that it is
ritis, optic neuropathy, and orbital inflammation were present in simply the limited expression of a systemic vasculitis. The involved
5%. Caucasians made up 61 to 75% of patients. Vision at presenta- organ and vessel type should be included in the name, for example
tion differs between patients with anterior versus posterior disease, cutaneous small-vessel vasculitis, and arguably for the most part,
with 25% of the former having 20/50 or worse vision, 60% of the retinal vasculitis, discussed previously (Jennette et al. 2013).
latter. Vision of 20/200 or worse was present at presentation in 15
and 34%, respectively. Patients with BD had a 45% rate per eye year Cutaneous small-vessel vasculitis
of developing any sort of ocular complication; a12% rate per eye Cutaneous small-vessel vasculitis (CSVV) is also known as cutane-
year of progressing to 20/50 or worse vision; and a 9% rate per eye ous leukocytoclastic vasculitis, hypersensitivity vasculitis, allergic
year of progressing to 20/200 or worse. The most common reasons vasculitis, and necrotizing vasculitis. Ocular manifestations have
for loss of vision were inflammatory haze and cataract (Kacmaz been reported. Uveitis and retinal vasculitis with retinitis have
et al. 2008). As mentioned, BD is much more common outside the been reported in one patient each (Tsai et  al. 1993; Corwin and
US, especially in Asia and the Middle East. While reporting their Baum 1982), with local or systemic treatment with glucocorticoids

(a) (b)

Fig. 11.5 (a) Retinal vasculitis with associated haemorrhage and retinitis in a patient with Behçet’s disease. (b) Fluorescein angiography of same area as Figure 11.5a,
revealing hyperfluorescence along involved vessels, indicating active vasculitis.
138 SECTION 3 clinical manifestations common to vasculitis

sufficient to control the disease. Peripheral ulcerative keratitis has Rheumatoid arthritis
been reported (Keenan et al. 2011; Doutre et al. 1986) as has bilat-
The most common ocular manifestation of rheumatoid arthri-
eral episclertis (Bollinger et al. 2009).
tis (RA) is keratoconjunctivitis sicca. Scleritis is seen in 1–6% of
Erythema elevatum diutinum (EED) is a rare dermatosis con-
patients with RA and conversely RA is the most common systemic
sidered to be a variant of CSVV, which has been associated with
association found in patients with scleritis. Peripheral ulcerative
a variety of other autoimmune conditions, including seropositive
keratitis may also occur (McCarthy et al. 1992; Foster et al. 1984).
rheumatoid arthritis, ulcerative colitis, Crohn’s disease, granulo-
While most series report that 1% of patients with RA develop scle-
matosis with polyangiitis, relapsing polychondritis, pyoderma
ritis, 14% of patients with rheumatoid vasculitis do so (Foster et al.
gangrenosum, type 1 diabetes mellitus, and gluten-sensitive enter-
1984). Retinal microvasculopathy has been reported in patients
opathy (Lekhanont et  al. 2011). Eight cases of ocular manifes-
with rheumatoid vasculitis, but the occurrence is uncommon
tations in the setting of EED have been reported in the English
(Meyer et al. 1978).
literature, including four cases of peripheral ulcerative keratitis
(Casanova et al. 2001; Takiwaki et al. 1998), and one case each of Sarcoid
Terrien’s marginal degeneration (Shimazaki et al. 1998), inflam-
matory peripheral keratitis without corneal thinning (Aldave et al. Sarcoidosis is a granulomatous systemic disease affecting all tis-
2003), nodular scleritis with panuveitis (Mitamura et  al. 2004), sues and organ systems (with the exception of the adrenal glands,
and pseudopterygium with progressive keratolysis (Lekhanont which are rarely involved). While the pathogenesis is unknown,
et al. 2011). the hallmark of sarcoid is the presence of non-caseating granulo-
mas, suggesting either a foreign material or infectious agent as the
inciting cause, perhaps in the setting of a permissive immunologi-
Vasculitis associated with systemic disease cal background. In patients with sarcoidosis, ocular manifestations
Systemic lupus erythematosus occur in up to 60% (Herbort et al. 2009). A survey of 50 Caucasian
Systemic lupus erythematosus (SLE) can affect any portion of the patients with biopsy-proven lung sarcoidosis in Greece reported
eye, ocular adnexa, or periorbital tissue. Orbital involvement is episcleritis in 16%, granulomatous iris nodules in 18%, periphlebi-
rare and manifests, as in other cases of orbital inflammation, with tis in 16%, periarteritis in 16%, and branch retinal vein occlusion in
proptosis, orbital pain, and pain with and restriction of extraocu- 14% (Pefkianaki et al. 2011). ACCESS (A Case–Control Etiologic
lar movement. The most common ocular manifestation of SLE is Study of Sarcoidosis) reported that the incidence of sarcoidosis
keratoconjunctivitis sicca, the hallmark being a decreased pro- varies across ethnic groups, African Americans having the highest
duction of the aqueous tear film. Proinflammatory markers such rate (35–80 per 100 000), followed by northern Europeans (15–20
as IL-17 can be found in the tear film of SLE patients (Oh et al. per 100 000), European Americans (3–10 per 100 000), Southern
2011; Frith et  al. 1990). Disease may range from mild to severe, Europeans (1–5 per 100  000), and Japanese (1–2 per 100  000).
with the latter potentially resulting in serious surface complica- Differences also exist in the clinical presentation and disease course
tions including symblepharon formation, forniceal foreshorten- among various ethnic groups. African Americans are more likely to
ing, and exposure keratopathy. Episcleritis is a completely benign, present with extrathoracic involvement in addition to thoracic dis-
non-visually threatening inflammation of the superficial episcleral ease. European Americans and Northern Europeans typically expe-
venous plexus. In contrast, scleritis is an excruciatingly painful and rience an acute course. Japanese patients are most likely to present
vision-threatening condition. Corneal disease may occur, usually with ocular involvement. African American patients have frequent
secondary to KCS, but peripheral ulcerative keratitis may also occur involvement of the eyes. Women are more likely to have eye and
and may herald uncontrolled systemic disease. Lupus retinopathy neurological involvement than men (Baughman et al. 2001). When
occurs in up to 29% of patients with active systemic disease with the eye is involved, the anterior segment is the most commonly
a strong correlation between the development of retinopathy and affected, seen in 85% of patients with ocular sarcoidosis. Anterior
CNS disease (Stafford-Brady et  al. 1988). SLE patients may have segment manifestations include granulomas and cicatrizing con-
concomitant antiphospholipid antibodies, including anticardi- junctivitis; anterior uveitis, usually granulomatous and chronic;
olipin and lupus anticoagulant. Positive antiphospholipid antibody iris nodules; band keratopathy and scleritis. The posterior segment
titres were found in 77% of patients with SLE and retinal involve- may be involved in 25% of patients and include vitritis, interme-
ment versus only 29% of SLE patients without retinal disease had diate uveitis, posterior uveitis, and panuveitis. Periphlebitis mani-
positive titres (Montehermoso et al. 1999). The presence of these fests as yellow perivenous exudates. Other manifestations include
antibodies increases the risk of occlusive vascular disease. Lupus choroidal nodules and exudative retinal detachment (Bonfioli and
choroidopathy is a rare ocular manifestation thought to be due Orefice 2005).
to uncontrolled hypertension (Hannouche et  al. 1995), immune
complex deposition in the choriocapillaris (Schwartz and Roberts
References
1983), and/or antiretinal pigment epithelium antibodies (Matsuo Akova, Y.A., Jabbur, N.S., and Foster, C.S. (1993). Ocular presentation of
et al. 1987). It is a marker of disease activity and thus should proba- polyarteritis nodosa. Clinical course and management with steroid and
cytotoxic therapy. Ophthalmology, 100, 1775–81.
bly be considered an indication for aggressive immunosuppression.
Albini, T., Zamir, E., Read, R., Smith, R., See, R., and Rao, N. (2005).
The optic nerve may be affected with development of optic neuritis Evaluation of subconjunctival triamcinolone for nonnecrotizing ante-
and/or ischaemic optic neuropathy. Eye movement abnormalities rior scleritis. Ophthalmology, 112, 1814–20.
have been reported in almost one-third of patients with SLE, usu- Aldave, A.J., Shih, J.L., Jovkar, S., and Mcleod, S.D. (2003). Peripheral kera-
ally secondary to ischaemic microvascular disease of the brainstem titis associated with erythema elevatum diutinum. American Journal
(Keane 1995). ofOphthalmology, 135, 389–90.
CHAPTER 11 ocular manifestations of systemic vasculitis 139

Ameli, F., Phang, K.S., and Masir, N. (2011). Churg-Strauss syndrome pre- Darrell, R.W., Wagener, H.P., and Kurland, L.T. (1962). Epidemiology of uvei-
senting with conjunctival and eyelid masses:  a case report. Medical tis. Incidence and prevalence in a small urban community. Archives of
Journal of Malaysia, 66, 517–9. Ophthalmology, 68, 502–14.
Arida, A., Kyprianou, M., Kanakis, M., and Sfikakis, P.P. (2010). The diag- Davis, M.D., Daoud, M.S., Kirby, B., Gibson, L.E., and Rogers, R.S., 3rd
nostic value of ultrasonography-derived edema of the temporal artery (1998). Clinicopathologic correlation of hypocomplementemic and
wall in giant cell arteritis: a second meta-analysis. BMC Musculoskeletal normocomplementemic urticarial vasculitis. Journal of the American
Disorders, 11, 44. Academy of Dermatology, 38, 899–905.
Ausayakhun, S., Louthrenoo, W., and Aupapong, S. (2002). Ocular diseases Doutre, M.S., Beylot, C., Morel, P., Dallot, A., Lagoutte, F., Beylot, J., and
in patients with rheumatic diseases. Journal of the Medical Association of Bioulac, P. (1986). [Ophthalmologic manifestations of leukocytoclastic
Thailand, 85, 855–62. vasculitis. Apropos of 3 case reports]. Annales de Dermatologie et de
Baughman, R.P., Teirstein, A.S., Judson, M.A., Rossman, M.D., Yeager, H. Venereologie, 113, 419–25.
Jr., Bresnitz, E.A., Depalo, L., Hunninghake, G., Iannuzzi, M.C., Johns, Falk, R.J., Gross, W.L., Guillevin, L., Hoffman, G., Jayne, D.R., Jennette, J.C.,
C.J., McLennan, G., Moller, D.R., Newman, L.S., Rabin, D.L., Rose, C., Kallenberg, C.G., Luqmani, R., Mahr, A.D., Matteson, E.L., Merkel, P.A.,
Rybicki, B., Weinberger, S.E., Terrin, M.L., Knatterud, G.L., Cherniak, R., Specks, U., and Watts, R. (2011a). Granulomatosis with polyangiitis
and Case Control Etiologic Study Of Sarcoidosis Research Group (2001). (Wegener’s): an alternative name for Wegener’s granulomatosis. Annals
Clinical characteristics of patients in a case control study of sarcoidosis. of the Rheumatic Diseases, 70, 704.
American Journal of Respiratory and Critical Care Medicine, 164, 1885–9. Falk, R.J., Gross, W.L., Guillevin, L., Hoffman, G., Jayne, D.R., Jennette, J.C.,
Birnbaum, A.D., Jiang, Y., Vasaiwala, R., Tessler, H.H., and Goldstein, D.A. Kallenberg, C.G., Luqmani, R., Mahr, A.D., Matteson, E.L., Merkel, P.A.,
(2012). Bilateral simultaneous-onset nongranulomatous acute anterior Specks, U., and Watts, R. (2011b). Granulomatosis with polyangiitis
uveitis:  clinical presentation and etiology. Archives of Ophthalmology, (Wegener’s): an alternative name for Wegener’s granulomatosis. Journal
130, 1–6. of the American Society of Nephrology, 22, 587–8.
Bley, T.A., Reinhard, M., Hauenstein, C., Markl, M., Warnatz, K., Hetzel, A., Falk, R.J., Gross, W.L., Guillevin, L., Hoffman, G.S., Jayne, D.R., Jennette, J.C.,
Uhl, M., Vaith, P., and Langer, M. (2008). Comparison of duplex sonog- Kallenberg, C.G., Luqmani, R., Mahr, A.D., Matteson, E.L., Merkel, P.A.,
raphy and high-resolution magnetic resonance imaging in the diagnosis Specks, U., Watts, R.A., American College of Rheumatology, American
of giant cell (temporal) arteritis. Arthritis and Rheumatism, 58, 2574–8. Society of Nephrology and European League Against Rheumatism
Bollinger, K., Medina, C., and Perez, V.L. (2009). Bilateral episcleritis (2011c). Granulomatosis with polyangiitis (Wegener’s):  an alterna-
as a manifestation of cutaneous leukocytoclastic vasculitis. Ocular tive name for Wegener’s granulomatosis. Arthritis and Rheumatism,
Immunology and Inflammation, 17, 23–5. 63, 863–4.
Bonfioli, A.A. and Orefice, F. (2005). Sarcoidosis. Seminars in Ophthalmology, Font, R.L., Mehta, R.S., Streusand, S.B., O’Boyle, T.E., and Kretzer, F.L. (1983).
20, 177–82. Bilateral retinal ischemia in Kawasaki disease. Postmortem findings and
Borchers, A.T. and Gershwin, M.E. (2012). Giant cell arteritis:  a review electron microscopic observations. Ophthalmology, 90, 569–77.
of classification, pathophysiology, geoepidemiology and treatment. Ford, R.G. and Siekert, R.G. (1965). Central nervous system manifestations
Autoimmunity Reviews, 11, A544–54. of periarteritis nodosa. Neurology, 15, 114–22.
Boyev, L.R., Miller, N.R., and Green, W.R. (1999). Efficacy of unilateral versus Foster, C.S., Forstot, S.L., and Wilson, L.A. (1984). Mortality rate in rheu-
bilateral temporal artery biopsies for the diagnosis of giant cell arteritis. matoid arthritis patients developing necrotizing scleritis or periph-
American Journal of Ophthalmology, 128, 211–5. eral ulcerative keratitis. Effects of systemic immunosuppression.
Breuer, G., Nesher, R., and Nesher, G. (2009a). Effect of biopsy length on Ophthalmology, 91, 1253–63.
the rate of positive temporal artery biopsies. Clinical and Experimental Frith, P., Burge, S.M., Millard, P.R., and Wojnarowska, F. (1990). External
Rheumatology, 27(Suppl. 52), S10–13. ocular findings in lupus erythematosus: a clinical and immunopatho-
Breuer, G.S., Nesher, G., and Nesher, R. (2009b). Rate of discordant findings logical study. British Journal of Ophthalmology, 74, 163–7.
in bilateral temporal artery biopsy to diagnose giant cell arteritis. Journal Ghanchi, F.D. and Dutton, G.N. (1997). Current concepts in giant cell (tem-
of Rheumatology, 36, 794–6. poral) arteritis. Survey of Ophthalmology, 42, 99–123.
Casanova, F.H., Meirelles, R.L., Tojar, M., Martins, M.C., Rigueiro, M.P., and Golnik, K.C. (2004). Neuro-ophthalmologic manifestations of systemic
De Freitas, D. (2001). Autoimmune keratolysis in a patient with leuko- disease: rheumatologic/Inflammatory. Ophthalmology Clinics of North
cytoclastic vasculitis: unusual erythema elevatum diutinum with granu- America, 17, 389–96, vi.
lomatous pattern. Cornea, 20, 329–32. Gonzalez, E.B., Varner, W.T., Lisse, J.R., Daniels, J.C., and Hokanson, J.A.
Caster, J.C., Shetlar, D.J., Pappolla, M.A., and Yee, R.W. (1996). Microscopic (1989). Giant-cell arteritis in the southern United States. An 11-year
polyangiitis with ocular involvement. Archives of Ophthalmology, retrospective study from the Texas Gulf Coast. Archives of Internal
114, 346–8. Medicine, 149, 1561–5.
Cid, M.C., Font, C., Oristrell, J., De La Sierra, A., Coll-Vinent, B., Lopez-Soto, Gonzalez-Gay, M.A., Garcia-Porrua, C., Llorca, J., Hajeer, A.H., Branas, F.,
A., Vilaseca, J., Urbano-Marquez, A., and Grau, J.M. (1998). Association Dababneh, A., Gonzalez-Louzao, C., Rodriguez-Gil, E., Rodriguez-Ledo,
between strong inflammatory response and low risk of developing visual P., and Ollier, W.E. (2000). Visual manifestations of giant cell arteritis.
loss and other cranial ischemic complications in giant cell (temporal) Trends and clinical spectrum in 161 patients. Medicine, 79, 283–92.
arteritis. Arthritis and Rheumatism, 41, 26–32. Gonzalez-Gay, M.A., Martinez-Dubois, C., Agudo, M., Pompei, O., Blanco,
Cioffi, G.A., Granstam, E., and Alm, A. (2003). Ocular circulation. In Adler’s R., and Llorca, J. (2010). Giant cell arteritis: epidemiology, diagnosis,
Physiology of the Eye, 10th edn. (ed. P.L. Kaufman and A. Alm). Mosby, and management. Current Rheumatology Reports, 12, 436–42.
St. Louis. Gritz, D.C. and Wong, I.G. (2004). Incidence and prevalence of uveitis in
Cohen, S.M., Kokame, G.T., and Gass, J.D. (1996). Paraproteinemias asso- northern California: The northern California epidemiology of uveitis
ciated with serous detachments of the retinal pigment epithelium and study. Ophthalmology, 111, 491–500.
neurosensory retina. Retina, 16, 467–73. Guillevin, L., Durand-Gasselin, B., Cevallos, R., Gayraud, M., Lhote,
Corwin, J.M. and Baum, J. (1982). Iridocyclitis in two patients with hypocom- F., Callard, P., Amouroux, J., Casassus, P., and Jarrousse, B. (1999).
plementemic cutaneous vasculitis. American Journal of Ophthalmology, Microscopic polyangiitis: clinical and laboratory findings in eighty-five
94, 111–3. patients. Arthritis and Rheumatism, 42, 421–30.
Danesh-Meyer, H.V., Savino, P.J., Eagle, R.C. Jr., Kubis, K.C., and Sergott, Guillevin, L., Lhote, F., and Gherardi, R. (1997). Polyarteritis nodosa, micro-
R.C. (2000). Low diagnostic yield with second biopsies in suspected scopic polyangiitis, and Churg-Strauss syndrome: clinical aspects, neu-
giant cell arteritis. Journal of Neuro-ophthalmology, 20, 213–5. rologic manifestations, and treatment. Neurologic Clinics, 15, 865–86.
140 SECTION 3 clinical manifestations common to vasculitis

Hannouche, D., Korobelnik, J.F., Cochereau, I., Hayem, G., Beaudreuil, J., Karam, E.Z., Muci-Mendoza, R., and Hedges, T.R. 3rd (1999). Retinal find-
Meyer, O., and Hoang-Xuan, T. (1995). Systemic lupus erythemato- ings in Takayasu’s arteritis. Acta Ophthalmologica Scandinavica, 77,
sus with choroidopathy and serous retinal detachment. International 209–13.
Ophthalmology, 19, 125–7. Keane, J.R. (1995). Eye movement abnormalities in systemic lupus erythema-
Harman, L.E. and Margo, C.E. (1998). Wegener’s granulomatosis. Survey of tosus. Archives of Neurology, 52, 1145–9.
Ophthalmology, 42, 458–80. Kedhar, S.R., Belair, M.L., Jun, A.S., Sulkowski, M., and Thorne, J.E. (2007).
Haynes, B.F., Fishman, M.L., Fauci, A.S., and Wolff, S.M. (1977). The ocular Scleritis and peripheral ulcerative keratitis with hepatitis C virus-related
manifestations of Wegener’s granulomatosis. Fifteen years experience cryoglobulinemia. Archives of Ophthalmology, 125, 852–3.
and review of the literature. American Journal of Medicine, 63, 131–41. Keenan, J.D., Mandel, M.R., and Margolis, T.P. (2011). Peripheral ulcera-
Hayreh, S.S. (1998). Masticatory muscle pain: an important indicator of giant tive keratitis associated with vasculitis manifesting asymmetrically as
cell arteritis. Special Care in Dentistry, 18, 60–5. fuchs superficial marginal keratitis and terrien marginal degeneration.
Hayreh, S.S., Podhajsky, P.A., and Zimmerman, B. (1998a). Occult giant cell Cornea, 30, 825–7.
arteritis:  ocular manifestations. American Journal of Ophthalmology, Klien, B.A. (1968). Ischemic infarcts of the choroid (Elschinig spots). A
125, 521–6. cause of retinal separation in hypertensive disease with renal insuf-
Hayreh, S.S., Podhajsky, P.A., and Zimmerman, B. (1998b). Ocular manifes- ficiency. A  clinical and histopathologic study. American Journal of
tations of giant cell arteritis. American Journal of Ophthalmology, 125, Ophthalmology, 66, 1069–74.
509–20. Kohler, U. (1988). [Unusual course of Goodpasture syndrome]. Klinische
Herbort, C.P., Rao, N.A., Mochizuki, M., and Members Of Scientific Monatsblätter für Augenheilkunde, 192, 354–7.
Committee Of First International Workshop On Ocular Sarcoidosis Kremer, I. and Blumenthal, M. (1997). Excimer phototherapeutic keratec-
(2009). International criteria for the diagnosis of ocular sarcoido- tomy for corneal subepithelial cryoglobulin deposits. Journal of Cataract
sis: results of the first International Workshop On Ocular Sarcoidosis and Refractive Surgery, 23, 1119–21.
(IWOS). Ocular Immunology and Inflammation, 17, 160–9. Kremer, I., Wright, P., Merin, S., Weiss, J., Pick, A.I., and Kaufman, H. (1989).
Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., Hallahan, C.W., Lebovics, R.S., Travis, Corneal subepithelial monoclonal kappa IgG deposits in essential cryo-
W.D., Rottem, M., and Fauci, A.S. (1992). Wegener granulomatosis: an globulinaemia. British Journal of Ophthalmology, 73, 669–73.
analysis of 158 patients. Annals of Internal Medicine, 116, 488–98. Ku, J.H., Ali, A., Suhler, E.B., Choi, D., and Rosenbaum, J.T. (2012).
Hsu, C.T., Kerrison, J.B., Miller, N.R., Goldberg, M.F. (2001). Choroidal Characteristics and visual outcome of patients with retinal vasculitis.
infarction, anterior ischemic optic neuropathy, and central retinal artery Archives of Ophthalmology, 130, 1–6.
occlusion from polyarteritis nodosa. Retina, 21, 348–51. Kubal, A.A. and Perez, V.L. (2010). Ocular manifestations of ANCA-associated
Iannetti, L., Zito, R., Bruschi, S., Papetti, L., Ulgiati, F., Nicita, F., Del Balzo, vasculitis. Rheumatic Diseases Clinics of North America, 36, 573–86.
F., and Spalice, A. (2012). Recent understanding on diagnosis and man- Lamprecht, P. and Gross, W.L. (2004). A brief history of Wegener’s granu-
agement of central nervous system vasculitis in children. Clinical and lomatosis:  on limited, localized, and generalized forms of the dis-
Developmental Immunology, 2012, 698327. ease:  comment on the article by the Wegener’s Granulomatosis
Jabs, D.A., Mudun, A., Dunn, J.P., and Marsh, M.J. (2000a). Episcleritis and Etanercept Trial Research Group. Arthritis and Rheumatism, 50, 334–5;
scleritis:  clinical features and treatment results. American Journal of author reply 335–6.
Ophthalmology, 130, 469–76. Lekhanont, K., Patarakittam, T., Mantachote, K., Waiyawatjamai, P., and
Jabs, D.A., Nussenblatt, R.B., Rosenbaum, J.T., and Standardization of Vongthongsri, A. (2011). Progressive keratolysis with pseudopterygium
Uveitis Nomenclature Working Group (2005). Standardization of uveitis associated with erythema elevatum diutinum. Ophthalmology, 118,
nomenclature for reporting clinical data. Results of the First International 927–33.
Workshop. American Journal of Ophthalmology, 140, 509–16. Margolis, R., Kosmorsky, G.S., Lowder, C.Y., and Schoenfield, L. (2007).
Jabs, D.A., Rosenbaum, J.T., Foster, C.S., Holland, G.N., Jaffe, G.J., Louie, J.S., Conjunctival involvement in Churg-Strauss syndrome. Ocular
Nussenblatt, R.B., Stiehm, E.R., Tessler, H., Van Gelder, R.N., Whitcup, Immunology and Inflammation, 15, 113–5.
S.M., and Yocum, D. (2000b). Guidelines for the use of immunosuppres- Matsuo, T., Nakayama, T., Koyama, T., and Matsuo, N. (1987). Multifocal
sive drugs in patients with ocular inflammatory disorders: recommenda- pigment epithelial damages with serous retinal detachment in systemic
tions of an expert panel. American Journal of Ophthalmology, 130, 492–513. lupus erythematosus. Ophthalmologica, 195, 97–102.
Jacob, J.L., Polomeno, R.C., Chad, Z., and Lapointe, N. (1982). Ocular mani- Mauriello, J.A. Jr., Stabile, C., and Wagner, R.S. (1986). Dacryocystitis follow-
festations of Kawasaki disease (mucocutaneous lymph node syndrome). ing Kawasaki’s disease. Ophthalmic Plastic and Reconstructive Surgery,
Canadian Journal of Ophthalmology, 17, 199–202. 2, 209–11.
Jennette, J.C., Falk, R.J., Andrassy, K., Bacon, P.A., Churg, J., Gross, W.L., McCarthy, J.M., Dubord, P.J., Chalmers, A., Kassen, B.O., and Rangno,
Hagen, E.C., Hoffman, G.S., Hunder, G.G., Kallenberg, C.G., et  al. K.K. (1992). Cyclosporine A for the treatment of necrotizing scleritis
(1994). Nomenclature of systemic vasculitides. Proposal of an interna- and corneal melting in patients with rheumatoid arthritis. Journal of
tional consensus conference. Arthritis and Rheumatism, 37, 187–92. Rheumatology, 19, 1358–61.
Jennette, J., Falk, R., Bacon, P., Basu, N., Cid, M., Ferrario, F., Flores-Suarez, L., Messmer, E.M. and Foster, C.S. (1999). Vasculitic peripheral ulcerative kera-
Gross, W., Guillevin, L., Hagen, E., Hoffman, G., Jayne, D., Kallenberg, titis. Survey of Ophthalmology, 43, 379–96.
C., Lamprecht, P., Langford, C., Luqmani, R., Mahr, A., Matteson, Meyer, E., Scharf, J., Miller, B., Zonis, S., and Nahir, M. (1978). Fundus lesions
E., Merkel, P., Ozen, S., Pusey, C., Rasmussen, N., Rees, A., Scott, D., in rheumatoid arthritis. Annals of Ophthalmology, 10, 1583–4.
Specks, U., Stone, J., Takahashi, K., and Watts, R. (2013). 2012 Revised Mihara, M., Hayasaka, S., Watanabe, K., Kitagawa, K., and Hayasaka, Y.
International Chapel Hill Consensus Conference nomenclature of vas- (2005). Ocular manifestations in patients with microscopic polyangiitis.
culitides. Arthritis and Rheumatism, 65, 1–11. European Journal of Ophthalmology, 15, 138–42.
Kacmaz, R.O., Kempen, J.H., Newcomb, C., Gangaputra, S., Daniel, E., Mitamura, Y., Fujiwara, O., Miyanishi, K., et  al. (2004). Nodular scleritis
Levy-Clarke, G.A., Nussenblatt, R.B., Rosenbaum, J.T., Suhler, E.B., and panuveitis with erythema elevatum diutinum. American Journal of
Thorne, J.E., Jabs, D.A., Foster, C.S., and Systemic Immunosuppressive Ophthalmology, 137, 368–70.
Therapy For Eye Diseases Cohort Study Group (2008). Ocular inflam- Montehermoso, A., Cervera, R., Font, J., Ramos-Casals, M., Garcia-Carrasco,
mation in Behcet disease: incidence of ocular complications and of loss M., Formiga, F., Callejas, J.L., Jorfan, M., Grino, M.C., and Ingelmo,
of visual acuity. American Journal of Ophthalmology, 146, 828–36. M. (1999). Association of antiphospholipid antibodies with retinal
CHAPTER 11 ocular manifestations of systemic vasculitis 141

vascular disease in systemic lupus erythematosus. Seminars in Arthritis Rosenbaum, J.T., Ku, J., Ali, A., Choi, D., and Suhler, E.B. (2012). Patients
and Rheumatism, 28, 326–32. with retinal vasculitis rarely suffer from systemic vasculitis. Seminars in
Morgan, C.M., Foster, C.S., D’Amico, D.J., and Gragoudas, E.S. (1986). Arthritis and Rheumatism, 41, 859–65.
Retinal vasculitis in polyarteritis nodosa. Retina, 6, 205–9. Sainz de la Maza, M., Molina, N., Gonzalez-Gonzalez, L.A., Doctor, P.P.,
Myers, J.P., Di Bisceglie, A.M., and Mann, E.S. (2001). Cryoglobulinemia Tauber, J., and Foster, C.S. (2012). Clinical characteristics of a large cohort
associated with Purtscher-like retinopathy. American Journal of of patients with scleritis and episcleritis. Ophthalmology, 119, 43–50.
Ophthalmology, 131, 802–4. Sanchez, N.P., Winkelmann, R.K., Schroeter, A.L., and Dicken, C.H. (1982).
Netter, F.H. (1989). Atlas of human anatomy. Ciba-Geigy Corporation, The clinical and histopathologic spectrums of urticarial vasculitis: study of
Summit, New Jersey. forty cases. Journal of the American Academy of Dermatology, 7, 599–605.
Oh, J.Y., Kim, M.K., Choi, H.J., Ko, J.H., Kang, E.J., Lee, H.J., Wee, W.R., Saulsbury, F.T. (2001). Henoch-Schonlein purpura. Current Opinion in
and Lee, J.H. (2011). Investigating the relationship between serum Rheumatology, 13, 35–40.
interleukin-17 levels and systemic immune-mediated disease in patients Schmidt, W.A., Kraft, H.E., Vorpahl, K., Volker, L., and Gromnica-Ihle, E.J.
with dry eye syndrome. Korean Journal of Ophthalmology, 25, 73–6. (1997). Color duplex ultrasonography in the diagnosis of temporal arte-
Ohno, S., Miyajima, T., Higuchi, M., Yoshida, A., Matsuda, H., Saheki, Y., ritis. New England Journal of Medicine, 337, 1336–42.
Nagamatsu, I., Togashi, T., and Matsumoto, S. (1982). Ocular manifes- Schwartz, M.M. and Roberts, J.L. (1983). Membranous and vascular choroi-
tations of Kawasaki’s disease (mucocutaneous lymph node syndrome). dopathy: two patterns of immune deposits in systemic lupus erythema-
American Journal of Ophthalmology, 93, 713–7. tosus. Clinical Immunology and Immunopathology, 29, 369–80.
Ozcakar, Z.B., Yalcinkaya, F., Altugan, F.S., Kavaz, A., Ensari, A., and Ekim, Shimazaki, J., Yang, H.Y., Shimmura, S., and Tsubota, K. (1998). Terrien’s
M. (2013). Hypocomplementemic urticarial vasculitis syndrome in marginal degeneration associated with erythema elevatum diutinum.
three siblings. Rheumatology International, 33, 763–6. Cornea, 17, 342–4.
Pagnini, I., Zannin, M.E., Vittadello, F., Sari, M., Simonini, G., Cimaz, R., and Sinico, R.A., Di Toma, L., Maggiore, U., Tosoni, C., Bottero, P., Sabadini,
Zulian, F. (2012). Clinical features and outcome of Cogan syndrome. E., Giammarresi, G., Tumiati, B., Gregorini, G., Pesci, A., Monti, S.,
Journal of Pediatrics, 160, 303–7 e1. Balestrieri, G., Garini, G., Vecchio, F., and Buzio, C. (2006). Renal
Pagnoux, C., Seror, R., Henegar, C., Mahr, A., Cohen, P., Le Guern, V., involvement in Churg-Strauss syndrome. American Journal of Kidney
Bienvenu, B., Mouthon, L., Guillevin, L., and French Vasculitis Study Diseases, 47, 770–9.
Group (2010). Clinical features and outcomes in 348 patients with pol- Smith, J.R., Mackensen, F., and Rosenbaum, J.T. (2007). Therapy insight: scle-
yarteritis nodosa: a systematic retrospective study of patients diagnosed ritis and its relationship to systemic autoimmune disease. Nature Clinical
between 1963 and 2005 and entered into the French Vasculitis Study Practice Rheumatology, 3, 219–26.
Group database. Arthritis and Rheumatism, 62, 616–26. Sohn, E.H., Wang, R., Read, R.W., Roufas, A., Teo, L., Moorthy, R., Albini, T.,
Pagnoux, C., Stubbe, M., Lifermann, F., Decaux, O., Pavic, M., Berezne, Vasconcelos-Santos, D.V., Dustin, L.D., Zamir, E., Chee, S.P., Mccluskey,
A., Delacroix-Szmania, I., Meaux-Ruault, N., Bienvenu, B., Cabane, J., P., Smith, R., and Rao, N. (2011). Long-term, multicenter evaluation of
and Guillevin, L. (2011). Wegener’s granulomatosis strictly and per- subconjunctival injection of triamcinolone for non-necrotizing, nonin-
sistently localized to one organ is rare: assessment of 16 patients from fectious anterior scleritis. Ophthalmology, 118, 1932–7.
the French Vasculitis Study Group database. Journal of Rheumatology, Stafford-Brady, F.J., Urowitz, M.B., Gladman, D.D., and Easterbrook, M.
38, 475–8. (1988). Lupus retinopathy. Patterns, associations, and prognosis.
Panja, M. and Mondal, P.C. (2004). Current status of aortoarteritis in India. Arthritis and Rheumatism, 31, 1105–10.
Journal of the Association of Physicians of India, 52, 48–52. Suhler, E., Lloyd, M., Choi, D., Rosenbaum, J., and Austin, D. (2008). Incidence
Pefkianaki, M., Androudi, S., Praidou, A., Sourlas, V., Zakynthinos, E., and prevalence of uveitis in Veterans Affairs Medical Centers of the
Brazitikos, P., Gourgoulianis, K., and Daniil, Z. (2011). Ocular dis- Pacific Northwest. American Journal of Ophthalmology, 146, 890–6.
ease awareness and pattern of ocular manifestation in patients with Takanashi, T., Uchida, S., Arita, M., Okada, M., and Kashii, S. (2001).
biopsy-proven lung sarcoidosis. Journal of Ophthalmic Inflammation Orbital inflammatory pseudotumor and ischemic vasculitis in
and Infection, 1, 141–5. Churg-Strauss syndrome: report of two cases and review of the litera-
Perez, V.L., Chavala, S.H., Ahmed, M., Chu, D., Zafirakis, P., Baltatzis, S., ture. Ophthalmology, 108, 1129–33.
Ocampo, V., and Foster, C.S. (2004). Ocular manifestations and concepts Takiwaki, H., Kubo, Y., Tsuda, H., et al. (1998). Peripheral ulcerative kera-
of systemic vasculitides. Survey of Ophthalmology, 49, 399–418. titis associated with erythema elevatum diutinum and a positive rheu-
Perumal, B., Black, E.H., Levin, F., and Servat, J.J. (2012). Non-infectious matoid factor: a report of three cases. British Journal of Ophthalmology,
orbital vasculitides. Eye (Lond), 26, 630–9. 138, 893–7.
Peter, J., David, S., Danda, D., Peter, J.V., Horo, S., and Joseph, G. (2011). Telander, D.G., Holland, G.N., Wax, M.B., and Van Gelder, R.N. (2006).
Ocular manifestations of Takayasu arteritis. Retina, 31, 1170–8. Rubeosis and anterior segment ischemia associated with systemic cryo-
Pless, M., Rizzo, J.F. 3rd, Lamkin, J.C., and Lessell, S. (2000). Concordance globulinemia. American Journal of Ophthalmology, 142, 689–90.
of bilateral temporal artery biopsy in giant cell arteritis. Journal of Thorne, J.E., Hernandez, M.I., Rencic, A., and Nousari, H.C. (2002). Severe
Neuro-ophthalmology, 20, 216–8. scleritis and urticarial lesions. American Journal of Ophthalmology,
Power, W.J., Rodriguez, A., Neves, R.A., Lane, L., and Foster, C.S. (1995). 134, 932–4.
Disease relapse in patients with ocular manifestations of Wegener gran- Thorne, J.E. and Jabs, D.A. (2001). Ocular manifestations of vasculitis.
ulomatosis. Ophthalmology, 102, 154–60. Rheumatic Diseases Clinics of North America, 27.
Reeves, S.W., Sloan, F.A., Lee, P.P., and Jaffe, G.J. (2006). Uveitis in the Tovilla-Canales, J.L. (1998). Ocular manifestations of giant cell arteritis.
elderly: epidemiological data from the National Long-term Care Survey Current Opinion in Ophthalmology, 9, 73–9.
Medicare Cohort. Ophthalmology, 113, 307 e1. Tsai, J.C., Forster, D.J., Ober, R.R., and Rao, N.A. (1993). Panuveitis and
Riono, W.P., Hidayat, A.A., and Rao, N.A. (1999). Scleritis: a clinicopatho- multifocal retinitis in a patient with leucocytoclastic vasculitis. British
logic study of 55 cases. Ophthalmology, 106, 1328–33. Journal of Ophthalmology, 77, 318–20.
Rodriguez, A., Calonge, M., Pedroza-Seres, M., Akova, Y.A., Messmer, E.M., Tugal-Tutkun, I., Onal, S., Altan-Yaycioglu, R., Huseyin Altunbas, H., and
D’Amico, D.J., and Foster, C.S. (1996). Referral patterns of uveitis in a Urgancioglu, M. (2004). Uveitis in Behcet disease:  an analysis of 880
tertiary eye care center. Archives of Ophthalmology, 114, 593–9. patients. American Journal of Ophthalmology, 138, 373–80.
142 SECTION 3 clinical manifestations common to vasculitis

Verity, D.H., Wallace, G.R., Vaughan, R.W., and Stanford, M.R. (2003). Yagci, A. (2012). Update on peripheral ulcerative keratitis. Clinics in
Behcet’s disease:  from Hippocrates to the third millennium. British Ophthalmology, 6, 747–54.
Journal of Ophthalmology, 87, 1175–83. Yamabe, H., Ozawa, K., Fukushi, K., Kubota, H., Ohsawa, H., Akitsu, H.,
Watson, P.G., and Hayreh, S.S. (1976). Scleritis and episcleritis. British Journal Chiba, N., Seino, S., Miyata, M., Onodera, K., et al. (1988). IgA nephrop-
of Ophthalmology, 60, 163–91. athy and Henoch-Schonlein purpura nephritis with anterior uveitis.
Watts, R.A., Carruthers, D.M., and Scott, D.G.I. (1995). Epidemiology of sys- Nephron, 50, 368–70.
temic vasculitis: changing incidence or definition? Seminars in Arthritis Zamir, E., Bodaghi, B., Tugal-Tutkun, I., See, R.F., Charlotte, F., Wang, R.C.,
and Rheumatism, 25, 28–34. Wechsler, B., Lehoang, P., Anteby, I., and Rao, N.A. (2003). Conjunctival
Wisnieski, J.J. (2000). Urticarial vasculitis. Current Opinion in Rheumatology, ulcers in Behcet’s disease. Ophthalmology, 110, 1137–41.
12, 24–31. Zamir, E., Read, R.W., Smith, R.E., Wang, R.C., and Rao, N.A. (2002). A pro-
Wisnieski, J.J., Baer, A.N., Christensen, J., Cupps, T.R., Flagg, D.N., Jones, spective evaluation of subconjunctival injection of triamcinolone ace-
J.V., Katzenstein, P.L., Mcfadden, E.R., Mcmillen, J.J., Pick, M.A., et al. tonide for resistant anterior scleritis. Ophthalmology, 109, 798–805;
(1995). Hypocomplementemic urticarial vasculitis syndrome. Clinical discussion 805–7.
and serologic findings in 18 patients. Medicine (Baltimore), 74, 24–41.
CHAPTER 12

Cardiopulmonary
manifestations of vasculitis
Stephen K. Frankel and Marvin I. Schwarz

Introduction 1992; Lynch et al. 2004). Furthermore, upper and lower respiratory
tract disease are independent risk factors for disease relapse after
Cardiopulmonary disease occurs often in systemic vasculitis, con- remission (Hogan et al. 2005). Upper respiratory tract disease is
tributing significantly to morbidity and mortality. The clinical pres- also characteristic of EGPA (approximately 70%) and presents as
entation is highly variable and may range from an abnormal chest allergic or eosinophilic rhinitis and/or sinusitis (Guillevin 1999a;
radiograph in an asymptomatic patient to a catastrophic event such Guillevin et al. 1988; Lanham et al. 1984). Upper respiratory tract
as diffuse alveolar haemorrhage (DAH) or sudden death. Since involvement is much less common in microscopic polyangitiis,
patients can first present with isolated (or seemingly isolated) occurring in fewer than 15% of patients.
pulmonary or cardiac disease, there may be confusion with more
common entities such as malignancy, infection, connective tissue Asthma and airways disease
disease, drug toxicity, and embolic phenomena. Alternatively, car- Asthma is present in almost all patients with EGPA. While the
diopulmonary manifestations may arise later in the disease course, duration and severity of the asthma is variable, it generally pre-
or there can be the simultaneous onset of cardiopulmonary mani- cedes the onset of the systemic vasculitis by several years (an aver-
festations with other systemic manifestations of disease. Both car- age of 7–8 years) and is frequently severe and/or steroid-requiring
diovascular and pulmonary involvement have been associated with (Guillevin 1999a; Solans et  al. 2001; Guillevin et  al. 1996). That
an increased risk for disease relapse in ANCA-associated vasculitis said, although maturity-onset asthma and severe asthma are more
(Pagnoux et al. 2008; Walsh et al. 2012; Hogan et al. 2005; Mukhtyar common in EGPA, the asthma in these patients otherwise remains
et al. 2008). Cardiac involvement in eosinophilic granulomatosis indistinguishable from chronic asthma. While systemic corticos-
with polyangiitis (EGPA, formerly Churg–Strauss syndrome) car- teroids represent effective therapy for severe asthma, the long-term
ries an increased risk of death, and there is evidence that lung dis- side-effects associated with oral corticosteroids make it desirable to
ease in patients with granulomatosis with polyangiitis also carries minimize the total cumulative dose required to control all aspects
a worse prognosis (Guillevin et  al. 1996; Guillevin et  al. 1999a; of the disease. The introduction of a steroid-sparing cytotoxic
Guillevin et al. 2011; Reinhold-Keller et al. 2000). agent may be indicated to control the ‘vasculitic’ manifestations of
EGPA (see Chapter 32); however, an equally aggressive approach
The scope of cardiopulmonary needs to be taken in optimizing therapies for the management of
complications severe asthma and sinus disease. Management strategies that may
help with the control of the severe asthma in EGPA patients may
Upper respiratory tract disease include high-dose inhaled corticosteroids, long-acting bronchodi-
Chronic rhinitis and sinusitis are common medical problems lator therapy, environmental modification and allergen avoidance,
usually resulting from allergic, infectious, or anatomic causes anticholinergic agents, anti-IgE therapy, nasal saline irrigation,
(Table  12.1). Nevertheless, in patients with refractory, ulcerative nasal corticosteroids, antireflux therapies, short-acting rescue
or destructive disease, or in whom other more common causes bronchodilators, as well as oral corticosteroids. Status asthmaticus
have been excluded, the possibility of vasculitis, and in particular contributes to the morbidity and mortality in patients with EGPA,
ANCA-associated vasculitis, should be considered. but appears to account for less than 10% of deaths.
Most patients (>85%) with granulomatosis with polyangiitis In granulomatosis with polyangiitis, there is involvement
(GPA, the entity formerly known as Wegener’s granulomatosis) of the trachea and bronchi in approximately 60% of patients.
will develop upper respiratory tract disease and present with rhi- Manifestations include granulomatous airway lesions, subglottic,
norrhoea, epistaxis, septal perforation, congestion, ear pain, hear- tracheal or bronchial stenosis, tracheo-/bronchomalacia, ulcera-
ing loss, mastoiditis, sinusitis, ulcerations, bony destruction and/ tions, haemorrhage, pseudotumour, and/or segmental or lobar
or deformities involving the nose, ears, or sinuses (Reinhold-Keller collapse from airway narrowing or occlusion (Daum et al. 1995).
2000; Specks 2003; Anderson et  al. 1992). Sinus imaging reveals Patients report shortness of breath, wheezing, exercise intoler-
abnormalities in 85% of patients (Fauci et al. 1983; Hoffman et al. ance, voice change, hoarseness, difficulty speaking or swallowing,
144 SECTION 3 clinical manifestations common to vasculitis

Table 12.1 Cardiopulmonary manifestations of selected vasculitides

Granulomatosis Microscopic Eosinophilic granulomatosis Kawasaki’s Polyarteritis Takayasu’s Behçet’s


with polyangiitis polyangiitis with polyangiitis disease nodosa arteritis disease
Upper airway disease +++ ++ +++ − − − −
Asthma and airways +++ − +++ − − − −
disease
Infiltrates +++ ++ +++ − + + +
Nodules and cavities +++ + ++ − + − +
Alveolar haemorrhage ++ ++ + − + − +
Pulmonary artery − + − − − +++ ++
abnormalities
Pleural disease ++ + ++ − + + +
Thromboembolic disease ++ − − − − + ++
Arrhythmias and ++ + ++ ++ + + +
conduction delays
Cardiomyopathy, ++ ++ ++ ++ ++ ++ +
myocarditis, and heart failure
Coronary arteritis ++ ++ ++ ++ ++ ++ +
Pericardial disease ++ ++ ++ ++ + + +
Infection ++ ++ ++ ++ ++ ++ ++
Drug toxicity ++ ++ ++ ++ ++ ++ ++

− Not reported or data unavailable.


+ Rare, but reported.
++ Well characterized and accepted disease manifestation.
+++ Characteristic and frequent disease manifestation.

haemoptysis, and/or chest discomfort. Subglottic or tracheal steno- however, the sensitivity of the flow volume loop is limited. Flexible
sis may present as a threatened airway and can be fatal (Figure 12.1). fibreoptic bronchoscopy with direct visualization is the single most
Pulmonary function testing may be helpful in detecting large air- useful test for evaluation of the airway. Depending upon the loca-
way involvement. Truncation of the inspiratory, expiratory, or both tion and severity of the airway complication, a combination of
portions of the flow volume loop may, respectively, represent a immunosuppressive therapy, interventional pulmonary techniques
variable extrathoracic, variable intrathoracic, or fixed obstruction; such as laser ablation, dilatation, local corticosteroid injection and
airway stenting, and/or surgery may be considered (Utzig et  al.
2002; Hoffman et al. 2003).

Parenchymal lung disease


Pulmonary infiltrates in vasculitis may represent primary vas-
culitic inflammation and necrosis, such as the granulomatous
inflammation and necrotizing vasculitis of granulomatosis with
polyangiitis or the eosinophilic pneumonia and necrotizing vascu-
litis of EGPA. Infection, DAH, heart failure/ pulmonary oedema,
and drug-induced lung disease may also present with radiographic
infiltrates otherwise indistinguishable from active vasculitis. While
the specific radiographic pattern may suggest one of the above (i.e.
diffuse alveolar filling would be more suggestive of DAH while
focal reticulonodular infiltrates would suggest infection or primary
vasculitic involvement), all possibilities must be considered by the
bedside clinician.
Pulmonary parenchymal disease is common in granulomatosis
with polyangiitis (80%), and the chest radiograph may show dif-
fuse infiltrates, focal consolidation, nodules, cavities, or atelectasis
from bronchial involvement. It is most often bilateral and hetero-
Fig. 12.1 CT image of tracheal stenosis in a patient with granulomatosis with geneous, and the infiltrates may wax, wane, migrate, and otherwise
polyangiitis. evolve over time. High-resolution computed tomography (HRCT)
CHAPTER 12 cardiopulmonary manifestations of vasculitis 145

outcome (Hogan et al. 1996). Classically, patients with DAH pre-


sent with haemoptysis, radiographic alveolar infiltrates, and a
decreased haematocrit or haemoglobin (Figure 12.4). Patients most
commonly report shortness of breath and haemoptysis, although
up to one-third of patients will not complain of haemoptysis at the
time of presentation (Zamora et al. 1997). Other symptoms include
chest discomfort, cough, fatigue, malaise, and/or fever. The chest
radiograph demonstrates patchy, bilateral, alveolar infiltrates that
appear as ground-glass attenuation, and/or alveolar consolida-
tion, on high-resolution computed tomography (HRCT) imaging.
The infiltrates are usually diffuse, but may have a lower lobe pre-
dilection. Initially, unilateral or focal infiltrates may be seen, but
ultimately evolve into the diffuse pattern. With recurrent DAH,
reticular infiltrates and septal line thickening may develop due to
fibrosis. While some authors report the presence of an elevated dif-
fusing capacity of carbon monoxide (DLCO) more than 30% above
normal predicted values as helpful in the diagnosis of DAH, it is
rarely feasible to perform extensive pulmonary function testing on
Fig. 12.2 CT image of multiple, bilateral nodules in a patient with
granulomatosis with polyangiitis.

of the chest gives a more detailed and comprehensive characteri- (a)


zation of the pulmonary parenchyma and may show a variety of
changes, including focal consolidation, ground-glass attenuation,
cavitary disease, nodular or micronodular disease, septal thicken-
ing, bronchovascular cuffing, pleural disease, and tracheobronchial
complications (Reuter et al. 1998; Aberle et al. 1990; Maskell et al.
1993; Sheehan et  al. 2003; Papiris et  al. 1992; Weir et  al. 1992).
Similarly, 40–75% of patients with EGPA have radiographic infil-
trates and up to 90% have abnormal CT imaging (Chumbley et al.
1977; Buschman et al. 1990; Choi et al. 2000; Worthy et al. 1998;
Guillevin 1999a; Abu-Shakra et al. 1994; Lanham et al. 1984). The
most common findings on chest imaging are bilateral, heterogene-
ous, patchy infiltrates characterized by ground-glass attenuation
and/or focal consolidation. Microscopic polyangiitis may also pre-
sent with diffuse pulmonary infiltrates representative of DAH in up
to 30% of patients. Likewise, other vasculitides such as giant cell
arteritis, Takayasu’s arteritis, Henoch-Schönlein purpura, cryoglo-
bulinaemia, and Behçet’s disease may produce pulmonary infil-
trates (discussed in Section Specific Vasculitides).
(b)
Nodules and cavities
The appearance of lung nodules and/or cavitary disease evokes a
differential diagnosis that includes malignancy, infection (in partic-
ular atypical infections such as those seen with fungi, mycobacteria,
Nocardia sp., Actinomyces sp., and polymicrobial infections), septic
emboli, sarcoidosis, connective tissue disease (e.g. the necrobiotic
nodules of rheumatoid arthritis), and ANCA-associated vasculitis
(Figures 12.2 and 12.3). A study by Cordier et al. of 77 patients with
granulomatosis with polyangiitis found nodular disease in 69% of
patients, and in 43% cavities were detected (Cordier et al. 1990).
Another CT study of patients with granulomatosis with polyangii-
tis found nodular disease in 90% with cavitation in 48% (Lee et al.
2003). The nodules vary in size (0.3–10 cm), can be limited in num-
ber, or multiple and bilateral (Cordier et al. 1990).

Diffuse alveolar haemorrhage


Diffuse alveolar haemorrhage (DAH) is a life-threatening com- Fig. 12.3 (a) Chest radiograph and (b) CT image of cavitary lesions in a patient
plication of vasculitis and an independent risk factor for poor with granulomatosis with polyangiitis.
146 SECTION 3 clinical manifestations common to vasculitis

(a) (b)

Fig. 12.4 Chest radiographs of a patient with diffuse alveolar haemorrhage. (a) On admission with haemoptysis. (b) Three days later with fulminant disease and
respiratory failure.

patients with acute, diffuse alveolar haemorrhage. The differential and mechanical ventilation), correction of any coagulopathy or
diagnosis of DAH mirrors the differential diagnosis of the more bleeding diathesis, and treatment of the underlying vasculitis.
general finding of dyspnoea/ respiratory distress accompanied by Given that DAH frequently represents life-threatening disease,
bilateral, alveolar infiltrates and includes pneumonia, drug reac- intravenous, high-dose corticosteroids are recommended, as
tion, cardiogenic and non-cardiogenic pulmonary oedema, acute is early introduction of plasmapheresis for severe or refractory
lupus pneumonitis, acute eosinophilic pneumonia, acute hyper- disease (Nachman et al. 1996; Klemmer et al. 2003; Gaskin and
sensitivity pneumonitis, acute interstitial pneumonitis, and acute Pusey 2001). Cyclophosphamide or rituximab therapy should
respiratory distress syndrome. also be initiated; however, the exact timing of administration
The diagnosis of DAH is made at time of bronchoscopy. The of either of these agents in the setting of critical illness remains
bronchoscope is in a ‘wedge’ position in a segmental or subseg- somewhat controversial given their potential for serious compli-
mental bronchus, and serial aliquots of sterile saline of 30–60 ml cations in an otherwise tenuous patient, and, in the case of rituxi-
each are instilled and withdrawn via the bronchoscope (for a total mab, slow onset of action and its removal from the circulation by
volume of 100–300 ml) (Meyer et al. 2012). The diagnosis is then concurrent plasmapheresis. (Lee and Specks 2004). Interestingly,
firm when the recovered bronchoalveolar lavage fluid demonstrates the Rituximab versus Cyclophosphamide for ANCA-Associated
a persistently haemorrhagic return (Figure 12.5). Nevertheless, the Vasculitis (RAVE) trial found that rituximab was non-inferior
finding of DAH is best described as a clinical finding or syndrome, when compared with cyclophosphamide in patients with
and in and of itself does not represent a final clinical diagnosis.
Rather, the presence of DAH carries an extended differential diag-
nosis as to the specific aetiology of the DAH. Pathologically, DAH
may represent pulmonary capillaritis (a small-vessel vasculitis of
the lung), bland haemorrhage or diffuse alveolar damage with
haemorrhage, and distinguishing among these histologies requires
surgical lung biopsy.
The differential diagnosis for DAH is extensive but focuses heav-
ily upon the vasculitides (Table 12.2). Pulmonary–renal syndrome
refers to those patients with both DAH and glomerulonephri-
tis, and 80% of these patients will ultimately be diagnosed with
an ANCA-associated vasculitis or Goodpasture’s syndrome. The
remainder of these cases comprise a mix of connective tissue dis-
eases, cryoglobulinaemia, Henoch-Schönlein purpura, and infec-
tion/ postinfectious complications (Jayne 1998; Niles et al. 1996).
Goodpasture’s syndrome mimics the ANCA-associated vascu-
litides limited to the lungs and/or kidneys as it is characterized by
DAH and crescentic glomerulonephritis, and in 10–20% of cases is
pANCA positive (Rodriguez et al. 2002). Fig. 12.5 Diffuse alveolar haemorrhage is diagnosed during bronchoscopy when
Treatment of DAH secondary to vasculitis includes supportive serial aliquots of lavagate demonstrate a persistently bloody return. Photograph
care (including oxygen and, if necessary, endotracheal intubation courtesy of Gregory P. Cosgrove, MD.
CHAPTER 12 cardiopulmonary manifestations of vasculitis 147

Table 12.2 Differential diagnosis of diffuse alveolar haemorrhage catastrophic results. Pulmonary artery aneurysms are unusual,
and when they occur they are most commonly the result of an iat-
Capillaritis rogenic complication (e.g. right heart catheterization) or trauma;
Pauci-immune disease/ANCA-associated vasculitides however, when neither iatrogenic injury nor trauma are present,
Granulomatosis with polyangiitis Behçet’s disease, Hughes–Stovin syndrome, and, to a lesser extent,
Microscopic polyangiitis Takayasu’s arteritis should be considered. Stenoses and occlu-
Eosinophilic granulomatosis with polyangiitis sions of the pulmonary arteries are more commonly seen with
Idiopathic pauci-immune pulmonary capillaritis Takayasu’s arteritis. The most common presenting symptom of
Immune complex-mediated disease pulmonary artery aneurysm is haemoptysis, but patients may also
Goodpasture’s syndrome present with an abnormal radiograph, cough, dyspnea, fever, chest
Essential cryoglobulinaemia discomfort, signs and symptoms of a mass effect of the aneurysm,
Henoch-Schönlein purpura or a more fulminant bleeding complication. Pulmonary artery
Behçet’s disease aneurysms may range in size from 1 to 10 cm and can be single
Primary connective tissue diseases
or multiple. While direct angiography was traditionally used to
Systemic lupus erythematosus image pulmonary artery aneurysms, it has largely been replaced
Antiphospholipid syndrome by CT angiography and magnetic resonance angiography (MRA)
Other connective tissue diseases (Berkmen 1998).
There are no randomized, controlled trials that evaluate medi-
Drug-induced lung injury
cal treatment of pulmonary artery aneurysms secondary to vascu-
Penicillamine
Chemotherapeutic agents
litis; however, immunosuppression (corticosteroids and cytotoxic
Propylthiouracil agents), embolization, surgery, anticoagulation, and antiplatelet
therapies have all been tried (Uzun et al. 2005). Of these, immu-
Haematopoietic stem cell transplantation nosuppression seems to confer the greatest benefit, and in the set-
Acute lung transplant rejection
ting of massive haemoptysis, embolization of the feeding vessel
Bland haemorrhage is indicated. While thrombus formation may be common within
Idiopathic pulmonary haemosiderosis pulmonary artery aneurysms, anticoagulation carries considerable
Sleep apnoea (severe) risk, and the thrombi are often adherent and organized, so that it is
Coagulopathy reasonable to avoid the use of anticoagulation (Uzun et al. 2005).
Cardiac disease
Mitral stenosis Pulmonary hypertension
Heart failure
Pulmonary arterial hypertension is a rare complication of vasculitis
Drug and inhalation-induced lung injury but case reports do exist (Lie 1996; Roncoroni et al. 1992). Primary
Chemotherapeutic agents plexogenic arteriopathy, thromboembolic disease, pulmonary
Trimellitic anhydride hypertension secondary to parenchymal lung disease, constrictive
Amiodarone
pericarditis, tamponade, restrictive cardiomyopathy, and heart fail-
Nitrofurantoin
ure should all be considered as potential aetiologies of pulmonary
Pulmonary veno-occlusive disease hypertension in the vasculitis patient.
Haematopoietic stem cell transplantation
Diffuse alveolar damage
Venous thromboembolic disease
Acute respiratory distress syndrome Venous thromboembolic disease (VTE) is well-described in
Acute exacerbation of fibrosing interstitial lung disease antiphospholipid syndrome (ALS) and the primary connective tis-
Drug-induced lung injury (e.g. cocaine/ crack lung) sue diseases, most often in systemic lupus erythematosus (SLE).
Similarly, excess cases of VTE occur in granulomatosis with poly-
angiitis as reported by the WeCLOT investigators in conjunction
with the WGET trial, in which they found 29 cases of VTE among
generalized active or severe vasculitis, and, on subgroup analy- 180 patients with granulomatosis with polyangiitis. This is an inci-
sis, rituximab was specifically found to be equally efficacious for dence of 7.0 per 100 person-years, which is comparable to patients
the management of alveolar haemorrhage (Stone et  al. 2010). with a known history of prior VTE and more than 20 times that of
Respiratory failure and the need for mechanical ventilation is a the general population (Merkel et al. 2005; WGET Research Group
poor prognostic factor (Guillevin et al. 1999b; Lauque et al. 2000). 2005). Venous and arterial thromboembolic disease is also common
Finally, the literature reports several cases in which recombinant in Behçet’s disease. The Italian Behçet’s disease cohort found deep
factor VII was used successfully to aid haemostasis in patients venous thrombosis in 21% of patients and superficial thrombo-
with severe, uncontrolled alveolar haemorrhage (Betensley and phlebitis in 10.9% (Pipitone et al. 2004). Intracardiac (right-sided)
Yankaskas 2002; Henke et al. 2004). thrombus is an uncommon but reported complication of Behçet’s
disease (Mogulkoc et  al. 2000). While the incidence of venous
Pulmonary artery aneurysms, thromboembolic disease remains unknown in other vasculitides, it
pseudoaneurysms, stenoses, and occlusions is prudent for the bedside clinician to remain sensitized to the pos-
Medium- and large-vessel vasculitides can affect the vessels of the sibility of venous thromboembolic disease in this patient population
heart and lungs, as well as the aorta and its branches, often with as a whole.
148 SECTION 3 clinical manifestations common to vasculitis

Pleural disease 1995; Siburian et al. 1993; Fong et al. 1992; Hosenpud et al. 1986).
Treatment requires therapy directed at the vasculitis as well as the
Pleural effusions have been reported in the ANCA-associated
conduction disorder/ arrhythmia itself.
vasculitides, polyarteritis nodosa, Behçet’s disease, and giant
cell arteritis. If present, they are of small volume (Cordier et al. Systolic and diastolic dysfunction,
1990; James 1988). If a pleural effusion is identified in a vasculitis
cardiomyopathy, and heart failure
patient, other considerations for its origin include infection (i.e. a
parapneumonic effusion or empyema), heart failure, or thrombo- Primary vasculitic involvement of the myocardial arterioles and
embolic disease. venules may accompany inflammation and granuloma formation
within the heart muscle resulting in (endo)myocarditis (Pagnoux
Infection and Guillevin 2005). While cardiac involvement can be asympto-
matic, it may also lead to systolic or diastolic dysfunction, cardio-
Infection, particularly pneumonia and sepsis, are important causes
myopathy, and heart failure. Ischaemic cardiomyopathy and heart
of morbidity and mortality in patients with vasculitis. The diagno-
failure may also occur as a result of vasculitis-associated coronary
sis is frequently delayed or missed because of overlap between the
artery disease (see Section Coronary Artery Disease). Other causes
signs and symptoms of infection, disease activity, and drug toxicity.
of heart failure in vasculitis include valvular heart disease, aortic
This is most likely in patients receiving immunosuppressive therapy
arch disease, and long-standing vasculitis-induced hypertension.
in whom severe disease, atypical organisms (fungi, mycobacteria,
Cardiomyopathy has been shown to correlate with poor outcomes
Pneumocystis jiroveci, and Nocardia sp., as well as routine bacte-
(Guillevin 1996). Given the high morbidity and mortality associ-
rial and viral agents) and atypical presentations of infection can be
ated with cardiac vasculitis, cardiac involvement qualifies as severe
expected. Corticosteroids have been shown to independently increase
or life-threatening disease, and a more-intensive immunosuppres-
the risk of infectious complications (Stuck et al. 1989). A long-term
sive regimen (e.g. cyclophosphamide plus corticosteroids or rituxi-
outcome study found that infection was the single most common
mab plus corticosteroids) is recommended.
cause of death in patients with ANCA-associated vasculitis, repre-
senting 48% of all deaths (Flossmann et al. 2011). Active vasculitis Valvular heart disease
was the second most common cause of death and accounted for 19%
of deaths. These data emphasize the infectious risk even more than While unusual in small-vessel vasculitis, valvular heart disease
an earlier retrospective analysis of 595 vasculitis patients that found has been described. Valvular heart disease is more common with
that 26% of deaths were caused by infection and that this was second large-vessel disease, particularly Takayasu’s arteritis. In Takayasu’s
only to complications of the vasculitis (58%) (Bourgarit et al. 2005). arteritis, 17–55% of patients will develop aortic insufficiency
It should also be noted that the presence of vasculitis disease activ- (Maksimowicz-McKinnon and Hoffman 2004; Numano and
ity does not exclude the possibility of concomitant infection, and, in Kobayashi 1999).
point of fact, infection may drive disease activity and disease activity Coronary artery disease
and its resultant damage to the respiratory tract and other end organs
Coronary arteritis (primary vasculitic involvement of the coro-
may further patient susceptibility to recurrent infections. Ultimately,
nary arteries) is most often seen in vasculitides that affect the
one cannot over-emphasize the importance of the bedside clinician
medium-sized vessels. Coronary artery aneurysms or ectasia
remaining vigilant to the possibility of infection.
develop in 15–25% of untreated children with Kawasaki’s dis-
Drug toxicity ease (Newburger et al. 2004). Approximately 10% of patients with
PAN will develop clinically significant coronary arteritis, although
As with infection, drug toxicity can often be mistaken for vasculitic
autopsy studies reveal a 50–62% incidence of coronary artery
disease activity. Methotrexate is associated with a drug-induced
involvement (Schrader et al. 1985; Holsinger et al. 1962). Similarly,
pneumonitis in 2–10% of patients, but pulmonary complications
5–25% of patients with Takayasu’s arteritis will develop coronary
have also been reported with other immunosuppressive agents
arteritis (Lie 1998). Coronary arteritis may involve the epicardial
as well (Frankel 2003). Given the extensive number of medica-
portion of the vessel or the smaller myocardial/ intramural por-
tions associated with pulmonary toxicity and the rapid evolution
tion of the vessel. Coronary arteritis may lead to narrowing, steno-
of the field, Drs Foucher and Camus of the Groupe d’Etudes de
sis, ectasia, aneurysm formation, thrombosis, and/or dissection in
la Pathologie Pulmonaire Iatrogène have developed the website
the affected vessel(s) and result in myocardial infarction, ischae-
<http://www.pneumotox.com>, which maintains up-to-date
mic cardiomyopathy, and/or heart failure (Pagnoux and Guillevin
information regarding medication-associated pulmonary toxicity.
2005). It should also be noted that therapy (i.e. corticosteroids) may
accelerate underlying atherosclerosis. Therefore, atherosclerotic
Conduction abnormalities, arrhythmia, heart disease must also be considered in the differential diagnosis
and sudden death of patients with vasculitis and coronary artery disease.
Conduction delay, bundle branch block, sinus node dysfunction,
and heart block are complications of vasculitis and are presumed Pericardial disease
due to endomyocardial or coronary vasculitis (Wilcke et al. 2003; Pericarditis may occur with any of the vasculitides, and is one of
Handa et al. 1997; Yokoi et al. 1992). Moreover, arrhythmias occur; the more common cardiac manifestations of the small-vessel,
they are most often sinus tachycardia and supraventricular tach- ANCA-associated vasculitides. While pericarditis is often an inci-
ycardia (Mirone et al. 1997; Schiavone et al. 1985). Occasionally, dental finding, clinically significant pericarditis and pericardial
more serious arrhythmias such as ventricular tachycardia and effusions occur, as do, albeit less commonly, constrictive pericardi-
sudden death have been reported (Nakada 1996; Haney et  al. tis, haemorrhagic pericarditis, and tamponade (Grant et al. 1994).
CHAPTER 12 cardiopulmonary manifestations of vasculitis 149

The great vessels antibodies, partial thromboplastin time, Russel viper venom
time, anti-beta2 glycoprotein-1 antibodies, hepatitis serologies,
The large-vessel vasculitides and Behçet’s disease affect the great
and cryoglobulins). If Goodpasture’s syndrome is a considera-
vessels of the chest leading to aneurysm formation, dissection,
tion, anti-glomerular basement membrane antibodies should be
thrombosis, stenosis, rupture, or occlusion. The development of
obtained. Patients with confirmed vasculitis should also have a
in situ thrombus can also lead to systemic embolic phenomena.
baseline electrocardiogram as part of their diagnostic work up.
Stenotic lesions of the aorta or its main branches are common in
Takayasu’s arteritis and may present with limb claudication, syn- Pulmonary function testing and
cope or near-syncope, visual disturbances, hypertension, bruits, arterial blood gas analysis
or diminished or absent pulses (Mwipatayi et al. 2005; Kerr et al.
Measurement of lung function by physiological testing and gas
1994; Hall et al. 1985; Lupi-Herrera et al. 1977; Nakao et al. 1967;
exchange are necessary to assess the integrity of the respiratory sys-
Sharma et al. 1996). CT and MRI/MRA are the imaging modalities
tem and characterize and quantify impairment, both initially and
of choice to identify and characterize great and large-vessel vascu-
longitudinally. Lung volumes, spirometry, bronchodilator response,
litides (Choe and Lee 1998). Positron emission tomography (PET)
diffusing capacity (DLCO), and room air blood gas analysis form
scanning appears to provide considerable information regarding
the core physiological assessment. Given the wide array of poten-
inflammation and disease activity in the vessel wall (Kissin and
tial pulmonary manifestations of vasculitis, obstructive, restrictive
Merkel 2004; Blockmans 2011).
or mixed patterns of ventilatory impairment with an increased,
Hypertension decreased or normal DLCO are all potentially consistent with an
underlying vasculitis. The tests, however, cannot reflect a diagnosis
Hypertension occurs in the context of vasculitis through a num-
of vasculitis, nor do they permit a specific pulmonary manifesta-
ber of mechanisms, including large-vessel vasculitis resulting in
tion to be attributed to vasculitis.
renal artery stenosis and as a complication of glomerulonephritis.
Hypertension is common in PAN and Takayasu’s arteritis. Imaging studies
Imaging studies are a key component of the initial cardiopulmo-
Clinical evaluation nary evaluation and longitudinal monitoring of the disease. Chest
History and physical radiographs and particularly CT scanning of the chest may iden-
The cardiopulmonary manifestations of vasculitis will often tify pulmonary disease even when clinically silent and reveal its
present with non-specific chest symptoms such as shortness of extent and pattern. CT of the sinuses may be helpful in character-
breath, exercise intolerance, cough, chest pain or chest discom- izing upper respiratory tract involvement. While MRI is not useful
fort, wheezing/ asthma, tachypnoea, increased work of breathing, for lung imaging, it is ideal for imaging of the pulmonary arteries
or haemoptysis. Alternatively, patients may present with general and great vessels of the chest (Chapter 18). CT angiography is an
cardiovascular symptoms such as light-headedness, syncope, alternative modality for examining the pulmonary arteries. Direct
hypertension, tachycardia, palpitations, or, in catastrophic cases, catheter-guided pulmonary angiography may also be performed
heart failure or sudden death. A  detailed and thorough history on occasion, but is an invasive procedure and as such carries some
and physical examination is central to any evaluation, and the risk and has largely been replaced by CTA and MRA. Some inves-
cardiopulmonary evaluation is no exception. Careful attention to tigators are currently evaluating positron emission tomography as
seemingly unrelated symptoms, a comprehensive review of sys- a modality for the evaluation of large vessel vasculitis, but its role
tems, and a detailed physical examination may reveal evidence of remains to be determined.
extrathoracic disease, which in turn may suggest that a presenting With regard to cardiac imaging, echocardiography is useful for
cardiopulmonary complaint may in fact be related to an underly- suspected cardiac involvement and may detect systolic or diastolic
ing vasculitis, primary connective tissue disease, or other systemic dysfunction, a prior myocardial infarction, cardiomyopathy, val-
autoimmune disorder. vular heart disease, coronary artery aneurysms, and/or pericardial
disease. Echocardiography remains the first-line screening tool of
Laboratory testing choice for the evaluation of cardiac involvement in vasculitis, but
Laboratory testing may be either for diagnostic purposes or it pays to recall that the sensitivity of echocardiography is limited.
for characterizing the extent and severity of a known case Conversely, the role of cardiac MRI (cMRI) has slowly been increas-
of vasculitis. Recommended studies often include ‘routine’ ing in the management of cardiac vasculitis, but specific indica-
screening laboratories such as a complete blood count (CBC), tions and its overall contribution to management decisions remain
metabolic panel, blood urea nitrogen, serum creatinine, uri- unclear, especially given the unknown specificity of cMRI for clini-
nalysis with microscopic sediment examination, liver function cally significant vasculitis (Manning et al. 2002; Marmursztejn et al.
tests, erythrocyte sedimentation rate, and C-reactive protein. 2009; Tacke et  al. 2011). Holter monitoring may be used for the
Determination of ANCA status and anti-proteinases-3 (PR3) assessment of potential arrhythmias. Coronary angiography will
and antimyeloperoxidase (MPO) antibody status is often indi- accurately assess the coronary vessels, but is invasive and may not
cated (Chapters  6 and 7). If the differential diagnosis includes reliably reflect disease in smaller branches.
one of the primary connective tissue diseases, antiphospholipid
syndrome or cryoglobulinaemia, serologies are ordered accord- Invasive studies and biopsy
ingly (antinuclear antibodies, rheumatoid factor, anti-cyclic While the diagnosis of vasculitis can be established on clinical
citrullinated peptide, antitopoisomerase (Scl-70), anti-SS-A/ grounds supported by characteristic radiographic or serologi-
Ro, anti-SS-B/La, antiphospholipid antibodies, anticardiolipin cal testing, tissue acquisition is frequently necessary for definitive
150 SECTION 3 clinical manifestations common to vasculitis

diagnosis. In patients with skin or upper airway involvement, easy et al. 2004). Although cardiac involvement is considered uncom-
access and low morbidity make these a reasonable choice for initial mon in MPA, a study of 85 patients with MPA revealed heart failure
biopsy in spite of limited diagnostic yields. Biopsy of the upper air- in 17.6% and pericarditis in 10% (Guillevin et al. 1999b).
ways is diagnostic in 21–70% of patients, sinus tissue being more
likely to be diagnostic than nasal tissue (Devaney et al. 1990; Colby
et al. 1991; Del Buono and Flint 1991). Eosinophilic granulomatosis with
Bronchoscopy is the procedure of choice for the evaluation and polyangiitis (chapter 32)
diagnosis of DAH and is useful in excluding infection and focal Eosinophilic granulomatosis with polyangiitis (EGPA) must be
airway lesions. Airway lesions are relatively common in granu- differentiated from other lung diseases associated with asthma
lomatosis with polyangiitis but are rarely seen in other vasculitic and eosinophilia, such as chronic eosinophilic pneumonia, aller-
disorders. Even when visualized, biopsy of endobronchial lesions gic bronchopulmonary mycosis, drug reactions, hypereosinophilic
yields diagnostic tissue in only 18% of patients (Daum et al. 1995). syndrome, parasitic infections, and isolated asthma/ atopic disease.
Transbronchial biopsies are rarely useful in pulmonary vasculitis Moreover, EGPA in an atopic individual may be manifest as gas-
(Schnabel et al. 1997). trointestinal disease (perforation, ischaemia, bleeding) or cardiac
Percutaneous renal biopsy establishes the existence of rapidly disease (cardiomyopathy, conduction abnormalities). Interestingly,
progressive glomerulonephritis (Chapter 16). A histopathological recent investigations have elucidated two distinct EGPA pheno-
diagnosis of pauci-immune necrotizing glomerulonephritis reflects types, an ANCA-positive phenotype characterized by renal disease,
an ANCA-positive vasculitis in the majority of cases (Hauer et al. alveolar haemorrhage, and peripheral nervous system involvement
2002; Weiss and Crissman 1985). (a ‘vasculitic’ phenotype) and an ANCA-negative phenotype char-
Surgical lung biopsy, if necessary, usually establishes a acterized by more severe pulmonary and cardiac involvement (an
definitive diagnosis in the suspected vasculitis patient (Travis ‘eosinophilic’ phenotype) (Sable-Fourtassou et al. 2005; Sinico et al.
et al. 1991; Specks 2003; Frankel et al. 2005). Tissue should be 2005; Sinico and Bottero 2009).
formalin-fixed for haematoxylin and eosin staining and special The use of leukotriene antagonist therapy in the management
stains, frozen for immunofluorescent studies, and cultured for of severe asthma in the setting of EGPA remains controversial,
microbiology. although the severity of the asthma associated with EGPA fre-
Endomyocardial biopsies are occasionally done, but given the quently argues for their use. With the wide-spread introduction of
potential risks and low diagnostic yield, it is used sparingly. leukotriene receptor antagonists (LTRA), a number of case series
appeared suggesting that LTRA might promote a biological conver-
Specific vasculitides sion towards a EGPA phenotype. Later analysis, however, seemed
to support the argument that the introduction LTRA permitted
Granulomatosis with polyangiitis (chapters 29–31) successful tapering of oral corticosteroids in patients with latent
Granulomatosis with polyangiitis (the entity formerly known EGPA, ‘unmasking’ the disease. In 2010, an analysis of the Food
as Wegener’s granulomatosis) involves the upper airways, tra- and Drug Administration’s Adverse Event Reporting System data-
cheobronchial tree, pleural space, pulmonary vasculature, and base found 163 cases of EGPA associated with an LTRA and that
pulmonary parenchyma as outlined in Section The Scope of the majority of these cases could not be explained by either cor-
Cardiopulmonary Complications. Approximately 10% of patients ticosteroid withdrawal or pre-existing EGPA (Bibby et  al. 2010).
with granulomatosis with polyangiitis will have lung-limited dis- Hence, the associations between LTRA and EGPA remain difficult
ease at the time of presentation. Cardiac complications, while to fully characterize pending further study.
uncommon, occur in 5–15% of patients, and include coronary Cardiac manifestations are found in 50–65% of patients with
arteritis, (endo-)myocarditis, cardiomyopathy, valvular heart dis- EGPA and cardiac disease accounts for 33–83% of its deaths
ease, conduction abnormalities, arrhythmia, and pericardial dis- (Chumbley et al. 1977; Lanham et al. 1984; Bourgarit et al. 2005;
ease (Korantzopoulos et al. 2004; Goodfield et al. 1995; Lynch et al. Dennert et  al. 2010). Cardiac involvement in EGPA is often
2004; Grant et al. 1994). A literature review conducted by Forstot expressed as a cardiomyopathy as a result of either endomyocarditis
et al. found that the most common manifestations of cardiac dis- with myocardial eosinophilic infiltrates or coronary arteritis with
ease in granulomatosis with polyangiitis are pericarditis (50%), ischaemic injury to the myocardium. Myocardial fibrosis, conges-
coronary arteritis (50%), myocarditis (25%), valvulitis or endocar- tive heart failure, valvular heart disease, myocardial infarction, con-
ditis (21%), conduction system granulomas (17%), AV node arte- duction delays, arrhythmias, pericardial disease, and sudden death
ritis (13%), sinus node arteritis (13%), and myocardial infarction have also been described (Kozak et al. 1995). Patients with cardiac
(11%) (Forstot et al. 1980). involvement in EGPA are more likely to be ANCA negative com-
pared to those without cardiac complications (Sable-Fourtassou
Microscopic polyangiitis (chapter 26) et al. 2005).
Pulmonary manifestations occur in 20–30% of patients, DAH
accounting for most of them (Lane et  al. 2005; Collins and Polyarteritis nodosa (chapter 25)
Quismorio 2005; Lauque et  al. 2000). Other lung complications As noted in Section The Scope of Cardiopulmonary Complications,
found in microscopic polyangiitis (MPA) include isolated radio- coronary arteritis is common in PAN and can result in myocardial
graphic infiltrates, pulmonary arterial aneurysms, pulmonary infarction, ischaemic cardiomyopathy, or death (Pagnoux et  al.
fibrosis, and bronchiolitis, though it is likely that at least some of 2010). Pulmonary alveolar involvement with classic PAN is rare
these are also the result of recurrent and chronic alveolar haemor- but DAH, radiographic infiltrates and pleural effusion have been
rhage (Collins and Quismorio 2005; Brugiere et al. 1997; Homma described.
CHAPTER 12 cardiopulmonary manifestations of vasculitis 151

Kawasaki’s disease (chapter 28) (Gelsomino et al. 2005; Nesi et al. 2002; Evans et al. 1995; Evans
et  al. 1994; Klein et  al. 1975). Lung involvement is unusual, but
In children with Kawasaki’s disease, 15–25% will develop coro-
there are reports of cough, hoarseness, sore throat, radiographic
nary artery aneurysms if diagnosis and treatment are delayed
infiltrates including one case with multiple pulmonary nodules,
(Newburger et al. 2004). A significant percentage of patients with
pleural effusions, granulomatous lung disease, pulmonary artery
cardiac involvement will subsequently develop myocarditis, con-
stenosis, obstruction and/or thrombosis, DAH, and multiple pul-
duction delays, arrhythmia, and/or pericarditis. Mitral regurgitation
monary infarctions (Olopade et al. 1997; Karam and Fulmer 1982;
and aortic regurgitation also occur. While in general the prognosis
Ladanyi and Fraser 1987; Bradley et al. 1984; Huong Dle et al. 2003;
for Kawasaki’s disease is excellent, myocardial infarction, heart
Radhamanohar 1991). Coronary arteritis secondary to GCA has
failure, and sudden death represent catastrophic complications of
also been reported (Kumar et al. 2002).
Kawasaki’s disease, and the majority of deaths from Kawasaki’s dis-
ease are due to coronary complications (Kato et al. 1986).
Behçet’s disease (chapter 34)
Pulmonary artery aneurysms are the most common lung complica-
Takayasu’s arteritis (chapter 24)
tion of Behçet’s disease and occur in 1–8% of patients (Erkan et al.
Takayasu’s arteritis affects the pulmonary arteries in approximately 2001) (Figure 12.6). Pulmonary artery aneurysms affect predomi-
50% of cases (Vanoli et al. 1999; Yamada et al. 2000) and may cause nantly young men and carry a poor prognosis with a 25–50% mor-
pulmonary artery stenosis, occlusion, aneurysm with rupture, or pul- tality rate (Hamuryudan et al. 1994; Seyahi et al. 2012). Pulmonary
monary hypertension. Takayasu’s arteritis affects the coronary arter- artery thrombus may occur with or without pulmonary artery aneu-
ies in 5–25% of cases, most commonly at the level of the coronary rysms. Other pulmonary manifestations of Behçet’s disease include
ostitia where they arise from the aorta, and such involvement may pulmonary nodules, atelectasis, organizing pneumonia, thrombo-
lead to myocardial infarction or sudden death (Lie 1987; Lie 1998). embolic disease, DAH due to capillaritis, pleural effusions, abnor-
Dilatation of the aortic root and aortic insufficiency are relatively mal pulmonary function testing, and pneumonia (Uzun et al. 2005).
common. An echocardiographic study of 78 patients with Takayasu’s Cardiovascular complications occur in 6% of Behçet’s patients and
arteritis and giant cell arteritis identified left ventricular systolic dys- may include pericarditis, endocarditis and valvular incompetence
function in 18% of patients (Pfizenmaier et al. 2004). Interestingly, or prolapse, intracardiac thrombosis, myocardial infarction, endo-
left ventricular dysfunction did not correlate with haemodynamic myocardial fibrosis, aortitis, and myocardial aneurysm (Atzeni
variables, aortic regurgitation, or systemic hypertension, suggesting et al. 2005; Gurgun et al. 2002; Mirone et al. 1997; Geri et al. 2012;
that primary cardiac inflammation was responsible for a significant Saadoun et  al. 2012). Other cardiovascular complications may
proportion of the cases. Finally, great and large-vessel involvement, include great or large vessel aneurysms with rupture, cardiomyo-
including thoracic and abdominal aortic aneurysm, stenosis, dissec- pathy, heart failure, repolarization abnormalities, and arrhythmias.
tion, rupture, coarctation and occlusion, are a hallmark of Takayasu’s
arteritis and are discussed in more detail in Chapter 24. Henoch–schönlein purpura (IgA vasculitis)
(chapter 39)
Giant cell arteritis (chapter 23) A retrospective study of Henoch–Schönlein purpura (HSP) found
Giant cell arteritis (GCA) involves the thoracic aorta in 8–10% of that 2.4% of patients, all adults, had pulmonary involvement
patients and can cause serious or fatal complications (Evans et al. (Nadrous et  al. 2004). Based upon this and previously reported
1994). Aortitis with aortic aneurysm formation, ectasia, rupture, cases, DAH appears to be the most common pulmonary manifesta-
and/or dissection occur, and aneurysmal dilatation of the aortic tion of HSP, although one case of pulmonary fibrosis was identified
root may lead to aortic regurgitation and congestive heart failure (Nadrous et al. 2004; Vats et al. 1999; Olson et al. 1992).

(a) (b)

Fig. 12.6 (a) CT image and (b) angiography of pulmonary artery aneurysms in a patient with Behçet’s disease.
152 SECTION 3 clinical manifestations common to vasculitis

Mixed cryoglobulinaemia (chapter 40) polyangiitis. American Journal of Respiratory and Critical Care Medicine,
155, 739–42.
Pulmonary involvement is rare with cryoglobulinaemic vascu- Buschman, D.L., Waldron, J.A., and King, T.E. (1990). Churg Strauss pul-
litis, although case reports of DAH and acute lung injury are monary vasculitis:  high resolution computed tomography scanning
recorded (Suzuki et al. 2003; Gomez-Tello et al. 1999). Infiltrates and pathologic findings. American Review of Respiratory Disease, 142,
and mild lung function abnormalities identified by pulmonary 458–61.
function testing may be seen as well (Viegi et al. 1989; Ferri et al. Choe, Y.H. and Lee, W.R. (1998). Magnetic resonance imaging diagnosis of
1998). Congestive heart failure, myocardial infarction, pericar- Takayasu arteritis. International Journal of Cardiology, 66, S175–9.
dial disease, and valvular heart disease have been reported with Choi, Y.H., Im, J.-G., Han, B.K., Kim, J.-H., Lee, K.Y., and Myong, N.H. (2000).
Thoracic manifestations of Churg-Strauss syndrome. Chest, 117, 117–24.
cryoglobulinaemia.
Chumbley, L.C., Harrison, E.G. Jr., and Deremee, R.A. (1977). Allergic gran-
Secondary vasculitis in primary connective ulomatosis and angiitis (Churg-Strauss syndrome). Report and analysis
of 30 cases. Mayo Clinic Proceedings, 52, 477–84.
tissue diseases (chapter 33)
Colby, T.V., Tazelaar, H.D., Specks, U., and Deremee, R.A. (1991). Nasal
Connective tissue diseases may present with a number of car- biopsy in Wegener’s granulomatosis. Human Pathology, 22, 101–4.
diopulmonary manifestations and complications, and approxi- Collins, C.E. and Quismorio, F.P. Jr. (2005). Pulmonary involvement in
mately 20% of patients with a primary connective tissue disease microscopic polyangiitis. Current Opinion in Pulmonary Medicine, 11,
(CTD) have a form of lung disease. Potential complications 447–51.
include interstitial lung disease, pulmonary hypertension, Cordier, J.-F., Valeyre, D., Guillevin, L., Loire, R., and Crechot, J.-M. (1990).
pleural disease, airways disease, thromboembolic, disease, dia- Pulmonary wegener’s granulomatosis. A clinical and imaging study of
77 cases. Chest, 97, 906–12.
phragmatic and respiratory muscle weakness, diffuse alveolar
Daum, T.E., Specks, U., Colby, T.V., Edell, E.S., Brutinel, M.W., Prakash, U.B.,
haemorrhage, pericardial disease, myocarditis, valvular heart
and Deremee, R.A. (1995). Tracheobronchial involvement in Wegener’s
disease, cardiomyopathy, coronary arteritis, conduction delays, granulomatosis. American Journal of Respiratory and Critical Care
and arrhythmias. Medicine, 151, 522–6.
Del Buono, E.A. and Flint, A. (1991). Diagnostic usefulness of nasal biopsy in
References Wegener’s granulomatosis. Human Pathology, 22, 107–10.
Aberle, D.R., Gamsu, G., and Lynch, D. (1990). Thoracic manifesta- Dennert, R.M., Van Paassen, P., Schalla, S., Kuznetsova, T., Alzand, B.S.,
tions of Wegener granulomatosis:  diagnosis and course. Radiology, Staessen, J. A., Velthuis, S., Crijns, H.J., Tervaert, J.W., and Heymans, S.
174, 703–9. (2010). Cardiac involvement in Churg-Strauss syndrome. Arthritis and
Abu-Shakra, M., Smythe, H., Lewtas, J., Badley, E., Weber, D., and Keystone, Rheumatism, 62, 627–34.
E. (1994). Outcome of polyarteritis nodosa and Churg-Strauss syn- Devaney, K.O., Travis, W.D., Hoffman, G., Leavitt, R., Lebovics, R., and Fauci,
drome. An analysis of twenty-five patients. Arthritis and Rheumatism, A.S. (1990). Interpretation of head and neck biopsies in Wegener’s gran-
37, 1798–803. ulomatosis. A pathologic study of 126 biopsies in 70 patients. American
Anderson, G., Coles, E.T., Crane, M., Douglas, A.C., Gibbs, A.R., Geddes, Journal of Surgical Pathology, 14, 555–64.
D.M., et  al. (1992). Wegener’s granulomatosis:  a series of 265 British Erkan, F., Gul, A., and Tasali, E. (2001). Pulmonary manifestations of Behçet’s
cases seen between 1975 and 1985. A  report by a sub-committee of disease. Thorax, 56, 572–8.
the British Thoracic Society Research Committee. Quarterly Journal of Evans, J.M., Bowles, C.A., Bjornsson, J., Mullany, C.J., and Hunder, G.G.
Medicine, 83, 427–38. (1994). Thoracic aortic aneurysm and rupture in giant cell arteritis.
Atzeni, F., Sarzi-Puttini, P., Doria, A., Boiardi, L., Pipitone, N., and Salvarani, A descriptive study of 41 cases. Arthritis and Rheumatism, 37, 1539–47.
C. (2005). Behçet’s disease and cardiovascular involvement. Lupus, Evans, J.M., O’Fallon, W.M., and Hunder, G.G. (1995). Increased incidence
14, 723–6. of aortic aneurysm and dissection in giant cell (temporal). arteritis.
Berkmen, T. (1998). MR angiography of aneurysms in Behcet disease: a report A population-based study. Annals of Internal Medicine, 122, 502–7.
of four cases. Journal of Computer Assisted Tomography, 22, 202–6. Fauci, A.S., Haynes, B.F., Katz, P., and Wolff, S.M. (1983). Wegener’s granulo-
Betensley, A.D. and Yankaskas, J.R. (2002). Factor VIIA for alveolar hemor- matosis: propsective clinical and therapeutic expeience with 85 patients
rhage in microscopic polyangiitis. American Journal of Respiratory and for 21 years. Annals of Internal Medicine, 98, 76–85.
Critical Care Medicine, 166, 1291–2. Ferri, C., La Civita, L., Longombardo, G., Zignego, A.L., and Pasero, G.
Bibby, S., Healy, B., Steele, R., Kumareswaran, K., Nelson, H., and Beasley, (1998). Mixed cryoglobulinaemia: a cross-road between autoimmune
R. (2010). Association between leukotriene receptor antagonist therapy and lymphoproliferative disorders. Lupus, 7, 275–9.
and Churg-Strauss syndrome: an analysis of the FDA AERS database. Flossmann, O., Berden, A., De Groot, K., Hagen, C., Harper, L., Heijl, C.,
Thorax, 65, 132–8. Hoglund, P., Jayne, D., Luqmani, R., Mahr, A., Mukhtyar, C., Pusey,
Blockmans, D. (2011). PET in vasculitis. Annals of the New York Academy of C., Rasmussen, N., Stegeman, C., Walsh, M., and Westman, K. (2011).
Sciences, 2011, 1228, 64–70. Long-term patient survival in ANCA-associated vasculitis. Annals of the
Bourgarit, A., Le Toumelin, P., Pagnoux, C., Cohen, P., Mahr, A., Le Guern, Rheumatic Diseases, 70, 488–94.
V., Mouthon, L., and Guillevin, L. (2005). Deaths occurring during the Fong, C., Schmidt, G., Cain, N., Cranswick, P., and Tonkin, A.M. (1992).
first year after treatment onset for polyarteritis nodosa, microscopic Churg-Strauss syndrome, cardiac involvement and life threatening ven-
polyangiitis, and Churg-Strauss syndrome:  a retrospective analysis tricular arrhythmias. Australian and New Zealand Journal of Medicine,
of causes and factors predictive of mortality based on 595 patients. 22, 167–8.
Medicine (Baltimore), 84, 323–30. Forstot, J.Z., Overlie, P.A., Neufeld, G.K., Harmon, C.E., and Forstot, S.L.
Bradley, J.D., Pinals, R.S., Blumenfeld, H.B., and Poston, W.M. (1984). Giant (1980). Cardiac complications of Wegener granulomatosis: a case report
cell arteritis with pulmonary nodules. American Journal of Medicine, 77, of complete heart block and review of the literature. Seminars in Arthritis
135–40. and Rheumatism, 10, 148–54.
Brugiere, O., Raffy, O., Sleiman, C., Groussard, O., Rothchild, E., Mellot, Frankel, S.K. (2003). Drug-induced lung disease. In Current diagnosis and
F., Jebrak, G., Mal, H., Roue, C., Pariente, R., and Fournier, M. (1997). treatment in pulmonary medicine (ed. M.E. Hanley and C.H.Welsh).
Progressive obstructive lung disease associated with microscopic Lange Medical Books/McGraw-Hill, New York.
CHAPTER 12 cardiopulmonary manifestations of vasculitis 153

Frankel, S.K., Cosgrove, G.P., and Brown, K.K. (2005). Small vessel vasculitis Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., Hallahan, C.W., Lebovics, R.S., Travis,
of the lung. Chronic Respiratory Disease, 2, 75–84. W.D., Rottem, M., and Fauci, A.S. (1992). Wegener’s granulomatosis: An
Gaskin, G. and Pusey, C. (2001). Plasmapheresis in antineutrophil cytoplas- analysis of 158 patients. Annals of Internal Medicine, 116, 488–98.
mic antibody-associated systemic vasculitis. Therapeutic Apheresis, 5, Hoffman, G.S., Thomas-Golbanov, C.K., Chan, J., Akst, L.M., and Eliachar,
176–81. I. (2003). Treatment of subglottic stenosis, due to Wegener’s granu-
Gelsomino, S., Romagnoli, S., Gori, F., Nesi, G., Anichini, C., Sorbara, C., lomatosis, with intralesional corticosteroids and dilation. Journal of
Stefano, P., and Gensini, G.F. (2005). Annuloaortic ectasia and giant cell Rheumatology, 30, 1017–21.
arteritis. Annals of Thoracic Surgery, 80, 101–5. Hogan, S.L., Falk, R.J., Chin, H., Cai, J., Jennette, C.E., Jennette, J.C., and
Geri, G., Wechsler, B., Thi Huong Du, L., Isnard, R., Piette, J.C., Amoura, Nachman, P.H. (2005). Predictors of relapse and treatment resistance in
Z., Resche-Rigon, M., Cacoub, P., and Saadoun, D. (2012). Spectrum of antineutrophil cytoplasmic antibody-associated small-vessel vasculitis.
cardiac lesions in Behcet disease: a series of 52 patients and review of the Annals of Internal Medicine, 143, 621–31.
literature. Medicine (Baltimore), 91, 25–34. Hogan, S.L., Nachman, P.H., Wilkman, A.S., Jennette, J.C., and Falk, R.J.
Gomez-Tello, V., Onoro-Canaveral, J.J., De La Casa Monje, R.M., (1996). Prognostic markers in patients with antineutrophil cytoplasmic
Gomez-Casero, R.B., Moreno Hurtrez, J.L., Garcia-Montes, M., and autoantibody-associated microscopic polyangiitis and glomerulone-
Armas, L.C. (1999). Diffuse recidivant alveolar hemorrhage in a patient phritis. Journal of the American Society of Nephrology, 7, 23–32.
with hepatitis C virus-related mixed cryoglobulinemia. Intensive Care Holsinger, D.R., Osmundson, P.J., and Edwards, J.E. (1962). The heart in peri-
Medicine, 25, 319–22. arteritis nodosa. Circulation, 25, 610–8.
Goodfield, N.E., Bhandari, S., Plant, W.D., Morley-Davies, A., and Sutherland, Homma, S., Matsushita, H., and Nakata, K. (2004). Pulmonary fibrosis in
G.R. (1995). Cardiac involvement in Wegener’s granulomatosis. British myeloperoxidase antineutrophil cytoplasmic antibody-associated vas-
Heart Journal, 73, 110–5. culitides. Respirology, 9, 190–6.
Grant, S.C., Levy, R.D., Venning, M.C., Ward, C., and Brooks, N.H. Hosenpud, J.D., Mcanulty, J.H., and Niles, N.R. (1986). Unexpected myocar-
(1994). Wegener’s granulomatosis and the heart. British Heart Journal, dial disease in patients with life threatening arrhythmias. British Heart
71, 82–86. Journal, 56, 55–61.
Guillevin, L., Cohen, P., Gayraud, M., Lhote, F., Jarrousse, B., and Casassus, Huong Dle, T., Andreu, M.R., Duhaut, P., Godeau, P., and Piette, J.C.
P. (1999a). Churg-Strauss syndrome. Clinical study and long-term (2003). Intra-alveolar haemorrhage in temporal arteritis. Annals of the
follow-up of 96 patients. Medicine, 78, 26–37. Rheumatic Diseases, 62, 189–90.
Guillevin, L., Durand-Gasselin, B., Cevallos, R., Gayraud, M., Lhote, F., James, D.G. (1988). ‘Silk route disease’ (Behçet’s disease). Western Journal of
Callard, P., Amouroux, J., Casassus, P., and Jarrousse, B. (1999b). Medicine, 148, 433–7.
Microscopic polyangiitis: clinical and laboratory findings in eighty-five Jayne, D.R. (1998). Pulmonary-renal syndrome. Seminars in Respiratory and
patients. Arthritis and Rheumatism, 42, 421–30. Critical Care Medicine, 19, 69–77.
Guillevin, L., Houng Du, L.T., Godeau, P., Jais, P., and Wechsler, B. Karam, G.H., and Fulmer, J.D. (1982). Giant cell arteritis presenting as inter-
(1988). Clinical findings and prognosis of polyarteritis nodosa and stitial lung disease. Chest, 82, 781–4.
Churg-Strauss angiitis:  A  study in 165 patients. British Journal of Kato, H., Ichinose, E., and Kawasaki, T. (1986). Myocardial infarction in
Rheumatology, 27, 258–64. Kawasaki disease: clinical analyses in 195 cases. Journal of Pediatrics,
Guillevin, L., Lhote, F., Gayraud, M., Cohen, P., Jarrousse, B., Lortholary, 108, 923–7.
O., Thibult, N., and Casassus, P. (1996). Prognostic factors in polyarte- Kerr, G.S., Hallahan, C.W., Giordano, J., Leavitt, R.Y., Fauci, A.S., Rottem,
ritis nodosa and churg-strauss syndrome. A prospective study in 342 M., and Hoffman, G.S. (1994). Takayasu’s arteritis. Annals of Internal
patients. Medicine, 75, 17–28. Medicine, 120, 919–29.
Guillevin, L., Pagnoux, C., Seror, R., Mahr, A., Mouthon, L., and Le Toumelin, Kissin, E.Y. and Merkel, P.A. (2004). Diagnostic imaging in Takayasu arteri-
P. (2011). The Five-Factor Score revisited: assessment of prognoses of tis. Current Opinion in Rheumatology, 16, 31–7.
systemic necrotizing vasculitides based on the French Vasculitis Study Klein, R.G., Hunder, G.G., Stanson, A.W., and Sheps, S.G. (1975). Large
Group (FVSG). cohort. Medicine (Baltimore), 90, 19–27. artery involvement in giant cell (temporal). arteritis. Annals of Internal
Gurgun, C., Ercan, E., Ceyhan, C., Yavuzgil, O., Zoghi, M., Aksu, K., Cinar, Medicine, 83, 806–12.
C.S., and Turkoglu, C. (2002). Cardiovascular involvement in Behçet’s Klemmer, P.J., Chalermskulrat, W., Reif, M.S., Hogan, S.L., Henke, D.C., and
disease. Japanese Heart Journal, 43, 389–98. Falk, R.J. (2003). Plasmapheresis therapy for diffuse alveolar hemor-
Hall, S., Barr, W., Lie, J.T., Stanson, A.W., Kazmier, F.J., and Hunder, G.G. rhage in patients with small vessel vasculitis. American Journal of Kidney
(1985). Takayasu’s arteritis:  a study of 32 North American patients. Diseases, 42, 1149–53.
Medicine, 64, 89–99. Korantzopoulos, P., Papaioannides, D., and Siogas, K. (2004). The heart in
Hamuryudan, V., Yurdakul, S., Moral, F., Numan, F., Tuzun, H., Tuzuner, N., Wegener’s granulomatosis. Cardiology, 102, 7–10.
Mat, C., Tuzun, Y., Ozyazgan, Y., and Yazici, H. (1994). Pulmonary arte- Kozak, M., Gill, E.A., and Green, L.S. (1995). The Churg-Strauss syndrome.
rial aneurysms in Behçet’s syndrome: a report of 24 cases. British Journal A case report with angiographically documented coronary involvement
of Rheumatology, 33, 48–51. and a review of the literature. Chest, 107, 578–80.
Handa, R., Wali, J.P., Aggarwal, P., Wig, N., Biswas, A., and Kumar, A.K. Kumar, P., Velissaris, T., Sheppard, M.N., and Pepper, J.R. (2002). Giant cell
(1997). Wegener’s granulomatosis with complete heart block. Clinical arteritis confined to intramural cornary arteries. Unforseen hazards myo-
and Experimental Rheumatology, 15, 97–9. cardial protection. Journal Cardiovascular Surgery (Torino), 43, 647–9.
Haney, I., Beghetti, M., Mccrindle, B.W., and Gow, R.M. (1995). Ventricular Ladanyi, M. and Fraser, R.S. (1987). Pulmonary involvement in giant cell
arrhythmia complicating Kawasaki disease. Canadian Journal of arteritis. Archives of Pathology and Laboratory Medicine, 111, 1178–80.
Cardiology, 11, 931–3. Lane, S.E., Watts, R.A., Shepstone, L., and Scott, D.G. (2005). Primary sys-
Hauer, H.A., Bajema, I.M., Van Houwelingen, H.C., Ferrario, F., Noel, L.H., temic vasculitis: clinical features and mortality. QJM, 98, 97–111.
Waldherr, R., Jayne, D.R., Rasmussen, N., Bruijn, J.A., and Hagen, E.C. Lanham, J., Elkon, K., Pusey, C., and Hughes, G. (1984). Systemic vasculitis
(2002). Renal histology in ANCA-associated vasculitis: differences between with asthma and eosinophilia: a clinical approach to the Churg-Strauss
diagnostic and serologic subgroups. Kidney International, 61, 80–9. syndrome. Medicine, 63, 65–81.
Henke, D.C., Falk, R.J., and Gabriel, D.A. (2004). Successful treatment of dif- Lauque, D., Cadranel, J., Lazor, R., Pourrat, J., Ronco, P., Guillevin, L., and
fuse alveolar hemorrhage with activated factor VII. Annals of Internal Cordier, J. F. (2000). Microscopic polyangiitis with alveolar hemorrhage.
Medicine, 140, 493–4. A study of 29 cases and review of the literature. Groupe d’Etudes et de
154 SECTION 3 clinical manifestations common to vasculitis

Recherche sur les Maladies “Orphelines” Pulmonaires (GERM“O”P). Nadrous, H.F., Yu, A.C., Specks, U., and Ryu, J.H. (2004). Pulmonary involve-
Medicine (Baltimore), 79, 222–33. ment in Henoch-Schonlein purpura. Mayo Clinics Proceedings, 79,
Lee, A.S. and Specks, U. (2004). Pulmonary capillaritis. Seminars in 1151–7.
Respiratory and Critical Care Medicine, 25, 547–55. Nakada, T. (1996). Ventricular arrhythmia and possible myocardial ischemia
Lee, K.S., Kim, T.S., Fujimoto, K., Moriya, H., Watanabe, H., Tateishi, U., in late stage Kawasaki disease: patient with a normal coronary arterio-
Ashizawa, K., Johkoh, T., Kim, E.A., and Kwon, O.J. (2003). Thoracic gram. Acta Paediatrica Japonica, 38, 365–9.
manifestation of Wegener’s granulomatosis: CT findings in 30 patients. Nakao, K., Ikeda, M., Kimata, S.-I., Niitani, H., Miyahara, M., Ishimi, Z.-I.,
European Radiology, 13, 43–51. Hashiba, K., Takeda, Y., Ozawa, T., Matsushita, S., and Kuramochi, M.
Lie, J.T. (1987). Coronary vasculitis. Archives of Pathology and Laboratory (1967). Takayasu’s arteritis: clinical report of eight-four cases and immu-
Medicine, 111, 224–33. nological studies of seven cases. Circulation, 35, 1141–55.
Lie, J.T. (1996). Isolated pulmonary Takayasu arteritis:  clinicopathologic Nesi, G., Anichini, C., Pedemonte, E., Tozzini, S., Calamai, G., Montesi, G.
characteristics. Modern Pathology, 9, 469–74. F., and Gori, F. (2002). Giant cell arteritis presenting with annuloaortic
Lie, J.T. (1998). Pathology of isolated nonclassical and catastrophic mani- ectasia. Chest, 121, 1365–7.
festations of Takayasu arteritis. International Journal of Cardiology, 66, Newburger, J.W., Takahashi, M., Gerber, M.A., Gewitz, M.H., Tani, L.Y.,
S11–21. Burns, J.C., Shulman, S.T., Bolger, A.F., Ferrieri, P., Baltimore, R.S.,
Lupi-Herrera, E., Sanchez-Torres, G., Marchushamer, J., Mispireta, J., Wilson, W.R., Baddour, L.M., Levison, M.E., Pallasch, T.J., Falace, D.A.,
Horwitz, S., and Vela, J.E. (1977). Takayasu’s arteritis. Clinical study of and Taubert, K.A. (2004). Diagnosis, treatment, and long-term manage-
107 cases. American Heart Journal, 93, 94–103. ment of Kawasaki disease: a statement for health professionals from the
Lynch, J. P. 3rd, White, E., Tazelaar, H., and Langford, C.A. (2004). Wegener’s Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease,
granulomatosis: evolving concepts in treatment. Seminars in Respiratory Council on Cardiovascular Disease in the Young, American Heart
and Critical Care Medicine, 25, 491–521. Association. Pediatrics, 114, 1708–33.
Maksimowicz-Mckinnon, K. and Hoffman, G.S. (2004). Large-vessel vasculi- Niles, J.L., Bottinger, E.P., Saurina, G.R., Kelly, K.J., Pan, G., Collins, A.B.,
tis. Seminars in Respiratory and Critical Care Medicine, 25, 569–79. Mccluskey, R.T. (1996). The syndrome of lung hemorrhage and nephritis
Manning, W.J., Stuber, M., Danias, P.G., Botnar, R.M., Yeon, S.B., and is usually an ANCA-associated condition. Archives of Internal Medicine,
Aepfelbacher, F.C. (2002). Coronary magnetic resonance imaging: cur- 156, 440–5.
rent status. Current Problems in Cardiology, 27, 275–333. Numano, F. and Kobayashi, Y. (1999). Takayasu arteritis—beyond pulseless-
Marmursztejn, J., Vignaux, O., Cohen, P., Guilpain, P., Pagnoux, C., Gouya, ness. Internal Medicine, 38, 226–32.
H., Mouthon, L., Legmann, P., Duboc, D., and Guillevin, L. (2009). Olopade, C.O., Sekosan, M., and Schraufnagel, D.E. (1997). Giant cell arte-
Impact of cardiac magnetic resonance imaging for assessment of ritis manifesting as chronic cough and fever of unknown origin. Mayo
Churg-Strauss syndrome: a cross-sectional study in 20 patients. Clinical Clinics Proceedings, 72, 1048–50.
and Experimental Rheumatology, 27, S70–6. Olson, J.C., Kelly, K.J., Pan, C.G., and Wortmann, D.W. (1992). Pulmonary
Maskell, G.F., Lockwood, C.M., and Flower, C.D. (1993). Computed tomog- disease with hemorrhage in Henoch-Schoenlein purpura. Pediatrics, 89,
raphy of the lung in Wegener’s granulomatosis. Clinical Radiology, 48, 1177–81.
377–80. Pagnoux, C. and Guillevin, L. (2005). Cardiac involvement in small and
Merkel, P.A., Lo, G.H., Holbrook, J.T., Tibbs, A.K., Allen, N.B., Davis, J.C. medium-sized vessel vasculitides. Lupus, 14, 718–22.
Jr., Hoffman, G.S., McCune, W.J., St Clair, E.W., Specks, U., Spiera, R., Pagnoux, C., Hogan, S.L., Chin, H., Jennette, J.C., Falk, R.J., Guillevin, L., and
Petri, M., and Stone, J.H. (2005). Brief communication: high incidence Nachman, P.H. (2008). Predictors of treatment resistance and relapse in
of venous thrombotic events among patients with Wegener granuloma- antineutrophil cytoplasmic antibody-associated small-vessel vasculi-
tosis:  the Wegener’s Clinical Occurrence of Thrombosis (WeCLOT). tis: comparison of two independent cohorts. Arthritis and Rheumatism,
Study. Annals of Internal Medicine, 142, 620–6. 58, 2908–18.
Meyer, K.C., Raghu, G., Baughman, R.P., Brown, K.K., Costabel, U., Du Bois, Pagnoux, C., Seror, R., Henegar, C., Mahr, A., Cohen, P., Le Guern, V.,
R.M., Drent, M., Haslam, P.L., Kim, D.S., Nagai, S., Rottoli, P., Saltini, Bienvenu, B., Mouthon, L., and Guillevin, L. (2010). Clinical features
C., Selman, M., Strange, C., and Wood, B. (2012). An official American and outcomes in 348 patients with polyarteritis nodosa:  a systematic
Thoracic Society clinical practice guideline: the clinical utility of bron- retrospective study of patients diagnosed between 1963 and 2005 and
choalveolar lavage cellular analysis in interstitial lung disease. American entered into the French Vasculitis Study Group Database. Arthritis and
Journal of Respiratory and Critical Care Medicine, 185, 1004–14. Rheumatism, 62, 616–26.
Mirone, L., Altomonte, L., Ferlisi, E.M., Zoli, A., and Magaro, M. (1997). Papiris, S.A., Manoussakis, M.N., Drosos, A.A., Kontogiannis, D.,
Behçet’s disease and cardiac arrhythmia. Clinical Rheumatology, 16, Constantopoulos, S. H., and Moutsopoulos, H.M. (1992). Imaging of
99–100. thoracic Wegener’s granulomatosis: the computed tomographic appear-
Mogulkoc, N., Burgess, M.I., and Bishop, P.W. (2000). Intracardiac thrombus ance. American Journal of Medicine, 93, 529–36.
in Behçet’s disease: a systematic review. Chest, 118, 479–87. Pfizenmaier, D.H., Al Atawi, F.O., Castillo, Y., Chandrasekaran, K., and
Mukhtyar, C., Flossmann, O., Hellmich, B., Bacon, P., Cid, M., Cohen-Tervaert, Cooper, L.T. (2004). Predictors of left ventricular dysfunction in
J.W., Gross, W.L., Guillevin, L., Jayne, D., Mahr, A., Merkel, P.A., Raspe, patients with Takayasu’s or giant cell aortitis. Clinical and Experimental
H., Scott, D., Witter, J., Yazici, H., and Luqmani, R.A. (2008). Outcomes Rheumatology, 22, S41–5.
from studies of antineutrophil cytoplasm antibody associated vasculi- Pipitone, N., Boiardi, L., Olivieri, I., Cantini, F., Salvi, F., Malatesta, R., La
tis: a systematic review by the European League Against Rheumatism Corte, R., Triolo, G., Ferrante, A., Filippini, D., Paolazzi, G., Sarzi-Puttini,
systemic vasculitis task force. Annals of the Rheumatic Diseases, 67, P., Restuccia, G., and Salvarani, C. (2004). Clinical manifestations of
1004–10. Behçet’s disease in 137 Italian patients: results of a multicenter study.
Mwipatayi, B.P., Jeffery, P.C., Beningfield, S.J., Matley, P.J., Naidoo, N.G., Clinical and Experimental Rheumatology, 22, S46–51.
Kalla, A.A., and Kahn, D. (2005). Takayasu arteritis: clinical features and Radhamanohar, M. (1991). Multiple pulmonary infarctions caused by giant
management: report of 272 cases. ANZ Journal of Surgery, 75, 110–7. cell arteritis. Postgraduate Medical Journal, 67, 491.
Nachman, P.H., Hogan, S.L., Jennette, J.C., and Falk, R.J. (1996). Reinhold-Keller, E., Beuge, N., Latza, U., Degroot, K., Rudert, H., Nolle,
Treatment response and relapse in antineutrophil cyctoplasmic B., Heller, M., and Gross, W. (2000). An interdisciplinary approach
autoantibody-associated microscopic polyangiitis and glomerulone- to the care of patients with wegener’s granulomatosis. Arthritis and
phritis. Journal of the American Society of Nephrology, 7, 33–9. Rheumatism, 43, 1021–32.
CHAPTER 12 cardiopulmonary manifestations of vasculitis 155

Reuter, M., Schnabel, A., Wesner, F., Tetzlaff, K., Risheng, Y., Gross, W.L., versus cyclophosphamide for ANCA-associated vasculitis. New England
and Heller, M. (1998). Pulmonary Wegener’s granulomatosis: correla- Journal of Medicine, 363, 221–32.
tion between high-resolution CT findings and clinical scoring of disease Stuck, A.E., Minder, C.E., and Frey, F.J. (1989). Risk of infectious complica-
activity. Chest, 114, 500–6. tions in patients taking glucocorticoids. Review of Infectious Diseases,
Rodriguez, W., Hanania, N., Guy, E., and Guntupalli, J. (2002). 11, 954–63.
Pulmonary-renal syndromes in the intensive care unit. Critical Care Suzuki, R., Morita, H., Komukai, D., Hasegawa, T., Nakao, N., Ideura, T., and
Clinics, 18, 881–95, x. Yoshimura, A. (2003). Mixed cryoglobulinemia due to chronic hepatitis
Roncoroni, A.J., Alvarez, C., and Molinas, F. (1992). Plexogenic arteriopathy C with severe pulmonary involvement. Internal Medicine, 42, 1210–4.
associated with pulmonary vasculitis in systemic lupus erythematosus. Tacke, C.E., Kuipers, I.M., Groenink, M., Spijkerboer, A.M., and Kuijpers,
Respiration, 59, 52–6. T.W. (2011). Cardiac magnetic resonance imaging for noninvasive
Saadoun, D., Asli, B., Wechsler, B., Houman, H., Geri, G., Desseaux, K., assessment of cardiovascular disease during the follow-up of patients
Piette, J.C., Huong Du, L.T., Amoura, Z., Salem, T.B., Cluzel, P., Koskas, with Kawasaki disease. Circulation Cardiovascular Imaging, 4, 712–20.
F., Resche-Rigon, M., and Cacoub, P. (2012). Long-term outcome of Travis, W.D., Hoffman, G.S., Leavitt, R.Y., Pass, H.I., and Fauci, A.S. (1991).
arterial lesions in Behcet disease:  a series of 101 patients. Medicine Surgical pathology of the lung in Wegener’s granulomatosis. American
(Baltimore), 91, 18–24. Journal of Surgical Pathology, 15, 315–33.
Sable-Fourtassou, R., Cohen, P., Mahr, A., Pagnoux, C., Mouthon, L., Jayne, Utzig, M.J., Warzelhan, J., Wertzel, H., Berwanger, I., and Hasse, J. (2002).
D., Blockmans, D., Cordier, J.-F., Delaval, P., Puechal, X., Lauque, Role of thoracic surgery and interventional bronchoscopy in Wegener’s
D., Viallard, J.-F., Zoulim, A., Guillevin, L., and French Vasculitis granulomatosis. Annals of Thoracic Surgery, 74, 1948–52.
Study Group (2005). Antineutrophil cytoplasmic antibodies and the Uzun, O., Akpolat, T., and Erkan, L. (2005). Pulmonary vasculitis in Behçet’s
Churg-Strauss syndrome. Annals of Internal Medicine, 143, 632–8. disease: A cumulative analysis. Chest, 127, 2243–53.
Schiavone, W.A., Ahmad, M., and Ockner, S.A. (1985). Unusual cardiac com- Vanoli, M., Castellani, M., Bacchiani, G., Cali, G., Mietner, B., Origgi, L., and
plications of Wegener’s granulomatosis. Chest, 88, 745–8. Scorza, R. (1999). Non-invasive assessment of pulmonary artery involve-
Schnabel, A., Holl-Ulrich, K., Dalhoff, K., Reuter, M., and Gross, W.L. (1997). ment in Takayasu’s arteritis. Clinical and Experimental Rheumatology,
Efficacy of transbronchial biopsy in pulmonary vaculitides. European 17, 215–8.
Respiratory Journal, 10, 2738–43. Vats, K.R., Vats, A., Kim, Y., Dassenko, D., and Sinaiko, A.R. (1999).
Schrader, M.L., Hochman, J.S., and Bulkley, B.H. (1985). The heart in pol- Henoch-Schonlein purpura and pulmonary hemorrhage: a report and
yarteritis nodosa:  a clinicopathologic study. American Heart Journal, literature review. Pediatric Nephrology, 13, 530–4.
109, 1353–9. Viegi, G., Fornai, E., Ferri, C., Di Munno, O., Begliomini, E., Vitali, C.,
Seyahi, E., Melikoglu, M., Akman, C., Hamuryudan, V., Ozer, H., Hatemi, G., Melocchi, F., Bombardieri, S., and Paoletti, P. (1989). Lung function
Yurdakul, S., Tuzun, H., Oz, B., and Yazici, H. (2012).Pulmonary artery in essential mixed cryoglobulinemia: a short-term follow-up. Clinical
involvement and associated lung disease in Behçet disease: a series of 47 Rheumatology, 8, 331–8.
patients. Medicine (Baltimore), 91, 35–48. Walsh, M., Flossmann, O., Berden, A., Westman, K., Hoglund, P., Stegeman, C.,
Sharma, B.K., Jain, S., and Sagar, S. (1996). Systemic manifestations of and Jayne, D. (2012). Risk factors for relapse of antineutrophil cytoplasmic
Takayasu arteritis:  the expanding spectrum. International Journal of antibody-associated vasculitis. Arthritis and Rheumatism, 64, 542–8.
Cardiology, 54, S149–154. Weir, I.H., Muller, N.L., Chiles, C., Godwin, J.D., Lee, S.H., and Kullnig, P. (1992).
Sheehan, R.E., Flint, J.D., and Muller, N.L. (2003). Computed tomography Wegener’s granulomatosis:  findings from computed tomography of the
features of the thoracic manifestations of Wegener granulomatosis. chest in 10 patients. Canadian Association of Radiologists Journal, 43, 31–4.
Journal of Thoracic Imaging, 18, 34–41. Weiss, M.A. and Crissman, J.D. (1985). Segmental necrotizing glomerulone-
Siburian, G., Hashimoto, Y., and Numano, F. (1993). Ventricular arrhythmias phritis: diagnostic, prognostic, and therapeutic significance. American
in Takayasu arteritis. International Journal of Cardiology, 40, 243–9. Journal of Kidney Diseases, 6, 199–211.
Sinico, R.A. and Bottero, P. (2009). Churg-Strauss angiitis. Best Practice and Wegener's Granulomatosis Etanercept Trial (WGET) Research Group
Research Clinical Rheumatology, 23, 355–66. (2005). Etanercept plus standard therapy for Wegener’s granulomatosis.
Sinico, R.A., Di Toma, L., Maggiore, U., Bottero, P., Radice, A., Tosoni, C., New England Journal of Medicine, 352, 351–61.
Grasselli, C., Pavone, L., Gregorini, G., Monti, S., Frassi, M., Vecchio, F., Wilcke, J.T., Nielsen, P.K., and Jacobsen, T.N. (2003). Reversible complete
Corace, C., Venegoni, E., and Buzio, C. (2005). Prevalence and clinical heart block due to Wegener’s granulomatosis. International Journal of
significance of antineutrophil cytoplasmic antibodies in Churg-Strauss Cardiology, 89, 297–8.
syndrome. Arthritis and Rheumatism, 52, 2926–35. Worthy, S.A., Muller, N.L., Hansell, D.M., and Flower, C.D. (1998).
Solans, R., Bosch, J.A., Perez-Bocanegra, C., Selva, A., Huguet, P., Alijotas, J., Churg-Strauss syndrome: the spectrum of pulmonary CT findings in 17
Orriols, R., Armadans, L., and Vilardell, M. (2001). Churg-Strauss syn- patients. AJR American Journal of Roentgenology, 170, 297–300.
drome: outcome and long-term follow-up of 32 patients. Rheumatology, Yamada, I., Nakagawa, T., Himeno, Y., Kobayashi, Y., Numano, F., and
40, 763–71. Shibuya, H. (2000). Takayasu arteritis:  diagnosis with breath-hold
Specks, U. (2003). Pulmonary vasculitis. In Interstitial lung disease, 4th edn contrast-enhanced three-dimensional MR angiography. Journal of
(ed. M.I. Schwarz and T.E. King). B. C. Decker, Hamilton. Magnetic Resonance Imaging, 11, 481–7.
Stone, J. H., Merkel, P.A., Spiera, R., Seo, P., Langford, C.A., Hoffman, G.S., Yokoi, K., Akaike, M., Nishiuchi, T., Kawai, H., and Saito, S. (1992). Scar for-
Kallenberg, C.G., St Clair, E.W., Turkiewicz, A., Tchao, N.K., Webber, mation in the cardiac conduction system of a patient with Takayasu’s
L., Ding, L., Sejismundo, L.P., Mieras, K., Weitzenkamp, D., Ikle, D., arteritis. Cardiology, 81, 378–83.
Seyfert-Margolis, V., Mueller, M., Brunetta, P., Allen, N.B., Fervenza, Zamora, M.R., Warner, M.L., Tuder, R., and Schwarz, M.I. (1997). Diffuse
F.C., Geetha, D., Keogh, K. A., Kissin, E.Y., Monach, P.A., Peikert, alveolar hemorrhage and systemic lupus erthematosus (SLE).: clinical
T., Stegeman, C., Ytterberg, S.R., and Specks, U. (2010). Rituximab presentation, histology, survival and outcome. Medicine, 76, 192–202.
CHAPTER 13

Vasculitic neuropathy
Shin J. Oh

Introduction 1984a; Bell and Weddell 1984b). In addition, there is a poorly


developed smooth muscle layer (McManis et al. 1993), so periph-
Peripheral neuropathy is a common and important clinical mani- eral nerve tissue is virtually incapable of autoregulating blood flow
festation of systemic necrotizing vasculitis (SNV). Vasculitic neu- and is susceptible to the small changes in perfusion pressure that
ropathy is the result of ischaemic damage due to occlusion of blood may occur with a vasculitic process. Because of this vulnerability, a
vessels associated with an inflammatory process in the vessel walls central fascicular fibre loss does occur in vasculitic neuropathy and
of the vasa nervorum. It can occur either as a manifestation of mul- is regarded as a typical pathological feature of ischaemic neuropa-
tisystem involvement or as an independent disease process, such as thy (Dyck et al. 1972).
non-systemic vasculitic neuropathy. Though vasculitic neuropathy Dyck et al. (1972) found in patients with vasculitic neuropathy
is relatively rare, its recognition is important because it is poten- associated with rheumatoid arthritis that the most vulnerable area
tially treatable. In the past few decades, remarkable progress has of the major nerves is in the mid-upper arm and mid-thigh levels,
been made in various aspects of vasculitic neuropathy. This chapter presumably because they are in a watershed zone of poor perfusion.
presents an overview of the classical concepts. At these levels, they found focal areas of nerve fibre degeneration,
largely in the centres of fascicles (central fascicular degeneration).
Vulnerability of peripheral nerves Distal to these levels, central fascicular degeneration became more
pronounced, with more fascicles becoming affected and broaden-
to vasculitic neuropathy ing of the lesions in the fascicles already damaged. In view of the
The hallmark of all SNV is inflammation and necrosis of the striking similarity between central fascicular degeneration in vas-
blood vessels, typically involving medium- and small-sized arter- culitic neuropathy and that seen in diabetic ophthalmoplegia, in
ies. Since the vasa nervorum in the peripheral nerve falls into the which ischaemia is also implicated, this pattern of fibre degenera-
spectrum of small-sized arteries and arterioles, it is not surprising tion may be typical of ischaemic lesions in the nerve.
that peripheral neuropathy is a common manifestation of SNV. The
vessels responsible for vasculitic neuropathy are predominantly the
arterioles of the epineurium, which typically have a diameter rang-
Pathology of vasculitic neuropathy
ing from 30 to 300 µm (Conn and Dyck 1984; Dyck et al. 1972). To render a definite diagnosis of vasculitic neuropathy, the unmis-
Typical lumina measure 10–25 µm for transperineurial arterioles takable histological features of vasculitis must be present: active,
and 10–20 µm for epineurial capillaries (Beggs et al. 1991; Dyck inactive, or healed necrotizing changes and infiltration of inflam-
et al. 1985; Giannini and Dyck 1993). matory cells within the vessel wall. Several histological types of
The microvasculature of peripheral nerves has several unique arterial changes are described in vasculitic neuropathy (Table 13.1).
features (McManis et al. 1993). There is a rich anastomotic blood These are described in this Section.
supply through two functionally distinct vascular systems:  the
extrinsic system composed of the epineurial vessels and regional Perivascular infiltration of inflammatory
arteries, arterioles, and venules, and the intrinsic system of the lon- mononuclear cells
gitudinal microvessels within the fascicles themselves. These two This is an early and mild arterial insult according to Dyck et  al.
systems are linked by a complex system of interconnecting ves- (1972) and occurs without any intramural necrosis or cellular infil-
sels, which provide high blood flow in the baseline state (McManis tration (Figure 13.1a). This alone is not enough to be diagnostic
et al. 1993; Sladky et al. 1985). This rich blood supply, along with of vasculitis because a similar finding is observed in inflamma-
the capacity of nerves to function relatively well with anaerobic tory neuropathies, especially in acute forms. However, there are
metabolism, makes peripheral nerves relatively resistant to ischae- some histological features that are helpful in differentiating these
mia. Only with intensive involvement of the vasa nervorum does disorders:  in vasculitic neuropathies, axonal degeneration is the
ischaemic damage occur. predominant finding, whereas in inflammatory neuropathies, seg-
On the other hand, some characteristics of the endoneuria1 ves- mental demyelination and endoneurial inflammatory cells are typi-
sels actually render nerves susceptible to ischaemia. The endoneu- cal findings. An exception has been found in vasculitic neuropathy
rial capillaries are larger and more widely spaced, particularly in the associated with HIV; inflammatory cells are often present in the
central fascicular regions, than in other tissues (Bell and Weddell endoneurial space and small endoneurial blood vessels are affected
158 SECTION 3 clinical manifestations common to vasculitis

Table 13.1 Pathological criteria of definite and probable vasculitis


Definite Probable
Wees et al. Type I: active vasculitis—definite fibrinoid necrosis with Type III: the presence of perivascular infiltration of inflammatory cells and active axonal
(1981) infiltration of inflammatory cells within the vessel wall, or degeneration without any definite intramural infiltration of inflammatory cells
Type II: inactive (or healed) vasculitis—concentric fibrous
scarring and intramural thickening of the vessel walls,
and minimal intramural and perivascular infiltration of
inflammatory cells
Dyck et al. 1. Inflammation of the vessel wall and 1. Mural or perivascular inflammation without tissue necrosis, with or without intimal
(1987) proliferation or
2. Necrosis of the wall 2. Evidence of necrosis or previous necrosis of the arterioles wall without inflammation,
with or without intimal proliferation and with or without focal or multifocal fibre
degeneration.
Hawke et al. 1. Vessel wall necrosis and an inflammatory cell reaction An inflammatory reaction in or around a vessel with a diameter greater than 30 µm
(1991)
Davies 1. Vessel wall necrosis with perivascular or transmural 1. Perivascular inflammatory cell in the medium-sized ( >120 µm) vessels with
(1994) infiltration of inflammatory cells or prominent acute axonal degeneration or focal nerve damage with aberrant
regenerating nerve cell clusters or
2. Perivascular inflammatory infiltrates with evidence of 2. Small-vessel cuffing with segmental fibre degeneration or acute axonal degeneration
previous vessel wall necrosis (fibrous obliteration with in 40% of fibres in the teased nerve
or without recanalization, disruption of the internal
elastic lamina or haemosiderin within the vessel wall
Collins 1. Transmural inflammatory cell infiltration in at least 1. Perivascular inflammatory cells and at least one other supportive pathological
et al. (2000) one blood vessels combined with signs of vascular features: vascular thickening and sclerosis, narrowing or obliteration of the lumen,
injury such as fibrinoid necrosis, endothelial disruption, thrombosis with or without recanalization, epineurial capillary proliferation,
fragmentation of the internal elastic lamina, or periadventitial haemosiderin, asymmetric nerve fibre loss, or Wallerian-like
haemorrhage into the vessel wall degeneration
Vital (2006) Necrotizing vasculitis: vessel wall infiltration by Perivascular infiltrates in the epi- or perineurium when associated with at least one of
inflammatory cells associated with the areas of fibrinoid the following lesion: regenerating small vessels, endoneurial purpura, asymmetric nerve
necrosis sometimes replaced by fibrosis fibre loss, and asymmetrical distribution of acute axonal degeneration with ovoids well
visible on semi-thin sections

Said et al. (1988) Transmural infiltration of small epineurial, perineurial arteries with inflammatory cells; leukocytoclasia; fibrinoid necrosis; destruction of the internal elastic lamina; occlusion
of the lumen; and the usual sparing of adjacent venules.
Kissel et al. (1985) Segmental wall necrosis and transmural inflammatory cell infiltration in at least one epineurial blood vessel. (Perivascular inflammation, central fibre atrophy or selective
fascicular atrophy is excluded.)
Vital (2006) Microvasulitis: inflammatory cells in the vessel walls and sometimes around them, in the absence of vessel wall necrosis in small vessels with few smooth-muscle fibres and no
internal elastic lamina.

(Said 1995). Thus, we make a diagnosis of ‘probable vasculitis’ when adventitia are typically found. Active vasculitis represents a Type
perivascular infiltrates of inflammatory cells are present together I lesion in our classification (Wees et al. 1981). With active vasculi-
with axonal degeneration, there is a decreased population of mye- tis, haemorrhage into surrounding tissue may occur, sometimes in
linated fibres, and clinical findings are compatible with vasculitis a perineurial or subperineurial crescentic pattern.
(Wees et al. 1981). This is a Type III lesion according to our clas-
sification (Wees et al. 1981) (Table 13.1). ‘Inactive vasculitis’
This is manifested by chronic changes including concentric fibrous
‘Active vasculitis’ scarring and thickening of the intima and media with minimal
This is represented by acute changes, including fibrinoid necrosis intramural and perivascular infiltrates of inflammatory cells, lym-
of the media with intramural and perivascular infiltrates of inflam- phocytes, and plasma cells (Figure 13.1d). Splitting and actual over-
matory cells, polymorphonuclear leukocytes, lymphocytes, and growth of the internal elastic membrane usually accompany this
eosinophils (Figure 13.1b,c). Polymorphonuclear leukocytes may lesion, which is Type II in our classification (Wees et al. 1981).
be prominent, but usually lymphocytes predominate in the vessel
wall and perivascular area. Eosinophils are present in vasculitis of Healed vasculitic lesions
various aetiologies but marked eosinophilic infiltration suggests These are indicative of previous severe injury to the arterial wall.
eosinophilic granulomatosis with polyangiitis (EGPA, formerly They are characterized by perivascular and intramural fibrosis
known as Churg–Strauss syndrome) (Midroni and Bilbao 1995; with fragmentation of the internal elastic membrane and narrow-
Oh et al. 1986). Together with these cardinal findings, dissolution ing, occlusion, and calcification of the lumen or recanalization of
of the internal elastic membrane and oedema or thickening of the the previously occluded lumen. Haemosiden-laden macrophages,
CHAPTER 13 vasculitic neuropathy 159

(a) (b)

(c) (d)

(e) (f)

Fig. 13.1 (a) Perivascular collection of mononuclear inflammatory cells (Type III) in the small arteriole in the epineurial space. There is no intramural infiltration of
inflammatory cells or fibrinoid necrosis. Paraffin section H & E, 250 ×. (b) Active vasculitis (Type I) in the larger arteriole in the epineurial space. Intramural infiltration
of mononuclear cells and fibrinoid necrosis of muscular and adventitial layers and near occlusion due to an intimal thickening. The arteriole is enlarged due to active
vasculitic process. Paraffin section. H & E, 100 ×. (c) Active vasculitis (Type I) in the arteriole in the perimysial space in muscle. Intimal thickening, intramural infiltrations of
mononuclear inflammatory cells, and fibrinoid necrosis of muscular and adventitial layers of the arteriole. The arteriole is enlarged due to active vasculitic process. Frozen
section. H & E. 100 ×. (d) Inactive vasculitis (Type II). Thick intimal layer and scattered intramural inflammatory cells in the muscular and adventitial layers of small arteriole
in the epineurial space. Paraffin section H & E, 100 ×. (e) Prominent myelin-digestion chambers (vacuoles) indicative of axonal degeneration in one nerve fascicle. Myelin
remnants (ovoids) are stained red. Frozen section. Modified trichrome. 100 ×. (f) Selective fascicular atrophy indicative of ischaemic neuropathy. Loss of myelinated fibres in
one half of the fascicle (arrow) is due to occlusion of vasa nervorum supplying blood to this section of fascicle. Myelinated fibres are stained red. 100 ×.

indicative of old haemorrhage, may cluster in a periadvential loca- helps differentiate these two different processes. Fragmentation
tion. There are no perivascular or intramural inflammatory cells. of the internal elastic membrane is suggestive of healed vasculitis.
This lesion may mimic atherosclerotic changes; however, care- Midroni and Bilbao (1995) occasionally observed miniature bun-
ful study of the vessels with connective tissue and elastin stains dles of aberrant regenerating axons, reminiscent of a traumatic
160 SECTION 3 clinical manifestations common to vasculitis

neuroma. Schroder (1986) has drawn attention to the reactive pro- Table 13.2 Distribution of definite and probable vasculitis
liferation of capillaries that can occur in the epineurium after a vas-
cular insult, although this observation is not specific to vasculitis. Type of vasculitis No. of cases Definite Probable
Thus, these findings should be regarded as clues suggestive of the
Wees et al. (1981) Systemic 17 15a 1
presence of remote vasculitis, but not indicative or diagnostic of
vasculitis. Dyck et al. (1987) Systemic 45 26 13b
Non-systemic 20 5 15
Axonal degeneration
Hawke et al. (1991) Systemic 34 30 3c
Axonal degeneration is the predominant pattern and is due to the
ischaemic damage of the nerve (Figure 13.1e) (Hawke et al. 1991; Davies (1996) Systemic 76 57 19
Loveshin and Kernohan 1948; Wees et al. 1981; Vital 2006). Said Non-systemic 25 19 6
et al. (1988b) observed axonal degeneration in an average of 65%
Claussen et al. (2000) Systemic 45 33d 12
of nerve fibres. The degree of axonal degeneration depends on the
activity of the vasculitic process. Prominent axonal degeneration is Collins et al. (2000) Systemic 25 15 10
invariably seen with active vasculitic lesions and is best observed Non-systemic 11 7 4
in the longitudinal cuts on frozen sections. Late in the disease, the Vital (2006) Combined 109 85e 24
process of axonal degeneration is more complete and few, if any,
fibres remain. a Type I in 14 cases and type II in 1 case. In 1 case, vasculitis in muscle biopsy.
b In 6, nerve biopsy is non-diagnostic. Definite vasculitis in other tissues.

Patterns of degeneration c In 1 case, nerve biopsy is non-diagnostic. However, autopsy showed definite vasculitis in

other tissues.
Various patterns of degeneration of fibres are noted, ranging from d Type I, in 27 cases and Type II in 6 cases.

central fascicular degeneration to total fascicular atrophy depend- e 60 with necrotizing vasculitis and 25 with microvasculitis.

ing on the severity of neuropathy (Figure 13.1f). Although central


fascicular degeneration is typical of ischaemic neuropathy, total
fascicular atrophy is also typical as a result of the total occlusion of criteria of vasculitis are too stringent to be clinically useful in vas-
arterioles that supply blood to the involved fascicle. culitic neuropathy (Table 13.1). In about one-quarter of patients
It should be emphasized that any combination of these changes with vasculitic neuropathy, the criteria for definite vasculitic neu-
may be found in a single sural nerve biopsy, indicating an ongoing ropathy are lacking (Table 13.2). This is most probably due to the
process (Hawke et al. 1991). In most cases, the nerve lesions appear absence of medium- and small-sized arteries in the nerve, which
to result from the summation of lesions of different ages of blood are predominantly involved in SNV. Although the criteria for
vessels (Fujimura et al. 1991). In autopsy series (Dyck et al. 1972; probable vasculitis differ from author to author, perivascular infil-
Loveshin and Kernohan 1948), all these changes could be seen tration (cuffing) of inflammatory cells must be present. This find-
along the course of an involved peripheral nerve. Dyck et al. (1972) ing alone is not sufficient for the diagnosis of vasculitis because it is
stated that chronic changes are usually seen in nerve of patients a non-specific finding in nerve pathology. This is especially true in
with long-standing, non-progressive neuropathy, and acute lesions the context of chronic inflammatory demyelinating polyneuropa-
in patients with clinically acute neuropathy. thy (CIDP). Predominant axonal degeneration favours vasculitis,
It is not possible to diagnose specific vasculitic syndromes from whereas segmental demyelination and endoneurial inflammatory
nerve biopsy specimens. In general, the calibre of involved vessels cells favour CIDP. Because of this, we recommend that perivas-
may allow one to assign a given biopsy to one of two broad groups cular infiltration of inflammatory cells and axonal degeneration
(Chalk et al. 1993a). Vasculitic involvement of larger (100–250 µm) be considered as the minimal diagnostic criteria for probable
epineurial arterioles is a typical feature of polyarteritis nodosa vasculitis.
(PAN), EGPA, granulomatosis with polyangiitis (GPA, formerly The criteria for probable vasculitic neuropathy are appropriate to
known as Wegener’s granulomatosis), rheumatoid vasculitis, and use in the clinical diagnosis of vasculitic neuropathy. Three studies
probably microscopic polyangiitis (MPA). Predominant involve- showed that all patients with probable vasculitis on nerve biopsy
ment of smaller (<100 µm) epineurial arterioles is more suggestive were proven to have systemic vasculitis by other means. In one
of Sjögren’s syndrome, systemic lupus erythematosus (SLE), and patient with Type III nerve biopsy findings in Wees’s series (1981),
non-systemic vasculitis of nerves (Chalk et al. 1993a). Involvement angiography of the celiac axis showed typical microaneurysms of
of epineurial veins occurs more commonly in GPA and EGPA than PAN. In Dyck’s 13 cases of probable vasculitic neuropathy, vascu-
in PAN (Lie 1990). Vasculitis of endoneurial vessels is uncommon litis was confirmed by liver and kidney biopsies and post-mortem
and would theoretically be classified with the hypersensitivity vas- findings (Dyck et al. 1987). In Hawke’s series, vasculitis was con-
culitides (leukocytoclastic vasculitis) according to Midroni and firmed in each case by muscle biopsy, kidney biopsy, or autopsy
Bilbao (1995). In HIV-induced vasculitic neuropathy, mononu- (Hawke et al. 1991).
clear cells are often present in the endoneurial space and vasculitic
change is seen more often in endoneurial vessels (Calabrese et al.
1989; Said et al. 1987). Pathogenesis of vasculitic neuropathy
Although a definite histological diagnosis of vasculitis in the It is generally accepted that the nerve fibre degeneration in this
nerve should be based on the demonstration of definite vasculitis disorder is ischaemic secondary to vasculitis in the vasa nervorum
(active or inactive vasculitis (Type I and II lesions)), the concept of (Asbury and Johnson 1978; Dyck et al. 1972). However, there are
probable vasculitis in the nerve is introduced because the definite different opinions with regard to the appearance of the consequent
CHAPTER 13 vasculitic neuropathy 161

nerve alteration following ischaemia. Dyck et  al. (1972) did not leads to activation of inflammatory mechanisms and infiltration
believe that ‘infarction’ occurs in nerve. Their view was based on a of polymorphonuclear leukocytes with subsequent tissue necrosis
detailed study of the involved nerves in rheumatoid vasculitic neu- producing the pathological picture of leukocytoclasis (Fauci et al.
ropathy, which did not reveal any evidence of infarction, defined 1978). More recent studies confirmed that this mechanism is also
as a circumscribed area of necrosis of all cellular elements leading operative in vasculitic neuropathy. Kissel et al. (1989) found immu-
to liquefaction with a border of macrophages (such as occurs in nofluorescent evidence for immune complex involvement in 85%
the brain). They maintained that the ‘infarcts of nerves’ described of biopsies in their series, and only in those vessels that also had
by Kernahan and Woltman (1938) were not infarcts, but prob- intense cellular infiltrates. In Hawke’s series (Hawke et al. 1991),
ably Renault’s corpuscles. On the other hand, Asbury and Johnson immunofluorescent staining was positive on nerves in 100% of 20
(1978) believe that frank infarct necrosis of the nerve does occur in patients. In 16 cases, immunoglobulin, complement C3, and fibrin-
vasculitic, amyloid, and diabetic neuropathies. ogen were present in endoneurial or perineurial blood vessels. In
The pattern of neuropathic involvement in vasculitic neuropathy four cases, only C3 and fibrinogen were found. In control nerves,
depends on the extent and temporal progression of the ischaemic occasional faint fluorescence was detected with C3 on endoneurial
changes (Fathers and Fuller 1996). Mononeuropathy multiplex, capillaries, but there was no fluorescent staining of larger epineu-
the classic pattern of neuropathy in vasculitic neuropathy, is due rial or perineurial vessels. There were four cases without cellular
to lesions in larger vessels leading to whole nerve trunk infarc- invasion of the sural nerve in which there was immunofluorescent
tion in nerves scattered throughout the body (Figure 13.2). On the evidence of immune complex deposition. Panegyres et al. (1990)
other hand, asymmetrical polyneuropathy (overlapping monon- also observed immune complex deposits in 13 (72%) of 18 cases.
europathy multiplex) results from a confluence of patchy discrete Kissel et  al. (1989) have questioned the role of immune com-
infarction of smaller vessels in many individual peripheral nerves, plexes, as they found that cellular infiltrates in vascular lesions
together with superimposed mononeuropathy from damage of were composed predominantly of T cells (71%) and macrophages
whole nerve trunks. Symmetrical polyneuropathy is a consequence (27%). Two-thirds of the T cells were of the CD8+ subset. In con-
of a more diffuse peripheral nerve ischaemia due to multiple small trast, B cells and polymorphonuclear leukocytes were extremely
lesions, which are more likely to affect longer nerves. rare, and a leukocytoclastic reaction was never observed. In con-
The classic theory of the pathogenesis of immune-mediated vas- trast, Panegyres et al. (1990) found the ratio of lymphocytes to mac-
culitis is that immune complex deposition in blood vessel walls rophages to vary from 3:1 to 1:23, with substantial numbers of B
lymphocytes in the three nerves in which lymphocyte subtyping
was performed. Neither group found any correlation between the
(a) (b) (c) types of inflammatory cells and the presence or type of associated
systemic disease. On the basis of these findings, Kissel et al. (1989)
suggested a cytotoxic T-cell-mediated process as a primary mecha-
nism of vascular damage in peripheral nerve vasculitis.

Biopsy diagnosis of vasculitic neuropathy


Nerve biopsy in diagnosis of vasculitic neuropathy
Nerve biopsy is often indicated in vasculitis and vasculitic neuropa-
thy for three reasons (Oh 1990): (1) peripheral nerves, including
the sural nerve, are commonly involved in vasculitic neuropathy
(Wees et al. 1981); (2) the diagnostic yield is high when the sural
nerve conduction is abnormal (Claussen et  al. 2000; Wees et  al.
1981); and (3)  vasculitis and vasculitic neuropathy are treatable.
In Oh’s series of 385 nerve biopsies over a 16-year period, vascu-
litic neuropathy was the most commonly diagnosed specific form
of neuropathy, accounting for 12% (Oh 1990). When clinical fea-
tures suggest the possibility of systemic vasculitis, detailed neuro-
logical and electrophysiological tests are mandatory. Asymptomatic
peripheral neuropathy was detected by the nerve conduction study
(NCS) in an average of 10% of patients with vasculitic neuropathy
(see Section Asymptomatic Vasculitic Neuropathy) (Claussen et al.
2000). When the patient has vasculitic neuropathy by neurological
and electrophysiological tests, nerve biopsy is definitely indicated
Fig. 13.2 (a) Mononeuropathy multiplex: right peroneal, left ulnar, and if obtaining other tissues for diagnosis is not feasible or warranted.
right superficial radial sensory radial neuropathy, occurring due to ischaemic Three nerves most suitable for biopsy are the sural, superficial fibu-
degeneration of the whole nerve of these individual nerves. Shaded area of the lar (peroneal), and superficial radial nerves. The most commonly
nerve represents the ischaemic degeneration. (b) Asymmetrical polyneuropathy
due to more patch damage in the scattered many nerves, but not corresponding
biopsied nerve is the sural nerve because it is easily identifiable,
to the named nerves. Lined areas represent milder degree of involvement. is commonly involved in vasculitic neuropathy, and can reliably
(c) Symmetrical polyneuropathy due to multiple small lesions in the nerve be tested electrophysiologically. On the other hand, Said, Kissel,
fascicles, which are more likely to affect longer nerves. and Vital recommend superficial fibular (peroneal) nerve biopsy
162 SECTION 3 clinical manifestations common to vasculitis

because it allows simultaneous muscle biopsy from the adjacent (1995), characteristic lesions may be present on segments of the
peroneus brevis muscle through a single incision on the lateral arteries as short as 50  µm. Thus, in cases of suspected vasculitic
aspect of the leg (Kissel and Mendell 1992; Said et al. 1988b; Vital neuropathy, recommendations are to cut and stain as many sec-
2006). Oh and colleagues prefer sural nerve biopsy because they tions of the biopsied nerve as possible.
have found the superficial fibular (peroneal) nerve to be less reli-
able in the nerve conduction testing and much harder to locate ana- Muscle biopsy in diagnosis
tomically than the sural nerve. of vasculitic neuropathy
To increase the diagnostic yield in vasculitic neuropathy, the fol- Muscle was the first tissue chosen for diagnosis of vasculitis and
lowing guidelines and precautions are recommended. was the favoured diagnostic biopsy site until the nerve biopsy was
First, the sural nerve should be tested electrophysiologically and introduced in 1968 (Dyck and Lofgren 1968). In 1952, Maxeiner
the more impaired sural nerve biopsied. In one report, vasculitic et  al. reported a 13% diagnostic sensitivity of vasculitis in 136
neuropathy was diagnosed by sural nerve biopsy in 100% of 15 muscles biopsies among 106 cases suspected of PAN and 35%
patients with sural nerve conduction abnormalities. Of these, 14 sensitivity in 26 cases of documented PAN. They also stated that
had vasculitis and one had probable vasculitis (Wees et al. 1981). the likelihood of positive biopsy was higher when a local lesion
Based on this finding, Wees et al. (1981) stated that abnormality in (nodule or tenderness) was present, compared with random sam-
the sural nerve conduction was prerequisite to the demonstration pling (30% versus 8%, respectively). In 1955, Garcia et al. reported
of vasculitis on biopsy, but this concept has been challenged. Seven seven cases of polyarteritis diagnosed by calf muscle biopsy, all of
cases of sural nerve biopsy-proven vasculitis with normal sensory which were later confirmed at autopsy. In the 1960s, Bleehen and
nerve conduction were reported (Davidson and Sundstrom 1988; Lovelace et al. described the technique of superficial fibular (pero-
Hawke et  al. 1991; Midroni and Bilbao 1995). In a series of 44 neal) sensory nerve and fibularis (peroneus) brevis muscle biopsy
confirmed cases of vasculitis the NCS on the biopsied sural nerve in polyarteritis (Bleehan et al. 1963; Lovelace 1964). Despite the
were abnormal in 38 of 39 patients tested (Claussen et al. 2000). general availability of nerve biopsy, muscle biopsy remained the
One patient with vasculitis had nerve conduction velocities (NCV) main diagnostic procedure for vasculitis at some medical centres
within the normal range (2 SD from the mean), but a relatively until the 1980s.
decreased amplitude in sensory compound nerve action potential Dahlberge et al. (1989) compared various diagnostic tests in 40
(CNAP) when compared with the same nerve on the other side. cases of suspected SNV and reported a diagnostic sensitivity of 19%
Among the three other cases with normal NCS of the biopsied sural among 26 muscle biopsies. Among 10 patients with proven SNV,
nerve, there was no evidence of vasculitis (Claussen et al. 2000). they found 50% sensitivity and concluded that the muscle biopsy
Thus, abnormal sural nerve conduction strongly correlates with the was the most valuable of the relatively safe procedures. However,
finding of vasculitis, suggesting that this test can be used to estimate they did not utilize nerve biopsy in any of their cases. Dyck et al.
the likelihood of a positive nerve biopsy. Normal sural nerve con- (1987) reported 25% diagnostic sensitivity of muscle biopsy in 12
duction does not preclude the diagnosis of vasculitis on the sural cases of proven vasculitis neuropathy. Said et al. (1988b) reported
nerve biopsy, but does decrease the likelihood of a positive biopsy. that fibularis (peroneus) brevis muscle biopsy had a diagnostic
Thus, when the sural nerve conduction is normal, we recommend sensitivity of 75% among 83 proven cases of vasculitic neuropa-
electrophysiological testing on the superficial fibular (peroneal) thy. This figure was exceptionally high and defied explanation. It is
and superficial radial sensory nerves, as these alternative nerves worth noting, however, that the same authors reported in 1995 that
may be biopsied. We have successfully used this approach to con- muscle biopsy had a diagnostic sensitivity of 54%, a figure more
firm vasculitis in two patients. Kissel and Mendell (1992) had to in agreement with the general consensus of other reports. At the
perform only two superficial radial nerve biopsies over a 15-year University of Alabama at Birmingham, a 17% diagnostic sensitiv-
period, both of which proved sufficient for diagnosis. In regard to ity of muscle biopsy was found among 115 cases of suspected SNV
the superficial fibular (peroneal) nerve, there are no published data and 42% sensitivity among 45 cases of proven vasculitic neuropathy
thus far that correlate the nerve conduction data and nerve biopsy (Claussen et al. 2000). Among eight studies, seven (all except Said’s
findings. 1988b study) have shown that nerve biopsy has a higher diagnostic
The second recommendation is that a biopsy specimen of the sensitivity than does muscle biopsy (Table 13.3).
entire cross-section of the nerve should be obtained. There is no Thus, in general, the diagnostic value of muscle biopsy in patients
role for fascicular nerve biopsy in suspected vasculitic neuropathy with suspected SNV is disappointingly low, indicating that this
because the vasa nervorum are located in the epineurial and peri- should not be the first choice of biopsy. If clinical findings and elec-
neurial space, which may not be obtained by fascicular biopsy. This trophysiological studies on the sural, superficial fibular (peroneal),
was unquestionably documented in the study by Dyck et al. (1972); and superficial radial nerves are normal and an affected nerve is not
all eight whole nerve biopsies demonstrated vasculitic neuropa- accessible for biopsy, then the muscle biopsy may be the only choice
thy while three of six fascicular biopsies failed to show vasculitic for tissue diagnosis.
change. In selecting a muscle for biopsy, clinical and needle electromyo-
Third, although this is not well documented, it seems a prudent graphy (EMG) data are useful. In one series, abnormal needle EMG
practice to perform the sural nerve biopsy early because glucocor- findings in the biopsied muscle did not discriminate between posi-
ticoid treatment may alter the histological features of vasculitides. tive and negative biopsies, but a trend was found between the pres-
Fourth, it is necessary to cut multiple sections from different ence of fibrillation and positive sharp waves and vasculitis on the
levels of the specimen since vasculitis is multifocal and segmen- muscle biopsy (Claussen et al. 2000). On this basis, it is preferable
tal. It has been our repeated experience that only a few sections of to biopsy a muscle that shows fibrillation and positive sharp waves
the biopsied nerve show the diagnostic change. According to Said on the needle EMG study.
CHAPTER 13 vasculitic neuropathy 163

Table 13.3 Diagnostic sensitivity of nerve and muscle biopsies in vasculitic neuropathy

Authors Patients No. cases Nerve biopsy Muscle biopsy Comments


Dyck et al. (1987) SNV, Biopsy proven VN 45 58% (26/45) 25% (3/12) Sural/gastrocnemius
Said et al. (1988) SNV, Biopsy proven VN 83 51% 75% Superficial fibular (peroneal)/peroneus brevis
Said (1995) SNV, Biopsy proven VN ? 62% 54% Same as above?
Claussen et al. (2000) SNV, suspectedc 115 39% 17% Sural in all/anterior tibialis in 2/3 of cases
SNV, biopsy proven VN 45 100% 42%
Collins et al. (2000) SNVd, suspected 70 27% 16% Superficial fibular (peroneal)/peroneus brevis
SNV, biopsy proven VN 22 90% 50%
Said (2005) SNV, biopsy proven VN 425 76.5% 59.5% Superficial fibular (peroneal)/peroneus brevis
Maxeiner et al. (1952) SNV, suspected 136a 13%
SNV, biopsy proven 26 35%
Dahlberg et al. (1989) SNV, suspected 26b 1 9%
Rapport (1993) SNV, suspected 29 20% Sural nerve biopsy
Cruz Martinez et al. (1988) SNV, biopsy proven VN 14 86% 79%
Dyck et al. (1987) NSV, biopsy proven VN 20 100% All by the sural nerve biopsy
Davies et al. (1996) NSV; biopsy proven VN 25 100% All by the sural nerve biopsy
Collins et al. (2003) NSV, biopsy proven VN 48 100% Sural in 30; SP/PB in 19; Sup radial in 2
Kararizou (2004) NSV, biopsy proven VN 22 100% All by the sural nerve biopsy
Vital (2006) CV, suspected 178e 62% Superficial fibular (peroneal)/peroneus brevis
Biopsy proven VNf 85 73% 59%
a 136 muscle biopsies from 106 cases suspected of polyarteritis nodosa.
b 26 muscle biopsies.
c Neuropathy/myopathy in 66 and no neuromuscular syndromes in 15. Others: no clinical information.
d Including 7 cases of NSV.
e Negative biopsy in 68 (38%) of 178 suspected vasculitis cases.
f Definite vasculitis cases only. Muscle biopsy: improved the yield of definite vasculitis by 27%.
SNV, systemic necrotizing vasculitis; NSV, non-systemic vasculitis; VN, vasculitic neuropathy; CV, combined systemic and non-systemic vasculitis.

Diagnostic sensitivity of simultaneous biopsy of nerve and muscle is widely advocated when considering
nerve and muscle biopsy vasculitis in the differential diagnosis. We usually do sural nerve
biopsy together with biopsy of the anterior tibialis or gastrocne-
The diagnostic sensitivity of simultaneously obtained nerve and
mius muscle. Another method is to perform the nerve biopsy first
muscle biopsy depends solely on the criteria used for patient
and, depending on the findings in the nerve, consider performing
selection. Among those with suspected SNV or vasculitic neurop-
muscle biopsy later.
athy, the diagnostic sensitivity averages 30% for the nerve biopsy
in contrast to 16% for the muscle biopsy (Table  13.3). Among
patients with confirmed vasculitic neuropathy, the diagnostic sen- Vasculitic neuropathy in systemic
sitivity averages 74% for the nerve biopsy and 55% for the mus- necrotizing vasculitis
cle biopsy. These findings indicate the superiority of nerve biopsy
over the muscle biopsy. Our data strongly support the higher sen- Incidence and risk factors
sitivity of nerve biopsy (39% of all cases) compared with muscle The exact incidence and prevalence of vasculitic neuropathy are
biopsy (17% of all cases) for patients with suspected and definite unknown. An approximation of the frequency of vasculitic neurop-
vasculitides. In none of our cases was the muscle biopsy positive athy can be ascertained from a report of the American College of
for vasculitis when the nerve biopsy was negative, but in three Rheumatology subcommittee on classification of vasculitis (Bloch
cases the muscle biopsy was more specific and resulted in a defi- et al. 1990; Hunder et al. 1990). In this study, 1000 new cases of vas-
nite diagnosis, increasing the diagnostic yield from 29 to 31%. culitis were detected at 48 medical centres over a 5.5-year period.
Thus our results support the finding that muscle biopsy is com- Since peripheral neuropathy occurs in up to 70% of patients with
plimentary to nerve biopsy in the detection of vasculitis. Previous primary vasculitic syndromes, approximately 700 new cases of vas-
studies also found that adding muscle biopsy to the nerve biopsy culitis neuropathy were estimated at 48 medical centres over the
increased the positive diagnostic yield by six to 27% (Hawke et al. study period, or three cases per year per centre. This figure closely
1991; Vital 2006). On the basis of these findings, the combined approximates that of several published series (Table 13.4).
164 SECTION 3 clinical manifestations common to vasculitis

Table 13.4 Frequency of cases with vasculitic neuropathy Table 13.5 Systemic diseases associated with vasculitic neuropathy

Authors Sources No of cases Yearsa Cases/year Vasculitic (%) Prevalence Frequency of


of diseases neuropathy
Wees and Oh Nerve/muscle biopsy 17 5 3.4
(1978) Primary vasculitic disease
Polyarteritis nodosa Rare 50–70
Dyck et al. (1987) Medical record/nerve 65b 22 2.9
biopsy Churg–Strauss syndromea Rare 64
Wegener’s granulomatosisb Rare 25
Said et al. (1988) Nerve/muscle biopsy 100 15 6.6 Giant cell arteritis Common in elderly 5–14
Oh (1990) 385 nerve/muscle 46 16 2.9 Microscopic polyangiitis Rare 7
biopsy ANCA-associated vasculitis Rare 8

Hawke et al. (1991) 1024 nerve biopsy 34 12 2.8 Rheumatoid diseases


Rheumatoid arthritis Common 10
Kissel (1994) 350 nerve biopsy 16 10 1.6 Systemic lupus erythematosus Common 2–18
Davies et al. (1996) 1559 nerve biopsy 25c 9 2.7 Sjögren’s syndrome Common 9
Progressive systemic sclerosis Uncommon 1.5
Collins et al. (2000) SPN/PBM biopsyd 22 10 2.2
Behçet’s syndrome Rare 5
Collins et al. (2003) Nerve/muscle biopsy 48c 20 2.4 Hypereosinophilic syndrome Rare 14
Kararizou et al. Nerve biopsy 22c 20 1.1 Hypersensitivity vasculitis Common 10
(2005) Other conditions with vasculitis
Said (2005) Nerve biopsy 425 34 12.5 Cryoglobulinaemia Rare 50
Malignancy Common Rare
Vital (2006) Nerve/muscle biopsy 109 15 7.3 HIV infection Variable 2
a Duration of years these cases were collected. Lyme disease Variable <1
b 45 systemic necrotizing vasculitic neuropathy and 20 non-systemic necrotizing vasculitic
a Churg–Strauss syndrome: eosinophilic granulomaotis with polyangiitis (EGPA).
neuropathy. b Wegener’s granulomatosis: graunomaotisis with polyangiitis (GPA).
c Non-systemic vasculitic neuropathy.
d SFN/PBM, superficial fibular (peroneal) nerve/ peroneal brevis muscle biopsy.

Vasculitic neuropathy is rare compared with other types of multiplex have symptoms for a shorter period (mean 9.2 weeks)
peripheral neuropathy. The most common cause for neuropathy in than patients with either an asymmetrical neuropathy (mean 32
the United States is diabetes mellitus. One study showed that 35% weeks) or a symmetrical neuropathy (mean 20 weeks) (Hawke
of 100 elderly (over 65 years of age) patients with disabling neu- et  al. 1991). This is probably due to clinicians’ higher index of
ropathy had one form or another of vasculitic neuropathy proved suspicion of vasculitic neuropathy in a setting of rapidly evolv-
by the nerve biopsy (Chia et al. 1996). According to our experience, ing mononeuropathy multiplex than in one of slowly progressing
this figure may be too high. asymmetrical or symmetrical polyneuropathy. Vasculitis-induced
The frequency of peripheral neuropathy in SNV syn- ischaemic changes in the nerve are not a selective process, and so
dromes ranges from 25% in EGPA to 70% in PAN (Table 13.5). both sensory and motor fibres are usually affected. All patients in
Peripheral neuropathy can be the initial manifestation of PAN. Hawke’s series (1991) had a sensorimotor peripheral neuropathy;
The frequency of peripheral neuropathy in the primary rheu- however, pure sensory neuropathy has also been reported (see
matic diseases ranged from 1.5% in progressive systemic scle- Section Sensory Neuropathy), so sensory neuropathy alone does
rosis to 18% in SLE. Most, though not all, cases of peripheral not rule out vasculitic neuropathy. Nerves in the legs are affected
neuropathy in primary rheumatic diseases are due to vasculitic more often than nerves in the arms. Pain and dysaesthesia are
neuropathy. common in vasculitic neuropathy, present in 53–70% of patients
(Hawke et al. 1991; Kissel et al. 1985). A typical clinical picture is
Clinical manifestations deep aching pain in the affected limb with subsequent develop-
Clinical clues and laboratory features suggesting vasculitis, includ- ment of unpleasant paraesthesia in the cutaneous distribution of
ing vasculitic neuropathy, are discussed in Chapter XX. Vasculitic the affected nerve. In vasculitic neuropathy due to SNV, common
neuropathy affects males and females equally and is most com- systemic symptoms are fever, anorexia, weight loss, and fatigue
mon in older individuals. The time course of disease varies from a (Kissel et al. 1985; Wees et al. 1981; Suppiah 2011). Systemic symp-
rapid onset over days to weeks (25% develop in less than 4 weeks) toms usually develop at the same time as vasculitic neuropathy but
to a more gradual evolution over many months (50% more than may precede it by more than 6 months (Hawke et al. 1991; Suppiah
2 months) (Hawke et al. 1991). When onset of disease is acute, it 2011). In non-vasculitic neuropathy, systemic features are absent
may mimic Guillain-Barré syndrome (GBS) (Suggs et  al. 1992). (Davies et al. 1996; Dyck et al. 1987; Kissel et al. 1985). In 1981,
In vasculitic neuropathy due to SNV, it is extremely rare to have a Wees published the first paper reporting asymmetrical and sym-
disease course of longer than 1 year (Hawke et al. 1991; Kissel et al. metrical polyneuropathy as manifestations of vasculitic neuropa-
1985; Wees et al. 1981) but in non-systemic vasculitic neuropathy, a thy, and polyneuropathy as the most common form of vasculitic
period of more than 12 months before diagnosis is not uncommon neuropathy. Since then it has been well accepted that there are
(Davies et  al. 1996). In general, patients with mononeuropathy three main patterns of neuropathy though the relative frequency
CHAPTER 13 vasculitic neuropathy 165

Table 13.6 Distribution of neuropathy patterns in vasculitic neuropathya

1981 1985 1991 1986 1995 2000 2002 1987 1996 2003 2005
Wees Kissel Hawks Bouche Midroni Claussen Collins Dyck Davies Collins Kararizou Total Total Total
et al. et al. et al. et al. and Bilbao et al. et al. et al. et al. et al. et al.
SNV SNV SNV SNV SNV SNV S/NSNVc NSVN NSVN NSDN NSVN SNV NSVN
Mononeuropathy 3 2 16 9 12 10 3d 12 10 6 9 55 (29%) 37 (32%) 92 (30%)
multiplex
Mononeuropathy (2)b (5) (7) (7) (14)
Asymmetrical 4 8 10 7 5 4 18d 4 7 37 4 56 (30%) 52 (45%) 108 (36%)
polyneuropathy
Symmetrical 3 6 8 3 14 21 1 4 8 5 8 56 (30%) 25 (22%) 81 (27%)
polyneuropathy
Sensory (1) (8) (2) (1) (11) (1) (13)
Asymptomatic 7 0 3 1 9 20 (12%) 0 20 (7%)
neuropathy
Myopathy (2) (3) (5) 0 (5)
Total cases 17 16 34 22 32 44 22 20 25 48 22 187 115 302
a These are compiled from the reports that classified the neuropathy patterns into three categories: mononeuropathy multiplex, asymmetrical polyneuropathy, and symmetrical

polyneuropathies. Said et al. (1988) and Cruz Marinez et al. (1988) series are excluded.
Said: 13 mononeuropathy, 62 mononeuropathy multiplex, 19 symmetrical neuropathy, and 6 no neuropathy.
Cruz Martinez: 5 mononeuropathy multiplex, 7 symmetrical polyneuropathy, 3 asymptomatic.
Vital (2006): Total cases: 178; all cases had peripheral neuropathy; mononeuropathy multiplex in 81, sensory neuropathy in 30, sensori-motor neuropathy in 19, and mononeuropathy in
7 cases.
b 2 cases are included in 3 cases of mononeuropathy multiplex.
c 7 cases had NSVN.
d This includes asymmetrical lumbar plexopathy.

SNV, systemic necrotizing vasculitic neuropathy; NSNV, non-systemic necrotizing vasculitic neuropathy.

varies from study to study (Table 13.6). A review of the literature cases of PAN. Cohen et al. (1980) used mononeuropathy multi-
confirms the finding that polyneuropathy (asymmetrical or sym- plex as a criterion for the diagnosis of PAN. Since mononeuropa-
metrical) is common, being observed in 55% of cases. The classical thy multiplex can be due to multiple causes, including multifocal
pattern of mononeuropathy multiplex was seen only in one-third motor and motor-sensory demyelinating neuropathies, this is no
of patients. Recognition of this concept is important; one should longer justifiable (Table 13.7). Of note, multifocal demyelinating
not exclude the possibility of vasculitic neuropathy because of the neuropathy is more common than vasculitic mononeuropathy
absence of mononeuropathy multiplex. multiplex in our clinic.

Mononeuropathy multiplex Mononeuropathy


Mononeuropathy multiplex has been touted as the classic mani- Mononeuropathy multiplex, by definition, requires the involve-
festation of peripheral neuropathy and the most common neu- ment of at least two nerves. There have been several reports of
rological manifestation in PAN. Mononeuropathy multiplex is mononeuropathy as a manifestation of vasculitic neuropathy
defined as the involvement of two or more individual nerves (Bouche et al. 1986; Said et al. 1988b; Wees et al. 1981). In 1981,
in more than one extremity, for example the ulnar nerve in Wees et  al. reported two cases of mononeuropathy among 17
one arm and the fibular (peroneal) nerve in a leg. In vasculitic patients with systemic vasculitic neuropathy: peroneal neuropa-
neuropathy, this is due to lesions in larger arterioles leading to thy in a patient with rheumatoid arthritis and anterior interosse-
whole-nerve trunk infarction. This ‘true mononeuropathy mul- ous neuropathy in a patient with PAN (Wees et al. 1981). In both
tiplex’ is thought to be the most distinctive pattern in vasculitic cases, NCS showed diffuse neuropathy and abnormal sural nerve
neuropathy (Kissel and Mendell 1992). Chalk et al. (1993a) wrote conduction. One group found five (11%) cases of mononeuropa-
the following: ‘Usually, vasculitic neuropathy presents as the syn- thy among 44 cases of vasculitic neuropathy (Claussen et al. 2000).
drome of multiple mononeuropathies, in which single periph- Said et al. (1988b) reported 13 cases of mononeuropathy among
eral nerves or cranial nerves are affected in succession. Abrupt 100 cases of vasculitic neuropathy:  peroneal neuropathy in 11,
onset of weakness, prickling paraesthesia, or sensory loss, often femoral neuropathy in one, and ulnar neuropathy in one. Bouche
associated with local pain, is characteristic. Occasional patients et al. (1986) also reported three cases of mononeuropathy: sciatic
develop a flurry of neuropathic deficits in several limbs during a neuropathy in one, peroneal nerve palsy in one, and ulnar neurop-
single day.’ In Ford and Siekert’s series (1965), 54% of patients had athy in one. This indicates that mononeuropathy is a manifestation
mononeuropathy multiplex. In Guillevin’s series (Guillevin et al. of vasculitic neuropathy in about 10% of patients with systemic
1992), mononeuropathy multiplex was reported in 70% of 182 necrotizing vasculitis.
166 SECTION 3 clinical manifestations common to vasculitis

Asymmetrical polyneuropathy presentation represents the most difficult diagnostic challenge for
Asymmetrical polyneuropathy is characterized by an asymmetri- clinicians because of a low index of suspicion of vasculitic neurop-
cal distribution of motor and sensory deficits between two limbs, athy. In Hawke’s series (Hawke et al. 1991), patients with monon-
for example symmetrical sensory polyneuropathy with left pero- europathy multiplex had a shorter period (mean 9.2 weeks) before
neal neuropathy. Kissel and Mendell (1992) termed such a pattern diagnosis than did patients with symmetrical polyneuropathy (mean
‘overlapping mononeuropathy multiplex.’ In their series, this was 20.4 weeks) or asymmetrical polyneuropathy (31.6 weeks). Most
the most common clinical presentation in vasculitic neuropathy, patients with vasculitic symmetrical polyneuropathy have mixed
but in a compiled series it was seen in 23% of cases (Table 13.6). sensorimotor findings. However, pure sensory symmetrical poly-
Many clinicians, usually non-neurologists, have labelled this pat- neuropathy has been reported in a few cases (see Section Sensory
tern ‘mononeuropathy multiplex,’ using loose criteria for mon- Neuropathy). A pure motor symmetrical polyneuropathy classically
oneuropathy multiplex. Thus, confusion has resulted as to the seen in acute or chronic inflammatory demyelinating polyneuropa-
frequency of mononeuropathy multiplex in vasculitic neuropathy. thy has not been reported in vasculitic neuropathy.
Asymmetric polyneuropathy should not be confused with true Sensory neuropathy
mononeuropathy multiplex, which has a distinct list of differential It has been claimed that in rheumatoid arthritis, sensory polyneu-
diagnoses (Table 13.7). Asymmetrical polyneuropathy results from ropathy is not due to vasculitis (Conn and Dyck 1984). However,
simultaneous patchy discrete infarctions of smaller vessels in many Wees et al. (1981) reported one RA patient with symmetrical sen-
individual peripheral nerves, together with superimposed monon- sory polyneuropathy who had a definite vasculitis on the sural nerve
europathies due to damage of whole nerve trunks. biopsy; Moore and Fauci (1981) reported one patient with sensory
Symmetrical polyneuropathy polyneuropathy and two patients with cutaneous sensory neuropa-
Symmetrical polyneuropathy is characterized by an ascending and thy. Subsequently, Dyck et al. (1987) described one case of sensory
distal, symmetrical, ‘stocking-glove’ type sensory loss with flaccid neuropathy with non-systemic vasculitic neuropathy (NSVN) and
distal weakness. The classical examples of symmetrical polyneurop- Lacomis et  al. (1997) reported one case of sensory neuropathy
athy are metabolic and alcoholic neuropathies. Thus, vasculitic neu- and vasculitis. In a recent analysis of a series of SNV, there were
ropathy has not usually been considered in the differential diagnosis ten (23%) examples of sensory polyneuropathy among 44 vascu-
of this clinical finding. Since the 1980s, however, several studies litic neuropathy cases. Puechal et al. (1995) reported 15 patients
have showed that vasculitis can cause symmetrical polyneuropathy with sensory neuropathy among 32 with vasculitic neuropathy
(Table 13.6). In fact, a symmetrical polyneuropathy pattern is seen and rheumatoid arthritis. Suppliah et al. (2011) reported sensory
in one-third of patients with vasculitic neuropathy. If this occurs as neuropathy in five of 269 patients with granulomatosis with poly-
the end result of extensive mononeuropathies multiplex, it is easier angiitis, but not in microscopic polyangiitis in ANCA-associated
to understand the pathogenesis of symmetrical polyneuropathy. vasculitis. These findings indicate that a sensory polyneuropathy
However, such a history is lacking in most patients. Thus, it is likely does not rule out vasculitic neuropathy, as was previously claimed.
that symmetrical polyneuropathy is a consequence of a more diffuse Collins et al. (2000) reported pure or predominant sensory neu-
peripheral nerve ischaemia due to multiple small lesions that are ropathy in 10% of 36 cases with histologically proven vasculitis by
more likely to affect longer nerves. A symmetrical polyneuropathy the superficial fibular (peroneal) nerve and peroneal brevis muscle
biopsy. Collins et  al. (2003) also reported ‘predominantly purely
sensory findings’ in 13% of 48 patients with non-systemic vascu-
Table 13.7 Various diseases with mononeuropathy multiplex litic neuropathy. We reported 17 (16%) cases of sensory vasculitic
neuropathy among 106 cases with histologically proven vasculitic
Categories Diseases neuropathy (Seo et  al. 2004). In 65% of cases, sensory vasculitic
neuropathy was associated with systemic vasculitis. The most com-
Ischaemic Vasculitic neuropathy mon clinical presentation was symmetrical polyneuropathy, seen
Diabetic neuropathy in 53% of cases. The most common nerve conduction pattern was
Amyloid neuropathy diffuse neuropathy of axonal degeneration. Sural nerve biopsy was
diagnostic in 88% of cases. In two cases, muscle biopsy was neces-
Inflammatory Multifocal motor sensory demyelinating neuropathy sary for the definite diagnosis of vasculitis. Non-systemic sensory
Multifocal motor neuropathy vasculitic neuropathy is usually benign. Of 11 patients followed for
Sarcoidosis longer than 2 years, none developed motor weakness due to neu-
ropathy. Kararizou et al. (2005) reported eight cases of sensory neu-
Brachial plexus neuropathy ropathy among 22 patients with NSVN. These studies indicate that
Lumbosacral plexus neuropathy sensory vasculitic neuropathy does exist and sensory neuropathy
Lymphomatoid granulomatosis can be due to vasculitic neuropathy.
Infectious Leprosy Asymptomatic vasculitic neuropathy
Subclinical involvement detected only by nerve conduction studies,
Lyme disease
called asymptomatic vasculitic neuropathy, was described in 1981
HIV by Wees et al. Among 17 patients with SNV, there were seven (41%)
Hereditary Hereditary liability to pressure palsy who did not show any clinical signs of peripheral neuropathy: two
had myopathy, three had generalized weakness, and two had com-
Malignant disease Lymphomatous neuropathy
plaints of weakness. Systemic symptoms and laboratory findings
CHAPTER 13 vasculitic neuropathy 167

were suggestive of PAN, which led to the nerve conduction study. et al. 1991; Kissel et al. 1985; Wees et al. 1981). NCS were found
In all these patients, a diffuse neuropathy was detected by the nerve to be abnormal in all patients with evidence of vasculitic neuropa-
conduction study and vasculitis was confirmed by the sural nerve thy on physical examination. The most common nerve conduction
biopsy. Subsequent studies confirmed the existence of asympto- abnormality was abnormal sensory nerve conduction (79–100% of
matic vasculitic neuropathy. Among 22 patients with vasculitic patients), with absent CNAP being most frequently observed. Sural
neuropathy, Bouche et al. (1986) found three (14%) with myalgia, nerve conduction abnormality was common, seen in 80–100% of
but no definite clinical sign of peripheral nerve involvement, who cases. Sensory nerve conduction in median and ulnar nerves was
had a neuropathic pattern on the electrophysiological study. Cruz abnormal in 70% of cases. Mixed nerve conduction was abnormal
Martinez et al. (1988) reported similar findings in their series of in the median nerves in 30% and in the ulnar nerves in 40% of
15 patients with biopsy-proven vasculitis; two patients had elec- the cases. Motor nerve conduction was less affected than sensory
trophysiological but not clinical evidence of neuropathy. Said et al. nerve conduction. NCV was slow in median and ulnar nerves in
(1988b) reported six patients with no clinical neuropathy but with 8%, in peroneal nerves in 83%, and in posterior tibial nerves in
spontaneous pain in muscles, among 100 biopsy-proven cases of 53% of cases. Motor NCV was either normal or minimally slow,
vasculitic neuropathy. Among 81 suspected cases of SNV, Claussen suggesting axonal neuropathy. As expected with axonal degenera-
et al. (2000) found seven (9%) who had systemic features without tion, the compound muscle action potential (CMAP) amplitude
neuropathy; in all five patients who underwent NCS, there was evi- was typically reduced. Bouche et al. (1986) found reduced CMAP
dence of neuropathy. Four (57%) of these seven patients were found amplitude in all his cases. Compared with the upper extremities,
to have vasculitis on nerve biopsy. These studies suggest that it is the lower extremities were more involved, with the most com-
useful to evaluate patients suspected of SNV with an electrophysi- monly affected nerves being the peroneal and sural. In the upper
ological study, even if there is no clinical evidence of neuropathy. extremities, the ulnar nerves were much more commonly involved
The electrophysiological tests should include sensory nerve con- than the median nerves. This suggests that the distribution of
duction studies and nerve conduction studies in the lower extremi- nerve infarction in mononeuropathy multiplex is not random.
ties, since these tests are extremely sensitive. If neuropathy is found, The needle EMG classically shows a denervation process in the
then biopsy of the involved nerve can follow immediately and fro- distal muscles in diffuse neuropathy. In cases of mononeuropathy
zen sections can confirm the diagnosis of vasculitic neuropathy. or mononeuropathy multiplex, this was confined to the involved
muscles. Fibrillations and positive sharp waves (PSW) were most
Electrophysiological findings commonly observed, as expected in axonal degeneration. Motor
Nerve conduction studies play three important roles in the diag- unit potentials (MUP) were relatively normal with little evidence
nosis of peripheral nerve vasculitis. The first is to detect periph- of the high-amplitude long-duration MUPs that characterize
eral neuropathy among patients suspected of systemic necrotizing re-innervation. Most patients had a strikingly reduced motor unit
vasculitis but who do not have neurological symptoms. Adequate recruitment pattern in clinically affected muscles, with sparse
nerve conduction tests were able to detect asymptomatic periph- numbers of MUPs firing in isolation at a rapid rate on maximum
eral neuropathy among such patients in 6–41% of cases, as dis- effort. Conduction block, an electrophysiological hallmark of
cussed in Section Asymptomatic Vasculitic Neuropathy. The segmental demyelination, has rarely been reported in vasculitic
second important role relates to the selection of a nerve for biopsy. neuropathy. Among 65 cases, Dyck et  al. (1987) reported mild
In one series, the sural nerve was most commonly involved and focal conduction block in two. We found conduction block in one
thus proved ideal for biopsy. When the sural nerve conduction nerve in nine of 47 cases. In none of these cases was a multifocal
is normal, electrophysiological testing of the superficial fibular conduction block observed, nor was any other electrophysiologi-
(peroneal) and superficial radial sensory nerves is recommended. cal manifestation of demyelination found. Hawke et al. observed
If the testing is abnormal, these nerves may be biopsied. Studies conduction block in three patients, two of whom had mononeu-
comparing the nerve conduction abnormalities and the diagnostic ropathy multiplex. Ropert and Metral (1990) reported five cases
yield of superficial fibular (peroneal) nerve and superficial radial of conduction block in necrotizing vasculitis; however, their data
nerve are not available at this time. The third important role is were not conclusive in view of the extremely low distal CMAP in
detection of asymptomatic nerve involvement not obvious by the four cases. Jamieson et al. (1991) reported a patient with necrotiz-
clinical evaluation among individuals with neurological or mus- ing vasculitis who showed a multifocal conduction block in the
cular disease. Wees et al. (1981) identified diffuse neuropathy by first study but classic axonal neuropathy in a later study. These
NCS in two patients with mononeuropathy, in one patient with findings suggest that conduction block may be present in a few
mononeuropathy multiplex, and in two cases of myopathy. In four nerves transiently during the acute denervating stage of vascu-
of five patients with mononeuropathy multiplex, Cruz Martinez litic neuropathy. Widespread asymmetrical conduction block in
found more diffuse signs of neuropathy than suggested by clinical the presence of relatively well-preserved distal CMAP is strongly
findings. Bouche reported a similar observation in patients with suggestive of an inflammatory demyelinating neuropathy. The
mononeuropathy multiplex. In our experience, the most com- needle EMG can also detect myopathy, either symptomatic or
mon nerve conduction abnormality in patients with vasculitic asymptomatic, in some patients with vasculitic neuropathy. This
neuropathy is diffuse neuropathy, rather than mononeuropathy is because myositis or myopathy is not uncommon in many col-
multiplex. For this reason, the lack of clear-cut mononeuropathy lagen vascular diseases. Wees et al. (1981) found a myopathic pat-
multiplex on nerve conduction studies cannot be used to exclude tern (many small-amplitude and short-duration MUPs with early
a vasculitic neuropathy. Nerve conduction abnormalities in vas- recruitment) in two patients with clinical evidence of myopathy,
culitic neuropathies are typical of axonal degeneration (Bouche in two patients with generalized weakness, and in one patient with
et  al. 1986; Cruz Martinez et  al. 1988; Dyck et  al. 1987; Hawke mononeuropathy. Bouche et al. (1986) found a myopathic pattern
168 SECTION 3 clinical manifestations common to vasculitis

in one case of mononeuropathy multiplex, in six cases of diffuse 20–40% of MPA, 20–40% of pauci-immune crescentic glomerulo-
neuropathy, and in one patient without clinical signs of neurop- nephritis, and 35% of EGPA (Radice and Sinico 2006). Cryoglobulin
athy. Needle EMG findings may be helpful in selecting the spe- assays may be useful, as vasculitic neuropathy is common in all
cific muscle for biopsy. In Claussen et al. (2000), abnormal needle forms of cryoglobulinaemia (Nemni et al. 1988). This is especially
EMG findings in the biopsied muscle did not allow discrimina- true in patients with hepatitis C virus-associated cryoglobulinaemia
tion between biopsy-positive and biopsy-negative vasculitis cases. (Khella et al. 1995). In HIV patients, vasculitic neuropathy is rare,
There was, however, a trend toward correlation between the pres- usually occurring before the development of AIDS (Calabrese et al.
ence of fibrillation and positive sharp waves and vasculitis in the 1989; Fuller et al. 1993). A few patients with Lyme disease have also
muscle biopsy, suggesting that selection of a muscle on this basis been reported to have mononeuropathy multiplex with a vasculi-
may result in a higher diagnostic yield of vasculitis. tis demonstrated pathologically (Meier et al. 1989). Thus, if clinical
findings are compatible, HIV and Lyme titres should be ordered.
Laboratory findings CSF is usually normal in vasculitic neuropathy except for mild
There is no specific laboratory marker for vasculitic neuropathy elevation of protein in one-third of patients. On the other hand,
(Table 13.8). Thus, the laboratory evaluation is directed toward iden- abundant cells and high protein are the classical CSF pattern in
tifying the underlying causes of vasculitis or a serological abnor- HIV vasculitic neuropathy (Said 1995).
mality that may point toward a specific vasculitic syndrome. Two Sakai et al. (2005) reported that mean plasma VEGF level in five
important laboratory tests in SNV are the erythrocyte sedimenta- patients with vasculitic neuropathy (one patient with NSVN) was
tion rate (ESR) and antineutrophil cytoplasmic (ANCA) antibody significantly higher than in the healthy controls, GBS, CIDP, and
tests. An elevated ESR is its most consistent and common abnormal- amyotrophic lateral sclerosis (ALS) patients. Plasma VEGF level
ity. It is almost invariably elevated in PAN, GPA, and giant cell arte- was increased in four of five patients. In two patients, a marked
ritis and therefore is useful diagnostically in these disorders (Haynes decrease in plasma VEGF and in one patient, a mild decrease was
et al. 1986). ANCA test is positive in 37 (47%) of 78 patients with noted after treatment. In view of small number, further studies with
SNV (Mathew et al. 2007). cANCA is positive in 80–90% of GPA, a larger study population are necessary.

Table 13.8 Laboratory findings in vasculitic neuropathy

Systemic necrotizing vasculitis Non-systemic necrotizing vasculitis

1981 1985 1991 1986 2000 1987 1996 2003

Wees et al. Kissel et al. Hawke et al. Bouch et al. Collins et al. Dyck et al. Davies et al. Collins et al.
(N = 17) (N = 16) (N = 14) (N = 22) (N = 25) (N = 20) (N = 25) (N = 48)
High sedimentation rate 14 (82%) 14 (87%) 6 (43%) 19 (86%) 17/22 (77%) 8 (40%) 10 (40%) 22 (25%)
Leucocytosis 6 14 14/23 (61%) 0 11 (23%)
Leucopenia 3
Anaemia 9 14/22 (64%) 15 (31%)
RF (+) 9 4 7 2/11a 10/18 (55%) 1/6 2/18 10 (20%)
Low complement 5/10 4/19 (21%) 5 (11%)
Eosinophilia 3 8
Increased GG 3 4/13 13/18
High CPK 3
Hepatitis B antigen 1/5 3/9 6/18
ANA titre (+) 4 3 8 7/20 (35%) 3/11 4/20 19 (39%)
High serum creatinine 1 3
Proteinuria 2
Cryoglobulin 1 0/7
CSF
Protein 1/7 (14%) 4/9 (44%) 8/17 (47%) 5/19 (26%)
Cellb 0 0 1/19
Oligoclonal band 1 1
a 2/11, positive in 2 cases out of 11 tested cases.
b 3 HIV patients had abundant cell and protein in CSF (Said).

Kararizou (2005): Elevated SR (>30 mm/h) in 4/22 (18%) cases and positive ANA in 2/22 (11%) cases.
CHAPTER 13 vasculitic neuropathy 169

Diagnosis frequent healing of vasculitic lesions including leg ulcers and neu-
ropathy, a lower incidence of relapse, fewer serious complications,
In non-systemic vasculitic neuropathy, a definite diagnosis of
and a lower mortality rate with intermittent cyclophosphamide
vasculitic neuropathy is based on the nerve biopsy (Table  13.9)
plus methylprednisolone than with other treatments. The precise
because, by definition, no other organ is involved. There is, how-
regimen for an individual patient depends on the extent, severity,
ever, a huge difference between two series with regard to the ratio
and tempo of the vasculitic neuropathy and other organ involve-
of definite vasculitis to probable vasculitis in the nerve biopsy. In
ment. Usually, a combination of prednisone 60 mg a day and oral
the series of Dyck et al. (1987), definite vasculitis was found only in
cyclophosphamide 150 mg a day is recommended as the initial
25% of cases, whereas in Davies’s series (1996), it was identified in
regimen. Some prefer intravenous methylprednisone therapy fol-
76% of cases. This difference between the two figures is not easily
lowed by oral prednisone therapy. For cyclophosphamide therapy,
explained. The histological diagnosis of vasculitic neuropathy was
some advocate a pulse intravenous therapy. In principle, the com-
confirmed by the nerve biopsy in 82–100% of patients with systemic
bined therapy is continued until significant improvement occurs
necrotizing vasculitis. The next most productive tissue was muscle
and the clinical condition stabilizes. This may take 6–12 months. At
biopsy. In rare cases, the tissue diagnosis was made by the liver or
this point, prednisone is slowly tapered and a switch of cyclophos-
kidney biopsy or at autopsy. In a few cases, celiac angiography was
phamide to azathioprine 2 mg/kg per day is recommended follow-
used to diagnose vasculitic neuropathy (Table 13.9). Though Said
ing the current practice in ANCA-associated vasculitis (Jayne et al.
(1995) stated that the tissue diagnosis of vasculitis was easily made
2003). Azathioprine is then slowly tapered. Some prefer methotrex-
by specific skin biopsy, the consensus is that this is less diagnostic
ate over azathioprine. Considering an experience with cyclophos-
and not necessarily indicative of systemic involvement (Lie 1990).
phamide (Langford and Sneller 1997; Rasmussen et al. 1996), it is
Treatment reasonable to continue azathioprine treatment up to 1 year after all
signs of the disease have subsided. This optimistic goal may not eas-
There have not been any prospective randomized controlled trials
ily be achievable in vasculitic neuropathy, in which many persistent
of therapy specific for vasculitic neuropathy. However, two rand-
symptoms and signs may be present. Thus, the recommendation is
omized controlled trials of various combinations of therapies for
to continue the maintenance dose of azathioprine for 1 year after
ANCA-associated vasculitis have been published (Stone et al. 2010;
vasculitic neuropathy becomes inactive or maximum improve-
Jones 2010). Thus, present therapies are based on collective experi-
ment has been achieved. If relapse occurs, either during tapering
ence in systemic and ANCA-associated vasculitis in general.
or maintenance of prednisone or azathioprine, the drugs must be
In systemic necrotizing vasculitis, the standard treatment pro-
increased to the previous dose. According to Hawke et al. (1991),
tocol over the past 40  years was cyclophosphamide and gluco-
vasculitic neuropathy is generally treated for more than 2 years.
corticoids. In patients with neurological involvement of vasculitis
Rituximab, a B-cell-depleting anti-CD20 monoclonal antibody,
(10 CNS and 15 peripheral neuropathy), Moore and Fauci (1981)
emerged as an alternative for cyclophosphamide in treatment
reported that virtually no extension or progression of disease
of vasculitis to reduce serious adverse reactions associated with
activity was seen in any patient receiving an adequate course of
cyclophosphamide treatment. Two randomized trials comparing
cyclophosphamide for a reasonable duration of time. In systemic
rituximab versus cyclophosphamide in ANCA-associated vas-
rheumatoid vasculitis, Scott and Bacon (1984) reported more
culitis have been published (Stone et al. 2010; Jones et al. 2010).

Table 13.9 Diagnostic methods for vasculitic neuropathy

Authors Total cases Nerve biopsya Muscle biopsy Others


Wees and Oh 1981 17 15 (82%) 3 1 celiac angiographyb
Kissel et al. 1985 16 160
Dyck et al. 1985 45 39 (87%) 3 (7%) 1 6: liver, kidney, post-mortem
13 had probable vasculitis.
Said et al. 1988b 95 42 (44%) 53c 0
Hawke et al. 1991 34 33 (97%) 2 (6%) 3 based on probable vasculitisd
1 renal biopsy; 1 autopsy
Claussen et al. 2000 45 45 (100%) 3 (7%) 9 based on Type III
Collins et al. 2000 22e 19 (86%) 4 (18%)
Vital 2006 85 62 (73%) 42 (59%) Definite vasculitis cases alone
a This includes the definite and probable vasculitis by the pathology criteria except Kissel and Said series in which the definite vasculitis criteria was only used.
b Type III. In this case, celiac angiography.
c In 16, this is the only tissue biopsied.
d Probable vasculitic neuropathy: 3. Diagnosis is confirmed by muscle biopsy in 2 and 1 renal biopsy.
e 7 cases with NSVN were included. In 3 cases with SVN, diagnosis is confirmed by muscle biopsy:
nerve biopsy was negative in 2 and nerve was not found in 1. One additional case with NSVN had definite
vasculitis in muscle.
170 SECTION 3 clinical manifestations common to vasculitis

Stone’s study (Stone et  al. 2010)  in 197 ANCA-positive patients neuropathy was age; older age predicts a worse outcome (Hawke
with either GPA or MPA showed an induction of remission in et al. 1991). Chang et al. (1984) found that patients with no recur-
64% of rituximab group as compared with 53% in the control daily rence of systemic vasculitis within 18 months of initial appearance
cyclophosphamide group. Jones’s study (Jones et  al. 2010)  in 44 of mononeuropathy multiplex have a good prognosis. Puechal
patients with newly diagnosed ANCA-associated renal vasculitis et  al. (1995) showed that vasculitic neuropathy in rheumatoid
showed a sustained remission in 76% in the rituximab group as arthritis is an indicator of poor prognosis; the overall survival
compared with 82% in the control intravenous cyclophosphamide rate at 5 years was 57%. They also found that cutaneous vasculi-
group. Both studies concluded that rituximab therapy is equal to tis, extensive mononeuropathy multiplex (more than three limbs
cyclophosphamide for induction of remission. Stone’s study (Stone involved), and depressed C4 level are indicators of a poor prog-
et  al. 2010)  even suggested that rituximab might be superior in nosis. Four studies have addressed the issue of long-term survival
relapsing disease. From these studies, one can conclude that rituxi- in vasculitic neuropathy. The survival rate was found to be 37%
mab is an attractive alternative for cyclophosphamide in the treat- (Hawke et al. 1991), 57% (Puechal et al. 1995), and 60% at 5 years
ment of SVN. (Chang et al. 1984) and 71% at 6 years (Dyck et al. 1987). Death
The recovery of function in vasculitic neuropathy is slow and was usually attributed to vasculitis (Dyck et al. 1987) and usually
lags considerably behind control of the vasculitis because axonal occurred within 6 months after onset of illness (Hawke et al. 1991).
regeneration must occur. In some patients with systemic vascu- There is no good explanation for the markedly different survival
litides, serum markers for control of vasculitis are not necessar- rates between Hawke’s series and Dyck’s series, except that in the
ily beneficial as a therapeutic index for vasculitic neuropathy. former, most deaths were due to non-vasculitic causes, and in
The role of nerve conduction studies in this regard has not been Dyck’s series most were due to vasculitis.
assessed. Therefore, a careful neurological evaluation is the best With treatment, vasculitic neuropathy has been found to have a
measure available for the determination of disease status in vas- good prognosis in terms of functional recovery. Moore and Fauci
culitic neuropathy. In addition to immunosuppressive treatment, (1981) reported that even those patients with severe peripheral
aggressive physical and occupational therapy is usually indicated neuropathies improved greatly over time with treatment of the
during the recovery stage of the disease, to improve the patient’s underlying diseases, physical therapy, and rehabilitation. Dyck et al.
range of motion of joints, optimize functional status and main- (1987) reported that two-thirds of living patients were better than
tain strength. To manage neuropathic pain, the use of gabapentin, at their first evaluation. Complete recovery was reported in 11%
clonazepam, carbamazepine, or amitriptyline may be required. (Hawke et al. 1991) to 21% (Chang et al. 1984) over a 3- to 5-year
Plasma exchange is known to be indicated as an adjunct ther- follow-up period. On recovery, patients were able to ambulate with-
apy in ANCA-associated vasculitis patients with severe renal out assistance in 58% (Chang et al. 1984) to 84% of cases (Hawke
failure (Jayne et al. 2007). In an open trial of eight patients with et al. 1991). Relapse was reported in 25% of cases in Chang’s series
rheumatoid vasculitis, a course of plasma exchange given with and in 0% in Hawke’s series. Relapse was most likely due to prema-
conventional immunosuppression resulted in more rapid healing ture cessation of prednisone or cytotoxic medications or subopti-
of skin lesions than in patients treated with immunosuppressives mal doses with these drugs. Despite reasonable functional recovery,
alone (Winkelstein et al. 1984). The peripheral neuropathy in these many patients had persistent sensory symptoms, especially in the
patients, however, did not improve. lower limbs (Davies 1994).
There is a clear evidence of benefit from intravenous immuno- Outcome has been much better in more recent series (Mathew
globulin (IVIg) in the treatment of coronary artery abnormali- et al. 2007). Initial stabilization with standard steroid and immu-
ties related to Kawasaki’s disease and ANCA-associated vasculitis nosuppressive therapy was achieved in 86 (91%) of 94 patients
(Aries et  al. 2005). One randomized, placebo-controlled trial in with SVN and 1-year survival rate was 90.3%. The most remark-
ANCA-associated systemic vasculitis with persistent disease activ- able outcome was reported recently from the European Vasculitic
ity showed that a single course of IVIg reduced disease activity Study Group in four different immunotherapy combination trials
in persistent ANCA-associated vasculitis, but this effect was not (Suppiah et al. 2011) These identified neuropathy in 40 (8%) of 506
maintained beyond 3 months (Jayne et al. 2000). There were two vasculitis patients at baseline and all 40 achieved remission from
reports of a favourable outcome with IVIg in vasculitic neuropathy active neuropathy within 9 months, 14 (35%) without long-term
(Schifitto et al. 1997; Levy et al. (2003). In a patient with HIV-related damage and 26 (65%) with chronic neuropathy (Suppiah et  al.
vasculitic mononeuropathy multiplex, clinical improvement was 2011). Ten (25%) out of 49 died within 5 years compared with 80
achieved and maintained for 19 months with combined treatment (18%) out of 450 without neuropathy.
with high-dose glucocorticoids and monthly IVIg for 4  months
(Schfitto et al. 1997). Levy et al. (2003) also reported a resolution of
neuropathy in 4 of 6 patients with peripheral neuropathy with ‘vas- Non-systemic vasculitic neuropathy
culitis’ (three confirmed vasculitis by the nerve biopsy) with IVIg Vasculitis confined to peripheral nerves is termed non-systemic
treatment. vasculitic neuropathy (NSVN) (Dyck et  al. 1987). Though this
entity is described first in 1938 by Keroohan and Woltman in a
Prognosis 48-year-old man with necrotizing vasculitis restricted to the
Studies of prognosis in vasculitic neuropathy are limited. Several peripheral nervous system at autopsy, the concept of localized
studies of PAN did not find that peripheral neuropathy was asso- peripheral nerve system vasculitis was reintroduced in 1985
ciated with a poorer outcome (Cohen et  al. 1980; Fronert and when Kissel et al. reported seven cases of NSVN among 16 cases
Sheps 1967; Sack et  al. 1975). Among the various factors ana- of biopsy-proven vasculitic neuropathy. None of their cases had
lysed, the only significant prognostic factor identified in vasculitic any other organ involvement or abnormal serological tests. Dyck
CHAPTER 13 vasculitic neuropathy 171

et al. (1987) reported 20 patients with NSVN among 65 cases of in the epineurial arterioles, not very different from SVN. Both
necrotizing vasculitis. Davies et al. (1996) reported their experi- showed axonal degeneration as the predominant pathological find-
ence with 25 patients with NSVN. Said et al. (1988b) reported 32 ing and selective fascicular atrophy or central atrophy as ischae-
cases of peripheral neuropathy alone among 100 patients with mic changes. Immune complexes in the vessel walls were observed
necrotizing vasculitis. Collins et  al. (2003) reported 48 patients as commonly in NSVN as in SVN (Davies et al. 1996). High ESR
with NSVN at presentation. Unlike other groups, this group (>40  mm/h) was noted in 25–50% of cases (Collins et  al. 2003;
includes patients with constitutional symptoms such as weight Kissel et al. 1985). Hepatitis-B antigen, monoclonal gammopathy,
loss and unexplained fever. Weight loss was observed in 35% of and cryoglobulin were absent in NSVN (Dyck et al. 1987). Collins
cases and unexplained fever in 15%. This raises a question whether et al. (2003) reported monoclonal gammopathy in three (8%; 3/37
all of Collins’s cases represent pure NSVN cases. Kararizou et al. cases) cases of patients older than 50 years. According to the 2010
(2005) reported 22 cases of NSVN. It is worth noting that patients Peripheral Nerve Society guidelines, proposed exclusionary criteria
with NSVN, because of the absence of systemic symptoms or other for NSVN-favouring the alternate diagnosis of systemic SVN were
organ involvement, are likely to see a neurologist. This is in con- clinicopathological evidence of other-organ involvement, ANCAs,
trast to SVN patients who are likely to be seen by a primary care cryoglobulins, sedimentation rate at or above 100 mm/h, and med-
physician or rheumatologist. ical condition/ drug predisposing to systemic vasculitis (Collins
NSVN represents about 25% of cases of vasculitic neuropathy et al. 2010).
(Davies et al. 1996; Dyck et al. 1987; Said et al. 1988b). In NSVN, Nerve biopsy is necessary for diagnosis of NSVN. Without nerve
there are no signs of systemic or organ involvement, such as biopsy, vasculitis cannot be reliably differentiated from other rap-
weight loss, fever, anorexia, or kidney failure (Table 13.10). There idly progressive neuropathies because many cases of NSVN appear
is no difference in the types of neuropathy, but Dyck et al. (1987) symmetrical and serological markers are usually absent.
observed that there are some differences between NSVN and SVN In contrast to SNV, NSVN is relatively benign; the disease is indo-
at the microscopic level. In NSVN, smaller perineurial arterioles lent and protracted over years and appears not to be life-threatening
were more often involved, the severity of pathology was less, and (Dyck et al. 1987). Davies et al. (1996) observed that only one of
probable vasculitis (perivascular infiltration of cells without intra- their 25 patients died during the 3-year follow-up period. The
mural infiltration of cells or fibrinoid necrosis) was more common majority had a monophasic course, but 32% had at least one relapse
in NSVN. during the follow-up period. Said (1995) followed 29 patients over
Dyck et al. (1987) believed that an underlying indolent necrotiz- an average of 6 years; 37% of these developed systemic manifesta-
ing vasculitis of the small epineurial arterioles was responsible. tions, 37% died an average of 3.3 years after the onset of neuropathy
Davies et al. (1996) observed the vasculitic features predominantly (eight from systemic manifestations and three from infection), and
24% had one or more relapses of neuropathy. Among 48 cases of
NSVN with longer than 6 months’ follow-up period, Collins et al.
Table 13.10 Pathological, clinical, and laboratory differences between (2003) reported a relapse of neuropathy in 38% of cases, spread of
systemic necrotizing (SNV) and non-systemic necrotizing (NSNV) vasculitis to skin in 6%, and death in 21%. Only half of these deaths
vasculitic neuropathies were related to vasculitis as the cause. Five-year survival rate was
85–87% in Said’s and Collins’ series. Patients in Said’s and Collins’
SNV NSNV series had a worse prognosis than did those in Dyck’s series, per-
haps due to inclusion of some cases of SVN.
Pathological differencesa A Cochrane review concluded that there is no randomized or
Commonly involved Epineurial arteriole Perineurial smaller
quasirandomized control treatment trials in NSVN (Vrancken
arteriole
et al. 2011). With glucocorticoid treatment in all cases and addi-
Types of vasculitis More often definite More common probable
Severity More severe Less severe
tional immunosuppressive medications in half the cases, there
Axonal degeneration No difference No difference was significant improvement in the disability scale (Davies et al.
Ischaemic change No difference No difference 1996). In several of Dyck’s cases (Dyck et  al. 1987), prednisone
appeared to halt progression of the disease. On this basis, Chalk
Clinical featuresa Essentially the same Essentially the same
et al. (1993a) stated that, in a substantial number of patients with
Laboratory features NSVN, the relatively favourable outlook might make the risks of
High ESR Common Rare immunosuppressive therapy difficult to justify. Kissel and Mendell
High ANA Common Extremely rare (1992) also stated that treatment with glucocorticoids alone may
High RF Common Extremely rare be appropriate. On the other hand, Collins et al. (2010) compared
Hepatitis B antigen 20% None
the outcome results between 28 patients treated with corticosteroid
ANCA 47%* None
alone and 20 with combination therapy (corticosteroids and cyclo-
Prognosis Serious Benign phosphamide). They found the combination therapy was more
Diagnosis Can be diagnosed Nerve biopsy: sine effective than corticosteroid monotherapy in inducing remission
from Other tissue qua non (95% versus 61%) and improving disability (85% versus 57%), with
Duration of disease Weeks to months Months to years
trends towards a reduced relapse rate (29% versus 59%), chronic
pain (44% versus 71%), and 5-year mortality. Thus, there is one
Treatment Cyclophosphamide Steroid alone? study that supports combination therapy in NSVN. For the treat-
Prognosis Serious Benign ment of NSVN, Peripheral Nerve Society guidelines recommend
* Mathew 2007 the following: (1) corticosteroid monotherapy for at least 6 months
172 SECTION 3 clinical manifestations common to vasculitis

is considered first-line; (2) combination therapy should be used for Table 13.11 Differential diagnosis between paraneoplastic and
rapidly progressive NSVN and patients who progress on steroid systemic vasculitic neuropathy
monotherapy; (3) immunosuppressive options include cyclophos-
phamide, azathiopine, and methotrexate; (4)  cyclophosphamide Paraneoplastic Systemica
is indicated for severe neuropathies, generally administered in i.v.
Sex Male predominance Equal
pulses to reduce cumulative dose and side-effects; (5) in patients
achieving clinical remission with combination therapy, mainte- Age 51–78 years (mean: 60) 19–84 years (mean: 62)
nance therapy should be continued for 18–24 months with azathio- Associated disease SCLC and lymphoma: 50% Rheumatological
prine or methotrexate (Collins et al. 2010). disease: 35%
There is controversy about the nature of NSVN: an organ-specific
Onset Subacute Subacute
vasculitis (Davies et al. 1996; Dyck et al. 1987) versus a mild form of
systemic vasculitis (Kissel et al. 1989; Said et al. 1988b). Said et al. Clinical features Mixed neuropathy Mostly mixed
(1988b) and Collins et al. (2003) argued against an organ-specific Varied; symmetrical Varied; symmetrical
vasculitis because of the demonstration of necrotizing arteritis in
polyneuropathy to polyneuropathy
muscle specimens of 81% of his patients with peripheral neuropa-
thy alone and the development of systemic manifestations in 37% mononeuropathy to mononeuropathy
during the follow-up period. Their argument may be tainted by the multiplex
fact that not all his patients had NSVN, as discussed above in this Systemic symptoms Rare Common
section. Davies et al. (1996) supported an organ-specific vasculi- and signs
tis theory because the good prognosis of NSVN was not correlated CNS symptoms Rare Rare
with a less-severe neuropathy than that seen in systemic vasculi- and signs
tis. Many of their NSVN patients had a very severe neuropathy,
Laboratory High ESR High ESR
whereas many patients with severe SNV had a relatively mild neu-
ropathy or none at all. Anti-Hu antibody Rare None
CSF findings High protein with normal cells Usually normal
Paraneoplastic vasculitic EMG/NCV Axonal neuropathy Axonal neuropathy
neuropathy Biopsy Microvasculitis Necrotizing vasculitis
Paraneoplastic vasculitis (PV) is rare and usually presents as cuta- Nerve: muscle (70%)b Nerve/muscle (42%)a
neous leukocytoclastic vasculitis in association with leukaemia and
Immunotherapy Moderately effective
lymphomas (Sanche-Guerrero et al. 1990).
Paraneoplastic vasculitic neuropathy (PVN) is rarer. Eighteen Tumour therapy Some benefit
patients with PVN have been reported in the literature thus far a From Claussen et al. 2000.
(Oh 1997; Turner et al. 2003; Ansari et al. 2004; Choi et al. 2013 b 70% of muscle biopsies showed vasculitis.

Muley et al. 2008; Correia et al. 2011). One case had PVN as EGPA
paraneoplastic syndrome (Correia et al. 2011). At UAB, only two
without glucocorticoids alone were associated with improvement
(1.3%) among 151 patients with biopsy-proven vasculitic neurop-
in two-third of patients.
athy over the past 25 years had PVN. Diverse cancers have been
reported in association with PV, the most common being small cell
lung cancer (SCLC) and lymphoma, which accounted for 54% of Vasculitic neuropathy in HIV
cases (Table  13.11). Malignancy was found 1–18  months before Vasculitic neuropathy is rare in HIV, occurring in 0.1–3% of
neuropathy in 42% of cases and 2–29 months after the diagnosis patients with AIDS-related complex or AIDS (see Chapter  37).
of neuropathy in another 42% of cases. PVN is generally a disease Prior to 1997, 27 cases were reported (Brannagan 1997). It was at
of the older population. The neuropathy is usually subacute and times the first manifestation of HIV and painful (Bradley 1996).
progressive, ranging from a few days to 8  months. Symmetrical The predominant manifestation of HIV-vasculitic neuropathy
polyneuropathy is the most common pattern of neuropathy; asym- was distal and symmetric polyneuropathy (eight of 18 cases) and
metrical polyneuropathy and mononeuropathy multiplex are less asymmetrical polyneuropathy (six cases) with weight loss, myal-
common. PVN is invariably characterized by mixed motor and gia, weakness, and leg tenderness (Gehardi 1993; Said et al. 1988b);
sensory neuropathy. Systemic symptoms are lacking. The most mononeuropathy multiplex was least common. In most patients,
prominent abnormal laboratory findings were high ESR (80% of vasculitic neuropathy was not associated with other organ involve-
cases) and a high CSF protein content (91%). Anti-Hu antibodies ment and was usually monophasic without relapse or remission.
were noted in three patients, prompting a search for occult SCLC Pathological studies showed inflammation and fibrinoid necrosis of
in both, which was found in one. pANCA was positive in two of arteries smaller than those typically affected in SNV. Endoneurial
three recent cases (Choi et  al. 2012; Correia et  al. 2011; Muley inflammatory cells were prominent. Active necrotizing lesions on
et al. 2008). NCS were typical of axonal degeneration. The diagno- biopsy did not coexist with healed lesions. Necrotizing vasculitis
sis of vasculitis was made in all cases by inspection of a peripheral may occur with CD4 counts ranging from 14 to 540. The ESR is
nerve biopsy that showed microvasculitis (nine cases), or necrotiz- usually elevated. CSF is usually characterized by increased protein
ing vasculitis (seven cases). Vasculitis was also common (70%) content and pleocytosis. Cryoglobulinaemia, which may be a cause
on muscle biopsy. Chemotherapy and cyclophosphamide with or of vasculitis, has also been described in several patients with HIV
CHAPTER 13 vasculitic neuropathy 173

infection and mononeuropathy multiplex. Clinical improvement neurogenic atrophy was observed in all cases and inflammatory
followed glucocorticoid therapy in nine of 11 cases (Gisselbrecht myopathy in one.
et  al. 1997). Plasmapheresis followed by zidovudine (AZT) was Thus, these studies showed that inflammatory vasculopathy is
effective in three patients (Brannagan 1997). There are also some observed in 20–100% of nerve biopsies in diabetic neuropathy,
data to suggest that AZT may be prophylactic for HIV-vasculitic especially in patients with proximal diabetic neuropathy (diabetic
neuropathy (Brannagan 1997). amyotrophy), and that microvasculitis seems to be a common fea-
ture. These studies also suggested that prednisone or IVIg is effec-
Vasculitic neuropathy with tive therapy in these patients. In summary, the present consensus is
that inflammatory vasculopathy, possibly immune mediated, plays
diabetes mellitus an important role in the pathogenesis of diabetic amyotrophy.
In recent years, there has been a flurry of reports of inflammatory
vasculopathy in diabetes mellitus. In 1984, Bradley et al. reported Vasculitic neuropathy with hepatitis
six patients, three of whom were diabetic, with painful lumbosacral
plexopathy, elevation of ESR, and epineurial lymphocyte infiltra- C virus infection
tion. Prednisone, alone or in conjunction with cyclophosphamide, Hepatitis C virus infection is associated with cryoglobulinaemia and
led to eventual improvement in the three diabetic patients. None associated vasculitis. Apartis et al. (1996) studied 15 patients with
had systemic vasculitis or cancer. The nerve in the lumbar plexus mixed cryoglobulinaemia and peripheral neuropathy, 10 of whom
in two autopsy cases showed perivascular epineurial inflammation had distal symmetric polyneuropathy and five of whom had mon-
with infiltration of adjacent endoneurium (Younger 2011; Raff oneuropathy multiplex. Of these, seven with distal symmetric poly-
et al. 1968). Younger et al. (1986) found microvasculitis (inflam- neuropathy and three with mononeuropathy multiplex had positive
matory infiltration of the walls of blood vessels measuring 70 µm hepatitis C serology. Necrotizing vasculitis was found in two of nine
or less) with scattered cells in the endoneurium in the sural nerve nerve biopsies from the HCV-positive patients, and interferon-α
biopsy in 12 (60%) of 20 patients (four with distal peripheral neu- apparently improved peripheral neuropathy in two. Khella et  al.
ropathy, six with proximal diabetic neuropathy and two patients (1995) described a patient with mononeuropathy multiplex, hepati-
with mononeuropathy multiplex). Perivascular lymphocytic infil- tis C virus infection, and mixed cryoglobulinaemia who had axonal
tration alone was seen in eight (40%) patients (two with distal and degeneration by EMG and vasculitis of the epineurial vessels on
six with proximal diabetic neuropathy). In addition, three patients sural nerve biopsy. High-dose prednisone and one course of plas-
had focal ischaemia. Axonal degeneration was seen in 10 tested mapheresis was tried with minimal symptomatic improvement.
nerves. In eight patients (seven with microvasculitis and one with Interferon-α was started at a dose of 3 million units three times per
severe perivasculitis), clinical improvement was obtained with week. Nine months after treatment with interferon-α, the patient
IVIg. Said et al. (1994) reported endoneurial inflammatory cells was free from neuropathy. David et al. (1996) described a person
in four patients and vasculitis in the epi- or perineurial vessels with non-systemic vasculitic mononeuropathy multiplex, cryoglob-
and ischaemic changes in two of 10 patients with painful diabetic ulinaemia, and hepatitis C virus infection, which was stabilized with
proximal neuropathy who had biopsy of the intermediate femoral prednisone/ cyclophosphamide/ interferon-α treatment. Initially,
cutaneous nerve. The two patients with vasculitis improved with immunosuppressive therapy was selected over interferon-α alone;
prednisone therapy. In 10 patients with proximal diabetic neu- interferon-α was added 2–3  months later, in part as an adjunct
ropathy, Krendal et al. (1995) reported perivascular lymphocytic therapy for the neuropathy. It was during these few months that the
infiltrates in the epineurial small vessels, in either the femoral patient’s condition stabilized and subsequently improved. An antivi-
cutaneous nerve (two) or the sural nerve (three). Neurological ral regimen, ideally a combination of interferon plus ribavirin, is the
improvement with immunosuppressive therapy (often includ- first-line treatment for HCV-induced cryoglobulinaemic vasculitis
ing IVIg) was noted. Llewelyn et  al. (1998) reported that, in 15 when the severity of its manifestations is mild-to-moderate. In case
cases of diabetic amyotrophy, biopsy of the intermediate cutane- of severe and life-threatening manifestation, immunotherapy (alone
ous nerve of the thigh showed epineurial microvasculitis in three or in addition to antiviral agents) is recommended (Chiche et al.
and non-vasculitic epineurial inflammatory infiltrate in another 2012). Among the immunotherapies, rituximab emerged as more
case. In a further case, microvasculitis was found in both the sural efficient than traditional immunosuppressive treatments (De vita
nerve and a quadriceps muscle biopsy specimen. Dyck et al.(1999) et al. 2012; Sneller et al. 2012).
found that in distal nerve biopsy in 33 patients with diabetic lum-
bosacral radiculoplexopathy (diabetic amyotrophy), epineurial Vasculitic neuropathy associated with
perivascular inflammation in all cases, microvasculitis (transmu-
ral infiltration of inflammatory cells without fibrinoid necrosis) other diseases
in 13, necrotizing vasculitis in two, and previous bleeding in 19 Malignant atrophic papulosis (Kohlmeier–Degos arteritis) is a rare,
(58%) of cases. They concluded that the primary event in diabetic often fatal, systemic vasculitis, primarily involving the skin, gastro-
amyotrophy is a microscopic vasculitis and weight loss may be a intestinal tract, and brain. Asbury and Johnson (1978) reported a
constitutional symptom of necrotizing vasculitis. In 15 patients person in whom skin, brain, and peripheral nerves were the pri-
with diabetic amyotrophy, Kelkar et al. (2000) found healed vas- mary targets, and sural nerve biopsy showed perineurial small
culitis in one, microvasculitis in the epineurial space in four, and arteries with subintimal proliferation, vessel wall thickening, lumi-
perivascular inflammatory cells in six of 13 intermediate femoral nal narrowing, and minimal perivascular inflammation.
cutaneous nerve biopsies and two sural nerve biopsies. In 15 mus- Schoene et al. (1981) reported a patient with Creutzfeldt–Jakob
cle biopsies (14 rectus femoris and one gastrocnemius muscle), disease and typical spongiform encephalopathy, but with unusual
174 SECTION 3 clinical manifestations common to vasculitis

clinical manifestations. The patient had a purpuric, non-pruritic Bloch, D.A., Michael, B.A., Hunder, G.G., et al. (1990). The American College
papular rash and polyneuropathy; sural nerve biopsy showed suba- of Rheumatology 1990 criteria for the classification of vasculitis. Patients
cute vasculitis. and methods. Arthritis and Rheumatism, 33, 1068–73.
Bouche, P., Leger, J.M., Travers, M.A., Cathala, H.P., and Castaigne, P. (1986).
Oh reported sarcoid polyneuropathy showing granulomatous
Peripheral neuropathy in systemic vasculitis: clinical and electrophysi-
vasculitis in epineurial small arteries, which produced almost ologic study of 22 cases. Neurology, 36, 1598–602.
complete occlusion of the lumen. Perivascular infiltrate of lym- Bradley, W.G., Chad, D., Verghese, J.P., et  al. (1984). Painful lumbosacral
phocytes and perineurial granuloma were also found (Oh 1979). plexopathy with elevated erythrocyte sedimentation rate:  A  treatable
Granulomatous vasculitis in sarcoid neuropathy was reported in inflammatory syndrome. Annals of Neurology, 15,457–64.
three additional cases (Vital et al. 1982; Gelassi et al. 1984; Iwata Bradley, W.G. and Verma, A. (1996). Painful vasculitic neuropathy in HIV-1
et al. 1993). Said et al. (2002) observed necrotizing vasculitis in the infection. Neurology, 47, 1446–1451.
nerve biopsy in seven of 11 cases of sarcoid neuropathy and sug- Brannagan, T.H. (1997). Retroviral-associated vasculitis of the nervous sys-
gested the possibility of ischaemic nerve lesion as the pathogenic tem. Neurologic Clinics, 15, 927–44.
mechanism of sarcoid neuropathy Calabrese, L.H., Estes, M., Yen-Lieberman, B., et  al. (1989). Systemic vas-
culitis in association with human immunodeficiency virus infection.
Human T-lymphotrophic virus I (HTLV-l) can cause T-cell leu-
Arthritis and Rheumatism, 32, 569–76.
kaemia, myelopathy, and polymyositis. The pathology of HTLV-I Camponovo, F. and Meier, C. (1986). Neuropathy of vasculitic origin in a
myelopathy is characterized by perivascular inflammation, but case of Garin-Boujadour-Bannwarth syndrome with positive borrelia
vasculitis is not seen. Similar perivascular inflammation is seen in antibody response. Journal of Neurology, 233, 69–72.
some patients with peripheral neuropathy (Said et al. 1988a). A sin- Chalk, C.H., Dyck, P.J., and Conn, D.L. (1993a). Vasculitic neuropathy. In
gle paper described three patients with HTLV-I and biopsies show- Diseases of peripheral nervous system, pp. 1424–36. W.B. Saunders,
ing necrotizing vasculitis (Vernat et al. 1994). Neuropathy occurs Philadelphia.
in 36–40% of patients with symptomatic late Lyme disease. Sural Chalk, C.H., Hamburger, H.A., and Dyck, P.J. (1993b). Antineutrophil cyto-
nerve biopsy in this disorder is characterized by perivascular col- plasmic antibodies in vasculitis peripheral neuropathy. Neurology, 43,
1826–7.
lections of inflammatory cells and axonal degeneration in a major-
Chang, R.W., Bell, C.L., and Hallett, M. (1984). Clinical characteristics
ity of cases (Halpern et al. 1987; Vallat et al. 1987). A few patients and prognosis of vasculitic mononeurpathy multiplex. Archives of
with Lyme disease had mononeuritis multiplex with vasculitis Neurology, 41, 618–21.
demonstrated pathologically (Camponovo and Meier 1986; Meier Chia, L.A.F., Lacroix, C., Adams, D., Plante, V., and Said, G. (1996).
et al. 1989). Characteristic nerve biopsy findings include perivascu- Contribution of nerve biopsy findings to the diagnosis of disabling
lar infiltration of inflammatory cells in the epineurial arterioles and neuropathy in the elderly. A  retrospective review of 100 consecutive
veins and axonal degeneration without necrotizing lesions of the patients. Brain, 119, 1091–8.
vessel walls (Meier et al. 1989). Active leprous neuropathy, once the Chiche, L., Bataille, S., Kaplantski, G., and Jourde, N. (2012). The place of
most common cause of neuropathy in the world, is invariably asso- immunotherapy in the management of HCV-induced vasculitis:  an
update. Clinical and Developmental Immunology, 2012, 315167.
ciated with non-necrotizing lymphocytic vasculitis (Said 1994). In
Choi, H.S., Kim, D.H., Yang, S.N., Sung, H.J., and Choi, S.J. (2013). A case
patients with multibacillary leprosy, vasculitis is associated with of paraneoplastic vasculitic neuropathy associated with gastric cancer.
infection of the endothelial cells by Mycobacterium leprae. Clinical Neurology and Neurosurgery, 115, 218–21.
Claussen, G.C., Thomas, T.D., Goyne, C., Vazquez, L.G., and Oh, S.J. (2000).
References Diagnostic value of nerve and muscle biopsy in suspected vasculitis
Ansari, J., Nagabhushan, N., Syed, R., Bomanji, J., Bacon, C.M., and Lee, S.M. cases. Journal of Clinical Neuromuscular Disease, 1, 117–23.
(2004). Small cell lung cancer associated with anti-Hu paranoeplastic Cohen, R.D., Conn, D.L., and Ilstrup, D.M. (1980). Clinical features, progno-
sensory neuropathy and peripehral nerve microvasculitis: case report sis, and response to treatment in polyarteritis. Mayo Clinic Proceedings,
and literature review. Clinical Oncology, 16, 71–6. 55, 146–55.
Apartis, E., Leger, M., Musset, L., et al. (1996). Peripheral neuropathy asso- Collins, M.P., Dyck, P.J., Gronseth, G.S., et al. (2010). Peripheral nerve society
ciated with essential mixed cryoglobulinaemia:  a role for hepatitis C guideline on the classification, diagnosis investigation, and immusup-
virus infection? Journal of Neurology Neurosurgery and Psychiatry, pressive therapy of non-systemic vasculitic neuropathy: executive sum-
60, 661–6. mary. Journal of the Peripheral Nervous System, 15, 176–84.
Aries, P.M., Hellmich, B., and Gross, W.L. (2005). Intravenous immunoglob- Collins, M.P., Mendell, J.R., Periquet, M.I., et al. (2000). Superficial fibular
ulin therapy in vasculitis. Clinical Review in Allergy and Immunology, (peroneal) nerve/poeroneus brevis muscle biopsy in vasculitic neuropa-
25, 237–45. thy. Neurology, 55, 636–43.
Asbury, A.K. and Johnson, P.C. (1978). Pathology of peripheral nerve. W.B. Collins, M.P., Periquet, M.I., Mendell, J.R., Sahenk, Z., Nagaraja, H.N., and
Saunders, Philadelphia. Kissel, J.T. (2003). Nonsystemic vasculitic neuropathy: insights from a
Beggs, J., Johnson, P.C., Olafsen, A., et al. (1991). Transperineurial arterioles clinical cohort. Neurology, 61, 623–30.
in human sural nerve. Journal of Neuropathology and Experimental Conn, D.L. and Dyck, P.J. (1984). Angiopathic neuropathy in connective tis-
Neurology, 50, 704–18. sue disease. In Peripheral neuropathy (ed. P.J. Dyck, P.K. Thomas, E.H.
Bell, M.A. and Weddell, A.G.M. (1984a). A descriptive study of the blood Lambert, and R. Bunge), pp. 2027–43. W.B. Saunders, Philadelphia.
vessels of the sciatic nerve in the rat, man and other mammals. Brain, Correia, Cda. C, Teixeira, H.M., and Melo, R.V. (2011). Vasculitic neuropathy
107, 871–98. presenting as Churg-Strauss pararneoplastic syndrome: a rare associa-
Bell, M.A. and Weddell, A.G.M. (1984b). A morphometric study of tion. Arquivos de Neuro-psiquiatria, 69, 994–5.
intrafascular vessels of mammalian sciatic nerve. Muscle Nerve, 7, Cruz Martinez, A., Barbado, F.J., Ferrer, M.T., et al. (1988). Electrophysiological
524–34. study in systemic necrotizing vasculitis of the polyarteritis nod as a
Bleehen, S.S., Lovelace, R.E., and Cotton, R.E. (1963). Mononeuritis mul- group. Electromyography and Clinical Neurophysiology, 28, 167–73.
tiplex in polyarteritis nodosa. Quarterly Journal of Medicine, 32, Dahlberg, P., Lockhart, J., and Overholt, E. (1989). Diagnostic studies for sys-
193–209. temic necrotizing vasculitis: sensitivity, specificity, and predictive value
CHAPTER 13 vasculitic neuropathy 175

in patients with multisystem disease. Archives of Internal Medicine, Hawke, S.H.B., Davies, L., Pamphlett, R., Guo, Y.P., Pollard, J.D., and McLeod,
149, 161–5. J.G. (1991). Vasculitic neuropathy. A  clinical and pathological study.
David, W.S., Peine, C., Schlesinger, P., and Smith, S.A. (1996). Nonsystemic Brain, 114, 2175–90.
vasculitic mononeuropathy multiplex, cryoglobulinemia, and hepatitis Haynes, B.F., Allen, N.B., and Fauci, A.S. (1986). Diagnostic and therapeu-
C. Muscle and Nerve, 19, 1596–602. tic approach to the patient with vasculitis. Medical Clinics of North
Davidson, J.R. and Sundstrom, W.R. (1988). Sural nerve biopsy in systemic America, 70, 355–68.
necrotizing vasculitis. Arthritis and Rheumatism, 31,149–50. Hunder, G.G., Arend, W.P., Bloch, D.A., et al. (1990). The American College
Davies, L. (1994). Vasculitic neuropathy. Baillliéré Clinical Neurology. 3, of Rheumatology 1990 criteria for the classification of vasculitis.
193–210. Arthritis and Rheumatism, 33, 1065–7.
Davies, L., Spies, J., Pollard, J., and McLeod, J. (1996). Vasculitis confined to Iwata, M., Kondo, M., Ando, M., et  al. (1993). [Peripheral polyneuropa-
peripheral nerves. Brain, 119, 1441–8. thy due to sarcoidosis in a patient with intrathoracic, ocular and skin
De Vita, S., Quartuccio, L., Isola, M., et al. (2012). A randomized controlled lesions]. [Japanese]. Nihon Kyobu Shikkan Gakkai Zasshi. Japanese
trial of rituximab for the treatment of severe cryoglobulinemic vasculi- Journal of Thoracic Diseases, 31, 1050–5.
tis. Arthritis and Rheumatism, 64, 843–53. Jamieson, P., Giuliani, M., and Martinez, J. (1991). Necrotizing angiopathy
Dyck, P.J., Benstead, T.J., Conn, D.L., Stevens, J.C., and Windebank, A.J. presenting with multifocal conduction blocks. Neurology, 41, 442–4.
(1987). Non-systemic vasculitic neuropathy. Brain, 110, 843–54. Jayne, D.J., Chapel, H., Adu, D., Misbah, S., O’Donoghue, D., Scott,
Dyck, P.J., Conn, D.L., and Okazaki, H. (1972). Necrotizing angiopathic neu- D., and Lockwood, C.M. (2000). Intravenous imunoglobuin for
ropathy: three dimensional morphology of fiber degeneration related to ANCA-associated systemic vasculitis with persistent disease activity.
sites of occluded vessels. Mayo Clinics Proceedings, 47, 461–75. QJM, 93, 433–9.
Dyck, P.J., Hansen, S., Karnes, J., et al. (1985). Capillary number and percent- Jayne, D.J., Rasmussen, N., Andrassy, K., Bacon, P., et  al. (2003). A ran-
age closed in human diabetic sural nerve. Proceedings of the National domized trial of maintenance therapy for vasculitis associated with
Academy of Science USA, 82, 2513–17. antineutrophil cytoplasmic autoantiobides. New England Journal of
Dyck, P.J. and Lofgren, E.P. (1968). Nerve biopsy. Choice of nerve, method, Medicine, 349, 36–44.
symptoms and usefulness. Medical Clinics of North America, 52, Jayne, D.R.W., Gaskin, G., Rasmussen, N., Abramowicz, D., et  al. (2007).
885–93. Randomized trial of plasma exchange or high-dose methylprednisolone
Dyck, P.J., Norell, J.E., and Dyck, P.J. (1999). Microvasculitis and ischemia as adjunctive therapy for severe renal vasculitis. Journal of the American
in diabetic lumbosacral radiculoplexus neuropathy. Neurology, 53, Society of Nephrology, 18, 2180–8.
2113–21. Jones, R.B., Tervaert, W.C., Hauser, T., et  al. (2010). Rituximab versus
Fathers, E. and Fuller, G.N. (1996). Vasculitic neuropathy. British Journal of cyclsophamide in ANCA-associated renal vasculitis. New England
Hospital Medicine, 55, 643–7. Journal of Medicine, 363, 211–10.
Fauci, A.S., Haynes, B.F., and Katz, P. (1978). The spectrum of vasculitis: clin- Kararizou, E., Davaki, P., Karandreas, N., Davou, R., and Vassilopoulos, D.
ical, pathologic, immunologic and therapeutic considerations. Annals of (2005). Nonsystemic vasculitic neuropathy: a clinicopathological study
Internal Medicine, 89, 660–76. of 22 cases. Journal of Rheumatology, 32, 853–8.
Ford, R. and Sickert, R. (1965). Central nervous system manifestations of Kelkar, P., Masood, M., and Parry, G.J. (2000). Distinctive pathologic findings in
periarteritis nodosa. Neurology, 15, 114–22. proximal diabetic neuropathy (diabetic amyotrophy). Neurology, 55, 83–8.
Fronert, P.P. and Sheps, S.G. (1967). Long-term follow-up study of periarteri- Kernahan, J.W. and Woltman, H.W. (1938). Periarteritis nodosa: a clinico-
tis nodosa. American Journal of Medicine, 43, 8–14. pathologic study with special reference to nervous system. Archives of
Fujimura, H., Lacroix, C., and Said, G. (1991). Vulnerability of nerve fibers Neurological Psychiatry, 39, 655–86.
to ischemia. A quantitative light and electron microscope study. Brain, Khella, S.L., Frost, S., Hermann, G.A., et al. (1995). Hepatitis C infection,
114, 731–3. cryoglobulinemia, and vasculitic neuropathy. Treatment with interferon
Fuller, G.N., Jacobs, J.M., and Guiloff, R.J. (1993). Nature and incidence of alpha: case report and literature review. Neurology, 45, 407–11.
peripheral nerve syndromes in HIV infection. Journal of Neurology Kissel, J.T., Eiethman, J.L., Omerza, J., Rammohan, K.W., and Mendell, J.R.
Neurosurgery and Psychiatry, 56, 372–81. (1989). Peripheral nerve vasculitis: immune characterization of the vas-
Garcia, R., Godlewski, W., Gruner, J., et al. (1955). Sur les formes multinevri- cular lesions. Annals of Neurology, 25, 291–7.
tiques et polynevritiques de la perarterite noueuse; Etude de 7 observa- Kissel, J.T. and Mendell, J.R. (1992). Vasculitic neuropathy. Neurology
tions inedites. Annals of Medicine, 56, 113–47. Clinics, 10, 761–81.
Gelassi, G., Gibertoni, M., Mancini, A., et  al. (1984). Sarcoidosis of the Kissel, J.T., Slivka, A.P., Warmolts, J.R., and Mendell, J.R. (1985). The clinical
peripheral nerve: Clinical, electrophysiological and histological study of spectrum of necrotizing angiopathy of the peripheral nervous system.
two cases. European Journal of Neurology,23, 459–65. Annals of Neurology, 18, 251–7.
Gherardi, R., Belec, L., Mhiri, C., Gray, F., Lescs, M.C., Sobel, A., Guillevin, Krendal, D.A., Costigan, D.A., and Hopkins, L.C. (1995). Successful treat-
L., and Wechsler, J. (1993). The spectrum of vasculitis in human immu- ment of neuropathy in patients with diabetes mellitus. Archives of
nodeficiency virus-infected patients—A clinopathologic evaluation. Neurology, 52, 1053–61.
Arthritis and Rheumatism, 36, 1164–74. Lacomis, D., Giuliani, M.J., Steen, V., and Powell, H.C. (1997). Small fiber
Giannini, C. and Dyck, P.J. (1993). Ultrastructural morphometric features of neuropathy and vasculitis. Arthritis and Rheumatism, 40, 1173–7.
human sural nerve endoneurial microvessels. Journal of Neuropathology Langford, C. and Sneller, M.C. (1997). New developments in the treatment
and Experimental Neurology, 52, 361–9. of Wegener’s granulomatosis, polyarteritis nodosa, microscopy polyarte-
Gisselbrecht, M., Cohen, O., Lortholary, O., et al. (1997). HIV -related vas- ritis and Churg-Strauss. Current Opinion in Rheumatology, 7, 26–30.
culitis: Clinical presentation and therapeutic approach on six patients. Levy, Y., Uziel, Y., Zandman, G.G., Amital, H., Sherer, Y., Langavitz, P.,
AIDS, 11, 121–3. Goldman, B., and Shoenfeld, Y. (2003). Intravenous immunoglobu-
Guillevin, L., Lhote, F., Jarrousse, B., et al. (1992). Treatment of polyarteritis lins in peripheral neuropathy associated with vasculitis. Annals of the
nodosa and Churg-Strauss syndrome. A  meta-analysis of 3 prospec- Rheumatic Disease, 62, 1221–3.
tive controlled trials including 182 patients over 12 years. Annales de Lie, J. (1990). Illustrated histopathologic classification criteria for selected
Médecine Interne (Paris), 143, 405–16. vasculitis syndromes. Arthritis and Rheumatism, 33, 1074–87.
Halpern, J.J., Little, B.W., Coyle, P.K., and Dattwyler, R.J. (1987). Lyme dis- Llewelyn, J.G., Thomas, P.K., and King, R.H. (1998). Epineurial microvasculi-
ease: cause of treatable peripheral neuropathy. Neurology, 37, 1700–6. tis in proximal diabetic neuropathy. Journal of Neurology, 245, 159–65.
176 SECTION 3 clinical manifestations common to vasculitis

Lockwood, C.M. (1996). New treatment strategies for systemic vasculitis: the Said, G., Coulon-Goeau, E., Lacroix, E., and Moulonguer, A. (1994). Nerve
role of intravenous immune globulin therapy. Clinical and Experimental biopsy findings in different patterns of proximal diagetic neuropathy.
Immunology, 1, 77–82. Annals of Neurology, 35, 559–69.
Lovelace, R.E. (1964). Mononeuritis multiplex in polyarteritis nodosa. Said, G., Goulon-Goeau, E., Lacroix, E., et al. (1988a). Inflammatory lesions
Neurology, 14, 434–42. of peripheral nerve in a patient with human T-lymphorrophic virus type
Loveshin, L.L. and Kernahan, J.W. (1948). Peripheral neuritis in periarteritis I-associated myelopathy. Annals of Neurology, 24, 275–7.
nodosa: a clinocopathological study. Archives of Internal Medicine, 82, Said, G., Lacroix, C., Plante-Bordeneuve, V., et al. (2002). Nerve granulomas
321–38. and vasculitis in sarcoid peripheral neuropathy: a clinicopathological
Mathew, L., Talbot, K., Love, S., et al. (2007). Treatment of vasculitic periph- study of 11 patients. Brain, 125, 264–75.
eral neuropathy: a retrospective analysis of outcome. QJM, 100, 41–51. Said, G., Lacroix, E., Andrieu, J.M., et  al. (1987). Necrotizing arteritis in
Maxeiner, S., McDonald, J., and Kirklin, J. (1952). Muscle biopsy in the diag- patients with inflammatory neuropathy and immunodeficiency virus
nosis of periarteritis nodosa: an evaluation. Surgical Clinics of North infection. Neurology, 37 (Suppl.), 176.
America, 32, 1223–35. Said, G., Lacroix-Ciaudo, E., Fujimura, H., Bias, E., and Faux, N. (1988b).
McManis, P.G., Low, P.A., and Lagerlund, T.D. (1993). Microenvironment of The peripheral neuropathy of necrotizing arteritis: a clinicopathological
nerve: blood flow and ischemia. In Peripheral neuropathy (ed. P.J. Dyck study. Annals of Neurology, 23, 461–5.
and P.R. Thoma), pp. 453–75, 3rd edn. W.B. Saunders, Philadelphia. Sakai, K., Komai, K., Yanase, D., and Yamada, M. (2005). Plasma VEGF as a
Meier, C., Grahmann, F., Engelhat, A., and Dumas, M. (1989). Peripheral marker for the diagnosis and treatment of vasculitic neuropathy. Journal
nerve disorders in lyme borreliosis: nerve biopsy studies in eight cases. of Neurology Neurosurgery and Psychiatry, 76, 296.
Acta Neuropathol (Berlin), 79, 271–178. Sanche-Guerrero, J., Gutrre-Urena, S., Vidaller, A., et al. (1990). Vasculitis as
Midroni, G. and Bilbao, J.M. (1995). Biopsy diagnosis of peripheral neuropa- a paraneoplastic syndrome. Report of 11 cases and review of the litera-
thy. Butterworth-Heinemann, Boston. ture. Journal of Rheumatology, 17, 1458–62.
Moore, P.M. and Fauci, A.S. (1981). Neurologic manifestations of systemic Schifitto, G., Barbano, R.L., Kieburtz, K.D., Cohn, S.E., and Zwillich, S.H.
vasculitis. American Journal of Medicine, 71, 517–24. (1997). HIV related vasculitic mononeuropathy multiplex:  a role for
Muley, S.A., Brown, K., and Parry, G.J. (2008). Paraneoplastic vasculitic IVIG? Journal of Neurology Neurosurgery and Psychiatry, 63, 255–6.
neuropathy related to carcinoid tumor. Journal of Neurology, 255, Schoene, W.E., Masters, E.L., Gibbs, E.J., Gajdusek, D.E., Tyler, H.R., Moore,
1085–7. F.D., and Dammin, G.J. (1981). Transmissible spongiform encephalopa-
Nemni, R., Corbo, M., Fazio, R., Quattrini, A., Comi, G., and Canal, N. thy (Creutzfeld Jakob disease). Atypical clinical and pathological find-
(1988). Cryoglobulinaemic neuropathy: a clinical, morphological and ings. Archives of Neurology, 38, 473–7.
immunocytochemical study of eight cases. Brain, 111, 541–52. Schroder, J.M. (1986). Proliferation of epineural capillaries and smooth muscle
Oh, S.J. (1990). Diagnostic usefulness and limitations of the sural nerve cells in angiopathic peripheral neuropathy. Acta Neuropathology, 72, 29–37.
biopsy. Yonsei Medical Journal, 31, 1–26. Scott, D.G.I. and Bacon, P.A. (1984). Intravenous cyclophosphamide and
Oh, S.J. (1997). Paraneoplastic vasculitis of the peripheral nervous system. methylprednisolone in treatment of systemic rhematoid vasculitis.
Neurologic Clinics, 15, 849–63. American Journal of Medicine, 76, 377–84.
Oh, S.J., Herrara, G., and Spalding, D.M. (1986). Eosinophilic vasculitic neu- Seo, J.H., Ryan, H.F., Claussen, G.C., Thomas, T.D., and Oh, S.J. (2004).
ropathy in the Churg-Strauss syndrome. Arthritis and Rheumatism, 29, Sensory neuropathy in vasculitis. A clinical, pathlogic, and electrophysi-
1173–5. ologic study. Neurology, 63, 874–8.
Panegyres, P.K., Iumbergs, P.E., Leong, A.S.Y., and Bourne, A.J. (1990). Sladky, J.T., Greenberg, J.H., and Brown, M.J. (1985). Regional perfusion in
Vasculitis of peripheral nerve and skeletal muscle:  clinopathological normal and ischemic rat sciatic nerves. Annals of Neurology, 17, 191–5.
correlation and immunopathic mechanisms. Journal of Neurological Sneller, M.C., Hu, Z., and Langford, C.A. (2012). A randomized controlled trial of
Science, 100, 193–202. rituximab following failure of antiviral therapy for hepatitis C virus-associated
Puechal, X., Said, G., Hilliquin, P., et al. (1995). Peripheral neuropathy with cryoglobulinemic vasculitis. Arthritis and Rheumatism, 64, 835–42.
necrotizing vasculitis in rheumatoid arthritis. A clinicopathologic and Stone, J.H., Merkel, P.A., Spiera, R., et al. (2010) Rituximab versus cyclsophos-
prognosis study of thirty-two patients. Arthritis and Rheumatism, 38, phamide for ANCA-associated vasculitis. New England Journal of
1618–29. Medicine, 363, 221–32.
Radice, A. and Sinico, R.A. (2006). Antineurotrophile cytoplasmic antibodies Suggs, S.P., Thomas, T.D., Joy, J.L., Lopez-Mendez, A., and Oh, S.J. (1992).
(ANCA). Autoimmunity, 38, 93–103. Vasculitic neuropathy mimicking Guillain-Barre syndrome. Arthritis
Raff, M.C., Sangalang, V., and Asbury, A.K. (1968). Ischemic mononeuropa- and Rheumatism, 35, 975–8.
thy multiplex associated with diabetes mellitus. Archives of Neurology, Suppiah, R., Hadden, R.D.M., Batra, R., Arden, N.K., Collins, M.P., Guillevin,
18, 487–99. L., Jayne, D.R.W., Luqmani, R.A. (2011). Peripheral neuropathy in
Rasmussen, N., Jayne, D.R.W., Abramowicz, D., et al. (1996). European thera- ANCA-associated vasculitis:  outcome from the European Vasculitis
peutic trials in ANCA associated systemic vasculitis: Disease scoring, Study Group trials. Rheumatology,50, 2214–22.
consensus regimens and proposed clinical trials. Sarcoidosis, 13, 29–34. Turner, M.R., Warren, J.D., Jacobs, J.M., et al. (2003). Microvasculitic para-
Richter, E., Schanbel, E., Csernok, E., et  al. (1994). Treatment of proteinemic polyneuropathy and B-cell lymphoma. Journal of the
ANCA-associated systemic vasculitis with high-dose intravenous Peripheral Nervous System, 8, 100–7.
immunoglobulin. Arthritis and Rheumatism, 37, S3–S3. Vallat, J.M., Hugon,J., Lubveau, M., Lebourtet, M.J., Dumas, M., and
Ropert, A. and Metral, S. (1990). Conduction block in neuropathies with Desproges-Gottgeron, R. (1987). Tickbite meningoradiculoneuritis: clini-
necrotizing vasculitis. Muscle Nerve, 13, 102–5. cal, electrophysiolgic, and histologic findings in 10 cases. Neurology, 37,
Sack, M., Cassidy, J.T., and Bole, G.G. (1975). Prognostic factors in polyarte- 749–53.
ritis. Journal of Rheumatology, 2, 411–20. Vernant, J.C., Smadja, D., Deforge-Lasseur, C., Cabre, P., Beulsson, G.,
Said, G. (1994). Inflammatory neuropathies associated with known infec- Neisson-Vernant, C., and Desgranges, C. (1994). Vasculitis and neuro-
tions (HIV, leprosy, Chagas’s disease, Lyme disease). Baillière's Clinical logic manifestations related to HTLV-1. Presse Médicale, 23, 1421–5.
Neurology, 3, 149–71. Vital, C., Aubertin, J., Ragnault, M., Amigues, H., Mouton, L., and Bellance,
Said, G. (1995). Vasculitic neuropathy. Baillière's Clinical Neurology, 4, R. (1982). Sarcoidosis of the peripheral nerve: a histological and ultas-
489–503. tructural study of two cases. Acta Neuropathol, 58, 111–4.
CHAPTER 13 vasculitic neuropathy 177

Vital, C., Vital, A., Canron, M.H., Jaffre, A., Vitallard, J.F., Ragnaud, J.M., Winkelstein, A., Starz, T.W., and Agarwal, A. (1984). Efficacy of combined
Brechenmacher, C., and Lagueny, A. (2006). Combined nerve and mus- therapy with plasmapheresis and immunosuppressants in rheumatoid
cle biopsy in the diagnosis of vasculitic neuropathy. A  16-year retro- vasculitis. Journal of Rheumatology, 11, 162–6.
spective study of 202 cases. Journal of the Peripheral Nervous System Younger, D.S. (2011). Diabetic lumbosacral radiculoplexus neuropathy:  a
11, 20–9. postmortem studied patient and review of the literature. Journal ofNeu-
Vrancken, A.F.J.E., Hughes, R.A.C., Said, G., et al. (2011). Immunosuppressive rology, 258, 1364–7.
treatment for non-systemic vasculitic neuropathy. Cochrane Library Younger, D.S., Rosoklija, G., Hays, A.P., Trojaborg, W., and Latov, N.
2011, Issue 9. (1986). Diabetic peripheral neuropathy:  a clinicopathologic and
Wees, S.J., Sunwoo, L.N., and Oh, S.J. (1981). Sural nerve biopsy in systemic immunohistochemical analysis of sural nerve biopsies. Muscle Nerve,
necrotizing vasculitis. American Journal of Medicine, 71, 525–32. 19, 722–7.
CHAPTER 14

Vasculitic manifestations in
the gastrointestinal tract
Gaafar Ragab

Introduction The microbiome


Gastrointestinal (GI) vasculitis used to be underrepresented in the Lederberg (2000) defined the term microbiome as the ecologi-
medical literature, in spite of its significant morbidity and mortal- cal communities of commensal, symbiotic, and pathological
ity. There is, however, an increased awareness of the importance micro-organisms that share our body space. They contribute to our
of early diagnosis and prompt management to avoid its potentially health and disease (Lederberg and MacCray 2001), among which
serious sequelae. autoimmune disorders occupy a prominent position (Chervonsky
In order to better understand the manifestations and conse- 2010). Humans can be regarded as a ‘super organism’ composed
quences of vasculitic lesions in the GI tract, and the different of human and non-human cells and their genomes (Scher and
approaches to diagnosis, it is necessary to briefly review the anat- Abramson 2011). The largest and most complex component is
omy and physiology of the GI tract. It is also important to under- intestinal bacteria, which includes as many as 1012 cells per 1 g of
stand the various pathogenic mechanisms causing bowel ischaemia, faeces in the average human individual (Savage 1977).
and the wide range of differential diagnosis. Advances in metagenomic technologies have recently begun to
The recent developments in our understanding of the human gut unravel the human gut microbiota. It is estimated that each individ-
microbiome is also expected to contribute to our knowledge of the ual houses at least 160 such species from a consortium of 1000 to
role of the GI tract in autoimmunity, and its potential contribution 1500 prevalent bacterial species (Qin et al. 2010). Our knowledge of
to future therapeutic approaches. species and functional composition of the human gut microbiome
is still based on very few cohorts and little is known about variation
Anatomic considerations across the world (Arumugam et al. 2011).
The intestinal microbes are taxonomically complex and con-
Blood supply to the abdominal organs is provided by three major stitute an ecological dynamic community (microbiota) that has
unpaired vessels arising from the abdominal aorta, namely the long been believed to exert a strong impact on human physiol-
celiac trunk, and the superior and inferior mesenteric arteries ogy. They are also involved in the maturation and proliferation
(Geboes et al. 2001). of human intestinal cells, helping to maintain their homeostasis,
The bowel vasculature should be viewed as a single functional but they can cause various diseases, such as inflammatory bowel
unit. Whether ischaemia develops depends mainly on the amount disease (Hattori and Taylor 2009) and rheumatoid arthritis (RA)
of blood that flows into a diseased artery from other arter- (Scher and Abramson 2011). Intestinal micro-organisms have
ies (Cognet et al. 2002). Multiple visceral arterial lesions may be also been incriminated in some vasculitic events such as Henoch–
found in patients without symptoms, while others with a single Schönlein purpura (HSP) (Roza et  al. 1983; Rasmussen 1982;
lesion have pain (Roobottom and Dubbins 1993). The occurrence Chen et al. 2005).
of painful ischaemia will depend upon the site of the lesion, for A role for the intestinal microenvironment, particularly, in the
example whether it is downstream from an anastomosis or not. The ‘putative’ localized GI vasculitis, has been hypothesized, and may
ability of a patient to develop collaterals may be compromised if explain why surgical resection puts an end to this inflammatory
there are other vascular abnormalities. The tempo of progression of process (Raza et al. 1999).
the lesion will also play a role. For example in Takayasu’s arteritis,
severe occlusion may occur before there is a chance for anastomotic
collaterals to develop (Cognet et al. 2002). Pathophysiology of GI tract circulation
Thanks to lavish collateral connections, proximal mesen- Bowel ischaemia can be precipitated by under-perfusion or occlu-
teric venous occlusion does not usually result in severe bowel sion; the initial lesions are followed by an inflammatory response
ischaemia, unlike occlusion of distal mesenteric veins, which due to the release of mediators such as platelet -activating factor and
causes infarction of the bowel wall with serious consequences cytokines, including tumour necrosis factor (TNF-α) (Bradbury
(Rademaker 1998). et  al. 1993; Kuroda et  al. 1994), from activated neutrophils,
180 SECTION 3 clinical manifestations common to vasculitis

macrophages, platelets, mast cells, and endothelium. They result in virus); 11 Churg–Strauss syndrome (now called eosinophilic gran-
further damage to the bowel wall. ulomatosis with polyangiitis); six Wegener’s granulomatosis (now
Acute bowel ischaemia passes through three stages. The first called granulomatosis with polyangiitis); four microscopic poly-
stage, referred to as reversible ischaemic enteritis or colitis, is char- angiitis; and three rheumatoid arthritis associated vasculitis. GI
acterized by mucosal necrosis, erosions, ulcerations, and sometimes tract manifestations were present at, or occurred within 3 months
haemorrhage (Haglund and Bergqvist 1999). The second stage of diagnosis in 50 (81%) patients. Symptoms were abdominal
shows deeper damage with necrosis reaching the deep submucosa. pain in 61 (97%); nausea or vomiting in 21 (34%); diarrhoea in 17
Stricture formation may ensue from healing by fibrosis (Whitehead (27%); haematochezia or melena in 10 (16%); and haematemesis
1976). The third stage is transmural bowel wall necrosis or infarc- in four (6%). Endoscopic detection of gastroduodenal ulceration
tion and is associated with a high mortality rate. Superinfection was noted in 17 (27%) patients, oesophageal ulceration in seven
after the breakdown of the mucosal barrier will further contribute (11%) patients, and colorectal ulceration in six (10%) patients,
to the process of wall necrosis (Musemeche et  al. 1986; Ouellet but histological signs of vasculitis were found in only three colon
et al. 1988). biopsies. Twenty-one (34%) patients had a surgical abdomen; 11
Toxic proteases, bacteria and their toxins, as well as free radicals (18%) developed peritonitis; nine (15%) had bowel perforations;
produced in the reperfusion phase will contribute to severe intesti- 10 (16%) bowel ischaemia/ infarction; four (6%) intestinal occlu-
nal necrosis. Serious systemic effects may result, including: hepatic sion; six (10%) acute appendicitis; five (8%) cholecystitis; and
and renal dysfunction; myocardial and circulatory changes; bone three (5%) acute pancreatitis. Some patients had more than one
marrow failure; increased risk of sepsis; disseminated intravascu- condition, and 16 (26%) patients died. The respective 10-month
lar coagulation; and multiple organ failure (Harward et al. 1993; and 5-year survival rate were 71% and 56% for the 21 surgical
Jamieson et  al. 1993; Fontes et  al. 1994; Landow and Anderson patients; whereas they rose to 94% and 82% for the 41 patients
1994; Smith et al. 1994). without a surgical abdomen. Peritonitis, perforations, ischaemia
or infarctions, and intestinal occlusion were the only GI tract
manifestations significantly associated with increased mortality
Manifestations of GI tract vasculitis in multivariate analysis. For the 15 patients with these conditions,
GI tract vasculitis has a limited repertoire of clinical symptoms 6-month and 5-year survival rates were 60% and 46%, respectively.
and signs. The extent and course of disease depend on the size and None of the other GI tract or extraintestinal vasculitis related
location of the affected vessel and the histological characteristics of symptoms or angiographic abnormalities was predictive of sur-
the lesion. Radiological findings in various types of vasculitis often gical complications or poor outcome. They noted, however, that
overlap considerably and therefore have limited value in specific prognosis has dramatically improved over the 30-year period rep-
diagnosis. Nevertheless, the possibility of vasculitis should be con- resented by this study, and attributed this to better management
sidered whenever: mesenteric ischaemic changes occur in young with prompt surgical intervention when indicated, and the com-
patients; are noted at unusual sites such as stomach, duodenum, or bined use of glucocorticoids and immunosuppressive drugs.
rectum; concomitantly involve the small and large intestine; or are
associated with genitourinary involvement. Knowledge of systemic
clinical manifestations in affected patients may suggest a specific Imaging
diagnosis (Ha et al. 2000). Despite the technical developments of non-invasive imaging
Acute bowel ischaemia with transmural infarction causes local modalities such as CT, MRI, and ultrasound, selective angiogra-
complications such as bleeding, perforation, abscess formation, and phy of mesenteric arteries is still the gold standard in diagnosing
peritonitis. It is clear that partial mural bowel ischaemia may also peripheral splanchnic vessel disease (Trompeter et al. 2002).
result in similar consequences upon disruption of the mucosal bar- The diagnosis of chronic mesenteric ischaemia requires confi-
rier. Whether arterial or venous, other causes of mesenteric ischae- dent elimination of other conditions because this condition often
mia must be kept in mind. Causes of non-occlusive reduction of does not manifest as one or more arterial stenoses on vascular
mesenteric blood supply must be appreciated as well. imaging. Barium enema may show thumbprinting due to submu-
Symptoms of GI tract vasculitis include:  abdominal angina, cosal oedema or haemorrhage. This sign is fairly specific for ischae-
defined as postprandial abdominal pain with weight loss and ano- mic bowel (Sultan et al. 1999).
rexia; changes in bowel habits and vomiting are less common. In Duplex ultrasound is an accurate screening test for proximal
the stomach, vasculitis may present as incapacitating gastroparesis arterial stenosis or occlusion. A  peak systolic velocity greater
with vomiting, postprandial heaviness, and delayed gastric empty- than 275 cm/s seems to be highly specific for significant superior
ing. Casey et al. (1993) advised investigating the visceral arteries, mesenteric artery (SMA) stenosis (Moneta et  al. 1993)  and an
at least with Doppler ultrasound, in patients with chronic gastric end-diastolic velocity greater than 45 cm/s may be even more accu-
symptoms unresponsive to pharmacotherapy. Cachexia suggesting rate (Perko 2001). Blunting of fasting and postprandial differences
a malignancy can occur with severe malabsorption or if the patient in peak systolic velocity in the SMA (as assessed by ultrasound
eats less to avoid triggering the pain. As these symptoms are not duplex scanning) was reported in patients with high-grade stenosis
uncommon, and tend to develop insidiously, the diagnosis may be (Gentile et al. 1995), suggesting failure of the mesenteric blood flow
delayed. to adapt to meal-related circulatory needs.
Pagnoux et al. (2005) reviewed the medical record of 62 patients Ripollés et al. (2005) studied sonographic findings in 58 patients
with systemic small and medium-sized vessel vasculitis and GI with proven ischaemic colitis, and reported a prospective sensitiv-
tract involvement followed between 1981 and 2002. Vasculitides ity of sonography for the characterization of colonic abnormalities
were distributed as follows:  38 PAN (21 related to hepatitis B of 93.5% (58/62 patients). Segmental involvement was observed
CHAPTER 14 vasculitic manifestations in the gastrointestinal tract 181

in 28%. Altered pericolic fat was the only sonographic variable approaches should be initiated in cases with expected positive out-
significantly associated with the presence of transmural necrosis. come, and if the timing allows. Observation should be in an ER
Contrast-enhanced MR angiography has been useful for detection setting, and surgical decisions should not be delayed to minimize
of significant arterial stenosis (Meaney et al. 1997). An important morbidity and mortality.
advantage, compared to CT angiography, is its relative safety.
Arterial-phase CT scans and CT angiograms are of great value
in the assessment of mesenteric arteries. They are helpful in iden-
Regional (localized) vasculitis
tifying the site, level, and cause of bowel ischaemia. Portal venous of the gastrointestinal tract
scans have additional advantages of depicting mesenteric veins, This describes an entity of localized lesions, which are not part of a
allowing better assessment of the bowel wall itself, and providing systemic process at the time of its presentation. Pathological exam
greater accuracy in the detection of perforation, abscess formation, may reveal that the lesion belongs to an established diagnostic cat-
and peritonitis (Wiesner et al. 2003). Acute bowel ischaemia may egory (Burke et al. 1995; Juliano et al. 2009; Raza et al. 1999).
cause various morphological changes, including: homogenous or GI vasculitis has an excellent prognosis when compared to sys-
heterogeneous hypo- or hyperattenuating wall thickening; dilata- temic variants after treatment with glucocorticoids and selective
tion; abnormal or absent wall enhancement; mesenteric stranding; surgery. Attention to this possibility saves the patient the risk of
vascular engorgement; ascites; pneumatosis; and portal venous gas unnecessary cytotoxic therapy. Raza et al. (1999) emphasized three
(Bartnicke and Balfe 1994; Ha et al. 2000). Acute bowel ischaemia points to be considered in this regard: (1) thorough exclusion of
may affect the small or large bowel and may be diffuse or localized, active systemic vasculitis, (2) informed consent by the patient, and
segmental or focal, and superficial or transmural; therefore it can (3) regular follow-up to exclude new activity. There is no consensus
mimic various diseases (Horton et al. 1999; Horton et al. 2000). in the literature on the duration of this follow-up.
Byun et al. (1999) accepted the presence of at least three of the
following signs on abdominal CT as diagnostic of ischaemic bowel
disease: bowel wall thickening; a target sign; dilation of an intesti- Takayasu’s arteritis
nal segment; engorgement of vessels; and increased attenuation of In Takayasu’s arteritis (TA), non-specific gastrointestinal symptoms,
mesenteric fat. There are three major pitfalls by which CT detection such as anorexia, nausea, vomiting, and loss of weight, may take
of bowel ischaemia may be missed. First, a colonic segment with place during the prepulseless phase. True gastrointestinal morbidity
superficial mucosal ischaemia may react with spasticity, which may resulting from arterial stenosis (Hall et al. 1985) and organ ischaemia
be misinterpreted as simple contraction. Second, bowel wall thick- is rare. Acute abdominal signs as the first manifestation, although
ness is different in a widely distended ischaemic colonic segment unusual, have been reported (Hands and Murie 1991; Cornejo et al.
than it is in a less-distended, normal colonic segment. Thus, a bowel 2002). Chronic mesenteric ischaemia presenting with weight loss
wall thickness of 3–5 mm in a distended colonic segment, which and postprandial pain has also been reported (Bongard et al. 1992;
would be normal for a non-distended segment, should be inter- Vega Sáenz et al. 1995; Tyagi et al. 1997; Mehta et al. 2004; Desai
preted as thickened. Third, there is a tendency to misinterpret bowel et al. 2004). The pain is typically postprandial, gradually increasing
wall dilatation and air–fluid level as ileus or pseudo-obstruction in severity, and then slowly resolving, and may be relieved by vom-
(Wiesner et al. 2003). In spite of these pitfalls, the sensitivity of CT iting. There is sometimes significant weight loss, and the patient
for the diagnosis of acute bowel ischaemia (82%) was comparable may develop a fear of eating (Mehta et al. 2004). Angeli et al. (2001)
to that of angiography (87.5%) when the two modalities were com- reviewed the literature for the prevalence of main aortic branch
pared directly (Klein et al. 1995). involvement (stenosis or occlusion) at angiography and cited the fol-
Non-invasive diagnostic investigations are very helpful and lowing in descending order: superior mesenteric (17–29%), inferior
increasingly promising; they are better used as screening tests for mesenteric (4–27%), and celiac (9–18%) (Figure 14.1). Koyama and
avoiding unnecessary angiograms. The diagnosis of ischaemia con- his team (1995) reported a case of hepatic artery aneurysm. Vanoli
tinues to rest on a careful medical history. Prompt surgical or inter- et  al. (2005) collected data of 104 Italian patients with TA. They
ventional decisions should not be delayed. reported the percentage of vascular involvement of any lesion, in
patients receiving full aortography, for the celiac trunk, the superior
Differential diagnosis and and inferior mesenteric arteries to be 21.4, 31.58, and 9.43, respec-
tively. In spite of its limitations, sonographic exploration of the aorta
management and its major branches is an excellent screening modality for pri-
Cases suspected of acute GI vasculitis should be promptly evalu- mary diagnosis (Figure 14.2), and thanks to its low invasiveness and
ated to exclude other causes of acute abdominal pain, as well as cost can be used for long-term follow-up of patients (Angeli et al.
non-vasculitic causes of mesenteric ischaemia. When the diag- 1999). Other techniques include CT, MRI, angiography, and positron
nosis of systemic vasculitis is established, the differential diagno- emission tomography (PET). The latter’s lack of spatial resolution
sis should include treatment-associated complications (Hokama improved with the use of PET-CT (Luna Alcala et al. 2009).
et  al. 2012)  such as opportunistic infections, for example CMV Intervention in cases with significant vessel involvement is usu-
(cytomegalovirus), in which case endoscopic findings may mimic ally multidisciplinary. Several approaches include surgery (Esato
those of vasculitis (Hokama et al. 2010; Hoshino et al. 2011). Also et al. 1982; Koyama et al. 1995; Desai et al. 2004) and radiological
non-steroidal anti-inflammatory drugs (NSAIDs) may cause pep- intervention (Bongard et al. 1992; Tyagi et al. 1997; Mehta et al.
tic ulcers, diaphragm disease (a circumferential ulcer causing a 2004). (Figure 14.3) Glucocorticoids should be used judiciously in
diaphragm-like stricture) (Hokama et  al. 2005), and other vari- the perioperative or peri-interventional period (Vega Sáenz et al.
ous types of enteropathy. Medical measures and interventional 1995; Mehta et al. 2004).
182 SECTION 3 clinical manifestations common to vasculitis

(a) (b)

Fig. 14.1 (a) Oblique parasagittal reformatted image using maximum intensity projection (MIP) technique for the abdominal aorta showing significant narrowings
at the origin of the celiac axis and superior mesenteric arteries (arrows). (b) CT angiogram using volume rendering technique for the abdominal aorta showing medial
patchy calcifications (arrowheads).

Giant cell arteritis branches, and a thick hypoechoic wall, referred to as a halo, where
thickening was diffuse and symmetric. This halo is similar to that
GI vasculitis is less common in GCA than in other vasculitides described by Schmidt (2000) and Schmidt et al. (1999). Trimble and
(Srigley and Gardiner 1980). Most reports of GCA involving the Weisz (2002) reported an 87-year-old male with infarction of the
GI tract are of small intestinal disease, in the form of infarction sigmoid colon secondary to GCA. The patient had an acute surgical
or perforation (Strigley and Gardiner 1980; Krant and Ross 1992; abdomen and at surgery the colon was markedly distended, and an
Smith et al. 1988). Lockhart and Robbin (2003) described a case of ischaemic segment of the sigmoid colon was resected. Microscopic
abdominal angina with other non-specific symptoms. The patient examination showed early infarction and GCA was noted through-
had an antecedent history of vasculitis suggestive of GCA. Her out the sections of the associated mesocolon. Heneghan et al. (1998)
physical examination was unremarkable. Angiogram suggested and Goulding et al. (2010) reported cases of hepatic involvement in
diffuse arterial abnormalities including those in the SMA. An GCT. Lee and her colleagues (2011) described a 65-year-old woman
intraoperative ultrasonography (US) image of the SMA demon- who presented with recurrent GI symptoms, fever, and a tender
strated marked narrowing with near occlusion as it divided into its right upper abdominal quadrant. She had raised inflammatory
markers and worsening liver biochemistry. Liver biopsy demon-
strated non-caseating, epithelioid cell, granulomatous inflamma-
tion of medium-sized arterioles within the portal tracts. The patient
responded dramatically within 3 days of high-dose prednisolone.
Before discharge, a temporal artery biopsy confirmed the diagnosis
of GCA. The authors recommended that GCA should be consid-
ered in a setting of fever, GI symptoms, and raised ESR in those
older than 50 years.

Polyarteritis nodosa
GI tract involvement has been reported in polyarteritis nodosa
(PAN) by many investigators, with percentages ranging from 14 to
53% (Frohnert and Sheps 1967; Fortin et al. 1995; Guillevin et al.
1995). Most older series have included patients with MPA and
Churg–Strauss syndrome (CSS) as well as those with PAN. GI tract
manifestations are among the most serious expressions of PAN.
Guillevin et al. (1995) studied the nature of incidence of GI mani-
Fig. 14.2 Arterial duplex examination of the aorta, celiac artery, and superior festation in 53 patients with PAN and CSS. Eighteen of the 53 cases
mesenteric arteries in a case of TA showing narrowing of SMA (arrow). (34%) had GI tract manifestations; these were considered among
CHAPTER 14 vasculitic manifestations in the gastrointestinal tract 183

(a) (b1) (b2)

Fig. 14.3 Lateral CT aortogram—a case of TA. (a) Before intervention showing complete occlusion of the celiac trunk starting from its ostium (red arrows) with
collateral filling of the hepatic and left gastric arteries which are markedly attenuated ‘thread like’. Occluded splenic artery and showing three stenotic lesions of SMA.
(b) After intervention: applied three stents are seen along the proximal portion of superior mesenteric artery; the proximal two stents are patent with no mural based
thrombi or intraluminal filling defects, while the distal stent shows re-stenosis causing about 60% luminal reduction (green arrows). Distally the artery is patent showing
normal calibre with no stenotic or occlusive lesions. No interval appreciable changes regarding the previously noted complete occlusion of the celiac trunk. The obtained
sections were processed to obtain angiographic images. Courtesy of Dr Loai Ezzat, Professor of Radiology, Cairo University.

the symptoms revealing PAN in seven (13.2%) of the 53 cases. Six pancreas (Griffith and Vural 1951; Ito et al. 1991), and the appendix
of the 18 patients with GI tract manifestations had definite organ (Moyama 1988; Ozcay et al. 2003) have all been reported. PAN may
involvement related to vasculitis. Abdominal pain without char- be detected at cholecystectomy or appendectomy in the absence
acteristic organ involvement or surgical abdomen was present in of other disease manifestations (Blidi et al. 1996). This apparently
12 of 18 patients. Furthermore, among the subgroup of patients isolated vasculitis usually appears as a necrotizing lesion with fibri-
with haemorrhage, perforation, digestive tract surgery due to PAN, noid necrosis but occasionally is granulomatous. How often these
intractable abdominal pain, and weight loss greater than 20% of findings represent a true systemic vasculitis as opposed to localized
normal weight attributable to GI tract ischaemia, there was a sig- inflammatory process of no significance is disputed. Vasculitis in
nificantly lower 10-year survival rate compared to patients without an organ removed for reasons other than systemic vasculitis should
GI tract manifestations (Guillevin et al. 1995). be interpreted cautiously and should prompt further history and
Guillevin and his group included GI involvement among the work-up for systemic disease if clinically suspicious.
items constituting the five-factor score, which can predict the 5-year
survival rate of patients with PAN. Stanson and his co-workers
(2001) reviewed the positive angiographic findings in 56 consecu-
tive patients with PAN. They reported the following arterial bed
distribution: gastric 33%, gastroduodenal 8%, hepatic 89%, SMA
97%, and inferior mesenteric artery (IMA) 100%. Angiographic
findings included aneurysms, ectasia, or occlusive disease. The true
frequency is difficult to determine because of the non-specificity of
findings and small sampling size. Jee et al. (2000) described multiple
aneurysms at the jejunal, ileal, right colic, and mid-colic branches of
the SMA, IMA, hepatic, and left gastric arteritis. Mesenteric infarc-
tion, perforation, or aneurysmal rupture in PAN is disastrous, and
relapses after treatment can take place(Guillevin et al. 1995; Levine
et al. 2002; Fauci 1978). Gastric and oesophageal perforations are
rare (Gourgoutis et al. 1971). Histological changes in the affected
arteries in PAN may include aneurysms with hyalinosis, focal neo-
vascularization, infiltration of mononuclear cells and neutrophils,
and focal capillaritis (Figure  14.4). Involvement of gallbladder
(Blidi et al. 1996; Gorgun et al. 2002; Parangi et al. 1991; Ito et al. Fig. 14.4 Polyarteritis nodosa. Microscopic examination (H & E staining) of celiac
1991; Nøhr et al. 1989; Kumar et al. 2003; Fernandes et al. 2005), the artery aneurysm, surgically removed from a patient with polyarteritis nodosa.
184 SECTION 3 clinical manifestations common to vasculitis

The liver is a common site of involvement in PAN. The vascu- recovered, but 50% developed coronary aneurysms despite early
lar changes result in aneurysm formation or rupture, infarction, or intravenous gammaglobulin (IVIg) treatment.
interstitial hepatitis. Hekali et al. (1991) compared the angiographi- Intestinal pseudo-obstruction has been reported to occur in
cally observed visceral aneurysms in 71 PAN patients to those of 2–3% of children with KD (Miyake et  al. 1987; Wheeler et  al.
other diseases with similar findings (seven from severe arterial 1990). Persistent vomiting, which is uncommon in KD, may be a
hypertension and three from rheumatoid arthritis). The mean clue to diagnosis (Akikusa et al. 2004). It tends to be seen early in
number of hepatic aneurysms was higher in PAN patients, and five pseudo-obstruction (Miyake et al. 1987), in contrast to mechani-
of 17 PAN patients had numerous and large aneurysms. In spite of cal bowel obstruction due to strictures, where vomiting generally
its common involvement, hepatic disease may remain undetected appears 2–4 weeks after the acute illness (Murphy et al. 1987; Mele
(Mowrey 1954). A constellation of presentations betraying hepatic and Evans 1996). The pathogenesis of this entity is felt to relate to
disease was, nonetheless, cited: haematoma (Fitchett and Oakley myenteric plexus. The absence of mesenteric vessel abnormalities
1975; Ayers and Fitchett 1992); intrahepatic bleeding and haemo- upon imaging suggests small-vessel disease (Miyake et  al, 1987;
bilia (Yazici et al. 1997); and haemorrhage (Brandstetter et al. 1995; Franken et al. 1979). The treatment varies from simple bowel rest
Ozcay et al. 2003). These may be further complicated by peritonitis (Wheeler et al. 1990) to intravenous glucocorticoids (Miyake et al.
or intestinal haemorrhage. Hepatic sequelae of HBV infection are 1987). In three cases, IVIg was given for pseudo-obstruction. In two
well appreciated. The incidence of HBV infection is decreasing and cases, it was administered concomitant with bowel symptoms, and
this is reflected in its diminishing role as a causative agent in PAN. in both there seems to have been a subsequent rapid resolution of
Endoscopy may detect ischaemic areas and ulcers. Total symptoms (Zulian et al. 2003; Fang et al. 2001). In the third case, the
abdominal angiography leads to more accurate appraisal of organ intestinal symptoms had resolved with conservative management
involvement (Ewald 1987), especially of occult aneurysms. This by the time the child was recognized as having KD and given IVIg
intervention should be considered in anticipation of the risk (Akikusa 2004). Pseudo-obstruction raises concern for coronary
of rupture of large aneurysms (Stanson et al. 2001). CT and MR artery disease, since in the study of Miyake et al. (1987), five of the
imaging may show bowel wall thickening with the so-called target seven cases who developed this were found to have coronary artery
sign, mesenteric vessel engorgement and haziness, ascites, or small disease. Similarly, Zulian and his group (2003) detected coronary
bowel obstruction, which may give a beak-like luminal narrowing artery aneurysm in one-half of their 10 cases presenting with surgi-
(Jee et al. 2000). cal abdomen, of whom three were felt to have pseudo-obstruction
Surgery is obligatory in case of perforation, haemorrhage, infarc- as a marker of significant vasculitis. Abnormal liver function tests
tion, pancreatitis, appendicitis, or cholecystitis. Not infrequently seem to be a common theme among studies examining potential
multiple perforations are found. Relapses are frequent, and immu- predictors of IVIg failure (Fukunishi et al. 2000; Han et al. 2000;
nosuppressive therapy may increase the likelihood of infections Durongpisitkul et al. 2003). Tremoulet et al. (2008) reported a rise
and delayed healing. Intensive medical management is necessary. in IVIg-resistant KD in one well-defined clinical population.
Glucocorticoid doses should not be lowered, nor should cyclophos-
phamide administration be postponed in the perioperative period. Granulomatosis with polyangiitis
Intravenous pulse cyclophosphamide is preferable to oral treat-
ment because of its more rapid onset of action, and its reliable (wegener’s granulomatosis)
delivery to the blood. Its administration is recommended intraop- The frequency of GI involvement in granulomatosis with polyangii-
eratively or immediately postoperatively to control the disease at tis (GPA) is uncertain as the few well-documented cases are selec-
the time of surgery (Guillevin 1999). tively biased towards more aggressive cases (Haworth and Pusey
1984; Storesund et al. 1998). In the early autopsy study conducted
Kawasaki’s disease by Walton (1958), necrotizing intestinal arteriolitis was found in
24% of 56 cases. Hoffman et al. (1992) analysed 158 patients and
The GI manifestations of Kawasaki’s disease (KD) have been reported no intestinal manifestations. Haworth and Pusey (1984)
described as early or late (Zulian et  al. 2003). Diagnosis may be found abdominal symptoms in four of 45 patients in their records.
challenging in patients with atypical or incomplete KD (Zulian et al. Müller-Ladner (2001) reported granulomatous gastritis. Storesund
2003). GI tract involvement may include small bowel obstruction et al. (1998) discussed the clinical and pathological presentations of
(Murphy et al. 1987; Mele and Evans 1996), focal colitis (Chung severe intestinal involvement in GPA. They found two females and
et al. 1996), bowel infarction (Mercer and Carpenter 1981), intes- four males, one of whom developed two episodes of bowel manifes-
tinal pseudo-obstruction (Akikusa et al. 2004), and hepatobiliary tations necessitating immediate surgical interventions. The average
disease (Cody et al. 1996). Zulian and his group (2003) described 10 age at onset of intestinal symptoms was 43.3 years (26–55 years)
children (4.6%) among a cohort of 219 KD patients who had severe and the first signs of their manifestations developed within the
abdominal complaints coincident with their disease onset. The first 2 years of illness. Acute pain, peritonitis, or distension was the
most common symptoms at onset were acute abdominal pain and main clinical picture in six of seven events, whereas the seventh
distension, vomiting, hepatomegaly, and jaundice. Haematemesis presented with profuse diarrhoea with blood and mucus. The small
was present in one; toxic shock syndrome requiring intensive care bowel was involved in two, the large intestine in three, and both in
admission occurred in four. Five patients had laparotomy, three two of seven episodes.
had percutaneous transhepatic biliary drainage, and one needed GI Histologically, vasculitis, ischaemia, inflammation, and ulcera-
endoscopy. Postoperative diagnosis was gallbladder hydrops with tion predominated, while granulomas were rarely identified.
cholestasis in five, paralytic ileus in three, appendicular vasculitis in Surgery was performed in six episodes, and perforation was seen
one, and haemorrhagic duodenitis in one. All patients completely four times. The author believed that the manifestations were
CHAPTER 14 vasculitic manifestations in the gastrointestinal tract 185

(a) (b)

Fig. 14.5 Mesenteric ischaemia. (a and b) Contiguous axial CT sections through the umbilical region of the same patient demonstrating irregular mural thickening of
one of the small intestinal loops (arrows) with multiple air fluid levels. It is seen adherent to the anterior abdominal wall (arrowheads).

associated with the disease process rather than related to the pre- granulomas and heavy eosinophilic infiltration in the small and
ceding treatment with immunosuppressive drugs. Pickhardt and medium-sized arteries and veins, whereas eosinophilic gastroen-
Curran (2001) described a case of fulminant enterocolitis that teritis is distinguished from CSS by the lack of vasculitis.
proved to be due to GPA. Their patient was also exceptional in The clinical picture consists of pain, ileus, ischaemic bowel dis-
having severe GI disease without concomitant renal involvement. ease, bleeding ulcers, and bloody diarrhoea. It may be so severe as
Steele and his co-workers (2001) described a young woman who to cause acute abdomen necessitating surgical interference (Kurita
presented initially to hospital with GI symptoms and then devel- et al. 1994; Sharma et al. 1996; Kaneki et al. 1998). The small intes-
oped severe colitis and haemorrhage prior to the occurrence of res- tine is the most common site of involvement (Sharma et al. 1996;
piratory tract symptoms, rapidly progressive renal failure, and nasal Nakamura et  al. 2002). Involvement of the colon has also been
septal perforation. Sokol et al. (1984) noted an unusual presenta- reported (Ohwada et al. 1997; Kim et al. 2000; Suzuki et al. 2005),
tion of GPA disease in a 16-year-old black woman with diarrhoea, and the duodenum can be the site of inflammation detected by
fever, weight loss, abdominal pain, arthralgias, and mouth ulcers, upper endoscopy (Nishie et al. 2003). Suzuki and his group (2005)
suggesting the diagnosis of inflammatory bowel disease. Biopsy reported a case of colon erosion, acalculous cholecystitis, and liver
specimens of rectal mucosa, oro- and nasopharynx, and skin con- abscesses that responded to antibiotic therapy. In the review of lit-
clusively established the diagnosis of GPA. The author emphasized erature in this report, only six cases of cholecystitis and one of liver
the value of taking biopsy specimens of oral lesions. abscess were cited. Nakamura et al. (1991) reported melena caused
The CT findings of GPA of the bowel are not unique to the by ruptured hepatic aneurysm into the intrahepatic bile ducts. The
disease. They share features such as multifocal or diffuse bowel occurrence of pancreatitis, haemorrhage, or perforation is predic-
wall thickening, abnormal wall enhancement, mesenteric vas- tive of poor outlook (Guillevin et al. 1999). Although abdominal
cular engorgement, and ascites with other small-vessel vasculitis complaints rarely appear as initial symptoms, this entity should be
(Figure 14.5). Nonetheless, establishing the diagnosis of progres- considered when the patient with abdominal pain presents with
sive vasculitis through CT is useful in directing therapy (Pickhard eosinophilia, asthma, or allergic rhinitis (Nishie et  al. 2003), or
and Curran 2001). during glucocorticoid withdrawal in asthmatic patients when the
so-called ‘forme frustes’ CSS may be encountered (Churg 2001;
Eosinophilic granulomatosis with Vaglio et al. 2012). GI involvement increases the risk of relapse in
polyangiitis (Churg–Strauss syndrome) CSS (Pavone et al. 2006).

The GI tract is the third most common site of involvement in


Churg–Strauss syndrome (CSS) (~50% of patients), after lung Microscopic polyangiitis
and skin (Kurita et  al. 1994). Pathological findings include mul- GI symptoms of microscopic polyangiitis (MPA) were identified
tiple intestinal ulcerations with or without perforation, ischaemic when this entity was recognized as distinct from PAN. Lhote et al.
change, intestinal obstruction, and ileus (Kurita et al. 1994; Sharma (1996) reported abdominal pain in 32–58% and digestive tract
et al. 1996). Intestinal perforation has been reported as a rare com- bleeding in 29% of cases. In 1999, Guillevin et al. retrospectively
plication of CSS (Sharma et al. 1996). In Japan, however, intestinal analysed 85 MPA patients and reported GI involvement in 30.6%.
perforation is frequently reported, with the small intestine being They believed that MPA is often a generalized disease characterized
the most common site (Shimamoto et al. 1990). Histologically, CSS by multivisceral involvement. In their study, GI tract involvement
can be differentiated from PAN by the presence of extravascular was as frequent as in PAN and CSS. It may be severe, and in two
186 SECTION 3 clinical manifestations common to vasculitis

patients was responsible for death. Guillevin and his co-workers CT features include bowel wall thickening with the target
reported abdominal pain in 23 patients, melena in four, haematem- sign and engorgement of mesenteric vessels with the comb sign
esis in three, bowel perforation in one, cholecystitis in two, appen- (Hokama et al. 2008).
dicitis in one, and pancreatitis in one, and death occurred in two In the study of Bagai and his group (2001), four of their seven
patients. When HBV or HCV infection was present, there was no patients on glucocorticoids had no recurrence, and in two the GI
clinical evidence that the viral infection was responsible for the vas- symptoms were milder. Parenteral glucocorticoids have been advo-
culitis. Patients with elevated transaminase level had no detectable cated for severe abdominal pain, but they should be used with cau-
viral antigens, antibodies to virus, or molecular biological evidence tion if there is active GI bleeding (Chen et al. 2005). Ronkainen
of virus replication. MPA has been cited as a cause of oral lesions et al. (2006), found that early prednisone therapy in IgAV resulted
(Shiboski et al. 2002), massive intestinal bleeding (Ueda et al. 2001), in a decreased abdominal pain score severity and duration. In their
extended colonic ulcerations, and haemorrhage with crypt abscess systematic review, Weiss et al. (2007) showed that CS lowered the
formation (Tsai et al. 2004), cholecystitis (Juliano et al. 2009), as odds of recurrent abdominal pain as well as surgical intervention.
well as focal rectal capillaritis manifesting as painless rectal bleed-
ing (Komanduri et al. 2002). Behçet’s syndrome
The frequency of GI tract involvement in Behçet’s syndrome (BS)
IgA Vasculitis (Henoch–Schönlein purpura) varies in different countries: Turkey 2.8–5%, Iran 4%, Israel Arabs
GI involvement occurs in approximately two-thirds of children 0%, Egypt 20%, Kuwait 21%, Scotland 50%, and Japan 50–60%
with IgA vasculitis (IgAV) (Katz et  al. 1991; Bagai 2001; Tizard (Yazici et  al. 1980; Xavier et  al. 2012; Jankowski et  al. 1992; El
1999). It usually is evident as abdominal pain. Abdominal symp- Menyawi et al. 2009; Jaber et al. 2002; Mousa et al. 1986; Shimizu
toms precede the typical rash in approximately 36% of cases et al. 1979; Oshima et al. 1963). Symptoms of GI involvement of BS
(Lanzkowsky 1992; Choong and Beasley 1998). Other presenting are mainly dysphagia, abdominal pain, nausea, vomiting, flatulence,
symptoms include nausea, vomiting, and diarrhoea, which may and diarrhoea, sometimes with blood (see Chapter 35). Oesophageal
be accompanied by melena that is believed to be associated with involvement is reported to range from 2 to 11% of cases, although
increased risk of renal disease (Tizard 1999). IgAV is believed to this may be an underestimate (Bayraktar et al. 2000). Oesophageal
be triggered by a respiratory tract infection; however, there are lesions include ulcers (linear, oval, or round), usually in the mid-
reports that incriminate GI tract pathogens in that role. The list dle third of the oesophagus (Ozenc et al. 1990; Yashiro et al. 1986),
includes Shigella (Roza et  al. 1983), Yersinia (Rasmussen 1982), fistulae (Asaoka et  al. 1990); pseudomembranous oesophagitis
Campylobacter (Lind et al. 1994), and Clostridium difficile enteri- (Bottomley et al. 1992); or varices related to portosystemic anasto-
tis (Boey et al. 1997). Chen et al. (2005) described characteristic moses (Bayraktar et al. 1995). The gastric mucosa appears to be the
endoscopic findings in a patient with HSP. These include discrete least affected part of the GI tract; the most frequent gastric lesions
coin-like petechiae, haemorrhagic erosion, and skip hyperaemic are aphthous ulcers (Kasahara et al. 1981; Lakhanpal et al. 1985). In
and ecchymotic lesions, seen in the gastric antrum, caecum, ile- the duodenum, aphthous ulcers have been described both clinically
ocaecal valve, and sigmoid colon. (Ozenc et  al. 1990)  and in post-mortem studies (Kasahara et  al.
In their review of intra-abdominal manifestations of IgAV, 1981). Resistance to medical treatment is remarkable, and perfo-
Choong and Beasly (1998) found that major complications of ration may complicate these ulcers (Testini et al. 1996). Although
abdominal involvement develop in 4.6% (range 1.3–13.6%), cit- gastric outlet obstruction can occur, there is not usually a deformity
ing intussusception as by far the most common complication. It is of the duodenum (Ozenc et al. 1990). Intestinal BS occurs in two
confined to the small bowel in 58% of cases. Bowel ischaemia and forms: mucosa inflammation and ischaemia. Small-vessel disease
infarction, intestinal perforation, fistula formation, late ileal stric- underlies ulceration, whereas large-vessel involvement of a mesen-
ture, acute appendicitis, massive upper GI bleeding, pancreatitis, teric artery or its major branches leads to ischaemia and infarction
hydrops of gallbladder, and pseudomembranous colitis are infre- (Bayraktar et al. 2000). The small intestine is the most frequently
quent. Involvement of the ileum or ascending colon can mimic involved extraoral part of the GI tract in BS (Kasahara et al. 1981).
acute appendicitis and leads to unnecessary appendectomy (Katz The lesions are most commonly found in the terminal ileum and
et al. 1991). the caecum, and less frequently in the colon. The lesions are com-
Frank appendicitis due to vasculitis was also reported (Kim monly aphthous ulcers, or they may be deep round or oval ulcers
et  al. 2005). The descending duodenum and the terminal ileum with a punched-out appearance, whereas the longitudinal ulcers
are frequently involved, with endoscopic characteristics of diffuse are rare. Lee et al. (1997) reported 37 persons with intestinal BS, of
mucosal lesions:  petechiae, haemorrhagic erosions, and ulcers whom 26 were surgically managed. A solitary ulcer was observed
(Esaki et al. 2002). in 22 cases (60%) while multiple ulcers were present in 15 cases.
Factor XIII activity has been reported to be low in HSP, espe- Chang et al. (2000) described a case of ileocaecal ulcer and ceco-
cially with severe GI disease (De Mattia et al. 1995). Ultrasound caecal fistula. Bowel perforation is unusual but serious (Kaklamani
may reveal generalized thickening of intestinal wall, ascites, intus- et al. 1998; Ghate and Jorizzo 1999).
susception (Chen et al. 2005), distended appendix surrounded by Rectal and anal involvement are quite rare, and may share fea-
hyperechoic inflamed fat (Kim 2004), or features consistent with tures with ulcerative colitis, both clinically and histopathologically
acalculous cholecystitis (Hoffmann et al. 2004). Ultrasonography (Hamza 1993) (Figure 14.6). Vasculitis of small veins and venules is
complements serial clinical assessment, clarifies the nature of common, characterized by the presence of crypt abscesses, submu-
involvement, and reduces the risks of unnecessary surgery (Choong cosal fibrosis, and an inflammatory reaction with an intact mucosa
and Beasley 1998). was described (Sayek et al. 1991).
CHAPTER 14 vasculitic manifestations in the gastrointestinal tract 187

(a) (b) (c)

Fig. 14.6 Behçet’s syndrome. Portions of colonic mucosa showing focal crypt distortion, surface mucosal erosions (a), and necrotic debris heavily infiltrated
by neutrophils (b). The mucosal tissue shows focal crypt distortion and moderate infiltration of lamina propria by neutrophils and mononuclear cells (c).

The differential diagnosis of intestinal BS includes Crohn’s dis- as well as arterial occlusions, stenosis, and arteriovenous fistula
ease, ulcerative colitis, caecal tuberculosis, caecal amoebiasis, have been described (Bayraktar et al. 1993). Intestinal amyloidosis
benign tumours, and drug-induced damage. It is often difficult to may be responsible for diarrhoea and malabsorption. The amyloid
distinguish between BS and inflammatory bowel disease, as they present is type AA. It tends to occur about a decade after the disease
share common extraintestinal features: oral ulcers, uveitis, erythema onset (Melikoglu et al. 2001).
nodosum, and arthritis (Sakane 1999). BS can be distinguished Upper GI tract radiological examination is helpful in showing
from Crohn’s disease by having ulcers that, characteristically, do deep ulcers, intestinal stenoses, or fistulas (Asaoka et  al. 1990).
not show a granuloma. Ulcers in BS are deep and associated with a Pyloric stenosis in the absence of duodenal deformity is character-
vasculitis that is usually a venulitis. Fistula formation and perfora- istic. The colonic lesions of BS can be seen using double-contrast
tion tend to occur early in the course of BS. The colonic ulcers of BS barium studies. Colonic haustrations are well preserved in BS and
are deeper than those of ulcerative colitis. ulcers can be detected (Bayraktar et al. 2000).
The ileocaecal region is the most frequently involved location; The EULAR recommendations for the management of gastroin-
while in ulcerative colitis the disease usually starts at the rectum testinal BS include sulfasalazine, corticosteroids, azathioprine, tha-
and moves proximally (Bayraktar et al. 2000). Acute pancreatitis lidomide, TNF-α blockers, and surgery (Hatemi et al. 2008). It is
was reported by Le Thi Huong et al. (1992), and Lakhanpal et al. noteworthy that some cases of intestinal BS are considered among
(1985) described pathological features of pancreatitis in five cases the serious manifestations (Sfikakis et al. 2007). In a review of the
of a large Japanese autopsy registry (170 cases). Biochemical evi- data on 369 patients who received TNF-α inhibitors for GI dis-
dence of pancreatitis is a relatively common finding in BS, even in ease, robust clinical response was noted in 100% of patients (Arida
the absence of other diagnostic clues (Bayraktar et al. 2000). et al. 2011).
The most common hepatic complication of BS is Budd–Chiari Surgery is considered for perforation and persistent bleeding.
syndrome (BCS) (Bayraktar et al. 1993; Bismuth et al. 1990). BS Invasive surgical procedures often result in excessive inflammatory
accounts for 42.4% of the cases of BCS with recognizable underly- cell infiltration into the treated tissues, with subsequent anasto-
ing disease in Turkey (Bayraktar et al. 1993). Goubran et al. (1999) motic leakage. Intermediate doses of glucocorticoids are given to
reported 30 cases of BCS; in 25 an aetiological factor could be iden- the patients for several days postoperatively (Sakane et al. 1999).
tified. Four patients (16%) had BS (one with a concomitant pro- The creation of a stoma is preferred over primary anastomosis
tein C deficiency). BCS presents with hepatomegaly, leg oedema, because of the high rate of intestinal leak, perforation, and fistula-
ascites, and venous dilatation over the trunk. With thrombosis tion at anastomotic sites (Bayraktar et al. 2000). Lee et al. (1997),
involving a long segment of inferior vena cava (IVC), the outcome commenting on the high recurrence rate (46.1%), strongly recom-
is much worse than when it involves the hepatic portion of IVC. mended periodic follow-up with radiography and endoscopy. They
Other hepatobiliary conditions reported in BS include:  hepato- advised examination of the entire bowel at the time of operation
megaly due to steatosis or congestion; cirrhosis; acute and chronic and that bowel resection should include a generous normal margin.
hepatitis; cholelisthiasis; acute cholecystitis; toxic hepatitis; hepatic
abscess (Lakhanpal et  al. 1985; Manna et  al. 1985); and primary Thromboangiitis obliterans
biliary cirrhosis (Jankowski et al. 1992). Aphthous lesions on the
hepatic capsule (Barrier et al. 1982) and a case with resemblance (buerger’s disease)
to small-duct primary sclerosing cholangitis (Hisaoka 1994) were Although some authors claim that GI manifestations in throm-
reported. Splenomegaly is noted in 20% of patients even in the boangiitis obliterans (TAO), especially early in the disease, are
absence of portal hypertension (Soysal et al. 1990). Lakhanpal et al. misdiagnosed (Adem et al. 2002), there is some controversy about
(1985) discovered 37 cases of splenic involvement in a total of 170 whether TAO causes GI problems (Hassoun et al. 2001). Intestinal
studied at autopsy. They reported splenitis, congestion, spleno- TAO occurs mostly in men, but there are reports of it affecting
megaly, haemosiderosis, infarction, and autosplenectomy in order women (Raat et  al. 1993; Lie 1998). Visceral involvement may
of descending frequency. Peritonitis may be encountered in BS. appear simultaneously with limb ischaemia (Cho et  al. 2005)  or
Saccular and fusiform arterial aneurysms of medium-sized arteries may precede it (Broide et  al. 1993; Adem et  al. 2002), which
188 SECTION 3 clinical manifestations common to vasculitis

(a) (b) (c)

Fig. 14.7 A case of TAO, surgically resected intestinal loop showing: (a) Artery with a cellular thrombus with early organization (star). The arterial wall is highly inflamed
as well as the perivascular tissues involving vein (V) and nerve (N) (H&E ×200). (b) Inflammation of the wall of small vessels (arrows) (H&E ×200). (c) Medium-sized
arteriole with organizing cellular thrombus (T), non-disrupted elastic lamina (arrows), and mild inflammation of the arterial wall with no evidence of fibrinoid necrosis
(HE ×200). Courtesy of Dr Soheir Mahfouz.

suggests that TAO should be considered in young male smokers vascular lesions were progressive and rendered the patients vul-
with unexplained abdominal pain. Multiple sites are often involved, nerable to infarction during periods of decreased cardiac output.
and there is a tendency to recur (Pfitzmann et al. 2002; Siddiqui Okuda et al. (1990) reported two patients with RA complicated by
et al. 2001), or have occlusion of vascular grafts (Schellong et al. severe attacks of enterocolitis, presumably due to mesenteric vas-
1994). The SMA, followed by the celiac trunk, is the artery most culitis (Figure 14.8).
frequently involved (Ito et  al. 1993; Pfitzmann et  al. 2002; Raat
et al. 1993; Schellong et al. 1994; Hassoun et al. 2001), although Systemic lupus erythematosus
the IMA (Nunez Garcia et al. 1998) and hepatic artery (Adem et al. Lupus enteritis is a serious presentation of systemic lupus erythe-
2002) can also be affected. Attention to the possibility and prompt matosus (SLE), with a prevalence rate ranging between 0.2 and 5.3%
intervention is required to avoid this potentially life-threatening (Medina et al. 1997; Drenkard et al. 1997). Lee et al. (2002) retro-
complication of visceral TAO (Deitch et  al. 1981; Siddiqui et  al. spectively studied 175 SLE patients, 38 (22%) of whom had at least
2001) (Figure 14.7). one episode of acute abdominal pain. Lupus enteritis, based on CT
findings, was found in 17 patients, comprising 45% of those patients
with acute abdominal pain. Histological findings may include
Vasculitis associated with inflammatory infiltrates or hyalinization of capillary walls with a
connective tissue disease wire-loop appearance reminiscent of lupus nephritis (Figure 14.9).
Vasculitis of rheumatoid arthritis The spectrum of lupus enteritis included pain and haemorrhage
Rheumatoid vasculitis is a relatively uncommon complication of
rheumatoid arthritis (RA). Pagnoux et  al. (2005) reported three
RA-associated vasculitis patients among their series of patients
(5%). Rheumatoid vasculitis has been reported to cause: multiple
small bowel ulceration (Takeuchi and Kuroda 2000); pancolitis
and discrete colonic ulcerations following glucocoticoid treat-
ment (Burt et  al. 1983); appendicitis (van Laar et  al. 1998); and
widespread microaneurysms of celiac and extraintestinal small
and medium-sized vessels (Hitter et al. 1988). Achkar et al. (1995)
described a patient with serious intra-abdominal haemorrhage
from a ruptured aneurysm of the inferior pancreaticoduodenal
artery in the setting of rheumatoid vasculitis. Jacobsen et al. (1985)
reported a case of mesenteric arteritis complicated by perforation
of the ileum with generalized peritonitis. Perforation due to intes-
tinal infarction was also reported in the small intestine (Tsai 1980),
and in a combined ileal and sigmoid infarction (Babian et al. 1998).
Another complication of rheumatoid vasculitis is stricture forma-
tion in the small bowel (Kuehne et al. 1992), or the colon (Keating
et al. 1998). Marcolongo et al. (1979) studied gastric, colonic, and
rectal biopsies obtained from patients with RA. Histological changes
were characterized by partial or complete loss of superficial epithe-
lium, inflammatory cellular infiltrate, and the presence of vasculitic
lesions. McCurley and Collins (1984) described three patients with
complicated RA and bowel infarction in which the distal mesen- Fig. 14.8 Acute intestinal obstruction. Axial CT scan showing dilated jejunal
teric vessels were occluded by endarteritis characterized by intimal bowel loops with multiple fluid levels. The patient had emergency surgical
proliferation without vessel wall necrosis or inflammation. These resection and 2 years later developed RA.
CHAPTER 14 vasculitic manifestations in the gastrointestinal tract 189

(a) (b)

Fig. 14.9 Endoscopically obtained section of ileum in a case of systemic lupus erythematosus (H & E scanning), showing infiltration by inflammatory cells. Note
hyalinization of the capillary wall with an appearance reminiscent of the ‘wire-loop’ in lupus nephritis (white arrow). The patient was diagnosed with lupus enteritis. (a) ×
400; (b) × 800.

with gastritis, diarrhoea, haemorrhage, perforation, infarction, and Plain radiography may show intraperitoneal free air, pneumatosis
ileus with ileitis. Colitis presents with similar changes as well as cystoide intestinalis, ileus, or a pseudo-obstruction pattern. Barium
intussusception leading to pain and obstruction (Hoffman and Katz enema may show thumbprinting due to submucosal oedema or
1980). Other less frequent manifestations include:  oesophageal haemorrhage, which is fairly specific for ischaemic bowel (Sultan
dysmotility and arteritis (Gastrucci et al. 1990; Harvey et al. 1954); et al. 1999). With insidious onset of symptoms, evaluation should
protein-losing enteropathy (Pelletier et al. 1992); oedematous villi incorporate ultrasound and abdominal CT. Abdominal ultrasound
and lymphangiectasia (Tian et al. 2010); massive ischaemic colitis may show bowel wall thickening (Shiohira et al. 1993). Abdominal
(Kistin et al. 1978); and perforated colonic diverticulae (Zizic et al. CT may show changes diagnostic of ischaemic bowel disease as
1997). The rectum can be involved in gangrenous ischaemic coli- noted in Section Imaging, including bowel wall thickening, target
tis (Reissman et al. 1994); stenosis (Palvio and Christensen 1987); sign, dilation of intestinal segments, prominent engorgement of
ulceration (Amit et al. 1999); and perforation with necrosis of the mesenteric vessel with a palisade pattern ‘the comb sign’ (Lee et al.
rectosigmoid (Lazaris et al. 2003), although rectal involvement is 2010), and increased attenuation of mesenteric fat. Angiography is
rare (14% of cases) owing to its rich blood supply (Lee et al. 2002). not useful as the disease affects the small vessels. Endoscopy may be
Lee and his group (2002) found lupus enteritis to be the most com- helpful and radioisotope scanning using gallium and indium white
mon cause of acute abdominal pain. It was the initial manifestation cell scanning has been used to highlight areas of inflammation and
of SLE in six of 17 cases (35.3%). The jejunum and ileum were the sepsis (Sultan et al. 1999). Laparoscopy or laparotomy may be con-
sites most commonly affected, being involved in 80% and 85% of sidered (Al-Hakeem 1998).
cases, respectively; 90% of cases had involvement in multiple vas- Medical treatment was successfully reported with high-dose
cular territories, and four cases relapsed. prednisolone in intestinal vasculitis (Medina et al. 1997; Cabrera
There is disagreement in the literature regarding the associa- et  al. 1994; Turner et  al. 1996)  and intravenous cyclophospha-
tion between intestinal infarction and presence of antiphospho- mide (Grimbacher et  al. 1998; Turner et  al. 1996). Small intes-
lipid antibodies (Medina et al. 1997; Sanchez-Guerrero et al. 1992; tinal ischaemia, infarction, and perforation of any segment
Lee et al. 2002) and its relationship to lupus activity (Medina et al. require emergency surgery. Large bowel ischaemia may be treated
1997; Lee et al. 2002). Leukopenia is frequently reported to cor- conservatively, and laparoscopy may be considered, as it is less
relate with the occurrence of lupus enteritis (Lee et al. 2002; Sultan invasive, and less consequential when compared to open surgery
et al. 1999). Microscopically, there are no pathognomonic findings (Medina et al. 1997).
suggestive of SLE; the appearance varies from segmental oedema
to discrete ulceration, gangrene, and perforation (Sasamura et al. Dermatomyositis
1991; Grimbacher et  al. 1998). Histologically, both small-vessel Dermatomyositis was reported to cause vasculitis involving small
arteritis and venulitis have been described. Associated findings arteries and capillaries, resulting in ischaemia and necrosis in any
include degeneration of the media of small arteries, fibrinoid part of the GI tract with serious consequences (Eshraghi et al. 1998).
necrosis of vessel walls, old thrombosis, phlebitis, and monocyte
infiltrate in the lamina propria. Acute and chronic inflammatory
infiltrate and punched out ulcers with oedematous mucosa can be Miscellaneous forms of vasculitis
detected on colonoscopy (Grimbacher et al. 1998; Hokama et al. Lopez et  al. (1980) studied leukocytoclastic (hypersensitivity)
2012). Pneumatosis cystoide intestinalis may also occur (Cabrera vasculitis (LCV) on skin biopsy in 18 patients who were younger
et al. 1994) although it is a non-specific finding. This may result than 16 years of age, and 75 older patients with LCV. Significant
in hepatic venous gas with a high mortality rate (Tian et al. 2010). GI manifestations at presentation or exacerbation of vasculitis
190 SECTION 3 clinical manifestations common to vasculitis

Adem, C., Benamouzig, R., Royer, I., et  al. (2002). [Buerger’s disease or
thromboangiitis obliterans revealed by an enteric ischemia. Case report
and literature review]. Gastroentérologie Clinique et Biologique, 26,
409–11.
Akikusa, J.D., Laxer, R.M., and Friedman, J.N. (2004). Intestinal pseudoob-
struction in Kawasaki disease. Pediatrics, 113, e504–6.
Al-Hakeem, M.S. and McMillen, M.A. (1998). Evaluation of abdominal
pain in systemic lupus erythematosus. American Journal of Surgery,
176, 291–4.
Amit, G., Stalnikowicz, R., Ostrovsky, Y., et al. (1999). Rectal ulcers: a rare
gastrointestinal manifestation of systemic lupus erythematosus. Journal
of Clinical Gastroenterology, 29, 200–2.
Angeli, E., Vanzulli, A., Venturini, M., et  al. (2001). The role of radiology
in the diagnosis and management of Takayasu’s arteritis. Journal of
Nephrology, 14, 514–24.
Arida, A., Fragiadaki, K., Giavri, E., et  al. (2011). Anti-TNF agents for
Behçet’s disease: analysis of published data on 369 patients. Seminars in
Arthritis and Rheumatism, 41, 61–70.
Fig. 14.10 Findings of non-specific vasculitis. Surgically obtained section of Arumugam, M., Raes, J., Pelletier, E., et al. (2011). Enterotypes of the human
ileum in a case of leukocytoclastic vasculitis (H & E staining). Small vessels show gut microbiome. Nature, 473(7346), 174–80.
wall infiltration with mononuclear cells and neutrophils (white arrow). × 200. Asaoka, M., Sakai, Y., Kimura, J., et al. (1990). [A case of tracheoesophageal
fistula in Behcet’s disease repaired with pericardial patch and gastric
roll]. Nihon Kyobu Geka Gakkai Zasshi, 38, 1549–53.
Ayers, A.B. and Fitchett, D.H. (1976). Hepatic haematoma in polyarteritis
were more frequent in the younger age group (66%), compared to nodosa. British Journal of Radiology, 49, 184–5.
the older patients (26%). No consistent radiological findings were Babian, M., Nasef, S., and Soloway, G. (1998). Gastrointestinal infarc-
noted. Duodenal and peritoneal biopsies suggested vasculitis in six tion as a manifestation of rheumatoid vasculitis. American Journal of
patients. Four patients needed exploratory laparotomy. Only one Gastroenterology, 93, 119–20.
patient had intestinal infarction, and two patients with acute abdo- Bagai, A., Albert, S., and Shenoi, S.D. (2001). Evaluation and therapeutic out-
men responded promptly to intravenous glucocorticoids. They come of palpable purpura. Indian Journal of Dermatology, Venereology
suggested that early recognition and treatment with glucocorti- and Leprology, 67, 320–3.
coids could avoid surgery. Vlahos et al. (2005) presented a case of Barrier, J., Cerbelaud, P., Raffi, F., et al. (1982). [Aphthous hepatic lesions and
Behçet’s syndrome]. Semaine des Hôpitaux, 58, 2395–6.
localized LCV of the left colon with megacolon. Diffuse mucosal
Bartnicke, B.J. and Balfe, D.M. (1994). CT appearance of intestinal ischemia
thickening of the antrum, duodenum, and jejunum were reported and intramural hemorrhage. Radiologic Clinics of North America, 32,
in a person with LCV (Nagarajan et al. 2005). Histopathological 845–60.
examination of tissue from patients with LCV and GI involvement Bayraktar, Y., Balkanci, F., Kansu, E., et  al. (1993). Budd-Chiari syn-
may reveal non-specific findings of vasculitis (Figure 14.10). drome: analysis of 30 cases. Angiology, 44, 541–51.
Cryoglobulinaemic vasculitis (CV) may involve the oesophagus, Bayraktar, Y., Balkanci, F., Kansu, E., et al. (1995). Cavernous transforma-
stomach, small intestine, or colon, resulting in GI haemorrhage, tion of the portal vein:  a common manifestation of Behcet’s disease.
intestinal ischaemia, acute pancreatitis, or cholecystitis. If there American Journal of Gastroenterology, 90, 1476–9.
is significant involvement of the major vessels, lift-threatening Bayraktar, Y., Ozaslan, E., and Van Thiel, D.H. (2000). Gastrointestinal mani-
festations of Behcet’s disease. Journal of Clinical Gastroenterology, 30,
complications, including perforation may occur. GI involvement
144–54.
is often seen in the context of multiorgan failure (catastrophic
Bismuth, E., Hadengue, A., Hammel, P., et al. (1990). Hepatic vein thrombo-
syndrome) (Saccardo et al. 2012), which has a high mortality rate sis in Behcet’s disease. Hepatology, 11, 969–74.
(Garas et al. 1996). Blidi, M., Quang Tri, N., Cassan, P., et al. (1996). [Acute cholecystitis in peri-
As an example of vasculitis associated with lymphoproliferative arteritis nodosa. 8 cases]. Annales de Médecine Interne (Paris), 147,
disease, LaBerge and Kerlan (2002) reported a case of lymphocytic 304–12.
lymphoma presenting with vasculitis. Their patient died of diffuse Boey, C.C., Ramanujam, T.M., and Looi, L.M. (1997). Clostridium
bowel ischaemia several days after laparotomy. Among this group, difficile-related necrotizing pseudomembranous enteritis in association
enterocolic lymphocytic phlebitis (ELP) is a rare cause of GI tract with Henoch-Schonlein purpura. Journal of Pediatric Gastroenterology
and Nutrition, 24, 426–9.
ischaemia, involving only the mural and mesenteric veins, which
Bongard, O., Schneider, P.A., Krahenbuhl, B., et  al. (1992). Transluminal
are surrounded by a lymphocytic and sometimes granulomatous
angioplasty of the aorta, renal and mesenteric arteries in Takayasu arteri-
infiltrate. The mesenteric arterial system and the systemic vascula- tis: report of two cases. European Journal of Vascular Surgery, 6, 567–71.
ture are characteristically spared (Abraham et al. 2004). Bottomley, W.W., Dakkak, M., Walton, S., et al. (1992). Esophageal involve-
ment in Behcet’s disease. Is endoscopy necessary? Digestive Diseases
References and Sciences, 37, 594–7.
Abraham, S.C., Solem, C.A., Hauser, S.C., et  al. (2004). Chronic antral Bradbury, A.W., Murie, J.A., and Ruckley, C.V. (1993). Role of the leucocyte
ulcer associated with gastroduodenal lymphocytic phlebitis. American in the pathogenesis of vascular disease. British Journal of Surgery, 80,
Journal of Surgical Pathology, 28, 1659–63. 1503–12.
Achkar, A.A., Stanson, A.W., Johnson, C.M., et  al. (1995). Rheumatoid Brandstetter, K., Schroder, W., and Bautz, W. (1995). [Massive intrahe-
vasculitis manifesting as intra-abdominal hemorrhage. Mayo Clin patic hemorrhage as the initial manifestation of panarteritis nodosa].
Proceedings, 70, 565–9. Rontgenpraxis, 48, 209–11.
CHAPTER 14 vasculitic manifestations in the gastrointestinal tract 191

Broide, E., Scapa, E., Peer, A., et al. (1993). Buerger’s disease presenting as Ewald, E.A., Griffin, D., and McCune, W.J. (1987). Correlation of angio-
acute small bowel ischemia. Gastroenterology, 104, 1192–5. graphic abnormalities with disease manifestations and disease severity
Burke, A.P., Sobin, L.H., and Virmani, R. (1995). Localized vasculitis of the in polyarteritis nodosa. Journal of Rheumatology, 14, 952–6.
gastrointestinal tract. American Journal of Surgical Pathology, 19, 338–49. Fang, S.B., Lee, H.C., Huang, F.Y., et al. (2001). Intestinal pseudo-obstruction
Burt, R.W., Berenson, M.M., Samuelson, C.O., et  al. (1983). Rheumatoid followed by major clinical features of Kawasaki disease: report of one
vasculitis of the colon presenting as pancolitis. Digestive Diseases and case. Acta Paediatrica Taiwanica, 42, 111–4.
Sciences, 28, 183–8. Fauci, A.S., Haynes, B., and Katz, P. (1978). The spectrum of vasculitis: clini-
Byun, J.Y., Ha, H.K., Yu, S.Y., et  al. (1999). CT features of systemic lupus cal, pathologic, immunologic and therapeutic considerations. Annals of
erythematosus in patients with acute abdominal pain:  emphasis on Internal Medicine, 89, 660–76.
ischemic bowel disease. Radiology, 211, 203–9. Fernandes, S.R., Samara, A.M., Magalhaes, E.P., et al. (2005). Acute cholecysti-
Cabrera, G.E., Scopelitis, E., Cuellar, M.L., et al. (1994). Pneumatosis cys- tis at initial presentation of ployarteritis nodosa. Clinical Rheumatology,
toides intestinalis in systemic lupus erythematosus with intestinal vas- 24,625–7.
culitis:  treatment with high dose prednisone. Clinical Rheumatology, Fitchett, D.H. and Oakley, C.M. (1975). Perihepatic haematoma in polyarte-
13, 312–6. ritis nodosa. Proceedings of the Royal Society of Medicine, 68, 805–6.
Casey, K.M., Quigley, T.M., Kozarek, R.A., et  al. (1993). Lethal nature of Fontes, B., Moore, F.A., Moore, E.E., et  al. (1994). Gut ischemia induces
ischemic gastropathy. American Journal of Surgery, 165, 646–9. bone marrow failure and increases risk of infection. Journal of Surgical
Castrucci, G., Alimandi, L., Fichera, A., et  al. (1990). [Changes in Research, 57, 505–9.
esophageal motility in patients with systemic lupus erythema- Fortin, P.R., Larson, M.G., Watters, A.K., et  al. (1995). Prognostic factors
tosus:  an esophago-manometric study]. Minerva Dietologica e in systemic necrotizing vasculitis of the polyarteritis nodosa group—a
Gastroenterologica, 36, 3–7. review of 45 cases. Journal of Rheumatology, 22, 78–84.
Chang, H.K., Kim, J.S., and Chung, H.R. (2000). Ileocecal ulcer with a cecocecal Franken, E.A., Jr, Kleiman, M.B., Norins, A.L., et  al. (1979). Intestinal
fistula in Behcet’s disease. Korean Journal of Internal Medicine, 15, 99–101. pseudo-obstruction in mucocutaneous lymph-node syndrome.
Chen, M.J., Wang, T.E., Chang, W.H., et  al. (2005). Endoscopic find- Radiology, 130, 649–51.
ings in a patient with Henoch-Schonlein purpura. World Journal of Frohnert, P.P. and Sheps, S.G. (1967). Long-term follow-up study of periarte-
Gastroenterology, 11, 2354–6. ritis nodosa. American Journal of Medicine, 43, 8–14.
Chervonsky, A.V. (2010). Influence of microbial environment on autoim- Fukunishi, M., Kikkawa, M., Hamana, K., et  al. (2000). Prediction of
munity. Nature Immunology, 11, 28–35. non-responsiveness to intravenous high-dose gamma-globulin ther-
Cho, Y.P., Kang, G.H., Han, M.S., et al. (2005). Mesenteric involvement of apy in patients with Kawasaki disease at onset. Journal of Pediatrics,
acute-stage Buerger’s disease as the initial clinical manifestation: report 137, 172–6.
of a case. Surgery Today, 35, 499–501. Gaburri, P.D., Chebli, J.M., Attalla, A., et al. (2005). Colonic ulcers in pro-
Choong, C.K. and Beasley, S.W. (1998). Intra-abdominal manifestations of pylthiouracil induced vasculitis with secondary antiphospholipid syn-
Henoch-Schonlein purpura. Journal of Paediatrics and Child Health, drome. Postgraduate Medical Journal, 81, 338–40.
34, 405–9. Garas, G. and Morgan, C.A. (1996). Hepatitis C and mixed cryoglobulinae-
Chung, C.J., Rayder, S., Meyers, W., et al. (1996). Kawasaki disease presenting mia: a case of primary gastrointestinal necrotizing vasculitis. Australian
as focal colitis. Pediatric Radiology, 26, 455–7. and New Zealand Journal of Medicine, 26, 110–11.
Cognet, F., Ben Salem, D., Dranssart, M., et al. (2002). Chronic mesenteric Geboes, K., Geboes, K.P., and Maleux, G. (2001). Vascular anatomy of the
ischemia:  imaging and percutaneous treatment. Radiographics, 22, gastrointestinal tract. Best Practice and Research Clinical gastroenterol-
863–79; discussion 79–80. ogy, 15, 1–14.
Cornejo, R., Gatica, H., Segovia, E., et al. (2002). [Intestinal necrosis as clini- Gentile, A.T., Moneta, G.L., Lee, R.W., et al. (1995). Usefulness of fasting and
cal presentation of Takayasu arteritis]. Revista Médica de Chile, 130, postprandial duplex ultrasound examinations for predicting high-grade
1159–64. superior mesenteric artery stenosis. American Journal of Surgery,
De Mattia, D., Penza, R., Giordano, P., et al. (1995). von Willebrand factor 169, 476–9.
and factor XIII in children with Henoch-Schonlein purpura. Pediatric Ghate, J.V. and Jorizzo, J.L. (1999). Behcet’s disease and complex aphthosis.
Nephrology, 9, 603–5. Journal of the American Academy of Dermatology, 40, 1–18; quiz 9–20.
Deitch, E.A. and Sikkema, W.W. (1981). Intestinal manifestation of Buerger’s Gorgun, E. and Ozmen, V. (2002). Acalculous gangrenous cholecystitis in
disease: case report and literature review. American Surgeon, 47, 326–8. a young adult: a gastrointestinal manifestation of polyarteritis nodosa.
Desai, K.A., Cotter, N.M., Doucet, L., et  al. (2004). Pathology case of the Surgical Laparoscopy Endoscopy and Percutaneous Techniques, 12,
month. 39-year-old woman with abdominal pain and weight loss. 359–61.
Takayasu’s arteritis (TA). Journal of the Louisiana State Medical Society, Goubran, H.A., Omar, A.A., El-Maghallawy, A.A., et al. (1999). A look into
156, 230–4; quiz 4. the etiology of 30 adults with Budd-Chiari syndrome. Hepatology,
Drenkard, C., Villa, A.R., Reyes, E., et al. (1997). Vasculitis in systemic lupus 30, 573A.
erythematosus. Lupus, 6, 235–42. Goulding, J., Pitcher, A., Piris, J., et al. (2010). Fevers, headache and abnor-
Durongpisitkul, K., Soongswang, J., Laohaprasitiporn, D., et  al. (2003). mal liver function in a 68-year-old man. Journal of the Royal Society of
Immunoglobulin failure and retreatment in Kawasaki disease. Pediatric Medicine, 103, 295–6.
Cardiology, 24, 145–8. Griffith, G.C. and Vural, I.L. (1951). A correlation of clinical and postmortem
El Menyawi, M.M., Raslan, H.M., and Edrees, A. (2009). Clinical features of findings in seventeen cases. Circulation, 3, 481–91.
Behçet’s disease in Egypt. Rheumatology International, 29, 641–6. Grimbacher, B., Huber, M., von Kempis, J., et al. (1998). Successful treatment
Esaki, M., Matsumoto, T., Nakamura, S., et  al. (2002). GI involvement in of gastrointestinal vasculitis due to systemic lupus erythematosus with
Henoch-Schönlein purpura. Gastrointestinal Endoscopy, 56, 920–3. intravenous pulse cyclophosphamide: a clinical case report and review
Esato, K., Noma, F., Kurata, S., et  al. (1982). Mesenteric infarction in of the literature. British Journal of Rheumatology, 37, 1023–8.
Takayasu’s arteritis treated by thromboendarterectomy and intestinal Guillevin, L. (1999). Treatment of classic polyarteritis nodosa in 1999.
resection. Japanese Journal of Surgery, 12, 130–4. Nephrology, Dialysis Transplantation, 14, 2077–9.
Eshraghi, N., Farahmand, M., Maerz, L.L., et al. (1998). Adult-onset dermat- Guillevin, L., Lhote, F., Gallais, V., et al. (1995). Gastrointestinal tract involve-
omyositis with severe gastrointestinal manifestations: case report and ment in polyarteritis nodosa and Churg-Strauss syndrome. Annales de
review of the literature. Surgery, 123, 356–8. Médecine Interne (Paris), 146, 260–7.
192 SECTION 3 clinical manifestations common to vasculitis

Ha, H.K., Lee, S.H., Rha, S.E., et al. (2000). Radiologic features of vasculitis Hoshino, K., Shibata, D., Miyagi, T., et al. (2011). Cytomegalovirus-associated
involving the gastrointestinal tract. Radiographics, 20, 779–94. gastric ulcers in a patientwithdermatomyositis treated with steroid and
Haglund, U. and Bergqvist, D. (1999). Intestinal ischemia—the basics. cyclophosphamide pulse therapy. Endoscopy, 43 (Suppl. 2), E277–8.
Langenbecks Archives of Surgery, 384, 233–8. Ito, M., Nihei, Z., Ichikawa, W., et al. (1993). Intestinal ischemia resulting
Hall, S., Barr, W., Lie, J.T., et al. (1985). Takayasu arteritis. A study of 32 North from Buerger’s disease: report of a case. Surgery Today, 23, 988–92.
American patients. Medicine (Baltimore), 64, 89–99. Ito, M., Sano, K., Inaba, H., et  al. (1991). Localized necrotizing arteritis.
Hamza, M. (1993). [Proctitis in Behçet disease. Study of 6 cases]. Revue du A report of two cases involving the gallbladder and pancreas. Archives
Rhumatisme (ed. Française), 60, 925–7. of Pathology and Laboratory Medicine, 115, 780–3.
Han, R.K., Silverman, E.D., Newman, A., et al. (2000). Management and out- Jaber, L., Milo, G., Halpern, G.J., et al. (2002). Prevalence of Behçet’s disease
come of persistent or recurrent fever after initial intravenous gamma in an Arab community in Israel. Annals of the Rheumatic Diseases,
globulin therapy in acute Kawasaki disease. Archives of Pediatrics and 61, 365–6.
Adolescent Medicine, 154, 694–9. Jacobsen, S.E., Petersen, P., and Jensen, P. (1985). Acute abdomen in rheu-
Hands, L. and Murie, J.A. (1991). Takayasu’s arteritis: a rare cause of acute matoid arthritis due to mesenteric arteritis. A case report and review.
abdomen in a Caucasian male. European Journal of Vascular Surgery, Danish Medical Bulletin, 32, 191–3.
5, 217–9. Jamieson, W.G., DeRose, G., Harris, K.A., et al. (1993). Myocardial and cir-
Harvey, A.M., Shulman, L.E., Tumulty, P.A., et al. (1954). Systemic lupus ery- culatory performance during the ischemic phase of superior mesenteric
thematosus: review of the literature and clinical analysis of 138 cases. artery occlusion. Canadian Journal of Surgery, 36, 435–9.
Medicine (Baltimore), 33, 291–437. Jankowski, J., Crombie, I., and Jankowski, R. (1992). Behçet’s syndrome in
Harward, T.R., Brooks, D.L., Flynn, T.C., et al. (1993). Multiple organ dys- Scotland. Postgraduate Medical Journal, 68, 566–70.
function after mesenteric artery revascularization. Journal of Vascular Jee, K.N., Ha, H.K., Lee, I.J., et al. (2000). Radiologic findings of abdomi-
Surgery, 18, 459–67; discussion 67–9. nal polyarteritis nodosa. AJR American Journal of Roentgenology, 174,
Hassoun, Z., Lacrosse, M., and De Ronde, T. (2001). Intestinal involvement 1675–9.
in Buerger’s disease. Journal of Clinical Gastroenterology, 32, 85–9. Juliano, J., Wilson, K.D., and Gertner, E. (2009). Vasculitis of the gall-
Hatemi, G., Silman, A., Bang, D., et al. (2009). Management of Behçet dis- bladder:  case report and spectrum of disease. Journal of Clinical
ease:  a systematic literature review for the European League Against Rheumatology, 15, 75–7.
Rheumatism evidence-based recommendations for the management of Kaklamani, V.G., Vaiopoulos, G., and Kaklamanis, P.G. (1998). Behçet’s
Behçet disease. Annals of the Rheumatic Diseases, 68, 1528–34. Disease. Seminars in Arthritis and Rheumatism, 27, 197–217.
Hattori, M. and Taylor, T.D. (2009). The human intestinal microbiome: a new Kaneki, T., Kawashima, A., Hayano, T., et  al. (1998). Churg-Strauss syn-
frontier of human biology. DNA Research, 16, 1–12. drome (allergic granulomatous angitis) presenting with ileus caused by
Haworth, S.J. and Pusey, C.D. (1984). Severe intestinal involvement in ischemic ileal ulcer. Journal ofGastroenterology, 33, 112–6.
Wegener’s granulomatosis. Gut, 25, 1296–300. Kasahara, Y., Tanaka, S., Nishino, M., et al. (1981). Intestinal involvement in
Hekali, P., Kajander, H., Pajari, R., et al. (1991). Diagnostic significance of Behçet’s disease: review of 136 surgical cases in the Japanese literature.
angiographically observed visceral aneurysms with regard to polyarteri- Diseases of the Colon and Rectum, 24, 103–6.
tis nodosa. Acta Radiologica, 32, 143–8. Katz, S., Borst, M., Seekri, I., et  al. (1991). Surgical evaluation of
Heneghan, M.A., Feeley, K.M., DeFaoite, N., et al. (1998). Granulomatous Henoch-Schonlein purpura. Experience with 110 children. Archives of
liver disease and giant-cell arteritis. Digestive Diseases and Sciences, 43, Surgery, 126, 849–53; discussion 53–4.
2164–7. Keating, J.P., King, B.R., Kenwright, D.N., et al. (1998). Vasculitis-induced
Hisaoka, M., Haratake, J., and Nakamura, T. (1994). Small bile duct abnor- colonic strictures:  report of two cases. Diseases of the Colon and
malities and chronic intrahepatic cholestasis in Behçet’s syndrome. Rectum, 41, 1316–21.
Hepatogastroenterology, 41, 267–70. Kim, S.J., Chung, I.K., Chung, I.S., et al. (2000). The clinical significance of
Hitter, E., Williame, L., Chappel, R., et al. (1988). Abdominal microaneu- upper gastrointestinal endoscopy in gastrointestinal vasculitis related to
rysms in rheumatoid arthritis. British Journal of Rheumatology, 27, scrub typhus. Endoscopy, 32, 950–5.
239–40. Kistin, M.G., Kaplan, M.M., and Harrington, J.T. (1978). Diffuse ischemic
Hoffman, B.I. and Katz, W.A. (1980). The gastrointestinal manifestations of colitis associated with systemic lupus erythematosus—response to sub-
systemic lupus erythematosus:  a review of the literature. Seminars in total colectomy. Gastroenterology, 75, 1147–51.
Arthritis and Rheumatism, 9, 237–47. Klein, H.M., Lensing, R., Klosterhalfen, B., et al. (1995). Diagnostic imaging
Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., et al. (1992). Wegener granulomato- of mesenteric infarction. Radiology, 197, 79–82.
sis: an analysis of 158 patients. Ann Intern Med, 116, 488–98. Komanduri, S., Jakate, S., and Keshavarzian, A. (2002). Focal rectal capil-
Hoffmann, J.C., Cremer, P., Preiss, J.C., et al. (2004). Gallbladder involvement laritis: microscopic polyangiitis presenting as painless rectal bleeding.
of Henoch-Schonlein purpura mimicking acute acalculous cholecystitis. Journal of Clinical Gastroenterology, 35, 157–9.
Digestion, 70, 45–8. Koyama, M., Tanaka, M., Shimizu, M., et al. (1995). Surgical treatment of
Hokama, A., Kishimoto, K., Ihama, Y., et al. (2012). Endoscopic and radio- mesenteric infarction, thoracoabdominal aortic aneurysm, and proper
graphic features of gastrointestinal involvement in vasculitis. World hepatic aneurysm in a middle-aged woman with Takayasu’s arteritis.
Journal of Gastrointestinal Endoscopy, 4, 50–6. Journal of Cardiovascular Surgery (Torino), 36, 337–41.
Hokama, A., Shimoji, K., Nakamura, M., et al. (2008). An uncommon cause Krant, J.D. and Ross, J.M. (1992). Extracranial giant cell arteritis restricted to
of haematochezia in an adult with skin rash. Gut, 57, 1430–47. the small bowel. Arthritis and Rheumatism, 35, 603–4.
Hokama, A., Taira, K., Yamamoto, Y., et al. (2010). Cytomegalovirus gastritis. Kuehne, S.E., Gauvin, G.P., and Shortsleeve, M.J. (1992). Small bowel stric-
World Journal of Gastrointestinal Endoscopy, 2, 379–380. ture caused by rheumatoid vasculitis. Radiology, 184, 215–6.
Hokama, A., Tanaka, K., Nakamoto, M., et al. (2005). An unusual cause of Kumar, B., Krishnani, N., and Misra, R. (2003). Isolated necrotizing vasculi-
rectal bleeding in a patient with rheumatoid arthritis. Gut, 54, 1061–71. tis of gallbladder: a report of two cases and review of literature. Indian
Horton, K.M., Corl, F.M., and Fishman, E.K. (1999). CT of nonneoplastic Journal of Pathology and Microbiology, 46, 429–31.
diseases of the small bowel: spectrum of disease. Journal of Computer Kurita, M., Niwa, Y., Hamada, E., et  al. (1994). Churg-Strauss syndrome
Assisted Tomography, 23, 417–28. (allergic granulomatous angiitis) with multiple perforating ulcers of the
Horton, K.M., Corl, F.M., and Fishman, E.K. (2000). CT evaluation of the small intestine, multiple ulcers of the colon, and mononeuritis multi-
colon: inflammatory disease. Radiographics, 20, 399–418. plex. Journal of Gastroenterology, 29, 208–13.
CHAPTER 14 vasculitic manifestations in the gastrointestinal tract 193

Kuroda, T., Shiohara, E., Homma, T., et al. (1994). Effects of leukocyte and mesenteric ischemia. Journal of Magnetic Resonance Imaging,
platelet depletion on ischemia--reperfusion injury to dog pancreas. 7, 171–6.
Gastroenterology, 107, 1125–34. Medina, F., Ayala, A., Jara, L.J., et  al. (1997). Acute abdomen in systemic
LaBerge, J.M. and Kerlan, R.K. Jr (2002). SCVIR 2002 Film Panel case 1: vas- lupus erythematosus:  the importance of early laparotomy. American
culitis associated with lymphoproliferative disease. Journal of Vascular Journal of Medicine, 103, 100–5.
and Interventional Radiology, 13, 529–32. Mehta, R., Deepak, John, A., et al. (2004). Takayasu arteritis presenting as
Lakhanpal, S., Tani, K., Lie, J.T., et al. (1985). Pathologic features of Behcet’s chronic mesenteric ischemia. Indian Journal of Gastroenterology,
syndrome: a review of Japanese autopsy registry data. Human Pathology, 23, 73–4.
16, 790–5. Mele, T. and Evans, M. (1996). Intestinal obstruction as a complication of
Landow, L. and Andersen, L.W. (1994). Splanchnic ischaemia and its role Kawasaki disease. Journal of Pediatric Surgery, 31, 985–6.
in multiple organ failure. Acta Anaesthesiologica Scandinavica, 38, Melikoglu, M., Altiparmak, M.R., Fresko, I., et al. (2001). A reappraisal of
626–39. amyloidosis in Behcet’s syndrome. Rheumatology (Oxford), 40, 212–5.
Lanzkowsky, S., Lanzkowsky, L., and Lanzkowsky, P. (1992). Mercer, S. and Carpenter, B. (1981). Surgical complications of Kawasaki dis-
Henoch-Schoenlein purpura. Pediatrics in Review, 13, 130–7. ease. Journal of Pediatric Surgery, 16, 444–8.
Lavelle, E.C., Murphy, C., O’Neill, L.A., and Creagh, E.M. (2010). The role of Miyake, T., Kawamori, J., Yoshida, T., et  al. (1987). Small bowel
TLRs, NLRs, and RLRs in mucosal innate immunity and homeostasis. pseudo-obstruction in Kawasaki disease. Pediatric Radiology, 17, 383–6.
Mucosal Immunology, 3, 17–28. Moneta, G.L., Lee, R.W., Yeager, R.A., et al. (1993). Mesenteric duplex scan-
Lazaris, A., Papanikolaou, I.S., Theodoropoulos, G.E., et al. (2003). Ischaemic ning: a blinded prospective study. Journal of Vascular Surgery, 17, 79–
necrosis of the rectum and sigmoid colon complicating systemic lupus 84; discussion 5–6.
erythematosus. Acta Gastro-Enterologica Belgica, 66, 191–4. Mousa, A.R., Marafie, A.A., Rifai, K.M., et  al. (1986). Behçet’s disease in
Lederberg, J. (2000). Infectious history. Science, 288, 287–93. Kuwait, Arabia. A report of 29 cases and a review. Scandinavian Journal
Lederberg, J. and McCray, A.T. (2001). ‘Ome sweet ‘omics—A genealogical of Rheumatology, 15, 310–32.
treasury of words. Scientist, 15, 8–9. Mowrey, F.H. and Lundberg, E.A. (1954). The clinical manifestations of
Lee, C., Lee, T., Lee, S-H., et al. (2010). GI vasculitis associated with systemic essential polyanglitis (periarteritis nodosa), with emphasis on the
lupus erythematosus. Gastrointestinal Endoscopy, 72, 618–9. hepatic manifestations. Annals of Internal Medicine, 40, 1145–64.
Lee, C.K., Ahn, M.S., Lee, E.Y., et al. (2002). Acute abdominal pain in sys- Muldoon, J., O’Riordan, K., Rao, S., et al. (1996). Ischemic colitis secondary
temic lupus erythematosus:  focus on lupus enteritis (gastrointestinal to venous thrombosis. A rare presentation of cytomegalovirus vasculitis
vasculitis). Annals of the Rheumatic Diseases, 61, 547–50. following renal transplantation. Transplantation, 61, 1651–3.
Lee, K.S., Kim, S.J., Lee, B.C., et al. (1997). Surgical treatment of intestinal Müller-Ladner, U. (2001). Vasculitides in the gastrointestinal tract. Best
Behçet’s disease. Yonsei Medical Journal, 38, 455–60. Practice and Research Clinical Gastroenterology, 15, 59–82.
Lee, S., Childerhouse, A., Moss, K. (2011). Gastrointestinal symptoms and Murphy, D.J., Jr, Morrow, W.R., Harberg, F.J., et  al. (1987). Small bowel
granulomatous vasculitis involving the liver in giant cell arteritis: a case obstruction as a complication of Kawasaki disease. Clinical Pediatrics
report and review of the literature. Rheumatology, 50, 2316–7. (Phila), 26, 193–6.
Le Thi Huong, D., Wechsler, B., Dell’Isola, B., et al. (1992). Acute pancreatitis Musemeche, C.A., Kosloske, A.M., Bartow, S.A., et al. (1986). Comparative
in Behcet’s disease. Digestive Diseases and Sciences, 37, 1452–3. effects of ischemia, bacteria, and substrate on the pathogenesis of intes-
Levine, S.M., Hellmann, D.B., and Stone, J.H. (2002). Gastrointestinal tinal necrosis. Journal of Pediatric Surgery, 21, 536–8.
involvement in polyarteritis nodosa (1986-2000): presentation and out- Nagarajan, S., Friedrich, T., Garcia, M., et al. (2005). Gastrointestinal leu-
comes in 24 patients. American Journal of Medicine, 112, 386–91. kocytoclastic vasculitis:  an adverse effect of sirolimus. Pediatric
Lhote, F., Cohen, P., Genereau, T., et  al. (1996). Microscopic polyangii- Transplantation, 9, 97–100.
tis: clinical aspects and treatment. Annales de Médecine Interne (Paris), Nakamura, S., Yokoi, Y., Suzuki, S., et al. (1991). A case of melena caused by a
147, 165–77. hepatic aneurysm ruptured into the intrahepatic bile duct in a patient with
Lie, J.T. (1998). Visceral intestinal Buerger’s disease. International Journal of allergic granulomatous angiitis. Japanese Journal of Surgery, 21, 471–5.
Cardiology, 66 (Suppl. 1), S249–56. Nakamura, Y., Sakurai, Y., Matsubara, T., et al. (2002). Multiple perforated
Lind, K.M., Gaub, J., and Pedersen, R.S. (1994). Henoch-Schonlein pur- ulcers of the small intestine associated with allergic granulomatous angi-
pura associated with Campylobacter jejuni enteritis. Case report. itis: report of a case. Surgery Today, 32, 541–6.
Scandinavian Journal of Urology and Nephrology, 28, 179–81. Nishie, M., Tomiyama, M., Kamijo, M., et  al. (2003). Acute chol-
Lockhart, M.E. and Robbin, M.L. (2003). Case 58:  giant cell arteritis. ecystitis and duodenitis associated with Churg-Strauss syndrome.
Radiology, 227, 512–5. Hepatogastroenterology, 50, 998–1002.
Lopez, L.R., Schocket, A.L., Stanford, R.E., et  al. (1980). Gastrointestinal Nøhr, M., Laustsen, J., and Falk, E. (1989). Isolated necrotizing panarteritis of
involvement in leukocytoclastic vasculitis and polyarteritis nodosa. the gallbladder: case report. Acta Chirurgica Scandinavica, 155, 485–7.
Journal of Rheumatology, 7, 677–84. Nunez Garcia, A., Lopez Cubero, L., Rico Zalba, L., et  al. (1998).
Luna Alcala, A., Jiménez, I.R., and Botella, E.R. (2009). Imaging techniques Thromboangiitis obliterans (Buerger’s disease) with intestinal
for the evaluation of systemic manifestations of vasculitis. Reumatología involvement--a case report. Angiology, 49, 489–92.
Clinica, 5, 178–82. Ohwada, S., Yanagisawa, A., Joshita, T., et  al. (1997). Necrotizing
Manna, R., Ghirlanda, G., Bochicchio, G.B., et al. (1985). Chronic active hep- granulomatous vasculitis of transverse colon and gallbladder.
atitis and Behcet’s syndrome. Clinical Rheumatology, 4, 93–6. Hepatogastroenterology, 44, 1090–4.
Marcolongo, R., Bayeli, P.F., and Montagnani, M. (1979). Gastrointestinal Okuda, Y., Takasugi, K., Imai, A., et  al. (1990). [Two cases of rheumatoid
involvement in rheumatoid arthritis:  a biopsy study. Journal of arthritis complicated with vasculitis-induced ischemic enterocolitis].
Rheumatology, 6, 163–73. Ryumachi, 30, 403–7, 8; discussion 7–8.
McCurley, T.L. and Collins, R.D. (1984). Intestinal infarction in rheuma- Oshima, Y., Shimizu, T., Yokohari, R., et  al. (1963). Clinical Studies on
toid arthritis. Three cases due to unusual obliterative vascular lesions. Behçet’s Syndrome. Annals of the Rheumatic Diseases, 22, 36–45.
Archives of Pathology and Laboratory Medicine, 108, 125–8. Ouellet, J.Y., Duprat, G. Jr, Laperriere, J., et al. (1988). Pyocolon: an unusual
Meaney, J.F., Prince, M.R., Nostrant, T.T., et al. (1997). Gadolinium-enhanced manifestation of colon ischemia. Canadian Association of Radiology
MR angiography of visceral arteries in patients with suspected chronic Journal, 39, 235–7.
194 SECTION 3 clinical manifestations common to vasculitis

Ozcay, N., Arda, K., Sugunes, T., et al. (2003). Polyarteritis nodosa presenting Saccardo, F., Castelnovo, L., and Monti, G. (2012). Emergency in
with necrotising appendicitis and hepatic aneurysm rupture. Turkish Cryoglobulinemia: Clinical and Therapeutic Approach. In HCV infec-
Journal of Gastroenterology, 14, 68–70. tion and cryoglobulinemia (ed. F. Dammacco). Springer-Verlag, Italia.
Ozenc, A., Bayraktar, Y., and Baykal, A. (1990). Pyloric stenosis with Sakane, T., Takeno, M., Suzuki, N., et  al. (1999). Behçet’s disease. New
esophageal involvement in Behcet’s syndrome. American Journal of England Journal of Medicine, 341, 1284–91.
Gastroenterology, 85, 727–8. Sanchez-Guerrero, J., Reyes, E., and Alarcon-Segovia, D. (1992). Primary
Pagnoux, C., Mahr, A., Cohen, P., et al. (2005). Presentation and outcome of antiphospholipid syndrome as a cause of intestinal infarction. Journal of
gastrointestinal involvement in systemic necrotizing vasculitides: analy- Rheumatology, 19, 623–5.
sis of 62 patients with polyarteritis nodosa, microscopic polyangiitis, Sasamura, H., Nakamoto, H., Ryuzaki, M., et al. (1991). Repeated intestinal
Wegener granulomatosis, Churg-Strauss syndrome, or rheumatoid ulcerations in a patient with systemic lupus erythematosus and high
arthritis-associated vasculitis. Medicine (Baltimore), 84, 115–28. serum antiphospholipid antibody levels. Southern Medical Journal,
Palvio, D.H. and Christensen, K.S. (1987). Systemic lupus erythematosus 84, 515–7.
with rectal stenosis simulating tumour or diverticulosis. Case report. Savage, D.C. (1977). Microbial ecology of the gastrointestinal tract. Annual
Acta Chirurgica Scandinavica, 153, 63–5. Review of Microbiology, 31, 107–33.
Parangi, S., Oz, M.C., Blume, R.S., et al. (1991). Hepatobiliary complications Sayek, I., Aran, O., Uzunalimoglu, B., et al. (1991). Intestinal Behçet’s dis-
of polyarteritis nodosa. Archives of Surgery, 126, 909–12. ease:  surgical experience in seven cases. Hepatogastroenterology,
Pavone, I., Grasselli, C., Chieric, E., et al. (2006). Outcomes and prognostic 38, 81–3.
factors during the course of primary small-vessel vasculitides. Journal of Schellong, S.M., Bernhards, J., Ensslen, F., et  al. (1994). Intestinal type of
Rheumatology, 33, 1299–1306. thromboangiitis obliterans (Buerger’s disease). Journal of Internal
Pelletier, S., Ekert, P., Landi, B., et al. (1992). [Exudative enteropathy in dis- Medicine, 235, 69–73.
seminated lupus erythematosus]. Annales de Gastroentérologie et Scher, J.U. and Abramson, S.B. (2011).The microbiome and rheumatoid
d'Hépatologie (Paris), 28(6-7), 259–62. arthritis. Nature Review Rheumatology, 7, 569–578.
Perko, M.J. (2001). Duplex ultrasound for assessment of superior mesenteric Schmidt, W.A. (2000). Doppler ultrasonography in the diagnosis of giant
artery blood flow. European Journal of Vascular and Endovascular, 21, cell arteritis. Clinical and Experimental Rheumatology, 18 (Suppl.
106–17. 20), S40–2.
Pfitzmann, R., Nussler, N.C., Heise, M., et  al. (2002). [Mesenteric artery Schmidt, W.A., Kraft, H.E., Borkowski, A., et al. (1999). Color duplex ultra-
occlusion as a rare complication of thromboangiitis obliterans]. Chirurg, sonography in large-vessel giant cell arteritis. Scandinavian Journal of
73, 86–9. Rheumatology, 28, 374–6.
Pickhardt, P.J. and Curran, V.W. (2001). Fulminant enterocolitis in Wegener’s Sfikakis, P.P., Markomichelakis, N., Alpsoy, E., et al. (2007). Anti-TNF ther-
granulomatosis: CT findings with pathologic correlation. AJR American apy in the management of Behçet’s disease—review and basis for recom-
Journal of Roentgenology, 177, 1335–7. mendations. Rheumatology (Oxford), 46, 736–41.
Pipitone, N., Olivieri, I., and Salvarani, C. (2012). Recommendations of Sharma, M.C., Safaya, R., and Sidhu, B.S. (1996). Perforation of small
the Italian society of rheumatology for the treatment of the primary intestine caused by Churg-Strauss syndrome. Journal of Clinical
large-vessel vasculitis with biological agents. Clinical and Experimental Gastroenterology, 23, 232–5.
Rheumatology, 30 (Suppl. 70), S139–61. Shiboski, C.H., Regezi, J.A., Sanchez, H.C., et al. (2002). Oral lesions as the
Qin, J., Li, R., Raes, J., Arumugam, M., Burgdorf, K. S., Manichanh, C., et al. first clinical sign of microscopic polyangiitis: a case report. Oral Surgery
(2010). A human gut microbial gene catalogue established by metagen- Oral Medicine Oral Pathology Oral Radiology and Endodontics, 94,
omic sequencing. Nature, 464, 59−65. 707–11.
Raat, H., Stockx, L., Broeckaert, L., et  al. (1993). Mesenteric involvement Shimamoto, C., Hirata, I., Ohshiba, S., et al. (1990). Churg-Strauss syndrome
of thromboangiitis obliterans (Buerger’s disease) in a woman. Journal (allergic granulomatous angiitis) with peculiar multiple colonic ulcers.
Belge de Radiologie, 76, 245–6. American Journal of Gastroenterology, 85, 316–9.
Rademaker, J. (1998). Veno-occlusive disease of the colon—CT findings. Shimizu, T., Ehrlich, G.E., Inaba, G., et al. (1979). Behçet’s disease (Behçet’s
European Radiology, 8, 1420–1. syndrome). Seminars in Arthritis and Rheumatism, 8, 223–60.
Rasmussen, N.H. (1982). Henoch-Schonlein purpura after yersiniosis. Shintaku, M., Inoue, N., Sasaki, M., et al. (1991). Cytomegalovirus vasculitis
Archives of Disease in Childhood, 57, 322–3. accompanied by an exuberant fibroblastic reaction in the intestine of an
Raza, K., Exley, A.R., Carruthers, D.M., et al. (1999). Localized bowel vasculi- AIDS patient. Acta Pathologica Japonica, 41, 900–4.
tis: postoperative cyclophosphamide or not? Arthritis and Rheumatism, Shiohira, Y., Uehara, H., Miyazato, F., et al. (1993). [Vasculitis-related acute
42, 182–5. abdomen in systemic lupus erythematosus—ultrasound appearances in
Reissman, P., Weiss, E.G., Teoh, T.A., et al. (1994). Gangrenous ischemic coli- lupus patients with intra-abdominal vasculitis]. Ryumachi, 33, 235–41.
tis of the rectum: a rare complication of systemic lupus erythematosus. Siddiqui, M.Z., Reis, E.D., Soundararajan, K., et al. (2001). Buerger’s disease
American Journal of Gastroenterology, 89, 2234–6. affecting mesenteric arteries: a rare cause of intestinal ischemia—a case
Ripollés, T., Simo, L., Martinez-Perez, M.J., et  al. (2005). Sonographic report. Vascular Surgery, 35, 235–8.
findings in ischemic colitis in 58 patients. AJR American Journal of Smith, F.C., Gosling, P., Sanghera, K., et al. (1994). Microproteinuria predicts
Roentgenology, 184, 777–85. the severity of systemic effects of reperfusion injury following infrarenal
Ronkainen, J., Koskimies, O., Ala-Houhala, M., et al. (2006). Early prednisone aortic aneurysm surgery. Annals of Vascular Surgery, 8, 1–5.
therapy in Henoch-Schönlein purpura:  a randomized, double-blind, Smith, J.A., O’Sullivan, M., Gough, J., et al. (1988). Small-intestinal perfora-
placebo-controlled trial. Journal of Pediatrics, 149, 241–7. tion secondary to localized giant-cell arteritis of the mesenteric vessels.
Roobottom, C.A. and Dubbins, P.A. (1993). Significant disease of the celiac British Journal of Rheumatology, 27, 236–8.
and superior mesenteric arteries in asymptomatic patients: predictive Sokol, R.J., Farrell, M.K., and McAdams, A.J. (1984). An unusual presenta-
value of Doppler sonography. AJR American Journal of Roentgenology, tion of Wegener’s granulomatosis mimicking inflammatory bowel dis-
161, 985–8. ease. Gastroenterology, 87, 426–32.
Roza, M., Galbe, M., Gonzalez Baschwitz, C., et al. (1983). Henoch-Schoenlein Soysal, M., Denizci, U., Alhan, S., et al. (1990). Splenic size in Behçet’s syn-
purpura after shigellosis. Clinical Nephrology, 20, 269. drome. British Journal of Rheumatology, 29, 497–8.
CHAPTER 14 vasculitic manifestations in the gastrointestinal tract 195

Srigley, J.R. and Gardiner, G.W. (1980). Giant cell arteritis with small bowel Ueda, S., Matsumoto, M., Ahn, T., et al. (2001). Microscopic polyangiitis com-
infarction. A case report and review of the literature. American Journal plicated with massive intestinal bleeding. Journal of Gastroenterology,
of Gastroenterology, 73, 157–61. 36, 264–70.
Stanson, A.W., Friese, J.L., Johnson, C.M., et  al. (2001). Polyarteritis Vaglio, A., Moosig, F., and Zwerina, J. (2012). Churg-Strauss syn-
nodosa: spectrum of angiographic findings. Radiographics, 21, 151–9. drome: update on pathophysiology and treatment. Current Opinion in
Steele, C., Bohra, S., Broe, P., et al. (2001). Acute upper gastrointestinal haem- Rheumatology, 24, 24–30.
orrhage and colitis: an unusual presentation of Wegener’s granulomato- van Laar, J.M., Smit, V.T., de Beus, W.M., et al. (1998). Rheumatoid vasculitis
sis. European Journal of Gastroenterology and Hepatology, 13, 993–5. presenting as appendicitis. Clinical and Experimental Rheumatology,
Storesund, B., Gran, J.T., and Koldingsnes, W. (1998). Severe intestinal 16, 736–8.
involvement in Wegener’s granulomatosis:  report of two cases and Vanoli, M., Daina, E., Salvarani, C., et al. (2005). Takayasu’s Arteritis: A study
review of the literature. British Journal of Rheumatology, 37, 387–90. of 104 Italian patients. Arthritis and Rheumatism, 53, 100–7.
Sultan, S.M., Ioannou, Y., and Isenberg, D.A. (1999). A review of gastroin- Vega Sáenz, J.L., Baraias Martinez, J.M., and Torres Almendros, M. (1995).
testinal manifestations of systemic lupus erythematosus. Rheumatology Ischemic colitis as manifestation of Takayasu arteritis. Gastroenterología
(Oxford), 38, 917–32. y Hepatología, 18, 326–9.
Suzuki, M., Nabeshima, K., Miyazaki, M., et al. (2005). Churg-Strauss syn- Vlahos, K., Theodoropoulos, G.E., Lazaris, A., et al. (2005). Isolated colonic
drome complicated by colon erosion, acalculous cholecystitis and liver leukocytoclastic vasculitis causing segmental megacolon:  report of a
abscesses. World Journal of Gastroenterology, 11, 5248–50. rare case. Diseases of the Colon and Rectum, 48, 167–71.
Takeuchi, K. and Kuroda, Y. (2000). [Rheumatoid vasculitis with multiple Walton, E.W. (1958). Giant-cell granuloma of the respiratory tract (Wegener’s
intestinal ulcerations: report of a case]. Ryumachi, 40, 639–43. granulomatosis). British Medical Journal, 2 (5091), 265–70.
Testini, M., Verzillo, F., Paradiso, V., et al. (1996). The surgical treatment of Weiss, P.F., Feinstein, J.A., Luan, X., et al. (2007). Effects of corticosteroid on
vascular and gastrointestinal implications in Behçet’s disease. Report of Henoch-Schönlein purpura: a systematic review. Pediatrics, 120, 1079–87.
four cases. Panminerva Medica, 38, 185–9. Wheeler, R.A., Najmaldin, A.S., Soubra, M., et al. (1990). Surgical presen-
Tian, X.P. and Zhang, X. (2010). Gastrointestinal involvement in systemic tation of Kawasaki disease (mucocutaneous lymph node syndrome).
lupus erythematosus:  insight into pathogenesis, diagnosis and treat- British Journal of Surgery, 77, 1273–4.
ment. World Journal of Gastroenterology, 16, 2971–7. Whitehead, R. (1976). The pathology of ischemia of the intestines. Pathology
Tizard, E.J. (1999). Henoch-Schonlein purpura. Archives of Disease in Annual, 11, 1–52.
Childhood, 80, 380–3. Wiesner, W., Khurana, B., Ji, H., et al. (2003). CT of acute bowel ischemia.
Trimble, M.A. and Weisz, M.A. (2002). Infarction of the sigmoid colon sec- Radiology, 226, 635–50.
ondary to giant cell arteritis. Rheumatology (Oxford), 41, 108–10. Xavier, J., Shahram, F., Davatchi, F., et al. (2012). Associated study of IL10 and
Trompeter, M., Brazda, T., Remy, C.T., et al. (2002). Non-occlusive mesen- IL23R-IL12RB2 in Iranian patients with Behcet’s disease. Arthritis and
teric ischemia: etiology, diagnosis, and interventional therapy. European Rheumatism, 64, 2761–72.
Radiology, 12, 1179–87. Yashiro, K., Nagasako, K., Hasegawa, K., et al. (1986). Esophageal lesions in
Tsai, C.N., Chang, C.M., Chuang, C.H., et  al. (2004). Extended colonic intestinal Behcet’s disease. Endoscopy, 18, 57–60.
ulcerations in a patient with microscopic polyangiitis. Annals of the Yazici, H., Tuzun, Y., Pazarli, H., et  al. (1980). Behcet’s disease as seen in
Rheumatic Diseases, 63, 1521–2. Turkey. Haematologica, 65, 381–3.
Tsai, J.T. (1980). Perforation of the small bowel with rheumatoid arthritis. Yazici, Z., Savci, G., Parlak, M., et al. (1997). Polyarteritis nodosa present-
Southern Medical Journal, 73, 939–40. ing with hemobilia and intestinal hemorrhage. European Radiology, 7,
Turner, H.E., Myszor, M.F., Bradlow, A., et al. (1996). Lupus or lupoid hepa- 1059–61.
titis with mesenteric vasculitis. British Journal of Rheumatology, 35, Zizic, T.M., Shulman, L.E., and Stevens, M.B. (1975). Colonic perforations in
1309–11. systemic lupus erythematosus. Medicine (Baltimore), 54, 411–26.
Tyagi, S., Verma, P.K., Kumar, N., et al. (1997). Stent angioplasty for relief of Zulian, F., Falcini, F., Zancan, L., et al. (2003). Acute surgical abdomen as
chronic mesenteric ischemia in Takayasu arteritis. Indian Heart Journal, presenting manifestation of Kawasaki disease. Journal of Pediatrics,
49, 315–8. 142, 731–5.
CHAPTER 15

Renal manifestations
Cees G.M. Kallenberg and
Jan Willem Cohen Tervaert

Renal involvement is common in both secondary and primary vas- or develop in patients with GCA, other conditions should be sus-
culitides. Particularly in the antineutrophil cytoplasmic antibody pected. Diagnostic work-up, including ANCA testing and a renal
(ANCA)-associated primary vasculitides, renal involvement is one biopsy if indicated, should be performed in order to reveal the
of the major manifestations of the disease and has a great impact underlying cause of the observed renal abnormalities. Treatment
on prognosis. Vasculitis may even be restricted to the kidneys, as should be directed at the underlying cause or condition.
in idiopathic ANCA-associated necrotizing crescentic glomerulo-
nephritis. In this chapter, the renal manifestations of the primary Takayasu’s arteritis
vasculitides are discussed. In addition, the clinical course, histo- Takayasu’s arteritis has been defined as a granulomatous inflam-
pathological findings, and pathogenesis, as well as prognosis and mation of the aorta and its major branches. In contrast to GCA,
treatment of renal involvement are described. patients are generally younger than 50 years and female (Jennette
et al. 2013). The arteritis in its acute stage is characterized by infil-
Renal involvement in large-vessel tration of the vessel wall with mononuclear cells, granulocytes,
vasculitides and, sometimes, giant cells. The inflammatory process leads to
obstructive changes due to intimal proliferation and medial fibro-
Giant cell arteritis sis. Although the vessels in the upper part of the body are affected
Giant cell arteritis (GCA) is defined, according to the 2012 Chapel more frequently, extension of the disease process to the abdominal
Hill Consensus Conference (Jennette et  al. 2013), as a granu- vessels occurs. Extension to the level of the renal arteries may result
lomatous arteritis of the aorta and its major branches. It often in renovascular hypertension and renal insufficiency, generally
involves the temporal artery, occurs in patients generally older than mild, due to obstruction of the ostia of the renal arteries or involve-
50 years, and is frequently associated with polymyalgia rheumatica. ment of the renal arteries themselves (Lagneau and Michel 1985).
In general, renal involvement is rare. Although mild proteinuria Hypertension, although difficult to detect in cases of pulseless dis-
and microscopic haematuria, sometimes with red cell casts, have ease, and renal insufficiency, should raise suspicion of involvement
been mentioned in less than 10% of patients (Klein et  al. 1975), of the renal arteries.
renal insufficiency is rare in GCA. Using more-sensitive imaging Mild proteinuria and haematuria may occur, but overt glomeru-
techniques it has been found that GCA not infrequently involves lar or interstitial disease is uncommon. Secondary AA amyloidosis
the aorta and its branches, including the renal arteries although has been described in a few cases (Wada et al. 1999). In addition,
renovascular hypertension is rare (Cid et al. 2009). Secondary renal a few cases of concomitant IgA nephropathy have been described
amyloidosis has been described in a few cases of GCA and should but this may be only coincidental (Cavatorta et al. 1995). Signs of
be suspected in persons with a long history of GCA who develop glomerular or interstitial disease suggest the coexistence of another
proteinuria and renal failure (Escriba et al. 2000). condition necessitating diagnostic work-up.
Overlapping syndromes of GCA with other forms of systemic vas- The active stage of large-vessel vasculitis, that is either GCA or
culitis do occur. Cases of GCA in conjunction with ANCA-positive Takayasu’s arteritis is treated with glucocorticoids, sometimes in
crescentic glomerulonephritis have been described (Logar et  al. combination with immunosuppressive drugs. Renal artery stenosis
1994; Muller et al. 2004). Classical polyarteritis affecting the kid- can be treated by interventional radiology techniques (Lovaria et al.
neys has also been documented together with GCA (O’Neill et al. 1999) or by reconstructive surgery. Results of surgery are usually
1976). It is not clear at present whether the occurrence of other satisfactory, although complex and repeat revascularization proce-
forms of vasculitis in patients with GCA is based on a pathophysi- dures are often needed. One series reported cure of hypertension in
ologically relevant association. The distinction made between the 12 of 19 patients (63%), improvement in six (31%), and no change
different forms of vasculitis is somewhat arbitrary and overlapping in one patient (6%); one of these 19 patients died due to a complica-
forms do frequently occur. tion of the procedure (Kieffer et al. 1990). A study on 27 patients
What conclusion can be drawn? Renal involvement is not a with Takayasu’s arteritis-induced renal artery stenosis demon-
characteristic finding of GCA. When signs of renal involvement, strated patency of the grafts in 90% of patients after 5 years, mak-
such as proteinuria, haematuria, or renal insufficiency, are present ing renal revascularization a first choice in this condition (Weaver
198 SECTION 3 clinical manifestations common to vasculitis

et al. 2004). Although results of open revascularization are better, day). Milder cases, with musculoskeletal involvement only, can be
endovascular intervention is possible as well with a 5-year primary treated with prednisone alone. Hypertension must be treated, but
patency rate of 49% (Ham et al. 2010). one should be watchful for the development of acute renal failure
induced by ACE inhibitors (Wang et  al. 1996). Major haemor-
Renal involvement in rhages can be treated by interventional radiology techniques such
medium-vessel vasculitides as embolization (Schouffoer et al. 1998).
Kawasaki’s disease
Kawasaki’s disease (KD) is a form of arteritis involving large,
ANCA-associated small-vessel
medium-sized, and small arteries. Primary renal involvement is vasculitides
rare, but acute renal failure may occur, either due to fluid exudation Antineutrophil cytoplasmic antibodies were first described as
from the intravascular to the extravascular space (prerenal insuf- sensitive and specific markers for granulomatosis with polyangii-
ficiency) or nephritis, which has not been well defined (Senzaki tis (GPA, formerly Wegener’s granulomatosis) by van der Woude
et al. 1994). Sonography showed enlarged kidneys with increased et al. (1985). The autoantibodies in GPA produce a characteristic
cortical echogenicity in a few cases (Nardi et al. 1985). Histological cytoplasmic staining pattern (cANCA) by indirect immunofluo-
findings in three cases suggested immune complex disease (Salcedo rescence (IIF) on ethanol-fixed neutrophils. The target antigen of
et al. 1988), interstitial nephritis (Veiga et al. 1992), and mesan- cANCA is proteinase 3, a third serine protease (besides elastase
gial sclerosis (Joh et  al. 1997), respectively. Also, three cases of and cathepsin G) from azurophilic granules (Goldschmeding et al.
transient nephrotic syndrome have been described (Krug et  al. 1989). Shortly after the detection of cANCA in GPA, it was found
2012). Post-mortem studies on 18 patients who died of acute KD that many patients with related conditions, such as microscopic
showed infiltration of IgA plasma cells in different organs, includ- polyangiitis (MPA), eosinophilic granulomatosis with polyangii-
ing the kidney, suggesting a triggering agent from the respiratory tis (EGPA, formerly Churg–Strauss syndrome), and idiopathic
tract inducing a systemic IgA response (Rowley et al. 2000). Taken necrotizing crescentic glomerulonephritis (NCGN), had autoanti-
together, renal pathology in KD has not been well defined. bodies that produced a perinuclear staining pattern (pANCA) on
ethanol-fixed neutrophils. The target antigen of pANCA in those
Classic polyarteritis nodosa conditions is myeloperoxidase (MPO) (Kallenberg et al. 1994). The
Classic polyarteritis nodosa (PAN) is characterized by necrotizing specificity of autoantibodies to either proteinase 3 (PR3) or MPO
inflammation of medium-sized or small arteries; glomerulone- in the aforementioned conditions, as well as the similarity of their
phritis or vasculitis in arterioles, capillaries, or venules is absent. renal histopathological findings (see Section PR3-ANCA versus
Tests for antineutrophil cytoplasmic antibodies (ANCA) are gener- MPO-ANCA Associated Glomerulonephritis), justify their classi-
ally negative. Classic PAN affects muscular arteries, frequently at fication as ANCA-associated vasculitides (Kallenberg et al. 1994,
bifurcations and in a segmental distribution. Necrotizing arteritis Jennette et al. 2013).
occurs in the acute stage with infiltration of the vessel wall with First, we discuss the renal manifestations of this group of disor-
neutrophils, monocytes, and lymphocytes, and sometimes eosin- ders, then deal with their pathophysiology, especially the role of
ophils. The lesions differ in age, varying from fibrinoid necrosis, ANCA, and finally review treatment of their renal manifestations.
which is frequent in the acute stage, to obliterative changes from
intimal and medial proliferation, in the healing stages. Cortical Granulomatosis with polyangiitis
infarcts result from ischaemia due to acute or chronic obliteration. Granulomatosis with polyangiitis (GPA) is defined by the 2012
In necrotic areas, aneurysms may occur and rupture, resulting in Chapel Hill Consensus Conference as a granulomatous inflam-
renal, perirenal, and retroperitoneal haemorrhage (Chandrakantan mation involving the respiratory tract and necrotizing vasculitis
and Kaufman 1999). Hypertension, of renovascular origin, is pre- affecting small to medium-sized vessels. Renal involvement occurs
sent in at least 50% of patients (Cohen et al. 1980). Furthermore, frequently in GPA and necrotizing glomerulonephritis is common
there may be symptoms, such as flank pain and haematuria, related (Jennette et al. 2013). At first presentation, around 50% of patients
to haemorrhage from ruptured (micro)aneurysms. Haematuria show clinical signs of renal involvement such as proteinuria, uri-
is present in most of the cases and some degree of proteinuria nary sediment abnormalities, and diminished renal function. The
(mostly mild) and renal insufficiency is frequent. A renal biopsy remaining patients present with extrarenal granulomatous inflam-
may show characteristic findings in intrarenal arteries. Due to the mation, with more or less overt vasculitis, particularly in the upper
risk of haemorrhage from rupturing aneurysms, a renal biopsy is airways. More than 50% of the patients who present without
not advocated as a first choice for diagnosis. Renal angiography renal manifestations develop clinical evidence of renal involve-
can detect (micro)aneurysms as well as non-aneurysmal lesions, ment during the course of their disease (Hoffman et  al. 1992;
such as perfusion defects, collateral arteries, and delayed empty- Reinhold-Keller et al. 2000). Although the clinical course of renal
ing of small renal arteries. The sensitivity and specificity of renal involvement in GPA is highly variable, the typical presentation of a
aneurysms for a diagnosis of PAN were 43% and 69%, respectively, patient with GPA consists of a period of insidious onset with upper
as shown in a study of 25 children with PAN (Brogan et al. 2002). airway symptoms, general malaise, myalgias, and arthralgias. This
When non-aneurysmal lesions were included in the analysis sensi- stage may last for months to years, after which a more acute phase
tivity rose to 80% but specificity fell to 50%. may occur with systemic vasculitis particularly manifested in the
Treatment of classic PAN with major organ involvement, such kidneys. Clinically, this course is described as rapidly progressive
as the kidneys, consists of glucocorticoids (prednisone 1 mg/ glomerulonephritis (RPGN). RPGN is characterized by a rapid
kg per day) accompanied by cyclophosphamide (2–3 mg/kg per decline in renal function together with signs of glomerulonephritis,
CHAPTER 15 renal manifestations 199

that is proteinuria (which is generally mild and does not progress outcome: percentage of normal glomeruli were predictive for a bet-
to nephrotic syndrome) and microscopic haematuria with cel- ter outcome and chronic acute tubulointerstitial lesions for a worse
lular casts (Fauci et  al. 1983; Hoffman et  al. 1992). Outcome of outcome (de Lind van Wijngaarden et al. 2006).
ANCA-associated renal vasculitis is variable. In a series of 246 new The pauci-immune character contrasts with the linear staining
patients with this condition, 28% reached end-stage renal failure seen in patients with RPGN as part of anti-glomerular basement
and 47% of these latter patients died. Cumulative patient sur- membrane (anti-GBM) disease, and with the granular staining seen
vival in this cohort of 246 patients was 82% at 1 year and 76% at in RPGN as part of immune-complex mediated glomerulonephritis
5 years. Mortality was associated with older age, initial creatinine of systemic autoimmune diseases and infection-related conditions
level greater than 200 μmol/l and sepsis (Booth et al. 2003). During (Jennette and Falk 1994; Jennette et al. 1989; Ronco et al. 1983).
the last decade, outcome of ANCA-associated glomerulonephritis In some cases, IgA nephropathy appears late in the course of the
has improved due to both better treatment and earlier diagnosis disease (Andrassy et al. 1992).
(Hilhorst et al. 2013).
Histopathologically, the initial phase is characterized by endothe- Microscopic polyangiitis
lial swelling with infiltration of polymorphonuclear granulocytes Microscopic polyangiitis (MPA) has been defined as necrotizing
followed by fibrinoid necrosis of the capillary wall and intracapillary vasculitis with few or no immune deposits affecting small ves-
thrombosis. Next, cellular crescents develop along with extension of sels of all types. Necrotizing glomerulonephritis is common, as
the vasculitic process. Crescents mature and lead to destruction of is pulmonary capillaritis. MPA is distinguished from classic PAN
Bowman’s capsule (Figure 15.1). Periglomerular inflammation fol- (see Chapter  21). In MPA, the major sites of the vasculitic pro-
lows, not infrequently accompanied by (pseudo)granuloma forma- cess are arterioles, capillaries, and venules. ANCA are present in
tion. The interstitium is also involved in most cases, with infiltration most instances and are directed to MPO in about 70% of the cases,
of lymphocytes, monocytes, plasma cells, and polymorphonuclear while only 20% of cases have antibodies to PR3, and around 10%
leukocytes. Granuloma formation occurs, particularly in the inter- are ANCA-negative (Kallenberg et al. 1994; Chen and Kallenberg
stitium, in about 10% of the cases. Necrotizing vasculitis is seen at 2010). MPA frequently presents as a renal–pulmonary syndrome in
times in the interstitial small arteries and in more advanced stages, which rapidly progressive glomerulonephritis occurs together with
sclerosis. Crescents become fibrous and global, or diffuse glomeru- lung haemorrhage, which may lead to pulmonary insufficiency
losclerosis develops. Most importantly, direct immunofluorescence (Jennette 1991). Other organ systems may be involved as well,
studies do not clearly demonstrate immune deposits along the glo- particularly the skin, evident as palpable purpura. Mononeuritis
merular capillary wall. Some IgM and C3 deposits can be seen, par- multiplex may also occur. Renal lesions cause proteinuria, which is
ticularly in Bowman’s space, but a typical granular or linear staining generally mild; haematuria with red cell casts; and varying degrees
pattern of the glomerular capillary wall is lacking. Based on these of renal insufficiency. Renal insufficiency may appear rapidly, par-
findings, the renal histology has been described as ‘pauci-immune’ ticularly in cases with a renal–pulmonary syndrome, but in many
glomerulonephritis. ANCA-associated glomerulonephritis has cases there is an insidious onset of renal insufficiency, especially
been classified into four categories:  focal, crescentic, mixed, and in anti-MPO-associated disease (Franssen et al. 1995). Differences
sclerotic (Berden et al. 2010). This classification is of clinical use in the clinical course between patients with anti-PR3-associated
as it independently predicts renal outcome, in particular develop- disease versus those with anti-MPO-associated NCGN are dis-
ment of end-stage renal disease (Chang et al. 2012; Hilhorst et al. cussed in Section PR3-ANCA versus MPO-ANCA Associated
2013). In patients presenting with severe renal involvement (serum Glomerulonephritis.
creatinine >500  μmol/l) renal histology was predictive for renal In patients with MPA, renal lesions histopathologically resemble
those in patients with GPA. The more insidious clinical course of
renal lesions in patients with MPA versus those with GPA is, how-
ever, reflected in a larger number of chronic lesions, such as glomer-
ulosclerosis and interstitial fibrosis at presentation (Franssen et al.
1995). As in NCGN that is part of GPA, immune deposits are scarce
in renal biopsies from patients with MPA. As such, the condition is
designated as ‘pauci-immune’ NCGN (Stilmant et al. 1979).

Eosinophilic granulomatosis with polyangiitis


Eosinophilic granulomatosis with polyangiitis (EGPA) has been
defined by the Chapel Hill Group as an eosinophil-rich inflamma-
tion with granulomatous inflammation involving the respiratory
tract and necrotizing vasculitis affecting small to medium-sized
vessels, in association with asthma and blood eosinophilia
(Jennette et al. 2013). Renal involvement occurs in about 50% of
EGPA patients. The clinical presentation of renal disease is not dif-
ferent from that in MPA and GPA: microscopic haematuria with
Fig. 15.1 Granulomatosis with polyangiitis in a 72-year-old male, admitted
cellular casts, proteinuria and varying degrees of renal insufficiency
with fever, multiple pulmonary nodules on chest radiograph, microscopic
erythrocyturia, mild proteinuria, and a serum creatinine concentration of (Eustace et al. 1999; Lanham et al. 1984). The most striking his-
135 μmol/l. Renal biopsy shows a periglomerular round cell infiltration, destruction tological finding is the widespread infiltration of activated eosino-
of Bowman’s capsule, and an extracapillary crescent. Silver methenamine, H&E, ×50. phils, particularly in the interstitium, accompanied by granuloma
200 SECTION 3 clinical manifestations common to vasculitis

formation. In the majority of cases, glomeruli show focal or diffuse the neutrophil-activating potential of IgG fractions from their
necrotizing and crescentic glomerulonephritis. By direct immuno- PR3-ANCA and MPO-ANCA patients (see Section Pathophysiology
fluorescence, immune deposits are generally absent, which classi- of the ANCA-associated Glomerulonephritides) (Franssen et  al.
fies glomerulonephritis in EGPA as pauci-immune (Clutterbuck 1999). They found that the PR3-ANCA containing fractions had a
et al. 1990; Gaskin et al. 1991). In some cases, however, there are significantly higher potential to induce neutrophil activation than
findings more consistent with classical PAN with necrotizing arte- the MPO-ANCA fractions. These data suggest that the presence
ritis of medium-sized arteries and aneurysm formation. of PR3-ANCA induces a more acute inflammatory response com-
There is no consensus on the role of ANCA in EGPA. In general, pared with MPO-ANCA. Their data (reviewed in Franssen et al.
about 50–60% of patients are ANCA-positive. Of those positive 2000) argue for a distinction between PR3-ANCA-associated vas-
for ANCA, most have autoantibodies to MPO (Cohen et al. 1995; culitis and MPO-ANCA-associated vasculitis. Other investigators
Cohen Tervaert et al. 1991a; Eustace et al. 1999). ANCA-negative also observed more frequent relapses in PR3-ANCA-associated
EGPA patients tend to present with limited disease, whereas EGPA vasculitis compared to MPO-ANCA-associated vasculitis, which is
patients positive for ANCA usually present with major organ also in favour of PR3-ANCA- and MPO-ANCA-associated vasculi-
involvement, frequently including the kidneys (Cohen Tervaert tis being different disease entities (Booth et al. 2003; Slot et al. 2004;
et al. 1991a; Cohen Tervaert et al. 1991b). The dichotomy of EGPA Lionaki et al. 2012). Also, the different associations of MPO-ANCA
into an ANCA-negative disease connected with the hypereosino- and PR3-ANCA positivity with HLA-class II antigens suggest that
philic syndrome and an ANCA-positive disease with small-vessel MPO-ANCA- and PR3-ANCA-associated vasculitis, rather than
vasculitis including the kidneys has been highlighted (Sinico et al. MPA and GPA, are different diseases (Lyons et al. 2012).
2005; Kallenberg 2005).
Pathophysiology of the ANCA-associated
Idiopathic necrotizing and crescentic glomerulonephritides
glomerulonephritis As already discussed, ANCA directed to either MPO or PR3 are
As already discussed, idiopathic necrotizing and crescentic glo- closely associated with pauci-immune necrotizing crescentic glo-
merulonephritis (NCGN) is the characteristic renal histopatholog- merulonephritis, either idiopathic or as part of GPA, MPA, and
ical manifestation in ANCA-associated small-vessel vasculitides. EGPA (Kallenberg et al. 1994). It has been shown that patients with
A number of patients present with pauci-immune NCGN without PR3-ANCA-associated disease have an eightfold increased risk
extrarenal manifestations of systemic vasculitis. In most of these, for relapse once ANCA is persistently or intermittently positive
ANCA are positive and directed against MPO. This condition, after induction of remission (Stegeman et al. 1994). Longitudinal
therefore, can be considered as a form of MPA limited to the kid- observations also point to a relationship between changes in level
ney. Patients present with the characteristic findings of NCGN: rap- of the autoantibodies and changes in disease activity of the asso-
idly progressive glomerulonephritis with haematuria and cellular ciated disorders, as rises in titres of ANCA appear to precede
casts, mild proteinuria, and renal insufficiency. In addition, arthral- clinical disease activity (Cohen Tervaert et  al. 1989; Egner and
gia, myalgia, and general malaise are common. Signs of systemic Chapel 1990; Jayne et al. 1995; Boomsma et al. 2000). Treatment
vasculitis may develop later on, but the majority do not develop based on changes in ANCA titres resulted in the prevention of
systemic vasculitis. The histopathological findings in the kidneys disease relapses (Cohen Tervaert et al. 1990). These studies were
are indistinguishable from those observed in patients with MPA. not confirmed by others (Kerr et al. 1993; Finkielman et al. 2007).
This also underscores that idiopathic NCGN can be considered as A meta-analysis of these studies showed a likelihood ratio of 2.84
a renal-limited form of MPA (Jennette 1998; Jennette et al. 1989; (CI 1.65–4.90) for a rise in ANCA levels as a predictor of an ensu-
Stilmant et al. 1979). ing relapse (Tomasson et al. 2012). These data somewhat suggest,
but certainly do not prove, that ANCA are pathogenic in patients
PR3-ANCA versus MPO-ANCA associated with ANCA-associated pauci-immune glomerulonephritis. The
glomerulonephritis observation of pulmonary haemorrhage and renal involvement in
Are there clinically relevant differences between PR3-ANCA and a neonate with MPO-ANCA in cord blood from a mother with a
MPO-ANCA associated glomerulonephritis? Franssen et al. (1995) previous history of MPO-ANCA vasculitis is, however, certainly
retrospectively analysed clinical features, pattern of pretreatment suggestive of a pathogenic role for MPO-ANCA (Bansal and
renal function loss, renal morphology, and outcome in a series of 46 Tobin 2004). More recently, a pathogenic epitope was described
patients with PR3-ANCA-associated disease and 46 patients with for MPO-ANCA. This linear epitope, consisting of amino acids
MPO-ANCA-associated disease. The mean number of affected 447–459, was not only exclusive for active disease but was also
organs in the PR3-ANCA group exceeded that of the MPO-ANCA detected in the total Ig fraction of ANCA-negative patients, reac-
group (3.9 versus 1.4). Granuloma formation occurred more fre- tivity being masked by ceruloplasmin. Furthermore, antibodies
quently in PR3-ANCA patients (Franssen et al. 1998). More relapses against this epitope had pathogenic properties as they were able
were seen in the PR3-ANCA group (Franssen et al. 1995; Franssen to activate neutrophils in vitro and induce nephritis in mice (Roth
et al. 1998). Although the prevalence of renal disease did not dif- et al. 2013).
fer between these groups, pretreatment renal function deteriorated What other data are available to support a pathophysiological role
significantly faster in PR3-ANCA patients than in MPO-ANCA for ANCA? In vitro, ANCA are able to activate neutrophils to pro-
patients. Also, the activity index of the renal biopsies was higher in duce reactive oxygen species and to release lytic enzymes such as
the PR3-ANCA patients, whereas the chronicity index was lower. elastase and PR3 (Falk et al. 1990). Neutrophils must be in a state of
Kidney survival was not different between the groups. In order to preactivation (‘primed’) to be activated by ANCA. Priming occurs
find an explanation for these differences, these investigators studied in the presence of low amounts of proinflammatory cytokines such
CHAPTER 15 renal manifestations 201

as tumour necrosis factor-α (TNF-α) or interleukin-1. During system, resulting in attraction of neutrophils. Those neutrophils are
priming, the target antigens of ANCA, that is PR3 and MPO, are subsequently activated by ANCA and degrade the immune com-
expressed at the cell surface and are therefore accessible for inter- plexes that were initially present.
action with ANCA. Interestingly, neutrophils, ex vivo, can also The potential of ANCA to augment an inflammatory in vivo
express PR3, but not MPO, on their surface in a resting state with- reaction has been demonstrated by Heeringa et  al. (1996) in an
out priming (mPR3). Percentages of mPR3-expressing neutro- animal model of anti-glomerular basement membrane (anti-GBM)
phils differ between individuals and seem genetically determined disease. They injected a subclinical dose of heterologous anti-GBM
(Schreiber et al. 2003). High levels of mPR3-expressing neutrophils antibodies in rats, which resulted in deposition of immunoglobu-
are found in patients with GPA and are associated with relapsing lins along the GBM without severe glomerulonephritis. In rats
disease (Rarok et al. 2002). In vitro, mPR3-expressing neutrophils immunized with human MPO, which develop anti-MPO antibod-
can be stimulated by anti-PR3 antibodies without priming (van ies cross-reactive to their own MPO, severe necrotizing and cres-
Rossum et  al. 2004). The interaction between neutrophils and centic glomerulonephritis developed after injection of a subclinical
ANCA occurs only when neutrophils are adherent to a surface, a dose of anti-GBM antibody. These experiments show the phlogis-
process in which β2-integrins are involved (Reumaux et al. 1995). tic potential of ANCA. The most convincing model for the patho-
This process is assumed to occur in vivo at the endothelial surface. genicity of ANCA comes from studies in MPO-deficient mice.
Indeed, activated neutrophils adherent to the endothelium are These mice were immunized with mouse MPO and their spleno-
observed in renal biopsies from patients with ANCA-associated cytes were transferred to immunodeficient mice which develop
NCGN (Brouwer et al. 1994). ANCA-induced neutrophil activa- severe necrotizing and crescentic glomerulonephritis, granuloma-
tion involves not only binding of the antibodies via their F(ab)2 tous inflammation, and systemic necrotizing vasculitis (Xiao et al.
fragments to surface expressed PR3 or MPO, but also via interac- 2002). Passive transfer of anti-MPO IgG alone also resulted in focal
tion of their Fc regions with Fc receptors on neutrophils, particu- NCGN with a paucity of Ig deposition. Lesions in the latter model
larly with the FcγRIIa (Porges et al. 1994). FcγRIIa is the only Fc could be aggravated by injection of lipopolysaccharide (Huugen
receptor that interacts with IgG2, but it also has a particular affinity et al. 2005).
for the IgG3 subclass. Interestingly, the increase in neutrophil acti- Further studies showed that the alternative pathway of comple-
vating capacity of serum IgG fractions from remission to relapse in ment is involved as well. Recipient mice deficient for either factor B
patients with PR3-ANCA-positive GPA correlates with increases of of the alternative pathway or factor C5 of the common effector path-
the IgG3 subclass of ANCA in those fractions, and not with that of way of complement did not develop lesions (Xiao et al. 2007) and
the other subclasses (Mulder et al. 1995). In addition, renal relapses antibodies against C5 were protective in this model (Huugen et al.
of GPA are particularly associated with increases of the IgG3 sub- 2007). Based on these findings, a trial with a blocking agent for
class of ANCA, although IgG1 and IgG4 subclasses of ANCA are the C5a receptor is underway in patients with ANCA-associated
present as well (Brouwer et al. 1991). These data suggest that the glomerulonephritis. These data demonstrate the pathogenic poten-
IgG3 subclass of ANCA plays an important role in neutrophil acti- tial of MPO-ANCA. A comparable approach using PR3-deficient
vation via Fcγ receptor interactions. mice did, however, not result in necrotizing systemic vasculitis
In vitro studies using endothelial monolayers have also shown (Pfister et al. 2004), although injection of PR3-ANCA did induce
that neutrophils, in the presence of ANCA, are able to adhere to mild pauci-immune proliferative glomerulonephritis and mild
and lyse endothelial cells (Ewert et  al. 1992; Savage et  al. 1992). pulmonary vasculitis in 40% of mice with a humanized immune
Elegant studies by Savage and colleagues demonstrated that ANCA system. However, granulomatous inflammation did not develop
are able to induce stable adherence of rolling neutrophils to lay- in this model (Little et  al. 2012). Granuloma formation suggests
ers expressing adhesion molecules (Radford et al. 2000). Whether the involvement of cell-mediated immunity. Indeed, T-cell reac-
endothelial cells themselves express ANCA antigens, such as PR3, tivity towards PR3, in particular a Th-17 response, was observed
has been a subject of controversy. Data from Mayet et  al. (1993, in patients with PR3-ANCA positive GPA, and renal activity was
1996) suggest that endothelial cells express PR3, particularly when reflected by the presence of effector T-cells in the urine (Abdulahad
activated, and that ANCA can bind to surface PR3 resulting in et al. 2009; Abdulahad et al. 2011). Thus, experimental data strongly
up-regulation of adhesion molecules and further cellular activation. suggest a pathogenic role for ANCA whereas the clinical expression
Others, however, have not been able to confirm PR3 expression by of ANCA-associated vasculitides requires, besides the humoral
endothelial cells (King et al. 1995), but have demonstrated that PR3 autoimmune response, a cellular response as well (Abdulahad et al.
binds to endothelial cells via a specific receptor (Taekema-Roelvink 2011; Wilde et al. 2011) (Figure 15.2).
et al. 2000).
More definite evidence for a pathophysiological role of ANCA Renal biopsy in ANCA-associated
may come from animal models. Passive transfer of ANCA in pri- (renal) vasculitides
mates or the induction of an autoimmune response to MPO in rats Before renal biopsy is considered, the involvement of the kidneys
does not result in renal lesions (Brouwer et al. 1993). When, how- and the degree of disease activity should be assessed. Tests neces-
ever, the products of activated neutrophils (lytic enzymes, MPO, sary to assess renal involvement include serum creatinine and cre-
and its substrate H2O2) are perfused into the renal artery in rats atinine clearance, urinalysis to determine if proteinuria is present,
immunized with MPO, severe pauci-immune NCGN develops and examination of urinary sediment to look for (dysmorphic)
(Brouwer et  al. 1993). These studies suggest that, initially, cati- erythrocytes and cellular casts. A renal biopsy is mandatory in case
onic proteins such as MPO and PR3 from activated neutrophils a specific diagnosis has not yet been made. Even if a diagnosis of
adhere to the glomerular capillary wall and are bound by their cog- GPA has been made, a renal biopsy is strongly advocated if the lab-
nate antibodies. The immune complexes activate the complement oratory abnormalities mentioned in section Granulomatosis with
202 SECTION 3 clinical manifestations common to vasculitis

Expansion of
Infection Teffs and Tems
Neutrophil Teffs and Tems Regulatory T cells
Release of
proinflammatory Effector T cells
cytokines Disturbed
Chemotaxis Priming Dysbalance Giant cells
function
Teffs and Tems
a ANCA Plasma cells
b
Dendritic cells
C5a, CD28null T cells
chemokines
ANCA-production Effector memory
proinflammatory T cells
cytokines Macrophage
Migration
B cells
In situ ICX l layer
formation Endothelia C5a
+ C5a receptor
complement
activation CD11b
Fc receptor
Adhesion
c Tissue inflammation molecule
HEV Proinflammatory
cytokines
ANCA
Cytokine
receptor
B cells trigger B cells trigger
PR3/MPO
DCs trigger DCs trigger
Tertiary
NET formation
lymphoid organ Granuloma

Fig. 15.2 A schematic representation of the pathogenic role of ANCA in AAV. Two pathways contributing to disease mechanisms in antineutrophil cytoplasmic
antibody (ANCA)-associated vasculitis (AAV) are depicted. (a) The ‘classic neutrophil pathway’ has been studied and confirmed by several groups. This pathway causes
necrotizing vasculitis. (b) We propose an additional ‘T-cell pathway’ that mainly causes granulomatous inflammation and promotes necrotizing vasculitis. Infections are
the starting point of both pathways; infections trigger priming of neutrophils (a), up-regulation of adhesion molecules on endothelial cells, and expansion of circulating
effector T cells (b). Primed neutrophils show increased surface expression of ANCA antigens and adhesion molecules. ANCA binding activates the neutrophil in the
following ways: (1) enhancing vessel wall adherence and transmigration capacity; (2) production and release of oxygen radicals; and (3) degranulation and release of
enzymes including myeloperoxidase (MPO) and proteinase-3 (PR3) (a). Transient immune complexes are formed locally by binding of ANCA to PR3/MPO sticking to
endothelial cells. Subsequently, complement is activated, which further promotes neutrophil degranulation. This all adds to the development of necrotizing vasculitis.
Whether this specific cascade is also applicable to disease pathogenesis in ANCA-negative AAV patients remains unclear. The expanded effector memory T cells (Tems)
are not sufficiently regulated by regulatory T cells (Tregs, b). This leads to dysbalance in the homeostasis of Tregs and Tems, resulting in further release of proinflammatory
cytokines promoting neutrophil priming (a); moreover, ANCA production is enhanced by further T-cell/B-cell interaction. (c) Expanded circulating Tems migrate into
target organs such as the lungs or the kidney. Within tissues, Tems drive granuloma formation, which is considered an ‘executioner’ of tissue destruction. Granulomas
are composed of numerous cell types such as T cells, B cells, giant cells, and dendritic cells (DCs). Moreover, ANCA production occurs in granulomas. Possibly, tertiary
lymphoid organs (TLOs) are ‘local controllers’ of tissue inflammation, as induction of Tregs is thought to take place in TLOs. Neutrophil extracellular trap (NET) formation
occurs in lesions as a consequence of neutrophil apoptosis and degranulation. DNA and serine proteases are deployed in these NETs. NET-derived products activate
DCs and B cells by sensing via Toll-like receptors (TLRs). Interferon (IFN-α) production by DCs might have an impact on local immune regulation; it has been shown
to impair Tregs in function. Although major efforts were made to unravel the pathogenesis of AAV, some missing links remain. The origin of ANCA is unexplained so
far. If and how microbial agents and/or T-cell dysregulation finally lead to the development of ANCA needs to be investigated further. HEV, high endothelial venules;
ICX, immunocomplexes. Reproduced with permission from Wilde, B., van Paassen, P., Witzke, O., Cohen Tervaert, J.W. New pathophysiological insights and treatment of
ANCA-associated vasculitis. Kidney International 2011, 79, 599–612. © Nature Publishing Group

Polyangiitis are present, in order to look for the vasculitis, to assess patients, particularly adults, renal lesions may progress to end-stage
activity and damage due to scarring. For treatment recommenda- renal disease (Fogazzi et al. 1989). The diagnosis of IgA vasculitis is
tions, see the chapters dealing with specific disease. made on clinical grounds together with characteristic small-vessel
vasculitis with IgA1-dominant immune deposits. In the circulation,
Other forms of small-vessel vasculitis IgA immune complexes of intermediate to large size can be detected
with renal involvement (Hilhorst et  al. 2011). The classification criteria of the American
IgA vasculitis (formerly Henoch–Schönlein purpura) was defined College of Rheumatology, to be applied for the classification of
by the Chapel Hill Consensus Conference (Jennette et al. 2013) as patients with already proven vasculitis, include palpable purpura,
vasculitis with IgA1-dominant immune deposits affecting small an age of less than 20 years at onset, bowel angina, and wall granu-
vessels, in other words capillaries, venules, or arterioles. Renal locytes on biopsy (Mills et al. 1990), but, remarkably, do not include
involvement has been reported in 33% of children with IgA vascu- IgA deposits.
litis and in 63% of adults (Rieu and Noel 1999). The disease is usu- Renal involvement is characterized by granular mesangial IgA
ally self-limiting and without serious sequela, but in a minority of deposits, frequently in combination with complement C3 and
CHAPTER 15 renal manifestations 203

(a) (b)

Fig. 15.3 Nephritis in IgA vasculitis. (a) Renal biopsy of a 45-year-old female with recurrent cutaneous vasculitis and episodes of gastrointestinal vasculitis of 2 years
duration, which was taken because of recent onset erythrocyturia and severe proteinuria. Renal biopsy shows a focal and segmental extracapillary crescentic
glomerulonephritis of recent origin. Silver methenamine, H&E ×50. (b) Staining with monoclonal anti-IgA1 antibodies shows diffuse glomerular, granular, deposits in the
mesangia and locally in the capillary wall. Insert: staining for C3 shows a similar pattern. Immunofluorescence, anti-IgA, ×125.

fibrinogen (Figure  15.3). IgA deposits are predominantly of the (Dammacco and Sansonno 1997). This is, however, only true in
IgA1 subclass, the IgA is polymeric, and both light chains are pre- countries with a high prevalence of hepatitis C, whereas in coun-
sent. In view of the relationship between upper respiratory tract tries with a low prevalence of hepatitis C, type II cryoglobulinae-
infections and relapses of IgA vasculitis, it has been suggested that mic vasculitis is really a primary form of vasculitis (really essential
dysregulation in IgA-class immune responsiveness is basic to the mixed cryoglobulinaemia) (Cohen Tervaert et al. 2007).
pathogenesis of IgA vasculitis. The pathogenesis of IgA vasculitis Cryoglobulinaemic vasculitis is a prototype of an immune com-
has not yet been fully elucidated (Feehally and Allen 1999). Varying plex mediated vasculitis, in which circulating cryoglobulins are
degrees of glomerular injury are present; in some cases, there are demonstrable in serum, complement levels C3 and, particularly, C4
only minimal abnormalities at light microscopy in the presence of are decreased, and immune deposits are present in the vessel walls
typical IF findings (Heaton et al. 1977; Meadow et al. 1972; Davin, (Gorevic et al. 1980) (Figures 15.4). The kidneys are involved in
2011). Mesangial proliferation is present in 25% of biopsies without 25–50% of the patients. Cryoglobulinaemic membranoproliferative
endocapillary proliferation and crescent formation. The remaining glomerulonephritis is particularly prevalent in patients with type
70–75% show more extensive mesangial proliferation with focal to II mixed cryoglobulins (D’Amico et al. 1989). By light microscopy,
diffuse endocapillary proliferation accompanied by crescents and eosinophilic deposits are visible within the glomerular capillaries,
membranoproliferative glomerulonephritis. Results of histopatho- together with massive infiltration of monocytes and polymorpho-
logical examination of renal lesions in IgA vasculitis are important nuclear leukocytes. Extracapillary proliferation may occur. Direct
as the severity of the lesions is a major prognostic determinant. immunofluorescence reveals deposits of IgM, IgG, C1q, and C3
Large proportions of glomeruli with crescents correlate with (D’Amico et  al. 1989); vasculitis of the small and medium-sized
increased risk for developing end-stage renal disease (Yoshikawa renal arteries may be present.
et al. 1981). The clinical picture is characterized by an insidious onset, a
Clinically, permanent renal impairment does not develop in case slowly progressive course, and some degree of proteinuria, haema-
of normal urinalysis during the first 6 months after presentation as turia, hypertension, and renal insufficiency. Nephrotic syndrome
deduced from a systemic review covering 12 studies of 1133 chil- develops in a minority of patients. In 25–30% of patients, a more
dren. It occurred in 1.6% of patients with isolated urinary abnor- acute presentation occurs with acute renal insufficiency together
malities during this period, and in 19.5% of patients who developed with proteinuria, haematuria, and hypertension. Oliguria or anuria
a nephritic or nephrotic syndrome (Narchi 2005). Another study is rare.
also showed that proteinuria at 1-year follow-up and advanced his-
tological findings in the renal biopsy are the most discriminative
factors for a poor prognosis (Edström Halling et al. 2010).
Conclusion
In the large-vessel vasculitides, renal involvement is relatively rare.
Vasculitis of medium-sized arteries may affect the kidneys at the
Cryoglobulinaemic renal vasculitis level of the renal artery and its bifurcations. When the glomeru-
In the Chapel Hill Consensus Conference, cryoglobulinaemic vas- lar capillaries are involved, vasculitis is classified as small-vessel
culitis was classified as one of the primary vasculitides (Jennette vasculitis. The group of small-vessel ANCA-associated diseases
et al. 2013). Many cases of type II and III cryoglobulinaemic vascu- frequently shows renal involvement, which is a major factor in out-
litis are related to infection with hepatitis C virus (HCV) (Johnson come. Kidney disease is also frequent in IgA vasculitis and cryo-
et al. 1994; Misiani et al. 1992) and HCV-related proteins or RNA globulinaemic vasculitis. Classification of the renal vasculitides is
are present not only in the cryoglobulins but also in the vessel wall important for prognosis and treatment.
204 SECTION 3 clinical manifestations common to vasculitis

(a) (b)

Fig. 15.4 Essential type 2 cryoglobulinaemia. (a) Diffuse proliferation of mesangial cells and subendothelial deposition of eosinophilic material with or without mesangial (matrix)
interposition is shown. Silver methenamine, H&E ×125. (b) Anti C3 polyclonal antibodies. Diffuse glomerular granular deposition of immune complexes consisting of IgG, IgM, and
Clq (not shown) as well as C3 in the mesangial area is seen as well as subendothelial deposition and occasionally intracapillary aggregates. Immunofluorescence, ×125.

Brouwer, E., Huitema, M.G., Klok, P.A., Cohen Tervaert, J.W., Weening, J.J.,
Acknowledgements and Kallenberg, C.G.M. (1993). Anti-myeloperoxidase associated pro-
The authors acknowledge Professor van Breda Vriesman, and Henk liferative glomerulonephritis: an animal model. Journal of Experimental
van Rie (both Department of Clinical Immunology, University Medicine, 177, 905–14.
Hospital Maastricht, Maastricht, The Netherlands) for providing Brouwer, E., Huitema, M.G., Mulder, A.H.L., Heeringa, P., van Goor, H.,
the figures of renal biopsies. Cohen Tervaert, J.W., Weening, J.J., and Kallenberg, C.G.M. (1994).
Neutrophil activation in vitro and in vivo in Wegener’s granulomatosis.
Kidney International, 45, 1120–31.
References Cavatorta, F., Campisi, S., Trabassi, E., Zollo, A., and Salvidio, G. (1995). IgA
Abdulahad, W.H., Kallenberg, C.G., Limburg, P.C., and Stegeman, C.A. nephropathy associated with Takayasu’s arteritis: report of a case and
(2009). Urinary CD4+ effector memory T cells reflect renal disease review of the literature. American Journal of Nephrology, 15, 165–7.
activity in antineutrophil cytoplasmic antibody-associated vasculitis. Chandrakantan, A. and Kaufman, J. (1999). Renal hemorrhage in polyarte-
Arthritis and Rheumatism, 60, 2830–8. ritis nodosa: diagnosis and management. American Journal of Kidney
Abdulahad, W.H., Lamprecht, P., Kallenberg, C.G. (2011). T-helper cells as Diseases, 33, e8.
new players in ANCA-associated vasculitides. Arthritis Research and Chang, D.Y., Wu, L.H., Liu, G., Chen, M., Kallenberg, C.G., and Zhao,
Therapy, 13, 236. M.H. (2012). Re-evaluation of the histopathologic classification of
Andrassy, K., Waldherr, R., Erb, A., and Ritz, E. (1992). De novo glomerulo- ANCA-associated glomerulonephritis: a study of 121 patients in a single
nephritis in patients during remission from Wegener’s granulomatosis. center. Nephrology Dialysis and Transplantation, 27, 2343–9.
Clinical Nephrology, 38, 295–8. Chen, M. and Kallenberg, C.G. (2010). ANCA-associated vascu-
Bansal, P.J. and Tobin, M.C. (2004). Neonatal microscopic polyangiitis sec- litides—advances in pathogenesis and treatment. Nature Reviews in
ondary to transfer of maternal myeloperoxidase-antineutrophil cytoplas- Rheumatology, 6, 653–64.
mic antibody resulting in neonatal pulmonary hemorrhage and renal Cid, M.C., Prieto-González, S., Arguis, P., Espígol-Frigolé, G., Butjosa, M.,
involvement. Annals of Allergy Asthma and Immunology, 93, 398–401. Hernández-Rodríguez, J., Segarra, M., Lozano, E., and García-Martínez,
Berden, A.E., Ferrario, F., Hagen, E.C., Jayne, D.R., Jennette, J.C., Joh, K., A. (2009). The spectrum of vascular involvement in giant-cell arteri-
Neumann, I., Noël, L.H., Pusey, C.D., Waldherr, R., Bruijn, J.A., and Bajema, tis: clinical consequences of detrimental vascular remodelling at differ-
I.M. (2010). Histopathologic classification of ANCA-associated glomerulo- ent sites. APMIS (Suppl). 127, 10–20.
nephritis. Journal of the American Society of Nephrology, 21, 1628–36. Clutterbuck, E.J., Evans, D.J., and Pusey, C.D. (1990). Renal involvement in
Boomsma, M.M., Stegeman, C.A., van der Leij, M.J., Oost, W., Herman, S.J., Churg-Strauss syndrome. Nephrology, Dialysis, Transplantation, 5, 161–7.
Kallenberg, C.G.M., Limburg, P.C., and Cohen Tervaert, J.W. (2000). Cohen, P., Guillevin, L., Baril, L., Lhote, F., Noel, L.H., and Lesavre, P. (1995).
Prediction of relapses in Wegener’s granulomatosis by measurement Persistence of antineutrophil cytoplasmic antibodies (ANCA) in asymp-
of anti neutrophil cytoplasmic antibody levels; a prospective study. tomatic patients with systemic polyarteritis nodosa or Churg-Strauss
Arthritis and Rheumatism, 43, 2025–33. syndrome:  follow up of 53 patients. Clinical and Experimental
Booth, A.D., Almond, M.K., Burns, A., Ellis, P., Gaskin, G., Neild, Rheumatology, 13, 193–8.
G.H., Plaisance, M., Pusey, C.D., Jayne, D.R. (2003). Outcome of Cohen, R.D., Conn, D.L., and Ilstrup, D.M. (1980). Clinical features, progno-
ANCA-associated renal vasculitis: a 5-year retrospective study. American sis and response to treatment in polyarteritis. Mayo Clinic Proceedings,
Journal of Kidney Diseases, 41, 776–84. 55, 146–55.
Brogan, P.A., Davies, R., Gordon, I., Dillon, M.J. (2002). Renal angiography Cohen Tervaert, J.W., Goldschemeding, R., Elema, J.D., Borne, A.E.G. Kr von
in children with polyarteritis nodosa. Pediatric Nephrology, 17, 277–83. dem, and Kallenberg, C.G.M. (1991a). Antimyeloperoxidase antibodies
Brouwer, E., Cohen Tervaert, J.W., Horst, G., Huitema, M.C., Giessen, M. van in the Churg Strauss Syndrome. Thorax, 46, 70–1.
der, Limburg, P.C. and Kalleberg, C.G.M. (1991). Predominance of IgG4 Cohen Tervaert, J.W., Huitema, M.G., Hené, R.J., Sluiter, W.J., The, T.H., van
subclass of anti-neutrophil cytoplasmic autoantibodies in patients with der Hem, G.K., and Kallenberg, C.G.M. (1990). Prevention of relapses
Wegener’s Granulomatosis and clinically related disorders. Clinical and in Wegener’s granulomatosis by treatment based on antineutrophil cyto-
Experimental Immunology, 83, 379–86. plasmic antibody titre. Lancet, 336, 709–11.
CHAPTER 15 renal manifestations 205

Cohen Tervaert, J.W., Limburg, P.C., Elema, J.D., Huitema, M.G., Horst, G., Franssen, C.F.M., Gans, R., Kallenberg, C.G.M., Hageluken, C., and Hoorntje,
The, T.H., and Kallenberg, C.G.M. (1991b). Detection of autoantibodies S.J. (1998). Disease spectrum of patients with antineutrophil cytoplas-
against myeloid lysosomal enzymes: a useful adjunct to classification of mic autoantibodies of defined specificities: distinct differences between
patients with biopsy-proven necrotizing arteritis. American Journal of patients with anti-proteinase 3 and anti-myeloperoxidase autoantibod-
Medicine, 91, 59–66. ies. Journal of Internal Medicine, 244, 209–16.
Cohen Tervaert, J.W., van Paassen, P., and Damoiseaux, J. (2007). Type II Franssen, C.F.M., Huitema, M.G., Muller Kobold, A.C., Oost-Kort, W.,
cryoglobulinemia is not associated with hepatitis C infection: the Dutch Limburg, P.C., Tiebosch, A., Stegeman, C.A., Kallenberg, C.G.M., and
experience. Annals of the New York Academy of Sciences, 1107, 251–8. Tervaert, J.W. (1999). In vitro neutrophil activation by antibodies to pro-
Cohen Tervaert, J.W., Woude, F.J. van der, Fauci, A.S., Ambrus, J.L., Velosa, teinase 3 and myeloperoxidase from patients with crescentic glomeru-
J., Keane, W.F., Meijer, S., Giessen, M. van der, The, T.H., Hem, G.K., lonephritis. Journal of the American Society of Nephrology, 10, 1506–15.
van der and Kallenberg, C.G.M. (1989). Association between active Franssen, C.F.M., Stegeman, C.A., Kallenberg, C.G.M., Gans, R.O.B., de Jong,
Wegener’s Granulomatosis and anticytoplasmic antibodies. Archives of P.E., Hoorntje, S.J., and Cohen Tervaert, J.W. (2000). Antiproteinase
Internal Medicine, 149, 2461–5. 3-and anti-myeloperoxidase-associated vasculitis. Kidney International,
D’Amico, G., Colasanti, G., Ferrario, F., and Sinico, R.A. (1989). Renal 57, 2195–206.
involvement in essential mixed cryoglobulinemia. Kidney International, Gaskin, G., Clutterbuck, E.J., and Pusey, C.D. (1991). Renal disease in the
35, 1004–14. Churg-Strauss syndrome. Contributions to Nephrology, 94, 58–65.
Dammacco, F. and Sansonno, D. (1997). Mixed cryoglobulinemia as a model Goldschmeding, R., van der Schoot, C.E., ten Bokkel Huinink, D., Hack, C.E.,
of systemic vasculitis. Clinical Reviews in Allergy and Immunology, 15, van den Ende, M.E., Kallenberg, C.G.M., and von dem Borne, A.E.G.Kr.
97–119. (1989). Wegener’s Granulomatosis autoantibodies identify a novel
Davin, J.C. (2011). Henoch-Schonlein purpura nephritis: pathophysiology, diisopropylfluorophosphate-binding protein in the lysosomes of normal
treatment, and future strategy. Clinical Journal of the American Society human neutrophils. Journal of Clinical Investigation, 84, 1577–87.
of Nephrology, 6, 679–89. Gorevic, P.D., Kassab, H.J., Levo, Y., Kohn, R., Meltzer, M., Prose, P., and
de Lind van Wijngaarden, R.A., Hauer, H.A., Wolterbeek, R., Jayne, D.R., Franklin, E.C. (1980). Mixed cryoglobulinemia:  clinical aspects and
Gaskin, G., Rasmussen, N., Noël, L.H., Ferrario, F., Waldherr, R., Hagen, long-term follow-up of 40 patients. American Journal of Medicine,
E.C., Bruijn, J.A., and Bajema, I.M. (2006). Clinical and histologic deter- 69, 287–308.
minants of renal outcome in ANCA-associated vasculitis: A prospective Ham, S.W., Kumar, S.R., Wang, B.R., Rowe, V.L., and Weaver, F.A. (2010).
analysis of 100 patients with severe renal involvement. Journal of the Late outcomes of endovascular and open revascularization for nonath-
American Society of Nephrology 17, 2264–74. erosclerotic renal artery disease. Archives of Surgery, 145, 832–9.
Edström Halling, S., Söderberg, M.P., and Berg, U.B. (2010). Predictors of Heaton, J.M., Turner, D.R., and Cameron, J.S. (1977). Localization of glo-
outcome in Henoch-Schönlein nephritis. Pediatric Nephrology, 25, merular ‘deposits’ in Henoch-Schönlein nephritis. Histopathology, 1,
1101–8. 93–104.
Egner, W. and Chapel, H.M. (1990). Titration of antibodies against neutro- Hilhorst, M., van Paassen, P., van Breda Vriesman, P., and Cohen Tervaert,
phil cytoplasmic antigens is useful in monitoring disease activity in sys- J.W. (2011). Immune complexes in acute adult-onset Henoch-Schonlein
temic vasculitides. Clinical and Experimental Immunology, 82, 244–9. nephritis. Nephrology Dialysis and Transplantation, 26, 3960–7.
Escriba, A., Morales, E., Albizua, E., Herrero, J.C., Ortuno, T., Carreno, A., Hilhorst, M., Wilde, B., van Paassen, P., Winkens, B., van Breda Vriesman,
Dominguez-Gil, B., and Praga, M. (2000). Secondary (AA-type) amy- P., Cohen Tervaert, J.W.:  Limburg Renal Registry. (2013). Improved
loidosis in patients with polymyalgia rheumatica. American Journal of outcome in anti-neutrophil cytoplasmic antibody (ANCA)-associated
Kidney Diseases, 35, 137–40. glomerulonephritis:  a 30-year follow-up study. Nephrology Dialysis
Eustace, J.A., Nadasdy, T., and Choi, M. (1999). The Churg Strauss syndrome. Transplantation, 28, 373–9.
Journal of the American Society of Nephrology, 10, 2048–55. Heeringa, P., Brouwer, E., Klok, P.A., Huitema, M.G., Born, J. van den, Weening,
Ewert, B.H., Jennette, J.C., and Falk, R.J. (1992). Anti-myeloperoxidase anti- J.J., and Kallenberg, C.G.M. (1996). Autoantibodies to myeloperoxidase
bodies stimulate neutrophils to damage human endothelial cells. Kidney aggravate mild anti-glomerular-basement-membrane-mediated glo-
International, 41, 375–83. merular injury in the rat. American Journal of Pathology, 149, 1695–706.
Falk, R.J., Terrell, R.S., Charles, L.A., and Jennette, J.C. (1990). Anti-neutrophil Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., et al. (1992). Wegener’s granulomato-
cytoplasmic autoantibodies induce neutrophils to degranulate and pro- sis: an analysis of 158 patients. Annals of Internal Medicine, 116, 488–98.
duce oxygen radicals in vitro. Proceedings of the National Academy of Huugen, D., van Esch, A., Xiao, H., Peutz-Kootstra, C.J., Buurman, W.A.,
Sciences of the United States of America, 87, 4115–9. Tervaert, J.W., Jennette, J.C., Heeringa P. (2007). Inhibition of comple-
Fauci, A., Haynes, B.F., Katz, P., and Wolff, S. (1983). Wegener’s granulomato- ment factor C5 protects against anti-myeloperoxidase antibody-mediated
sis: prospective clinical and therapeutic experience with 85 patients for glomerulonephritis in mice. Kidney International, 71, 646–54.
21 years. Annals of Internal Medicine, 98, 76–85. Huugen, D., Xiao, H, van Esch, A., Falk, R.J., Peutz-Kootstra, C.J., Buurman,
Feehally, J. and Allen, A.C. (1999). Pathogenesis of IgA nephropathy. Annals W.A., Tervaert, J.W., Jennette, J.C., and Heeringa, P. (2005). Aggravation
Medicine Interne (Paris), 150, 91–8. of anti-myeloperoxidase antibody-induced glomerulonephritis by bac-
Finkielman, J.D., Merkel, P.A., Schroeder, D., Hoffman, G.S., Spiera, R., St. terial lipopolysaccharide: role of tumor necrosis factor-alpha. American
Clair, E.W., Davis, J.C.Jr, McCune, W.J., Lears, A.K., Ytterberg, S.R., Journal of Pathology, 167, 47–58.
Hummel, A.M., Viss, M.A., Peikert, T., Stone, J.H., and Specks, U. Jayne, D.R.W., Gaskin, G., Pusey, C.D., and Lockwood, C.M. (1995). ANCA
(2007). Antiproteinase 3 antineutrophil cytoplasmic antibodies and dis- and predicting relapse in systemic vasculitis. Quarterly Journal of
ease activity in Wegener’s granulomatosis. Annals of Internal Medicine, Medicine, 88, 127–33.
147, 611–9. Jennette, J.C. (1991). Antineutrophil cytoplasmic autoantibody-associated
Fogazzi, G.B., Pasquali, S., Moriggi, M., Casanova, S., Damilano, I., Mihatsch, disease: a pathologist’s perspective. American Journal of Kidney Diseases,
M.J., Zuccheli, P., and Ponticelli, C. (1989). Long-term outcome of 18, 164–70.
Schönlein-Henoch nephritis in the adult. Clinical Nephrology, 31, 60–6. Jennette, J.C. (1998). Crescentic glomerulonephritis. In Heptinstall’s pathol-
Franssen, C.F.M., Gans, R.O.B., Arends, B., Hageluken, C., Wee, P.M. ogy of the Kidney (ed. J.C. Jennette, J.L. Olson, M.M., Schwartz, and F.G.
ter, Gerlag, P.G.G., and Hoorntje, S.J. (1995). Differences between Silva), pp. 625–56. Lippincott-Raven, Philadelphia.
anti-myeloperoxidase and anti-proteinase 3 associated renal disease. Jennette, J.C. and Falk, R.J. (1994). The pathology of vasculitis involving the
Kidney International, 47, 193–9. kidney. American Journal of Kidney Diseases, 24, 130–41.
206 SECTION 3 clinical manifestations common to vasculitis

Jennette, J., Falk, R., Bacon, P., Basu, N., Cid, M., Ferrario, F., Flores-Suarez, L., K.G. (2012). Genetically distinct subsets within ANCA-associated vas-
Gross, W., Guillevin, L., Hagen, E., Hoffman, G., Jayne, D., Kallenberg, culitis. New England Journal of Medicine, 367, 214–23.
C., Lamprecht, P., Langford, C., Luqmani, R., Mahr, A., Matteson, E., Mayet, W.J., Csernok, E., Szymkowiak, C., Gross, W.L., and Meyer zum
Merkel, P., Ozen, S., Pusey, C., Rasmussen, N., Rees, A., Scott, D., Specks, Büschenfelde, K.H. (1993). Human endothelial cells express protein-
U., Stone, J., Takahashi, K., Watts, R. (2013). 2012 Revised International ase 3, the target antigen of anti-cytoplasmic antibodies in Wegener’s
Chapel Hill Consensus Conference Nomenclature of Vasculitides. Granulomatosis. Blood, 82, 1221–9.
Arthritis and Rheumatism, 65, 1–11. Mayet, W.J., Schwarting, A., Orth, T., Duchmann, R., and Meyer zum
Jennette, J.C., Wilkman, A.S., and Falk, R.J. (1989). Anti-neutrophil cyto- Büschenfelde, K.H. (1996). Antibodies to proteinase 3 mediate expres-
plasmic autoantibody-associated glomerulonephritis and vasculitis. sion of vascular cell adhesion molecule-1 (VCAM-1). Clinical and
American Journal of Pathology, 135, 921–30. Experimental Immunology, 103, 259–67.
Joh, K., Kanetsuna, Y., Ishikawa, Y., Aizawa, S., Naito, L., and Sado, Y. (1997). Meadow, S.R., Glasgow, E.F., White, R.H., Moncrieff, M.W., Cameron, J.S.,
Diffuse mesangial sclerosis associated with Kawasaki disease: analysis of and Ogg, C.S. (1972). Schönlein-Henoch nephritis. Quarterly Journal of
alpha chains (alpha 1-alpha 6) of human type 1 in the renal basement Medicine, 41, 241–58.
membrane. Virchows Archiv, 430, 489–94. Mills, J.A., Michel, B.A., Bloch, D.A., et al. (1990). The American College of
Johnson, R.J., Willson, R., and Yamabe, K. (1994). Renal manifestations of Rheumatology 1990 criteria for the classification of Henoch-Schonlein
hepatitis C virus infection. Kidney International, 46, 1255. Purpura. Arthritis and Rheumatism, 33, 1114–21.
Kallenberg, C.G.M. (2005). Churg Strauss Syndrome: just one disease entity? Misiani, R., Bellavita, P., Fenili, D., et al. (1992). Hepatitis C virus infection
Arthritis and Rheumatism, 52, 2589–93. in patients with essential mixed cryoglobulinemia. Annals of Internal
Kallenberg, C.G.M., Brouwer, E., Weening, J.J., and Cohen Tervaert, J.W. Medicine, 117, 573–7.
(1994). Anti-neutrophil cytoplasmic antibodies: current diagnostic and Mulder, A.H., Stegeman, C.A., and Kallenberg, C.G.M. (1995). Activation
pathophysiological potential. Kidney International, 46, 1–15. of granulocytes by anti-neutrophil cytoplasmic antibodies (ANCA) in
Kerr, G.R., Fleischer, T.H.A., Hallahan, C.D., et al. (1993). Limited prognostic Wegener’s granulomatosis: a predominant role for the IgG3 subclass of
value of changes in antineutrophil cytoplasmic antibody titer in patients ANCA. Clinical and Experimental Immunology, 101, 227–32.
with Wegener’s granulomatosis. Arthritis and Rheumatism, 36, 365–71. Muller, E., Schneider, W., Kettritz, U., Schmidt, W.A., Luft, F.C., and Gobel,
Kieffer, E., Piquois, A., Bertal, A., Bletry, O., and Godeau, P. (1990). U. (2004). Temporal arteritis with pauci-immune glomerulonephritis: a
Reconstructive surgery of the renal arteries in Takayasu’s disease. Annals systemic disease. Clinical Nephrology, 62, 384–6.
of Vascular Surgery, 4, 156–65. Narchi, H. (2005). Risk of long term renal impairment and duration of
King, W.J., Adu, D., Daha, M.R., et  al. (1995). Endothelial cells and renal follow-up recommended for Henoch-Schönlein purpura with normal
epithelial cells do not express the Wegener’s autoantigen, proteinase 3. or minimal urinary findings: a systematic review. Archives of Diseases in
Clinical and Experimental Immunology, 102, 98–105. Children, 90, 916–20.
Klein, R.G., Hunder, G.G., Stanson, A.W., and Sheps, S.C. (1975). Large Nardi, P.M., Haller, J.O., Friedman, A.P., Slovis, T.L., and Schaffer, R.M.
artery involvement in giant cell (temporal) arteritis. Annals of Internal (1985). Renal manifestations of Kawasaki’s disease. Pediatric Radiology,
Medicine, 83, 806. 15, 116–18.
Krug, P., Boyer, O., Balzamo, E., Sidi, D., Lehnert, A., and Niaudet, P. (2012). O’Neill, Jr W.M., Hammar, S.A., and Bloomer, H.A. (1976). Giant-cell arteri-
Nephrotic syndrome in Kawasaki disease:  a report of three cases. tis with visceral angiitis. Archives of Internal Medicine, 136, 1157.
Pediatric Nephrology, 9, 1547–50 Pfister, H., Ollert, M., Frohlich, L.F., Quintanilla-Martinez, L., Colby, T.V.,
Lagneau, P. and Michel, J.B. (1985). Renovascular hypertension and Specks, U., and Jenne, D.E. (2004). Antineutrophil cytoplasmic autoan-
Takayasu’s disease. Journal of Urology, 134, 876. tibodies against the murine homolog of proteinase 3 (Wegener autoanti-
Lanham, J.G., Elkon, K.B., Pusey, C.D., and Hughes, G.R. (1984). Systemic gen) are pathogenic in vivo. Blood, 104, 1411–8.
vasculitis with asthma and eosinophilia:  a clinical approach to the Porges, A.J., Redecha, P.B., Kimberly, W.T., Csernok, E., Gross, W.L.,
Churg-Strauss syndrome. Medicine, 63, 65–81. and Kimberly, R.P. (1994). Anti-neutrophil cytoplasmic antibodies
Lionaki, S., Blyth, E.R., Hogan, S.L., Hu, Y., Senior, B.A., Jennette, C.E., engage and activate human neutrophils via Fc gamma RIIa. Journal of
Nachman, P.H., Jennette, J.C., and Falk, R.J. (2012). Classification of Immunology, 153, 1271–80.
antineutrophil cytoplasmic autoantibody vasculitides: the role of anti- Radford, D.J., Savage, C.O.S., and Nash, G.B. (2000). Treatment of rolling
neutrophil cytoplasmic autoantibody specificity for myeloperoxidase neutrophils with antineutrophil cytoplasmic antibodies causes conver-
or proteinase 3 in disease recognition and prognosis. Arthritis and sion to firm integrin-mediated adhesion. Arthritis and Rheumatism, 43,
Rheumatism, 64, 3452–62. 1337–45.
Little, M.A., Al-Ani, B., Ren, S., Al-Nuaimi, H., Leite, M., Jr, Alpers, C.E., Rarok, A.A., Stegeman, C.A., Limburg, P.C., and Kallenberg, C.G.M. (2002).
Savage, C.O., and Duffield, J.S. (2012). Anti-proteinase 3 anti-neutrophil Neutrophil membrane expression of proteinase 3 is related to relapse
cytoplasm autoantibodies recapitulate systemic vasculitis in mice with a in PR3-ANCA-associated vasculitis. Journal of the American Society of
humanized immune system. PLoS One, 7, e28626. Nephrology, 13, 2232–8.
Logar, D., Rozman, B., Vizjak, A., Ferluga, D., Mulder, A.H.L., and Kallenberg, Reinhold-Keller, E., Beuge, N., Latza, U., de Groot, K., Rudert, H., Nolle, B.,
C.G.M. (1994). Arteritis of both internal carotid arteries in a patient Heller, M., and Gross, W.L. (2000). An interdisciplinary approach to the
with focal crescentic glomerulonephritis and antineutrophil cytoplas- care of patients with Wegener’s granulomatosis: long-term outcome in
mic antibodies (c-ANCA). British Journal of Rheumatology, 33, 167–9. 155 patients. Arthritis and Rheumatism, 43, 1021–32.
Lovaria, A., Nicolini, A., Meregaglia, D., Saccheri, S., Rivolta, R., Montanari, Reumaux, D., Vossebeld, P.J., Roos, D., and Verhoeven, A.J. (1995). Effect of
E., Morganti, A., and Rossi, P. (1999). Interventional radiology in the tumor necrosis factor-induced integrin activation on Fc gamma recep-
treatment of renal artery stenosis. Annals of Urology, 33, 146–55. tor II-mediated signal transduction: relevance for activation of neutro-
Lyons, P.A., Rayner, T.F., Trivedi, S., Holle, J.U., Watts, R.A., Jayne, D.R., phils by anti-proteinase 3 or anti-myeloperoxidase antibodies. Blood, 86,
Baslund, B., Brenchley, P., Bruchfeld, A, Chaudhry, A.N., Cohen Tervaert, 3189–95.
J.W., Deloukas, P., Feighery, C., Gross, W.L., Guillevin, L., Gunnarsson, Rieu, P. and Noel, L.H. (1999). Henoch/Schönlein nephritis in children and
I., Harper, L., Hrušková, Z., Little, M.A., Martorana, D., Neumann, adults:  morphological features and clinico-pathological correlations.
T., Ohlsson, S., Padmanabhan, S., Pusey, C.D., Salama, A.D., Sanders, Annals Medicine Interne (Paris), 150, 151–9.
J,S., Savage, C.O., Segelmark, M., Stegeman, C.A., Tesař, V., Vaglio, A., Ronco, P., Verroust, P., Mignon, F., et al. (1983). Immunopathologic stud-
Wieczorek, S., Wilde, B., Zwerina, J., Rees, A.J., Clayton, D.G., and Smith, ies of polyarteritis nodosa and Wegener’s granulomatosis: A report of
CHAPTER 15 renal manifestations 207

43 patients with 51 renal biopsies. Quarterly Journal of Medicine, 52, Taekema-Roelvink, M.E., Van Kooten, C., Heemskerk, E., Schroeijers, W.,
212–23. and Daha, M.R. (2000). Proteinase 3 interacts with a 111-KD mem-
Roth, A.J., Ooi, J.D., Hess, J.J., et al. (2013). Epitope specificity determines brane molecule of human umbilical vein endothelial cells. Journal of the
pathogenicity and detectability in ANCA-associated vasculitis. Journal American Society of Nephrology, 11, 640–8.
of Clinical Investigation, 123, 1773–83. Tomasson, G., Grayson, P.C., Mahr, A.D., Lavalley, M., and Merkel, P.A.
Rowley, A.H., Shulman, S.T., Mask, C.A., Finn, L.S., Terai, M., Baker, S.C., (2012). Value of ANCA measurements during remission to predict a
Galliani, C.A., Takahashi, K., Naoe, S., Kalelkar, M.B., and Crawford, relapse of ANCA-associated vasculitis—a meta-analysis. Rheumatology
S.E. (2000). IgA plasma cell infiltration of proximal respiratory tract, (Oxford), 51, 100–9.
pancreas, kidney, and coronary artery in acute kawasaki disease. Journal van der Woude, F.J., Rasmussen, N., Lobatto, S., Wiik, A., Permin, H., van
of Infectious Diseases, 182, 1183–91. Es, L.A., van der Giessen, M., van der Hem, G.K., and The, T.H. (1985).
Salcedo, J.R., Greenberg, L., and Kapur, S. (1988). Renal histology of mucocu- Autoantibodies to neutrophils and monocytes:  tool for diagnosis and
taneous lymph node syndrome (Kawasaki disease). Clinical Nephrology, marker of disease activity in Wegener’s Granulomatosis. Lancet, ii, 425–9.
29, 47–51. van Rossum, A.P., Rarok, A.A., Huitema, M.G., Fassina, G., Limburg, P.C.,
Savage, C.O., Pottinger, B.E., Gaskin, G., Pusey, C.D., and Pearson, J.D. and Kallenberg, C.G.M. (2004). Constitutive membrane expression of
(1992). Autoantibodies developing to myeloperoxidase and proteinase 3 proteinase 3 and neutrophil activation by anti-PR3 antibodies. Journal
in systemic vasculitis stimulate neutrophil cytotoxicity toward cultured of Leukocyte Biology, 76, 1162–70.
endothelial cells. American Journal of Pathology, 141, 335–42. Veiga, P.A., Pieroni, D., Baier, W., and Feld, L.G. (1992). Association of
Schouffoer, A.A., Siegert, C.E., Arend, S.M., Thompson, J., and van Oostaijen, Kawasaki disease and interstitial nephritis. Pediatric Nephrology, 6, 421–3.
J.A. (1998). Embolization of a ruptured aneurysm in classic polyarteritis Wada, Y., Nishida, H., Kohno, K., Tamai, O., Fujisawa, M., Katoh, S.,
presenting as perirenal hematoma. Archives of Internal Medicine, 158, Morimatsu, M., and Okuda, S. (1999). AA amyloidosis in Takayasu’s
1466–8. arteritis-long-term survival on maintenance haemodialysis. Nephrology
Schreiber, A., Busjahn, A., Luft, F.C., and Kettritz, R. (2003). Membrane Dialysis Transplantation, 14, 2478–81.
expression of proteinase 3 is genetically determined. Journal of the Wang, A.Y., Lai, K.N., Li, P.K., Leung, C.B., and Lui, S.F. (1996). Acute renal
American Society of Nephrology, 14, 68–75. failure induced by angiotensin converting enzyme inhibitor in a patient
Senzaki, H., Suda, M., Noma, S., Kawaguchi, H., Sakakihara, Y., and Hishi, T. with polyarteritis nodosa. Renal Failure, 18, 293–8.
(1994). Acute heart failure and acute renal failure in Kawasaki disease. Weaver, F.A., Kumar, S.R., Yellin, A.E., Anderson, S., Hood, D.B., Rowe, V.L.,
Acta Paediatrica Japonica, 36, 443–7. Kitridou, R.C., Kohl, R.D., and Alexancer, J. (2004). Renal revasculari-
Sinico, R.A., DiToma, L., Maggiore, U., Bottera, P., Radice, A., Tosoni, C., zation in Takayasu arteritis-induced renal artery stenosis. Journal of
Grasselli, C., Pavone, L., Gregorini, G., Monti, S., Frassi, M., Vecchio, F., Vascular Surgery, 3, 749–57.
Corace, C., Venegoni, E., and Buzio, C. (2005). Prevalence and clinical Wilde, B., van Paassen, P., Witzke, O., and Tervaert, J.W. (2011). New patho-
significance of antineutrophil cytoplasmic antibodies in Churg-Strauss physiological insights and treatment of ANCA-associated vasculitis.
syndrome. Arthritis and Rheumatism, 52, 2926–35. Kidney International, 79, 599–612.
Slot, M.C., Tervaert J.W., Boomsma, M.M., and Stegeman, C.A. (2004). Xiao, H., Heeringa, P., Hu, P., Liu, Z., Zhao, M., Aratani, Y., Maeda, N., Falk,
Positive classic antineutrophil cytoplasmic antibody (C-ANCA) titer R.J., and Jennette, J.C. (2002). Antineutrophil cytoplasmic autoantibod-
at switch to azathioprine therapy associated with relapse in proteinase ies specific for myeloperoxidase cause glomerulonephritis and vasculitis
3-related vasculitis. Arthritis and Rheumatism, 51, 269–73. in mice. Journal of Clinical Investigation, 110, 955–63.
Stegeman, C.A., Cohen Tervaert, J.W., Sluiter, W.J., Manson, W.L., de Jong, Xiao, H., Schreiber, A., Heeringa, P., Falk, R.J., and Jennette, J.C. (2007).
P.E., and Kallenberg, C.G.M. (1994). Association of nasal carriage of Alternative complement pathway in the pathogenesis of disease medi-
Staphylococcus aureus and higher relapse rate in Wegener’s granuloma- ated by anti-neutrophil cytoplasmic autoantibodies. American Journal
tosis. Annals of Internal Medicine, 120, 12–17. of Pathology, 170, 52–64.
Stilmant, M.M., Bolton, W.K., Sturgill, B.C., et  al. (1979). Crescentic glo- Yoshikawa, N., White, R.H., and Cameron, J.S. (1981). Prognostic signifi-
merulonephritis without immune deposits: clinicopathologic features. cance of the glomerular changes in Henoch Schönlein nephritis. Clinical
Kidney International, 15, 184–95. Nephrology, 16, 223–9.
CHAPTER 16

Digital ischaemia and


Raynaud’s phenomenon
K. Kwasind Huston, John H. Stone,
and Fredrick M. Wigley

Introduction knowledge has led to many exciting and new treatment modalities
for Raynaud’s phenomenon and digital ischaemia.
Insufficient blood flow to the distal extremities often constitutes
a patient’s first clear indication of an underlying illness. The Definitions
severity of this process can range from a mild nuisance to critical
ischaemia and tissue destruction. For patients, even mild symp- Historical perspective
toms can cause great emotional distress, especially in the set- In 1862, Raynaud described a condition ‘which in the ordinary lan-
ting of diagnostic uncertainty. For physicians, digital ischaemia guage is designated under the name of dead finger’. He described
poses a diagnostic challenge with high stakes. Failure to diagnose 31 patients who suffered from varying combinations and degrees
the cause promptly, and institute effective therapy prolongs the of digital pallor, cyanosis, and symmetrical gangrene, all of which
patient’s discomfort and leads to the loss of tissue viability. Few he attributed to prolonged vasospasm. To Raynaud, the symmetry
clinical events are more painful to observe than the slow death of the process that he observed strongly implicated hyperactivity
of a digit or limb stricken with insufficient blood flow, awaiting of the sympathetic nervous system as its cause. On inspection of
either auto amputation or surgical removal. The urgency of digi- the cases, it is clear that substantial heterogeneity existed within
tal ischaemia often dictates that treatment based on the proper this group of patients. Whereas some suffered extensive gan-
interpretation of clinical clues must begin before the definitive grene, others experienced only a ‘slight inconvenience . . . which
diagnosis is secure. Accurate knowledge of disease mechanisms often passes unperceived so that it does not require any treatment’
allows rational treatment decisions even when diagnostic infor- (Raynaud 1862).
mation is incomplete. The malady first described by Raynaud became known as
For general practitioners, few clinical findings are more strongly Raynaud’s disease. This unfortunate label persists today in
associated with rheumatic disease than digital ischaemia and medical jargon, albeit with a variety of usages. In the late 1800s,
Raynaud’s phenomenon. At the same time, few syndromes are Hutchinson noted that there were several distinct causes of sym-
the source of greater confusion regarding terminology, aetiology, metrical gangrene, including heart failure and senile atherosclero-
and management. More than 130  years after Maurice Raynaud sis (Hutchinson 1901). He was the first to note the development
first described ‘l’asphyxie locale et de la gangréne symétrique des of scleroderma several years after the onset of Raynaud’s disease
extrémités’ (local asphyxia and symmetrical gangrene of the digits) (Hutchinson 1896). In view of the varied causes of Raynaud’s dis-
(Raynaud 1862), there remains a tendency to lump many forms of ease, Hutchinson suggested Raynaud’s phenomenon as an alterna-
digital ischaemia under the rubric of Raynaud’s disease. Because all tive term. He wrote: ‘All who have studied in any detail the cases
aetiologies of digital ischaemia are not alike, and because therapeu- which have been grouped together under the name of Raynaud’s
tic approaches to this problem vary according to the precise cause, malady will admit that the time has arrived when they ought to
an essential step in patient management is distinguishing between be classified. They are not all alike, nor do they tend to the same
vasculitic, vasospastic, thrombotic, and other causes of digital results’ (Hutchinson 1901).
ischaemia. Over the ensuing decades, other influential medical thinkers
In this chapter, the focus is on digital ischaemia and Raynaud’s addressed the issue initially framed by Raynaud. Allen and Brown
phenomenon related to systemic rheumatic conditions, particularly (1932a) noted that Raynaud had recanted the strong association
vasculitis and the connective tissue diseases (scleroderma, systemic between ‘local asphyxia’ of the digits and symmetrical gangrene in
lupus erythematous (SLE), and related conditions). There has been his later writings. Specifically, Raynaud had characterized asphyxia
dramatic progress in defining the pathophysiological events lead- and gangrene as two degrees of the same malady, with gangrene
ing to vasospasm and vasculopathy over the last several years. This often absent. In 1932, they defined Raynaud’s disease by the
210 SECTION 3 clinical manifestations common to vasculitis

following criteria: (1) intermittent attacks of colour changes in the (a)


acral regions; (2) symmetrical or bilateral involvement; (3) absence
of clinical evidence of occlusive lesions in the peripheral arteries;
(4) minimal trophic changes or gangrene; (5) presence of the dis-
order for at least 2 years; and (6) no evidence of a secondary cause.
In a description of 265 patients diagnosed with Raynaud’s disease
at the Mayo Clinic, Allen and Brown concluded that only 147 met
their criteria for this diagnosis. In addition to the relative lack of
gangrene in the disorder they called Raynaud’s disease, they also
commented on a characteristic absence of pain ‘ordinary of no sig-
nificance’ (Allen and Brown 1932b). Raynaud’s disease as delineated
by Allen and Brown corresponds to primary Raynaud’s phenom-
enon in today’s nomenclature (see Section Current Nomenclature).
Lewis and Pickering, contemporaries of Allen and Brown,
believed that consensus on a definition for Raynaud’s disease
could never be achieved and proposed that the term be discarded
(Lewis and Pickering 1933). In its place, they proposed the consid- (b)
eration of two major categories of digital ischaemia: (1) Raynaud’s
phenomenon (corresponding to Allen and Brown’s criteria for
Raynaud’s disease); and (2) gangrene/ necrosis, an entirely separate
(but non-uniform) entity. To Lewis and Pickering, Raynaud’s phe-
nomenon signified ‘active and intermittent closure of small arter-
ies of the order of those supplying the digits, a closure manifesting
itself clinically in a pallid or fully cyanotic state of the affected skin’
(Lewis and Pickering 1933). The authors emphasized that this phe-
nomenon (illustrated in Figure 16.1) should not be regarded as the
result of only one set of circumstances.
In contrast, with regard to the second major category, digital
gangrene/ necrosis, Lewis and Pickering stated that digital artery
vasospasm was ‘not an essential or even a usual precursor of ’ that
condition (Lewis and Pickering 1933). Rather, they asserted, digital
gangrene/ necrosis stemmed from a permanent structural change,
resulting in ‘plugging’ of the culprit arteries. Though lacking proof, Fig. 16.2 Digital infarctions occurring in Wegener’s granulomatosis and Buerger’s
disease. (a) Wegener’s granulomatosis. (b) Thromboangiitis obliterans (Buerger’s
Lewis and Pickering (1933) hypothesized that ‘intimal changes, disease).
including thrombosis, are usually or always responsible for the
obliteration of small arteries leading to necrosis’.
Under the heading of conditions associated with gangrene, Lewis distinctions, they anticipated the modern definitions of primary
and Pickering included not only disorders leading to major digital and secondary Raynaud’s phenomenon.
infarction (Figure 16.2a and 16.2b), but also the minute necrotic
foci that we now term digital pitting (Figure 16.3). In making these Current nomenclature
At least two parallel systems of nomenclature exist in the classifica-
tion of Raynaud’s phenomenon (Table 16.1). In the United States,

Fig. 16.1 Raynaud’s phenomenon, representing digital artery vasospasm


leading to pallor. In prolonged vasospasm, pallor gives way to cyanosis, followed
by rubor as circulation is restored. Fig. 16.3 Digital pitting in a patient with diffuse systemic sclerosis.
CHAPTER 16 digital ischaemia and raynaud’s phenomenon 211

Table 16.1 Nomenclature schemes for Raynaud’s phenomenon young women, among family members of people with Raynaud’s
phenomenon, and in geographic areas with cold temperatures.
Underlying disorder United States Europe Results have been published from a study focused on the
incidence and natural history of Raynaud’s phenomenon in a
No Primary Raynaud’s Primary Raynaud’s disease
phenomenon
community-based population derived from the Framingham
Offspring Study Cohort (Suter et al. 2005). In that report of 717
Yes Secondary Raynaud’s Raynaud’s syndrome women and 641 men, the incidence, over a period of 7 years, was
phenomenon 2.2% in women and 1.5% in men. Interestingly, the symptoms of
Raynaud’s rarely interfered with daily activities and remitted in
objection has existed to use of the stigma ‘Raynaud’s disease’ for 64% of affected patients.
patients in whom vasospasm is the principal pathophysiological Many case reports document that Raynaud’s phenomenon can
mechanism. Thus, primary Raynaud’s phenomenon is the preferred be associated with vasculitis or be a presenting sign of a disease
designation for the occurrence of cold-induced vasospasm that is process with vasculitis (Andreu-Sanchez et al. 1991). Furthermore,
not associated with a disease or known cause (LeRoy and Medsger most review articles list vasculitis as a cause of Raynaud’s phenom-
1992). Secondary Raynaud’s phenomenon is the term of vasos- enon; however, few surveys have measured the true frequency of
pasm induced by cold in the setting of an underlying disorder. In vasculitis as the cause of Raynaud’s phenomenon in patients pre-
Europe, Raynaud’s phenomenon is the blanket term for any type senting with that complaint. Most reports of associations between
of cold-induced digital vasospasm (Belch 1990), and the concept vasculitis and Raynaud’s phenomenon must be regarded with cau-
of Raynaud’s phenomenon is divided into Raynaud’s syndrome tion because of the failure to obtain definitive diagnoses of vasculi-
(associated with an underlying condition) and primary Raynaud’s tis; loose usage of the term vasculitis (in particular, a tendency for
disease (where no underlying condition is present). certain connective tissue disorders, for example scleroderma and
Lack of consensus regarding these issues will probably persist SLE, to be labelled as such); confusion resulting from alterations in
until the precise causes of cold-induced vasospasm (probably mul- vasculitis classification schemes (Jennette et al. 1994; Lie 1994); and
tifactorial) are defined more clearly. Another source of confusion imprecise definitions of what is meant by Raynaud’s phenomenon.
results from the imprecise application of the moniker Raynaud’s Primary Raynaud’s phenomenon is the most common diagnosis
(whether disease, syndrome, or phenomenon) to conditions in in most surveys. Raynaud’s phenomenon associated with a connec-
which vasospasm is not the principal event—or perhaps not even tive tissue disease (scleroderma, SLE, inflammatory myopathies) is
a minor one. Most forms of vasculitis fall under this category. the second most common category reported. Among 100 patients
Although some degree of vasospasm may be present in the vascu- presenting to an academic centre with Raynaud’s phenomenon,
litic disorders, the principal event is narrowing of the blood ves- Porter et  al. reported vasculitis principally in conjunction with
sel lumen by necrotizing inflammation, frequently complicated by connective tissue diseases (Porter et al. 1976). A tabulation of 631
the occurrence of thrombi in the microvasculature. The impor- cases seen at the same institution found that 37.4% had primary
tance of this distinction is not semantic; digital ischaemia resulting Raynaud’s phenomenon, 37.2% had a connective tissue disease,
from vasculitic inflammation requires drastically different therapy 3.5% had hypersensitivity angiitis, 1.9% had Buerger’s disease, and
than does vasospasm due to primary or secondary Raynaud’s 20% had other disorders not associated with vasculitis (Friedman
phenomenon. et al. 1988). Thus, in that study, Raynaud’s phenomenon was attrib-
uted to vasculitis in fewer than 5% of the patients.
Similarly, another survey from 50 Italian medical centres found that
Epidemiology among 761 patients with Raynaud’s phenomenon, scleroderma was
Most studies of Raynaud’s phenomenon have been conducted at the most common cause (28.4%). Vasculitis, in contrast, was reported
academic medical centres, and thus the prevalence data are biased in less than 5% of cases (Grassi et al. 1998). A population-based survey
toward patients with more severe illnesses. Only recently have in South Carolina found that 32% of patients had primary Raynaud’s
population-based surveys estimated the prevalence of Raynaud’s phenomenon, 60% had Raynaud’s phenomenon and other health
phenomenon in the general population. The data vary depending problems, and 8% had evidence of a connective tissue disease (Maricq
on the diagnostic criteria, the survey technique, and the particular et al. 1990). Vasculitis was not reported in any of the patients who
characteristics of the population surveyed. described symptoms compatible with Raynaud’s phenomenon in that
Population-based surveys estimate the prevalence of Raynaud’s study. A study of 118 patients assessed the prevalence of secondary
phenomenon to be between 4.9% and 20.1% in women, and disorders in patients with Raynaud’s phenomenon that were referred
between 3.8% and 13.5% in men (Maricq et  al. 1993). Surveys to a rheumatology centre in Italy (De Angelis et al. 2003). Of the 118
across different ethnic groups consistently report figures in the patients, 52% had a secondary disorder (47% with scleroderma) but
range of 3–5%: from Spain, 3.2–4.7% (Riera et al. 1993); from the none had vasculitis. A more recent study from Serbia followed 3029
Netherlands, 2.9% (Bartelink et  al. 1992); from Japan, 3.0–3.4% patients with primary Raynaud’s phenomenon at baseline for a mean
(Inaba et al. 1993); from Turkey, 5.9% (Onbasi et al. 2005); from of 4.8 years (Pavlov-Dolijanovic et al. 2012). The authors reported that
the United States, 4.3–5.7% (Maricq et  al. 1989); and among 54.8% of patients still had primary Raynaud’s phenomenon at the end
urban-dwelling African-Americans, 4.0% (Gelber et  al. 1999). of the study and 37.1% developed a connective tissue disorder (8.66%
Outlying estimates include those from France, 13.5–20.1% (Marciq with scleroderma). Vasculitis developed in only 0.99% of the study
et al. 1993); Sweden, 15.9% (Leppert et al. 1987); Estonia, 7.9–8.3% population.
(Valter and Maricq 1997); and New Zealand, 11.5% (Purdie et al. In summary, the existing studies of Raynaud’s phenomenon docu-
2009). The prevalence of Raynaud’s phenomenon is higher among ment a moderately high prevalence of this condition. Estimates of the
212 SECTION 3 clinical manifestations common to vasculitis

prevalence of Raynaud’s phenomenon are stable across many popu- mechanistic insights may have direct implications for treatment of
lations. The true prevalence of vasculitis among patients with symp- Raynaud’s phenomenon.
toms of Raynaud’s, although not well known, is probably less than 5%. The endothelium of cutaneous blood vessels also influences local
blood flow in response to a variety of stimuli. The endothelium of
the vessel can be activated by a number of mechanisms, includ-
Pathogenesis ing: changes in the physical environment of the cell, such as shear
This section focuses on the pathogenesis of primary and secondary stress altered by local blood flow; alterations in the biochemical
Raynaud’s phenomenon. Specific information about vascular dys- milieu, such as regional hypoxia; circulating hormones; neurotrans-
function in the vasculitides is found elsewhere in this text. mitters released by autonomic or sensory nerves; autocoids from the
vessels themselves, such as bradykinin; mediators released from cir-
Physiological control of blood vessel calibre culating cells, such as platelet-derived serotonin and thromboxane A;
Maintenance of core body temperature is achieved partly by altera- and endothelial released factors such as prostacyclin or endothelin.
tion of blood flow to the skin surface through a dense array of arte-
riovenous (AV) shunts. During local or whole body cooling, blood Primary Raynaud’s phenomenon
flow to the skin is reduced via vasoconstriction of these shunts. In The pathophysiology of Raynaud’s phenomenon remains incom-
contrast, warm temperatures cause vasodilatation, thereby foster- pletely understood. The factors contributing to this condition are
ing heat exchange. The thermoregulatory vessels, which also pro- complex, and are probably different for the primary and second-
vide nutritional support to the digits, are influenced not only by ary forms of Raynaud’s phenomenon. As suggested by Lewis and
changes in temperature, but also by emotional stress and a variety Pickering (1933), most of the current evidence implicates a local
of other environmental factors. vascular defect rather than a central nervous system abnormality
These changes in blood flow are mediated primarily by reflex as the principal problem, with a resulting unfavourable balance
increases in sympathetic tone. Regulation of regional blood flow is between vasodilatory and vasoconstrictive stimuli.
complex, altered not only by neural signals, but also by hormonal Familial aggregation occurs in primary Raynaud’s phenomenon
influences and mediators released by both the endothelium and cir- (Freedman et al. 1999); approximately 30% of patients with primary
culating cells. Certain autonomic nerve endings release a group of Raynaud’s phenomenon have first-degree family members with the
neurotransmitters that mediate vasodilation. These include nitric condition. This finding suggests the possibility of a specific gene
oxide, vasoactive intestinal peptide, and acetylcholine. Cutaneous defect(s) among patients with primary Raynaud’s phenomenon.
vasculature is also sensitive to mediators released from sensory Indeed, it has been suggested that primary Raynaud’s phenomenon
fibres, including calcitonin gene related peptide (CGRP), substance is a trait rather than a disorder (Hadler 1998).
P, neurokinin A, and neuropeptide Y (Generini et al. 2005). In primary Raynaud’s phenomenon, exaggerated cold sensitivity
Cutaneous blood vessels are richly innervated by a variety of of the α2-adrenoceptors is thought to be a major factor in patients’
neuron subtypes. The most potent influences on the digital circula- symptoms, possibly via cold-induced translocation of the α2C adr-
tion are the α2-adrenergic receptors, with vasoconstriction of small energic receptor to the plasma membrane of regulatory cells. This
arteries under α2-adrenergic tone. The expression of α2-adrenergic hypothesis is supported by the inhibition of vasospastic attacks
receptors is influenced by temperature. At cold temperatures, in vitro by selective α2-adrenoceptor antagonists and increased sensitiv-
studies demonstrate up-regulation of α2-adrenergic receptors, as well ity to agonists in patients with primary Raynaud’s phenomenon
as an increased affinity of α2-adrenoceptors for norepinephrine, the (Freedman et al. 1993; Freedman et al. 1995). Additional studies with
major sympathetic neurotransmitter (Flavahan et al. 1985). Studies more-specific agonists and antagonists of α2-adrenoceptors may
in human volunteers have confirmed the role of α2-adrenoceptors elucidate the defect(s) present in primary Raynaud’s phenomenon.
in mediating cold-induced vasoconstriction (Coffman and Cohen Non-adrenergic defects in control of regional blood flow may
1988; Ekenvall et al. 1988). There are now at least three known α2- also contribute to primary Raynaud’s phenomenon. Non-adrenergic
adrenergic receptor subtypes, α2A, α2B, and α2C (Philipp et al. 2002). mechanisms may modulate the α2-adrenergic response by increasing
Interestingly, the α2C adrenergic receptor remains localized to intra- the number of receptors, or they may alter the activity of receptors by
cellular compartments under normal conditions where it is unavail- changes in the intercellular signal-transduction pathway. Increased
able for binding to its ligand; however, upon exposure to cold, the α2C production of endothelin-1 (Zamora et al. 1990), decreased produc-
receptor translocates from the Golgi network to the cell surface where tion of CGRP (perhaps secondary to loss of cutaneous innervation)
it is available for agonist-induced activation (Jeyaraj et  al. 2001). (Bunker et al. 1990) and impaired endothelial function (Bedarida
Receptor translocation is mediated by the RhoA/Rho kinase signal- et al. 1993) have all been reported in primary Raynaud’s phenomenon.
ling pathway, possibly in response to stimulation by reactive oxygen The protein tyrosine kinase signal transduction pathway has
species generated by smooth muscle cell mitochondria (Bailey et al. been implicated in Raynaud’s phenomenon. Blood vessels taken
2004; Bailey et al. 2005). Furthermore, inhibition of the α2C receptor from patients with primary or secondary Raynaud’s phenom-
has been shown to prevent cold-induced vasoconstriction, highlight- enon showed increased tyrosine phosphorylation in response
ing the importance of specific receptor subtypes in normal physiolog- to cold-induced contraction compared to control blood vessels
ical mechanisms of thermoregulation (Chotani et al. 2000). It is now (Furspan et al. 2004; Furspan et al. 2005). Both vessel contraction
thought that α2C receptors may function as vascular stress receptors and tyrosine phosphorylation could be inhibited by a protein tyros-
in response to mechanical (vibration), immunological (autoimmune ine kinase inhibitor leading to the conclusion that tyrosine phos-
disease), or chemical/ environmental triggers (Flavahan 2008). These phorylation mediates blood vessel reactivity in Raynaud’s patients.
triggers can up regulate the expression of α2C receptors or stimulate An epidemiological survey reports that women using unop-
their mobilization to the vascular smooth muscle cell surface. These posed oestrogen are more likely to have Raynaud’s phenomenon,
CHAPTER 16 digital ischaemia and raynaud’s phenomenon 213

suggesting a negative influence of oestrogen on digital vessels stem cells enter the vessel and differentiate into myofibroblasts with
(Fraenkel et  al. 1998); however, others have demonstrated that synthetic capacity. The mechanisms involved in vessel remodelling
the acute infusion of oestrogen improves cutaneous blood flow in are incompletely understood but are likely to involve an interplay
Raynaud’s phenomenon (Lekakis et al. 1998). There is in vitro and between extracellular matrix proteins such as elastin and fibrillin-1,
in vivo evidence to support the vasodilatory properties of oestrogen various cytokines including transforming growth factor-β, mem-
via the nitric oxide pathway (Generini et al. 2005). Despite these bers of the matrix metalloproteinase family of proteases, and reac-
findings, oestrogen has recently been shown to increase smooth tive oxygen species (Flavahan et al. 2003; Bou-Gharios et al. 2004;
muscle expression of α2C receptors and cold-induced vasoconstric- Filippov et al. 2005; Su et al. 2001). There is phenotypic alteration
tion (Eid et al. 2007). It is likely that oestrogen has multiple effects of the vascular smooth muscle cells with migration to the vascular
on the vasculature, which result in a dynamic compromise between intima, proliferation, and enhanced extracellular matrix protein
vasoconstricting and vasodilating mechanisms. At this point, the secretion with subsequent vessel dysfunction resulting in ischaemic
interaction between oestrogen and Raynaud’s remains unclear. damage to under-perfused tissue. Progress in elucidating the mech-
anisms of scleroderma vasculopathy is now being provided by ani-
Secondary Raynaud’s phenomenon mal models generated via aberrant expression of key transcription
Endothelial dysfunction and smooth muscle defects have also been factors in mice (Maurer et al. 2009; Asano et al. 2010). Hopefully,
implicated in secondary Raynaud’s phenomenon. Depending on the these exciting studies will provide significant insight into molecular
specific disease or type of vascular injury, there may be several rea- mechanisms of scleroderma vasculopathy.
sons for disruption of normal vascular tone. A disease process lead- The blood vessel lesions occur in the setting of inadequate new
ing to injury of the endothelium potentially diminishes production blood vessel formation despite elevated circulating levels of vascu-
of vasodilators such as nitric oxide. Dysfunctional endothelial cells lar endothelial growth factor (Flavahan et al. 2003). This suggests
also: promote intravascular thrombosis; enhance the release of medi- additional defects in down-stream regulation of angiogenesis but
ators that activate vascular smooth muscle cells; and promote inflam- whether this is due to defects in the endothelial cell response to
matory responses by increased expression of adhesion molecules. angiogenic stimuli or the presence of inhibitors of angiogenesis is
Most of the studies investigating the role of the endothelium in currently not known. Increased levels of the angiogenesis inhibi-
secondary Raynaud’s phenomenon have been performed in scle- tors endostatin and thrombospondin-1 have been reported in
roderma, a disease associated with a profound vasculopathy and scleroderma (Hebbar et al. 2000; Macko et al. 2002). In addition,
repeated digital ischaemic events that often lead to digital ulcers and recent studies suggest that there are inadequate numbers of circu-
amputation. In the digital arteries of scleroderma patients, there is lating bone marrow-derived endothelial precursors in scleroderma
luminal narrowing of more than 75% caused by intimal fibrosis (Kuwana et al. 2004). However, other investigators have reported
and occlusion of the lumen by thrombi. Evidence of endothelial higher levels of circulating endothelial precursors in scleroderma
cell activation is reported in cutaneous vessels (Freemont et  al. patients (Allanore et al. 2007; Avouac et al. 2008). This discrepancy
1992; Prescott et al. 1992). These findings suggest the presence of may be a result of technical issues in having a standard approach to
endothelial dysfunction (Pearson 1991). characterize these cells (Distler et al. 2009). Endothelial progenitor
Platelet activation is also evident in scleroderma, with increased cells are normally capable of repairing or re-modelling damaged
systemic release of thomboxane, a potent vasoconstricting prosta- vessels and impairment of this function could cause a defect in vas-
glandin (Reilly et al. 1986). Increased platelet adhesion, decreased culogenesis (Cipriani et al. 2007; Del Papa et al. 2006).
storage of von Willebrand factor, and decreased adenosine uptake While few studies have investigated directly the mediators of digital
are all manifestations of endothelial damage in scleroderma ischaemia and vasospasm in vasculitis, it is likely that a complex series
(Herrick et al. 1996). Infusion of L-arginine, the substrate for nitric of events occurs after direct injury to the endothelium, with profound
oxide production, can reverse the vasospasm in the hands of scle- disturbances in normal regulatory mechanisms. Vascular occlu-
roderma patients (Freedman et al. 1999), suggesting that defects in sion and damage reduce tissue perfusion, creating ischaemia. While
nitric oxide production may also exist in scleroderma, but work by ischaemia releases potent vasodilators, damaged vessels are unlikely
Flavahan reports normal endothelial function of scleroderma blood to have normal responses. Thus, it is not surprising to see secondary
vessels using cutaneous arteries isolated from biopsies of clinically vasospasm and Raynaud’s phenomenon in vasculitic diseases.
uninvolved skin (Flavahan et  al. 2000). These in vitro studies of
scleroderma vessels demonstrate a profound increase in sensitiv- Clinical evaluation
ity (up to 300-fold) to α2-adrenoceptor-mediator vasoconstriction
of smooth muscle, independent of endothelial mechanisms. This Differential diagnosis
study suggests that an early defect in scleroderma may exist in the Primary Raynaud’s is typically elicited by patient history but on occa-
vascular smooth muscle response, while later the endothelial cell sion the stress associated with a medical visit or entrance into a cold
damage occurs, causing a vasculopathy with progressive ischaemia examination room will precipitate an attack. This is especially true
that results from an imbalance between vessel loss and angiogenesis. for those patients who are more prone to frequent vasospasm. The
The vasculopathy in scleroderma is characterized by intimal pro- physician often has the luxury of time when considering potential
liferation thought to be associated with vascular smooth muscle diagnostic and treatment modalities in these patients. In contrast,
cells having a synthetic or ‘dedifferentiated’ phenotype. It is hypoth- the occurrence of critical digit ischaemia is a medical emergency.
esized that these cells differ from the normal ‘differentiated’ vascu- This condition requires a swift diagnostic evaluation and the prompt
lar smooth muscle cell responsible for vascular tone in that they institution of treatment, usually before the work-up is complete.
produce extracellular matrix proteins and are involved in vessel Before narrowing the focus to the rheumatic conditions, a wide
remodelling (Flavahan et al. 2003). It is also possible that circulating array of diagnostic categories must be excluded (Table 16.2).
214 SECTION 3 clinical manifestations common to vasculitis

Table 16.2 Differential diagnosis of digital ischaemia In approaching the patient, these four processes may be viewed
broadly as corresponding to vasculitic diseases, primary Raynaud’s
Primary Raynaud’s phenomenon Antiphospholipid Syndrome phenomenon, connective tissue diseases and clotting diatheses
Secondary Raynaud’s phenomenon Blood dyscrasias (particularly aPL-associated disorders), respectively. This con-
Systemic sclerosis Cryoglobulinaemia
struct is useful, because recognition of the underlying mechanism
will allow a logical approach to diagnosis and treatment decisions.
Systemic lupus erythematosus Paraproteinaemias
Because issues related to thrombosis are covered elsewhere in
Inflammatory myopathy Cold agglutinins
this text, the following sections focus on the clinical evaluation of
Rheumatoid arthritis Polycythaemia vera patients with digital ischaemia caused by the primary mechanisms
Sjögren’s syndrome of vasculitis, vasospasm, and vasculopathy.
Mixed connective tissue disease
Undifferentiated connective tissue Interpreting the colour changes
disease Primary Raynaud’s phenomenon is generally mild in nature. Exposure
Vasculitides Drugs and toxins to cold triggers pallor that extends from the digital tip to its middle or
Buerger’s disease Ergotamines base. Sharp colour demarcation indicates virtually complete closure
Wegener’s granulomatosis/ Vinblastine
of the digital artery and cutaneous arterioles. The pallor remains for
granulomatosis with polyangiitis a short period of time before cyanosis (a blue/gray or even black-
Bleomycin
Polyarteritis nodosa
ish discoloration) appears, caused by the pooling of slow-flowing,
Amphetamines
deoxygenated blood. The digital skin and other areas exposed to cold
Microscopic polyangiitis Vinyl chloride exposure (hands, arms, face, nose, ears, knees, feet) may appear mottled, and
Churg–Strauss syndrome/ eosinophilic Clonidine remain cyanotic for approximately 15–20 minutes after rewarming.
granulomatosis with polyangiitis
Carboplatin Following reversal of the vasoconstriction, an erythematous blush
Cryoglobulinaemic vasculitis
Gemcitabine occurs, caused by hyperaemic reperfusion of the digit.
Takayasu’s arteritis Many patients recall the cyanotic phase of Raynaud’s phenom-
Giant cell arteritis enon vividly but overlook the other phases, particularly the pallor
Structural arterial disease Occupational phase. Throughout the attack, patients feel symptoms of numbness,
Atherosclerosis Hand–arm vibration syndrome ‘pins and needles’, and discomfort, sometimes accompanied by a
Hypothenar hammer syndrome sense of limb clumsiness. As noted by Allen and Brown, pain is
uncommon in primary Raynaud’s phenomenon (Allen and Brown
Other 1932a).
Thoracic outlet syndrome Not every patient reports the classic triphasic colour changes
Reflex sympathetic dystrophy (white, blue, and red). Thus, a history of unusual sensitivity to cold
Hypothyroidism and episodic white and/or blue discoloration of the digital skin in
Cold injury (frostbite) response to cold is sufficient for a diagnosis of Raynaud’s phenom-
Paraneoplastic enon. A typical attack begins asymmetrically, in one or two fingers.
On continued exposure to cold, patients with primary Raynaud’s
phenomenon may develop pallor of all of the fingers of both hands.
An exhaustive description of the clinical evaluation for all causes The criteria for diagnosis of primary Raynaud’s include symmet-
of digital ischaemia is beyond the scope of this chapter, but the fol- ric vasospastic attacks precipitated by cold or emotional stress in
lowing points are pertinent. First, the clinical evaluation begins the absence of tissue necrosis. Patients with this disorder should
with a careful history. Following completion of the history, the pos- have no evidence of an underlying secondary cause for Raynaud’s
sibility of an underlying systemic inflammatory condition should and normal testing including autoantibodies and erythrocyte sedi-
be apparent to the examiner. During the physical examination, mentation rate. In addition, these patients should have normal nail-
particular attention may then be directed toward narrowing the fold capillaries. Cutaneous capillaries can be examined by placing
differential diagnosis and determining the severity of the disorder. a drop of immersion oil at the base of the patient’s fingernail and
A history of cigarette smoking, particularly when heavy, is relevant viewing the nailfold with an ophthalmoscope set to 10 to 40 diop-
not only to atherosclerosis, but also to Buerger’s disease (throm- ters or with a stereoscopic microscope. Abnormal capillaries with
boangiitis obliterans). Obstetrical histories of second or third tri- vessel dilatation or loss of capillary loops suggest a secondary cause
mester miscarriages may indicate the antiphospholipid antibody of Raynaud’s such as scleroderma, dermatomyositis, or systemic
(aPL) syndrome. Occupational histories may reveal risk factors for lupus erythematosus (Wigley 2002).
mechanically induced digital ischaemia, such as the hypothenar Persistent asymmetric attacks, particularly if associated with
hammer syndrome (Pineda et  al. 1985). Other forms of trauma, pain, prolonged ischaemia, or tissue ulceration, also suggest sec-
such as extreme cold exposure, also constitute information that ondary Raynaud’s phenomenon. These findings are suggestive of
must be elicited in the patient’s history. A history of weight loss or a more dramatic disease process affecting blood vessels. In con-
cachexia may provide a clue to an unrecognized malignancy. trast to the findings in primary Raynaud’s phenomenon, vessels in
Among systemic inflammatory illnesses leading to vessel occlu- patients with secondary causes such as scleroderma are chronically
sion, four major disease processes (often not mutually exclusive) ill with multiple abnormalities (see Section Pathogenesis) even in
pertain. These are:  (1)  necrotizing inflammation; (2)  vasospasm; the absence of acute vasospasm. Spasm of these narrowed, fibrotic
(3) non-inflammatory vasculopathy; and (4) thrombus formation. vessels leads to more startling perfusion deficits in these patients.
CHAPTER 16 digital ischaemia and raynaud’s phenomenon 215

The index and middle fingers are usually more sensitive to attacks
than the other fingers. The thumb is the least sensitive digit. Toes
as well as fingers may be affected, but finger involvement is usually
more intense. Rarely, a patient with secondary Raynaud’s phenom-
enon has attacks only in the feet.
In vasculitis, the presentation of digital ischaemia differs strik-
ingly from primary Raynaud’s phenomenon, but may be similar
in appearance to secondary Raynaud’s phenomenon. Vasculitis
patients with digital ischaemia present with acutely painful, swollen
fingers, and signs of tissue ischaemia. The process is usually sym-
metrical, but often there is a marked disparity between involvement
of the upper and lower extremities. The digit is often reddened
secondary to the dilatation of small blood vessels, an attempt to
foster collateral flow. The fingertips or the sites of subsequent tis-
sue infarction are initially pale, with rims of cyanosis mixed with
erythema. The involved digits are exquisitely tender to touch. Pain Fig. 16.5 Patient with limited systemic sclerosis, sclerodactyly, and telangiectasias.
often extends into the deep tissues of the fingers, hands, and, occa-
sionally, the arms. The pain may be so intense that the patient is ◆ Patients with scleroderma-spectrum illnesses may have scle-
unable to sleep, pacing the floor or hanging the ischaemic limb rodactyly, telangiectasias, hypopigmentation (in dark-skinned
over the edge of the bed in an attempt to improve local blood flow. individuals), or calcinosis cutis (Figure 16.5).
Although the ischaemia may wax and wane, it persists without ◆ Capillary loop dilatations, often visible to the naked eye
complete recovery, even with prolonged exposure to warmth. If (Figure 16.6), firmly place the underlying illness in the spectrum
digital ischaemia is not reversed by prompt, effective treatment, tis- of connective tissue disease. Normal capillary architecture sug-
sue infarction and gangrene ensue. gests primary Raynaud’s phenomenon.
Patients with vasculitis-associated digital ischaemia seldom recall
any pallor phase, noting instead the onset of pain followed by the ◆ The loss of digital pulp attests to previous digital infarctions
rapid onset of cyanosis and progression to gangrene. In vasculitis, (Figure 16.7), but this may occur in many connective tissue dis-
digital ulceration is a consequence of vascular occlusion, compli- orders as well as in the vasculitides. Thus, digital pulp atrophy
cated by elements of vasoconstriction. does not distinguish ischaemia due to vasculitis from that caused
by connective tissue diseases.
Physical examination clues ◆ A positive Allen’s test or the absence of pulses in the wrists or
The presence of specific findings on physical examination often feet suggests the presence of a process involving medium-sized
differentiate the four major pathophysiological processes in digital arteries.
ischaemia (vasculitis, vasospasm, vasculopathy, and thrombosis) ◆ In contrast to the vasospasm associated with connective tissue
with a high degree of specificity. Although the entire patient must diseases, necrotizing vasculitis commonly causes splinter haem-
be considered and examined thoroughly, careful attention to the orrhages (Figure 16.8).
hands may be particularly instructive:
◆ Patients with primary Raynaud’s typically have normal-appearing Specific clinical testing
digits in the absence of acute vasospasm. Several days elapse before results of most serological analyses
◆ Connective tissue disorders associated with digital ischaemia can are available and so treatment must begin before the entire diag-
result in marked cyanosis (Figure 16.4). nostic evaluation is complete, but a full work-up is essential to

Fig. 16.4 Cyanosis in a patient with systemic sclerosis. Fig. 16.6 Capillary loop dilatation in a patient with systemic sclerosis.
216 SECTION 3 clinical manifestations common to vasculitis

(a) (a)

(b) (b)

(c)

Fig. 16.7 Digital infarction (a), with subsequent loss of digital pulp (b).

knowledgeable patient management beyond the first few days. The


battery of blood and urine tests that is useful in the initial assessment
of patients with digital ischaemia is displayed in Table 16.3. Selected
imaging studies (particularly chest radiography), directed by find-
ings in the history, physical examination, and other evaluations, may
also be instructive. Increased acute phase proteins, as indicated by
an elevated erythrocyte sedimentation rate or C-reactive protein, are
very helpful in distinguishing digital ischaemia due to inflammatory
causes, that is systemic necrotizing vasculitis or vasculitis associated Fig. 16.8 Digital ischaemia in a patient with vasculitis. (a) Purple, cyanotic
discoloration on the palmar side. (b) Splinter haemorrhages. (c) Normal fingers
with a connective tissue disease, from the vasculopathy character-
again, several months after start of treatment. The splinter haemorrhages have
ized by intimal fibrosis and vasospasm associated with Raynaud’s nearly grown out.
phenomenon in scleroderma. The presence of a secondary disorder
will dictate further diagnostic evaluation.
Buerger’s disease). Somewhat surprisingly, 44 patients had evidence
Utility of angiography of atherosclerotic disease despite a mean age of 46.7 years. Of these
The usefulness of angiograms was studied in a group of 103 patients, 47% had laboratory evidence of dyslipidaemia. These
patients who presented with bilateral Raynaud’s phenomenon and results suggest a high prevalence of atherosclerosis in patients with
had no obvious underlying disease process (Van Vugt et al. 2003). Raynaud’s and this finding requires further investigation.
Angiography revealed vasospasm in 44 patients, peripheral embo- Angiography is not required for all patients with digital ischae-
lism in eight patients, and vasculitis in six patients (three with mia. In many cases, angiography only serves to confirm what both
CHAPTER 16 digital ischaemia and raynaud’s phenomenon 217

Table 16.3 Useful blood and urine tests in the evaluation of digital a compatible history, but the specificity of this finding is imper-
ischaemia fect. In the proper clinical setting, angiography of other vascular
beds (mesenteric or renal blood vessels) may confirm the diagno-
Blood sis of polyarteritis nodosa. Finally, angiography of the extremities
Complete blood count with differential sometimes provides useful information by indicating the extent
Serum electrolytes and chemistries of disease; substantial territory at risk may support more aggres-
Thyroid stimulating hormone sive treatment with immunosuppressive drugs. The extent of vessel
Lipid profile involvement, however, correlates imperfectly with the likelihood of
Erythrocyte sedimentation rate (or C-reactive protein)
treatment response. In the setting of vasculitis, the decision to add
a cytotoxic agent is probably founded more securely on evidence
Antinuclear antibody screen
of disease in vital organs or an inadequate response of the digital
Assays for precipitins: anti-Ro, -La, -Sm, -RNP,
ischaemia to the initial use of glucocorticoids, rather than to the
C3 and C4 complement levels extent of vessel involvement.
Topoisomerase III (Scl-70). antibodies
Antineutrophil cytoplasmic antibody assays (positive immunofluorescence
tests followed by ELISA for proteinase-3 and myeloperoxidase antibodies) Treatment
Antiphospholipid antibody tests (anticardiolipin antibodies, The principal mode of therapy in digital ischaemia associated with
anti-beta2-glycoprotein I antibodies, assays to detect lupus anticoagulants, rheumatic conditions is predicated on the perceived pathophysi-
rapid plasma reagin)a ology in each individual patient. Several guidelines are useful in
Cryoglobulins designing the approach to treatment:
Serum protein electropheresis ◆ For primary Raynaud’s phenomenon or for Raynaud’s phenom-
Urine enon caused by a connective tissue disease, treatment should
Urinalysis with microscopy be directed toward reversing vasospasm. An algorithm for the
Urine protein electropheresis treatment of vasospasm is described in Section Treatment of
Raynaud’s Phenomenon
a Additional investigations may be required if a primary thrombotic process is suspected.
◆ In some patients with connective tissue diseases (particularly
SLE, dermatomyositis, mixed connective tissue disease and
the patient and clinician already know (that the patient has insuf- undifferentiated connective tissue disease), necrotizing vasculi-
ficient blood flow to the extremities) but fails to clarify the cause tis may be superimposed on a predisposition to vasospasm. In
(Figure 16.9). In general, the careful, astute performance of a his- SLE, for example, episodes of Raynaud’s phenomenon are usu-
tory and physical examination, supplemented by selected diagnos- ally similar to those that occur in primary Raynaud’s phenom-
tic tests of the blood and urine, are more reliable than angiographic enon: relatively painless, uncomplicated, and not associated with
procedures in making these distinctions. ischaemia-induced tissue injury. When such a patient develops
The presence of corkscrew-shaped collateral blood vessels severe Raynaud’s attacks with threatened digital infarction, one
at the levels of the wrists or ankles suggests Buerger’s disease must consider the possibility of vasculitis (or thrombotic vascu-
(thromboangiitis obliterans) (see Chapter  38) in a patient with lar occlusion secondary to the aPL syndrome). Glucocorticoids
alone are usually sufficient to control vasculitis associated with a
connective tissue disease.
◆ For patients with digital ischaemia caused by a primary systemic
necrotizing vasculitis (Wegener’s granulomatosis, polyarteritis
nodosa, the Churg–Strauss syndrome, or others), immunosup-
pression is the cornerstone of treatment. In addition to gluco-
corticoids, these patients often require a cytotoxic agent such as
cyclophosphamide or B-cell targeted therapy with rituximab.
In such patients, trials of vasodilators may be attempted, but
such modalities are unlikely to achieve the dramatic effect they
sometimes have in disorders in which vasospasm is the principal
mechanism. Furthermore, the use of vasodilators in vasculitis
may lead to a steal phenomenon in which the healthy blood ves-
sels dilate but inflamed vessels cannot, which exacerbates ischae-
mia in inflamed tissues.
◆ In the acute setting, thrombosis of the microvasculature may
complicate digital ischaemia caused by either intense vasospasm
or necrotizing vasculitis. Treatment with heparin in the acute
phase (while waiting for vasodilators or immunosuppression
Fig. 16.9 Angiogram of a 59-year-old man with severe digital ischaemia caused
by vasculitis. His digital ischaemia resolved entirely with immunosuppression to take effect) may be prudent, even though few studies provide
(cyclophosphamide and glucocorticoids). The sharp ‘cut-off’ appearance to the unequivocal support for this practice. Obviously, anticoagulation
blood vessels cannot be distinguished from thrombosis. should not be employed in patients with pulmonary capillaritis,
218 SECTION 3 clinical manifestations common to vasculitis

mesenteric haemorrhage, bleeding into the central nervous sys- small. Another meta-analysis has been performed in patients with
tem, or other such precarious clinical situations. Long-term anti- Raynaud’s secondary to scleroderma (Thomopson et al. 2001). This
coagulation with coumadin or low-molecular-weight heparin is study included six randomized controlled trials of calcium channel
usually unnecessary. blockers (mostly nifedipine) versus placebo. Similar to the analysis
◆ Finally, certain specific forms of vasculitis, such as Buerger’s dis-
of primary Raynaud’s patients, this study demonstrated a signifi-
ease and cryoglobulinaemia, may require additional treatment cant reduction in frequency and severity of symptoms, which was
strategies (smoking cessation or antiviral therapy). Approaches determined to be of moderate clinical utility.
to these individual types of vasculitis are discussed elsewhere in Although few studies have used the sustained release prepara-
this textbook. tions of nifedipine, these agents or other newer dihydropyridine
calcium channel blockers are the initial pharmacological treatment
In Sections Treatment of Raynaud’s Phenomenon and Treatment of choice in practice, because of their greater ease of use (Sturgill
of Digital Ischaemia Caused by Systemic Necrotizing Vasculitis, we and Seibold 1998). Long-acting dihydropyridine calcium-channel
discuss separately the two major therapeutic approaches, vasodila- blockers, for example amlodipine, isradipine, and felodipine have
tion and immunosuppression. not been rigorously tested, but might be preferable to shorter-acting
agents in scleroderma patients because they have less negative ino-
Treatment of Raynaud’s phenomenon tropic effects (La Civita et al. 1993).
The treatment of Raynaud’s phenomenon consists of both phar- If sustained-release calcium-channel blockers are ineffective,
macological and non-pharmacological therapies. Avoidance of the a reasonable second approach is to try intermittent dosing with
cold and other triggering factors are usually sufficient to ameliorate shorter-acting preparations of the same agent, or to switch to a dif-
the problem in primary Raynaud’s phenomenon. The maintenance ferent calcium-channel blocker. The calcium-channel blocker dose
of a warm core body temperature (in addition to warm extremities) must be maximized before an agent is declared a failure. Some cli-
is a critical element in the treatment of this condition. nicians recommend combining different calcium-channel blockers,
Non-pharmacological interventions are important in secondary in recognition of the fact that there are distinct categories of these
Raynaud’s phenomenon as well. Indeed, such strategies are the cor- agents (Spedding and Vanhoutte 1993), but this strategy has not
nerstone of day-to-day management of this problem. Patients with been subjected to a therapeutic trial.
severe Raynaud’s phenomenon must reduce any undue demands A host of other vasodilating drugs, including nitrates (topi-
on the peripheral circulation. Resting the involved limb, reduc- cal and oral) and sympatholytic agents (reserpine, guanethidine,
ing repetitive motion or local trauma, and controlling environ- methyldopa, prazosin, phenoxybenzamine, phentolamine, and
mental factors such as ambient temperature and emotional stress others) have been employed in Raynaud’s phenomenon (Coffman
are essential supporting measures. Patients with severe Raynaud’s and Cohen 1987; McFayden et al. 1973; Russell and Lessard 1985;
phenomenon should also modify their work environments to Varadi and Lawrence 1969). Although many of these agents have
avoid cold temperatures, undue stress or repeated digital trauma. not been studied thoroughly, in general they are less useful than
Although relaxation and behaviour modification (cold avoid- calcium-channel blockers and often have substantial side effects.
ance and stress management), are important, these measures are Nevertheless, in critical situations, we sometimes combine one of
unlikely to be curative in patients with secondary Raynaud’s phe- these agents, for example prazosin, with a calcium-channel blocker.
nomenon. Biofeedback plays no important role in the treatment A systematic review examining the efficacy of prazosin over pla-
of severe Raynaud’s phenomenon (Raynaud’s Treatment Study cebo reported a modest treatment effect with this drug (Pope
Investigators 2000). et al. 1998).
The importance of smoking cessation was addressed by a study For patients with Raynaud’s phenomenon that is refractory to
of patients with Raynaud’s secondary to occupational exposure to non-pharmacological measures and to calcium-channel blockers
a vibratory stimulus (Cherniack et al. 2000). In a cross-sectional there are now several options including prostaglandins, phospho-
examination of 601 current and former vibratory tool users, symp- diesterase inhibitors, selective serotonin reuptake inhibitors, angio-
toms were found to be more severe in those who smoked compared tensin II receptor inhibitors, and angiotensin-converting enzyme
to non-smokers. In a long-term follow-up study of 199 affected inhibitors.
individuals who were all removed from the vibration exposure Results with an intravenous prostacyclin analogue (iloprost)
for 2 years, smokers were twice as likely as non-smokers to have have been encouraging. A double-blind, placebo-controlled study
continued severe vasospasm. Thus, even after removal from the of 131 scleroderma patients found that the weekly attack rate
stimulus that caused Raynaud’s, smokers experienced a delayed decreased by 39% in iloprost-treated patients, compared with
improvement in vasospasm compared with non-smokers. It is a 22% decrease in the placebo group (P  =  0.005) (Wigley et  al.
likely that smoking cessation is beneficial for patients with other 1994). A Raynaud’s phenomenon severity score decreased by 35%
forms of Raynaud’s phenomenon as well. in the iloprost group, compared with 20% in the placebo group
Calcium-channel blockers constitute the first-line pharma- (P = 0.011). The results of a 12-month randomized trial comparing
cological treatment for patients with Raynaud’s phenomenon. cyclic intravenous iloprost with nifedipine were reported in 2001
A meta-analysis of 18 trials examining calcium channel blockers (Scorza et al. 2001). Iloprost was given as an 8-hour infusion daily
for primary Raynaud’s was published in 2005 (Thompson and Pope for 5 days and subsequently as an 8-hour infusion every 6 weeks.
2005). The majority of trials used nifedipine compared with pla- The authors reported significant improvements in skin score and
cebo and an overall reduction in both frequency and severity of Raynaud’s severity in the iloprost group versus the nifedipine
symptoms was demonstrated. Although improvements were sta- group. A recent randomized, open-label study reported low-dose
tistically significant, the clinical efficacy was felt to be relatively iloprost to be equally effective as high-dose iloprost in patients
CHAPTER 16 digital ischaemia and raynaud’s phenomenon 219

with severe Raynaud’s phenomenon and scleroderma (Kawald 2004). Despite this finding, bosentan did not improve symptoms of
et  al. 2008). Intravenous iloprost is currently unavailable in the Raynaud’s phenomenon. A small randomized, placebo-controlled
United States, and two placebo-controlled trials of oral iloprost study also reported lack of improvement in the frequency, duration,
in scleroderma patients did not reveal statistically significant dif- and severity of Raynaud’s phenomenon in scleroderma patients
ferences between the treatment and placebo groups (Belch et al. treated with bosentan (Nguyen et al. 2010).
1995; Wigley et al. 1998a). There are few data regarding the util- In addition to drug therapy for Raynaud’s phenomenon, there has
ity of inhaled iloprost but an open-label study of 20 patients with long been an interest in complementary and alternative modalities
primary or secondary Raynaud’s phenomenon reported a possible such as biofeedback, acupuncture, low-level laser therapy, herbal
benefit in scleroderma but not primary Raynaud’s phenomenon treatment, L-arginine, and antioxidants for primary and secondary
patients (Pakozdi et al. 2008). Larger controlled studies are needed Raynaud’s. A recent meta-analysis of controlled trials examining
to evaluate this agent. these therapies was published (Malenfant et al. 2009). The authors
Sildenafil is a type 5 phosphodiesterase inhibitor that promotes determined that significant flaws in study design precluded any
smooth muscle cell relaxation and vasodilatation. It has been shown meaningful conclusions regarding the efficacy of these treatments.
to be efficacious in a randomized placebo-controlled trial of pul- Longer-term, more-rigorous trials are needed to evaluate the ben-
monary hypertension which included scleroderma patients (Galie efit of alternative modalities.
et al. 2005). There are also now several small studies reporting the In severe cases of Raynaud’s phenomenon that are refractory to
efficacy of this agent in treating patients with Raynaud’s phenom- oral agents, the use of intravenous prostacyclin (PGI2, epopros-
enon (Fries et al. 2005; Gore and Silver 2005; Brueckner et al. 2010; tenol), approved for use in primary pulmonary hypertension,
Herrick et  al. 2011). Tadalafil, another type 5 phosphodiesterase may be considered (Wigley et al. 1998b). This agent was beneficial
inhibitor, has generated recent interest due to its longer half-life. for scleroderma-associated pulmonary hypertension in a rand-
Two randomized, double-blind, placebo-control studies have omized controlled trial that also showed trends towards improved
been published but with opposing conclusions regarding efficacy Raynaud’s severity in these patients (Badesch et al. 2000)
(Schiopu et al. 2009; Shenoy et al. 2010). This may be explained by Some cases of Raynaud’s phenomenon are complicated by digital
differences in trial design. The negative study by Schiopu required ulcerations, with critical ischaemia and demarcation of the involved
patients to discontinue all vasodilators whereas the positive study digit(s). These patients may require hospitalization. Attempts to
by Shenoy allowed continued therapy with other vasodilators. maximize blood flow to the involved extremity should include all of
A subsequent randomized trial demonstrating efficacy of tadalafil the modalities described for the treatment of Raynaud’s phenom-
has been reported in abstract form (Agarwal et al. 2010). enon. Maintenance of rest in a warm environment is essential. In
Serotonin is released by activated platelets as well as nerve end- addition to calcium-channel blockers, we treat patients who have
ings and is a potent vasoconstrictor. There are several case reports ischaemic digital ulcers with aspirin, 325 mg/day. Surgical inter-
of serotonin reuptake inhibitors in the treatment of Raynaud’s as ventions may include temporary lumbar, cervical, or digital sym-
well as a randomized trial of fluoxetine compared with nifedipine pathetic blocks, to determine the potential of sympathectomy for
(Rey et  al. 2003; Garcia-Porrua et  al. 2004; Coleiro et  al. 2001). reversing acute vasopasm (Flatt 1983). We often employ repeated
Fluoxetine resulted in significantly improved frequency and sever- digital nerve blocks in the acute setting, using bupivicaine every 12
ity of Raynaud’s symptoms but nifedipine showed only a trend hours. Permanent sympathectomies may be attempted if the tem-
towards improvement. Follow-up studies are clearly indicated for porary blocks are successful. Digital sympathectomies are safer and
these agents. may be more useful than more proximal procedures (Zachary et al.
Angiotensin converting enzyme inhibitors have demon- 1997). Finally, even if the condition progresses to digital gangrene,
strated mixed results in several studies of these agents (Hummers surgical amputation of digits should be avoided acutely, and auto-
and Wigley 2003). Only one of three placebo controlled trials amputation should be allowed to occur.
reported benefit. A large multicenter, randomized, double-blind, Future therapies will probably be directed against more selected
placebo-controlled trial of the angiotensin converting enzyme targets. There have been reports of several novel agents for treat-
inhibitor quinapril demonstrated no beneficial effects on Raynaud’s ment of Raynaud’s phenomenon in the last several years. Five small
phenomenon or healing of digital ulcers (Gliddon et al. 2007). The uncontrolled studies of botulinum toxin A  treatment have been
results of this large well-performed study largely exclude angio- published (Iorio et al. 2012). This toxin is thought to relax vascular
tensin converting enzyme inhibitors from the armamentarium of smooth muscle cells by blocking depolarization. These small stud-
Raynaud’s therapy. The angiotensin II receptor blocker losartan ies all report improvement in Raynaud’s phenomenon after injec-
has shown efficacy in primary Raynaud’s but less so in scleroderma tion of botulinum toxin; however, lack of controls, small sample
patients. These agents have been used at our institution with mixed size, and retrospective nature of most of these trials preclude any
results. definitive determination of efficacy. A  new formulation of topi-
Endothelin is a small peptide produced by endothelial cells which cal nitroglycerin (MQX-503) was evaluated in a multicenter, ran-
has potent vasoconstrictive properties. The endothelin receptors domized, placebo-controlled study of 219 adults with primary or
can be found on various cell types including endothelial cells, secondary Raynaud’s phenomenon (Chung et al. 2009). This treat-
smooth muscle cells, and fibroblasts, and this peptide may play a ment was well tolerated and more effective than placebo in improv-
role in vascular remodelling and disease pathogenesis. Bosentan, ing symptoms of Raynaud’s phenomenon. Given their pleiotropic
an endothelin receptor antagonist, is effective in treating pulmo- and potentially beneficial effect on vasculature, statins have now
nary hypertension associated with scleroderma (Rubin et al. 2002). been evaluated in a randomized, double-blind, placebo controlled
In addition, this drug is effective in preventing new digital ulcers trial of atorvastatin versus placebo over 4 months in patients with
and improving hand function in scleroderma patients (Korn et al. scleroderma and secondary Raynaud’s phenomenon (Abou-Raya
220 SECTION 3 clinical manifestations common to vasculitis

et al. 2008). This trial reported significant benefit in measures of achieve a reduction in the frequency and severity of attacks. We
Raynaud’s and digital ulcer severity in the treatment group com- have found that some patients require higher than normal dosage,
pared to placebo. such as 20 mg of amlodipine daily, to obtain a therapeutic response.
A small study of an investigational agent has focused treatment It is important to set the proper therapeutic expectations with each
on advances in the pathophysiology of cold-induced vasospasm medication. Patients need to understand that the drugs used do
(Wise et al. 2004). This study examined the time to recovery of skin not cure or eliminate Raynaud’s phenomenon but can reduce the
temperature after a cold challenge in scleroderma patients given a frequency and severity of attacks and limit the occurrence of digi-
selective inhibitor of the α2C receptor. As discussed in the Section tal ischaemic ulcers. If calcium channel blockers at the maximum
Pathogenesis of this chapter, the α2C adrenergic receptor is thought tolerated dose are not sufficient to control symptoms, we will move
to mediate vasoconstriction in response to cold. Patients given the on to a second-line agent. Our practice is to utilize phosphodi-
study drug had shorter temperature recovery times compared to esterase inhibitors as second-line drugs unless there is a medical
patients given placebo. Larger studies are needed to define the reason to consider other agents, such as patients with angina who
role of this agent in Raynaud’s phenomenon, but this represents a might benefit from topical nitroglycerin or those with benign pro-
potential advance in selective targeting of drugs to the relevant dis- static hypertrophy who might benefit from prazosin. Sildenafil is
ease causing mechanism. Inhibitors of the RhoA/Rho kinase sys- typically started at 25 mg three times daily and titrated up to 50 mg
tem that allows translocation of the α2C receptor to the cell surface daily three times daily as tolerated to reach the desirable effect. For
are currently under investigation. The RhoA/Rho kinase inhibitor patients who have issues with insurance coverage and cost, we will
fasudil was studied in a double-blind, randomized, crossover labo- also consider tadalafil every other day in order to take advantage
ratory study of patients with scleroderma and Raynaud’s phenom- of its relatively long half-life. Patients are always warned to avoid
enon (Fava et al. 2012). This study found no improvement in the any nitrates when taking a phosphodiesterase inhibitor. If a first-
time to skin temperature recovery or digital blood flow after a cold or second-line drug is not effective, combination therapy will be
challenge. It is currently unknown whether this agent would be considered. Amlodipine with sildenafil is a common therapy in
efficacious in improving Raynaud’s phenomenon in the real world our clinics. A  calcium channel blocker plus nitroglycerin, losar-
setting. Lastly, antioxidants may be helpful in that oxidative stress tan, or prazosin, or even a combination of three or more agents, is
stimulates RhoA/Rho kinase signalling and mobilization of α2C to sometimes needed for difficult cases. In patients with a low blood
the cell surface (Bailey et. al. 2005). Preliminary studies with probu- pressure or depression or anxiety, a selective serotonin reuptake
col (Denton et al. 1999) and N-acetylcysteine (Sambo et al. 2001; inhibitor such as fluoxetine is often employed. Low-dose aspirin is
Rosato et al. 2009) suggest benefit of antioxidant therapy. typically added for patients with a history of digital ulcers. Clinical
trials are also considered, especially for patients with refractory
Our practice Raynaud’s phenomenon.
This section describes the author’s current practice based on evi- Patients who suffer critical digital ischaemia that is not relieved
dence sited above, biological considerations, and clinical experi- by oral agents will often be admitted to the hospital. Patients are
ence. When first discussing treatment with patients, we review the placed in a warm room, preferably away from a busy nursing sta-
importance of education and understanding. Patients are provided tion in order to minimize stress. The patient is then started on
with a brief overview of normal body temperature regulation prin- intravenous epoprostenol at 2 ng/kg/min which is then gradually
ciples as well as an educational overview of Raynaud’s phenomenon titrated to therapeutic effect or the development of side-effects such
mechanisms, stressing the importance of temperature variance. It as flushing or headaches. Patients are typically maintained on this
is our view that patient understanding is essential for good thera- infusion for 5 days while in the hospital. Although there is a lack
peutic outcomes. Patients must know what factors to avoid such of rigorous evidence, these patients without an undue bleeding risk
as handling cold beverages or the need to wear thin cotton gloves are also anticoagulated with heparin in order to minimize throm-
when visiting the grocery store. They are also instructed in meth- bosis caused by severe vasospasm and endothelial damage. In
ods to optimize core and peripheral body temperature. This may selected patients we move to digital sympathectomy when they are
include the use of hand warming packets inside of gloves or socks not responding to medical therapy. After discharge from the hos-
or running warm water over the hands to help resolve an attack pital, patients are maintained on oral vasodilators as well as anti-
of Raynaud’s phenomenon. We also strongly urge patients who platelet agents such as 81 mg aspirin. For those patients who suffer
smoke cigarettes that they must stop smoking or they can expect a digital gangrene, the necrotic digit is allowed to autoamputate in
poor outcome. Patients are given information resources to continue order to preserve as much viable tissue as possible. However, for
self-education after the visit and directed to review reliable websites patients who develop infection involving the necrotic tissue, surgi-
such as http://www.hopkinsscleroderma.org. cal removal is usually necessary.
Once patients demonstrate an understanding of underlying
principles, we move to a discussion of pharmacological options. Treatment of digital ischaemia caused
For patients with mild attacks and no history of digital ischaemic by systemic necrotizing vasculitis
ulcers, we often recommend against drug therapy. However, for Patients presenting with digital ischaemia of a suspected inflamma-
those with frequent and bothersome symptoms and for those with tory cause should be hospitalized. Analogous to the treatment of
digital ulcers, we begin treatment with a dihydropyridine calcium myocardial infarction, the first 24 hours after presentation to a phy-
channel blocker such as amlodipine. This drug is typically started sician may be critical in determining outcome. In addition to the
at 5 mg daily (2.5 mg in patients with low baseline blood pressure). attention that must be directed to treatment of the digital ischae-
We will warn patients of side-effects such as dizziness, hypotension, mia and to pain control, patients must be carefully assessed for the
or peripheral oedema. We then titrate up the dose as tolerated to extent and severity of disease in other organs. Patients presenting
CHAPTER 16 digital ischaemia and raynaud’s phenomenon 221

with digital ischaemia often have rapidly progressive disease of the touch for years, without advancement of the ischaemic process or
kidneys, lungs, nerves, and gastrointestinal tract. further threats to tissue viability. In such cases, a stable pattern of
The non-pharmacological treatments discussed with regard to digital coolness, that is involvement of the same fingers or toes from
the treatment of secondary Raynaud’s phenomenon are directly one visit to the next, rather than spread of involvement to previ-
applicable to the management of digital ischaemia in vasculitis ously uninvolved digits, is reassuring.
as well. A  warm environment is essential, and substantial nar- Vasodilator therapy can be considered in patients with vasculitis
cotic analgesia may be required for some weeks. The first line of who have Raynaud’s phenomenon or evidence of reversible vasos-
treatment for digital ischaemia associated with systemic necrotiz- pasm directly related to the inflammatory vascular disease. Caution
ing vasculitis is high-dose glucocorticoid therapy. In practice, we is recommended, however, as there is concern that vasodilator ther-
employ 3-day intravenous pulses of methylprednisolone in patients apy could decrease blood flow to affected tissue. If there is fixed ves-
with severe, digit- or limb-threatening ischaemia. In patients sel narrowing due to necrotizing inflammation of the blood vessel
with less severe ischaemia and no evidence of other critical organ wall, vasodilators may preferentially expand unaffected vessels cre-
involvement, we sometimes give lower glucocorticoid doses, such ating a vascular steal phenomenon, which could reduce perfusion
as methylprednisolone, 1 mg/kg/day, or oral daily prednisone. As a to tissue supplied by diseased vasculature.
rule, in the absence of significant contraindications to high doses Necrotic fingertips and toes may require many months for the
of glucocorticoids, it is proper to err on the side of administering painful process of autoamputation to occur. Months after the
too much glucocorticoid treatment in the first few days, because inflammatory process has been arrested, selected patients with dig-
the side-effects of such therapy are usually well-tolerated (or man- ital necrosis secondary to vasculitis may be candidates for surgical
ageable) in the short-term and are unlikely to be lasting. However, removal of necrotic tissue. This procedure provides substantial pain
careful attention must be paid to the management of potential relief and causes little loss of viable tissue, provided that the process
side-effects of longer-term glucocorticoid treatment such as osteo- of autoamputation has advanced sufficiently.
porosis prevention, careful glucose and blood pressure monitoring,
and prompt treatment of infections. References
Some patients with necrotizing vasculitis, particularly those Abou-Raya, A., Abou-Raya, S., and Helmii, M. (2008). Statins: Potentially
whose disease involvement is limited to the extremities, respond useful in therapy of systemic sclerosis-related Raynaud’s phenomenon
and digital ulcers. Journal of Rheumatology, 35, 1801–8.
well to glucocorticoids alone. These patients typically have dra-
Agarwal, V., Ghosh, P., Sharma, A., et  al. (2010). Efficacy of tadalafil in
matic responses within 24 hours of glucocorticoid administration. Raynaud’s phenomenon secondary to systemic sclerosis. Arthritis and
Others, however, manifest only moderate responses to glucocorti- Rheumatism, 62S, S872
coids and, at the completion of a 3-day methylprednisolone pulse, Aldoori, M., Bruce, W., and Dieppe, P. (1986). Nifedipine in the treatment of
fail to demonstrate sustained improvement. These patients, as well Raynaud’s syndrome. Cardiovascular Research, 20, 466–9.
as those with evidence of organ- or life-threatening disease in other Allanore, Y., Batteux, F., Avouac, J., et  al. (2007). Levels of circulating
systems, should be treated with cyclophosphamide or possibly endothelial progenitor cells in systemic sclerosis. Clinical Experimental
B-cell therapy with rituximab, in addition to glucocorticoids (Stone Rheumatology, 25, 60–6.
et al. 2010). For vasculitis patients receiving the combination of glu- Allen, E. and Brown, G. (1932a). Raynaud’s disease: a critical review of minor
cocorticoids and cyclophosphamide, we routinely administer tri- requisites for diagnosis. American Journal of the Medical Sciences, 183,
187–200.
methoprim–sulfamethoxazole (TMP/SMZ) (one double-strength
Allen, E. and Brown, G. (1932b). Raynaud’s disease:  A  clinical study of
tablet by mouth each day) as prophylaxis against Pneumocystis one hundred and forty-seven cases. Journal of the American Medical
jirovecii pneumonia. Patients who are allergic to TMP/SMZ may Association, 99, 1472–8.
receive instead dapsone 100 mg daily. Andreu-Sanchez, J., Martin-Santos, J., Isasi-Zaragoza, C., Trujillo-Castellanos,
Regardless of their initial response to glucocorticoids, all A., Cuende-Quintana, E., and Mulero, J. (1991). Raynaud’s phenomenon
patients with digital ischaemia must be followed carefully until as initial manifestation of cutaneous polyarteritis nodosa. Annals of the
their disease is under good control. Digital ischaemia may herald Rheumatic Diseases, 50, 48–50.
organ-threatening vasculitis in other systems that require cyclo- Asano, Y., Stawski, L., Hant, F., et al. (2010). Endothelial Fli1 deficiency impairs
phosphamide or rituximab, and such organ involvement may not vascular homeostasis. American Journal of Pathology, 176, 1983–98.
Avouac, J., Juin, F., Wipff, J., et al. (2008). Circulating endothelial progeni-
be evident at presentation. Consequently, we re-evaluate patients
tor cells in systemic sclerosis. Annals of the Rheumatic Diseases, 67,
within 1 week following discharge from the hospital. The duration 1455–60.
of immunosuppressive therapy for each patient must be individ- Badesch, D.B., Tapson, V.F., McGoon, M.D., et al. (2000). Continuous intra-
ualized, but may be predicted in large measure from the precise venous epoprostenol for pulmonary hypertension due to the sclero-
type of underlying vasculitis. Whereas the ANCA-associated vas- derma spectrum of disease. Annals of Internal Medicine, 132, 425–34.
culitides generally require concomitant glucocorticoids and cyclo- Bailey, S.R., Eid, A.H., Mitra, S., Flavahan, S., and Flavahan, N.A. (2004).
phosphamide or rituximab, other forms of vasculitis may respond Rho kinase mediates cold-induced constriction of cutaneous arteries.
adequately to prednisone alone. These issues are discussed in other Circulation Research, 94, 1367–74.
chapters of this book. Bailey, S.R., Mitra, S., Flavahan, S., and Flavahan, N.A. (2005). Reactive oxy-
gen species from smooth muscle mitochondria initiate cold-induced
In the weeks after the initiation of treatment, assessment of dis-
constriction of cutaneous arteries. American Journal of Physiology Heart
ease activity in patients with digital vasculitis presents a challeng- and Circulatory Physiology, 289, 243–50.
ing task. Blood vessels damaged by the acute vasculitic injury may Bartelink, M., Wollersheim, H., van de Lisdonk, E., et al. (1992). Prevalence
never return to their premorbid condition. Fingers involved by the of Raynaud’s phenomenon. Netherlands Journal of Medicine, 41, 149–52.
inflammation become cyanotic when exposed to cold for months Bedarida, G., Kim, D., Blaschke, T., et al. (1993). Venodilation in Raynaud’s
afterward, and even in normal temperatures may remain cool to disease. Lancet, 342, 1451–4.
222 SECTION 3 clinical manifestations common to vasculitis

Belch, J. (1990). The phenomenon, syndrome and disease of Maurice Flatt, A. (1983). Digital arterial sympathectomy. Journal of Hand Surgery,
Raynaud. British Journal of Rheumatology, 29, 162–5. 8, 283–8.
Belch, J., Capell, H., Cooke, E., et al. (1995). Oral iloprost as a treatment for Flavahan, N.A. (2008). Regulation of vascular reactivity in scleroderma.
Raynaud’s syndrome:  A  double-blind multicentre placebo-controlled Rheumatic Disease Clinics of North America, 34, 81–87.
study. Annals of the Rheumatic Diseases, 54, 197–200. Flavahan, N.A., Flavahan, S., Liu, Q., Wu, S., Tidmore, W., Wiener, C.M.,
Bombardier, C., Gladman, D., Urowitz, M., Caron, D., Chang, C., et  al. Spence, R.J., and Wigley, F.M. (2000). Increased α2-adrenergic con-
(1989). Derivation of the SLEDAI:  A  disease activity index for lupus striction of isolated arterioles in diffuse scleroderma. Arthritis and
patients. Arthritis and Rheumatism, 35, 630–40. Rheumatism, 43, 1886–90.
Bou-Gharios, G., Ponticos, M., Rajkumar, V., and Abranham, D. (2004). Flavahan, N.A., Flavahan, S., Mitra, S., and Chotani, M. (2003). The vascu-
Extra-cellular matrix in vascular networks. Cell Proliferation, 37, 207–20. lopathy of Raynaud’s phenomenon and scleroderma. Rheumatic Disease
Brueckner, C., Becker, M., Kroencke, T., et al. (2010). Effect of sildenafil on Clinics of North America, 29, 275–91.
digital ulcers in systemic sclerosis. Annals of the Rheumatic Diseases, 69, Flavahan, N.A., Lindblad, L.E., Verbeuren, T.J., et  al. (1985). Cooling
1475–78. and α1—and α2-adrenergic responses in cutaneous veins:  Role of
Bunker, C., Terenghi, G., Springall, D., et al. (1990). Deficiency of calcitonin receptor-reserve. American Journal of Physiology, 249, H950–5.
gene-related peptide in Raynaud’s phenomenon. Lancet, 336, 1530–3. Fraenkel, L., Zhang, Y., Chaisson, C.E., et al. (1998). The association of estro-
Cherniack, M., Clive, J., and Seidner, A. (2000). Vibration exposure, smoking, gen replacement therapy and the Raynaud’s phenomenon in postmeno-
and vascular dysfunction. Occupational and Environmental Medicine, pausal women. Annals of Internal Medicine, 129, 208–11.
57, 341–47. Freedman, R., Baer, R., and Mayes, M. (1995). Blockade of vasospastic
Chotani, M., Flavahan, S., Mitra, S., et al. (2000). Silent alpha(2C)-adrenergic attacks by α2-adrenergic but not α1-adrenergic antagonists in idiopathic
receptors enable cold-induced vasoconstriction in cutaneous arteries. Raynaud’s disease. Circulation, 92, 1448–51.
American Journal of Physiology Heart and Circulatory Physiology, 278, Freedman, R., Girgis, R., and Mayes, M. (1999). Acute effect of nitric oxide
H1075–83. on Raynaud’s phenomenon in scleroderma. Lancet, 354, 739.
Chung, L., Shapiro, L., Fiorentin, D., et al. (2009). MQX-503, a novel formula- Freedman, R., Moten, M., Migaly, P., et al. (1993). Cold-induced potentia-
tion of nitroglycerin, improves the severity of Raynaud’s phenomenon. tion of α2-adrenergic vasoconstriction in primary Raynaud’s disease.
Arthritis and Rheumatism, 60, 870–77. Arthritis and Rheumatism, 36, 685–90.
Cipriani, P., Guiducci, S., Miniati, I., et al. (2007). Impairment of endothelial Freemont, A., Hoyland, J., Fielding, P., et al. (1992). Studies of microvascu-
cell differentiation from bone marrow-derived mesenchymal stem cells. lar endothelium in uninvolved skin of patients with systemic sclero-
Arthritis and Rheumatism, 56, 1994–2004. sis: Direct evidence for a generalized microangiopathy. British Journal of
Coffman, J. and Cohen, R. (1987). Intra-arterial vasodilator agents to Dermatology, 126, 561–8.
reverse finger vasoconstriction. Clinical Pharmacological Therapy, 41, Friedman, E.I., Taylor, L.M. Jr., and Porter, J.M. (1988). Late-onset Raynaud’s
574–88. syndrome:  Diagnostic and therapeutic considerations. Geriatrics,
Coffman, J. and Cohen, R. (1988). Role of alpha-adrenoceptor subtypes 43, 59–70.
mediating sympathetic vasoconstriction in human digits. European Fries, R., Shariat, K., von Wilmowsky, H., and Bohm, M. (2005). Sildenafil
Journal of Clinical Investigation, 18, 309–13. in the treatment of Raynaud’s phenomenon resistant to vasodilatory
Coleiro, B., Marshall, S.E., Denton, C.P., et al. (2001). Treatment of Raynaud’s therapy. Circulation, 112, 2980–85.
phenomenon with the selective serotonin reuptake inhibitor fluoxetine. Furspan, P.B., Chatterjee, S., and Freedman, R.R. (2004). Increased tyros-
Rheumatology, 40, 1038–43. ine phosphorylation mediates the cooling-induced contraction and
De Angelis, R., Del Medico, P., Blasetti, P., and Cervini, C. (2003). Raynaud’s increased vascular reactivity of Raynaud’s disease. Arthritis and
phenomenon: clinical spectrum of 118 patients. Clinical Rheumatology, Rheumatism, 50, 1578–85.
22, 279–84. Furspan, P.B., Chatterjee, S., Mayes, M.D., and Freedman, R.R. (2005).
Del Papa, N., Quirici, N., Soligo, D., et al. (2006). Bone marrow endothelial Cooling-induced contraction and protein tyrosine kinase activity of iso-
progenitors are defective in systemic sclerosis. Arthritis and Rheumatism, lated arterioles in secondary Raynaud’s phenomenon. Rheumatology, 44,
54, 2605–15. 488–94.
Denton, C.P., Bunce, T.D., Darado, M.B., et al. (1999). Probucol improves Galie, N., Ghofrani, H.A., Torbicki, A., et al. (2005). Sildenafil citrate therapy
symptoms and reduces lipoprotein oxidation susceptibility in patients for pulmonary arterial hypertension. New England Journal of Medicine,
with Raynaud’s phenomenon. Rheumatology, 38, 309–15. 353, 2148–57.
Distler, J., Allanore, Y., Avouac, J., et al. (2009). EULAR Scleroderma Trials Garcia-Porrua, C., Margarinos, C.C., and Gonzalez-Gay, M.A. (2004).
and Research group statement and recommendations on endothelial Raynaud’s phenomenon and serotonin reuptake inhibitors. Journal of
precursor cells. Annals of the Rheumatic Diseases, 68, 163–8. Rheumatology, 31, 2090–91.
Eid, A.H., Maiti, K., Mitra, S., et al. (2007). Estrogen increases smooth muscle Gelber, A., Wigley, F., Stallings, R., Bone, L., Barker, A., Baylor, I., et  al.
expression of α2c-adrenoceptors and cold-induced constriction of cuta- (1999). Raynaud’s phenomenon in an inner-city African American com-
neous arteries. American Journal of Physiology Heart and Circulatory munity: Prevalence of symptoms, health status, and co-morbid cardio-
Physiology, 293, 1955–61. vascular disease. Journal of Clinical Epidemiology, 52, 441–6.
Ekenvall, L., Lindblad, L., Norbreck, O., et al. (1988). α-Adrenoceptors and Generini, S., Seibold, J., and Matucci-Cerinic, M. (2005). Estrogens and
cold-induced vasoconstriction in human finger skin. American Journal neuropeptides in Raynaud’s phenomenon. Rheumatic Disease Clinics of
of Physiology, 255, H1000–3. North America, 31, 177–86.
Fava, A., Wung, P., Wigley, F., et al. (2012). Efficacy of Rho kinase inhibi- Gliddon, A., Dore, C., Black, C., et al. (2007). Prevention of vascular damage
tor fasudil in secondary Raynaud’s phenomenon. Arthritis Care and in scleroderma and autoimmune Raynaud’s phenomenon. Arthritis and
Research, 64, 925–29. Rheumatism, 56, 3837–46
Filippov, S., Koenig, G.C., Chun, T., et al. (2005). MT1-matrix metallopro- Gore, J. and Silver, R. (2005). Oral sildenafil for the treatment of Raynaud’s
teinase directs arterial wall invasion and neointima formation by vascu- phenomenon and digital ulcers secondary to systemic sclerosis. Annals
lar smooth muscle cells. Journal of Experimental Medicine, 202, 663–71. of the Rheumatic Diseases, 64, 1387.
Finch, M., Dawson, J., Johnston, G., et al. (1986). The peripheral vascular Grassi, W., De Angelis, R., Lapadula, G., Leardini, G., and Scarpa, R. (1998).
effects of nifedipine in Raynaud’s syndrome associated with sclero- Clinical diagnosis found in patients with Raynaud’s phenomenon:  a
derms: A double-blind crossover study. Clinical Rheumatology, 5, 493. multicenter study. Rheumatology International, 18, 17–20.
CHAPTER 16 digital ischaemia and raynaud’s phenomenon 223

Hadler, N. (1998). ‘Primary Raynaud’s’ is not a disease or even a disorder; It’s Malenfant, D., Catton, M., and Pope, J. (2009). The efficacy of complemen-
a trait. Journal of Rheumatology, 25, 2291–4. tary and alternative medicine in the treatment of Raynaud’s phenom-
Hebbar, M., Peyrat, J.P., Hornez, L., et al. (2000). Increased concentrations enon. Rheumatology, 48, 791–95.
of the circulating angiogenesis inhibitor endostatin in patients with sys- Maricq, H., Carpentier, P., Weinrich, M., et al. (1993). Geographic variation
temic sclerosis. Arthritis and Rheumatism, 43, 889–93. in the prevalence if Raynaud’s phenomenon: Charleston, SC, USA, ver-
Herrick, A., Illingworth, K., Blann, A., et al. (1996). Von Willebrand factor, sus Tarentaise, Savoie, France. Journal of Rheumatology, 20, 70–6.
thrombomodulin thromboxane, B-thromboglobulin, and markers of Maricq, H., McGregor, A., Diat, F., Smith, E., Maxwell, D., LeRoy, C., et al.
fibrinolysis in primary Raynaud’s phenomenon and systemic sclerosis. (1990). Major clinical diagnoses found among patients with Raynaud’s
Annals of the Rheumatic Diseases, 55, 122–7. phenomenon from the general population. Journal of Rheumatology, 17,
Herrick, A., van den Hoogen, F., Gabrielli, A., et al. (2011). Modified-release 1171–6.
sildenafil reduces Raynaud’s phenomenon attack frequency in limited Maricq, H., Weinrich, M., Keil, J., et al. (1989). Prevalence of scleroderma
cutaneous systemic sclerosis. Arthritis and Rheumatism, 63, 775–82. spectrum disorders in the general population of South Carolina.
Hirschl, M., Katzenschlager, R., Francesconi, C., and Kundi, M. (2004). Arthritis and Rheumatism, 32, 998–1006.
Low level laser therapy in primary Raynaud’s phenomenon—results Maurer, B., Busch, N., Jungel, A., et al. (2009). Transcription factor fos-related
of a placebo controlled double blind intervention study. Journal of antigen-2 induces progressive peripheral vasculopathy in mice closely
Rheumatology, 31, 2408–12. resembling human systemic sclerosis. Circulation, 120, 2367–76.
Hummers, L.K. and Wigley, F.M. (2003). Management of Raynaud’s phenom- McFayden, I., Housley, E., and Macherson, A. (1973). Intra-arterial reserpine
enon and digital ischemic lesions in scleroderma. Rheumatic Disease administration on Raynaud’s syndrome. Archives of Internal Medicine,
Clinics of North America, 29, 293–313. 132, 256–60.
Hutchinson, J. (1896). Acro-scleroderma following Raynaud’s phenomenon. Mohrland, J., Porter, J., Smith, E., et  al. (1985). A multiclinic,
Clinical Journal, 7, 240. placebo-controlled, double-blind study of prostaglandin E1 in RP.
Hutchinson, J. (1901). Raynaud’s phenomenon. Medical Press, 123, 403–5. Annals of the Rheumatic Diseases, 44, 754–60.
Inaba, R., Maeda, M. Fujita, S., Kashiki, N., Komura, Y., Nagata, C., et al. Nguyen, V., Eisendle, K., Gruber, I., et al. (2010). Effect of the dual endothelin
(1993). Prevalence of Raynaud’s phenomenon and specific clinical signs receptor antagonist bosentan on Raynaud’s phenomenon secondary to
related to progressive systemic sclerosis in the general population of systemic sclerosis. Rheumatology, 49, 583–87.
Japan. International Journal of Dermatology, 32, 652–5. Onbasi, K., Sahin, I., Onbasi, O., Ustun, Y., and Koca, D. (2005). Raynaud’s
Iorio, M., Masden, D., and Higgins, J. (2012). Botulinum toxin A treatment phenomenon in a healthy Turkish population. Clinical Rheumatology,
of Raynaud’s phenomenon. Seminars in Arthritis and Rheumatism, 41, 24, 365–69.
599–603 Pakozdi, A., Howell, K., Wilson, H., et al. (2008). Inhaled iloprost for the
Jennette, J., Falk, R., Andrassy, K., et al. (1994). Nomenclature of systemic treatment of Raynaud’s phenomenon. Clinical and Experimental
vasculitides. Proposal of an international consensus conference. Arthritis Rheumatology, 26, 709.
and Rheumatism, 37, 187–92. Pavlov-Dolijanovic, S., Damjanov, N., Stojanovic, R., et  al. (2012).
Jeyaraj, S.C., Chotani, M.A., Mitra, S., Gregg, H.E., Flavahan, N.A., and Scleroderma pattern of nailfold capillary changes as predictive value
Morrison, K.J. (2001). Cooling evokes redistribution of α2C-adrenoceptors for the development of a connective tissue diseases. Rheumatology
from golgi to plasma membrane in transfected human embryonic kidney International, 32, 3039–45.
293 cells. Molecular Pharmacology, 60, 1195–1200. Pearson, J. (1991). The endothelium: Its role in scleroderma. Annals of the
Kawald, A., Burmester, G., Huscher, D., et al. (2008). Low versus high-dose Rheumatic Diseases, 50, 866–71.
iloprost therapy over 21 days in patients with secondary Raynaud’s phe- Philipp, M., Brede, M., and Hein, L. (2002). Physiological significance of
nomenon and systemic sclerosis. Journal of Rheumatology, 35, 1830–37. α2-adrenergic receptor subtype diversity: one receptor is not enough.
Korn, J.H., Mayes, M., Cerinic, M.M., et al. (2004). Digital ulcers in systemic American Journal of Physiology Regulatory, Integrative, and Comparative
sclerosis. Arthritis and Rheumatism, 50, 3985–93. Physiology, 283, R287–95.
Kuwana, M., Okazaki, Y., Yasuoka, H., Kawakami, Y., and Ikeda, Y. (2004). Pineda, C., Weisman, M., Bookstein, J., and Saltzstein, S. (1985). Hypothenar
Defective vasculogenesis in systemic sclerosis. Lancet, 364, 603–10. hammer syndrome. Form of reversible Raynaud’s phenomenon.
La Civita, L., Pitaro, N., Rossi, M., et al. (1993). Amlodipine in the treatment American Journal of Medicine, 79, 561–70.
of Raynaud’s phenomenon. British Journal of Rheumatology, 32, 524–5. Pope, J., Fenlon, D., Thompson, A., et  al. (1998). Prazosin for Raynaud’s
Lekakis, J., Mavrikakis, M., Papamichael, C., et al. (1998). Short-term estro- phenomenon in progressive systemic sclerosis. Cochrane Database of
gen administration improves abnormal endothelial function in women Systematic Reviews, 2, CD000956.
with systemic sclerosis and Raynaud’s phenomenon. American Heart Porter, J., Bardana, E., Baur, G., et  al. (1976). The clinical significance of
Journal, 136, 905–12. Raynaud’s syndrome. Surgery, 80, 756–64.
Leppert, J., Aberg, H., Ringqvist, I., and Sorensson, S. (1987). Raynaud’s phe- Prescott, R., Freemont, A., Jones, C., et al. (1992). Sequential dermal micro-
nomenon in a female population: Prevalence and association with other vascular and perivascular changes in the development of scleroderma.
conditions. Angiology, 38, 871–7. Journal of Pathology, 166, 255–63.
LeRoy, E. and Medsger, T. (1992). Raynaud’s phenomenon: A proposal for Purdie, G., Harrison, A., and Purdie, D. (2009). Prevalence of Raynaud’s phe-
classification. Clinical and Experimental Rheumatology, 10, 485–8. nomenon in the adult New Zealand population. New Zealand Medical
Lewis, T. and Pickering, G. (1933). Observations upon maladies in which the Journal, 122, 55–62.
blood supply to digits ceases intermittently or permanently, and upon Rademaker, M., Cooke, E., Almond, N., et al. (1989). Comparison of intra-
bilateral gangrene of digits; observations to so called ‘Raynaud’s disease’. venous infusions of iloprost and oral nifedipine in the treatment of RP
Clinical Science, 1,327–66. in patients with systemic sclerosis. British Medical Journal, 298, 561–3.
Lie, J. (1994). Nomenclature and classification of vasculitis: Plus ca change, Raynaud, M. (1862). De l’asphyxie locale et de la gangréne symétrique des
plus c’est la meme chose. Arthritis and Rheumatism, 37, 181–6. extrémités. Rignoux, Paris.
Macko, R.F., Gelber, A.C., Young, B.A., et  al. (2002). Increased circu- Raynaud’s Treatment Study Investigators (2000). Comparison of
lating concentrations of the counteradhesive proteins sparc and sustained-released nifedipine and temperature biofeedback for treat-
thrombospondin-1 in systemic sclerosis (scleroderma). Relationship ment of primary Raynaud phenomenon:  Results from a randomized
to platelet and endothelial cell activation. Journal of Rheumatology, 29, clinical trial with 1-year follow-up. Archives of Internal Medicine,
2565–70. 160, 1101–8
224 SECTION 3 clinical manifestations common to vasculitis

Reilly, I., Roy, L., and Fitzgerald, G. (1986). Biosynthesis of thromboxane Su, B., Mitra, S., Gregg, H., et al. (2001). Redox regulation of vascular smooth
in patients with systemic sclerosis and Raynaud’s phenomenon. British muscle cell differentiation. Circulation Research, 89, 39–46.
Medical Journal, 292, 1037–9. Suter, L.G., Murabito, J.M., Felson, D.T., and Fraenkel, L. (2005). The inci-
Rey, J., Cretel, E., Jean, R., Pastor, M.J., and Durand, J.M. (2003). Serotonin dence and natural history of Raynaud’s phenomenon in the community.
reuptake inhibitors, Raynaud’s phenomenon and erythromelalgia. Arthritis and Rheumatism, 52, 1259–63.
Rheumatology, 42, 601–2. Thompson, A.E. and Pope, J.E. (2005). Calcium-channel blockers for Primary
Riera, D., Vilardell, M., Vaque, J., et al. (1993). Prevalence of Raynaud’s phe- Raynaud’s phenomenon: a meta-analysis. Rheumatology, 44, 145–50.
nomenon in a healthy Spanish population. Journal of Rheumatology, Thompson, A.E., Shea, B., Welch, V., Fenlon, D., and Pope, J.E. (2001).
20, 66–9. Calcium-channel blockers for Raynaud’s phenomenon in systemic scle-
Rosato, E., Borghese, F., Pisarri, S., and Salsano, F. (2009). The treatment with rosis. Arthritis and Rheumatism, 44, 1841–47.
N-acetylcysteine of Raynaud’s phenomenon and ischemic ulcers therapy Valter, I. and Maricq, H. (1997). Prevalence of Raynaud phenomenon in Tartu
in sclerodermic patients. Clinical Rheumatology, 28, 1379–84. and Tartumaa, southern Estonia. Scandinavian Journal of Rheumatology,
Rubin, L.J., Badesch, D.B., Barst, R.J., et al. (2002). Bosentan therapy for pul- 26, 117–24.
monary arterial hypertension. New England Journal of Medicine, 346, Van Vugt, R.M., Kater, L., Dijkstra, P.F., Schardijn, G.H.C., Kastelein, J.J.P.,
896–903. and Bijlsma, J.W.J. (2003). The outcome of angiography in patients with
Russell, L. and Lessard, J. (1985). Prazosin treatment of Raynaud’s phenom- Rayndaud’s phenomenon: An unexpected role for atherosclerosis and
enon: A double-blind single crossover study. Journal of Rheumatology, hypercholesterolemia. Clinical and Experimental Rheumatology, 21,
12, 94–102. 445–50.
Sambo, P., Amico, D., Giacomelli, R., et  al. (2001). Intravenous Varadi, D. and Lawrence, A. (1969). Suppression of Raynaud’s phenomenon
N-acetylcysteine for treatment of Raynaud’s phenomenon secondary to by methyldopa. Archives of Internal Medicine, 124, 13.
systemic sclerosis: a pilot study. Journal of Rheumatology, 28, 2257–62. Wigley, F. (2002). Raynaud’s Phenomenon. New England Journal of Medicine,
Sauza, J., Kraus, A., Gonzales-Amaro, R., et al. (1984). Effect if the calcium 347, 1001–08.
channel blocker nifedipine in Raynaud’s phenomenon: A double-blind Wigley, F., Badesch, D., Rubin, L., et al. (1998a). The effect of epoprostenol
trial. Journal Rheumatology, 11, 362–8. (E) on Raynaud’s phenomenon (RP) in patients with scleroderma spec-
Schiopu, E., Hsu, V., Impens, A., et al. (2009). Randomized placebo-controlled trum of disease (SSD) and pulmonary hypertension (PH). Arthritis and
crossover trial of tadalafil in Raynaud’s phenomenon secondary to sys- Rheumatism, 41, S1243.
temic sclerosis. Journal of Rheumatology, 36, 2264–68. Wigley, F., Korn, J., Csuka, M., Medsger, T., Rothfield, N., Ellman, M., et al.
Scorza, R., Caronni, M., Mascagni, B., et  al. (2001). Effects of long-term (1998b). Oral iloprost in patients with Raynaud phenomenon secondary
cyclic iloprost therapy in systemic sclerosis with Raynaud’s phenom- to systemic sclerosis: A multicenter, placebo-controlled, double-blind
enon. A  randomized, controlled study. Clinical and Experimental study. Arthritis and Rheumatism, 41, 670–7.
Rheumatology, 19, 503–08. Wigley, F., Wise, R., Seibold, J., et al. (1994). Intravenous iloprost onfusion
Shenoy, P., Kumar, S., Jha, L., et al. (2010). Efficacy of tadalafil in secondary in patients with RP secondary to systemic sclerosis. Annals of Internal
Raynaud’s phenomenon resistant to vasodilator therapy. Rheumatology, Medicine, 120, 199–206.
49, 2420–28. Wise, R.A., Wigley, F.M., White, B., et al. (2004). Efficacy and tolerability of a
Spedding, M. and Vanhoutte, P. (1993). Nomenclature of calcium chan- selective α2C-adrenergic receptor blocker in recovery from cold-induced
nel and channel modulators. Journal of Cardiovascular Pharmacology, vasospasm in scleroderma patients. Arthritis and Rheumatism, 50,
22, 906–8. 3994–4001.
Stone, J., Merkel, P., Spiera, R., et al. (2010). Rituximab versus cyclophospha- Zachary, L., Rice-Puoci, F., and Ellman, M. (1997). Digital sympathectomy
mide for ANCA-associated vasculitis. New England Journal of Medicine, for fingertip ulcerations/infarctions in scleroderma patients. Arthritis
363, 221–32. and Rheumatism, 40, S1339.
Sturgill, M. and Seibold, J. (1998). Rational use of calcium-channel antago- Zamora, M., O’Brien, R., Rutherford, R., et al. (1990). Serum endothelin-1
nists in Raynaud’s phenomenon. Current Opinion in Rheumatology, concentrations and cold provocation in primary Raynaud’s phenom-
10, 584–8. enon. Lancet, 336, 1144–7.
SECTION 4

Imaging and
percutaneous
interventions
Angiography has traditionally been the imaging modality of choice in childhood, and the desirability of follow up imaging to deter-
for diagnosis of vasculitis disorders; however, it is increasingly mine the course of disease and its treatment. In the last few years,
replaced by non-invasive cross-sectional imaging studies in the cross-sectional imaging procedures have evolved more than angi-
evaluation of vasculitis, particularly large- and medium-vessel vas- ography, which nevertheless still provides superior resolution
culitis. These newer techniques are less invasive than conventional of small and peripheral vessels. The more static nature of angio-
angiography, and can portray the lumen and changes occurring not graphic techniques compared to cross-sectional imaging is evident
only in the lumen but within the vessel wall and surrounding tissue, in a comparison between the relevant chapters in the 1st edition of
often at an early stage of disease when medical intervention is most this text, and their present form, which shows only slight changes
effective. Unlike conventional angiography, cross-sectional imag- in the chapter on conventional angiography (Chapter  17), and
ing can also better disclose changes in affected organs. impressive advances in the chapter on cross-sectional imaging
The choice between imaging studies may be also influenced (Chapter 18) as well as a chapter on positron emission tomogra-
by availability of equipment and physicians skilled in the perfor- phy (Chapter 19).
mance and interpretation of various procedures. Other factors Angiography is necessary when endovascular therapy is being
include concern for exposure to ionizing radiation, particularly considered.
CHAPTER 17

Angiography and
percutaneous interventions
Souheil Saddekni and Rachel F. Oser

Introduction characterize syphilitic aneurysms, and may be found in patients


with negative syphilis serology.
The end result of the inflammatory process in vasculitis is a
broad range of vascular changes including stenoses, occlusions,
aneurysms, and pseudoaneurysms. The arterial vasculitides are Interventional radiology techniques
classified based on their aetiology or their anatomical vessel Percutaneous endovascular treatment has evolved since its incep-
involvement (see Chapter 1). The anatomic classification adopted tion in the late seventies and over the past three decades has
for angiographic purposes divides vasculitis into three catego- shown remarkable technical progress, starting from large-profile
ries: (1) large vessels, (2) medium and small vessels, and (3) ves- and compliant balloons to low-profile, high-pressure balloons,
sels of all sizes. Most vasculitis involving predominantly large balloon-mounted stents, self-expanding stents, laser-assisted
vessels are one of two types:  Takayasu’s arteritis and giant cell techniques, cutting balloons, atherectomy devices, and the clos-
arteritis. Vasculitis of medium and small vessels include polyarte- est technique to surgical bypass, the covered stent (or stent-graft).
ritis nodosa, microscopic angiitis and Buerger’s disease (throm- Percutaneous revascularization techniques took their position as
boangiitis obliterans). Many vasculitides involve vessels of all a more attractive, less invasive treatment for occlusive vascular
sizes, including vasculitis associated with connective tissue dis- disease and proved extremely useful, particularly in the treatment
eases, substance abuse, Behçet’s syndrome, and infection-related of atherosclerotic disease and fibromuscular dysplasias. However,
arteritis. many stenoses resulting from the fibrotic changes associated with
Diagnosis of vascular inflammatory disorders is sometimes dif- the inflammation associated with the vasculitides exhibited a less
ficult. The clinical features are variable and are often the result predictable response to balloon angioplasty, especially in the acute
of an occlusive process and abnormal blood flow through the phase. Once settled and the inflammation subsided, stenosis from
affected vessels, which may cause organ ischaemia, organ damage, vasculitis or the superimposed atherosclerotic disease are treated
and less frequently, vessel rupture. The occlusive process is not successfully with angioplasty and stenting. Here, techniques offer
unique to these disorders; in fact, it is more commonly encoun- an attractive non-surgical alternative for treating some lesions in
tered in atherosclerosis in adults or trauma, congenital, or clotting TA, providing relief of cerebral, upper extremity, or renal ischae-
disorders in younger patients. Therefore, the clinical presentation, mia. Newer stent grafts are being placed with increasing safety.
laboratory results, type and pattern of vascular involvement and These will undoubtedly have a great impact on large-vessel (aortic,
angiographic features are all important for definitive diagnosis subclavian, iliac, and femoral artery) reconstruction and revas-
and treatment. cularization, whereas finer instruments and techniques, such as
Certain characteristic patterns are highly specific and nearly diag- those used in the coronary circulation, will allow safer and more
nostic. For example, when stenoses or occlusions of the aortic arch widespread use of these modalities in smaller and more peripheral
or arch vessels are encountered, the only major diagnostic consid- vessels.
erations in adults are atherosclerotic disease (see Chapter 44), and When acute arterial occlusions are encountered, thrombolytic
giant cell arteritis (GCA) (see Chapter 23), and Takayasu’s arteritis therapy may reveal a focal underlying stenosis, which could then
(TA) in younger patients (see Chapter 24). Other characteristic find- be dilated. Thrombolysis may offer the only treatment available for
ings are multiple pulmonary aneurysms with large-vein thrombosis small-vessel thrombotic occlusions. On the other hand, emboliza-
in Behçet’s syndrome (BS) (see Figure 17.11). A relatively young tion techniques have been used to occlude Behçet’s disease pulmo-
male smoker with lower extremity ischaemia, and certain occlu- nary aneurysms not responsive to immunosuppression. Traditional
sive arteriographic changes, is most likely to have thromboangiitis surgical intervention for thoracic and abdominal aneurysms in
obliterans (TAO). Multiple small aneurysms in the renal or mes- Behçet’s disease is associated with high morbidity because many
enteric circulation are most commonly associated with polyarte- patients are on active drug therapy. Suture line pseudoaneurysm
ritis nodosa (PAN). Large-vessel saccular aneurysms, especially formation and thrombosis are frequent. These complications are
with surrounding fluid, are characteristic of mycotic aneurysms. avoided with endovascular therapy. Similarly, in treating PAN
Linear dystrophic (tree-bark) calcifications in the ascending aorta aneurysms, microcatheters and microcoils have been used for
228 SECTION 4 imaging and percutaneous interventions

selective embolization in what might have been catastrophic renal Raynaud’s phenomenon and no evidence of systemic disease can
or gastrointestinal microaneurysmal rupture. have peripheral small-vessel occlusions angiographically indistin-
As the role of interventional radiology in the management of guishable from those of thromboangiitis obliterans (TAO; formerly
local complications of systemic vasculitides has become more Buerger’s disease or connective tissue disease) (see Figure 17.21).
established, future interventions will hopefully complement and Treatment in these patients is directed towards relief of acute
benefit from immunological and cellular advances in treating this ischaemia (anticoagulation, thrombolysis, percutaneous translumi-
set of disorders. Immunosuppressive drugs or monoclonal anti- nal angioplasty (PTA), or surgical intervention) (see Figure 17.21).
bodies can perhaps be delivered locally (for example, loaded on At times, it is quite difficult to establish a final diagnosis.
a balloon catheter) to certain lesions to suppress hyperplasia and
reduce the likelihood of recurrent stenosis. Alternatively, these Takayasu’s arteritis
agents may be injected in a concentrated form through microcath-
eters into a limited arterial bed for regional treatment. Drug eluted Takayasu’s arteritis (TA) is a non-specific inflammation of unknown
stents have remarkably reduced poststenting re-stenosis rate in the aetiology affecting the aorta, its branches, and occasionally the pul-
coronary arteries. These other technical feats, however, have yet to monary arteries (see Chapter 24). The end result is marked fibro-
prove their clinical usefulness. sis and thickening of the arterial wall, which most often results
For the purpose of this chapter, we focus on current angiogra- in stenosis or occlusion and occasionally aneurysm formation
phy and intervention in vasculitis syndromes as encountered in a (Lupi-Herrera et al. 1977; Strachnan 1964).
university practice. Patients undergo angiography either to estab- Patients with TA have a diverse course. Some patients already
lish a diagnosis, to document the extent of involvement, or to treat have advanced arterial obstruction and collateral circulation when
the disease. Endovascular therapies are more applicable to large they first develop constitutional symptoms. At times, the first pres-
and medium-vessel diseases than to the small-vessel varieties such entation is during a ‘burned-out’ phase with stenoses and occlusions
as granulomatosis with polyangiitis (GPA; previously Wegener’s but without evidence of active inflammation (Hall et al. 1985). In
granulomatosis), eosinophilic granulomatosis with polyangiitis late stages, inflammatory aortic lesions resemble the more common
(EGPA; formerly Churg–Strauss syndrome), microscopic polyangii- atherosclerotic disease, which often gets superimposed and obscures
tis (MPA), IgA vasculitis (IgAV; formerly Henoch–Schönlein pur- the original inflammatory lesions. The diagnosis can be confirmed
pura), cryoglobulinaemic vasculitis, and leukocytoclastic vasculitis. with arterial biopsy, but biopsy is positive in only 35% of cases.
The diagnosis and treatment in these small-vessel diseases is usually
established on clinical, laboratory, and pathological grounds rather Angiographic evaluation of takayasu’s arteritis
than by angiography, and they are not discussed in this chapter. Angiographic abnormalities appear to follow certain patterns of
Some non-inflammatory syndromes present with a clinical pic- involvement. TA has been classified by Lupi-Herrera et al. (1975)
ture similar to vasculitis, such as acute ischaemic symptoms in the into four categories, based on anatomic distribution of the disease.
upper extremities. For example, patients with a clinical history of Type I involves the aortic arch and its branches; Type II affects the

(a) (b) (c)

Fig. 17.1 Takayasu’s arteritis—aneurysmal form. A 12-year-old child presented with abdominal pain and was found to have an elevated ESR. Anteroposterior projections
of the upper (a) and lower (b) abdominal aorta show a fusiform aneurysm of the upper abdominal aorta involving the origin of the celiac, superior mesenteric, and renal
arteries. (c) Lateral view.
CHAPTER 17 angiography and percutaneous interventions 229

descending thoracic and abdominal aorta without the arch. Types and contour irregularities occur because the degree of destruc-
III and IV are combinations of Types I and II either without or with tion of the arterial wall is not uniform (Lande and Berkman 1986).
pulmonary arterial involvement, respectively. Long-segment stenoses (more than 5 cm) are more common than
Inflammation is most prominent in the outer media and adventi- short ones (Yamato et  al. 1986). Within the thorax, the left sub-
tia (Nasu 1976), with progressive fibrosis and thickening occurring clavian artery is most often involved. The carotid arteries are the
in these layers. Aneurysms form when disruption of the vessel wall next most commonly involved (Figure  17.3). Coronary artery
is extensive and the adventitia is weakened (Figure 17.1). involvement is seen in approximately 10% of cases. Dilation of the
The most common angiographic abnormalities of the aorta and ascending aorta can lead to aortic insufficiency. Serpiginous, fine
its branches are stenoses (Nasu 1976) (Figure 17.2). Skip lesions vessels are seen in the wall of the aortic arch and the descending
occur often and the transition from normal vessel to diseased is aorta, which represent enlarged vaso vasorum providing collateral
often abrupt. The contour of the stenoses is smooth in two-thirds of blood flow. Within the abdomen, stenoses occur most often in the
the cases. In the active phase, however, inhomogeneous thickening abdominal aorta and next most often in the renal arteries which can

(a) (b)

(c)

Fig. 17.2 Takayasu’s arteritis—Type I (aortic arch and its branches). A 35-year-old female presented with claudication in both upper extremities. Physical examination
revealed weak pulses in both upper extremities and carotid bruits. (a) Arch aortogram shows almost complete occlusion of the proximal left subclavian artery (horizontal
arrow) and complete occlusion of the right subclavian artery (vertical arrow). (b) Antegrade filling of the right vertebral and the posterior circulation. There is reversed
flow in the left vertebral artery, which fills the left subclavian artery (subclavian steal syndrome). White arrows denote the direction of blood flow. (c) Diffuse narrowing of
right subclavian artery and complete occlusion of the right axillary artery (vertical arrow). The right brachial artery is reconstituted (horizontal arrow).
230 SECTION 4 imaging and percutaneous interventions

(a) (b)

(c) (d)

Fig. 17.3 Takayasu’s arteritis—Type III. A 31-year-old female with pulse discrepancy, bilateral carotid bruits, and elevated ESR. (a) Aortic arch study demonstrates bilateral
diffuse disease in the great vessels. There is complete occlusion of the right subclavian artery (white arrow). Selective left carotid (b) and innominate artery (c) injections
illustrate diffuse, severe, long segment stenosis (black arrowheads) of the common carotid arteries. The black arrow in (c) points to occluded right subclavian artery distal
to the origin of the vertebral artery. (d) Manual subtraction abdominal aortogram shows stenosis of the proximal right renal artery (vertical white arrow) and narrowing
of the infra-renal aorta.

cause renovascular hypertension. The superior mesenteric artery 1977). There is no correlation between the extent of the pulmonary
is the most frequently involved visceral artery (Park et al. 1989). arterial lesions and the extent of systemic arteritis.
Stenoses in mesenteric vessels can lead to abdominal pain, or vis-
ceral ischaemia, which may require intervention. In the pulmonary Interventional radiographic treatment
arteries, stenoses and occlusions are seen in any of the branch ves- in takayasu’s arteritis
sels, but most often in the upper lobes. Systemic collaterals pro- Medical treatment consists mainly of corticosteroids in the acute
vide flow to the lungs at times. Dilation of the pulmonary arteries phase with possible cytotoxic or immunosuppressive drugs later
is rare. Pulmonary involvement is asymptomatic in many patients, (see Chapter 24). Operative intervention in TA has been employed
although some demonstrate right heart strain (Lupi-Herrera et al. mainly to relieve cerebral ischaemia due to aortic arch syndrome
CHAPTER 17 angiography and percutaneous interventions 231

and to relieve renovascular hypertension caused by stenosis of hypertension (Saddekni et al. 1980). Despite resistance to balloon
the abdominal aorta or renal arteries (Inada et al. 1970; Kimoto inflation of the affected renal artery, the final result of this PTA
1979; Kusaba et al. 1973). Arterial aneurysms or aortic regurgi- was successful. This initial experience has been expanded in India,
tation may also require surgical intervention. Bypass grafts usu- where TA is more prevalent. Sharma and colleagues have explored
ally yield better outcome than endarterectomy procedures (Pajari some of the technical features involved in PTA of TA (Sharma et al.
et  al. 1986). The most important determinant of 5-year patency 1990). They found that the stenoses in TA were non-compliant and
of a graft is whether it was placed during active inflammation. resistant. The patients frequently complained of extreme pain with
Treatment with warfarin or platelet inhibitors plays a minor role dilation. The minimal involvement of the intima makes the pas-
in the outcome (Pajari et al. 1986). The 5-year patency rate was sage of a guide wire feasible, but the fibrotic element of the lesions
50% when grafts were placed during active inflammation and 88% creates significant resistance to balloon dilation (Castaneda-Zuniga
when placed during quiescent intervals. Angioplasty and/or stent- et  al. 1981; Rao et  al. 1993; Srur et  al. 1985). Prolonged dilation
ing is supplanting surgery as the primary treatment of stenotic is often required for TA. Immediate results of PTA of fibroplastic
occlusive disease. Serial imaging is often used to evaluate disease lesions may not appear satisfactory; however, delayed response
progression; once stenoses develop they do not resolve and they and improvement due to remodelling has been noted (Park et al.
most often worsen. 1989; Srur et al. 1985) (Figure 17.4). Because the lesions in TA are
The first PTA in TA was reported among other non-atherosclerotic often progressive, repeated percutaneous treatment for stenotic
lesions in 1980 by Saddekni et al. in a patient with renovascular disease is preferred over surgery. Neither angioplasty nor operative

(a) (b) (c)

(d) (e)

Fig. 17.4 Percutaneous interventions in Takayasu’s arteritis. A 38-year-old female who has severe hypertension resistant to medical therapy. (a) Abdominal angiogram
reveals a long tubular stenosis in the middle and distal main right renal artery (black arrowheads). A lower pole branch arises from the stenotic segment. Angiographically,
this lesion appears similar to fibromuscular dysplasia (see Figure 17.18). (b) Arch study confirms the diagnosis of Takayasu’s arteritis with complete occlusion (black
arrow) of the left subclavian artery distal to the origin of the left vertebral artery. (c) Percutaneous transluminal renal angioplasty was performed. Postangioplasty
arteriogram shows poor results with persistent, significant stenosis of the main renal artery and severe spasm involving several segmental branches of the renal artery.
Clinical observation and follow-up of this patient revealed significant improvement in her hypertension. One year later the patient presented with worsening of blood
pressure control. Repeat angiography was performed (d). Selective right renal arteriogram (d) shows improvement over the initial preangioplasty and the immediate
postangioplasty arteriogram (a and c). Repeat angioplasty was performed in view of haemodynamically significant residual stenosis (e). Repeat angioplasty of the
right renal artery (e) shows a limited immediate response but with improvement of the stenosis. Blood pressure control was good over the ensuing 2 years. This case
demonstrates limited immediate response to angioplasty but progressive remodelling and improvement.
232 SECTION 4 imaging and percutaneous interventions

intervention is recommended when there is evidence of active feasible, focal lesions involving the renal, or visceral arteries can
inflammation.1 Some studies show that PTA is successful in 90% be treated percutaneously (Rozenblit and Saddekni 1999). This can
of TA cases (Tyagi et  al. 1993), with long-term primary patency be done prior to surgery in an attempt to decrease the morbidity
rates in excess of 60% (Rao et al. 1993). Recurrent lesions can often of surgery or after bypass to treat newly developed focal lesions or
be treated easily with repeat angioplasty, with markedly prolonged recurrences.
secondary patency rates. Occluded segments often contain throm-
bus within the arterial lumen, and thrombolysis may reveal under-
lying stenosis amenable to angioplasty (Rao et al. 1993). Giant cell arteritis
Angioplasty/ stenting can play a complementary role to sur- Giant cell arteritis (GCA) and TA both affect large vessels (see
gery in some cases (Figure  17.5). While aortobifemoral bypass Chapter 23). GCA frequently, but not universally, affects the tem-
can be performed in cases where percutaneous treatment is not poral arteries (Jennette et al. 1994). Temporal artery involvement

(a) (b) (c)

(d) (e) (f)

Fig. 17.5 Surgical and percutaneous interventions in Takayasu’s arteritis—Type II. A 40-year-old male presented with renal failure and bilateral lower extremity
claudication. (a) IVDSA (intravenous digital subtraction angiography) shows that the abdominal aorta is patent to the level of the origin of the superior mesenteric artery
(SMA), but is totally occluded distally. There is probably proximal stenosis in the SMA (white arrow). Note the prominent retroperitoneal collaterals (black arrowheads)
and occluded renal arteries. (b) Reconstitution of normal appearing common femoral arteries (c fem). A supraceliac aorto-bifemoral graft was performed with satisfactory
flow in the celiac and SMA. Intraoperative examination of the abdominal aorta, adjacent lymph nodes, and soft tissues was consistent with aortitis. (c) Abdominal
aortogram showing proximal anastomosis of the aorto-bifemoral graft to the supraceliac aorta. The proximal splenic artery overlaps to obscure the stenosis at the origin
of the SMA. A replaced right hepatic artery arising from the SMA is a normal anatomical variant. (d) Oblique projection of the abdominal aorta shows severe stenosis
at the origin of the SMA (arrow head). (e) Angioplasty followed by stenting of the SMA ostial stenosis was performed. Arrows indicate stent. (f) After stenting, there is
excellent flow through the SMA and replaced right hepatic artery.

1 Editors note: PET scans can be valuable in disclosing persistence of otherwise ‘occult’ inflammation in Takayasu’s arteritis and other large-vessel disease
(see Chapter 19).
CHAPTER 17 angiography and percutaneous interventions 233

can also be seen in PAN and GPA (Jennette et  al. 1994). The the culprit vessel resulting in haemorrhage is not found, and more
defining feature of GCA is the initial active inflammatory pro- non-selective embolization based on CT scan findings is indicated
cess, which involves the inner media and inner elastic lamina (Chandrakantan and Kaufman 1999; Smith and Wernick 1989).
(Jennette et  al. 1994). Age at onset is useful in differentiating This should be even more practical with the advent of multide-
between TA and GCA; GCA is a much more common disease tector CT angiography, which is gaining favour in the workup in
than TA in persons beyond 50 years of age. Angiography in GCA vascular, arterial disorders and in the detection of the site of acute
is performed with special attention to the large arteries known to bleeding (extravasation). Coronary arterial involvement includes
be favoured sites of involvement. This disease has a predilection thrombosis, aneurysms, and irregularity. Approximately 60% of
for the aorta and its major branches, along with the extracranial patients with clinical cardiac symptoms had large-vessel coronary
branches of the carotid artery (Klein et al. 1975) (Figure 17.6). involvement and 40% had small-vessel involvement at autopsy
Approximately 14% of patients with GCA demonstrate large (Przybojewski 1981). Patients with PAN have a short duration of
artery involvement (Hamrin 1972; Klein et al. 1975). When there preinfarct angina, suggesting rapid progression of the lesion. Many
is involvement of only large vessels, it is difficult to differenti- of the myocardial infarctions in PAN patients are large but silent. In
ate between GCA and TA based on angiographic features; and several cases, the infarction occurred shortly after glucocorticoids
it is practically impossible if the aorta and large vessels are both were started. It has been suggested that the reparative process led to
involved. fibrosis occluding the culprit vessels.

Polyarteritis nodosa
Polyarteritis nodosa (PAN) is a systemic, necrotizing, predomi-
Kawasaki’s disease
nantly medium-sized-vessel vasculitis that classically involves Kawasaki’s disease (KD) is an acute inflammatory disease caus-
the renal and visceral arteries (Bron et al. 1995) (see Chapter 25). ing systemic vasculitis, which primarily affects infants and young
Coronary arteries are the next most commonly involved vessels; children. The diagnosis is established on clinical grounds and
microscopic arteries are not involved. Since no diagnostic sero- by angiography and echocardiography. Cardiovascular involve-
logical test exists, angiography can have an important role in the ment is frequent, with coronary artery aneurysms the most pre-
diagnosis of PAN. Organs involved with PAN may be inaccessi- dominant vascular abnormality, occurring in 15–20% of cases.
ble for biopsy and the biopsy of uninvolved sites has a poor yield Aneurysms may also involve other medium-sized vessels such as
(Provenzale and Allen 1996). The angiographic diagnosis of PAN the axillary, iliac, or renal arteries. For more details, see Chapter 28.
has a sensitivity and specificity of approximately 90%, a positive Cross-sectional imaging studies can be used advantageously for
predictive value (PPV) of 55%, and a negative predictive value diagnosis and follow-up of these patients, obviating the need for
(NPV) of 98% (Hekali et  al. 1991). The ‘gold standard’ for the invasive angiography.
diagnosis of PAN is demonstration of necrotizing inflammation of
medium-sized arteries (Easterbrook 1980).
Renal artery involvement (Figure 17.7), seen in 70% of patients,
Thromboangiitis obliterans
often causes hypertension and haematuria. Gastrointestinal Thromboangiitis obliterans, also referred to as Buerger’s disease, is
manifestations include abdominal pain, GI bleeding, and bowel a distinct vasculitis of medium and small-sized arteries and veins,
infarction. In PAN, the elastic lamina is destroyed and the media which initially involves the distal vessels of the legs and arms and
undergoes fibrinoid necrosis. Microaneuryms (1–5  mm) arise progresses proximally, either in a contiguous or skip-zone fashion.
from weakened, necrotic portions of the arterial wall. Aneurysms The first case was described in 1879 by Feliz von Winiwarter, an
are not pathognomonic of PAN and not all PAN patients have Austrian surgeon (von Winiwarter 1879). Leo Buerger published
microaneurysms. Small vessels tend to thrombose and can be seen his first group of patients, which served as the cornerstone for sub-
as occlusions in arteriography. Focal lesions tend to occur at vessel sequent study, in 1908 (Buerger 1908).
bifurcations. PAN is the most common cause of microaneurysms. The prevalence of TAO has decreased since its first description,
Aneurysms of visceral arteries have been demonstrated in 60–90% but has remained fairly constant over the past few decades. This
of PAN patients (Figure 17.8), but some patients with visceral aneu- may represent a refinement in diagnosis rather than a true decrease
rysms have diagnoses other than PAN (Bron et al. 1995; Fleming in prevalence. There is greater incidence of the disease in eastern
and Stern 1965). The mean number of visceral aneurysms in PAN Europe, Asia, and the Middle East. Some series have estimated
is higher than in other disorders. Glucocorticoid treatment can its prevalence among women as high as 20% (Olin et  al. 1990;
result in resolution of aneurysms. In PAN, healing lesions, chronic Yorukoglu et al. 1993), most probably reflective of the increased
stenoses, and acute aneurysm formation can all coexist (Hekali prevalence of smoking among women. When present, the severity
et al. 1991). of the disease is equal in both sexes (Lie 1987). For more details,
Spontaneous haemorrhage from a ruptured aneurysm in PAN see Chapter 37.
is seen most often in young men with a history of hypertension Histologically, vessels affected by TAO vary according to the age
who develop sudden, severe flank or abdominal pain (Capps and of the lesion. Acutely, vessels show severe inflammation involving
Klein 1970; Chandrakantan and Kaufman 1999). Transcatheter all three layers of the vessel wall. In the subacute phase, there is
selective embolization can be life saving and preserve renal func- progressive organization of an occlusive thrombus; in the chronic
tion at the same time. Angiographic embolization of haemor- phase, there is complete organization of an occlusive thrombus
rhage in the gastrointestinal tract has also been performed and with extensive recanalization, and adventitial and perivascular
can avert catastrophic outcomes (Herskowitz et al. 1993). At times, fibrosis. These chronic occlusions are almost always accompanied
234 SECTION 4 imaging and percutaneous interventions

(a) (b) (c)

(d) (e) (f)

(g)

Fig. 17.6 Giant cell arteritis (GCA). (a) Arch aortogram illustrates irregularities involving both subclavian arteries (black arrowheads). (b) Abdominal aortogram shows
proximal left renal artery stenosis (black arrowhead). (c) Diffuse disease in branches of the external carotid artery. (d) Diffuse irregular narrowing of the vertebral artery.
(e) Diffuse, severe stenosis of the intracranial segment of the right vertebral artery (arrowheads). (f) Diffuse, severe, tubular stenosis of the proximal half of the SMA
(white arrows). (g) The replaced right hepatic artery (black arrowheads) is a normal variation arising from the proximal SMA and is free of disease.
CHAPTER 17 angiography and percutaneous interventions 235

(a) (b)

Fig. 17.7 Polyarteritis nodosa (PAN) in the kidney. An 18-year-old female presents with cutaneous and nasal mucosal lesions suspicious for systemic vasculitis. Early
(a) and late (b) arterial phase of selective left renal arteriogram. Note the multiple, small, round aneurysms (arrows) characteristic of PAN.

(a) (b)

Fig. 17.8 Visceral PAN. A 50-year-old male with abdominal pain suspected to have vasculitis. (a) Multiple small aneurysms are seen in the cystic artery branch arising
from the right hepatic artery, pancreaticoduodenal branches of the gastroduodenal artery, and the left hepatic artery (black arrowheads). (b) Magnified view of the cystic
artery aneurysms.
236 SECTION 4 imaging and percutaneous interventions

by extensive collateral circulation, which accounts for some of the the proximal segment of an occluded vessel, are thought to be
characteristic angiographic findings of the disease (Tanaka 1998). fairly characteristic of TAO. These collaterals can be seen to run
Although there is no single angiographic feature specific for directly along a thrombosed segment of artery in a direct network
TAO, a typical constellation of radiological findings, in the proper of collaterals. This unusual pattern of collateral vessels has been
clinical setting, is diagnostic (Hagen and Lohse 1984; Rivera 1973; hypothesized to represent dilated vasa vasorum (Figures  17.9
Suzuki et al. 1996). The main angiographic findings are multiple, and 17.10). A second pattern of collateral vessels was described
bilateral focal areas of stenosis or occlusion, with normal proxi- to resemble a ‘tree root’ or ‘spider’s leg’. These collateral vessels
mal and intervening vessels. The patient is normally free of ath- originate from distally occluded vessels and can either re-open
erosclerotic changes and uninvolved vessels demonstrate smooth as direct collaterals distally, or terminate within the soft tissues.
lumen. The occlusions are usually of the cut-off variety. Corkscrew In the early stages of TAO, a ‘corrugated’ pattern is noted, which
collaterals often develop in TAO, but are not specific, as they are involves a short segment of a peripheral vessel. This is felt to either
also often seen in atherosclerotic disease. However, direct collat- represent alterations in a vessel prior to thrombotic occlusion, or
erals, which arise either from adjacent uninvolved vessels, or from a recanalized vessel.

(a) (c)

(b) (d)

Fig. 17.9 Thromboangiitis obliterans (TAO). A 42-year-old male smoker with a long history of bilateral calf claudication, presents with new onset, right foot rest pain.
Pathological examination of an amputated toe was consistent with TAO. (a) Normal-appearing vessels are seen above the knee. (b) Below the knee, there is severe
occlusive disease of all tibio-peroneal vessels. Only the left peroneal artery is patent. (c) Classic corkscrew collaterals. (d) Magnified view of the corkscrew collaterals.
CHAPTER 17 angiography and percutaneous interventions 237

(a) (b)

Fig. 17.10 Raynaud’s phenomenon in thromboangiitis obliterans. A 29-year-old female, heavy smoker with history of recurrent Raynaud’s phenomenon presents with
coldness and numbness in the extremities. Clinical evaluation revealed no evidence of systemic vasculitis. (a) Right hand arteriogram demonstrates occlusion of the distal
radial artery and all distal proper digital arteries. Similar findings were seen in the left hand. (b) Left foot arteriogram (anteroposterior projection) demonstrates occlusion
of the anterior tibial, dorsalis pedis, and multiple digital arteries.

The standing-wave pattern has a reported prevalence up to


75% in TAO (Hagen and Lohse 1984). This is characterized by
a ‘goose-trachea’ change in the outline of a larger arterial trunk,
most notably in the femoral–popliteal segment, and was originally
postulated to represent a preobstruction lesion with intimal hyper-
plasia that was specific for TAO (Shionoya et al. 1978). However,
it has also been shown that standing waves can be seen with any
distal obstruction, and represent flow phenomena relating to inflow
obstruction and the creation of backflow waves (Lehrer 1967). It
also is frequently noted angiographically in young arteries with no
associated pathology whatsoever. Thus while this may be a helpful
sign in diagnosing TAO, it is not specific.

Behçet’s syndrome
Behçet’s syndrome (BS) is a multisystem disorder affecting young
adults mostly from the Middle East and Far East (see Chapter 34).
Any large or small artery, vein, or organ may be involved in an unpre-
dictable combination (Chajek and Fainaur 1975). Venous lesions are
more common than large artery involvement, causing thrombosis of
veins such as the superior vena cava or inferior vena cava. Arterial
manifestations are rare, occurring in approximately 2.5% of patients
with longstanding disease. Arterial involvement can be divided into
three groups: the first group involves arterial occlusions, the second
group arterial aneurysms, and the third group pulmonary artery
aneurysms (Park et al. 1984). BS is virtually the only vasculitis lead-
ing to pulmonary arterial aneurysms (Lacombe et al. 1985), which
are usually multiple (Figure 17.11). In general, invasive angiography Fig. 17.11 Behçet’s syndrome. A large aneurysm (black arrowheads) is seen in the
should be avoided in BS, since vessel (artery and vein) punctures can right lower pulmonary artery, with occlusion of its segmental branches consistent
lead to thrombosis (Tunaci et al. 1995) or aneurysms. with thrombosis.
238 SECTION 4 imaging and percutaneous interventions

Pulmonary artery aneurysms in BS may rupture, with signifi-


cant morbidity; they may be treated successfully with medications.
Transcatheter embolization has been performed for patients with
symptomatic aneurysms that do not regress with glucocorticoid
treatment. Surgical bypass is complicated at times by aneurysm
formation at the anastomosis (Hamza 1987). Thus, as noted above,
angiography should generally be avoided in BS. Caval thrombosis
is also frequent and may preclude transcatheter pulmonary arterial
intervention (Tunaci et al. 1995).

Infection-related vasculitis
Inflammation of the arterial wall due to infection leads to various
manifestations, which depend largely on the size of the affected ves-
sel. Virtually all infectious organisms have been implicated in this
form of vasculitis (see Chapters 41 and 44). Aneurysms or pseu-
doaneurysms, typically called mycotic aneurysms, can be seen in
infection of vessels of all sizes.
There are several routes of infection of the vessel wall: (1) direct
extension from an intravascular source (endocarditis); (2) haema-
togenous seeding to diseased intima or vaso vasorum (bacteraemia
from any source); (3) direct extension from an extravascular source
(abscess, osteomyelitis); (4)  lymphangitic spread; and (5)  direct
Fig. 17.12 Salmonella aortitis. A 62-year-old male with fever and Salmonella
contamination from trauma, intravenous drug abuse, and osteomyelitis of the right ilium, presents with lower abdominal pain. Findings
instrumentation. on abdominal CT scan prompted aortogram. Abdominal aortogram (lateral
projection) shows a saccular aneurysm from the posterior wall of the infrarenal
Infectious aortitis aorta (black arrowheads). The patient was treated with resection of the aneurysm
Some infectious agents preferentially involve large vessels such as and left axillary artery to bifemoral bypass graft.
the aorta and pulmonary arteries. Salmonella can cause infectious
aortitis, but staphylococcal and streptococcal aortitis (Bronze et al.
1999) are more common. Treponemes and mycobacteria also have
a predilection for the aorta (see Chapter 41).
Salmonella aortitis
Salmonella aortitis is typically seen in elderly patients with
pre-existing atherosclerosis, aortic aneurysm, or a vascular graft.
It typically causes aneurysms of the thoracic (20%) or abdominal
aorta (80%) (Oskoui et al. 1993) (Figure 17.12). The clinical pres-
entation is often misleading and rupture is a frequent complication
(Flamand et al. 1992; Katz et al. 1992). The clinical diagnosis of an
infectious aneurysm is difficult unless it is very large or develops a
complication such as rupture. More than 90% of patients present
with fever and non-specific back and abdominal pain (Flamand
et al. 1992). These patients need to be identified as early as possible
to avoid complications. Treatment includes long-term antibiotics
and surgical resection of the aneurysm and extra-anatomic bypass
(Katz et  al. 1992). Surgical procedures may be difficult and only
50% of patients survive the perioperative period.
Syphilitic aortitis
Syphilitic aortitis is initiated by haematogenous spread of trep-
onemes and is a manifestation of tertiary syphilis. The ascending
aorta and the aortic arch are typically involved. The vaso vasorum
is destroyed by perivascular inflammation, which ultimately leads to
fibrosis of the aortic media. Syphilitic aortitis causes ascending aor-
tic aneurysms with or without valvular incompetence and coronary
Fig. 17.13 Drug abuse vasculitis. An 18-year-old male with history of i.v. drug
ostial stenosis. Linear calcification is present in 70% of patients. The abuse presents with severe pain in the fingertips immediately after attempted
term ‘tree-bark’ appearance has been associated with changes of the intravenous injection of a street drug. An arteriogram of the left hand shows
intima in syphilitic aortitis. Diagnosis can be difficult as there may be focal embolic occlusion in the proper digital (arrowhead) and common digital
no reliable history of syphilis and serological studies may be negative. arteries (arrow).
CHAPTER 17 angiography and percutaneous interventions 239

(a) (b) (c)

Fig. 17.14 Systemic lupus erythematosus (SLE). A 30-year-old white female with SLE presents with numb, cold hands and feet. (a) Luminal irregularity of the entire
brachial artery (white arrows). (b) Pelvic arteriogram shows luminal irregularity involving right external iliac artery (white arrows). (c) Occluded radial artery at the wrist
(large white arrow).

Drug abuse antiphospholipid antibodies and other factors, arterial occlusions


Intravenous drug abuse may lead to vascular complications related may be due to thrombosis or to vasculitis. The two aetiologies
to local injury or infections from unclean needles. Pseudoaneurysms may be indistinguishable in a given patient (Figure 17.15). Renal
or arteriovenous fistulae may form. Accidental injection of hyper- artery thrombosis or stenosis can occur in primary or secondary
tonic solution containing powder, or other foreign substances, into antiphospholipid syndrome.
the arterial system can result in multiple occlusions of digital arter-
ies (Figure 17.13). Raynaud’s phenomenon may follow this form of
injury (Yao et al. 1972). Long-term ingestion of certain compounds,
most notably ergotamine, causes diffuse arterial spasm reminiscent
of vasculitis.

Vasculitis secondary to connective


tissue diseases
Inflammatory vascular disease can occur with systemic lupus
erythematosus, rheumatoid arthritis, and scleroderma (see
Chapter 33). Angiographic appearances of these entities overlap
and definite diagnosis cannot be made on the basis of angiographic
features alone, though it provides corroborative information.

Systemic lupus erythematosus


Raynaud’s phenomenon is present in up to 30% of patients with
systemic lupus erythematosus (SLE). Vasculitis in patients with
SLE typically involves medium and small-sized vessels, although
Fig. 17.15 Systemic lupus erythematosus. A 39-year-old female with a history of
large vessels can also be involved. Angiographically, small and lupus vasculitis presented with left upper extremity ischaemic symptoms. Selective
medium-sized vessels show tapered or abrupt occlusions with left subclavian arteriogram shows abrupt cut-off in upper brachial artery with poor
sparse formation of collateral vessels (Figure  17.14). Because collateral flow consistent with acute thrombosis. She underwent thrombolysis with
hypercoagulability is common in SLE due to the presence of tissue plasminogen activator infusion with complete resolution of symptoms.
240 SECTION 4 imaging and percutaneous interventions

(a) (b) Rheumatoid arthritis


Vasculitis is seen in small number of patients with long-standing,
severe rheumatoid arthritis (RA). Angiographic examinations
show either occlusive or hyperaemic arterial abnormalities (Laws
et al. 1963). Most of the occlusive lesions occur at the level of the
digital arteries, although medium-sized vessels are also involved
(Figure 17.16). Hyperaemic changes tend to occur around regions
of synovial proliferation.

Scleroderma
In scleroderma, small vessels of the digits, kidneys and heart show
intimal thickening with fibrous deposition and occlusion. Over 80%
of patients have a history of Raynaud’s phenomenon. Angiography
demonstrates multiple focal stenoses and occlusion, especially in
small vessels. Arteriography of the hand shows severe involvement
of proper digital arteries, generally in the mid- or distal portion of
the vessel (Dabich et al. 1972).

Mimickers of vasculitis
A multitude of conditions may imitate vasculitis (see Chapter 44).
Fig. 17.16 Rheumatoid arthritis. A 49-year-old female with rheumatoid arthritis Hypothenar hammer syndrome (Figure 17.17) and other occupa-
presents with symptoms of ischaemia in the lower extremities. (a) Complete tional injuries, neurofibromatosis (Figure  17.18), thoracic outlet
occlusion of the popliteal artery at the level of joint line (black arrow) with syndrome, radiation injury, frostbite, and drug abuse can produce
collaterals (black arrowheads). (b) Reconstitution of the posterial tibial artery vascular changes mimicking vasculitis. Three disorders are com-
(black arrowheads), which is the only run-off vessel to the foot.
monly seen and merit brief discussion.

(a) (b)

Fig. 17.17 Trauma: hypothenar hammer syndrome. A construction worker presented with numbness of the right ring and little fingers. He had classic history of
repetitive trauma to the right hypothenar eminence. (a) Right hand arteriogram shows occlusion of the distal ulnar artery (double arrows). The single arrow points to the
hook of the hamate bone where occlusion of the ulnar artery begins. Retrograde thrombosis has occurred to the first significant collateral vessel (black arrowhead). The
radial artery fills the deep and superficial palmar arches and digital arteries. (b) Magnified view of the late arterial phase shows the shadow of the distal ulnar artery filled
with thrombus (arrows).
CHAPTER 17 angiography and percutaneous interventions 241

Fig. 17.18 Neurofibromatosis, type I: tubular stenoses. A 3-year-old male child


presented with hypertension underwent abdominal aortogram. The patient had
café au lait spots consistent with type I neurofibromatosis. Abdominal aortogram
shows severe tubular stenosis of the right main renal artery (black arrowhead).
A similar appearance can be seen in intimal fibroplasia and large-vessel vasculitides.

Fibromuscular dysplasia Fig. 17.19 Fibromuscular dysplasia: medial fibroplasia with mural aneurysm.


Arterial fibrous lesions are often detected during angiography A 55-year-old white female with long history of poorly controlled hypertension
underwent angiography to rule out renal artery stenosis. Selective right lower renal
of practically all branches of the aorta, particularly the renal
artery injection demonstrates the characteristic corrugated or ‘string of beads’
and carotid arteries. The aetiology of fibromuscular dysplasia is appearance (black arrowheads) involving the distal portion of the main renal
unknown. It is usually associated with renovascular hypertension artery prior to branching. Upper renal artery injection revealed similar pathology
and predominantly occurs in young Caucasian women. Harrison (not shown). Angioplasty of both renal arteries was performed and the patient
and McCormack (1971) developed a pathological classification remained normotensive.
based on the arterial layer involved. The most common type is
known as ‘medial fibroplasia with mural aneurysm’ and has the
classic ‘string of beads’ appearance (Figure  17.19). This appear- Neurofibromatosis is another cause of long tubular stenosis in
ance is diagnostic and requires no further investigation. This type the aorta with associated ostial stenosis of aortic branches, which
responds readily to balloon angioplasty and should not be stented. should be considered in the differential diagnosis. Midaortic syn-
The other type involve other layers of the arterial wall and appear as drome is usually discovered in childhood or adolescence (Stanley
focal or long smooth stenosis, which respond to angioplasty but it et al. 1981). Major vascular reconstruction is often required because
may exhibit partial response initially with remodelling later. Some these developmental lesions are difficult to dilate or stent.
lesions recoil immediately and require stenting when they recoil to
their severe stenotic state. They are typically lumped angiographi- Raynaud’s phenomenon
cally under the term ‘intimal’ (Figure  17.4A), stenoses that are Raynaud’s phenomenon (Raynaud 1862)  is episodic spasm of
impossible to differentiate angiographically from vasculitic lesions. small vessels of upper extremity precipitated by cold tempera-
Fibromuscular dysplasia is not a systemic disease. tures. Its classic clinical features are blanching of the hand lead-
ing to bluish discoloration, followed by pink coloration with
Midaortic syndrome symptoms of numbness, tingling, and pain on recovery (Klippel
Developmental narrowing of the abdominal aorta is also known 1991). Angiographically, this is characterized by multiple areas of
as abdominal aortic coarctation, aortic hypoplasia, or aortic dys- segmental but reversible spasm that improves with vasodilators
plasia. It is often associated with ostial stenoses of the renal and or poststress manoeuvre. Patients with Raynaud’s phenomenon
or visceral arteries, and is occasionally seen during evaluation of may have severe ischaemia and arterial thrombosis, but it is debat-
renovascular hypertension (Figure 17.20). The appearance is simi- able whether this represents a separate, but related, problem (see
lar to the abdominal form of Takayasu’s arteritis (Lewis et al. 1988). Figure 17.21 and Chapter 16).
242 SECTION 4 imaging and percutaneous interventions

(a) (b) (c)

(d) (e)

Fig. 17.20 Midaortic syndrome. A 19-year-old male with hypertension. Evaluation for systemic vasculitis was negative. Findings in this syndrome are similar to Takayasu’s
arteritis, but are seen only in the mid-abdominal aorta. (a and b) Anteroposterior and lateral views of the abdominal aortogram show nearly complete occlusion of the celiac
and superior mesenteric arteries with stenotic suprarenal and renal segments of the abdominal aorta (black arrows). Note the huge inferior mesenteric artery (black and
white arrows) providing collateral supply to branches of the superior mesenteric artery and stenosis in the right main renal artery (black arrowhead). (c) Selective right renal
arteriogram shows severe stenosis of the right main renal artery with extension to the ostia of the anterior and posterior divisions. (d) Right renal artery angioplasty. Note the
simultaneous angioplasty, after placing a balloon catheter in each division of the right main renal artery (the so-called ‘kissing balloon technique’). (e) After angioplasty, the
right renal arteriogram shows limited immediate success, as is common in Takayasu’s arteritis.
CHAPTER 17 angiography and percutaneous interventions 243

(a) (b) (c)

(d) (e) (f)

Fig. 17.21 Raynaud’s phenomenon. A 49-year-old female patient with a history of recurrent Raynaud’s phenomenon presented with acute ischaemia of the right
upper extremity. (a) Selective right subclavian artery injection shows complete occlusion of the upper right brachial artery (black arrowheads). (b) Occlusion of the
brachial, radial, and ulnar arteries. Small collaterals (black arrowheads) reconstitute the interosseous artery (black arrows). (c) Arteriogram showing circulation to the
distal right forearm and wrist demonstrates an occluded ulnar artery and partial reconstitution of the radial artery (black arrowhead). Collaterals reconstitute part of the
deep palmar arch (black arrows). The patient underwent thrombolysis with intra-arterial urokinase infusion overnight. The patient improved clinically, with no signs of
residual ischaemia. (d, e, and f) Post-thrombolysis arteriogram demonstrated complete resolution of the thrombosis within the right brachial artery (black arrowheads in
(d)). Other widely patent vessels (e) include the radial (black arrowheads), ulnar (curved arrows), and interosseous arteries (black arrows). The deep palmar arch (black
arrowheads) and its branches are well visualized (f). There is no significant underlying arterial disease. Evaluation of this patient over a period of several years consistently
failed to reveal an aetiology for recurrent Raynaud’s phenomenon and arterial thrombosis.
244 SECTION 4 imaging and percutaneous interventions

References Klein, R.G., Hunder, G.G., Stanson, A.W., and Sheps, S.G. (1975). Large
artery involvement in giant cell (temporal) arteritis. Annals of Internal
Bron, K.M., Strott, C.A., and Shapiro, A.P. (1995). The diagnostic value of Medicine, 83, 806–12.
angiographic observations in polyarteritis nodosa. A case of multiple
Klippel, J.H. (1991). Raymond’s phenomenon: the French tri-color. Archives
aneurysms in the visceral organs. Archives of Internal Medicine, 116, 450.
of Internal Medicine, 151, 2389–93.
Bronze, M.S., Shirwany, A., Corbett, C., and Schaberg, D.R. (1999). Infectious
Kusaba, A., Inokuchi, K., Kiyose, T., and Oka, N. (1973). Carotid reconstruc-
aortitis:  an uncommon manifestation of infection with Streptococcus
tion for Takayasu’s (pulseless) disease using open thromboendarterec-
pneumoniae. American Journal of Medicine, 107, 627–30.
tomy at aortocarotid Junction. Japanese Journal of Surgery, 3, 91–7.
Buerger, L. (1908). Thrombo-Angiitis obliterans:  A  study of the vascular
Lacombe, P., Frija, G., Parlier, H., Lang, F., Hamza, R., and Bismuth, V. (1985).
lesion leading to pre-senile spontaneous gangrene. American Journal of
Transcatheter embolization of multiple pulmonary artery aneurysms in
the Medical Sciences, 136, 567–80.
Behçet’s syndrome. Acta Radiologica Diagnosis, 26, 251–3.
Capps, J.H. and Klein, R.M. (1970). Polyarteritis nodosa as a cause of perire-
Lande, A. and Berkman, Y.M. (1986). Radiologic aspects of aortitis. In
nal and retroperitoneal hemorrhage. Radiology, 94, 143.
Aortitis: Clinical, pathologic, and radiographic aspects (ed. A. Lande, Y.M.
Castaneda-Zuniga, W.R., Formanek, A., Lillehei, R.C., Tadavarthy, M., and Berkman, and H.A. McAllister Jr.), pp. 81–143. Raven Press, New York.
Amplatz, K. (1981). Non-surgical treatment of Takayasu’s disease.
Laws, J.W., Lillie, J.G., and Scott, J.T. (1963). Arteriographic appearances in
Cardiovascular and Interventional Radiology, 4, 245–8.
rheumatoid arthritis and other disorders. British Journal of Radiology,
Chajek, T. and Fainaur, M. (1975). Behçet’s disease. Report of 41 cases and 36, 477.
review of the literature. Medicine, S4, 179–96.
Lehrer, H. (1967). The physiology of angiographic arterial waves. Radiology,
Chandrakantan, A. and Kaufman, J. (1999). Clinical nephrology teaching 89, 11.
case-renal hemorrhage in polyarteritis nodosa: Diagnosis and manage-
Lewis, III, V.D., Meranze, S.G., McLean, G.K., O’Neill, Jr., J.A., Berkowitz,
ment. American Journal of Kidney Diseases, 33, 1–3.
H.D., and Burke, D.R. (1988). The midaortic syndrome: Diagnosis and
Dabich, L., Bookstein, J.J., Zweifler, A., and Zarafonetis, C.J.D. (1972). Digital treatment. Cardiovascular Radiology, 167, 111–13.
arteries in patients with scleroderma. Archives of Internal Medicine,
Lie, J.T. (1987). Thromboangiitis obliterans (Buerger’s disease) in women.
130, 708.
Medicine, 66, 65–72.
Easterbrook, J.S. (1980). Renal and hepatic microaneurysms: Report of a new
Lupi-Herrera E., Sanchez-Torres, G., Horwitz, S., and Gutierrez, F.E. (1975).
entity simulating polyarteritis nodosa. Radiology, 137, 629.
Pulmonary artery involvement in Takayasu’s arteritis. Chest, 67, 69.
Flamand, F., Harris, K.A., DeRose, G., Karam, B., and Jamieson, W.G.
Lupi-Herrera E., Sanchez-Torres, G., Marcushamer, J., Mispireta, J., Horwitz,
(1992). Arteritis due to Salmonella with aneurysm formation: two cases.
S., and Vela, J.E. (1977). Takayasu’s arteritis. Clinical study of 107 cases.
Canadian Journal of Surgery, 35, 248–52.
American Heart Journal, 93, 94–103.
Fleming, R.J. and Stern, L.Z. (1965). Multiple intraparenchymal renal aneu-
Nasu, T. (1976). Aortitis syndrome:  pathologic aspect. Gendai Iryo, 8,
rysms in polyarteritis nodosa. Radiology, 84, 100.
1143–50.
Hagen, B. and Lohse, S. (1984). Clinical and radiologic aspects of Buerger’s
Olin, J., Young, J., Graor, R., Ruschhaupt, W., and Bartholomew, J. (1990).
disease. Cardiovascular and Interventional Radiology, 7, 283–93.
The changing clinical spectrum of thromboangiitis obliterans (Buerger’s
Hall, S., Barr, W., Lie, J.T., Stanson, A.W., Kazmier, F.J., and Hunder, G.G. disease). Circulation, 82 (Suppl. IV), 3–8.
(1985). Takayasu artertis. A  study of 32 North American patients.
Oskoui R., Davis W.A., and Gomes M.N. (1993). Salmonella aortitis: a report
Medicine, 64, 89–99.
of a successfully treated case with a comprehensive review of the litera-
Hamrin, B. (1972). Polymyalgia arteritica. Acta Medica Scandinavica ture. Archives of Internal Medicine, 153, 517–25.
Supplementum, 533, 1–131.
Pajari, R., Hekali, P., and Harjola, P.T. (1986). Treatment of Takayasu’s
Hamza, M. (1987). Large artery involvement in Behçet’s disease. Journal of Arteritis: An analysis of 29 operated patients. Thoracic and Cardiovascular
Rheumatology, 14, 554–9. Surgeon, 34, 176–81.
Harrison, E.G. Jr. and McCormack, L.J. (1971). Pathologic classifications Park, J.H., Han, M.C., and Bettmann, M.A. (1984). Arterial manifestations of
of renal arterial disease in renovascular hypertension. Mayo Clinic Behçet disease. American Journal of Radiology, 143, 821–5.
Proceedings, 46, 161–7.
Park, J.H., Han, M.C., Kim, S.H., Oh, B.H., Park, Y.B., and Seo, J.D. (1989).
Hekali, P., Kajander, H., Pajari, R., Stenman, S., and Somer, T. (1991). Takayasu Arteritis:  Angiographic findings and results of angioplasty.
Diagnostic significance of angiographically observed visceral aneu- American Journal of Roentgenology, 153, 1069–74.
rysms with regard to polyarteritis nodosa. Acta Radiologica, 32, 143–8.
Provenzale, J.M. and Allen, N.B. (1996). Neuroradiologic findings in pol-
Herskowitz, M.M., Flyer, M.A., and Sclafani, S.J.A. (1993). Percutaneous yarteritis nodosa. American Journal of Neuroradiology, 17, 1119–26.
transhepatic coil embolization of a ruptured intrahepatic aneurysm
Przybojewski, J.Z. (1981). Polyarteritis nodosa in the adult- report of a case
in polyarteritis nodosa. Cardiovascular and Interventional Radiology,
with repeated myocardial infarction and a review of cardiac involve-
16, 254–6.
ment. South African Medical Journal, 26, 512–18.
Inada, K., Katsummura, T., Hirai, J., and Sunada, T. (1970). Surgical treat-
Rao, S.A., Mandalam, K.R., Rao, V.R., Gupta, A.K., Joseph, S., Unni,
ment in the aortitis syndrome. Archives of Surgery, 100, 220–4.
M.N., Subramanyan, R., and Neelakandhan, K.S. (1993). Takayasu
Iwia, T. (1998). Buerger’s disease with intestinal involvement. Internal Journal Arteritis: Initial and long-term follow-up in 16 patients after percutane-
of Cardiology, 66 (Suppl. 1), S257–63. ous transluminal angioplasty of the descending thoracic and abdominal
Jennette, J.C., Falk, R.J., Andrassy, K., Bacon, P.A., Churg, J., Gross, W.L., aorta. Radiology, 189, 173–9.
Hagen, E.C., Hoffman, G.S., Hunder, G.G., Kallenberg, C.G.M., Raynaud, A.G.M. (1862). De l’asphyxie locale et de la gangrene symetrique
McCluskey, R.T., Sinico, R.A., Rees, J., van Es, L.A., Waldherr, R., and des extremities. (Translated by T. Barlow, New Sydenham Society,
Wiik, A. (1994). Nomenclature of systemic vasculitides, proposal of London, 1888). In Textbook of angiography of vascular disease, (ed. H.L.
an international consensus conference. Arthritis and Rheumatism, 37, Neiman and J.S.T. Yao), p. 365. Churchill Livingstone, New York.
187–92.
Rivera, R. (1973). Roentgenographic diagnosis of Buerger’s disease. Journal of
Katz, S.G., Andros, G., and Kohl R.D. (1992). Salmonella infection of the Cardiovascular Surgery, 14, 40–46.
abdominal aorta. Surgery Gynaecology and Obstetrics, 175, 102–6.
Rozenblit, G.N. and Saddekni, S. (1999). Percutaneous renal revasculari-
Kimoto, S. (1979). The history and present status of aortic surgery in Japan zation in children and adolescents with reno- vascular hypertension.
particularly for aortitis syndrome. Journal of Cardiovascular Surgery, 20, Techniques in Vascular and Interventional Radiology, 2, 84–90.
107–28.
CHAPTER 17 angiography and percutaneous interventions 245

Saddekni, S., Sniderman, K.W., Hilton, S., and Sos, T.A. (1980). Percutaneous Suzuki, S., Yamada, I., and Himeno, Y. (1996). Angiographic findings
transluminal angioplasty in non-atheroscelerotic lesions. American in Buerger Disease. Internal Journal of Cardiology, 54 (Suppl.),
Journal of Roentgenology, 135, 975–82. S189–95.
Sharma, S., Rajani, M., Kaul, U., Talwar, K.K., Dev, V., and Shrivastava, S. (1990). Tanaka, K. (1998). Pathology and pathogenesis of Buerger’s disease. Internal
Initial experience with percutaneous transluminal angioplasty in the man- Journal of Cardiology, 66 (Suppl. 1), S237–42.
agement of Takayasu’s arteritis. British Journal of Radiology, 63, 517–22. Tunaci, A., Berkmen, Y.M., and Gokmen, E. (1995). Thoracic involvement
Shionoya, S., Ban, J., Nakata, G., Matsubara, J., Hirai, M., and Kawai, S. in Behçet's disease: pathologic, clinical, and imaging features. American
(1978). Involvement of the iliac artery in Buerger’s disease. Journal of Journal of Roentgenology, 164, 51–6.
Cardiovascular Surgery, 19, 69–76. Tyagi, S., Singh, B., Kaul, U.A., Sethi, K.K., Arora, R., and Khalilullah, M.
Smith, D. and Wernick, R. (1989). Spontaneous rupture of a renal artery (1993). Balloon angioplasty for renovascular hypertension in Takayasu’s
aneurysm in polyarteritis nodosa: Critical review of the literature and arteritis. American Heart Journal, 125, 1386–93.
report of a case. American Journal of Medicine, 87, 464–7. von Winiwarter, F. (1879). Ueber eine eigenthumliche Form von Endarteritis
Stanley, J.C., Graham, L.M., Whitehouse, W.M., Jr., Zelenock, G.B., Erlandson, und Endophlebitis mit Gangren des Fusses. Archiv für Klinische
E.E., Cronenwett, J.L., and Lindenauer, S.M. (1981). Developmental Chirurgie, 23, 202–26.
occlusive disease of the abdominal aorta and the splanchnic and renal Yamato, M., Lecky, J.W., Hiramatsu, K., and Kohda, E. (1986). Takayasu arte-
arteries. American Journal of Surgery, 142, 190–6. ritis: Radiographic and angiographic findings in 59 patients. Radiology,
Strachnan, R.W. (1964). The natural history of Takayasu’s arteriopathy. 161, 329–34.
Quarterly Journal of Medicine, 33, 57. Yao, J.S.T., Goodwin, D.P., and Kenyon, J.R. (1970). Case of ergot poisoning.
Srur, M.F., Sos, T.A., Saddekni, S., Cohn, D.J., Rozenblit, G., and Wetter, E.B. British Medical Journal, 3, 86.
(1985). Intimal fibromuscular dysplasia and Takayasu’s arteritis: delayed Yorukoglu, Y., Ilgit, E., Zengin, M., Nazleil, K., Salman, E., and Yucel, E.
response to percutaneous transluminal renal angioplasty. Radiology, (1993). Thromboangiitis obliterans (Buerger’s disease in women—a
157, 657–60. reevaluation). Angiology, 44, 527–32.
CHAPTER 18

Cross-sectional imaging
in vasculitis
Enrique A. Sabater and Anthony W. Stanson

Introduction Ultrasonography
The traditional gold-standard imaging test for the diagnosis of vas- Ultrasonography (US) uses high-frequency sound waves transmitted
culitis is angiography, and angiographic findings are included in through the patient’s tissues and generates an image from the echoes
the diagnostic criteria of multiple vasculitides. While angiography reflected by the tissues and their interfaces. Gray scale or B-mode
demonstrates with high-resolution the changes affecting the lumen images provide anatomical detail and characterization of the tis-
of vessels, one of its main limitations is the inability to visualize the sues. Beyond anatomical imaging, Doppler ultrasonography uses
actual changes occurring in the blood vessel wall. This is where the the Doppler principle to analyse the ultrasound waves reflected by
pathological changes affecting the lumen of the vessel are occurring. moving blood and generate blood velocity waveforms. This can be
More importantly, mural changes may precede luminal changes, used to assess vascular haemodynamics, allowing quantitative assess-
making angiography abnormal at later stages of disease. For these ment of flow direction and velocity and qualitative assessment of flow
reasons, the non-invasive cross-sectional imaging modalities of waveforms for indirect evidence of proximal or distal disease. US is
ultrasonography (US), computed tomography (CT), and magnetic also used invasively in transoesophageal probes to evaluate the heart
resonance imaging (MRI) have assumed a more important role in and thoracic aorta, and in endovascular probes to evaluate large and
the diagnosis and follow-up of vasculitis, sometimes suggesting the medium-size vessels from the inside. Beyond continued improve-
diagnosis at earlier stages of disease when early treatment may have ments in the imaging technology, the most recent development in
greater impact on disease progression. ultrasound imaging is the use of intravascular agents in the form of
All three of these modalities, and in particular CT and MRI, have microbubbles, which has the potential to provide information about
seen continued advances, now allowing faster and higher resolu- vessel wall inflammation and, therefore, be useful for diagnosis and
tion scanning. With these improvements, CT and MRI can be used monitoring of vasculitis (Shalhoub et al. 2012).
to obtain high-quality angiographic images with minimally inva- Advantages of US include its widespread availability, low-cost,
sive techniques, matching the angiographic images of the vascular exam speed, and absence of contraindications. US avoids the need
lumen obtained with conventional angiography for evaluation of for ionizing radiation or i.v. contrast injection. It allows multiplanar
large and medium-size-vessel vasculitis. Conventional angiogra- real-time imaging and, for superficial vessels, excellent detail of the
phy, however, still provides better resolution than CT and MRI for vessel walls with better spatial resolution (approximately 0.1 mm)
imaging of small, less than 1.5 mm, peripheral vessels. Ultrasound than CT or MRI. Disadvantages of ultrasound for vascular imag-
provides excellent vessel detail to assess luminal and mural changes ing are due mostly to the difficulty in visualizing certain vessels
in peripheral and superficial vessels. adequately, either because of body habitus, depth of the vessel, or
The cross-sectional imaging modalities are also useful for dem- intervening anatomical structures blocking the vessel from an appro-
onstrating the end-organ effects and complications of the vascu- priate imaging window. US does not provide information on degree
litic process. Depending on the specific vasculitis, abnormalities of activity of the vasculitis. In addition, the quality of the examina-
can be demonstrated in any organ, most commonly in the lungs, tion and demonstration of findings is heavily dependent on the skill
heart, liver, kidneys, bowel, and brain. In the small-vessel vascu- and diligence of the sonographer, compared to the more reproduc-
litides where vessel changes are beyond the resolution of all imag- ible and relatively operator-independent images obtained by CT or
ing modalities, these cross-sectional modalities are still useful to MRI. For these reasons, US is most useful for evaluation of luminal
evaluate tissue damage. and vessel wall changes in superficial vessels, including the upper and
The non-specific clinical features of many vasculitides often lead lower extremities, the great vessels of the aortic arch, the temporal
to differential diagnoses that include an infectious disease or a artery, and, through transcranial Doppler, the circle of Willis.
malignant process. Cross-sectional imaging is frequently initially
performed with these diagnoses in mind. It is important to be cog- Computed tomography
nizant of the potential findings in vasculitis, to recognize them, and Computed tomography generates a cross-sectional image of the
suggest the diagnosis, which hopefully will redirect the patient’s body from a computer reconstruction of measurements of X-ray
evaluation and lead to early treatment of the disease. transmission through a thin slice of tissue. CT has evolved from
248 SECTION 4 imaging and percutaneous interventions

(a) (b)

(c) (d)

Fig. 18.1 CT Angiography. (a) A 64-slice CT scan through the mid-thigh showing inflammatory wall thickening and luminal narrowing of the superficial femoral arteries
(SFA) bilaterally (arrows) in a patient with giant cell arteritis. (b) Three-dimensional reconstruction of the same patient showing the extent of disease in the diffuse SFA
narrowing (arrows) with return to more normal luminal diameter in the popliteal artery (arrowhead). (c) Same patient after treatment showing decreased wall thickening
and improved luminal diameter of the SFA on axial images (arrows). (d) Three-dimensional reconstructions after treatment showing diffuse bilateral SFA narrowing
(arrows) that has improved in the interval.

single-row detector conventional scanners to the new generation of great advantage over catheter-based conventional angiography by
high-resolution multirow (currently 320 detection rows) detector avoiding the risk of complications at the arterial puncture site and
array scanners, which allow fast imaging with greater scan coverage at the arteries traversed with the catheters and wires. Disadvantages
and resolution. One key advantage of the current technology is the of CT, in comparison with MRI and US, include the use of ion-
ability to reconstruct the images in any imaging plane while pre- izing radiation, which in turn is less than the radiation dose from
serving the same spatial resolution (Figure 18.1). This advancement angiography, and the need to use iodinated contrast for CTA for
has led to the virtual replacement of the majority of diagnostic angi- adequate tissue contrast. Iodinated contrast has potentially det-
ography with either CT angiography (CTA) or MR angiography rimental effects on renal function and the potential for allergic
(MRA). For CTA, scanning is performed in the transverse (axial) reactions. Ideally, CT scanning in vasculitis should be performed
plane after a bolus infusion of contrast, and three-dimensional first without contrast, to improve detection of arterial calcium and
(3D) images are generated from software reconstructions of the detection of increased attenuation, as from haemorrhage/ haema-
axial scans. Advantages of CT include its widespread availabil- toma. Then, contrast-enhanced images should be obtained in the
ity, rapid scan acquisition (5 second whole body scan in the fast- arterial phase, to allow CTA reconstructions during peak vessel
est scanners), good spatial resolution, excellent demonstration of enhancement. A third acquisition should be obtained after a time
calcifications and bone detail, excellent imaging of parenchymal delay from the contrast injection to demonstrate the enhancement
changes and complications of vasculitis, and reproducible results. of organ parenchyma and arterial walls. Despite performing these
The examination is well tolerated by most patients. CTA only needs three acquisitions, exam times are considerably faster than with
a peripheral i.v. for the contrast bolus administration, which is a MRI. The disadvantage of this approach is the amount of radiation
CHAPTER 18 cross-sectional imaging in vasculitis 249

given to the patient, which can have long-term effects especially in exam, and contraindication of the exam in patients with pacemak-
younger patients. ers and other implantable devices susceptible to magnetic forces. (For
gadolinium toxicity see Section Organ Changes and Complications.)
Magnetic resonance imaging
MRI uses the magnetic properties of hydrogen atoms in body tis- Role of cross-sectional imaging in vasculitis
sues to generate an image. In general, MRI has limited spatial reso-
lution when compared to CT but superior contrast resolution. That Vascular changes
is, imaging sequences can be obtained to bring out the difference Cross-sectional imaging can be used to image the arterial and venous
between normal and diseased tissue and between tissues of differ- changes of vasculitis by demonstrating the lumen of the vessel and
ent composition. Advancements in MRI technology and higher mural changes. Vessel luminal changes, which can be demonstrated
magnetic field scanners have increased the resolution and the in large and medium-vessel vasculitis with angiographic techniques,
speed of MRI examinations. MRI also has the advantage of allow- include luminal irregularity, stenosis, occlusion, ectasia, aneurysm,
ing imaging in any plane. and dissection. Mural changes that can be demonstrated include
The imaging sequences used for MRI evaluation in a patient with wall thickening, calcifications, haematomas, and mural thrombus.
suspected vasculitis are constantly evolving as new techniques are Inflammation and oedema surrounding the vessel wall can also be
developed. Standard imaging sequences for MRI include the con- demonstrated with CT and, especially, with MRI. Contrast enhance-
ventional T1-weighted spin echo sequence, which is well suited ment of the vessel wall seen with CT or MRI is evaluated to aid in
for evaluating anatomy because of its better spatial resolution, and the detection of disease. Although this finding has been linked to
T2-weighted spin echo sequence, which is more time consuming disease activity in some studies, the correlation has not been reli-
but provides excellent tissue contrast without having to adminis- able and further, larger studies are needed. The ability to image the
ter intravenous contrast. Fat suppression with the T2-weighted vessel wall and to provide non- or minimally invasive angiographic
sequence can be used to improve vessel wall contrast. T2 fast spin images is the key advantage of cross-sectional imaging. Luminal and
echo sequences are used for faster T2-like imaging and can be com- mural changes can also be characterized as to distribution, location,
bined with cardiac gating and inversion recovery to improve ves- and morphology, which may provide clues for suggesting the dif-
sel wall images. Cardiac gating is used with spin echo techniques ferential diagnosis of the disease process. Mural changes and con-
to eliminate artefacts from cardiac motion and to depict anatomic trast enhancement can also be followed on repeat studies to assess
abnormalities better in the thoracic aortic region and arch branches. response to treatment or progression of disease (Figure 18.2).
Intravenous contrast may be used with T1-weighted sequences to On the venous side, some vasculitides have a significant venous com-
demonstrate abnormal vascularity in soft tissues or organs, includ- ponent. Ultrasonography is well established as the imaging method of
ing arterial walls. This requires low doses of gadolinium and, usu- choice for extremity deep venous thrombosis, with magnetic reso-
ally, a delay before the start of imaging. nance venography (MRV) and CT venography also being options.
MRI can be used for focused vascular imaging using magnetic Conventional venography is rarely used today, most often when the
resonance angiography (MRA) techniques, with the goal of gen- calf veins cannot be adequately seen by other methods and suspicion
erating images of the vessel lumen similar to conventional angiog- is high for thrombosis. MRI best demonstrates occlusion of the venous
raphy. Vascular imaging is often possible with non-invasive MRA drainage of the brain. Evaluation of intestinal or solid organ venous
time-of-flight or phase-contrast techniques. However, contrast drainage and superior vena cava/ inferior vena cava (SVC/IVC) is best
MRA using an intravenous bolus of gadolinium, although mini- demonstrated by cross-sectional techniques of CT or MRI.
mally invasive, has replaced in many areas the non-contrast MRA
sequences because it provides the best arterial detail without the Organ changes and complications
artefacts of non-contrast techniques, which in turn results in faster Cross-sectional imaging can provide additional insight into the
and more accurate exams. Contrast-MRA images are obtained with diagnosis of a vasculitis by demonstrating characteristic changes
precise timing after a fast bolus of larger amounts of gadolinium, and distribution of involvement of organs affected. In smaller-vessel
in contrast with contrast-enhanced MRI for organ parenchyma vasculitides where vessel changes are not detectable, the parenchy-
enhancement, described in this section. Images are obtained in a mal effects of the vasculitis are most often the only cross-sectional
three-dimensional acquisition, which allows viewing of the data imaging findings. Arterial flow compromise may be evident as
in any desired plane, optimizing detection of abnormalities, and ischaemic changes or infarcts in the affected organs. Venous outflow
depiction of anatomy (Hartnell 1999). After the MRA images of compromise will also result in ischaemic changes and congestion.
the vessel lumen are obtained, delayed postcontrast images are Arterial aneurysms may rupture, appearing as intraparenchymal or
obtained to assess thickening of the vessel wall and its enhance- subcapsular haemorrhage. The perivascular inflammatory process
ment, which is thought to correlate with active inflammation. By may also extend to the organ’s parenchyma and become prominent
using the technique of ECG-gated inversion recovery T1-weighted as areas of abnormal CT attenuation or MRI signal. Nodules and
gradient echo, the conspicuity of mural enhancement can be masses may be seen in vasculitides associated with granuloma for-
enhanced while reducing artefacts from vessel pulsatility and car- mation. CT and MRI may detect perivascular inflammation as infil-
diac motion (Desai et al. 2005). trative changes in the fat surrounding vessels, most commonly seen
Advantages of MRI include its versatile orientation of the imaging in the mediastinum, retroperitoneum, and mesentery. Rupture of
plane, use of non-ionizing radiation, lack of significant nephrotoxic- aneurysms and bleeding from infarcted organs may present as free
ity of gadolinium, and excellent tissue contrast. Disadvantages include haemorrhage or focal haematomas anywhere in the body.
the length of time of the exam, its poor imaging of arterial calcium/ The optimal cross-sectional imaging modality for a particular
calcifications, reduced spatial resolution, expense, more limited avail- patient depends on the body part to be imaged and the advantages of
ability, potential for claustrophobia limiting the tolerability of the each modality. Certain modalities are well established as providing
250 SECTION 4 imaging and percutaneous interventions

(a) (b)

(c)

Fig. 18.2 Wegener’s granulomatosis. (a) CT scan through the upper chest shows thickening around the lumens of the left subclavian and left carotid arteries (arrows)
with extension of inflammation into the mediastinal fat. (b) CT scan at the aortic arch level shows thickening along the left lateral wall of the aortic arch (arrow). (c) CT
scan taken through the aortic arch several months after therapy shows reduction in the degree of thickening of the left wall of the aortic arch (arrow).

the best information for a specific body part. For example, MRI is CT has the advantage of faster scanning with coverage of the entire
the modality of choice for imaging the brain and central nervous body in one scan, which may be an issue in some cases. MRI, on
system for changes of vasculitis. CT, including regular thickness the other hand, has the advantage of not requiring ionizing radia-
scanning and high-resolution thin section scans, is the established tion, the cumulative effect of which has become more of a concern,
best modality for studying lung disease. In most cases, neither of particularly in the younger patient who will require repeat imaging
these examples requires contrast administration. Other areas can for disease and treatment response evaluation.
be imaged with either modality, depending on the situation. For Selecting appropriate imaging modalities for the wide spectrum
example, the abdomen is frequently screened with ultrasound, of clinical settings is often challenging because complex technolo-
which can be definitive for some organ lesions, but often additional gies need to be matched to the complexity of disease conditions
information is needed from CT or MRI. CT is excellent for abdomi- and to the particular requirements of a given patient. Consultation
nal disease and is often also used for screening, but lesions can be between radiologists and referring clinicians is an important com-
missed without intravenous contrast administration. MRI can then ponent to achieve a thorough, efficient, and safe patient evaluation.
be used to demonstrate abdominal disease in patients with renal
insufficiency or severe iodinated contrast allergy, although concern Imaging of specific vasculitis
for nephrogenic systemic fibrosis as a complication of receiving
gadolinium in patients with severe renal insufficiency has curtailed Takayasu’s arteritis
the use of this imaging technique in this clinical situation. Another Takayasu’s arteritis is the ideal vasculitis for MRI and CT imaging
example is imaging of the orbit, in which CT best demonstrates with cross-sectional images and angiographic techniques. Because
bone changes, as in the destructive lesions found in granulomatosis large elastic arteries including the aorta, its major branches, and
with polyangiitis (Wegener’s granulomatosis). On the other hand, the pulmonary arteries are affected, mural changes are well within
MRI best demonstrates the soft tissue extension of disease and the spatial resolution of CT and MRI, which gives these imaging
characterizes best the abnormalities by its signal characteristics. modalities an important role in early diagnosis. The large calibre
CHAPTER 18 cross-sectional imaging in vasculitis 251

lumen. MRI can also detect wall thickening with the advantage
that no contrast is needed to distinguish between the wall and
the lumen of the artery (Figures 18.4 and 18.5) (Matsunaga et al.
1998). Infiltration of the adjacent mediastinal fat can sometimes
be observed with CT and MRI, corresponding to the perivascu-
lar infiltration by various cells occasionally seen around the vasa
vasorum.
CT and MRI findings have been used to try to document response
to treatment and therefore to predict which early lesion may pro-
gress to stenosis. Based on a small number of patients, MRI stud-
ies have shown resolution of arterial wall contrast enhancement,
oedema, and thickening after treatment, which may correlate with
prevention of stenotic lesions (Aluquin et al. 2002; Andrews 2002).
The largest series following CT findings before and after treatment
showed that about half the patients had absence of arterial wall
Fig. 18.3 Takayasu’s arteritis. CT scan through the upper abdomen shows thick, enhancement and a decrease in the amount of wall thickening after
contrast enhancing wall of the aorta (arrow) and an inner ring of low density treatment. In this study, thickening progressed in 25% of patients
(curved arrow) indicating the inner wall. (Paul et al. 2001).
Ultrasound can also demonstrate the mural and luminal changes
of this arteritis. In a study evaluating the brachiocephalic vessels,
of the thoracic aorta and its orientation perpendicular to the axial all patients had subclavian artery involvement (one-third of them
scanning plane make this artery ideally suited for CT evaluation bilaterally), and the majority (69%) had common carotid artery
(Chung et  al. 1996). In the younger affected population, athero- involvement, which was always bilateral. The mural thickening
sclerotic changes in the vessel wall are usually not present to con- tends to be homogeneous, circumferential, iso- or hyperechoic, and
fuse the diagnosis. The main advantage of cross-sectional imaging typically involves the proximal half to two-thirds of the common
over conventional angiography is the demonstration of mural carotid arteries when affected, with a sharp distal cut-off for the wall
changes, which have been noted to be more frequent than luminal changes (Figure 18.6). Rarely, patients may have internal carotid
changes and would be usually missed by conventional angiography artery involvement. Doppler evaluation is useful to demonstrate
(Yamada et al. 1998). In addition, the potential complications of the haemodynamic effects of the stenotic disease. Disadvantages
conventional angiography are avoided. of ultrasound include underestimation of subclavian artery disease
The significant feature of early disease is thickening of the aortic and difficulty imaging the proximal subclavian artery, which may
or large vessel wall. At this stage of the disease, conventional angi- result in a false-negative exam (Sun et al. 1996).
ography relies on detecting descending aortic wall thickening as a During the late phase, or pulseless stage, of the disease, occlu-
subtle increased distance between the intraluminal contrast and the sive arterial changes predominate as a result of pronounced fibrotic
air in the lung. Because the luminal diameter is not affected, angiog- thickening of the adventitia and intimal thickening. The diagnosis
raphy can easily miss the subtle mural changes of early disease. The of Takayasu’s arteritis is most often made during this stage of the
aorta and innominate artery are the most common sites for mural disease, frequently confirmed by angiography. CTA and MRA pro-
changes in the presence of a normal-appearing angiogram (Park vide excellent angiographic images of the aorta and major branches
et al. 1997). Normally, the aortic wall is less than 1 mm in thickness to make the diagnosis non- or minimally invasively (Figure 18.7).
and the outer edge of the aorta is sharply defined. In patients with
Takayasu’s arteritis, this sharp vessel definition is lost. CT without
contrast may demonstrate higher-attenuation thickened arterial
walls (Park 1996). This high attenuation may be due to prolifera-
tion of fibrous and connective tissue and increased calcium content
in the media and adventitia (Sharma et al. 1996). A ‘double ring’
pattern of aortic wall thickening has been described in CT with i.v.
contrast (Figure 18.3) (Park 1996; Hayashi et al. 1991). An inner,
poorly enhancing ring is thought to represent mucoid or gelatinous
swelling of the intima (Hayashi et al. 1991) or the low-attenuation
intima trapped between the increased attenuation of the lumen
and the media/ adventitia (Sharma et  al. 1996). The outer and
thicker well-enhanced ring is thought to represent active medial
and adventitial inflammation probably with enlarged vasa vaso-
rum. Enhancement may occur in the early arterial phase or may
be delayed (Park et al. 1997). With proper timing of the scanning
during the contrast bolus in the arterial phase, subtle thickening
can be detected with high sensitivity by CT (Matsunaga et al. 1997).
Timing is important because non-contrast or delayed images after Fig. 18.4 Takayasu’s arteritis. MRI cross-section through the mid-chest shows wall
contrast cannot reliably identify the aortic wall separate from the thickening (between the arrows) of the descending thoracic aorta.
252 SECTION 4 imaging and percutaneous interventions

(a) (b)

Fig. 18.5 Takayasu’s arteritis. (a) Transverse MRI through the mid-chest shows thickening of the wall of the descending thoracic aorta (arrow) with high signal intensity.
(b) MRA coronal view of the abdominal aorta with gadolinium enhancement shows subtle irregularity of the suprarenal aortic lumen (arrows) in addition to slight
expansion of the diameter.

(a) (b)

Fig. 18.6 Takayasu’s arteritis. (a) Ultrasound examination in longitudinal view of the common carotid artery shows circumferential, hypoechoic wall thickening measured
at 2.5 mm (arrow). Normal thickness is less than 0.8 mm. (b) This larger field of view shows the segmental distribution of disease. There is sharp cut-off of the wall
thickening involving the left half of the image (arrow) while there is no evidence of wall thickening to the right of the arrow.

(b)

(a)

Fig. 18.7 Takayasu’s arteritis. (a) MRI through the upper chest shows wall thickening of the innominate artery (arrow IA) and of the left subclavian artery (arrow LSCA).
Between these two is a stenotic remnant of the left common carotid artery. (b) MRI with oblique sagittal view shows stenosis of the left subclavian artery
(arrow LSCA) with circumferential wall thickening and severe stenosis of the left common carotid artery (arrow LCCA) from its origin.
CHAPTER 18 cross-sectional imaging in vasculitis 253

(Figure  18.12). CT best demonstrates this, because MRI misses


subtle calcifications due to the lack of signal. Calcification is usu-
ally fine, sharp, and pencil-like and involves the arch and descend-
ing thoracic aorta. This is consistent with the dystrophic type of
calcifications seen from deposits of calcium in scarred media and
intima. Takayasu’s arteritis should be strongly suspected when a
relatively young female patient is found to have this type of arterial
calcification. Degenerative type of calcifications may also be seen
when secondary atherosclerosis develops and is demonstrated by

(a)

Fig. 18.8 Takayasu’s arteritis. MRI through the mid-chest shows the descending
thoracic aorta (arrow) to have a small calibre and to be occluded.

Sensitivity and specificity for luminal changes in the thoracic aorta


and major branches has been shown to be 93% or greater by CT
when compared to angiography (Yamada et al. 1998). Takayasu’s
arteritis is the only aortitis resulting in stenosis/ occlusion of the
aorta (Figure  18.8). As in conventional angiography, findings
include long and diffuse or short and segmental irregular stenosis/
occlusions of major branches of the aorta near their origins. Skip
areas of involvement are common when imaging lumen irregular- (b)
ity/ stenosis but more diffuse mural involvement has been demon-
strated in the angiographically normal areas (Sharma et al. 1996).
Commonly involved are the left subclavian artery, left common
carotid artery (CCA), innominate artery, renal artery, celiac artery,
superior mesenteric artery, and the pulmonary artery (Figure 18.9).
Stenotic lesions of the thoracic aorta are more common than in the
abdominal aorta. Skip areas of involvement and abundant collater-
alization are frequently seen. Fusiform or sacular true aneurysms,
ectasia, and poststenotic aneurysms can be seen most commonly
in the aorta and also in aortic branches (Figures 18.10 and 18.11),
with ruptures also reported (Regina et  al. 1998). Cine MRI can
accurately depict the aortic insufficiency that often accompanies
dilatation of the ascending aorta (Matsunaga et al. 1998). Aortic
dissections have been reported in the descending thoracic aorta
but are rare and all reported cases had associated hypertension
in addition to the media degeneration of the disease (Matsunaga
et al. 1997). At ultrasound, diffuse homogeneous circumferential
thickening of the vessel wall in the proximal CCA can be seen, with
Doppler findings of increased flow velocity and turbulence through
the involved segments when stenotic. The distal common, internal,
and external carotid arteries are spared and demonstrate dampened Fig. 18.9 Takayasu’s arteritis. Coronal view of MRA with gadolinium injection
waveforms. shows: (a) segmental stenosis of the innominate (brachiocephalic) artery
Another finding during the late stage of the disease includes (arrows); and (b) the left common carotid artery (arrows LT CC ART) has marked
aortic wall calcifications, which occur after 5  years of disease narrowing of its lumen, as seen in a different projection.
254 SECTION 4 imaging and percutaneous interventions

(b)

(a)

Fig. 18.10 Takayasu’s arteritis. (a) CT scan through the upper chest shows expansion of the innominate artery (black arrow) and left common carotid artery (white
arrow) with surrounding thickening of the arterial walls. The left subclavian artery has a much smaller calibre (curved arrow) and its wall is also thickened. (b) CT scan
through the chest taken at the level below the aortic arch shows ectasia of the ascending and descending segments of the thoracic aorta (arrows). There is slight wall
thickening in the descending segment.

(a) (b)

Fig. 18.11 Takayasu’s arteritis. (a) Abdominal aortogram shows marked irregularity of the infrarenal aorta and a small saccular aneurysm on the left side (arrow). (b) CT
scan through the infrarenal aorta at the level of the eccentric saccular aneurysm (arrow).

rough, irregular, and heavy calcifications in the damaged intima Pulmonary arterial stenoses are specific for Takayasu’s arteritis
(Matsunaga et  al. 1997). It has been suggested that early and versus other forms of arteritis and can be demonstrated by CT or
delayed mural enhancement on CT represents active disease, while MRI. In the early phase of the disease, wall thickening has been
lack of mural enhancement, high precontrast attenuation, and wall detected when conventional angiography demonstrates a normal
calcifications are more common in inactive disease (Antoniou lumen. Involvement is more frequent in upper lobe pulmonary
et al. 1998). arteries and in segmental branches followed by subsegmental
CHAPTER 18 cross-sectional imaging in vasculitis 255

(a) (b)

Fig. 18.12 Takayasu’s arteritis. (a) CT scan through the upper chest shows scattered calcification in the wall of the descending thoracic aorta (arrow) indicating
end-stage Takayasu’s arteritis. (b) CT scan through the lower chest shows high-grade stenosis of the aorta (arrow).

branches (Park et  al. 1995). The main differential diagnosis for of luminal arterial changes in the thorax, upper extremities, and
these findings is chronic thromboembolic disease and, therefore, lower extremities, high-resolution multidetector CTA with 3D
abnormal findings in systemic arteries should be sought to make reconstructions and 3D MRA are suitable to use instead of con-
the differentiation. Later in the disease, findings include dilatation ventional angiography. Typical findings are the same as in conven-
of the central pulmonary arteries and a ‘pruned tree’ appearance tional angiography with stenosis, occlusions, and skip lesions. The
of peripheral pulmonary arteries. CT may show a flame-shaped typical long, smooth stenosis of the distal subclavian and axillary
termination of the pulmonary artery (Matsunaga et  al. 1997), arteries seen with this vasculitis can be clearly demonstrated with
which is also seen in aortic branches. Enlarged collateral com- CTA and MRA. CT and MRI are the best modalities for demon-
munication between the systemic and pulmonary arterial circula- strating aortic involvement with GCA, both for the initial diagnosis
tion should be sought as indirect evidence of haemodynamically and to follow patients for complications, which include aneurysms
significant pulmonary arterial involvement. On chest CT, espe- and dissections (Figure 18.13). A population-based cohort study
cially with high-resolution scanning, the pulmonary parenchyma demonstrated marked increased risk of aortic aneurysm develop-
may demonstrate mosaic attenuation, which is seen as patchy ment in GCA patients versus matched controls (Evans et al. 1995).
low attenuation areas thought to represent regional hypoperfu- Aneurysms were more commonly late sequelae of the disease,
sion. Pulmonary vessels within these areas of low attenuation may with median time of 5.8 years after the initial diagnosis for devel-
show reduced calibre and number compared to the surrounding opment of thoracic aortic aneurysm and 2.5 years for abdominal
lung. These parenchymal findings correlate with the presence of aortic aneurysm. When aortic root dilatation is present, cine MRI
pulmonary arteritis on angiography. Subpleural reticulolinear techniques may demonstrate the commonly associated aortic valve
changes and pleural thickening may be seen, but these findings insufficiency.
do not correlate with the presence of angiographic findings of Thickening of the wall of the aorta or proximal arch vessels in
arteritis (Takahashi et al. 1996). Also reported are bilateral, small, GCA is well demonstrated by CT or MRI (Figures 18.14, 18.15,
poorly marginated, centrilobular nodules and branching linear and 18.16a, b). The more typical involvement of medium-sized
opacities that resolve with treatment and recur with recurrence arteries can be diagnosed with higher resolution CT and MR.
of symptoms. These may represent distended pulmonary arteries Wall changes in smaller arteries of the extremities or in abdominal
receiving high-pressure systemic flow form bronchial artery col- aortic branches that are more affected by motion artefact can be
laterals (Hara et al. 1998). imaged with high-resolution CT. The main differential diagnosis
for mural changes in these vessels in the older patient population
Giant cell arteritis typically affected by GCA is atherosclerosis. Differentiation of
Giant cell arteritis (GCA), as is the case with Takayasu’s arteri- atherosclerosis from GCA remains a challenge in the early stages
tis, produces changes in the arterial wall and lumen that may of the disease. Features that suggest GCA include circumferen-
be demonstrated by CT, MRI, and US.1 The arterial segments tial wall thickening, the distribution of the vessels affected, and
involved are more distal than in Takayasu’s arteritis, which requires inflammatory changes in the surrounding fat. CT has the advan-
higher-resolution scanning to detect the findings. For evaluation tage over MRI in the detection of wall calcifications seen as part of
atherosclerotic changes.
Imaging features of GCA and Takayasu’s arteritis overlap and
1 Editor’s note: the nosology of Takayasu’s arteritis and giant cell arteritis is distinction between the two relies on the more central aortic and
based on differing ages of occurrence: TA is typically a disease seen in people branch origin involvement and the typical earlier onset of disease
under 50 years of age, and GCA is seen people aged 50 years and over. seen in Takayasu’s arteritis.
256 SECTION 4 imaging and percutaneous interventions

(a) (b)

Fig. 18.13 Giant cell arteritis with late development of aneurysm and dissection. (a) CT scan through the aortic arch shows a dissecting aneurysm with the true lumen
anterior (arrow) and false lumen posterior (curved arrow). (b) CT scan through the mid-chest level shows an aneurysm of the descending aortic segment at the caudal
end of the dissection (arrow). The pulmonary artery (P) is also aneurysmal.

with conflicting results. The main challenge remains in distinguish-


ing vasculitis from atherosclerosis. One study reported the presence
of a hypoechoic halo around the lumen of the temporal artery as
a characteristic finding in temporal arteritis not seen with athero-
sclerosis (Figure 18.17) (Schmidt et al. 1997). The halo disappeared
after a mean of 16 days of treatment with glucocorticoids and is
thought to represent wall oedema, rather than cellular infiltrate. In
this study, 12 of 16 biopsies that included the arterial segment with
a halo were positive (Schmidt et al. 1997). In another study, all of
the six positive biopsies had halos while two patients with halos
had negative biopsies (Venz et al. 1998). A more recent study of
32 patients with suspected GCA showed that absence of the halo
sign or inflammatory stenosis predicted with certainty a negative
biopsy, which could reduce the high percent of negative biopsies
being performed (LeSar et al. 2002). Similar halos have also been
Fig. 18.14 Giant cell arteritis. CT scan through the upper chest below the level reported in upper extremity and neck arteries with GCA (Schmidt
of the aortic arch shows ectasia of the ascending and descending segments and
et al. 1999; Skopinski et al. 1999). The most recent meta-analysis of
thickening of the wall of the aorta (arrows).
studies evaluating the halo sign in GCA found sensitivity and speci-
ficity of 68% and 91% for unilateral findings and 43% and 100%
for bilateral findings (Arida et al. 2010). These findings confirm the
A particular MRA imaging sequence appears to be effective to usefulness of the halo sign and suggest it should be incorporated
detect active vasculitis in the aorta for both GCA and TAK (prob- early in the diagnostic algorithm of GCA. Importantly, the 100%
ably also for patients with Behçet’s syndrome and Cogan’s syn- specificity of bilateral disease should confirm the diagnosis and
drome). It is a post-gadolinium, T1-weighted image sequence perhaps obviate the need to biopsy.
which by adjusting the inversion time can detect high signal inten- Other studies have found wall thickening or presence of a halo
sity in the aortic wall apparently indicating acute inflammatory to be uncommon and have focused on evaluation of blood veloc-
tissue damage (Figure  18.16c). The initial report described this ity by Doppler examination (Lauwerys et  al. 1997; Barrier et  al.
finding for Takayasu’s arteritis (Desai et al. 2005). 1982; Puechal et al. 1995). One such study demonstrated that a low
The superficial temporal artery is commonly involved with GCA distal temporal artery velocity best distinguishes between GCA
and represents an opportunity to make the diagnosis. Although this and polymyalgia rheumatica or normal controls (Lauwerys et al.
artery can be easily biopsied for diagnosis, the biopsy requires a 1997). Conflicting results have been reported on the positive or
long segment of artery, potential bilateral biopsies, and proper tim- negative predictive value of Doppler studies. To improve sensitiv-
ing relative to the course of the disease (Hunder and Weyand 1997). ity and specificity, some investigators have included examination
Investigators have used high-frequency ultrasound examination of of multiple arteries (temporal, facial, and ophthalmic) to obtain
this artery to make the diagnosis or to improve the utility of the a combined score (Puechal et  al. 1995). Additional studies with
biopsy, by identifying a shorter, ultrasonographically abnormal larger number of patients and different conditions are needed. In
arterial segment. Studies have looked at wall thickening, luminal the future, a combination of anatomical and Doppler ultrasound
irregularity, stenosis or occlusions, and changes in blood velocity, findings of multiple extracranial arteries may prove more reliable
CHAPTER 18 cross-sectional imaging in vasculitis 257

(a) (b)

(c)

Fig. 18.15 Giant cell arteritis. MR scans through the neck with three different sequences. (a) T1 sequence shows circumferential thickening of the left common carotid
artery (LCCA) wall (white arrow) compared to the normal imperceptible wall of the right common carotid artery (RCCA, black arrow). Low signal around the LCCA
represents inflammation extending into the perivascular fat. Normal, high T1 signal fat is seen around the RCCA. (b) T2 sequence shows markedly increased signal
intensity of the LCCA wall (black arrow) from inflammation compared to the normal RCCA (white arrow). (c) Gadolinium enhancement of the LCCA wall (black arrow)
is best appreciated by comparing with the precontrast image (a). Notice no significant enhancement of the normal RCCA (white arrow).

(a) (b) (c)

Fig. 18.16 A 76-year-old female with acute GCA. MRI and MRA of the chest: three imaging sequences. (a) T2-weighted, IIR fs, dark blood image shows thickening and
oedema of the aortic wall (arrows). (b) T1-weighted, postgadolinium lava image shows contrast enhancement of the aortic wall (arrow). (c) T1-weighted, postgadolinium
delayed enhancement with adjusted inversion time to maximize the signal intensity in the aortic wall showing inflammatory tissue damage. Illustrations courtesy of James
Glockner, MD, Mayo Clinic.
258 SECTION 4 imaging and percutaneous interventions

for determining the absence or presence of disease and aid in the


decision whether or not to perform a biopsy.

Polyarteritis Nodosa; Microscopic Polyangiitis


The 1990 ACR classification did not differentiate polyarteritis
nodosa (PAN) and microscopic polyangiitis (MPA). Although
they share symptoms, there are important differences in anatomi-
cal, clinical, and laboratory findings which reflect unique patho-
genetic processes. PAN involves mostly medium-sized arteries,
while MPA involves small arteries, venules, and capillaries. Their
dominant clinical manifestations are similar in that both cause neu-
rological and gastrointestinal symptoms, as well as constitutional
symptoms. Important clinical differences are that PAN does not
cause glomerulonephritis nor does it affect the lungs. They differ
also in pathogenesis: PAN may be related to viruses such as HBV
or malignancy such as hairy cell leukaemia and MPA is one of the
ANCA-associated diseases. Considerable overlap in imaging find-
ings is seen with other necrotizing vasculitis, such as granuloma-
Fig. 18.17 Giant cell arteritis. Transverse ultrasound examination of the tosis with polyangiitis (Wegener’s granulomatosis) (Figure  18.2)
superficial temporal artery shows circumferential wall thickening (between the and systemic lupus erythematosus. Spatial resolution limits the
arrows) and an hypoechoic halo (curved arrow). ability of cross-sectional imaging to detect subtle arterial changes

(a) (b) (c)

(d)
(f)

(e)

Fig. 18.18 Polyarteritis nodosa with multiorgan involvement. CT scans through the upper and mid-abdomen shows: (a) aneurysm of left gastric artery (arrow); (b) small
cortical infarction of the right kidney (curved arrow), small subcapsular hepatic hematoma (arrow), and small bowel wall thickening from ischaemia (between white
arrows); (c) aneurysm of the inferior mesenteric artery with perivascular inflammatory changes (arrow). Abdominal angiograms show: (d) lateral aortogram shows a
fusiform aneurysm of the left gastric artery (arrow); (e) renal arteriogram shows zone of infarction in the mid-right kidney (between the arrows) and diffuse narrowing of
a primary branch of the right renal artery (curved arrow), additional irregularities of the renal artery are also noted; (f) fusiform aneurysm of the inferior mesenteric artery
(arrow), multiple smaller aneurysms, and luminal irregularities are present in the distribution of the IMA.
CHAPTER 18 cross-sectional imaging in vasculitis 259

(a) (b)

Fig. 18.19 Polyarteritis nodosa. (a) MRA with gadolinium coronal view shows irregularity of the renal parenchyma on the right (arrows). The left kidney has similar disease
but is not well identified because it is only partially included in the section. (b) Left renal arteriogram shows diffuse irregularity and stenosis of the renal artery branches.

of stenosis, occlusions, and dilatations often seen by conventional These pulmonary changes are best demonstrated by CT. Cardiac
angiography in these smaller arteries. Improvements in technol- involvement may present with cardiac enlargement and with
ogy allow CTA to image microaneurysms in the 2–3 mm diam- changes of pericarditis, including pericardial thickening and/or
eter range, which typify PAN, but detection is difficult. With more effusions (Gunal et al. 1997). Cardiac involvement can be imaged
advanced disease, CTA and MRA can frequently detect arterial with echocardiography, MRI, or CT.
abnormalities (Figure 18.18a, c, d and f). CT mainly, but also MRI The liver and mesenteric arteries are also commonly involved
and US, do provide excellent visualization of the organ changes and with PAN and are a major cause of morbidity and mortality. In the
complications accompanying this necrotizing vasculitis. CTA also liver, aneurysms can be detected by cross-sectional imaging, most
provides a less-invasive and preferred method, compared to con- often at arterial branching points (Figure 18.20). As in the kidneys,
ventional angiography, for following patient response to treatment peripheral areas of infarction may be seen. Spontaneous rupture
by evaluating improvement in organ perfusion and regression of of aneurysms may occur, resulting in contained intraparenchymal
aneurysms. or subcapsular haematomas, or free intraperitoneal haemorrhage.
The kidney is involved in greater than 85% of patients with PAN Cross-sectional imaging is again helpful to exclude malignancy,
and MPA, and multiple findings have been described. CTA and adenoma, or trauma as the cause of haemorrhage (Holzknecht
MRA fail to demonstrate the typical intrarenal microaneurysms et al. 1997). The jejunum is the segment of bowel most commonly
of 1  mm or less, but may demonstrate larger intra- and extrare-
nal aneurysms as well as stenosis and occlusions of the main renal
artery and division branches. CT or MRI can demonstrate multiple
peripheral wedge-shaped areas of cortical ischaemia/ infarction
(Figures 18.18b, e and 18.19) (Moss and Bush 1992). An uncom-
mon presentation of PAN is a perinephric/ subcapsular haemor-
rhage or a retroperitoneal haemorrhage from rupture of a renal
artery aneurysm. Ultrasound always detects a perirenal abnor-
mality but often cannot distinguish haemorrhage from tumour or
abscess. CT and MRI are helpful in detecting the abnormality and
excluding common potential causes, including tumours and poly-
cystic kidney disease. If no underlying disease is identified, angiog-
raphy should be the next step to exclude vasculitis. If CT or MRI
identifies multiple aneurysms and cortical infarcts, a necrotizing
vasculitis should be highly suspected, PAN being the most com-
mon (Brkovic et al. 1996).
The chest is the area of next most common involvement with
MPA. Potential findings include wedge-shaped or round, periph-
eral pulmonary infiltrates of non-segmental distribution simulat- Fig. 18.20 Polyarteritis nodosa with ruptured aneurysm in the liver. CT scan
ing thromboembolic disease with infarction. Cavitation may be through the upper abdomen without contrast enhancement shows a large
seen with these infiltrates. Massive pulmonary oedema may be the aneurysm of a hepatic artery branch (arrow) with surrounding hematoma (curved
result of congestive heart failure from cardiac involvement in PAN. arrow) in the posterior aspect of the right lobe of the liver.
260 SECTION 4 imaging and percutaneous interventions

involved and CT may demonstrate bowel wall thickening and imaging modalities may detect these changes described for PAN in
oedema from ischaemia/ infarction (Figure 18.18b). CT is also the Section Polyarteritis Nodosa; Microscopic Polyangiitis.
best imaging modality to detect pneumoperitoneum, which can be
seen when infarction results in bowel perforation. Rheumatoid arthritis
Rheumatoid arthritis (RA) may be complicated by vasculitis that
Kawasaki’s disease can resemble the changes described above for PAN. CT can detect
Kawasaki’s disease (KD) has a definite predilection for the coronary visceral arterial involvement with aneurysms, stenosis, and occlu-
arteries. Cardiac catheterization is the gold standard for diagno- sions, and the complications of infarctions, haemorrhage, and
sis of coronary disease and echocardiography has been the most bowel perforations. Aortitis can also be seen with CT or MRI, with
used non-invasive imaging modality for diagnosis and follow-up. predilection for the ascending aorta and development of aortic
Cardiac-gated MRI and multidetector coronary CTA are useful valve insufficiency. Changes of pericarditis with pericardial thick-
to evaluate coronary pathology and cardiac function abnormali- ening and effusions may be demonstrated with CT, MRI, or US.
ties, and can be used to replace catheter-based coronary angiog-
raphy for the diagnosis and late evaluation of this disease. Ideally, Systemic lupus erythematosus and
non-invasive imaging may replace higher-risk invasive procedures antiphospholipid antibody syndrome
in this young population. Systemic lupus erythematosus (SLE) causes thrombotic vascular
Echocardiography can help establish the diagnosis of KD by changes, arterial or venous, in a significant number of patients.
detecting coronary artery dilatation or aneurysms. It has also been Venous thrombosis is most common in the deep venous system of
used to follow regression of the disease after treatment has been ini- the extremities where vascular ultrasound plays a primary role for
tiated. Ischaemic changes can also be indirectly assessed by evalu- detection, although deep venous thrombosis (DVT) is a non-specific
ation of regional wall motion (Takahashi 1997). Echocardiography finding. Venous thrombosis can also occur more centrally, charac-
has limitations when it comes to detecting distal aneurysms and teristically involving the renal veins, and has also been reported for
occlusive lesions. It is also more difficult to consistently perform in the vena cavae, mesenteric veins, the superior sagittal sinus, and
older children (Duerinckx et al. 1997). Transoesophageal echocar- other sites (Si-Hoe et al. 1997; Flusser et al. 1996). CT and MRI
diography has been used for the diagnosis and follow-up of adult are well suited for detection of renal vein or vena cava thrombosis.
KD when transthoracic echocardiography is unable to detect the CT findings related to renal vein thrombosis include a dilated renal
aneurysms (Habon et al. 1998). vein containing thrombus, IVC thrombus in 40 to 50% of patients,
MRI has many advantages for cardiac imaging. It can visualize the presence of pericapsular venous collaterals when changes are more
arterial lumen, the presence of intimal proliferation or thrombus, chronic, and renal parenchymal changes including enlargement
and detect flow. It also offers improved visualization of more distal and an abnormal enhancement pattern (Si-Hoe et al. 1997). MRI
aneurysms compared to echocardiography and better measurement can also demonstrate these findings, and is particularly useful when
of total length of long aneurysms, thereby stratifying patients to i.v. contrast material cannot be used due to renal insufficiency. MRI
treatment regimens and follow-up schedules (Duerinckx et al. 1997). is also preferred for brain imaging. Central venous thrombosis in
Advances in MRI technology, with faster speeds of image acquisi- the thorax, from SLE or antiphospholipid syndrome (APS), can be
tion, have led to increasing use of MRI for diagnosis and evaluation detected by CT or MRI (Alcazar-Ramirez et al. 1997). MRI has the
of KD of the coronary arteries. The largest series comparing MRI advantage of detecting abnormalities without i.v. contrast by using
to conventional angiography showed complete agreement of find- gradient echo sequences. Colour Doppler ultrasound is not useful
ings between the two modalities in 13 paediatric patients, ages 3 to for detecting SVC thrombosis but may be misleading when evaluat-
8 years (Mavrogeni et al. 2004). Although multidetector CTA could ing the subclavian and brachiocephalic veins where they cannot be
be used to evaluate the coronary arteries in early disease, MRI has compressed to establish thrombotic occlusion.
the significant advantage of avoiding ionizing radiation exposure Aortitis has been reported in cases of APS, detected by CT
in this young patient population. MRI can also demonstrate active and MRI (Seror et  al. 1998). These cases involved the infrarenal
inflammation and oedema in the walls of the vessels (McMahon abdominal aorta and showed segmental, circumferential aortic
et al. 2005). For diagnosis of early disease, coronary angiography can wall thickening with high attenuation on CT and also with infil-
be supplanted by the combination of echocardiography and MRI. tration of the periaortic fat. On MRI, aortic wall thickening with
Multidetector CTA has been used in the evaluation of late find- enhancement after gadolinium was seen. In the one case, conven-
ings of coronary KD in adolescents and young adults and has been tional angiography and ultrasound were negative. The usefulness of
shown to have excellent correlation with coronary angiography cross-sectional imaging for treatment follow-up was shown when
findings (Kanamaru et al. 2005). CT is the best imaging modality CT demonstrated resolution of changes after 6 to 8 weeks of glu-
for determining aneurysm size and also for follow-up of throm- cocorticoid therapy. Two cases of aortic thrombosis, one infrarenal
bus formation and calcifications associated with these aneurysms and one supraceliac, have been reported. Renal artery thrombosis
(Fuyama et al. 1996). At the later stages of the disease, when find- or stenosis can occur in primary or secondary APS.
ings of coronary stenosis and occlusions have implications for SLE commonly affects the kidneys. CT detects changes of severe
treatment, CTA has the improved resolution compared to MRI to disease by demonstrating diffuse enlargement or diminution in
detect these changes in peripheral arteries. renal size. CT may also demonstrate renal vein thrombosis and
Kawasaki disease can also result in aneurysms of the aorta and other potentially treatable causes of renal failure, including ureteral
in other major arteries, including iliac, axillary, and renal arteries. obstruction and abscesses. Spontaneous subcapsular renal haema-
Pericarditis can also occur with this disease. All cross-sectional tomas secondary to vasculitis have also been reported.
CHAPTER 18 cross-sectional imaging in vasculitis 261

In SLE patients presenting with abdominal pain, CT plays a key rupture, which is the leading cause of death in BS. Aneurysms are
role in excluding a surgical abdomen and preventing unnecessary difficult to treat because they tend to recur after surgical repair at the
exploratory laparotomy when the symptoms are due to mesenteric anastomotic sites (Akiyama et al. 1998; Berkmen 1998). With MRI,
vasculitis and serositis. Lupus mesenteric vasculitis (LMV) may T1 spin echo sequences are used to show the anatomy, locate the
mimic other more common lesions, and often presents with severe, aneurysm, and evaluate the surrounding area for additional lesions.
disabling abdominal pain. Delayed recognition is potentially fatal if Then, non-contrast MRA techniques may be performed, includ-
it leads to infarction and perforation. If instituted early, glucocor- ing time-of-flight and gradient echo sequences. The time-of-flight
ticoid treatment may stop progression. Abdominal radiographs are technique may underestimate the size of the aneurysm due to arte-
not helpful and barium studies are non-specific. The key CT finding fact produced by saturation of flow in areas of high turbulence, but
for this diagnosis is a palisade-like prominence of the mesenteric this is overcome by the information from the T1 spin-echo images
vessels leading to the involved bowel loop. Mesenteric vessels are (Berkmen 1998). If findings are not clear, gadolinium MRA or CTA
seen as thickened, non-tapering vessels, which appear increased may be used for improved visualization of luminal abnormalities.
in number because of their increased prominence (the comb sign) Arterial lesions occur most frequently in the aorta followed by the
(Ko et  al. 1997; Si-Hoe et  al. 1997). These findings may corre- pulmonary, femoral, popliteal, subclavian, and carotid arteries, with
late with inflammatory cell infiltration in the wall of vessels and occasional coronary artery involvement (Berkmen 1998; Park et al.
perivascular oedema resulting in vessel engorgement. The involved 1984). This is the only known acquired disorder causing pulmonary
bowel loop is usually in the superior mesenteric artery distribu- artery aneurysms and these are best demonstrated by pulmonary
tion and most often demonstrates symmetrical and circumferen- CTA or MRA (Greene et al. 1998). CT has the advantage of also dem-
tial bowel wall thickening (>3 mm, <1 cm), with a low-attenuation onstrating potentially associated lung parenchymal abnormalities,
middle layer between higher-attenuation inner and outer layers including regional differences in perfusion, air space nodules, and
(double halo or target sign). Specific CT findings for bowel infarc- pleural-based, wedge-shaped masses which may result from pulmo-
tion should be sought to avoid delay of surgery treatment. These nary infarction. The lobar arteries are involved half the time, and the
findings include pneumatosis intestinalis and air in the mesenteric main pulmonary arteries and segmental branches involved approxi-
veins or portal veins. Infarction may progress to perforation, which mately 25% of the time each (Tunaci et al. 1999). Aneurysms tend
can be seen by CT as free fluid or abscess formation with or without to disappear or at least regress after several months to years. Visceral
pneumoperitoneum. artery involvement is rare but has been reported, including a renal
artery aneurysm rupture identified by CT (Sueyoshi et  al. 1996).
Behçet’s syndrome Mesenteric vascular engorgement, similar to SLE changes described
Behçet’s syndrome (BS) lends itself to diagnostic imaging with CT, in Section Systematic Lupus Erythematosus and Antiphospholipid
MRI, and US due to its involvement of large veins and arteries. This Antibody Syndrome, has also been seen (Ha et al. 1998).
is one disease where conventional angiography should be avoided A case of internal carotid artery dissection imaged by colour-flow
if possible due to the higher risk of aneurysm formation at arterial Doppler US has been reported (Pannone et al. 1998). The authors
puncture sites. Venous punctures also carry the risk of initiating postulate that arterial dissections in BS may be due to the same vasa
venous thrombosis. Both US and non-contrast MRA techniques vasorum vasculitis suspected as the basis for aneurysm formation.
can be performed without puncture of the vascular system. Parenchymal abnormalities in BS secondary to small-vessel
Veins are involved more frequently than arteries in BS (25%). vasculitis can also be seen with CT and MRI, correlating with
Findings include a wide range of lesions, from subcutaneous areas of infarction/ ischaemia, haemorrhage, and inflammation.
thrombophlebitis, to DVT, to large vein occlusion and intracardiac Involvement of the brain and central nervous system is well evalu-
thrombus. Among large veins, the IVC is most frequently involved ated with MRI. Lesions are more common in the midbrain and
followed by the SVC, common iliac veins, and hepatic veins posterior forebrain structures and can be single or multifocal foci
(Terzioglu et al. 1998). Dural sinus thrombosis may occur. Venous with low T1 and high T2 signal. Diffusion weighted images (DWI)
occlusion may be accompanied by varix formation. The most com- can help differentiate BS ischaemic stroke, but lesions can also
mon lesion is superficial thrombophlebitis of the lower extremity. be hyperintense on DWI. Contrast enhancement pattern can be
Extremity DVT is best evaluated by ultrasound, including com- variable. The main differential diagnosis is multiple sclerosis with
pression images. If US is indeterminate due to poor visualization the location of lesions in the mesodiencephalon and grey matter
of extremity veins, MR venography techniques can be performed involvement favouring vasculitis. Chronic BS can result in brain-
without the need for contrast. CT venography techniques can also stem and cerebellar atrophy, which has high specificity for the con-
be performed but require peripheral i.v. injection of iodinated con- dition (Al-Araji and Kidd 2009).
trast. CT and MR produce excellent images of larger veins of the
thorax, abdomen, and pelvis. One limitation of MRV techniques is Thromboangiitis obliterans
difficulty distinguishing between absence of flow due to occlusion (buerger’s disease)
and very slow flow due to severe stenosis. Alternate MR sequences Thromboangiitis obliterans (TAO) affects small and medium-sized
may be employed to determine if the vein lumen is filled with arteries and veins, primarily in the extremities. Conventional angi-
thrombus. ography is frequently used to confirm the diagnosis and establish
Arterial involvement in BS includes arterial occlusions and aneu- the extent of the disease. However, very few reports in the litera-
rysm formation. Aneurysms occur more frequently than occlu- ture discuss cross-sectional imaging of TAO. MRA techniques may
sions and carry a poor prognosis. They are often multiple and may reach limits of spatial resolution when only small distal arteries of
show rapid progression leading to pseudoaneurysm formation and the hand or foot are involved, but newer contrast-enhanced MRA
262 SECTION 4 imaging and percutaneous interventions

techniques may prove adequate to demonstrate the findings. US


can be used in the diagnosis of extremity venous thrombosis and
Doppler ultrasonography can demonstrate tapered narrowing and
occlusions of extremity arteries. Findings are often bilateral and
one of the key observations is the absence of atheromatous plaque
at or proximal to the sites of occlusion (Fiessinger 1997). Other
sites of arterial involvement besides the extremities have been
reported, including mesenteric and renal arteries (Iwai 1998; Gomi
et al. 1978), and these would be amenable to diagnosis with MRA
and CTA.

Chronic periaortitis
Chronic periaortitis is caused by lymphocytic inflammation of the
adventitia of the aorta and occurs most frequently in the infrarenal
abdominal aorta.2 Patients present with pain, which may mimic
a ruptured aneurysm, generalized malaise, and elevated erythro- Fig. 18.21 Inflammatory aneurysm of the abdominal aorta. There is considerable
cyte sedimentation rates. Different named disorders have been thickening of the wall (arrows) of the infrarenal aortic aneurysm. Note there is
described, all involving the same inflammatory process but vary- relative sparing of the posterior wall. There is also considerable thrombus between
ing in the degree of inflammation and fibrosis, in the presence of the opacified lumen (curved arrow) and the thickened inflamed wall.
aneurysm, and in the involvement of adjacent organs. These disor-
ders are idiopathic retroperitoneal fibrosis, or mediastinal fibrosis,
and inflammatory aneurysm. CT is well established as the preferred Granulomatosis with polyangiitis (formerly
modality for making the diagnosis, since US may miss the find- wegener’s granulomatosis) and eosinophilic
ings. CT shows a thick, enhancing rind of soft tissue around the granulomatosis with polyangiitis (formerly
aorta (Figures 18.21 and 18.22). In inflammatory aneurysms, this churg–strauss syndrome)
confluent soft tissue thickening is located primarily anterolateral
with posterior sparing. Other types of retroperitoneal fibrosis tend These two diseases produce necrotizing vasculitis mostly limited
to produce more circumferential involvement around the aorta to small arteries; any organ system may become affected but on
and more irregular periaortic extension of the process. MRI is cross-sectional imaging studies, granulomatous lesions are not
also used to image periaortitis but different sequences are needed always distinguished from necrotic lesions of small-vessel vascu-
to delineate the lumen, the aortic wall, and periaortic soft tissue litis. In a few patients with these diseases, larger arteries may show
rind (Figure  18.23). In inflammatory aneurysms, T2-weighted imaging evidence typical of vasculitis: aortic wall thickening and
images may demonstrate a multilayered, or ‘onion skin’, appear- evidence of coronary artery vasculitis in the rare patients with
ance with variable signal intensities for the aneurysm and the adja- granulomatosis with polyangiitis (GPA) (Figure  18.2) and small,
cent inflammation. CT and MRI can also delineate adjacent organ false aneurysms in visceral artery branches and irregularities of
involvement, in particular vena cava stenosis/ occlusion and ure- intracranial arteries rarely in patients with eosinophilic granulo-
teral obstruction with hydronephrosis (Tennant et al. 1993; Arrive matosis with polyangiitis (EGPA). Small-vessel vasculitis affects
et al. 1995). branches that are too small to be detected by any imaging tech-
nique. However, the organs that are affected may show secondary
Cogan’s syndrome evidence of vasculitis: tissue damage and infarction. The imaging
Cogan’s syndrome is a rare inflammatory disorder of unknown aeti- modalities of CT and MR are excellent for detecting such abnor-
ology. The primary clinical manifestations are ocular inflammation malities; however, these types of findings lack the specificity of a
and audiovestibular disease. Systemic manifestations of vasculitis vasculitis diagnosis compared to the patterns of other types of vas-
occur in a small percentage of patients. Vasculitis may involve the culitis that affect large and medium-sized arteries. Cross-sectional
aortic valve, the aorta, the primary branches, and medium-sized imaging is also excellent to detect organ involvement by granu-
arteries. These areas of involvement can be identified with all lomatous lesions. Significant cardiac involvement occurs in both
imaging modalities. The findings are similar to those of GCA and GPA and EGPA. MR scanning is particularly effective to evalu-
TA:  inflammatory wall thickening and aneurysm formation of ate pathology of the heart: myocardium, pericardium, valves, and
the aorta, aortic valve insufficiency, wall thickening and stenoses coronary arteries.
of the medium-sized arteries, and occasionally pseudoaneurysm Patients with GPA have a high incidence of sinonasal and lung dis-
formation which may lead to rupture of abdominal visceral arter- ease. Bony erosion of the nasal septum and turbinates may be found,
ies. Organ involvement by small-vessel vasculitis may also occur. indicating necrosis. Membrane thickening of the sinuses and gran-
Vasculitis is preceded by eye and inner ear disease and, indeed, may ulomatous masses with bony erosion of sinus walls, including the
occur many months or years later (Vollertsen et al. 1986; Matteson orbital walls, may also occur. Bony thickening of the sinus walls may
and Stanson 2012). be seen, presumably as a reaction to inflammation. Abnormalities of
the bony structures are best seen with CT scans. Soft tissue changes
2Editors’s note: A significant number of reported cases of periaortitis and are better depicted by MRI; T2-weighted images distinguish inflam-
aortitis appear to be manifestations of IgG4-related systemic disease (Stone mation and T1 weighted images depict granulomatous masses.
and Khosroshahi 2011). Abnormalities in the lungs are common:  extensive infiltrates,
CHAPTER 18 cross-sectional imaging in vasculitis 263

(a) (b)

Fig. 18.22 Periaortitis with progression. (a) The aortic lumen is demarcated by calcification (arrow). The irregular thick wall beyond this is inflammation (curved arrow).
(b) Three months later there is marked increase in the degree of wall thickening (curved arrows).

(a) (b)

(c)

Fig. 18.23 Inflammatory aneurysm. (a) MRI T1 shows aneurysm of the infrarenal aorta. The thick rind (arrow) is difficult to distinguish from the lumen (curved arrow).
(b) MRI through the same level but with a sequence of gradient recalled echo shows high signal intensity of the lumens of the aorta (arrow) and IVC (curved arrow).
There is not clear distinction of the thick wall of the aorta from thrombus in the aneurysm. (c) MRA with gadolinium enhancement in the late phase shows high signal
intensity of the wall thickening (arrow). The lumen of the aorta, in the centre of the aneurysm, is partially enhanced and can be distinguished from the low signal intensity
mural thrombus (curved arrow).

nodules, and masses. The larger masses may show cavitation sec- thickening and enhancement as well as evidence of parenchymal
ondary to necrosis. Tracheal and bronchial wall thickening may lead lesions of ischaemia caused by vasculitis and granulomatous lesions.
to stenoses of the airways (Silvera et al. 2012). Involvement of the Patients with EGPA have similar organ involvement as those with
central nervous system is uncommon. MRI can detect meningeal GPA, but the sinonasal involvement is limited to sinus membrane
264 SECTION 4 imaging and percutaneous interventions

thickening, polyps, and fluid. Pulmonary abnormalities are much Fuyama, Y., Hamada, R., Uehara, R., Yano, I., Fujiwara, M., Matoba, M., et al.
less evident; infiltrates tend to clear and recur in different locations, (1996). Long-term follow up of abdominal aortic aneurysm complicat-
presumably related to eosinophilic infiltration and granuloma- ing Kawasaki disease: comparison of the effectiveness of different imag-
ing methods. Acta Paediatrica Japonica, 38, 252–5.
tous lesions rather that necrotizing vasculitis. Nodules do occur
Gomi, T., Ikeda, T., and Yuhara, M. (1978). Renovascular hypertension due to
but are small and not likely to show cavitation. Thin section CT Buerger’s disease. Japanese Heart Journal, 19, 308–14.
images of the chest show abnormalities in most patients, includ- Greene, R.M.E., Saleh, A., Taylor, A.K.M., Callaghan, M., Addis, B.J., Nzewi,
ing distal bronchial wall thickening and ground-glass attenuation O.C., et  al. (1998). Non-invasive assessment of bleeding pulmonary
(Bouhaouala et al. 2012). artery aneurysms due to Behçet disease. European Radiology, 8, 359–63.
Gunal, N., Kara, N., Cakar, N., Kocak, H., Kahramanyol, O., and Cetinkaya,
References E. (1997). Cardiac involvement in childhood polyarteritis nodosa.
Akiyama, K., Hirota, J., Ohkado, A., and Shiina, Y. (1998). Multivarious International Journal of Cardiology, 60, 257–62.
clinical manifestations of multiple pseudoaneurysms in Behçet’s disease. Ha, H.K., Lee, H.J., Yang, S.K., Ki, W.W., Yoon, K.H., Shin, Y.M., et al. (1998).
Journal of Cardiovascular Surgery, 39, 175–8. Intestinal Behçet syndrome: CT features of patients with and patients
Al-Araji, A. and Kidd, D.P. (2009). Neuro-Behçet’s disease:  epidemiology, without complications. Radiology, 209, 449–54.
clinical characteristics and management. Lancet Neurology, 8, 192–204. Habon, T., Toth, K., Keltai, M., Lengyel, M., and Palik, I. (1998). An adult
Alcazar-Ramirez, J.D., Fernandez-Nebro, A., Abarca, M., Salgado, F., De case of Kawasaki disease with multiplex coronary aneurysms and myo-
Haro-Liger, M., Rodriguez-Andreu, J., et al. (1997). Central vein throm- cardial infarction: the role of transesophageal echocardiography. Clinical
bosis in the antiphospholipid syndrome. Lupus, 6, 549–51. Cardiology, 21, 529–32.
Aluquin, V.P., Albano, S.A., Chan, F., Sandborg, C., Pitlick, P.T. (2002). Hara, M., Sobue, R., Ohba, S., Kitase, M., Sasaki, S., Ogino, H., et al. (1998).
Magnetic resonance imaging in the diagnosis and follow up of Takayasu’s Diffuse pulmonary lesions in early phase Takayasu arteritis pre-
arteritis in children. Annals of the Rheumatic Diseases, 61, 526–9. dominantly involving pulmonary artery. Journal of Computer Assisted
Andrews, J., Kaskard, D.O. (2002). Current management options in Behçet’s Tomography, 22, 801–3.
disease. Minerva Medica, 93, 335–45. Hartnell, G.G. (1999). Great vessels of the chest. In Magnetic resonance of the
Antoniou, A., Vlahos, L., and Mourikis, D. (1998). Abdominal Takayasu’s heart and great vessels: clinical applications (ed. J. Bogaert, A.J. Duerinckx
arteritis:  imaging with color duplex sonography. European Radiology, and F.E. Rademakers), pp. 246–8. Springer-Verlag, Berlin Heidelberg.
8, 547–9. Hayashi, H., Katayama, N., Takagi, R., Matsuoka, Y., and Matsunaga, N.
Arida, A., Kyprianou, M., Kanakis, M., and Sfikakis, P.P. (2010). The diag- (1991). CT analysis of vascular wall during the active phase of Takayasu’s
nostic value of ultrasonography-derived edema of the temporal artery aortitis. European Radiology, 1 (Suppl.), S239.
wall in giant cell arteritis: a second meta-analysis. BMC Musculoskeletal Holzknecht, N., Gauger, J., Helmberger, T., Stabler, A., and Reiser, M. (1997).
Disorders, 11, 44. Cross-sectional imaging findings in a case of polyarteritis nodosa with
Arrive, L., Correas, J.M., Leseche, G., and Ghebontni, L. (1995). Inflammatory ruptured hepatic artery aneurysm. American Journal of Roentgenology,
aneurysms of the abdominal aorta:  CT findings. American Journal of 169, 1317–9.
Roentgenology, 165, 1481–4. Hunder, G.G. and Weyand, C.M. (1997). Sonography in giant-cell arteritis.
Barrier, J., Potel, G., Renaut-Hovasse, H., Hanh, T.H., Peltier, P., Chamary, V., New England Journal of Medicine, 337, 1385–6.
et al. (1982). The use of Doppler flow studies in the diagnosis of giant Iwai, T. (1998). Buerger’s disease with intestinal involvement. International
cell arteritis. Selection of temporal artery biopsy site is facilitated. JAMA, Journal of Cardiology, 66, S257–63.
248, 2158–9. Kanamaru, H., Sato, Y., Takayama, T., Ayusawa, M., Karasawa, K., Sumitomo,
Berkmen, T. (1998). MR angiography of aneurysms in Behçet’s disease:  a N., Harada, K. (2005). Assessment of coronary artery abnormalities by
report of four cases. Journal of Computer Assisted Tomography, 22, 202–6. Multislice spiral computed tomography in adolescents and young adults
Bouhaouala, M.H., Bouslama, K., Charfi, M., Mhiri, N., Ben Romdhane, with Kawasaki disease. American Journal of Cardiology, 95, 522–5.
M.H., El Beji, M., and Hendaoui, L. (2012). Churg-Strauss syn- Ko, S.F., Lee, T.Y., Cheng, T.T., Ng, S.H., Lai, H.M., Cheng, Y.F., et al. (1997).
drome. In Systemic vasculitis, imaging features (ed. L. Hendaoui, CT findings at lupus mesenteric vasculitis. Acta Radiologica, 38, 115–20.
A.W. Stanson, M.H. Bouhaouala, and F. Joffre), pp. 285–299. Lauwerys, B.R., Puttemans, T., Houssiau, F.A., and Devogelaer, J.P. (1997).
Springer-Verlag, Berlin Heidelberg. Color Doppler sonography of the temporal arteries in giant cell arteritis
Brkovic, D., Moehring, K., Doersam, J., Pomer, S., Kaelble, T., and Riedasch, and polymyalgia rheumatica. Journal of Rheumatology, 24, 1570–4.
G. (1996). Aetiology, diagnosis and management of spontaneous perire- LeSar, C.J., Meir, G.H., DeMasi, R.J., Sood, J., Nelms, C.R., Carter, K.A., et al.
nal haematomas. European Urology, 29, 302–7. (2002). The utility of color duplex ultrasonography in the diagnosis of
Chung, J.W., Park, J.H., Im, J.G., Chung, M.J., Han, M.C., and Ahn, H. (1996). temporal arteritis. Journal of Vascular Surgery, 36, 1154–60.
Spiral CT angiography of the thoracic aorta. Radiographics, 16, 811–24. Mavrogeni, S., Papadopoulos, G., Douskou, M., Kaklis, S., Seimenis, I.,
Desai, M.Y, Stone, J.H., Foo, T.K., Hellman, D.B., Lima, J.A.C., and BAras, P., Nikolaidou, P., Bakoula, C., Karanasios, E., Manginas, A.,
Bluemke, D.A. (2005). Delayed contrast enhanced MRI of the aor- Cokkinos, D. D. (2004). Magnetic resonance angiography is equivalent
tic wall in Takayasu’s arteritis: initial experience. American Journal of to X-ray coronary angiography for the evaluation of coronary arteries
Roentgenology, 184, 1427–31. in Kawasaki disease. Journal of the American College of Cardiology, 43,
Duerinckx, A.J., Troutman, B., Allada, V., and Kim, D. (1997). Coronary MR 649–52.
angiography in Kawasaki disease. American Journal of Roentgenology, Matsunaga, N., Hayashi, K., Sakamoto, I., Matsuoka, Y., Ogawa, Y., Honjo,
168, 114–6. K., et al. (1998). Takayasu arteritis: MR manifestations and diagnosis
Evans, J.M., O’Fallon, W.M., and Hunder, G.G. (1995). Increased incidence of of acute and chronic phase. Journal of Magnetic Resonance Imaging, 8,
aortic aneurysm and dissection in giant cell (temporal) arteritis. Annals 406–14.
of Internal Medicine, 122, 502–7. Matsunaga, N., Hayashi, K., Sakamoto, I., Ogawa, Y., and Matsumoto, T.
Fiessinger, J.N. (1997). Thromboangiitis obliterans (Buerger’s disease). Revue (1997). Takayasu arteritis: protean radiologic manifestations and diag-
Du Rhumatisme (English Edition), 64, 281–2. nosis. Radiographics, 17, 579–94.
Flusser, D., Abu-Shakra, M., Baumgarten-Kleiner, A., Flusser, G., and Matteson, E.L. and Stanson, A.W. (2012). Cogan’s syndrome. In Systemic vascu-
Sukenik, S. (1996). Superior sagittal sinus thrombosis in a patient with litis—imaging features (ed. L. Hendaoui, A.W. Stanson, M.H. Bouhaouala,
systemic lupus erythematosus. Lupus, 5, 334–6. and F. Joffre), pp. 123–35. Springer-Verlag, Berlin Heidelberg.
CHAPTER 18 cross-sectional imaging in vasculitis 265

McMahon, C.J., Su, J.T., Taylor, M.D., Krishnamurthy, R., Muthupillai, R., Si-Hoe, C.K., Thng, C.H., Chee, S.G., Teo, E.K., and Chng, H.H. (1997).
Kovalchin, J.P., Vick, G.W. 3rd. (2005). Images in cardiovascular medi- Abdominal computed tomography in systemic lupus erythematosus.
cine. Detection of active coronary arterial vasculitis using magnetic Clinical Radiology, 52, 284–9.
resonance imaging in Kawasaki disease. Circulation, 112, e315–6. Silvera, S., Palangie, E., Guillevin, L., Hendaoui, L., and Legmann,
Moss, A.A. and Bush, W.H. (1992). The kidneys. In Computed tomography P. (2012). Imaging features of granulomatosis with polyangii-
of the body with magnetic resonance imaging (ed. A.A. Moss, G. Gamsu, tis (Wegener’s). In Systemic vasculitis, imaging features (ed. L.
and H.K. Genant), pp. 1003–5. W.B. Saunders Company, Philadelphia. Hendaoui, A.W. Stanson, M.H. Bouhaouala, and F. Joffre), pp. 259–78.
Pannone, A., Lucchetti, G., Stazi, G., Corvi, F., Ferguson, T.L., Massucci, M., Springer-Verlag, Berlin Heidelberg.
et al. (1998). Internal carotid artery dissection in a patient with Behçet’s Skopinski, S., Constans, J., Cherifi, H., Midy, D., Jarnier, P., Le Metayer, P.,
syndrome. Annals of Vascular Surgery, 12, 463–7. et  al. (1999). Inflammatory arteriopathy of the arms in the course of
Paul, J.F., Fiessinger, J.N., Sapoval, M., Hernigou, A., Mousseaux, E., Horton disease. Report of six cases. Journal des Maladies Vasculaires,
Emmerich, J., Piette, J.C. (2001). Follow-up electron beam CT for the 24, 45–8.
management of early phase Takayasu arteritis. Journal of Computer Stone, J. and Khosroshahi, A. (2011). Aortitis, periaortitis, and retroperito-
Assisted Tomography, 25, 924–31. neal fibrosis, as manifestations of IgG4-related systemic disease. Current
Park, J.H. (1996). Conventional and CT angiographic diagnosis of Takayasu Opinion Rheumatology, 23, 88–94.
arteritis. International Journal of Cardiology, 54, S165–71. Sueyoshi, E., Sakamoto, I., Hayashi, N., Fukuda, T., Matsunaga, N., Hayashi,
Park, J.H., Chung, J.W., Im, J.G., Kim, S.K., Park, Y.B., and Han, M.C. (1995). K., et al. (1996). Ruptured renal artery aneurysm due to Behçet’s disease.
Takayasu arteritis: evaluation of mural changes in the aorta and pulmo- Abdominal Imaging, 21, 166–7.
nary artery with CT angiography. Radiology, 196, 89–93. Sun, Y., Yip, P.K., Jeng, J.S., Hwang, B.S., and Lin, W.H. (1996).
Park, J.H., Chung, J.W., Lee, K.W., Park, Y.B., and Han, M.C. (1997). CT angi- Ultrasonographic study and long-term follow-up of Takayasu’s arteritis.
ography of Takayasu arteritis: comparison with conventional angiogra- Stroke, 27, 2178–82.
phy. Journal of Vascular and Interventional Radiology, 8, 393–400. Takahashi, K., Honda, M., Furuse, M., Yanagisawa, M., and Saitoh, K. (1996).
Park, J.H., Han, M.C., and Bettmann, M.A. (1984). Arterial manifestations of CT findings of pulmonary parenchyma in Takayasu arteritis. Journal of
Behçet’s disease. American Journal of Roentgenology, 143, 821–5. Computer Assisted Tomography, 20, 742–8.
Puechal, X., Chauveau, M., and Menkes, C.J. (1995). Temporal Doppler-flow Takahashi, M. (1997). Kawasaki disease. Current Opinion in Pediatrics, 9, 523–9.
studies for suspected giant-cell arteritis. Lancet, 345, 1437–8. Tennant, W.G., Hartnell, G.G., Baird, R.N., and Horrocks, M. (1993).
Regina, G., Fullone, M., Testini, M., Todisco, C., Greco, L., Rizzi, R., et al. Radiologic investigation of abdominal aortic aneurysm disease: com-
(1998). Aneurysms of the supra-aortic trunks in Takayasu’s disease. parison of three modalities in staging and the detection of inflammatory
Report of two cases. Journal of Cardiovascular Surgery, 39, 757–60. change. Journal of Vascular Surgery, 17, 703–9.
Schmidt, W.A., Kraft, H.E., Borkowski, A., and Gromnica-Ihle, E.J. (1999). Terzioglu, E., Kirmaz, C., Uslu, R., Sin, A., Kokuludag, A., Sagduyu, A., et al.
Color duplex ultrasonography in large-vessel giant cell arteritis. (1998). Superior vena cava syndrome together with multiple venous
Scandinavian Journal of Rheumatology, 28, 374–6. thrombosis in Behçet’s disease. Clinical Rheumatology, 17, 176–7.
Schmidt, W.A., Kraft, H.E., Vorpahl, K., Volker, L., and Gromnica-Ihle, E.J. Tunaci, M., Ozkorkmaz, B., Tunaci, A., Gul, A., Engin, G., and Acunas, B.
(1997). Color duplex ultrasonography in the diagnosis of temporal arte- (1999). CT findings of pulmonary artery aneurysms during treatment
ritis. New England Journal of Medicine, 337, 1336–42. for Behçet’s disease. American Journal of Roentgenology, 172, 729–33.
Seror, O., Fain, O., Dordea, M., Ghenassia, C., Coderc, E., and Sellier, N. Venz, S., Hosten, N., Nordwald, K., Lemke, A.J., Schroder, R., Bock, J.C., et al.
(1998). Aortitis with antiphospholipid antibodies: CT and MR findings. (1998). Use of high resolution color Doppler sonography in diagnosis of
European Radiology, 8, 1373–5. temporal arteritis. Rofo. Fortschritte auf dem Gebiete der Rontgenstrahlen
Shalhoub, J., Owen, D.R.J., and Leen, E.L.S (2012). Evaluation of carotid und der Neuen Bildgebenden Verfahren, 169, 605–8.
atherosclerosis and inflammation by contrast enhanced ultrasound. In Vollertsen, R.S., McDonald, T.J., Younge, B.R., Banks, P.M., Stanson, A.W.,
Systemic vasculitis, (ed. L. Hendaoui, A.W. Stanson, M.H. Bouhaouala, and Ilstrup, D.M. (1986). Cogan’s syndrome: 18 cases and a review of the
and F. Joffre), pp. 51–60. Springer-Verlag, Berlin Heidelberg. literature. Mayo Clinic Proceedings, 61, 344–61.
Sharma, S., Sharma, S., Taneja, K., Gupta, A.K., and Rajani, M. (1996). Yamada, I., Nakagawa, T., Himeno, Y., Numano, F., and Shibuya, H. (1998).
Morphologic mural changes in the aorta revealed by CT in patients Takayasu arteritis: evaluation of the thoracic aorta with CT. Radiology,
with nonspecific aortoarteritis (Takayasu’s arteritis). American Journal 209, 103–9.
of Roentenology, 167, 1321–5.
CHAPTER 19

PET imaging in vasculitis


Daniel Blockmans

Introduction an increased signal due to a technical artefact, which delineates the


contours of the human body (Figure 19.1). Increasingly, FDG-PET
Treatment of vasculitis often requires prolonged use of steroids and
scintigraphy is carried out in combination with a CT scan, which
other drugs affecting the immune system in seriously ill patients.
makes it possible to directly correlate a focus of increased FDG
Hence, the diagnosis of vasculitis and preferably the exact type of
consumption to an anatomical structure.
vasculitis should be firmly established. Proof of type is preferably
Pathologically increased FDG uptake can be observed in vari-
histological, for example a clearly inflamed medium layer of a tem-
ous conditions such as ischaemically threatened myocardial tissue
poral artery in giant cell arteritis (GCA) or a pauci-immune glo-
(Maes et al. 1994), cancer (Hawkins et al. 1992), or infection (Ichiya
merulonephritis in a patient with granulomatosis with polyangiitis
et al. 1996). Non-infectious inflammatory tissues also take up more
(GPA, formerly known as Wegener’s granulomatosis), but may
FDG, as in sarcoidosis (Brudin et al. 1994) or lymphadenopathy in
also be obtained by other means such as by classic angiography in
systemic lupus erythematous (Fey et al. 2004). Hence, FDG-PET
patients with Takayasu’s arteritis (TA) or classic polyarteritis nodosa
is a good method to search for causes of fever of unknown origin
(PAN), in whom it is hard to obtain biopsy proof of vasculitis.
(FUO), which can be the result of an underlying infection, tumour,
Temporal artery biopsy, moreover, cannot always be regarded
systemic disease, or miscellaneous process (Blockmans et al. 2001).
as the gold standard for diagnosing GCA. The negative predictive
Its use in patients with FUO has led to the observation that GCA,
value of a temporal artery biopsy taken in ideal circumstances is
which is a common cause of FUO in elderly patients, can be visual-
only in the range of 90% (Hall et al. 1983). In 1999, a variant of
ized on FDG-PET scintigraphy as an intense FDG uptake in the
GCA was described that was typified by involvement of the large
vessels and a higher likelihood of temporal artery sparing. In these
patients, the sensitivity of temporal artery biopsy was only 58%
(Brack et al. 1999). In patients with GPA without renal involvement,
histological proof of vasculitis is also often hard to obtain: nasal or
bronchoscopic biopsies frequently show necrosis or granulomatous
inflammation, and less often, vasculitis. In necrotizing small ves-
sel vasculitides (GPA and microscopic polyangiitis), the detection
of antineutrophil cytoplasmic antibodies (ANCA) directed against
proteinase-3 or against myeloperoxidase can substitute for the
lack of biopsy proof of vasculitis in patients with compatible clini-
cal pictures. In large and medium-sized-vessel vasculitis, there is
no laboratory test with high specificity for vascular inflammation.
Positron emission tomography (PET) with radioactive labelled
fluoro-18-deoxyglucose (FDG) may be an alternative way to dem-
onstrate vasculitis in a non-invasive manner and at an early state,
before stenosis and aneurysms—demonstrable on classic angiogra-
phy or magnetic resonance angiography—develop.
FDG is a positron-emitting glucose analogue with a half-life
of 110 minutes, which is transported across the capillary and cell
membranes in proportion to the rate of glucose uptake. It is phos-
phorylated by the enzyme hexokinase to FDG-6-phosphate, which
cannot be metabolized further, and it is trapped inside the cell.
Metabolically active cells consuming large amounts of sugar trap
large amounts of FDG, which enables them to be imaged. In the
normal human body, there is an intense FDG uptake in the brain,
which can only use glucose as its energy source, and in the myocar- Fig. 19.1 Normal FDG-PET scintigraphy showing FDG consumption in the brain
dium. FDG is excreted through the kidneys and, hence, the urinary and myocardium, and FDG accumulation in the urine bladder. The contours of
tract is readily visible. At the boundary of air and body, there is also the body are readily visible.
268 SECTION 4 imaging and percutaneous interventions

Fig. 19.2 Pathological FDG uptake in the thoracic aorta and the subclavian arteries in a 79-year-old female with GCA (thin arrows). There is also increased FDG uptake
in the shoulders (broad arrows), corresponding to associated PMR.

aorta and/or the subclavian, axillary, carotid, iliac, and femoral The subclavian arteries are the ones that most frequently take up
arteries. FDG on PET scan of persons with GCA (up to 75%), followed by
the abdominal and thoracic aorta in about half, while an increased
PET in large-vessel vasculitis FDG uptake in the axillary, carotid, iliac, and femoral arteries is
seen in 30–40% (Blockmans et  al. 2006). A  typical example of a
Giant cell arteritis and polymyalgia rheumatica positive PET scintigraphy in GCA is shown in Figure 19.2.
The studies on the use of FDG-PET in GCA have clearly shown The temporal arteries themselves cannot be visualized by
that GCA can involve many arteries beyond the temporal arter- FDG-PET scintigraphy, due to their small size (4  mm in diam-
ies. For many years, it has been known that GCA can involve eter required), their superficial localization, and the vicinity of
the aorta or other larger arteries, but it was thought to be rare. the FDG-consuming brain. So, purely cranial forms of GCA will
In 1999, before the widespread use of FDG-PET in GCA, Brack be missed by FDG-PET (Brodmann et al. 2004). The specificity of
et al. described a ‘large-vessel variant’ of GCA, in whom the tem- clearly increased FDG uptake in the thoracic aorta, the subclavian,
poral arteries were frequently spared but the large vessels were carotid, or axillary arteries is high for vasculitis, ranging between
involved (Brack et al. 1999). Using FDG-PET in biopsy-proven 95 and 100%. FDG uptake is typically symmetrical in GCA: both
GCA, we showed that more than half of patients had involve- subclavian or both iliac arteries take up FDG. The specificity of
ment of their larger thoracic arteries (Blockmans et al. 2000b). increased FDG uptake in the lower limbs (and perhaps also the
The exact frequency of involvement of a certain artery as judged abdominal aorta) for vasculitis is lower (70–80%), because these
by FDG-PET depends on the method used to classify an artery vascular territories are prone to atherosclerosis, which also has an
as ‘positive’. We and others have used a semiquantitative method, inflammatory component that can be responsible for false-positive
in which 0 stands for no FDG uptake, 1 for minimal (physio- vascular FDG uptake (Yun et al. 2001). One may assume that the
logical?) FDG uptake, 2 for clear uptake, and 3 for intense FDG intensity of the FDG uptake in atherosclerosis would be lower com-
uptake. Grades 2 and 3 are interpreted as pointing to vasculitis pared to that in vasculitis and would be more focal, while vasculi-
(Blockmans et al. 1999). This of course is imprecise, and others tis produces more linear FDG uptake. Very dramatic pictures of
have used ROI indexes (regions of interest, comparing counts to inflamed lower limb vasculitis in GCA as visualized by FDG-PET
background areas, e.g. the liver) (Hautzel et al. 2008; Lehmann scan have been published (Tato and Hoffmann 2006).
et al. 2011; Tezuka et al. 2012). For this semiquantitative evalu- As a rule, the intensity of vascular FDG uptake in GCA drops
ation, maximum standardized uptake values (SUVmax) are while the patient is being treated with steroids, paralleling the
assessed for predefined vascular ROI. The SUVmax are calcu- drop in inflammatory parameters. In some patients, however,
lated by dividing tissue activity by total activity injected. A ROC a clear vascular FDG uptake can still be detected after months
(receiver-operating characteristics) analysis can be performed to of steroid therapy and while the patient seems to be in complete
define a diagnostic cut-off value for vasculitis. remission (Blockmans et al. 2006). Vascular remodelling has been
CHAPTER 19 pet imaging in vasculitis 269

hypothesized to be the cause for this persistent FDG uptake, but


this has never been proven histologically. Once the patient has
taken steroids, pictures are harder to interpret and one cannot rely
entirely on results of FDG-PET scintigraphies to decide if a patient
is experiencing a relapse (Both et al. 2008). To judge if a patient is
relapsing, the results of the PET scan should be considered in con-
junction with the patients symptoms, the degree of inflammation
on blood sampling, and, eventually, the response to higher dose of
steroids. PET scans have no predictive role in defining the possibil-
ity of a future relapse, but patients with high vascular FDG uptake

Posterior
at diagnosis may be more prone to aortic dilatation in the years to

Left
come (Blockmans et al. 2008). This possible association has not yet
been confirmed by prospective studies.
In patients with GCA with complaints indicative of polymyal-
gia rheumatica (PMR), the presence of PMR correlated with an
increased FDG uptake in the shoulders, but not with a more-intense
FDG uptake in the subclavian or axillary arteries, as might have
been assumed. In patients with isolated PMR, in whom GCA had
been ‘excluded’ by a negative temporal artery biopsy, increased
FDG uptake in the shoulders and hips was found in about 90% of
patients, while half of them also showed an increased FDG uptake
in the spinous processes of the cervical and/or lumbar vertebrae
(Blockmans et al. 2007; Yamashita et al. 2012; Adams et al. 2012).
On magnetic resonance imaging of the vertebrae in PMR, an inter- Fig. 19.3 PET scintigraphy of a 74-year-old women who was diagnosed with
spinous bursitis could be shown, corresponding to the increased PMR 8 years earlier and experienced several relapses during steroid tapering. This
FDG uptake on PET scan (Salvarani et  al. 2008). Vascular FDG PET scan, taken on the occasion of her most recent relapse, shows increased
uptake in these patients with isolated PMR complaints could FDG uptake in the shoulders, hips, and spinous processes of cervical and lumbar
be detected in about 30%, especially in the subclavian arteries, vertebrae (arrows).
although of less intensity than in GCA patients. As in GCA, FDG
uptake does not disappear completely in all patients with PMR
under steroid treatment and, here also, FDG-PET scintigraphies experience, however, that FDG uptake is less intense than in GCA,
have no predictive role as to which patients will relapse. Increased probably reflecting a lower degree of vascular inflammation. As in
FDG uptake in the shoulders can be found in patients with rheu- GCA, vascular FDG uptake also decreases in most patients after the
matoid arthritis, periarthritis scapula–humeralis, or chondrocalci- start of steroid therapy.
nosis. FDG uptake in the cervical or lumbar spine combined with The role of FDG-PET scan in the assessment of activity of
vascular FDG uptake points to PMR. An example of a typical PET Takayasu’s arteritis has not been determined. In fact, larger pro-
scintigraphy in isolated PMR is shown in Figure 19.3. spective studies dealing with this problem are still lacking, due to
PET scanning has become a useful tool for the evaluation of the rarity of the disease and the limited availability of this technique
patients with atypical presentations of GCA or PMR, such as when in certain parts of the world. In earlier studies, there was a good
systemic complaints like fever or weight loss dominate the clinical pic- correlation between the results of FDG-PET and clinical activity of
ture, or even in asymptomatic patients with an unexplained elevation the disease (Webb et al. 2004; Andrews et al. 2004; Kobayashi et al.
of parameters of inflammation. All areas of increased FDG consump- 2005), while later studies showed less favourable results (Lee et al.
tion can be visualized by one single examination. For the detection 2009; Arnaud et al. 2009). Finally, two more recent retrospective
of GCA or PMR, a combined CT scan probably does not offer any studies seem to indicate that one indeed can rely on PET images
advantage over PET scintigraphy alone, because the larger thoracic to judge activity of the disease (Lee et al. 2012; Tezuka et al. 2012).
and limb arteries are easily recognizable on PET scintigraphy. If the
patient is examined on a combined PET/CT scan, the diameter of the Chronic periaortitis
aorta at the different levels can be measured. To judge the thickness of This form of vasculitis, which can be isolated or part of a broader
the aortic wall, a CT scan with higher resolution is needed. disease such as multifocal fibrosclerosis, sometimes related to high
IgG4 levels, can also be visualized by PET scintigraphy (Derdelinckx
Takayasu’s arteritis et al. 2000; Blockmans et al. 2002; Drieskens et al. 2002; Salvarani
The assessment of disease activity in Takayasu’s arteritis is dif- et al. 2005). Very intense FDG uptake surrounding the aorta and
ficult because clinical symptoms and biochemical parameters do common iliac arteries has been reported. During steroid or rituxi-
not always reflect the actual inflammatory state within the arte- mab treatment, FDG uptake normalizes (Derdelinckx et al. 2000;
rial wall. It is important to detect the disease early in its course Maritati et al. 2012). In the diagnosis and assessment of periaorti-
before irreversible structural wall changes such as stenosis or aneu- tis, a combined PET-CT scan is preferred over PET alone, because
rysms occur. Hara was the first to use FDG-PET in a patient with periaortitis is also easily visible on CT scan as a rim of inflamma-
Takayasu’s arteritis (Hara et al. 1999). As in GCA, FDG uptake in tion surrounding the aorta. Figure 19.4 shows PET and CT findings
the aorta and its primary branches can be readily seen. It is my in a 72-year-old male with abdominal periaortitis.
270 SECTION 4 imaging and percutaneous interventions

(a) (a)

(b) (b)

Fig. 19.4 (a) PET image showing increased FDG uptake surrounding the
abdominal aorta (arrow), which corresponds to a rim of inflammation (white Fig. 19.5 (a) FDG-PET scintigraphy in a 30-year-old male with known Behçet’s disease
arrow) around the aorta on CT scan (b). and persisting inflammation on biochemical testing. PET scan showed a focus of
intense FDG uptake in the right lung (arrow), which corresponded to an inflammatory
aneurysm of a branch of the right pulmonary artery (white arrow) on CT scan (b).

(b)

(a)

(c)

Fig. 19.6 (a) PET scintigraphy of a 55-year-old female patient in whom a diagnosis of GPA, limited to the upper airways, was made 1 year earlier, for which she was
treated with cyclophosphamide i.v. for 4 months and high-dose steroids, followed by azathioprine and a tapering course of steroids. Despite therapy, inflammatory
parameters remained high and a PET scan was performed, which showed a focus of increased FDG uptake in the upper pole of the left kidney (arrow). (b) On CT scan,
the differential diagnosis was made of a renal cell carcinoma or a GPA-associated granuloma (white arrow). (c) Kidney biopsy of this lesion revealed an inflammatory
process, in which neutrophilic polymorphonuclear leukocytes, histiocytes, and multinucleated giant cells (g) were abundant (haematoxylin eosin stain). (Courtesy of Prof.
E. Lerut, Department of Pathology, University Hospital Gasthuisberg, Leuven, Belgium).
CHAPTER 19 pet imaging in vasculitis 271

(a) (b)

Left
Posterior

Left
(c)

Fig. 19.7 (a) PET scintigraphy of a 39-year-old male in whom a diagnosis of limited GPA was made 32 months earlier. He was first treated with mycophenolate mofetil
and steroids for 6 months, the former was then switched to azathioprine. Thirteen months after diagnosis, he had a relapse manifested by pericarditis, and six i.v. pulses of
cyclophosphamide were given, followed again by azathioprine. Twenty-six months after diagnosis, a transitional cell carcinoma of the bladder was diagnosed, for which a
transurethral resection was performed, followed by mitomycin bladder instillations. In the work-up of this bladder tumour, a CT scan of the lungs was performed, which
showed a small tumour in the right lung; this had enlarged on imaging 6 months later. On PET scan this lesion took up FDG (arrow). Clinically, there were no signs of
GPA activity, and the patient was thought to be in remission of his GPA. (b) Fusion image of PET scan and CT scan, with white arrow pointing to the lesion. (c) Because
a metastasis of the transitional cell carcinoma was suspected, a transthoracic biopsy was performed, which showed inflammatory cells with centrally geographic
necrosis; this is rather typical for GPA. Note also scattered multinucleated giant cells (haematoxylin eosin stain). (Courtesy of Prof. E. Verbeken, Department of Pathology,
University Hospital Gasthuisberg, Leuven, Belgium).

PET in medium-vessel vasculitis PET in small-vessel vasculitis


Little has been published on this topic, due to the rarity of these Even more than in medium-sized-vessel vasculitis, the ves-
conditions (PAN and Kawasaki’s disease). In the few cases of PAN sels involved in small vessel vasculitis such as granulomato-
in whom we performed a PET scan, we could never visualize indi- sis with polyangiitis are far too small to be visualized by PET.
vidual inflamed vessels. Cutaneous involvement appears as an Glomerulonephritis is not visible on FDG-PET scanning.
intense signal at the site of the inflamed skin, but this of course is Granulomatous lesions in the lungs, sinuses, or kidney, however,
non-specific (Bleeker-Rovers et al. 2003). can be seen on PET scan (Blockmans et al. 2000a; Almuhaideb
In patients with Behçet’s disease, pulmonary artery aneurysms et al. 2011) and preferably on a combined PET-CT scan. Due to
and involvement of other larger arteries can be visualized on PET its nature, the differential diagnosis between vasculitis/granulo-
and further described on CT scan (Denecke et al. 2007; Loh et al. matosis, infection, or malignancy cannot be made by PET scan.
2010; Bin Cho et al. 2011). An example of such a lesion on PET scan In my experience, sequela lesions of GPA do not take up FDG.
in a patient with Behçet’s disease is given in Figure 19.5. Figures 19.6 and 19.7 show FDG images of granulomatous GPA
lesions in the kidney and lung, respectively.

multiorgan involvement of Wegener’s granulomatosis. British Journal of


References Radiology, 84, e202–4.
Adams, H., Raijmakers, P., and Smulders, Y. (2012). Polymyalgia rheumatica Andrews, J., Aj Nahhas, A., Pennell, D., et al. (2004). Noninvasive imaging
and interspinous FDG uptake on PET/CT. Clinical Nuclear Medicine, in the diagnosis and management of Takayasu’s arteritis. Annals of the
37, 502–5. Rheumatic Diseases, 63, 995–1000.
Almuhaideb, A., Syed, R., Iordnidou, L., Saad, Z., and Bomanji, J. (2011). Arnaud, L., Haroche, J., Malek, Z., et al. (2009). Is 18F-fluorodeoxyglucose
Fluorine-18-fluorodeoxyglucose PET/CT rare finding of a unique positron emission tomography scanning a reliable way to assess
272 SECTION 4 imaging and percutaneous interventions

disease activity in Takayasu arteritis? Arthritis and Rheumatism, 60, Fey, G., Jolles, P., Bucley, L., and Massey, G. (2004). 2-Deoxy-2-(18F)
1193–200. fluoro-D-glucose positron emission tomography uptake in systemic
Bin Cho, S., Yun, M., Lee, J., Kim, J. Shim, W., and Bang, D. (2011). Detection lupus erythematosus-associated adenopathy. Molecular Imaging and
of cardiovascular system involvement in Behçet’s disease using fluoro- Biology, 6, 7–11.
deoxyglucose positron emission tomography. Seminars in Arthritis and Hall, S., Persellin, S., Lie, J.T., O’Brien, P.C., Kurland, L.T., and Hunder, G.G.
Rheumatism, 40, 461–6. (1983). The therapeutic impact of temporal artery biopsy. Lancet, 2,
Bleeker-Rovers, C., Bredie, S., Van Der Meer, J., et al. (2003). F-18 fluorode- 1217–20.
oxyglucose positron emission tomography in diagnosis and follow-up Hara, M., Goodman, P., and Leder, R. (1999). FDG-PET finding in early-phase
of patients with different types of vasculitis. Netherlands Journal of Takayasu arteritis. Journal of Computer Assisted Tomography, 23, 16–8.
Medicine, 61, 323–9. Hautzel, H., Sander, O., Heinzel, A., et al. (2008). Assessment of large-vessel
Blockmans, D., Baeyens, H., Van Loon, R., Lauwers, G., and Bobbaers, H. involvement in giant cell arteritis with 18F-FDG PET; introducing
(2000a). Periaortitis and aortic dissection due to Wegener’s granuloma- a ROC-analysis-based cut-off ratio. Journal of Nuclear Medicine, 49,
tosis. Clinical Rheumatology, 19, 161–4. 1107–13.
Blockmans, D., Coudyzer, W., Vanderschueren, S., et al. (2008). Relationship Hawkins, R.A., Hoh, C., Glaspy, J., et al. (1992). The role of positron emission
between fluorodeoxyglucose uptake in the large vessels and late aortic tomography in oncology and other whole-body applications. Seminars
diameter in giant cell arteritis. Rheumatology, 47, 1179–84. in Nuclear Medicine, 22, 268–84.
Blockmans, D., De Ceuninck, L., Vanderschueren, S., Knockaert, Ichiya, Y., Kuwabara, Y., Sasaki, M., et  al. (1996). FDG-PET in infectious
D., Mortelmans, L., and Bobbaers, H. (2006). Repetitive lesions: the detection and assessment of lesion activity. Annals of Nuclear
18F-fluorodeoxyglucose positron emission tomography in giant cell Medicine, 10, 185–91.
arteritis: a prospective study of 35 patients. Arthritis andRheumatism, Kobayashi, Y., Ishii, K., Oda, K., et al. (2005). Aortic wall inflammation due
55, 131–7. to Takayasu arteritis imaged with 18F-FDG PET coregistered with
Blockmans, D., De Ceuninck, L., Vanderschueren, S., Knockaert, enhanced CT. Journal of Nuclear Medicine, 46, 917–22.
D., Mortelmans, L., and Bobbaers, H. (2007). Repetitive Lee, K., Cho, A., Choi, Y., et  al. (2012). The role of
18-fluorodeoxyglucose positron emission tomography in isolated poly- 18F-fluorodeoxyglucose-positron emission tomography in the assess-
myalgia rheumatica: a prospective study in 35 patients. Rheumatology, ment of disease activity in patients with Takayasu arteritis. Arthritis
46, 672–7. andRheumatism, 64, 866–75.
Blockmans, D., Knockaert, D., Maes, A., et al. (2001). Clinical value of (18F) Lee, S., Ryu, J., Kim, H., et al. (2009). Evaluation of disease activity using
fluoro-deoxyglucose positron emission tomography for patients with F-18 FDG PET-CT in patients with Takayasu arteritis. Clinical Nuclear
fever of unknown origin. Clinical Infectious Diseases, 32, 191–6. Medicine, 34, 749–52.
Blockmans, D., Maes, A., Stroobants, S., et  al. (1999). New arguments for Lehmann, P., Buchtala, S., Achajew, N., et  al. (2011). 18F-FDG PET as a
a vasculitic nature of polymyalgia rheumatica using positron emission diagnostic procedure in large vessel vasculitis—a controlled, blinded
tomography. Rheumatology, 38, 444–7. re-examination of routine PET scans. Clinical Rheumatology, 30, 37–42.
Blockmans, D., Stroobants, S., Maes, A., and Mortelmans, L. (2000b). Loh, H., Yung, G., Bui, C., Mansberg, R., and Comsa, M. (2010). Pulmonary
Positron emission tomography in giant cell arteritis and polymyalgia artery aneurysm with false-positive FDG PET in a patient with Behçet’s
rheumatica:  evidence for inflammation of the aortic arch. American disease. Clinical Nuclear Medicine, 35, 286–8.
Journal of Medicine, 108, 246–9. Maes, A., Flameng, W., Nuyts, J., et  al. (1994). Histological alterations in
Blockmans, D., Van Moer, E., Dehem, J., Feys, C., and Mortelmans, L. (2002). chronically hypoperfused myocardium: correlations with PET findings.
Positron emission tomography can reveal abdominal periaortitis. Circulation, 90, 735–45.
Clinical Nuclear Medicine, 27, 211–2. Maritati, F., Corradi, D., Versari, A., et  al. (2012). Rituximab therapy for
Both, M., Ahmadi-Simab, K., Reuter, M., et al. (2008). MRI and FDG-PET chronic periaortitis. Annals of the Rheumatic Diseases, 71, 1262–4.
in the assessment of inflammatory aortic arch syndrome in compli- Salvarani, C., Barozzi, L., Cantini, F., et al. (2008). Cervical intraspinous bur-
cated courses of giant cell arteritis. Annals of the Rheumatic Diseases, sitis in active polymyalgia rheumatica. Annals of the Rheumatic Diseases,
67, 1030–3. 67, 758–61.
Brack, A., Martinez-Taboada, V., Stanson, A., Goronzy, J.J., and Weyand C.M. Salvarani, C., Pipitone, N., Versari, A., et  al. (2005). Positron emission
(1999). Disease pattern in cranial and large-vessel giant cell arteritis. tomography (PET):  evaluation of chronic periaortitis. Arthritis and
Arthritis andRheumatism, 42, 311–7. Rheumatism, 53, 298–303.
Brodmann, M., Lipp, R., Passath, A., et al. (2004). The role of 2-18F-fluoro-2- Tato, F. and Hoffmann, U. (2006). Clinical presentation and vascular imag-
deoxy-D-glucose positron emission tomography in the diagnosis of ing in giant cell arteritis of femoropopliteal and tibioperitoneal arteries.
giant cell arteritis of the temporal arteries. Rheumatology, 43, 241–2. Analysis of four cases. Journal of Vascular Surgery, 44, 176–82.
Brudin, L., Valind, S., Rhodes, C., et  al. (1994). Flurine-18 deoxyglucose Tezuka, D., Haraguchi, G., Ishihara, T., et  al. (2012). Role of FDG
uptake in sarcoidosis measured with positron emission tomography. PET-CT in Takayasu Arteritis. Journal of the American College of
European Journal of Nuclear Medicine, 21, 297–305. Cardiology: Cardiovascular Imaging, 5, 422–9.
Denecke, T., Staeck, O., Amthauer, H., and Lopez Hänninen, E. (2007). PET/CT Webb, M., Chambers, A., Al-Nahhas, A., et al. (2004). The role of 18F-FDG
visualizes inflammatory activity of pulmonary artery aneurysms in Behçet PET in characterising disease activity in Takayasu arteritis. European
disease. European Journal of Medical and Molecular Imaging, 34, 370. Journal of Nuclear Medicine and Molecular Imaging, 31, 627–34.
Derdelinckx, I., Maes, A., Bogaert, J., Mortelmans, L., and Blockmans, D. Yamashita, H., Kubota, K., Takahashi, Y., et al. (2012). Whole-body fluoro-
(2000). Positron emission tomography in the diagnosis and follow-up of deoxyglucose positron emission tomography/computed tomography in
aortitis of the thoracic aorta. Acta Cardiologica, 55, 193–5. patients with active polymyalgia rheumatica:  evidence for distinctive
Drieskens, O., Blockmans, D., Van Den Bruel, A., and Mortelmans, L. (2002). bursitis and large-vessel vasculitis. Modern Rheumatology, 22, 705–11.
Riedel’s thyroiditis and retroperitoneal fibrosis in multifocal fibrosclero- Yun, M., Yeh, D., Araujo, L., Jang, S., Newberg, A., and Alavi, A. (2001). F-18
sis. Positron emission tomographic findings. Clinical Nuclear Medicine, FDG uptake in the large arteries. A new observation. Clinical Nuclear
27, 413–5. Medicine, 26, 314–9.
SECTION 5

Vasculitic diseases
and syndromes and
related disorders
CHAPTER 20

Approach to the diagnosis of


vasculitis in adult patients
Barri J. Fessler

The diagnosis of vasculitis is one of the most challenging tasks in and will discuss emerging advances that can be helpful in support-
the practice of medicine because of non-specific symptoms, over- ing the diagnosis.
lapping syndromes, lack of highly sensitive or specific diagnostic
tests, and the absence of generally accepted diagnostic criteria.
While classification criteria for the different vasculitic syndromes Initial approach to the diagnosis
have been proposed for research purposes (Arend et  al. 1990; The recognition that certain signs and symptoms may be due to an
Bloch et al. 1990; Calabrese et al. 1990; Hunder et al. 1990; Jennette underlying inflammatory vascular process is the first step in estab-
et al. 1994; Leavitt et al. 1990; Lightfoot et al. 1990; Masi et al. 1990; lishing a diagnosis of vasculitis. Clinical manifestations, including
Mills et al. 1990) the application of these criteria for diagnosis is rapidly progressive multisystem inflammatory disease, constitu-
not useful in the clinical setting (Rao et al. 1998; Sorensen et al. tional symptoms, chronic fevers, symptoms of tissue ischaemia
2000; Murchison et al. 2012) (see Chapter 1). Better understand- (especially in unusual patient populations such as in a young
ing of the aetiology, pathogenesis, clinical, and laboratory asso- woman), neuropathy, or suspicious skin lesions such as palpable
ciations has led to the 2012 revised international Chapel Hill purpura, should heighten suspicion for vasculitis. The patient’s
Consensus Conference on nomenclature of vasculitides (Jennette history and physical examination remain the most powerful diag-
et  al. 2013). Names were changed (eponyms replaced), defini- nostic tools available to the clinician. There is no single typical pres-
tions were updated, and categories not included in the initial 1994 entation for vasculitis; therefore recognition of patterns or clusters
Consensus Conference (Jennette et  al. 1994)  were added. There of signs and symptoms is critically important. Complaints of head-
remains a strong need for classification and diagnostic criteria to aches, scalp tenderness, jaw claudication, vision loss, and muscle
aid in the early diagnosis of vasculitis and facilitate research stud- pain/ stiffness raise the possibility of giant cell arteritis (Weyand
ies (see Chapter 1, reviewed in Waller et al. 2013). The diagnosis of and Goronzy 2003). New-onset cough with haemoptysis and hae-
a specific form of vasculitis ultimately depends on the identifica- maturia suggests the development of a pulmonary–renal syndrome
tion of particular patterns of clinical, radiographic, laboratory, and (Gallagher et al. 2002). Coupled with the presence of chronic sinus
histopathological features. involvement or asthma and eosinophilia, diagnoses such as gran-
The evaluation of a patient with symptoms suggestive of a vascu- ulomatosis with polyangiitis (GPA; previously called Wegener’s
litic syndrome must be individually tailored, based on the extent of granulomatosis) and eosinophilic granulomatosis with polyangiitis
organ involvement and tempo of the disease manifestations. Early (EGPA; previously called Churg–Strauss syndrome), respectively,
in the disease course, a vasculitic syndrome may cause vague con- emerge as possibilities (Lane et al. 2005a). The absence of a pulse
stitutional symptoms or involve only one organ system, making or blood pressure, or its unilateral decrease in an arm, suggests an
the timely diagnosis especially challenging. As other disease mani- occlusive vascular process such as Takayasu’s arteritis (Park et al.
festations evolve over time, morbidity and mortality associated 2005). The presence of Raynaud’s phenomenon and palpable pur-
with the syndrome increase; therefore, aggressive evaluation and pura in a patient with hepatitis C virus (HCV) infection raises the
prompt treatment is critically important in improving outcomes. possibility of cryoglobulinaemic vasculitis (Schott et  al. 2001).
The demonstration of vasculitis on tissue biopsy remains the gold Age of syndrome onset can also be informative. Giant cell arteritis
standard for diagnosis. In situations where the pretest probability generally develops in patients over 50 years of age (Machado et al.
of a vasculitic syndrome is high and biopsies cannot be obtained or 1988) whereas Kawasaki’s disease occurs almost exclusively in chil-
are negative either due to sampling error or prior treatment, sur- dren (Lane et al. 2005b).
rogate markers of vasculitis along with serology and imaging can A thorough history can help identify risk factors associated
support a diagnosis. Understanding the usefulness and limitations with the development of certain vasculitides, such as smoking or
of serologies, imaging studies, and biopsies is important in estab- exposure to second-hand smoke and thromboangiitis obliterans
lishing a correct diagnosis. To complicate matters further, there (Buerger’s disease), blood transfusions and HCV-associated
are numerous conditions that can mimic primary vasculitic syn- cryoglobulinaemia, or underlying diseases that can mimic vas-
dromes (Table 20.1, discussed further in Chapter 44). This chapter culitis such as atherosclerosis or malignancy. Information that
will focus on the approach to the diagnosis of vasculitis in adults should be specifically sought includes: illicit drug use; high-risk
276 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 20.1 Primary and secondary vasculitides sexual behaviour; prior thromboses; miscarriages; travel his-
tory; prior malignancies; operations; invasive procedures; den-
Primary Secondary and mimics tal work; medications; over the counter supplements or herbal
Large vessel involvement Infections preparations; occupation; hobbies; and medication and chemical
Giant cell arteritis Subacute bacterial endocarditis exposures.
Takayasu’s arteritis Syphilis Physical examination may provide clues to the presence of vas-
Behçet’s syndrome Hepatitis B culitis or one of its mimics. It can also help to delineate the extent
Medium and small vessel involvement Hepatitis C of clinical organ involvement and provide a framework for further
Polyarteritis nodosa Cytomegalovirus diagnostic testing. A thorough vascular exam, including palpation
Cutaneous polyarteritis Epstein–Barr virus of arterial pulses, auscultation for bruits, and blood pressure meas-
Granulomatosis with polyangiitis Human immunodeficiency virus urements in all four extremities, should be performed in all patients
Eosinophilic granulomatosis Meningococcaemia suspected of vasculitis. There are four forms of vasculitis that cause
with polyangiitis bruits: Takayasu’s arteritis, giant cell arteritis, Behçet’s syndrome,
Microscopic polyangiitis Tuberculosis and Cogan’s syndrome. Three forms of vasculitis lead to loss of a
Thromboangiitis obliterans Brucella
pulse:  Takayasu’s arteritis, giant cell arteritis, and thromboangii-
Cryoglobulinaemia Salmonella
tis obliterans (Stone et  al. 2001). Even in a patient who presents
Kawasaki’s disease Rocky Mountain spotted fever
with ‘only a rash,’ all systems must be examined to look for early
Behçet’s syndrome Medications
Primary angiitis of the CNS β-lactams
or occult involvement. As outlined in Table 20.2, any organ system
Cogan’s syndrome Sulfonamides can be involved and the clinical expression of vasculitis is diverse.
Predominately small vessel involvement Quinolones Many of these findings, when viewed in isolation, are non-specific
Cutaneous leukocytoclastic vasculitis Macrolides
Urticarial vasculitis Thiazides
Behçet’s syndrome Loop diuretics
IgA vasculitis Beta blockers Table 20.2 Physical examination findings that may suggest an
Phenytoin underlying vasculitis
Propylthiouracil
Selective serotonin reuptake Vital signs New-onset hypertension, lack of a pulse or blood pressure in
inhibitors an extremity, fever, unintentional weight loss
NSAIDs Head Scalp tenderness, swollen nodular and tender temporal artery,
Antitumour necrosis factor scalp necrosis
α inhibitors
Colony stimulating factors Eyes Retinal vasculitis, embolic phenomenon, episcleritis, scleritis,
(GM-CSF, G-GSF) dacrocystitis, visual field deficits, vision loss
Carbimazole Ears Chondritis, otitis media, hearing loss
Clopidogrel
Nose Friable mucosa, ulcerations, septal perforation, cobblestone
Montekulast
changes, swelling or collapse of the nasal bridge (saddle nose
Minocycline
deformity)
Drugs
Cocaine ± levamisole Oropharynx Mucosal ulcerations, hoarseness, stridor
Amphetamines Pulmonary Wheezing, crackles, dullness to percussion, pleural rub
Heroin
Connective tissue diseases Cardiac New or worsening heart murmur, decreased intensity of heart
Rheumatoid arthritis sounds, pericardial rub, irregular rhythm
Systemic lupus erythematosus Abdomen Tenderness, decreased bowel sounds, bruits, stool haemoccult
Sjögren’s syndrome positivity
Polymyositis
Genital Testicular tenderness, ulcerations on penis/scrotum or vulva/
Dermatomyositis
vagina
Other
Malignancy Vascular Bruits in the subclavian, carotid, axillary, brachial, abdominal,
Thrombotic thrombocy- femoral and/or popliteal arteries; absence of pulses in arms or
topenic purpura legs
Cardiac myxoma Skeletal Joint pain and/or synovitis
Cholesterol emboli syndrome
Atherosclerosis Neurological Muscle pain or weakness, single or multiple mononeuropathy,
Calciphylaxis polyneuropathy, mononeuritis multiplex, headaches, change in
Amyloidosis mental status, seizures, cranial neuropathy
Moyamoya disease Skin Petechial or purpuric rashes, cutaneous ulcerations, nodules,
Ehlers–Danlos syndrome digital infarcts, periungual erythema or infarcts, splinter
Fibromuscular dysplasia haemorrhages under fingernails, gangrene, erythema nodosa,
Antiphospholipid antibody pseudofolliculitis, livedo reticularis, Raynaud’s phenomenon,
syndrome urticaria (especially if it lasts ≥24 hours)
CHAPTER 20 approach to the diagnosis of vasculitis in adult patients 277

but, when interpreted in the context of the history and laboratory and systemic lupus erythematosus (Radice and Sinico 2005). If the
findings, can help to establish a diagnosis. For example, the find- ANCA immunofluorescence test is positive, then enzyme-linked
ing of new-onset hypertension is a frequent finding in the general immunosorbent assays (ELISA) testing for antibodies to serine
population, but when coupled with abdominal pain and monon- proteinase 3 (PR3) and myeloperoxidase (MPO) are necessary.
europathy multiplex, it suggests the possibility of polyarteritis These assays have a significantly higher positive predictive value
nodosa. Therefore, familiarity with the patterns of symptomatology than the immunofluorescence ANCA test (83% versus 45%). The
in vasculitic syndromes is essential. results of the ANCA test and anti-PR3/MPO antibodies should
be interpreted together to increase their usefulness. If the combi-
nation of tests (cANCA and anti-PR3 antibodies; or pANCA and
Laboratory testing anti-MPO antibodies) are positive, this is highly suggestive of GPA
Initially, basic laboratory tests consisting of a complete blood and MPA, respectively (Stone et al. 2000); however, between 10 and
count, serum creatinine, transaminases (ALT, AST), urinalysis, 20% of patients with classical GPA will have pANCA and anti-MPO
erythrocyte sedimentation rate (ESR), and C-reactive protein antibodies, and some patients with MPA or EGPA have cANCA
(CRP) should be obtained. Normochromic, normocytic anaemia positivity. A major caveat is that patients with cocaine-associated
is common in vasculitic syndromes. In addition, thrombocytosis vasculitis, and those exposed to levamisole (a veterinary antihel-
is frequently seen and is reflective of the acute phase response. In minthic medication used to process cocaine) may develop pANCA
contrast, thrombocytopenia and leukopenia are not expected in positivity by immunofluorescence at unusually high titres with
primary vasculitic syndromes and would suggest an alternate diag- low-titre anti-MPO antibodies, anti-PR3 antibodies, or antibod-
nosis. Eosinophilia may be observed in several of the vasculitic syn- ies to human neutrophil elastase, lactoferrin, or cathepsin G (see
dromes as well as vasculitis mimics. The presence of haematuria Chapter 44) (Graf et al. 2011). To further complicate the useful-
(with dysmorphic red blood cells on microscopic examination) or ness of the ANCA test, 10–50% of patients with biopsy-proven
proteinuria can indicate glomerular involvement prior to a rise in vasculitis will not have any positive serologies (Radice and Sinico
serum creatinine. Microscopic examination of the urine should be 2005). A study has shown that almost 10% of patients with both
performed on a fresh urine specimen to increase the potential to cANCA and anti-PR3 antibodies had diagnoses other than vas-
observe cellular casts, which degrade over time. Elevated transami- culitis, such as malignancy, infections, or other autoimmune dis-
nases may indicate liver involvement and would lead to supplemen- eases (McAdoo et  al. 2012). These findings may be due, in part,
tal testing for hepatitis B virus and HCV infection, both of which to increased use of ANCA testing in patient populations with low
can be associated with polyarteritis nodosa or cryoglobulinaemia. clinical suspicion of vasculitis, and/or recent changes to detection
Hepatitis B surface antigen can be seen in up to 10% of patients methods. Nonetheless, ANCA testing should not take the place
with polyarteritis nodosa; however, elevated transaminases are not of tissue confirmation of a diagnosis of GPA or MPA unless the
specific to liver involvement and can also be seen in muscle inflam- clinical findings are overwhelmingly classical and other causes have
mation or infarction and haemolysis. Creatine kinase and aldolase been thoroughly excluded.
should be checked if muscle involvement is suspected or if elevated Complement levels are useful in the differential diagnosis of glo-
transaminases are not felt to be related to liver inflammation. An merulonephritis; C3 and C4 levels are usually decreased in lupus
elevated ESR is frequently observed in vasculitis, but a normal nephritis, cryoglobulinaemia, or endocarditis but are typically nor-
or low ESR does not rule out its diagnosis. Up to 24% of patients mal in many vasculitic syndromes such as GPA or MPA (Hebert
with giant cell arteritis have a normal ESR (Salvarani and Hunder et al. 1991). Antiglomerular basement membrane (GBM) antibod-
2001; Weyand et al. 2000; Zweegman et al. 1993). Studies have sug- ies should be obtained in the evaluation of alveolar haemorrhage,
gested that C-reactive protein (CRP) is a more sensitive marker of normocomplementaemic glomerulonephritis, or a pulmonary–
a positive temporal artery biopsy than ESR, therefore both should renal syndrome (Herody et al. 1993).
be obtained (Walvick and Walvick 2011; Kermani et  al. 2012). Antinuclear antibodies (ANA) and rheumatoid factor (RF)
Nonetheless, an elevated ESR and/or CRP is non-diagnostic, also are also not useful screening tests for vasculitis because they can
being observed in malignancies and infections. be observed in any of the vasculitic syndromes and many of its
There are no specific laboratory tests to diagnose vasculi- mimics. The ANA should only be ordered if the diagnosis of sys-
tis. Neither antineutrophil cytoplasmic antibodies (ANCA) nor temic lupus erythematosus is being entertained based on history
complement levels should be used as screening tests. Proposed and physical examination findings. Similarly, the RF should only
indications for ANCA testing by indirect immunofluorescence be obtained if an underlying diagnosis of rheumatoid arthritis is
include: the presence of glomerulonephritis (especially if rapidly suspected based on symptoms and physical examination. The RF
progressive); pulmonary haemorrhage; cutaneous vasculitis with can be significantly elevated in patients with cryoglobulinaemia,
systemic features; multiple lung nodules; chronic destructive dis- Sjögren’s syndrome, and subacute bacterial endocarditis (Dorner
ease of the upper airways; long-standing sinusitis or otitis; subglot- et al. 2004).
tic or tracheal stenosis; peripheral neuropathy; and retro-orbital Laboratory tests are useful in excluding some of the second-
mass (Hagen et al. 1998; Mandl et al. 2002). A cytoplasmic stain- ary causes of vasculitis or its mimics. At least three sets of blood
ing pattern (cANCA) is seen most commonly in generalized GPA cultures should be obtained in any febrile patient with multiorgan
and the perinuclear staining (pANCA) is seen most commonly in system involvement to evaluate for endocarditis. If the clinical sus-
microscopic polyangiitis (MPA). picion for endocarditis is high, then echocardiography should also
Taken in isolation, a positive ANCA test does not establish a be performed, despite negative blood cultures. In the appropriate
diagnosis of vasculitis. ANCAs can be seen in a diverse spectrum clinical setting, specific serological tests for organisms can be help-
of conditions, including inflammatory bowel disease, infections, ful, such as tests for syphilis in the evaluation of aortitis.
278 SECTION 5 vasculitic diseases and syndromes and related disorders

Diagnostic tests to confirm In a patient with suspected pulmonary involvement, a radio-


graph of the chest can be informative if it demonstrates nodules,
suspicions of vasculitis opacities, or infiltrates; however, if it is normal, it does not exclude
After initial evaluation by history, physical examination, and basic a pulmonary process. High-resolution CT (HRCT) scanning of the
laboratory testing, the distribution of organ involvement should chest is much more sensitive, unmasking pulmonary abnormalities
suggest a differential diagnosis. However, this process is not static; in patients with GPA whose chest radiographs are normal (Reuter
new clinical manifestations can develop at any time and may change et al. 1998) (see Chapters 18 and 30). HRCT can help to make the
the diagnostic pathway. Secondary testing (Table  20.3) should distinction between active inflammation (‘ground-glass changes’)
focus on providing supportive evidence for a vasculitic process and and fibrosis. However, it cannot accurately make the distinction
excluding mimics. For example, in a patient presenting with promi- between infiltrates due to alveolar bleeding from vasculitis, infec-
nent symptoms of peripheral neuropathy or myopathy, electrodiag- tion, or medication toxicity. Biopsy of focal lesions in the lung
nostic tests (electromyography and nerve conduction tests) should should be pursued.
be obtained. If a neuropathy is documented, the distinctions among
infection, toxin, malignancy, metabolic, and inflammatory aetiolo- Tissue biopsy
gies need to be considered. If vasculitis remains in the differential,
then a nerve/ muscle biopsy would be considered if an accessible Ideally, tissue biopsy should be performed prior to treatment ini-
nerve is affected (see Chapter 13). Demonstration of vasculitis on tiation; however, if there is critical organ involvement, treatment
tissue biopsy is preferred to establish the diagnosis. If tissue cannot should never be delayed for diagnostic purposes. The location
be obtained (or is unrevealing) then directed imaging is indicated. of a biopsy is determined by suspicion for clinical involvement
To delineate areas of suspected organ involvement, imaging stud- and accessibility of tissue; for example, biopsy of one of the aor-
ies such as CT or MRI are frequently obtained (see Chapter 18). tic branches would not be feasible and therefore an imaging study
In addition, they can be used to direct the site of tissue biopsy would be utilized in place of biopsy. If several organ systems are
for focal lesions. MRI or CT is used for evaluation of ears, eyes, involved, choice of biopsy site is determined by the morbidity of the
nose, and throat involvement in a suspected vasculitic syndrome. procedure and the likelihood of obtaining disease-specific infor-
MRI can delineate mucosal inflammation, air–fluid levels, cavitary mation. Whereas a skin biopsy demonstrating leukocytoclastic
lesions, and retro-orbital inflammatory or fibrotic masses. CT scans vasculitis confirms the presence of vasculitis, it does not narrow
can demonstrate mucosal thickening, air–fluid levels, sclerosing down the differential diagnosis with respect to the specific type of
osteitis, bone thickening, and bone destruction (Lohrmann et al. systemic vasculitis present, such as polyarteritis nodosa, GPA, etc.
2006). In a patient with central nervous system involvement, MRI In contrast, a lung biopsy showing vasculitis, geographic necrosis,
of the brain with gadolinium can demonstrate areas of ischaemia, and palisading granulomas would suggest GPA (Travis et al. 1991).
infarcts, mass lesions, and meningeal enhancement. MR imaging of When biopsies reveal classic vasculitic changes, they are very
the brain is abnormal in 90% of patients with histologically proven informative; however, a normal biopsy does not exclude a diagno-
angiitis of the central nervous system (Calabrese et al. 1997), but a sis of vasculitis. Factors that can influence biopsy positivity include
normal MRI does not rule out central nervous system vasculitis. inadequate tissue sample and prior immunosuppressive treatment.
Further evaluation of the central nervous system includes a lumbar Certain vasculitides, such as giant cell arteritis, are characterized
puncture, cerebral angiogram, and brain biopsy. In the setting of a by patchy vascular inflammation, which results in normal areas of
normal MRI and normal cerebrospinal fluid, central nervous sys- vessel wall scattered between areas of vasculitis (skip lesions); if
tem vasculitis is rare (Calabrese et al. 1997). insufficient tissue is obtained, the vasculitis can be missed (Albert
et  al. 1976). Blind tissue biopsies (i.e. sampling of tissue that is
not clinically involved) have a low diagnostic yield for vasculitis
Table 20.3 Diagnostic tests used in evaluation of suspected vasculitis
(19% for nerve and 29% for muscle) (Albert et al. 1988). The dem-
onstration of vasculitis in a tissue specimen is not a final diagnosis.
Chest radiograph and high-resolution CT scan
It should be viewed as a sign of an underlying process, which must
Sinus radiograph and CT scan be interpreted in the context of the clinical, serological, and imag-
Orbital CT scan or MRI ing studies to establish a final diagnosis.
Electromyography and nerve conduction tests
Temporal artery biopsy
Echocardiography
In a patient presenting with symptoms suggestive of giant cell arteri-
Ultrasound of temporal arteries tis, the diagnostic procedure of choice is the temporal artery biopsy.
Brain MRI with angiogram However, giant cell arteritis can sometimes present with vague signs
and symptoms such as fevers, myalgias, and anaemia in the absence
Conventional angiography
of headaches, jaw claudication, etc. (Calamia and Hunder 1981;
MRI angiogram of aorta and branches Gonzalez-Gay et al. 2005). In patients over the age of 65 years, giant
CT Angiography cell arteritis is the aetiology for 16% of fevers of unknown origin
18 FGD-PET/CT imaging
(FUO) (Esposito and Gleckman 1978). Therefore, a temporal artery
biopsy may also be considered in the evaluation of FUOs in this older
Tissue biopsy: skin, nerve, muscle, lung, kidney, brain, temporal artery population (after malignancy and infections have been excluded).
CT, computed tomography; MRI, magnetic resonance imaging; FGD-PET, The decision regarding whether to perform unilateral or bilateral
fluorodeoxyglucose positron emission testing. temporal artery biopsy is controversial. Some advocate unilateral
CHAPTER 20 approach to the diagnosis of vasculitis in adult patients 279

biopsy with a contralateral biopsy if the initial biopsy is negative; and is completely non-diagnostic. Vasculitis and granulomas are
others perform routine bilateral biopsies in all patients. To increase very rarely demonstrated; therefore, it is of limited diagnostic value
the diagnostic yield of the biopsy, the artery specimen should be at for vasculitis.
least 1 cm (Ypsilantis et al. 2011). In specimens less than 0.5 cm in
length, the rate of positive findings is 19% versus 89% in specimens Renal biopsy
more than 2 cm in length (Breuer et al. 2009). Findings that predict In patients with multisystem disease and a rising creatinine or
a high probability of a positive temporal artery biopsy include jaw in the setting of proteinuria of unexplained aetiology or cellu-
claudication, new headaches, eye symptoms, and tender temporal lar casts, a renal biopsy should be considered for diagnostic and
artery (Smetana and Shmerling 2002) and elevated CRP (Walvick prognostic purposes (see Chapter  15). Standard stains (haema-
and Walvick 2011; Kermani et  al. 2012). Nevertheless, approxi- toxylin and eosin, silver, trichrome), immunofluorescence, and
mately 10% of patients with the clinical diagnosis of giant cell arte- electron microscopy should be performed on all specimens. Most
ritis have normal biopsies (Hall et al. 1983). commonly, the renal biopsy reveals glomerular disease, for exam-
ple focal segmental necrotizing glomerulonephritis or crescentic
Skin biopsy glomerulonephritis. True necrotizing arteritis or granulomas are
In patients with cutaneous involvement, a skin biopsy is easy to rarely seen on renal biopsy. In a patient with symptoms suggestive
obtain and has low morbidity. A skin biopsy can easily demonstrate of generalized GPA, the biopsy site of choice would be lung rather
small-vessel vasculitis. In addition, direct immunofluorescence of than kidney because there is a higher likelihood of finding vasculi-
skin can reveal IgA deposits in patients with IgA vasculitis (IgAV; tis and granulomas in the lung.
formerly known as Henoch–Schönlein purpura) (Van Hale et al.
1986) or prominent IgG deposition around blood vessels and the Vascular imaging studies
basement membrane zone in hypocomplementaemic urticarial
vasculitis (Mehregan et al. 1992). However, skin biopsies typically Angiography
provide a general diagnosis of inflammatory vascular disease rather Traditionally, angiography has been the diagnostic procedure of
than a precise disease entity. choice for detecting arterial stenoses, occlusions, and aneurysms in
large and medium-sized vessels within the central nervous system,
Lung biopsy chest, and abdomen (see Chapter  17). In addition, angiography
When lower respiratory tract abnormalities are present, obtaining may be used when biopsies are not feasible or are unrevealing. The
lung tissue is often essential. Transbronchial biopsy has a low diag- presence of multiple saccular aneurysms in more than one organ is
nostic yield for demonstrating vasculitis (Schnabel et al. 1997) but highly suggestive of vasculitis. However, a multitude of other con-
is useful in confirming the presence of pulmonary haemorrhage ditions can mimic angiographic vasculitic changes including: ath-
and evaluating for infectious or malignant aetiologies. Open lung erosclerosis; amyloidosis; vasospasm; infection; fibromuscular
biopsy is highly sensitive and specific for confirming a diagnosis dysplasia; Ehlers–Danlos syndrome; atrial myxoma; phaeochro-
of GPA (Hoffman et al. 1992; Mark et al. 1988), but there is sub- mocytoma; use of sympathomimetic drugs; and malignancy (see
stantial morbidity and mortality associated with the procedure. Chapter 44). Therefore, the results of the imaging study must be
Video-assisted thoracic surgery (VATS) for obtaining lung tissue interpreted in the context of the clinical setting. The specificity of
may provide an excellent alternative to open lung biopsy due to its brain angiography in differentiating vasculitis mimics from histo-
low morbidity and mortality (Ooi et al. 2005). logically defined angiitis of the central nervous system is less than
30% (Duna and Calabrese 1995). Several other caveats exist when
Nerve and muscle biopsy interpreting angiographic results. Angiography does not have ade-
In patients suspected of having nerve involvement, a nerve biopsy quate resolution for small vessels; therefore, conditions character-
should be considered (see Chapter 13). The yield of a nerve biopsy ized predominantly by small-vessel inflammation, such as IgAV,
is higher with site selection guided by clinical and electrodiagnostic may be overlooked. In addition, aneurysms take time to develop
findings. The diagnostic yield from a nerve and muscle biopsy that and consequently they may not be demonstrated when the angio-
is not clinically involved is only 19% and 29%, respectively (Albert gram is performed early in the disease course. Similarly, traditional
et al. 1988). Nevertheless, despite abnormal electrodiagnostic find- angiography only visualizes the lumen of the vessel; thickening of
ings, up to 45% of nerve biopsies can reveal no diagnostic pathol- the vessel wall, which may occur prior to the development of sten-
ogy (Pioro and Calabrese 1995; Rappaport et al. 1993). Biopsy of oses or aneurysms, would not be demonstrated on routine angi-
skeletal muscle along with nerve biopsy can increase the diagnostic ography. Limiting factors for performing angiography include the
yield for vasculitis (Collins et al. 2000; Stone et al. 2001). The sural presence of compromised renal function, which could be worsened
and superficial peroneal nerves are the most commonly biopsied following infusion of contrast dye, as well as risk for ischaemic
nerves, but the radial sensory nerve is also accessible to biopsy. complications. In patients with Behçet’s syndrome, angiography is
The morbidity associated with nerve biopsy is not trivial; infection, not recommended due to increased risk for aneurysm formation at
non-healing ulcers, and chronic pain at the incision site have been arterial puncture sites (Kingston et al. 1979).
observed (Gabriel et al. 2000; Rappaport et al. 1993).
Magnetic resonance imaging
Nasal or sinus biopsy For the evaluation of the aorta and its primary branches, magnetic
In patients with symptoms suggestive of GPA or EGPA, a sample of resonance imaging (MRI) is being performed more commonly now
nasal or sinus tissue can be examined although the diagnostic yield in place of traditional angiography (see Chapter 18). There is no
is very low. Acute and chronic inflammation is frequently observed risk of ionizing radiation or iodinated contrast material; therefore,
280 SECTION 5 vasculitic diseases and syndromes and related disorders

it can be used for serial imaging. It provides visualization of the ves- with increased uptake in and around hip and shoulder joints, ischial
sel lumen and also demonstrates vessel wall oedema and thickness tuberosities, and lumbar spinous processes; asymptomatic large ves-
(Flamm et al. 1998). Although MRI is felt to have excellent sensitiv- sel involvement may also be seen (Blockmans 2011; Yamashita et al.
ity and specificity for large-vessel vasculitis (Yamada et al. 2000) it is 2012). In addition, PET/ CT imaging may be useful in the diagno-
not without limitations. It may overestimate vascular occlusions, it sis and follow-up of the upper respiratory tract and lung lesions in
does not image small branch vessels, and vascular calcifications are patients with GPA (Ito et al. 2013) and vascular lesions in Behçet’s
poorly visualized (Yamada et al. 2000). In addition, disease activity syndrome (Trad et al. 2013); however, additional studies are needed
as measured by vessel wall oedema on MRI does not consistently to confirm these findings.
correlate with symptoms, acute phase reactants, or occurrence of
new anatomic changes on subsequent exams (Tso et al. 2002). The Conclusions
incorporation of angiography into MR imaging allows for more
detailed vascular characterization (Narvaez et al. 2005). Diagnosis of vasculitis is challenging. It first requires familiarity
with the clinical features of the different syndromes and narrowing
Computed tomography down the differential diagnosis based on the patient’s history and
Computed tomography (CT) scanning can provide details regard- physical examination. The next requirement is selection of appro-
ing the degree and nature of vessel wall thickening, for example priate tests to delineate areas of disease involvement with focus on
calcification and extent of disease (see Chapter 18). The addition demonstrating vasculitis, either through tissue biopsy or imaging
of three-dimensional reconstruction techniques contributes studies. Choice of biopsy site is determined by the morbidity of the
angiogram-like images, highlighting areas of stenosis, occlusion, procedure and the likelihood of obtaining disease-specific informa-
and aneurysmal dilation. tion. The type of imaging study is chosen based on size and location
of vessels involved. At the same time, secondary causes of vascu-
Ultrasound litis and mimics of vasculitis must be excluded. Because systemic
vasculitis can be life-threatening if untreated, a clinician oftentimes
Colour duplex ultrasonography provides better resolution than MRI
must institute several treatments concurrently, directed towards the
or CT scan for medium-sized peripheral arteries such as temporal,
suspected vasculitis as well as its mimics, while the investigation is
carotid, axillary, and femoral arteries (see Chapter 18). Blood flow,
in progress. If the clinical presentation is atypical, and tissue biopsy
wall thickening, and plaques can be visualized. Findings sugges-
or imaging is not obtained early in the disease course, a definitive
tive of vasculitis include hypoechoic dark wall swelling (halo sign),
diagnosis may never be established, leading to confusion about
increased flow velocity due to stenosis, and acute occlusion (Schmidt
long-term treatment and prognosis.
et al. 1997; Schmidt et al. 2003; Nesher et al. 2002). These findings
reflect inflamed arterial walls and are not specific for giant cell arteri- References
tis. In a meta-analysis of studies using ultrasonography for giant cell Albert, D.A., Rimon, D., and Silverstein, M.D. (1988). The diagnosis of
arteritis, the sensitivity of the halo sign was 69% and specificity was polyarteritis nodosa. I. A literature-based decision analysis approach.
82% compared with temporal artery biopsy (Karassa et al. 2005). Arthritis and Rheumatism, 31, 1117–27.
Rarely, other entities such as polyarteritis nodosa, GPA, EGPA, or Albert, D.M., Ruchman, M.C., and Keltner, J.L. (1976). Skip areas in temporal
amyloidosis can cause temporal artery inflammation. Ultrasound arteritis. Archives of Ophthalmology, 94, 2072–7.
is most effective in diagnosing new cases of giant cell arteritis; it is Arend, W.P., Michel, B.A., Bloch, D.A., Hunder, G.G., Calabrese, L.H.,
much less effective in detecting cases of recurrent disease. Edworthy, S. M., Fauci, A.S., Leavitt, R.Y., Lie, J.T., Lightfoot, R.W., Jr.,
et al. (1990). The American College of Rheumatology 1990 criteria for
PET Scanning the classification of Takayasu arteritis. Arthritis and Rheumatism, 33,
1129–34.
Positron emission tomography (PET) is a non-invasive, meta- Belhocine, T., Blockmans, D., Hustinx, R., Vandevivere, J., and Mortelmans,
bolic imaging modality, based on the regional distribution of 18 L. (2003). Imaging of large vessel vasculitis with (18).FDG PET: illusion
F-fluorodeoxyglucose, which is used extensively in the management or reality? A critical review of the literature data. European Journal of
of oncology patients. It is emerging as a useful tool in the diagno- Nuclear Medicine and Molecular Imaging, 30, 1305–13.
sis and follow up of large-vessel vasculitis (Blockmans 2011; Walter Bloch, D.A., Michel, B.A., Hunder, G.G., McShane, D.J., Arend, W.P.,
et al. 2005; Fuchs et al. 2012) (see Chapter 19). PET scanning allows Calabrese, L.H., Edworthy, S.M., Fauci, A.S., Fries, J.F., and Leavitt,
R.Y. (1990). The American College of Rheumatology 1990 criteria
visualization of the entire body in a single image and can dem-
for the classification of vasculitis. Patients and methods. Arthritis and
onstrate the distribution of vascular involvement. The PET signal Rheumatism, 33, 1068–73.
is weak for small arteries such as temporal arteries but strong for Blockmans, D. (2011). PET in vasculitis. Annals of New  York Academy of
branches extending from the aorta. It may be useful for assessing Sciences, 1228, 64–70.
disease activity and response to treatment in patients with giant cell Blockmans, D., De Ceuninck, L., Vanderschueren, S., Knockaert, D., Mortelmans,
arteritis and Takayasu’s arteritis (Blockmans et al. 2000; Blockmans L., and Bobbaers, H. (2006). Repetitive 18F-fluorodeoxyglucose positron
et al. 2006; Lee et al. 2012; Fuchs et al. 2012; Tezuka et al. 2012). emission tomography in giant cell arteritis:  a prospective study of 35
Additional investigation is needed to establish the sensitivity and patients. Arthritis and Rheumatism, 55, 131–7.
specificity of PET scanning in large-vessel vasculitis (Belhocine Blockmans, D., Stroobants, S., Maes, A., and Mortelmans, L. (2000). Positron
emission tomography in giant cell arteritis and polymyalgia rheumat-
et  al. 2003). An important application of the PET scan is in the
ica: evidence for inflammation of the aortic arch. American Journal of
evaluation of atypical presentations of vasculitis (Blockmans et al. Medicine, 108, 246–9.
2006), fever of unknown origin, and elevated acute phase reactants Breuer, G.S., Nesher, R., and Nesher, G. (2009). Effect of biopsy length on
(Lensen et al. 2013). PET imaging also demonstrates a unique dis- the rate of positive temporal artery biopsies. Clinical and Experimental
tribution of involvement in patients with polymyalgia rheumatica Rheumatology, 27 (1 Suppl. 52), S10–3.
CHAPTER 20 approach to the diagnosis of vasculitis in adult patients 281

Calabrese, L.H., Duna, G.F., and Lie, J.T. (1997). Vasculitis in the central American College of Rheumatology 1990 criteria for the classification of
nervous system. Arthritis and Rheumatism, 40, 1189–201. giant cell arteritis. Arthritis and Rheumatism, 33, 1122–8.
Calabrese, L.H., Michel, B.A., Bloch, D.A., Arend, W.P., Edworthy, S.M., Ito, K. Minamimoto, R., Yamashita, H., Yoshida, S., Morooka, M., Okasaki,
Fauci, A.S., Fries, J.F., Hunder, G.G., Leavitt, R.Y., Lie, J.T., et al. (1990). M., Mimori, A., and Kubota, K. (2013). Evaluation of Wegener’s granu-
The American College of Rheumatology 1990 criteria for the classifi- lomatosis using 18F-fluorodeoxyglucose positron emission tomogra-
cation of hypersensitivity vasculitis. Arthritis and Rheumatism, 33, phy/computed tomography. Annals of Nuclear Medicine, 27, 209–16.
1108–13. Jennette, J.C., Falk, R.J., Andrassy, K., Bacon, P.A., Churg, J., Gross, W.L.,
Calamia, K.T. and Hunder, G.G. (1981). Giant cell arteritis (temporal Hagen, E.C., Hoffman, G.S., Hunder, G.G., Kallenberg, C.G., et  al.
arteritis) presenting as fever of undetermined origin. Arthritis and (1994). Nomenclature of systemic vasculitides. Proposal of an interna-
Rheumatism, 24, 1414–18. tional consensus conference. Arthritis and Rheumatism, 37, 187–92.
Collins, M.P., Mendell, J.R., Periquet, M.I., Sahenk, Z., Amato, A.A., Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F.,
Gronseth, G.S., Barohn, R.J., Jackson, C.E., and Kissel, J.T. (2000). Flores-Suarez, L.F., Gross, W.L., Guillevin, L., Hagen, E.C., Hoffman,
Superficial peroneal nerve/peroneus brevis muscle biopsy in vasculitic G.S., Jayne, D.R., Kallenberg, C.G., Lamprecht, P., Langford, C.A.,
neuropathy. Neurology, 55, 636–43. Luqmani, R.A., Mahr, A.D., Matteson, E.L., Merkel, P.A., Ozen, S.,
Dorner, T., Egerer, K., Feist, E., and Burmester, G.R. (2004). Rheumatoid fac- Pusey, C.D., Rasmussen, N., Rees, A.J., Scott, D.G., Specks, U., Stone,
tor revisited. Current Opinion in Rheumatology, 16, 246–53. J.H., Takahashi, K., and Watts, R.A. (2013). 2012 revised International
Duna, G F. and Calabrese, L.H. (1995). Limitations of invasive modalities in Chapel Hill Consensus Conference Nomenclature of Vasculitides.
the diagnosis of primary angiitis of the central nervous system. Journal Arthritis and Rheumatism, 65, 1–11.
of Rheumatology, 22, 662–7. Karassa, F.B., Matsagas, M.I., Schmidt, W.A., and Ioannidis, J.P. (2005).
Esposito, A.L. and Gleckman, R.A. (1978). Fever of unknown origin in the Meta-analysis: test performance of ultrasonography for giant-cell arte-
elderly. Journal of the American Geriatrics Society, 26, 498–505. ritis. Annals of Internal Medicine, 142, 359–69.
Flamm, S.D., White, R.D., and Hoffman, G.S. (1998). The clinical application Kermani, T.A., Schmidt, J., Crowson, C.S., Ytterberg, S.R., Hunder, G.G.,
of ‘edema-weighted’ magnetic resonance imaging in the assessment of Matteson, E.L., and Warrington, K.J. (2012). Utility of erythrocyte sedi-
Takayasu’s arteritis. International Journal of Cardiology, 66 (Suppl. 1), mentation rate and C-reactive protein for the diagnosis of giant cell arte-
S151–9. ritis. Seminars in Arthritis and Rheumatism, 41, 866–71.
Fuchs, M., Briel, M., Daikeler, T., Walker, U.A., Rasch, H., Berg, S., Ng, Q.K., Kingston, M., Ratcliffe, J.R., Alltree, M., and Merendino, K.A. (1979).
Raatz, H., Jayne, D., Kötter, I., Blockmans, D., Cid, M.C., Prieto-González, Aneurysm after arterial puncture in Behçet’s disease. British Medical
S., Lamprecht, P., Salvarani, C., Karageorgaki, Z., Watts, R., Luqmani, Journal, 1, 1766–7.
R., Müller-Brand, J., Tyndall, A., and Walter, M.A. (2012). The impact Lane, S.E., Watts, R., and Scott, D.G. (2005a). Epidemiology of systemic vas-
of 18F-FDG PET on the management of patients with suspected large culitis. Current Rheumatology Reports, 7, 270–5.
vessel vasculitis. European Journal of Nuclear Medicine and Molecular Lane, S.E., Watts, R.A., Shepstone, L., and Scott, D.G. (2005b). Primary sys-
Imaging, 39, 344–53 temic vasculitis:  clinical features and mortality. Quarterly Journal of
Gabriel, C.M., Howard, R., Kinsella, N., Lucas, S., McColl, I., Saldanha, G., Medicine, 98, 97–111.
Hall, S.M., and Hughes, R.A. (2000). Prospective study of the usefulness Leavitt, R.Y., Fauci, A.S., Bloch, D.A., Michel, B.A., Hunder, G.G., Arend, W.P.,
of sural nerve biopsy. Journal of Neurology Neurosurgery and Psychiatry, Calabrese, L.H., Fries, J.F., Lie, J.T., Lightfoot, R.W. Jr, et al. (1990). The
69, 442–6. American College of Rheumatology 1990 criteria for the classification of
Gallagher, H., Kwan, J.T., and Jayne, D.R. (2002). Pulmonary renal syn- Wegener’s granulomatosis. Arthritis and Rheumatism, 33, 1101–7.
drome: a 4-year, single-center experience. American Journal of Kidney Lee, K., Cho, A., Choi, Y., Lee, S.W., Ha, Y.J., Jung, S.J., Park, M.C., Lee, J.D., Lee,
Diseases, 39, 42–7. S.K., and Park, Y.B. (2012). The role of 18F-fluorodeoxyglucose-positron
Gonzalez-Gay, M.A., Lopez-Diaz, M.J., Barros, S., Garcia-Porrua, C., emission tomography in the assessment of disease activity in patients
Sanchez-Andrade, A., Paz-Carreira, J., Martin, J., and Llorca, J. (2005). with Takayasu arteritis. Arthritis and Rheumatism, 64, 866–75.
Giant cell arteritis: laboratory tests at the time of diagnosis in a series of Lensen, K.J., Voskuyl, A.E., van der Laken, C.J., Comans, E.F., van
240 patients. Medicine (Baltimore), 84, 277–90. Schaardenburg, D., Arntzenius, A.B., Zwijnenburg, T., Stam, F.,
Graf, J., Lynch, K., Yeh, C.L., Tarter, L., Richman, N., Nguyen, T., Kral, A., Gompelman, M., Zant, F.M., van Paassen, A.Q., Voerman, B.J, Smit,
Dominy, S., and Imboden, J. (2011). Purpura, cutaneous necrosis, and anti- F., Anten, S., Siegert, C.E., Binnerts, A., and Smulders, Y.M. (2013).
neutrophil cytoplasmic antibodies associated with levamisole-adulterated 18F-Fluorodeoxyglucose positron emission tomography in elderly
cocaine. Arthritis and Rheumatism, 63, 3998–4001 patients with an elevated erythrocyte sedimentation rate of unknown
Hagen, E.C., Daha, M.R., Hermans, J., Andrassy, K., Csernok, E., Gaskin, G., origin. PLoS One, 8, e58917.
Lesavre, P., Ludemann, J., Rasmussen, N., Sinico, R.A., Wiik, A., and Lightfoot, R.W., Jr., Michel, B.A., Bloch, D.A., Hunder, G.G., Zvaifler, N.J.,
van der Woude, F.J. (1998). Diagnostic value of standardized assays for McShane, D.J., Arend, W.P., Calabrese, L.H., Leavitt, R.Y., Lie, J.T., et al.
anti-neutrophil cytoplasmic antibodies in idiopathic systemic vasculitis. (1990). The American College of Rheumatology 1990 criteria for the
EC/BCR Project for ANCA Assay Standardization. Kidney International, classification of polyarteritis nodosa. Arthritis and Rheumatism, 33,
53, 743–53. 1088–93.
Hall, S., Persellin, S., Lie, J.T., O’Brien, P.C., Kurland, L.T., and Hunder, G.G. Lohrmann, C., Uhl, M., Warnatz, K., Kotter, E., Ghanem, N., and Langer,
(1983). The therapeutic impact of temporal artery biopsy. Lancet, 2, M. (2006). Sinonasal computed tomography in patients with Wegener’s
1217–20. granulomatosis. Journal of Computer Assisted Tomography, 30, 122–5.
Hebert, L.A., Cosio, F.G., and Neff, J.C. (1991). Diagnostic significance of Machado, E.B., Michet, C.J., Ballard, D.J., Hunder, G.G., Beard, C.M., Chu,
hypocomplementemia. Kidney International, 39, 811–21. C.P., and O’Fallon, W.M. (1988). Trends in incidence and clinical pres-
Herody, M., Bobrie, G., Gouarin, C., Grunfeld, J.P., and Noel, L.H. (1993). entation of temporal arteritis in Olmsted County, Minnesota, 1950–
Anti-GBM disease: predictive value of clinical, histological and serologi- 1985. Arthritis and Rheumatism, 31, 745–9.
cal data. Clinical Nephrology, 40, 249–55. Mandl, L.A., Solomon, D.H., Smith, E.L., Lew, R.A., Katz, J.N., and Shmerling,
Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., Hallahan, C.W., Lebovics, R.S., Travis, R.H. (2002). Using antineutrophil cytoplasmic antibody testing to diag-
W.D., Rottem, M., and Fauci, A.S. (1992). Wegener granulomatosis: an nose vasculitis: can test-ordering guidelines improve diagnostic accu-
analysis of 158 patients. Annals of Internal Medicine, 116, 488–98. racy? Archives of Internal Medicine, 162, 1509–14.
Hunder, G.G., Bloch, D.A., Michel, B.A., Stevens, M.B., Arend, W.P., Calabrese, Mark, E.J., Matsubara, O., Tan-Liu, N.S., and Fienberg, R. (1988).
L.H., Edworthy, S.M., Fauci, A.S., Leavitt, R.Y., Lie, J.T., et al. (1990). The The pulmonary biopsy in the early diagnosis of Wegener’s
282 SECTION 5 vasculitic diseases and syndromes and related disorders

(pathergic) granulomatosis:  a study based on 35 open lung biopsies. Schott, P., Hartmann, H., and Ramadori, G. (2001). Hepatitis C
Human Pathology, 19, 1065–71. virus-associated mixed cryoglobulinemia. Clinical manifestations, his-
Masi, A.T., Hunder, G.G., Lie, J.T., Michel, B.A., Bloch, D.A., Arend, W.P., topathological changes, mechanisms of cryoprecipitation and options of
Calabrese, L.H., Edworthy, S.M., Fauci, A.S., Leavitt, R.Y., et al. (1990). treatment. Histology and Histopathology, 16, 1275–85.
The American College of Rheumatology 1990 criteria for the classifica- Smetana, G.W. and Shmerling, R.H. (2002). Does this patient have temporal
tion of Churg-Strauss syndrome (allergic granulomatosis and angiitis). arteritis? Journal of the American Medical Association, 287, 92–101.
Arthritis and Rheumatism, 33, 1094–100. Sorensen, S.F., Slot, O., Tvede, N., and Petersen, J. (2000). A prospective study
McAdoo, S.P., Hall, A., Levy, J., Salama, A.D., and Pusey, C.D. (2012). of vasculitis patients collected in a five year period: evaluation of the
Proteinase-3 antineutrophil cytoplasm antibody positivity in patients Chapel Hill nomenclature. Annals of the Rheumatic Diseases, 59, 478–82.
without primary systemic vasculitis. Journal of Clinical Rheumatology, Stone, J.H., Calabrese, L.H., Hoffman, G.S., Pusey, C.D., Hunder, G.G., and
8, 336–40. Hellmann, D.B. (2001). Vasculitis. A  collection of pearls and myths.
Mehregan, D.R., Hall, M.J., and Gibson, L.E. (1992). Urticarial vasculitis: a Rheumatic Disease Clinics of North America, 27, 677–728.
histopathologic and clinical review of 72 cases. Journal of the American Stone, J.H., Talor, M., Stebbing, J., Uhlfelder, M.L., Rose, N.R., Carson, K.A.,
Academy of Dermatology, 26, 441–8. Hellmann, D.B., and Burek, C.L. (2000). Test characteristics of immuno-
Mills, J.A., Michel, B.A., Bloch, D.A., Calabrese, L.H., Hunder, G.G., Arend, fluorescence and ELISA tests in 856 consecutive patients with possible
W.P., Edworthy, S.M., Fauci, A.S., Leavitt, R.Y., Lie, J.T., et al. (1990). The ANCA-associated conditions. Arthritis Care Research, 13, 424–34.
American College of Rheumatology 1990 criteria for the classification Tezuka, D., Haraguchi, G., Ishihara, T., Ohigashi, H., Inagaki, H., Suzuki,
of Henoch-Schönlein purpura. Arthritis and Rheumatism, 33, 1114–21. J., Hirao, K., and Isobe, M. (2012). Role of FDG PET-CT in Takayasu
Murchison, A.P., Gilbert, M.E., Bilyk, J.R., Eagle, R.C. Jr, Pueyo, V., Sergott, Arteritis. Journal of the American College of Cardiology: Cardiovascular
R.C., and Savino, P.J. (2012). Validity of the American College of Imaging, 5, 422–9.
Rheumatology criteria for the diagnosis of giant cell arteritis. American Trad, S., Bensimhon, L., El Hajjam, M., Chinet, T., Wechsler, B., and Saadoun,
Journal of Ophthalmology, 154, 722–9. D. (2013). (18)F-fluorodeoxyglucose-positron emission tomography
Narvaez, J., Narvaez, J.A., Nolla, J.M., Sirvent, E., Reina, D., and Valverde, scanning is a useful tool for therapy evaluation of arterial aneurysm in
J. (2005). Giant cell arteritis and polymyalgia rheumatica: usefulness of Behçet’s disease. Joint Bone Spine, 80, 420–3.
vascular magnetic resonance imaging studies in the diagnosis of aortitis. Travis, W.D., Hoffman, G.S., Leavitt, R.Y., Pass, H.I., and Fauci, A.S. (1991).
Rheumatology (Oxford), 44, 479–83. Surgical pathology of the lung in Wegener’s granulomatosis. Review of
Nesher, G., Shemesh, D., Mates, M., Sonnenblick, M., and Abramowitz, H.B. 87 open lung biopsies from 67 patients. American Journal of Surgical
(2002). The predictive value of the halo sign in color Doppler ultra- Pathology, 15, 315–33.
sonography of the temporal arteries for diagnosing giant cell arteritis. Tso, E., Flamm, S.D., White, R.D., Schvartzman, P.R., Mascha, E., and
Journal of Rheumatology, 29, 1224–6. Hoffman, G.S. (2002). Takayasu arteritis:  utility and limitations of
Ooi, A., Iyenger, S., Ferguson, J., and Ritchie, A.J. (2005). VATS lung biopsy in magnetic resonance imaging in diagnosis and treatment. Arthritis and
suspected, diffuse interstitial lung disease provides diagnosis, and alters Rheumatism, 46, 1634–42.
management strategies. Heart Lung Circulation, 14, 90–2. Van Hale, H.M., Gibson, L.E., and Schroeter, A.L. (1986). Henoch-Schönlein
Park, M.C., Lee, S.W., Park, Y.B., Chung, N.S., and Lee, S.K. (2005). Clinical vasculitis: direct immunofluorescence study of uninvolved skin. Journal
characteristics and outcomes of Takayasu’s arteritis:  analysis of 108 of the American Academy of Dermatology, 15, 665–70.
patients using standardized criteria for diagnosis, activity assessment, Waller R., Ahmed A., Patel I., and Luqmani R. (2013). Update on the classifica-
and angiographic classification. Scandinavian Journal of Rheumatology, tion of vasculitis. Best Practice and Research Clinical Rheumatology, 27, 3–17.
34, 284–92. Walter, M.A., Melzer, R.A., Schindler, C., Muller-Brand, J., Tyndall, A., and
Pioro, M. and Calabrese, L.H. (1995). How useful is sural nerve biopsy in Nitzsche, E.U. (2005). The value of [18F]FDG-PET in the diagnosis of
the diagnosis of systemic vasculitis? Arthritis and Rheumatism, 38, S339. large-vessel vasculitis and the assessment of activity and extent of disease.
Radice, A. and Sinico, R.A. (2005). Antineutrophil cytoplasmic antibodies European Journal of Nuclear Medicine and Molecular Imaging, 32, 674–81.
(ANCA). Autoimmunity, 38, 93–103. Walvick, M.D. and Walvick, M.P. (2011). Giant cell arteritis: laboratory pre-
Rao, J.K., Allen, N.B., and Pincus, T. (1998). Limitations of the 1990 American dictors of a positive temporal artery biopsy. Ophthalmology, 118, 1201–4.
College of Rheumatology classification criteria in the diagnosis of vascu- Weyand, C.M., Fulbright, J.W., Hunder, G.G., Evans, J.M., and Goronzy,
litis. Annals of Internal Medicine, 129, 345–52. J.J. (2000). Treatment of giant cell arteritis: interleukin-6 as a biologic
Rappaport, W.D., Valente, J., Hunter, G.C., Rance, N.E., Lick, S., Lewis, T., marker of disease activity. Arthritis and Rheumatism, 43, 1041–8.
and Neal, D. (1993). Clinical utilization and complications of sural nerve Weyand, C.M. and Goronzy, J.J. (2003). Giant-cell arteritis and polymyalgia
biopsy. American Journal of Surgery, 166, 252–6. rheumatica. Annals of Internal Medicine, 139, 505–15.
Reuter, M., Schnabel, A., Wesner, F., Tetzlaff, K., Risheng, Y., Gross, W.L., Yamada, I., Nakagawa, T., Himeno, Y., Kobayashi, Y., Numano, F., and
and Heller, M. (1998). Pulmonary Wegener’s granulomatosis: correla- Shibuya, H. (2000). Takayasu arteritis:  diagnosis with breath-hold
tion between high-resolution CT findings and clinical scoring of disease contrast-enhanced three-dimensional MR angiography. Journal of
activity. Chest, 114, 500–6. Magnetic Resonance Imaging, 11, 481–7.
Salvarani, C. and Hunder, G.G. (2001). Giant cell arteritis with low erythro- Yamashita, H., Kubota, K., Takahashi, Y., Minaminoto, R., Morooka, M.,
cyte sedimentation rate: frequency of occurence in a population-based Ito, K., Kano, T., Kaneko, H., Takashima, H., and Mimoiri, A. (2012).
study. Arthritis and Rheumatism, 45, 140–5. Whole-body fluorodeoxyglucose positron emission tomography/
Schmidt, D., Hetzel, A., Reinhard, M., and Auw-Haedrich, C. (2003). computed tomography in patients with active polymyalgia rheumat-
Comparison between color duplex ultrasonography and histology of the ica: evidence for distinctive bursitis and large-vessel vasculitis. Modern
temporal artery in cranial arteritis (giant cell arteritis). European Journal Rheumatology, 22, 705–11.
of Medicinal Research, 8, 1–7. Ypsilantis, E., Courtney, E.D., Chopra, N., Karthikesalingam, A., Eltayab,
Schmidt, W.A., Kraft, H.E., Vorpahl, K., Volker, L., and Gromnica-Ihle, E.J. M., Katsoulas, N., Tang, T.Y., and Ball, R.Y. (2011). Importance of speci-
(1997). Color duplex ultrasonography in the diagnosis of temporal arte- men length during temporal artery biopsy. British Journal of Surgery,
ritis. New England Journal of Medicine, 337, 1336–42. 98, 1556–60.
Schnabel, A., Holl-Ulrich, K., Dalhoff, K., Reuter, M., and Gross, W.L. (1997). Zweegman, S., Makkink, B., and Stehouwer, C.D. (1993). Giant-cell arteritis
Efficacy of transbronchial biopsy in pulmonary vasculitides. European with normal erythrocyte sedimentation rate: case report and review of
Respiratory Journal, 10, 2738–43. the literature. Netherlands Journal of Medicine, 42, 128–31.
CHAPTER 21

Vasculitis in infancy,
childhood, and adolescence
Ross E. Petty

The spectrum of vasculitis varies with age. The most common vas- Evaluation of children for disease activity
culitides of infancy and childhood are all but unknown in the adult
Evaluation of the Birmingham Vasculitis Activity Score (BVAS) in
population; those that are common in the aging population do not
paediatric patients indicated limited applicability to this age group
occur in young children. Most forms of vasculitis are uncommon
(Morishita et al. 2012). Demirkaya et al. (2012) evaluated the BVAS
or rare in childhood. The exceptions are Henoch–Schönlein pur-
and disease extent index in large groups of children with Henoch–
pura (HSP) and Kawasaki’s disease (KD), which are not only the
Schönlein purpura, Wegener’s granulomatosis, Takayasu’s arteritis,
most common vasculitides of childhood, but are probably the most
and polyarteritis nodosa, and concluded that although these tools
common systemic vasculitides in any age group, and account for
were able to differentiate among the different vasculitides, had good
approximately 90% of childhood vasculitis. This chapter will focus
convergent validity, and good responsiveness to change, further
on vasculitides that occur predominantly in children and on fea-
tures of childhood-onset of vasculitis that also occur in the adult
population.
Table 21.1 Paediatric Rheumatology European Society classification of
vasculitis in childhood
Classification of vasculitis in childhood
The Paediatric Rheumatology European Society (PReS) has pro- 1. Predominantly large-vessel vasculitis
posed a classification that is age-appropriate and was arrived at by a. Takayasu’s arteritis
the consensus of an international committee of paediatric rheuma- 2. Predominantly medium-sized vessel vasculitis
tologists and nephrologists (Ozen et al. 2006). This classification is a. Polyarteritis nodosa
based primarily on the size of affected vessels, and secondarily on b. Cutaneous polyarteritis nodosa
the presence or absence of granulomata (Table 21.1).
c. Kawasaki’s disease
Takayasu’s arteritis (TA) is the only vasculitis of childhood affect-
3. Predominantly small-vessel vasculitis
ing large arteries. KD is the most common of the medium-sized
vasculitides. The small-vessel vasculitides are classified as granu- a. Granulomatous
lomatous or non-granulomatous. Granulomatosis with polyangiitis i. Granulomatosis with polyangiitis (previously known as Wegener’s
(formerly called Wegener’s granulomatosis) is the most common granulomatosis)
granulomatous small-vessel vasculitis, and HSP the most common ii. Eosinophilic granulomatosis with polyangiitis (previously known as
non-granulomatous small-vessel vasculitis of childhood. Other Churg–Strauss syndrome)
vasculitides occur in childhood, but they are very rare, and from b. Non-granulomatous
limited knowledge of their characteristics, they appear to be similar i. Microscopic polyangiitis
to the diseases in adults. ii. Henoch–Schönlein purpura
iii. Isolated cutaneous vasculitis
Common characteristics of iv. Hypocomplementaemic urticarial vasculitis
childhood vasculitis 4. Other vasculitides
a. Behçet’s syndrome
Some vasculitides, such as HSP and KD, have very characteristic pres-
b. Vasculitis secondary to infection (including hepatitis B-associated PAN),
entations, but the diagnosis of vasculitis can be difficult; multiple organ malignancies, or drugs
systems can be affected and there are no specific diagnostic tests for
c. Isolated CNS vasculitis
most. Consideration of a possible diagnosis of vasculitis should be
d. Cogan’s syndrome
prompted by the presence of one or more features shown in Table 21.2.
The characteristic ages at onset of the vasculitides of child- e. Unclassified
hood are shown in Table  21.3. These are generalizations, and Reproduced from Annals of the Rheumatic Diseases, Ozen, S., Ruperto, N., Dillon M.J., et al.
exceptions occur. 65, 936–41, 2006, with permission from BMJ Publishing Group Ltd.
284 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 21.2 Features that suggest a vasculitis syndrome administrative data base (Yang et al. 2005). A much higher incidence
(20.4 per 100 000 children less than 17 years of age) was reported
Clinical features in a smaller, questionnaire-based study in the United Kingdom by
Fever, weight loss, fatigue of unknown origin Gardner-Medwin et al. (2002). In this English study, however, the
Skin lesions (purpura, urticaria, livedo reticularis, painful incidence was highest in children of Asian ethnic origin. The peak age
nodules, ulcers) of onset is between 5 and 7 years (Yang et al. 2005; Gardner-Medwin
Neurological abnormalities (headache, mononeuritis multiplex, et al. 2002). There is slight preponderance of affected males, although
focal CNS signs) after age 10 years one report noted a higher frequency in girls (Yang
Arthralgia, arthritis, myalgia, myositis, or serositis et al. 2005). In Taiwan, the highest incidence is in the winter months,
Hypertension, haematuria, unexplained renal failure and lowest in June and July (Yang et al. 2005). Similar seasonal varia-
Palpable aneurysms, absence or asymmetry of pulses tion has been noted in other case series.
Pulmonary infiltrates or haemorrhage Familial HSP is observed occasionally. Motoyama and Iitaka
Cardiac ischaemia, arrhythmias (2005) reported two family members (seven sets of siblings and
a mother–daughter pair) in 428 families with at least one affected
Laboratory abnormalities
member. In three pairs, HSP occurred less than 1  month apart.
High ESR or CRP
A  number of genetic associations have been reported but none
Anaemia, leukocytosis, especially with eosinophilia
appears to be strongly associated with HSP or with the nephritis of
Haematuria
this disease. An angiotensin converting enzyme (ACE) gene dele-
Antineutrophil cytoplasmic antibodies
tion has been associated with high ACE levels and HSP (Young
Elevated factor VIII related antigen (von Willebrand factor antigen)
et al. 2012). Other possible genetic associations include heterozy-
gous deficiency of C2, and, in those with nephritis, increased
frequency of HLA B35 and DRB1*01, DRB1*11, and DRB1*14
Table 21.3 Characteristic ages at onset of vasculitides in infancy,
childhood, and adolescence (Amoli et al. 2002). An increased frequency of the C4B*Q0 null
allele reported in Icelandic children with HSP suggests that inad-
equate functioning of the complement pathway in elimination
Age range Vasculitis
of immune complexes may predispose to HSP (Steffanson et  al.
Infancy Kawasaki’s disease 2005). Galactose-deficient IgA1, known to be increased in chil-
Early childhood Kawasaki’s disease dren with HSP nephritis (Kiryluk et al. 2011), may act as an anti-
Henoch–Schönlein purpura gen, resulting in the formation of large immune complexes of IgA1
and anti-IgA1, which deposit in the kidney (and presumably in
Late childhood Henoch–Schönlein purpura
other tissues). The subject is thoroughly reviewed by Twilt and
Granulomatosis with polyangiitis Benseler (2012).
Polyarteritis nodosa
Adolescence Granulomatosis with polyangiitis
Aetiology and pathogenesis
Polyarteritis nodosa
The cause of HSP is unknown; however, clinical evidence of a preced-
ing upper respiratory tract infection and occasional reports of associ-
Microscopic polyangiitis
ations with streptococci, mycoplasma, Bartonella henselae, and with
Churg–Strauss syndrome a variety of viruses suggest an infectious trigger. Reports document
Takayasu’s arteritis the rare occurrence of HSP following influenza A H1N1 immuni-
zations (Watanabe 2011). The high serum IgA levels (Trygstad and
Stiehm 1971; Saulsbury 1999) and the characteristic predominance
of IgA1 in the vasculitic lesions (Baart de la Faille-Kuyper et  al.
modifications would make them more useful in assessing disease
1973) also suggest the possibility of a transmucosal infectious aeti-
activity in childhood vasculitides.
ology. However, no organisms have been consistently linked to the
occurrence of HSP and the wide variety of bacterial and viral infec-
Henoch–Schönlein purpura tions that have been reported suggest that the effect of infection in
HSP is the most commonly reported vasculitis in children in general, rather than a specific infection, may be of more pathogenic
Europe, North America, and India. Data from other parts of the significance, possibly mediated via TLR 2 or 4 (Canpinar et al. 2010).
world are lacking, and in Japan, where KD is common, HSP may Several instances of HSP following treatment of arthritis, psoriasis,
be the second most common vasculitis of childhood. Schönlein or inflammatory bowel disease with the anti-TNF agents etanercept,
described the triad of arthritis, purpura, and abnormalities of the infliximab, or adalimumab have been reported in adults (Rahman
renal sediment (Schönlein 1837). Henoch added the fourth impor- et al, 2010). The author has observed one adolescent with HSP fol-
tant element, abdominal pain (Henoch 1974). lowing treatment of Crohn’s disease with infliximab. The pathogen-
esis of HSP has been reviewed by Lau et al. (2010).
Epidemiology and genetics
Although HSP occurs world-wide, incidence data from much Clinical manifestations
of the world are lacking. In Taiwan, an annual incidence of 12.9 The classification criteria for childhood Henoch–Schönlein pur-
(11.8–13.4) per 100  000 children less than 17  years of age was pura are shown in Table 21.4. The presence of purpura plus at least
reported in a study of 2759 cases over a 4-year period using an one other criterion is sufficient to make the diagnosis of HSP.
CHAPTER 21 vasculitis in infancy, childhood, and adolescence 285

Table 21.4 EULAR/PRINTO/PReS criteria for classification of Henoch– Table 21.5 Common manifestations of Henoch–Schönlein purpura in
Schönlein purpura. The presence of purpura plus at least one other childhood
criterion fulfills classification criteria for HSP
Manifestation Frequency (%)
Criterion Description Sensitivity (%) Specificity (%)
Purpura 100
Purpura Predominantly lower limb 89 86
Arthritis 82
Abdominal Acute diffuse colicky pain 61 64
Abdominal pain 63
pain (may include intussusception Gastrointestinal bleeding 33
and GI bleeding) Occult 23
Biopsy Skin: leukocytoclastic 93 89 Gross 10
vasculitis with predominant Duodenal obstruction 1
IgA deposition Intussusception 0.4
Kidney: proliferative Glomerulonephritis 40
glomerulonephritis with
predominant IgA deposition Haematuria 40

Arthritis or Acute onset of joint pain 78 42 Gross haematuria 7


arthralgia with or without swelling Proteinuria 25
Renal Proteinuria >0.3 g/24 h or 33 70 Nephrotic syndrome 3
involvement >30 mg/mmol (albumin:
Orchitis (% in boys) 9
creatinine ratio) on a spot
morning urine sample Seizures 2
Haematuria >5 RBC/hpf or
(From Saulsbury 1999 and Chang et al. 2004.)
≥2+ dipstick or red blood cell
casts in urinary sediment
Presence of Plus at least one other criterion 100 87
perforation, obstruction, and infarction may ensue. The small bowel
purpura
is most frequently affected by these complications. Intussusception
(Adapted from Ozen et al. 2006, 2010.) is rare (1 : 250) (Chang et al. 2004). It is reported to be ilioileal in
EULAR, European League Against Rheumatism; PRINTO, Paediatric Rheumatology more than two-thirds and ileocolic in the remainder (Chang et al.
International Trials Organisation; PReS, Paediatric Rheumatology European Society.
2004; Szer 1996). Ulceration of the stomach or duodenum has been
reported (Fu et al. 2005).
The classical clinical manifestations usually follow an upper respira- Renal disease, manifested by haematuria and proteinuria, occurs
tory tract infection and become evident over the course of a few days in 40–50% of patients, usually within the first month after onset of
or a week. The child may be quite well or have a low-grade fever and the rash. In the large series reported by Chang et al. (2005) micro-
malaise. The frequency of clinical manifestations is listed in Table 21.5. scopic haematuria occurred in 14.2%, and gross haematuria in 4.6%.
Palpable purpura occurs, predominantly on the buttocks Nephrotic syndrome occurred in less than 1%. Hypertension at
and lower extremities, and is the presenting complaint in first presentation was documented in 14% of children in one series;
three-quarters (Trapani et al. 2005). The lesions range in size from two-thirds of these had a normal urinalysis (Watson et al. 2012).
petechiae to coalescent purpura several centimetres in diam- Pulmonary haemorrhage has been reported in a small number of
eter. Larger lesions may ulcerate. Some lesions may be bullous. children with HSP (Chen et al. 2011). Peripheral or central nervous
Oedema may occur over the dorsum of the hands and feet and is system dysfunction, resulting from vascular obstruction, haemor-
a characteristic finding in the scalp, where it may be most evident rhage, or hypertension is another rare complication, and has been
on a dependent area of the head, particularly in young children. reviewed by Garzoni et al. (2009).
Scrotal oedema is rare.
Diagnosis and differential diagnosis
Arthralgias or arthritis occurs in approximately 80% of chil-
dren, and are the presenting complaint in 15% (Trapani et  al. According to the criteria in Table 21.4, a diagnosis of HSP can be
2005). It usually affects four or fewer large joints. In the series of made on the basis of the presence of palpable purpura with one
100 children reported by Saulsbury (1999), the feet and ankles or more of the following criteria: arthralgia or arthritis, abdomi-
were affected in 72%, knees in 50%, hands and wrists in 26%, and nal pain, and haematuria. Occasionally, however, biopsy of the skin
elbows in 10%. The swelling is characteristically periarticular and or kidney is necessary to confirm the diagnosis. The differential
the joint is tender and painful on motion. The entire course of the diagnosis includes microscopic polyangiitis (MPA), polyarteritis
joint disease usually begins within a week of the development of nodosa (PAN), IgA nephropathy, and acute haemorrhagic oedema
purpura, is usually limited to a few days to a week in duration, and of childhood.
resolves completely.
Abdominal pain occurs in up to two-thirds of children, and may
Laboratory investigations
be the presenting complaint in 12% (Trapani et al. 2005). Colicky Pathology
abdominal pain may be severe and be accompanied by vomit- The characteristic lesion is leukocytoclastic vasculitis of small cap-
ing and occult or, less commonly, gross blood in the stool. Bowel illaries and postcapillary venules in the skin, and focal or diffuse
286 SECTION 5 vasculitic diseases and syndromes and related disorders

proliferative glomerulonephritis. Deposition of IgA in affected ves- Prognosis


sels is highly characteristic (Ferrario and Rastaldo 2005).
The long-term prognosis is usually excellent, but HSP may have a
Laboratory investigations monocyclic or polycyclic course and recurrences (usually purpura
Leukocytosis and thrombocytosis are present. The erythrocyte and abdominal pain) occur in one-third to one-half of children.
sedimentation rate (ESR) and C reactive protein (CRP) are usu- Haematuria and proteinuria may persist for well over a year in
ally elevated, and serum IgA levels are elevated in one-half of the up to one-half of patients (Chang et  al. 2005). The development
patients. Factor VIII related antigen has been shown to be elevated of chronic renal failure in 5% may be predicted by the presence
in the acute phase of the disease in a small study of children with of hypertension and nephritic or nephrotic syndrome at onset.
HSP (de Mattia et  al. 1995). Components of the complement In such patients progressive renal dysfunction may occur over a
system are usually normal, although early in the disease course period of many years. In children with normal urinalysis and blood
activation of the alternative complement pathway is common. pressure at diagnosis, the risk of developing hypertension as a late
In general, there is no increased prevalence of autoantibodies, sequela is low (Nussinovitch et al. 2005).
although IgA antibodies to endothelial surface antigens may be
detected using human umbilical cord endothelial cells (Yang et al. Kawasaki’s disease
2002). It has been reported that antibodies to neutrophil cytoplas- The description by Kawasaki (1967) of the febrile exanthematous
mic antigens (ANCA) of the IgA, but not IgG isotype, are associ- illness that now bears his name, marked an important milestone,
ated with early HSP, but disappear in the convalescent stage of the although isolated examples of this disease had been previously
disease (Ozaltin et al. 2004). recognized, often diagnosed as fatal infantile polyarteritis nodosa
The presence of haematuria, proteinuria, and, occasionally cel- (Munro-Faure 1959). The disease is one of the most important
lular casts indicates the presence of glomerulonephritis. exanthematous illnesses of infancy and childhood, the second most
common childhood vasculitis world-wide, and the most common
Treatment cause of acquired heart disease in the industrialized world.
In most patients, supportive treatment, including analgesia, is all
that is required. Although glucocorticoids may rapidly reduce the Epidemiology and genetics
severity of the joint and skin disease, they are seldom required for the KD is a disease of young children; the peak age at onset in North
management of these manifestations. There is no strong evidence America is about 2 years (Holman et al. 2003) and more than 80%
that early treatment with glucocorticoids reduces the risk of renal have onset under the age of 5 years. Occasionally, older children
or gastrointestinal disease. In a double-blind placebo-controlled and adolescents have KD. Boys are affected more commonly than
study, Huber et al. (2004) compared the effect of prednisone on the girls (1.4 : 1.0). The incidence of KD varies widely throughout the
development or renal or gastrointestinal disease at 1 year. The treat- world, being highest in Japan. Detailed epidemiological studies
ment group of 20 children were given oral prednisone (2 mg/kg/ in Japan demonstrated a steady increase in the incidence over the
day for 2 weeks, followed by a weaning dose for 1 week). A similar past 14 years to 140 per 100 000 children under age 5 years, and it
group of children received supportive care (nasogastric suction, has been suggested that at present rates, 1 in every 150 children in
parenteral nutrition). There was no significant difference in the rate Japan will suffer from this disease (Burns et al. 2005).
of renal disease or gastrointestinal complications at 1 year, although One per cent of children with KD have an affected sibling
two children in the placebo group had intussusception compared to (Yanagawa et  al. 1995)  and concordance in twins is 14% (Harada
none in the prednisone-treated group. et  al. 1986). Interestingly, most affected twins developed the dis-
In the face of established renal disease, the value of glucocor- ease within 2 weeks of each other, suggesting an important role for
ticoids is controversial. There is some evidence that high-dose an environmental agent. In North America, children of Japanese,
methylprednisolone may minimize the effects of renal disease Southeast Asian, Korean, and Chinese ancestry are particularly at
(Niaudet and Habib 1998). A number of case series document the risk to develop KD. African Americans may have a lower risk of KD
effectiveness of cyclosporine in children with HSP complicated by than Americans of Asian ancestry, but have a higher frequency than
nephritis (Shin et  al. 2005b). In a small non-randomized study, is seen in American Caucasians (Davis et al. 1995). There is a sug-
azathioprine together with glucocorticoids was shown to improve gestion that African Americans who do get KD have a lower risk of
renal outcome of children with HSP and nephritis compared to coronary artery disease (Porcalla et al. 2005). The strongest genetic
treatment with glucocorticoids alone (Shin et al. 2005a). Dapsone association appears to be with the inositol 1,4,5-triphosphate (IP3)
was effective in eight children with HSP that had been refractory kinase-c (ITPKC) gene (Onouchi et al. 2008). In genome-wide asso-
to treatment with corticosteroids and had frequent relapses (Iqbal ciation screens, several loci of interest have been found on chro-
and Evans 2005). Recurrent purpura was particularly responsive mosomes 6 (in the HLA region), 8 (in the FAM167A-BLG region),
to this drug, but relapse upon cessation of dapsone was frequent. chromosome 20 (in the CD40 region), and in the FCGR2A gene
Saulsbury (2009) reported the response of two children with HSP (Onouchi et al. 2012).
to colchicine. There has been no demonstrated role for biological
therapy (anti-TNF, anti-IL1R, anti-IL6R, anti-CD20) in HSP. Aetiology and pathogenesis
Experience with other drugs including anticoagulants and intra- Three distinct epidemics of KD were observed in Japan, in 1979,
venous immunoglobulin is very limited, but has not shown any 1982, and 1986 (Yanagawa et  al. 2001; Yanagawa et  al. 1999).
dramatic effect. Renal transplantation has been required in a few A  seasonal relationship has been shown in Japan (Burns 2005).
patients with HSP, and the rate of recurrence in the transplanted Seasonality is less striking in North American studies but more
kidney is approximately one in three (Meulders et al. 1994). cases are seen in the winter months (Bell et  al.1983). It seems
CHAPTER 21 vasculitis in infancy, childhood, and adolescence 287

likely that some property common to many viruses and bacteria an exudate is not present, unless there is an intercurrent pharyngeal
(a superantigen) is the inciting agent (Yeung 2004). This possibil- infection.
ity is supported by restricted T-cell receptor variable region usage
Rash
(Abe et al. 1992) and by evidence derived from the Lactobacillus
caseii-induced murine model of KD (Duong et al. 2003). The rash of KD is polymorphous, usually macular or maculopap-
ular, but not vesicular or crusting. It may occur anywhere and is
Clinical manifestations often accentuated in the perineal area. Desquamation occurs earli-
est in the perineum (often within the first 7–10 days), but is usually
KD is a systemic inflammatory illness with a wide spectrum
most dramatic on the palms and soles where sheet-like desquama-
of evolving clinical and laboratory features. The most serious
tion begins at the tips of the digits and around the nails in the sec-
long-term consequence of the disease is the development of aneu-
ond to fourth weeks of illness.
rysms, particularly affecting the coronary arteries. The classifica-
tion criteria have evolved somewhat and the current recommended Lymphadenopathy
criteria are shown in Table 21.6. By history, transient unilateral cervical adenopathy is quite com-
The clinical manifestations of KD can be roughly divided into mon, but it is often an early feature and may have disappeared
three phases, although there is great variability in the sequence of before the child is seen by a physician. Ultrasonography reveals the
clinical findings. The first phase is characterized by the abrupt onset presence of two or more nodes in a single mass.
of fever lasting for 10–14 days (untreated) and often preceded or
accompanied by symptoms of an upper respiratory tract or gastro- Extremity changes
intestinal infection. During this time, changes in the lips and oral The palms and soles may be bright red and the dorsum of the hands
mucosa, conjunctivitis, rash, cervical adenitis, and arthritis may and feet are often oedematous.
develop. A  subacute phase lasts for 2–4 weeks (untreated) and is Other features
characterized by normalization of the temperature, disappearance Aside from sinus tachycardia accompanying the fever, clinical evi-
of mucosal changes, conjunctivitis, adenitis and arthritis, and by dence of cardiac involvement is usually absent. In some children
desquamation of the skin of the palms and soles. It is during the sub- congestive heart failure occurs early in the disease course as a result
acute phase that coronary artery aneurysms most often make their of myocarditis. Obstruction of aneurysms (especially those >8 mm
appearance. The third convalescent phase may last months, during in diameter) may result in infarction, arrhythmias, or sudden death.
which time the inflamed arteries heal, other manifestations of KD In addition to the clinical changes that constitute the criteria for
disappear, and there is normalization of the acute phase response. classification of KD, there are many other common abnormalities.
Fever Benseler, McCrindle, et al. (2005b) documented a viral or bacterial
A persistent fever of 39–40°C is uniformly present. It is minimally infection in one-third of children with KD. Baker and colleagues
responsive to antipyretics and not responsive to antibiotics. It is (2009) noted one or more gastrointestinal symptoms (vomiting,
usually accompanied, in young patients in particular, by marked diarrhoea, abdominal pain) in 61% and respiratory symptoms
irritability. (rhinorrhoea or cough) in 35% of children within 10 days prior to
the diagnosis of KD. Arthritis most often occurs in the recovery
Conjunctivitis phase of the disease and most commonly affects knees and ankles.
Bilateral bulbar, non-suppurative conjunctivitis, sometimes with It is transient and leaves no sequelae. Anterior uveitis frequently
perilimbal sparing, is present in about 90% of patients. It is often accompanies the conjunctivitis (Smith et al. 1989). Transient sen-
accompanied by anterior uveitis, demonstrable on slit-lamp exam- sorineural deafness has been reported (Knott et al. 2001). Sterile
ination, and in 10–20%, by keratitis, vitreous opacities, or papil- pyuria and gallbladder hydrops (Suddleson et al. 1987) are occa-
loedema (Kumagai et al. 1996). sional complications.
Changes in the lips and oral mucosa
Red, swollen, vertically cracked lips, erythema of the oropharynx, Differential diagnosis
or strawberry tongue occur in 85–90%. The throat may be sore, but In the young child who presents to the physician in the second week
of illness with typical clinical features, the diagnosis is quite straight
forward. More often than not, however, the physician is confronted
with a patient who has atypical or incomplete features of KD, espe-
Table 21.6 Classification criteria for Kawasaki’s disease
cially early in the disease course. In this circumstance a number of
viral or bacterial illnesses may be considered, including adenovirus,
Fever persisting for at least 5 days (mandatory criterion) Plus four of the
following five features:
parvovirus, herpesvirus, leptospirosis, streptococcal, and staphylo-
coccal infections. Toxic shock syndrome is rarely a consideration.
◆ Changes in the peripheral extremities or perineum In the child with protracted disease, systemic juvenile idiopathic
◆ Polymorphous exanthema
◆ Bilateral conjunctival injection arthritis and polyarteritis nodosa should be considered. Because
◆ Changes of lips or oral cavity: injection of oral and pharyngeal mucosa many febrile children are treated with antibiotics, a drug reaction
◆ Cervical lymphadenopathy (typically unilateral) may be confused with incomplete KD.
In the presence of coronary artery involvement (detected on echocardiography)
and fever, fewer than four of the remaining five criteria are sufficient). Histopathology
(Reproduced from Annals of the Rheumatic Diseases, Ozen, S., Ruperto, N., Dillon M.J., et al. Post-mortem studies show that medium-sized arteries, particularly,
65, 936–41, 2006, with permission from BMJ Publishing Group Ltd.) but not exclusively the coronary arteries, are the site of inflammation
288 SECTION 5 vasculitic diseases and syndromes and related disorders

characterized by fibrinoid necrosis (Fujiwara and Hamashuma et al. 2007) used AST ≥200, total bilirubin ≥0.9, and CRP ≥7 as
1978). IgA producing B cells and plasma cells, and macrophages predictors of non-response, allocating 1 point to each. A  score
are the characteristic infiltrating cells (Rowley et al. 1997). Later, of ≥2 predicted non-response with a sensitivity of 77% and speci-
aneurysms, thrombosis, and stenosis develop. Inflammation of the ficity of 86%. The utility of predictive scores in North American
myocardium, endocardium, and pericardium is common in the patients has been questioned by Sleeper et al. (2011).
early stages of the disease; fibrosis occurs later. Patients who fail to respond to a second IVIg administration
should receive intravenous methylprednisolone (IVMP) 30 mg/
Laboratory investigations kg/day for 3 consecutive days. Failure to maintain a response to
Marked elevation of ESR and serum CRP, and elevation of the white this regimen should prompt use of oral prednisone (2 mg/kg/day
blood cell count are characteristic in the acute phase of the disease. in divided doses with a tapering schedule) until evidence of inflam-
The platelet count is initially normal, or occasionally subnormal, mation has disappeared. Although early studies suggested that glu-
but becomes markedly elevated in the subacute phase of the disease cocorticoids were contraindicated in the treatment of KD, this is
when counts exceeding one million are not unusual. Factor VIII clearly not the case (Sundel et al. 2003; Lang et al. 2006). Ogata et al.
related antigen levels are increased in early KD and may reflect the (2011) reported the efficacy of methylprednisolone and IVIg com-
activity of the vasculitis (Falcini et al. 1999). Urinalysis may reveal pared with IVIg alone as initial therapy in children with KD who
sterile pyuria. Mild elevations of liver enzymes are common. The had Egami scores over 3. Those treated with IVMP and IVIg had
irritability and headache are reflected by increased protein and a much higher rate of response (86%) compared to those treated
cells, and decreased glucose in the cerebrospinal fluid (Dengler with IVIg alone (23%). However, Sleeper et  al. (2011) could not
et al. 1998). Autoantibodies to endothelial antigens and neutrophil demonstrate improved coronary artery outcomes in children pro-
cytoplasmic antigens are not characteristic of KD, at least early in spectively classified as being IVIg resistant and treated with both
the disease course (Guzman et al. 1994; Nash et al. 1995). IVIg and i.v. methylprednisolone.
The possible role of biologicals (instead of IVIg) has not been
Diagnostic imaging established. Based on the demonstration by Hui-Yuen et al. (2006)
Echocardiography is the standard tool used to demonstrate coro- that TNF was critical to the development of experimental KD in
nary artery dilatation or aneurysms, stenosis, thrombosis, or artery mice, the therapeutic use of anti-TNF agents is logical. Burns et al.
wall hyperechogenicity. Nuclear perfusion scans and conven- (2008) found that both infliximab and IVIg were equally effective
tional coronary arteriography are indicated in selected patients. in treating children who had not responded to an initial infusion
Magnetic resonance imaging has also been suggested as an effective of IVIg. Son et al. (2011) observed a shorter duration of fever and
non-invasive measure for following patients with coronary artery fewer days of hospitalization, but no differences in coronary artery
changes (Tacke et al. 2011). outcome, in patients in whom re-treatment was with infliximab
Electrocardiography may demonstrate evidence of pericarditis, compared to IVIg. Mori et al. (2012) demonstrated regression of
myocarditis, or myocardial ischaemia. It is recommended (Dajani coronary artery abnormalities (hyperechogenicity or dilatation) in
et al. 1994) that it be performed at the time of diagnosis and con- 18 of 20 patients who received infliximab (5 mg/kg) for retreatment
sidered again 6–8 weeks later. Twenty per cent or more of untreated of IVIg-resistant KD. Other approaches such as plasmapheresis
patients develop coronary artery aneurysms (Belay et  al. 2006), (Mori et al. 2012) and cytotoxic drugs have been little studied and
almost always between the second and eighth weeks of disease, their role is probably minimal.
although they occasionally develop earlier.
Follow-up
Treatment Monitoring children with KD is important in the acute and con-
Treatment of KD is initiated in hospital with intravenous immu- valescent phases of the disease. In those with coronary artery
noglobulin (IVIg)(2 g/kg) and aspirin (80–100 mg/kg/day). There abnormalities, much longer follow-up is needed. In the absence of
is usually a prompt response to this treatment with immediate coronary aneurysms, the child with KD can be considered to have
fall in fever, improvement in sense of well-being, and disappear- recovered when clinical signs and symptoms have disappeared and
ance of other signs over a few days. High-dose aspirin therapy is laboratory abnormalities, especially platelet count and ESR, have
continued until the platelet count rises, at which time the dose returned to normal. Until that time they should receive low-dose
is reduced to 2–3 mg/kg/day until the platelet count normalizes. aspirin and be evaluated every 2–4 weeks. The present of coronary
IVIg has been shown to lower the incidence of coronary artery artery changes necessitates more intensive observation. Depending
aneurysms (Newburger et  al. 1986). Unfortunately 10–20% of on the severity of the arterial lesion, patients may need restriction
children fail to respond to the first dose of IVIg. In that event, of physical activities, repeated echocardiography or cardiac stress
the diagnosis should be re-evaluated, and if KD is still consid- tests, or coronary angiography (Dajani et al. 1994). Management of
ered to be the correct diagnosis, a second dose of IVIg (2 g/ giant aneurysms requires prolonged use of anticoagulants.
kg) should be given. There have been several attempts to pre-
dict response to IVIg in Japanese children. The Egami score Outcome
(Egami et al. 2006) identified five predictors of non-response to With the exception of coronary artery disease, enduring complica-
IVIg:  age less than 6  months (1 point), duration of illness less tions of KD are rare. The outcome of KD has been dramatically
than 4  days (1 point), platelet count ≤30  × 1010/l (1 point), improved by treatment with IVIg, aspirin, and, when necessary,
CRP ≥8 mg/dl (2 points), and ALT ≥80 U/l (2 points). A total glucocorticoids. Treatment with IVIg has reduced the incidence
score of 3 or more identified the IVIg-resistant patients with a of coronary artery aneurysms to less than 5%. Most aneurysms
sensitivity of 78% and specificity of 76%. The Sano score (Sano eventually disappear, but it is likely that abnormalities of the
CHAPTER 21 vasculitis in infancy, childhood, and adolescence 289

endothelium persist, perhaps for decades. Aneurysms rarely rup- Table 21.7 Classification criteria for granulomatosis with polyangiitis
ture, but occlusion with thrombus, especially in the child with giant
aneurysms (>8 mm in diameter), may cause myocardial infarction The presence of three of the following criteria fulfils the classification criteria:
or sudden death, even years after the initial illness. ◆ Abnormal urinalysis (proteinuria > 0.3 g/24 h, >5RBC/hpf or RBC casts)
Long-term studies of mortality in patients who had KD are ◆ Granulomatous inflammation within artery wall or in perivascular site
beginning to emerge. A  Japanese study of more than 6000 indi- ◆ Chronic nasal and/or sinus inflammation, recurrent epistaxis, nasal septal
viduals who had KD between 1982 and 1992, and who were fol- perforation, or saddle nose deformity
lowed until 2001, showed that although early death occurred in ◆ Subglottic, tracheal, or endobronchial stenosis
29 patients during the acute phase, later deaths were not signifi- ◆ Nodules, cavities, or fixed infiltrates on chest radiography or CT scan
cantly increased (Nakamura et al. 2005). The long-term outcome ◆ Presence of anti-PR3 (cANCA) or anti-MPO (pANCA)
of patients who had giant aneurysms was reported by Suda et al.
(Adapted from Annals of the Rheumatic Diseases, Ozen, S., Pistorio, A., Iusan S., et al. 69,
(2011) in 76 patients with a median follow-up of 19 years (range 798–06, 2010, with permission from BMJ Publishing Group Ltd.)
2 months to 36 years). Survival rates were 95%, 88%, and 88% at 10,
20, and 30 years, respectively. Seven patients died, and one under-
went heart transplantation. Coronary artery bypass surgery has (2012) reported associations between HLA DP and antiproteinase
been reviewed by Kitamura et  al. (2009), and showed event-free 3 ANCA and between HLA DQ and antimyeloperoxidase ANCA.
outcome of 60% at 25 years. Aetiology and pathogenesis
The aetiology and pathogenesis are unknown, but probably involve
ANCA-associated vasculitides genetic predispositions, environmental antigens, complement
Antibodies to neutrophil cytoplasmic antigens are characteristic activation, neutrophil apoptosis, and neutrophil extracellular trap
of granulomatosis with polyangiitis (formerly called Wegener’s (NET) formation, and a dysregulated adaptive immune system
granulomatosis), microscopic polyangiitis, and eosinophilic gran- resulting in the production of ANCAs, which directly contribute
ulomatosis with polyangiitis (formerly called Churg–Strauss syn- to the development of the disease. The subject has been thoroughly
drome), all of which predominantly affect small vessels. They are reviewed by Cartin-Ceba et al. (2012).
all very uncommon in childhood, although the incidence of gran- Clinical manifestations
ulomatosis with polyangiitis appears to be increasing (Grisauru Constitutional symptoms such as fever and weight loss, coupled
et al. 2010). with evidence of sinopulmonary or renal disease and laboratory
Antineutrophil cytoplasmic antibodies evidence of inflammation, should raise the question of a systemic
vasculitis such as GPA. Signs and symptoms reflecting inflamma-
Antibodies that react specifically with granules in the cytoplasm tion of the sinuses, nose, and ears are the most common present-
of neutrophils or monocytes were first associated with segmental ing features (Table 21.8). Subglottic stenosis is quite characteristic
necrotizing glomerulonephritis by Davies et  al. (1982) and with of childhood-onset GPA, occurring in up to 50%, but is usually
granulomatosis with polyangiitis by van der Woude (1985). Initially not manifested until later in the disease course. Pulmonary infil-
identified as C (cytoplasmic) or P (perinuclear) on the basis of the trates, nodules, and haemoptysis are sometimes present at onset,
immunofluorescence staining pattern, specific antigens are now but increase in frequency with increasing disease duration.
identified as PR3 (proteinase 3)  for cANCA and MPO (myelop- Glomerulonephritis occurs in almost two-thirds. Cutaneous gran-
eroxidase) for pANCA. Although characteristic of this group of ulomas, often over the elbow or knee, may ulcerate. In some chil-
vasculitides, they are not specific and ANCAs are found in patients dren, inflammation of the nose results in a saddle nose deformity
with a wide variety of apparently unrelated disorders including and perforation of the nasal septum.
cystic fibrosis, inflammatory bowel disease, and autoimmune hepa-
titis (Rus and Handwerger 2000). Laboratory investigations
Elevated ESR, platelet count and white blood cell count are all
Granulomatosis with polyangiitis non-specific, but common findings in GPA. Elevation of Factor
Granulomatosis with polyangiitis (GPA) is a granulomatous vas- VIII related antigen (von Willebrand Factor Antigen) suggests the
culitis affecting small vessels of the respiratory tract, kidneys, and presence of active inflammation of blood vessels. The presence of
occasionally other sites. It is an important cause of pulmonary ANCA (chiefly cANCA directed against PR-3) is highly suggestive
haemorrhage in children and adolescents. The proposed criteria for of the diagnosis. Other autoantibodies, including antinuclear anti-
classification are shown in Table 21.7. bodies, are usually not present.
Chest radiographs show a range of abnormalities, including pleu-
Epidemiology and genetics
ral effusions, infiltrates, nodules, and cavitating lesions, even in the
The incidence of GPA appears to be increasing in adults (Watts
absence of pulmonary symptoms or signs. Computed tomography
et al. 2000; Mahr et al. 2006) and children (Grisauru 2010) and is
more clearly delineates these lesions. Radiographs may reveal opac-
approximately 0.64–1.2 /100 000 in children and adolescents. It is
ity of the sinuses indicating the presence of inflammation.
most common in late childhood and adolescence and affects girls
more frequently than boys (Cabral et al. 2009). It occurs more com- Treatment
monly in northern Europe than in southern Europe. Whether this GPA is a life-threatening disease, and early therapy with prednisone
reflects genetic or environmental factors is not known. GPA is very (1 mg/kg/day orally in divided doses) and cyclophosphamide
rarely familial. A  polymorphism of the PTPN22 gene appears to (2 mg/kg/day orally or 0.75 g/m2/month i.v.) for 6 months is the
be associated with GPA in adults (Jagiello et al. 2005). Lyons et al. initial treatment of choice. In the very sick child, glucocorticoids
290 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 21.8 Manifestations at disease onset: the ARChiVe cohort of with minimal disease. Rituximab (monoclonal antibody that binds
65 children with GPA CD20 on B lymphocytes) has shown promise in adults with GPA
and may decrease or eliminate the need for cyclophosphamide in
% some patients (Guerry et al. 2011).
Constitutional 89 Outcome
Malaise, fatigue 89 The outcome of patients with GPA has dramatically improved;
Fever 54 however, long-term outcome data are few. In a group of seven
Weight loss 43 adults with onset of GPA prior to age 16 years, and followed for at
Ear, nose, and throat 80 least 10 years, disease relapses were common, and long-term mor-
Nasal disease 65 bidity was significant:  infertility, malignancy, renal failure, nasal
Sinusitis 60 septal defect, corticosteroid side-effects, and, in one patient, death
Otitis/ mastoiditis 14 from pulmonary complications of GPA (Arulkumaran et al. 2011).
Subglottic disease 14
Hearing loss 11 Microscopic polyangiitis
Oral ulcers 9
Microscopic polyangiitis (MPA) is a necrotizing vasculitis
Pulmonary abnormalities 80 of small vessels characterized by few or no immune deposits
Dyspnoea 58 (pauci-immune) (Savage et al. 1985; Peco-Antic et al. 2006; Hattori
Chronic cough 52
et al. 2001; Besbas et al. 2000). MPA is similar to GPA in that it
Haemoptysis/ alveolar haemorrhage 45
affects both kidneys and lungs, but differs in that, unlike GPA,
Pleurisy 23
Oxygen dependency 19
the upper airways are not affected, and it is most strongly associ-
Radiographic abnormalities ated with pANCA (anti-MPO) rather than cANCA (anti-PR3) as
Nodules 42a in GPA.
Fixed infiltrates 23a Epidemiology and genetics
Abnormal pulmonary function tests 43b
MPA is rare in childhood and adolescence and there are no accu-
Ocular abnormalities 37 rate incidence data. In adults, the highest incidence rates have been
Non-specific red eye 15 reported in Japan (14.8/million), with lower incidence rates in
Conjunctivitis 9 Europe (Chen and Kallenberg 2010). In the paediatric age range it
Scleritis 5 is more common in girls and occurs predominantly in the second
Cutaneous abnormalities 35 decade of life. No genetic markers have been identified.
Palpable purpura/ petechiae 23
Aetiology and pathogenesis
Gastrointestinal abnormalities 42 No aetiology has been identified. Graves’ disease treated with
Non-specific abdominal pain 32
propylthiouracil has been associated with MPA in four children
Chronic nausea 17
(Fujieda et al. 2002). Higher frequencies have been reported in the
Musculoskeletal abnormalities 57 spring (Peco-Antic 2006), suggesting a possible infectious cause.
Arthralgia/ myalgia 54 There is some evidence that anti-MPO antibodies may play a direct
Arthritis 20 role in pathogenesis (Schlieben et al. 2005), although the antibody
Central nervous system abnormalities 25 in itself is insufficient to cause the disease.
Severe headache 14
Clinical manifestations
Dizziness 12
Constitutional symptoms (fever, weight loss) may be present for
Renal abnormalities 75 months before pulmonary or renal disease becomes evident. Renal
Abnormal urinalysis 75
involvement at onset occurs in all patients and ranges from asymp-
Biopsy-proven glomerulonephritis 52
tomatic microscopic haematuria and proteinuria to glomerulone-
Elevated serum creatinine 42
phritis, nephrotic syndrome, and renal failure. Hypertension occurs
a Of 62 children who underwent chest radiographs. in at least one-half of patients. Pulmonary disease presents as haem-
b Of 35 children who had pulmonary function tests performed.
optysis and chest radiographs show infiltrates in at least one-half.
(Reproduced from Arthritis and Rheumatism, Cabral, D.A., Uribe, A.G., Benseler, S., et al. 60,
3413–24. Copyright © 2009 by the American College of Rheumatology.)
Rashes, including purpura, occur in almost all patients. Headache
and seizures may occur and may reflect cerebral vasculitis.
can be administered as i.v. methylprednisolone (30 mg/kg/d; maxi- Laboratory investigations
mum 1 gram/day) on 1 to 5 consecutive days, followed by oral Inflammatory indices are increased non-specifically. ANCA with
prednisone, tapered according to clinical response. A number of specificity for myeloperoxidase is usually detected in significant
studies in adults with GPA have sought therapeutic pathways that titres. Renal histopathology shows focal segmental glomerular
reduce exposure to cyclophosphamide. Thus cyclophosphamide necrosis with fibrous and fibrocellular crescents (Peco-Antic et al.
induction, followed by maintenance with azathioprine was found 2006). The lesions are pauci-immune; immunoglobulin deposits
to be effective (Jayne et al. 2003) (see Chapter 31). Non-response are not detected in the lesions by immunofluorescence micros-
to i.v. cyclophosphamide should prompt a change to oral adminis- copy. Haematuria and proteinuria reflect nephritis and nephrotic
tration. Methotrexate is an effective agent for induction in patients disease.
CHAPTER 21 vasculitis in infancy, childhood, and adolescence 291

Treatment and outcome Classification


In addition to oral prednisone (1–2 mg/kg/day) or intravenous The PReS/EULAR criteria, for childhood PAN are shown in
methylprednisolone (30 mg/kg/day on 1–3 consecutive days; maxi- Table 21.9.
mum 1 gram/day), cyclophosphamide is usually recommended for
induction therapy, followed by maintenance with methotrexate, Epidemiology and genetics
azathioprine, or mycophenolate mofetil (Peco-Antic et  al. 2006; This disease occurs world-wide, but is most commonly reported
Chung and Seo 2010). No paediatric therapeutic trials have been from Japan and Turkey (Maeda et  al. 1997; Ozen et  al. 2004). It
reported. The outcome is variable and many patients require dialy- affects males and females with equal frequency and, in child-
sis or renal transplantation. hood, appears to be most common in late childhood and early
adolescence.
Churg–Strauss syndrome
Churg–Strauss syndrome (CSS), or allergic granulomatosis and Aetiology, pathogenesis, and pathology
angiitis, now called eosinophilic granulomatosis with polyangiitis The aetiology of PAN is unknown. The associations of PAN with
(EGPA) (Churg and Strauss 1951)  is a granulomatous vasculitis hepatitis B and C, reported in adults, do not appear to be a major
affecting small vessels predominantly in the lungs, usually on a factor in childhood-onset disease. Familial occurrence is rare
background of severe asthma and eosinophilia and accompanied by and no genetic markers have been identified. The histopathol-
anti-MPO ANCAs. It is the least common of the ANCA-associated ogy is characterized by segmental necrotizing vasculitis of small
vasculitides in childhood. There are probably fewer than 40 chil- and medium-sized arteries with fibrinoid necrosis. Destruction of
dren and adolescents with CSS reported (Boyer et al. 2006; Zwerina the internal elastic lamina results in aneurysm formation, which
et al. 2008). is most common in bifurcations of the mesenteric and renal
vasculature.
Clinical and laboratory manifestations
There are too few cases to allow development of paediatric cri- Clinical manifestations
teria, although some differences appear to exist between chil-
Fever, weight loss, and abdominal pain (reflecting ischaemia) mus-
dren and adults with this disease, and the American College of
cle pain, or tenderness, livedo reticularis, and painful subcutaneous
Rheumatology criteria for the classification of EGPA are usually
nodules, typically in the calves or soles of the feet, herald the onset of
used: a history of asthma, eosinophilia above 10%, mononeuropa-
the disease (Cassidy and Petty 2011) (Table 21.10). Arthritis occurs
thy/ polyneuropathy, history of allergy, non-fixed pulmonary infil-
in up to two-thirds of patients. Peripheral neuropathy or central
trates, paranasal sinus abnormality, and a biopsy with extravascular
nervous system disorders occur in approximately two-thirds.
eosinophils. The presence of four or more criteria has a sensitivity
Highly suggestive, but rare, manifestations include episcleritis and
of 85% and specificity of 99.7% in adult Churg–Strauss syndrome
scrotal pain secondary to vasculitis involving the testis.
(Masi et al. 1990).
In addition, cardiovascular disease (pericarditis, myocarditis, Investigations
myocardial infarction) occurs in approximately half of the patients
Markers of the acute phase response are abnormal. Autoantibodies
with CSS irrespective of age. Renal involvement, although rare in
are usually absent, although ANCAs are occasionally present, usu-
adults with CSS, was seen in up to 40% of children.
ally reacting with myeloperoxidase (pANCA). Arteriography or
Skin manifestations (petechiae, purpura) are a common early
magnetic resonance angiography confirm the presence of aneu-
presentation of CSS in childhood. ANCA positivity may be
rysms involving the renal, celiac, or mesenteric arteries. Excisional
less frequent in children than adults with CSS. Biopsy shows
biopsy of a subcutaneous nodules reveals the typical necrotizing
eosinophil-containing granulomata.
fibrinoid necrosis of a small muscular artery.
Treatment and outcome
Conventionally, children with CSS are treated with prednisone
(2  mg/kg/day in divided doses, or intravenous methylpredni- Table 21.9 Criteria for classification of childhood polyarteritis
solone (30 mg/kg/day, maximum 1 gram/day) on 3 consecutive nodosa
days, together with cyclophosphamide. Plasmapheresis has been
used in patients resistant to conventional therapy and rituximab Histological evidence (necrotizing vasculitis of small or medium-sized
may be an effective alternative to cyclophosphamide (Rich and muscular arteries) or angiographic evidence (aneurysm, stenosis, or occlusion
Brown 2012), but experience with the use of this drug in children of medium or small arteries), plus one of the following:
is very limited. Outcome, although much improved in recent ◆ Skin involvement: livedo reticularis, painful subcutaneous nodules,
years, is still somewhat guarded, and cardiovascular events may superficial or deep skin infarctions
lead to death. ◆ Myalgia: muscle pain or tenderness
◆ Hypertension: systolic/ diastolic blood pressure >95 centile for height
Childhood polyarteritis nodosa ◆ Peripheral neuropathy: glove or stocking sensory neuropathy; motor
mononeuritis multiplex
and cutaneous polyarteritis ◆ Renal disease: proteinuria (>0.3 g/24 h); haematuria (<5RBC/hpf or RBC
Polyarteritis nodosa (PAN) is a relapsing necrotizing vasculitis of casts); GFR <50% of normal
small and medium-sized muscular arteries, resulting in the forma- (Adapted from Annals of the Rheumatic Diseases, Ozen, S., Pistorio, A., Iusan S., et al. 69,
tion of aneurysms. 798–06, 2010, with permission from BMJ Publishing Group Ltd.)
292 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 21.10 Clinical abnormalities in 81 children with Table 21.11 Classification criteria for childhood Takayasu’s arteritis
polyarteritis nodosa
Angiography (conventional, CT, or MRI) of the aorta or its main branches
Clinical finding Frequency (%) and pulmonary arteries showing aneurysm/ dilatation, narrowing,
occlusion, or thickened arterial wall not due to fibromuscular dysplasia,
Fever 84 or similar causes; changes usually focal or segmental, plus one of the
Arthritis/ arthralgia 74 following:

Abdominal pain 68 Lost/ decreased/ unequal peripheral artery pulse(s)

Myalgia 67 Claudication: focal muscle pain induced by physical activity

Skin abnormalities Discrepancy of four limb systolic BP >10 mm Hg difference in any limb.
Rash 69 Audible murmurs or palpable thrills over large arteries
Oedema 20
Petechiae 17 Systolic/diastolic BP greater than 95th percentile for height

Mucous membrane abnormalities 9 Erythrocyte sedimentation rate >20 mm per hour or CRP above normal

Renal disease 25 (Adapted from Annals of the Rheumatic Diseases, Ozen, S., Ruperto, N., Dillon M.J., et al.
65, 936–41, 2006, with permission from BMJ Publishing Group Ltd.)
Nervous system abnormalities
Seizures 16
Others, including peripheral neuropathy 10
the Indian subcontinent. Although the incidence and prevalence
Cardiac disease 21 are uncertain, it is probably the third most common vasculitis in
Respiratory disease 7 childhood and adolescence world-wide. It is the most common
cause of renovascular hypertension in Asian children (Chugh and
Cervical adenopathy <5
Sakhuja 1992). It is twice as common in girls as in boys in most
Splenomegaly <5 reported series. Although TA has been reported in twins and
(Cassidy, J.T., and Petty, R.E. 2011)
other family members (Morishita et al. 2011), it is seldom familial.
HLA B-52 has been associated with TA in Japan, Korea, and India
(Yajima et al. 1994).
Treatment and outcome Aetiology, pathogenesis, and pathology
Prednisone (1–2 mg/kg/day in divided doses) is usually sufficient The aetiology is unknown but an immune pathogenesis is likely
to control this inflammatory vasculitis. Failure to respond within a (Arnaud et al. 2006).
few weeks should prompt the addition of azathioprine (2 mg/kg/ In acute disease there is a γδ T-cell, cytotoxic T-cell, and NK
day) or cyclophosphamide (2 mg/kg/day). Most disease is mono- cell infiltration of the vasa vasorum and destruction of the media
phasic, and most children with PAN do well, although myocar- and adventia through release of perforin, with intimal hyperplasia
dial infarction, persistent peripheral neuropathy, renal failure, and resulting in stenosis (Seko 2000). With time the elastic fibres of the
hypertension many occur. media are destroyed, leading to aneurysms, and fibrosis replaces the
smooth muscle of the media, leading to stenosis. B lymphocytes are
Cutaneous polyarteritis implicated by their presence in the inflammatory lesions (Arnaud
Bansal and Houghton (2010) reviewed the 119 published cases of et al. 2006). Granulomas with giant cells are present (Kothari 2002).
cutaneous polyarteritis in childhood, noting the presence of arthral- An association with a strongly positive Mantoux skin test and
gia or arthritis in 58%, myalgia or myositis in 53%, and cranial or tuberculosis is seen in some parts of the world where tuberculosis
peripheral nerve involvement in 14%. Absent from this group of is common (Hahn et al. 1998), but the significance of the associa-
children was evidence of visceral vasculitis. The characteristic cuta- tion is not clear (Castillo-Martinez and Amezcua-Guerra 2012).
neous lesions include livedo reticularis and painful subcutaneous An association with Sweet’s syndrome has been reported (Ma
nodules reflecting inflamed medium-sized muscular arteries. The et  al. 2011). Ulcerative colitis is occasionally associated with TA
disease is often associated with preceding streptococcal infection. (Balamtekin et al. 2009).
Histological examination demonstrated lesions like those seen in
polyarteritis nodosa. The outcome following treatment with pred- Clinical manifestations
nisone (0.5–1.0 mg/kg/day) is usually very favourable. TA is classified according to the distribution of aneurysms and ste-
nosis in vessels into type 1 (aortic arch and its branches, ~60%);
Takayasu’s arteritis type II (thoracoabdominal aorta and its branches, ~40%); type
Takayasu’s arteritis (TA) is characterized by inflammation of the III (lesions of both aortic and thoracoabdominal aorta and is
aorta and its major branches. Classification criteria for childhood branches, <1%); and type IV (with pulmonary artery involvement,
TA are shown in Table 21.11. <1%). Renal artery stenosis is present in about three-quarters
(Kothari 2002).
Epidemiology and genetics Symptoms and signs of TA reflect the pattern of vessel involve-
TA is rare in infancy, and most common in adolescence. It occurs ment. Hypertension and its manifestations are the most common
world-wide but most reports come from Japan, Southeast Asia, and presenting features, occurring in 21 of 23 children in a reported
CHAPTER 21 vasculitis in infancy, childhood, and adolescence 293

series (Stanley et al. 2003). Fever, weight loss, anorexia, night sweats, reported studies ranges from 10% (Shrivasatava 1995)  to 31%
headache, claudication, chest wall pain, and myalgias are common (Dabague and Reyes 1996). Causes of death in adults with TA include
presenting features. A neurological event such as a cerebrovascu- cardiac failure, renal failure, ruptured aneurysms, and stroke. Studies
lar accident may be the initial manifestation of the disease, and of long-term survival of juvenile-onset TA have not been reported.
occurs at some time during the course of the disease in one-quarter
to one-half of patients (Kothari 2002). On examination, hyper-
tension, absence of one or more peripheral pulses, tenderness of Childhood primary angiitis of
major arteries, and the presence of bruits over the aorta or its main the central nervous system
branches support the diagnosis. The characteristic wreath-like Isolated central nervous system vasculitis has emerged as an impor-
retinal vascular pattern originally described by Takayasu is rare, tant category of vasculitis in children and adolescents in the past
although other retinal changes, resulting from ischaemia, are pre- decade. Once believed to be very rare, it appears that many children
sent in up to one-half of patients (Chun et al. 2001). with idiopathic central nervous system disease have vasculitis or
inflammatory brain disease (Benseler 2006).
Investigations
The acute phase response is usually markedly abnormal. Levels of Definition and classification
von Willebrand factor antigen, angiotensin converting enzyme, Childhood primary angiitis of the central nervous system (cPACNS)
and other markers of inflammation correlate poorly with disease is defined as a newly acquired unexplained neurological or psychi-
activity (Hoffman and Ahmed 1998). Antinuclear antibodies, atric deficit with specific angiographic and/or histopathological fea-
rheumatoid factors, and ANCAs are absent, although antibodies tures of angiitis within the CNS in a child ≤18 years of age at onset
to endothelial cell antigens and anticardiolipin antibodies may be (Benseler 2006). Based on imaging criteria, two categories of pri-
present (Eichorn et al. 1996). It is a difficult challenge to determine mary CNS vasculitis are recognized: angiography-positive disease
the presence and degree of active inflammation in a child with TA. that affects medium and large vessels, and angiography-negative
Aside from signs and symptoms, and abnormalities of the acute disease that affects small vessels (Celluci and Benseler 2010).
phase reactants, which may not reliably reflect the extent of active Angiography-positive disease is further classified as being progres-
disease, imaging studies are probably the best way to monitor dis- sive or non-progressive.
ease activity by the demonstration of new lesions (Kothari 2002;
Stanley et al. 2003). Epidemiology and genetics
Treatment and outcome The majority of cases of cPACNS (66 children) have been
reported from one institution, and there are no generalizable
Studies of the treatment of TA in adults have been the guide for
data regarding incidence. It is widely believed that the disease
management of the disease in children and adolescents. Aggressive
has been under-recognized. In the group of 62 patients with
treatment of hypertension is critical. Antiplatelet doses of aspirin
angiography-positive cPACNS reported by Benseler (2006) 61%
may be useful in preventing intravascular clotting, particularly if
were male and the median age at diagnosis was 7.2 years (range
anticardiolipin antibodies are present. Treatment is initiated with
0.7–17.6 years).
prednisone in doses of 1–2 mg/kg/day, usually in divided doses or
intravenous methylprednisolone (30 mg/kg/day on 1–3 consecutive
days, maximum dose 1 gram/day), and cyclophosphamide (2 mg/
Clinical manifestations
kg/day orally, or monthly for 4–6 months in an intravenous dose of The presenting signs and symptoms reflect the location and extent
0.5–1 g/m2 per dose), followed by maintenance with oral or sub- of underlying vasculitis. Many children are asymptomatic and have
cutaneous methotrexate (0.35–0.65 mg/kg/week). Mycophenolate sudden unexplained onset of CNS abnormalities, or acute severe
mofetil has been used in some patients with benefit (although headache. Seizures are a common presenting feature of small-vessel
usually not complete remission) (Goel et  al. 2010; Al Abrawi (angiography-negative) cPACNS. In others behavioural abnormali-
et al. 2008). Studies of anti-TNF agents show promise (Hoffman ties may dominate the clinical presentation, and in these children
et al. 2004). Rituximab administration, resulting in B-lymphocyte subtle focal defects may be important indicators of underlying
depletion, has been reported to induce remission in adults with TA cPACNS. Large-vessel cPACNS more often presents with focal
(Hoyer et al. 2012). The use of the biologicals in the management of abnormalities (Table 21.12).
TA is increasing, but their role is still not certain. Surgical stenting
or bypassing of severely stenotic areas of artery may be required in Investigations
the child with uncontrolled hypertension or vascular compromise Indicators of systemic inflammation are often, but not always,
of the central nervous system. Therapy was reviewed by Liang and elevated. ANAs and ANCAs are not present. Hutchinson et  al.
Hoffman (2005). described at least one abnormal inflammatory marker in 76% of
Hypertension, cerebrovascular accident, and renal failure are the children with small-vessel childhood PACNS. Anticardiolipin anti-
major long-term complications. In most children and adolescents, bodies are sometimes found in low titre (Hutchinson et al. 2010).
TA is a chronic or life-long illness with periods of quiescence punc- Cellucci et al. (2012) suggested that von Willebrand factor antigen
tuated by signs or symptoms of vascular insufficiency or labora- is a potential biomarker for childhood PACNS, and is closely corre-
tory evidence of active disease. Outcome is probably significantly lated with disease activity. Cerebrospinal fluid examination revealed
improved in those who receive early treatment, before fibrosis has led pleocytosis and elevated protein in one-third. Five of 332 children
to stenosis, which is not responsive to medical treatment. Long-term thought to have a demyelinating disease, were subsequently found
anti-inflammatory treatment is usually necessary. Mortality in to have cPACNS (O’Mahony et al. 2013).
294 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 21.12 Comparison of characteristics at diagnosis in children with 2012). Neurological deficits associated with large-vessel PACNS are
angiography-positive and angiography-negative cPACNS often permanent, but some children with small-vessel disease may
make a complete recovery following immunosuppressive treatment
Angiography positive Angiography negative (Hutchinson et al. 2010).
(large-vessel cPACNS) (small-vessel cPACNS)
N = 144 N = 58
Diffuse abnormalities
Rare vasculitis or vasculitis-like
Reduced consciousness 5% 38% syndromes in childhood
Cognitive dysfunction 42% 69% Cogan’s syndrome
Memory deficit 32% 64%
Behaviour abnormality 38% 66% A syndrome of interstitial keratitis followed by neurovestibu-
Concentration deficit 34% 64% lary deficits, including deafness, is a very rare occurrence in
childhood (Kundell and Ochs 1980; Orsoni et  al. 2004). It is
Focal abnormalities
thought to be an autoimmune disorder and antibodies to anti-
Fine motor deficit 90% 54%
gens in the inner ear have been described (Lunardi et al. 2002).
Gross motor deficit 88% 22%
Hemiparesis 72% 14% Initial management with corticosteroids is usually effective,
Hemisensory deficit 64% 18% but leflunomide (Xie et  al. 2009)  and mycophenolate mofetil
Gait abnormality 86% 62% (Hautefort et al. 2009) have both been reported to be effective in
Cranial nerve deficit 64% 14% corticosteroid-resistant children.
Language deficit 52% 78%
Movement disorder 24% 17% Susac’s syndrome
Optic neuritis 0 10% Subacute encephalopathy, retinal arterial branch occlusion, and sen-
Seizures sorineural hearing loss with characteristic cystic lesions in the corpus
Focal 10% 69% callosum is a very rare disorder caused by a microangiopathy of the
Generalized 4% 33% brain, retina, and inner ear (Hahn et al. 2004). It may mimic a demy-
Status epilepticus 0 24% elinating disease or acute disseminated encephalomyelitis (ADEM).
Headache is usually prominent, and cognitive changes, psychiatric
(Data from the CNS Vasculitis Clinic, Hospital for Sick Children, Toronto. Cabral, D.A. and
Benseler, S: in Textbook of Pediatric Rheumatology, 6th edn, p. 539. Cassidy, J.T., Petty, R.E.,
symptoms, sudden visual loss, hearing loss, vertigo, and tinnitus are
Laxer, R.M., Lindsley, C.B., eds. (2011) Elsevier, Philadelphia.) characteristic. Treatment with corticosteroids and cyclophospha-
mide or plasmapheresis has been suggested (Hahn et al. 2004).

Imaging studies are essential for diagnosis and classification of Sneddon’s syndrome
PACNS. Computed tomography (CT) often fails to demonstrate Livedo racemosa followed by cerebrovascular infarction (Sneddon’s
lesions, and magnetic resonance imaging is the study of choice syndrome) results from endothelial proliferation and obliteration
for demonstrating parenchymal lesions, and magnetic resonance of small arteries and arterioles in the skin and brain. It is extremely
angiography for demonstrating vascular lesions (Aviv et al. 2006). rare in children (Gottlöber et al., 2000). It can be associated with
The subject is thoroughly reviewed by Twilt and Benseler (2012). antiphospholipid antibodies and systemic lupus erythematosus.
When clinical suspicion suggests PACNS, but all imaging studies Immunosuppressive and anticoagulant treatment has been used
are normal, brain biopsy, including the meninges, gray and white (Gottlöber et al. 2000).
matter, is indicated. Benseler et al. (2005a) described four children
in whom conventional angiograph, CT, MRI, and MRA were all References
normal, but in whom histopathological studies reveal transmural Abe, J., Kotzin, B.L., Jujo, K., et al. (1992). Selective expansion of T cells express-
lymphocytic infiltration of small and medium-sized blood vessels ing T-cell receptor variable regions V beta 2 and V beta 8 in Kawasaki dis-
in both white and gray matter, and leptomeninges. ease. Proceedings of the National Academy of Sciences USA, 89, 4066–70.
Al Abrawi, S., Fouillet-Desjonqueres, M., David, L., Barral, X., Cochat, P., and
Treatment Cimaz, R. (2008). Takayasu arteritis in children. Pediatric Rheumatology,
6, 1–5.
Management of the neurological manifestations such as seizures
Amoli, M.M., Thomson, W., Hajerer, A.H., et al. (2002). HLA B35 association
requires the expertise of a paediatric neurologist. Treatment of the with nephritis in Henoch Schönlein purpura. Journal of Rheumatology,
underlying vasculitis is usually initiated with prednisone (2 mg/kg/ 28, 1266–70.
day weaned over a 6-month period as indicated) and intravenous Arnaud, L., Kahn, J.E., Girszyn, N., Piette, A.M., and Bletry, O. (2006).
cyclophosphamide (500–750 mg/m2 every 4 weeks for 6 months). Takayasu’s arteritis: An update on physiopathology. European Journal of
Following this induction phase, treatment is continued with pred- Internal Medicine, 17, 241–6.
nisone on a tapering dose, and azathioprine (2–3 mg/kg/day) or Arulkumaran, N., Jawad, S., Smith, S.W., et al. (2011). Long-term outcome of
mycophenolate mofetil (800–1200 mg/m2/day) for a period of at paediatric patients with ANCA vasculitis. Pediatric Rheumatology, 9, 1–7.
least 18 months (Hutchinson et al. 2010). Aviv, R. I., Benseler, S.M., Silverman, E.D., et al. (2006). MR imaging and
angiography of primary CNS vasculitis of childhood. American Journal
of Neuroradiology, 27, 192–9.
Outcome
Baart de la Faille-Kuyper, E.H.Y., Kater, L, Kooiker, C.J., and Dorhout Mees,
Early studies indicated a very poor prognosis in children with CNS E.J. (1973). IgA deposits in cutaneous blood vessel walls and mesangium
vasculitis. Intracerebral haemorrhage may occur (Pistracher et al. in Henoch-Schönlein syndrome. Lancet, 1, 892–3.
CHAPTER 21 vasculitis in infancy, childhood, and adolescence 295

Baker, A.L., Lu, M., Minich, L., et al. (2009). Associated symptoms in the Chen, M. and Kallenberg, C.G.M. (2010).The environment, geoepidemiology
ten days before the diagnosis of Kawasaki disease. Journal of Pediatrics, and ANCA-associated vasculitis. Autoimmunity Reviews, 9, A293–A298.
154, 592–5. Chen, S.-Y., Chang, K.-C., Yu, M.-C., Asueh, S., and Ou,.L-S. (2011).
Balamtekin, N., Gűrakan, F., Ozen, S., Oguz, B., and Talim, B. (2009). Pulmonary hemorrhage associated with Henoch-Schönlein purpura in
Ulcerative colitis associated with Takayasu’s arteritis in a child. Acta pediatric patients: case report and review of the literature. Seminars in
Paediatrica, 98, 1368–71. Arthritis and Rheumatism, 41, 305–12.
Bansal, N-K. and Houghton, K.M. (2010). Cutaneous polyarteritis nodosa Chugh, K.S. and Sakhuja, V. (1992). Takayasu’s arteritis as a cause of renovas-
in childhood: A case report and review of the literature. Arthritis, 2010, cular hypertension in Asian countries. American Journal of Nephrology,
687547. 306, 464–5.
Belay, E.D., Maddox, R.A., Holman, R.C., Curns, A.T., Ballah, K., and Chun, Y.S., Park, S.-J., Park, I.K., Chung, H., and Lee, J. (2001). The clinical
Schonberger, L.B. (2006). Kawasaki syndrome and risk factors for and ocular manifestations of Takayasu arteritis. Retina, 21, 132–140.
contrary artery abnormalities. Pediatric Infectious Diseases Journal, Chung, S.A. and Seo, P. (2010). Microscopic polyangiitis. Rheumatic Disease
25, 245–9. Clinics of North America, 36, 545–58.
Bell, D.M., Morens, D.M., Holman, R.C., et al. (1983). Kawasaki syndrome in Churg, J. and Strauss, L. (1951). Allergic granulomatosis, allergic angiitis, and
the United States 1976–1980. American Journal of Diseases of Children, periarteritis nodosa. American Journal of Pathology, 27, 277–301.
137, 211–4. Dabague, J. and Reyes, P.A. (1996). Takayasu arteritis in Mexico. A 38-year
Benseler, S.M. (2006). Central nervous system vasculitis in children. Current clinical perspective through literature review. International Journal of
Rheumatology Reports, 8, 442–449. Cardiology, 54 (Suppl.), S103–9.
Benseler, S.M., de Veber, G., Hawkins, C., et al. (2005a). Angiography nega- Dajani, A.S., Taubert, E.A., Takahashi, M., et  al. (1994). Guidelines for
tive primary central nervous system vasculitis in children. Arthritis and long-term management of patients with Kawasaki disease. Report
Rheumatism, 52, 2159–67. from the Committee on Rheumatic Fever, Endocarditis and Kawasaki
Benseler, S.M., McCrindle, B.W., Silverman, E.D., Tyrrell, P.N., Wong, J., and Disease. Council on Cardiovascular diseases in the Young. American
Yeung, R.S.M. (2005b). Infections and Kawasaki disease; implications Heart Association. Circulation, 89, 916–22.
for coronary artery outcome. Pediatrics, 116, 760–6. Davies, D.J., Moran J.E., Niall, J.F., and Ryan, G.B. (1982). Segmental necrotiz-
Besbas, N., Ozen, S., Saatchi, U., et al. (2000). Renal involvement in polyarteritis ing glomerulonephritis with anti-neutrophil antibody: Possible arbovi-
nodosa: evaluation of 26 Turkish children. Pediatric Nephrology, 14, 327–7. rus aetiology? British Medical Journal, 285, 606.
Boyer, D., Vargas, S.O., Slattery, D., Rivera-Sanchez, Y.M., and Colin, A.A. Davis, R.L., Waller, P.L., Mueller, B.A., et al. (1995). Kawasaki syndrome in
(2006). Churg-Strauss syndrome in children: A clinical and pathologic Washington State. Race-specific incidence rates and residential proxim-
review. Pediatrics, 118, 914–20. ity to water. Archives of Pediatric and Adolescent Medicine, 149, 66–9.
Burns, J.C., Best, B.M., Mejias, A. et  al. (2008). Infliximab treatment of de Mattia, D., Penza, R., Giordano, P., et al. (1995). Von Willebrand factor
immunoglobulin-resistant Kawasaki disease. Journal of Pediatrics, and factor XIII in children with Henoch-Shonlein purpura. Pediatric
153, 833–8. Nephrology, 9, 603–5.
Burns, J.C., Cayan, D.R., and Tong, G. (2005). Seasonality and temporal clus- Demirkaya, E., Ozen, S., Pistorio, A., et  al. (2012). Performance of the
tering of Kawasaki syndrome. Epidemiology, 16, 220–5. Birmingham Vasculitis Activity Score and Disease Extent Index in
Cabral, D.A. and Benseler, S. (2011). Granulomatous vasculitis, microscopic childhood vasculitides. Clinical and Experimental Rheumatology, 30
polyangiitis and primary angiitis of the central nervous system. In (Suppl. 70), S162–78.
Textbook of pediatric rheumatology, 6th edn, (ed. J.T. Cassidy, R.E. Petty, Dengler, L.D., Capparelli, E.V., Bastian, J.F., et al. (1998). Cerebrospinal fluid
R.M. Laxer, and C.B. Lindsley), p. 539. Elsevier, Philadelphia. profile in patients with acute Kawasaki disease. Pediatric Infectious
Cabral, D.A., Uribe, A.G., Benseler, S., et al. (2009). Classification, presen- Diseases Journal, 17, 478–81.
tatin, and initial treatment of Wegener’s granulomatosis in childhood. Duong, T.T., Silverman, E.D., Bissessar, M.V., and Yeung, R.S. (2003).
Arthritis and Rheumatism, 60, 3413–24. Superantigenic activity is responsible for induction of coronary arte-
Canpinar, H., Özaltin, F., Bilginer, Y., Bakkaloğlu, A., and Özen, S. (2010). ritis in mice:  An animal model of Kawasaki disease. International
Toll-like receptors 2 and 4 surface expression reflects endotoxin tolerance Immunology, 15, 79–89.
in Henoch-Schönlein purpura. Turkish Journal of Pediatrics, 52, 22–7. Egami, K., Muta, H., Ishii, M., et al. (2006). Prediction of resistance to intra-
Cartin-Ceba, R., Peikert, T., and Specks, U. (2012). Pathogenesis of venous immunoglobulin treatment in patients with Kawasaki disease.
ANCA-associated vasculitis. Current Rheumatology Reports, 14, 481–93. Journal of Pediatrics, 149, 237–40.
Cassidy, J.T. and Petty, R.E. (2005). Polyarteritis nodosa and related vas- Eichorn, J., Sima, D., Thiel, B., et al. (1996). Anti-endoethelial cell antibodies
culitides. In Textbook of pediatric rheumatology, 5th edn (ed. J.T. in takayasu arteritis. Circulation, 94, 2396–401.
Cassidy, R.E. Petty, R.M. Laxer, and C.B. Lindsley), p.  514. Elsevier, Falcini, F., Generini, S., Pignone, A., et al. (1999). Are angiotensin convert-
Philadelphia. ing enzyme and von Willebrand factor circulating levels useful param-
Castillo-Martínez, D. and Amezcua-Guerra, L.M. (2012). Self-reactivity eters to monitor disease activity in Kawasaki disease? Endothelium, 6,
against stress-induced cell molecules:  the missing link between 209–15.
Takayasu’s arteritis and tuberculosis? Medical Hypotheses, 78, 485–8. Ferrario, F. and Rastaldo, M.P. (2005). Henoch-Schonlein nephritis. Journal
Celluci, T. and Benseler, S.M. (2010). Central nervous system vasculitis in of Nephrology, 18, 637–41.
children. Current Opinion in Rheumatology, 22, 590–7. Fu, K.I., Yagi, S., Mashimo, Y., Sugitani, K., et  al. (2005). Regression
Cellucci, T., Tyrell, P.N., Pullenayegum, E., and Benseler S.M. (2012). Von of helicobacter pylori-negative duodenal ulcers complicated by
Willebrand Factor antigen—a possible biomarker of disease activity in Schönlein-Henoch purpura with H. pylori eradication therapy. The first
childhood central nervous system vasculitis? Rheumatology (Oxford), report. Digestive Diseases and Sciences, 50, 381–4.
51, 1838–45. Fujieda, M., Hattori, M., Kurayama, H., and Koitabashi, Y. (2002). Clinical
Chang, W.L., Yang, Y.H., Lin, Y.T., and Chiang, B.L. (2004). Gastrointestinal features and outcomes of children with antineutrophil cytoplasmic
manifestations of Henoch-Schönlein purpura: a review of 261 patients. antibody-associated glomerulonephritis associated with propylthi-
Acta Paediatrica, 93, 1427–31. ouracil treatment. Journal of the American Society for Nephrology, 13,
Chang, W.-L., Yang, Y.-S., Wang, L.-C., et al. (2005). Renal manifestations in 437–445.
Henoch-Schönlein purpura: a ten year study. Pediatric Nephrology, 20, Fujiwara,H. and Hamashima, Y. (1978). Pathology of the heart in Kawasaki
1269–72. disease. Pediatrics, 61, 100–7.
296 SECTION 5 vasculitic diseases and syndromes and related disorders

Gardner-Medwin, J.M.M., Dolezalova, P., Cummins, C., and Southwood, T.R. Jayne, D., Rasmussen, N., Andrassy, K., et al. (2003). A randomized trial of
(2002). Incidence of Henoch-Schönlein purpura, Kawasaki disease and rare maintenance therapy for vasculitis associated with antineutrophil cyto-
vasculitides in children of different ethnic origins. Lancet, 360, 1197–1202. plasmic autoantibodies. New England Journal of Medicine, 349, 36–44.
Garzoni, L., Vanoni, F., Rizzi, M., et al. (2009). Nervous system dysfunction Kawasaki, T. (1967). Acute febrile mucocutaneous syndrome with lymphoid
in Henoch-Schonlein syndrome:  systematic review of the literature. involvement with specific desquamation of the fingers and toes in chil-
Rheumatology, 48, 1524–9. dren. Arerugi, 16, 178–222.
Goel, R., Danda, D., Mathew, J., and Edwin, N. (2010). Mycophenolate Kiryluk, K., Moldoveanu, Z., Sanders, J.T., et  al. (2011). Aberrant glyco-
mofetil in Takayasu’s arteritis. Clinical Rheumatology, 29, 329–32. sylation of IgA1 is inherited in both pediatric IgA nephropathy and
Gottlöber, P., Bezold, G., Schjaer, A., et al. (2000). Sneddon’s syndrome in a Henoch-Schönlein purpura nephritis. Kidney International, 80, 79–87.
child. British Journal of Dermatology, 142, 374–5. Kitamura, S., Tsuda, S., Kobayashi, J., Nakajima, H., Yoshikawa, Y., Yagihara,
Grisauru, S., Yuen, G.W.H., Miettunen, P.M., and Hamiwka, L.A. (2010). T., and Kada, A. (2009). Twenty-five-year outcome of pediatric coronary
Incidence of Wegener’s Granulomatosis in children. Journal of artery bypass surgery for Kawasaki disease. Circulation, 120, 60–8.
Rheumatology, 37, 440–2. Knott, P.D., Orloff, L.A., Harris, J.P., et  al. (2001). Sensorineural hearing
Guerry, M.J., Brogan, P., Bruce, I.N., et al. (2011). Recommendations for the loss and Kawaski disease:  a prospective study. American Journal of
use of Rituximab in anti-neutrophil cytoplasmic antibody-associated Otolaryngology, 22, 343–8.
vasculitis. Rheumatology (Oxford), 51, 634–43. Kothari, S.S. (2002). Takayasu’s arteritis in children—a review. Images in
Guzman, J., Fung, M., and Petty, R.E. (1994). Diagnostic value of Pediatric Cardiology, 9, 4–23.
anti-neutrophil and anti-endothelial cell antibodies in early Kawasaki Kumagai, N. and Ohno, S. (1996). Kawasaki disease. In Ocular immunity
disease. Journal of Pediatrics, 124, 917–20. and infection (ed. J.S. Pepose, G.M. Holland, and K.R. Wilhelmus), pp.
Hahn, J.S., Lannin, W.C., and Sarwal, M.M. (2004). Microangiopathy of 391–6. Mosby, St. Louis.
brain, retina and inner ear (Susac’s syndrome) in an adolescent female Kundell, S.P. and Ochs, H. D. (1980). Cogan Syndrome in childhood. Journal
presenting as acute encephalomyelitis. Pediatrics, 114, 276–81. of Pediatrics, 97, 96–8.
Hahn, H., Thomson, P.D., Kala, U., Beale, P.G., and Levin, S.E. (1998). A Lang, B.A., Yeung, R.S., Oen, K.G., et al. (2006). Corticosteroid treatment of
review of Takayasu’s arteritis in children in Gauteng, South Africa. refractory Kawasaki disease. Journal of Rheumatology, 33, 803–9.
Pediatric Nephrology, 12, 668–75. Lau, K.K., Suzuki, H., Novak, J., and Wyatt, R.J. (2010). Pathogenesis of
Harada, F., Sada, M., Kamiya, T., et al. (1986). Genetic analysis of Kawasaki Henoch-Schönlein purpura nephritis. Pediatric Nephrology, 25, 19–26.
syndrome. American Journal of Human Genetics, 39, 537–9. Liang, P. and Hoffman, G.S. (2005). Advances in the medical and surgi-
Hattori, M., Kurayama, H., and Koitabashi, Y. (2001). Antineutrophil cyto- cal treatment of Takayasu arteritis. Current Opinion in Rheumatology,
plasmic autoantibody-associated glomerulonephritis in children. 17, 16–24.
Journal of the American Society for Nephrology, 12, 1493–500. Lunardi, C., Bason, C., Leandri, M., et al. (2002). Autoantibodies to inner
Hautefort, C., Loundon, N., Montchilova, M., Marlin, S., Garabedian, E.N., ear and endothelial antigens in Cogan’s syndrome. Lancet, 360, 915–21.
and Ulinski, T. (2009). Mycophenolate as a treatment of steroid-resistant Lyons, P.A., Rayner, T.F., Trevedi, S., et al. (2012). Genetically distinct subsets
Cogan’s syndrome in childhood. International Journal of Pediatric with ANCA-associated vasculitis. New England Journal of Medicine, 367,
Otorhinolaryngology, 73, 1477–9. 214–23.
Henoch, E.H. (1974). About a peculiar form of purpura. Reprinted in Ma, E.H., Akikusa, J.D., MacGregor, D., Ng, J., and Su, J.C. (2011). Sweet’s
American Journal of Diseases of Children, 128, 78–9. syndrome with post-inflammatory elastolysis and Takayasu arteritis in
Hoffman, G.S. and Ahmed, A.E. (1998). Surrogate markers of disease activ- a child: a case report and literature review. Pediatric Dermatology, 29,
ity in patients with Takaysu arteritis. A  preliminary report from the 645–60.
International Network for the Study of Systemic Vasculitides. (INSSYS). Maeda, M., Kobayahi, M., Okamoto, S., et al. (1997). Clinical observation
International Journal of Cardiology, 66 (Suppl.1), S191–4. of 14 cases of childhood polyarteritis nodosa in Japan. Acta Paediatrica
Hoffman, G.S., Merkel, P.A., Brasington, R.D., Lenschow, D.J., and Lioang, P. Japonica, 39, 277–9.
(2004). Anti-tumor necrosis factor therapy in patients with difficult to Mahr, A.D., Neogi, T., and Merkel, P.A. (2006). Epidemiology of Wegener’s
treat Takayasu arteritis. Arthritis and Rheumatism, 50, 2296–304. granulomatosis. Lessons from descriptive studies and analyses of
Holman, R.C., Curns, A.T., Belay, E.D., Steiner, C.A., and Schonberger, L.B. genetic and environmental risk determinants. Clinical and Experimental
(2003). Kawasaki syndrome hospitalizations in the United States, 1997 Rheumatology, 24 (Suppl. 21), S82–91.
and 2000. Pediatrics, 112, 495–501. Masi S.T., Hunder, G.G., Lie, F.T., et  al. (1990). The American College of
Hoyer, B.F., Mumtaz, I.M., Loddenkemper, K., et al. (2012). Takayasu arteritis Rheumatology 1990 criteria for the classification of Churg-Strauss syn-
is characterized by disturbances of B cell homeostasis and responds to B drome (Allergic granulomatosis and angiitis). Arthritis and Rheumatism,
cell depletion therapy with Rituximab. Annals of the Rheumatic Diseases, 33, 1094–1100.
71,75–9. Mori, M., Imagawa, T., Hara, R., et al. (2012). Efficacy and limitation of inflix-
Huber, A.M., King, J., McLaine P., et  al. (2004). A randomized imab treatment for children with Kawasaki disease intractable to intra-
placebo-controlled trial of prednisone in early Henoch Schönlein pur- venous immunoglobulin therapy: Report of an open-label case series.
pura. BMC Medicine, 2, 7–14. Journal of Rheumatology, 39, 863–7.
Hui-Yuen, J.S., Duong, T.T., and Yeung, R.S.M. (2006). TNFα is necessary for Morishita, K., Li, S.C., Muscal, E., et al. (2012). Assessing the performance of
the induction of coronary artery inflammation and aneurysm formation the Birmingham Vasculitis Activity Score at diagnosis for antineutrophil-
in an animal model of Kawasaki disease. Journal of Immunology, 176, associated vasculitis in a registry for childhood vasculitis (ARChiVe).
6294–6301. Journal of Rheumatology, 29, 1088–94.
Hutchinson, C., Elbers, J., Halliday, W., et al. (2010). Treatment of small ves- Morishita, K.A., Rosendahl, K., and Brogan, P. (2011). Familial Takayasu
sel primary vasculitis in children: An open label cohort study. Lancet arteritis—a pediatric case and a review of the literature. Pediatric
Neurology, 9, 1078–84 Rheumatology, 9, 1–5.
Iqbal, H. and Evans, A. (2005). Dapsone therapy for Henoch-Schönlein pur- Motoyama, O. and Iitaka, K. (2005). Familial cases of Henoch-Schönlein pur-
pura. A case series. Archives of Disease of Childhood, 90, 985. pura in eight families. Pediatrics International, 47,612–15.
Jagiello, P., Aries, P., Arning, L., et al. (2005). The PTPN22 620W allele is a Meulders, Q., Pirson, Y., Cosyns, J.P., et al. (1994). Course of Henoch-Schönlein
risk factor for Wegener’s granulomatosis. Arthritis and Rheumatism, 52, nephritis after renal transplantation. Report of ten patients and review of
4039–43. the literature. Transplantation, 58, 1179–86.
CHAPTER 21 vasculitis in infancy, childhood, and adolescence 297

Munro-Faure, H. (1959). Necrotizing arteritis of the coronary vessels in Rich E.N. and Brown, K.K. (2012).Treatment of antineutrophil cytoplas-
infancy. Pediatrics, 23, 914–26. mic antibody- associated vasculitis. Current Opinion in Pulmonary
Nakamura, Y., Aso, E., Yashiro, M., et al. (2005). Mortality among persons Medicine,18, 447–54.
with a history of Kawasaki disease in Japan: can pediatricians safely dis- Rowley, A.H., Eckerley, C.A., Jack, H.M., et al. (1997). IgA plasma cells in vas-
continue follow-up of children with a history of the disease but without cular tissue of patients with Kawasaki syndrome. Journal of Immunology,
cardiac sequelae? Acta Paediatrica, 94, 429–34. 139, 5946–55.
Nash, M.C., Shah, V., Reader, J.A., and Dillon, M.J. (1995). Antineutrophil Rus, V. and Handwerger, B.S. (2000). Clinical value of antineutrophil cyto-
cytoplasmic antibodies and anti-endothelial cell antibodies are not plasmic antibodies. Current Rheumatology Reports, 2, 383–9.
increased in Kawasaki disease. British Journal of Rheumatology, Sano, T., Kurotobi, S., Matsuzaki, K., et  al. (2007). Prediction of
34, 882–7. non-responsiveness to standard high-dose gamma-globulin therapy in
Newburger, J.W., Takahashi, M., Burns, J.C., et  al. (1986). The treatment patients with acute Kawasaki disease before starting initial treatment.
of Kawasaki disease with intravenous gamma globulin. New England European Journal of Pediatrics, 166, 131–7.
Journal of Medicine, 315, 341–7. Saulsbury, F.T. (2009). Successful treatment of prolonged Henoch-Schönlein
Niaudet, P. and Habib, R. (1998). Methylprednisolone pulse therapy in the purpura with colchicine. Clinical Pediatrics, 48, 866–8.
treatment of severe forms of Schönlein-Henoch purpura nephritis. Saulsbury, F.T. (1999). Henoch-Schönlein purpura in children: Report of 100
Pediatric Nephrology, 12, 238–43. patients and review of the literature. Medicine (Baltimore), 78, 395–409.
Nussinovitch, N., Elishkevitz, K., Volovitz, B., and Nussinovitch, M. (2005). Savage, C.O., Winearls, C.G., Evans, D.J., et  al. (1985). Microscopic pol-
Hypertension as a late sequela of Henoch-Schönlein purpura. Clinical yarteritis: presentation, pathology and prognosis. Quarterly Journal of
Pediatrics (Philadelphia), 44, 543–7. Medicine, 56, 467–83.
Ogata, S., Ogihara, Y., Nomoto, K., et al. (2011). Clinical score and tran- Schlieben, D.J., Korbet, S.M., Kimura, R.E., Schwartz, M.M., and Lewis,
script abundance patterns identify Kawasaki disease patients who may E.J. (2005). Pulmonary-renal syndrome in newborn with placental
benefit from addition of methylprednisolone. Pediatric Research, 66, transmission of ANCAs. American Journal of Kidney Diseases, 45,
577–84. 758–61.
O’Mahony, J., Bar-Or, A., Arnold, D.L., et al. (2013). Masquerades of acquired Schönlein, J.L. (1837). Allgemeine unde Specielle Pathologie und Therapie, 3rd
demyelination in children: Experience in a national demyelinating dis- edn. Literatur-Vomptoinr, Herisau, Germany.
ease program. Journal of Child Neurology, 28, 184–97. http://jcn.sage- Seko, Y. (2000). Takayasu arteritis. Insights into immunopathology. Japanese
pub.com/content/early/2012/05/07/0883073812443006 Heart Journal, 41, 15–26.
Onouchi, Y., Gunji, T., Burns, J.C., et al. (2008). ITPKC functional polymor- Shin, J.I., Park, J.M., Kim, J.H., et al. (2005a). Can azathioprine and steroids
phism associated with Kawasaki disease susceptibility and formation of alter the progression of severe Henoch-Schönlein nephritis in children?
coronary artery aneurysms. Nature Genetics, 40, 35–42. Pediatric Nephrology, 20, 1087–92.
Onouchi, Y., Ozaki, K., Burns, J.C., et al. (2012). A genome-wide associa- Shin, J.L., Park, J.M., Shin, Y.H., et al. (2005b). Cyclosporin A therapy for
tion study identifies three new risk loci for Kawasaki disease. Nature severe Henoch-Schönlein nephritis with nephrotic syndrome. Pediatric
Genetics, 44, 517–21. Nephrology, 20, 1093–7.
Orsoni, J.G., Zavota, L., Vincenti, V., Pellistri, I., and Rama, P. (2004). Cogan Shrivasatava, S. (1995). Takayasu’s arteritis. American Heart Journal, 129,
syndrome in children: Early diagnosis and treatment is critical to prog- 1228–9.
nosis. American Journal of Ophthalmology, 137, 757–8. Sleeper, L.A., Minich, L.L., McCrindle, B.M., et  al. (2011). Evaluation of
Ozaltin, F., Bakkaloglu, A., Ozen, S., et al. (2004). The significance of IgA Kawasaki disease risk scoring systems for intravenous immunoglobulin
class of antineutrophil cytoplasmic antibodies (ANCA) in childhood resistance. Journal of Pediatrics, 158, 831–5.
Henoch-Schönlein purpura. Clinical Rheumatology, 23, 426–9. Smith, L.B., Newburger, J.W., and Burns, J.C. (1989). Kawasaki syndrome and
Ozen, S., Anton, J., Arisoy, N., et al. (2004). Juvenile polyarteritis: results the eye, Pediatric Infectious Disease Journal, 8, 116–8.
of a multicenter survey of 110 children. Journal of Pediatrics, 145, Son, M.B., Gauvreau, K., Burns, J.C., et al. (2011). Infliximab for intravenous
517–22. immunoglobulin resistance in Kawasaki disease: A retrospective study.
Ozen, S., Pistorio, A., Iusan S., et al. (2010). EULAR/PRINTO/PReS crite- Journal of Pediatrics, 158, 644–9.
ria for Henoch Schönlein purpura, childhood polyarteritis nodosa, Stanley, P., Roebuck, D., Barboza, A., et al. (2003). Takayasu’s arteritis in chil-
childhood Wegener granulomatosis and childhood Takayasu arteri- dren. Techniques in Vascular and Interventional Radiology, 6, 158–68.
tis: Ankara 2008. Final classification criteria. Annals of the Rheumatic Steffanson, T.V., Kolka, R.S., Sigurdardottir, S.L., et al. (2005). Increased fre-
Diseases, 69, 798–06. quency of C4B*Q0 alleles in patients with Henoch-Schönlein purpura.
Ozen, S., Ruperto, N., Dillon M.J., et al. (2006). EULAR/PReS endorsed con- Scandinavian Journal of Immunology, 61, 274–8.
sensus criteria for the classification of childhood vasculitides. Annals of Suda, K., Iemura, M., Nishiono, H., et  al. (2011). Long-term prognosis of
the Rheumatic Diseases, 65, 936–41. patients with Kawasaki disease complicated by giant coronary aneu-
Peco-Antic, A., Bonaci-Nikolic, B., Basta-Javonavic, G., et  al. (2006). rysms. Circulation 123, 1836–42.
Childhood polyangiitis associated with MPO-ANCA. Pediatric Suddleson, E.A., Reid, B., Woolley, M.M., and Takahashi, M. (1987). Hydrops
Nephrology, 21, 46–53. of the gallbladder associated with Kawasaki syndrome. Journal of
Pistracher, K., Gellner, V., Riegler, S., Schockler, B., Scarpatetti, M., and Pediatric Surgery, 22, 956–9.
Kurschel, S. (2012). Cerebral hemorrhage in the presence of primary Sundel, R.P., Baker, A.L., Fulton, D.R., and Newburger, J.W. (2003).
childhood central nervous system vasculitis. A review. Childs Nervous Corticosteroids in the initial treatment of Kawasaki disease: report of a
System, 28, 1141–8. randomized trial. Journal of Pediatrics, 142, 611–6.
Porcalla, A.R., Sable, C.A, Patel, K.M., et  al. (2005). The epidemiology of Szer, I. (1996). Henoch-Schönlein purpura: when and how to treat. Journal of
Kawasaki disease in an urban hospital; does African American race Rheumatology, 23, 1661–5.
protect against coronary artery aneurysms? Pediatric Cardiology, 26, Tacke, E., Kuipers, I.M., Groenink, M., Spijkerboer, A.M., and Kuijpers, T.W.
775–81. (2011). Cardiac magnetic resonance imaging for non-invasive assess-
Rahman, F.Z., Takhar, G.K., Roy, O., Shepherd, A., Bloom, S.L., and ment of cardiovascular disease during the follow-up of patients with
McCartney, S.A. (2010). Henoch-Schönlein purpura complicating adali- Kawasaki disease. Circulation Cardiovascular Imaging, 4, 712–20.
mumab therapy for Crohn’s disease. World Journal of Gastrointestinal Trapani, S., Micheli, A., Grisolia, F., et  al. (2005). Henoch-Schönlein pur-
Pharmacology and Therapy, 6, 119–22. pura in childhood: Epidemiological and clinical analysis of 150 cases
298 SECTION 5 vasculitic diseases and syndromes and related disorders

over a 5-year period and review of literature. Seminars in Arthritis and Yanagawa, H., Nakamura, Y., Ojima, T., et al. (1999). Changes in epidemic
Rheumatism, 35, 143–53. patterns of Kawasaki disease in Japan. Pediatric Infectious Disease
Trygstad, C.W. and Stiehm, E.R. (1971). Elevated IgA globulin in anaphylac- Journal, 18, 64–6.
toid purpura. Pediatrics, 476, 1023–8. Yanagawa, H., Nakamura, Y., Yashiro, M., et al. (2001). Incidence survey of
Twilt, M. and Benseler, S.M. (2012). The spectrum of CNS vasculitis in chil- Kawasaki disease in 1997 and 1998 in Japan. Pediatrics, 107, E33.
dren and adults. Nature Reviews Rheumatology, 8, 97–107. Yanagawa, H., Yashiro, M., Nakamura, Y., et  al. (1995). Nationwide sur-
van der Woude, F.J., Rasmussen, N., Lobatto, S., et al. (1985). Autoantibodies veillance of Kawasaki disease in Japan, 1984-1993. Pediatric Infectious
against neutrophils and monocytes:  tool for diagnosis and Diseases Journal, 14, 69–71.
marker of disease activity in Wegener’s granulomatosis. Lancet, Yang, Y.-H., Hung, C.F., Hsu, C.R, et al. (2005). A nationwide survey on epi-
1, 425–9. demiological characteristics of childhood Henoch-Schönlein purpura in
Watanabe, T. (2011). Henoch-Schönlein purpura following influenza vac- Taiwan. Rheumatology (Oxford), 44, 618–22.
cinations during the pandemic of influenza A  (H1N1). Pediatric Yang, Y.-U., Wang, S.J., Chuang, Y.-H., et al. (2002). The level of IgA anti-
Nephrology, 26, 795–8. bodies to human umbilical vein endothelial cells can be enhanced by
Watson, W, Richardson, A.R.W., Holt, R.C.L., Jones, C.A., and Beresford, TNF-α treatment in children with Henoch-Schönlein purpura. Clinical
M.W. (2012). Henoch Schonlein purpura.—A 5-year review and pro- and Experimental Immunology, 130, 352–7.
posed pathway. PLoS ONE, 7, e29512. Yeung, R.S.M., (2004). The etiology of Kawasaki disease:  a superantigen-
Watts, R.A., Lane, S.E., Bentham, G., and Scott, D.G.I. (2000). Epidemiology mediated process. Progress in Pediatric Cardiology, 19, 109–13.
of systemic vasculitis. Arthritis and Rheumatism, 43, 414–9. Young, H.L., Sung, J.C., Jong, D.J., and Gwan, G.S. (2012). Associations
Xie, L., Cai, Y., Bao, X, and Cao, L. (2009). Leflunomide for the successful between the angiotensin converting enzyme insertion/deletion poly-
management of juvenile Cogan’s syndrome. Clinical Rheumatology, 28, morphism and susceptibility to vasculitis: a meta-analysis. Journal of the
1453–5. Renin-Angiotensin- Aldosterone System, 13, 196–201.
Yajima, M., Numsno, F., Park, Y.B., and Sugar, S. (1994). Comparative Zwerina, J., Eger, G., Englbrecht, M., Manger, B., and Schott, G. (2008).
studies of patients with Takayasu arteritis in Japan, Korea and India. Churg-Strauss syndrome in childhood: A systematic literature review
Comparison of clinical manifestations, angiography, and HLA-B enti- and clinical comparison with adult patients. Seminars in Arthritis and
gen. Japanese Circulation Journal, 58, 9–14. Rheumatism, 39, 108–15.
CHAPTER 22

Assessment of disease
activity and damage
Raashid Luqmani

Introduction been, translated into clinical practice. We now have standards and
guidelines for the routine treatment for these less common but not
Purpose of assessment rare and potentially life-threatening diseases (Lapraik et al. 2007;
The complexity of the systemic vasculitides means that a structured Mukhtyar et al. 2009a; Mukhtyar 2009b). With the increasing use
approach to evaluation is important; in turn, it provides a rational of expensive biological therapies, funding agencies will be looking
basis to determine a treatment strategy. The clinical manifestations for evidence of response to treatment in order to justify the cost of
in patients with vasculitis are heterogeneous; as a result, patients treatment. Providing a robust framework for assessment of disease
may initially present to a wide variety of different specialists, is going to become increasingly important to ensure that patients
depending on their initial clinical manifestation. Over time, the receive the most appropriate care.
initial manifestations may be supplemented by more widespread
features (for example, patients with granulomatosis with polyangii- Concepts of assessment
tis might notice nasal crusting and discharge as the first features, Definitions of disease state should be reproducible, so that they
and subsequently develop hearing loss, or lung or renal disease). can be compared within the same patient over a period of time
In patients with established disease, despite initial good control, as well as across centres to provide a reliable way of documenting
relapses are common especially in small-vessel vasculitis. In other and evaluating progress, in order to assist in making any necessary
words, with or without treatment we should expect the clinical fea- adjustments to treatment for individual patients, and for measuring
tures to vary and we should be able to measure this change in order the status of patients for clinical trials and studies. Measurement
to make appropriate therapeutic decisions. Untreated multisystem of disease activity is often the starting point for treatment, which
vasculitis is usually fatal. The situation is now very different because is aimed at reducing activity as much as possible. We know that
we have effective therapies such as cyclophosphamide or rituximab, untreated multisystem vasculitis with very active disease will be
as a result of which, most patients survive their initial presenta- fatal. Most patients survive their vasculitis following initial therapy,
tion. However, we have not yet managed to cure the disease in the only to face the consequences of the long-term morbidity of vascu-
majority. Instead, these patients suffer chronic relapsing remitting litis. This includes relapses of disease activity, but also scarring from
episodes, requiring long-term care and follow-up. Therapies for vas- the disease itself, organ impairment or damage, and potential for
culitis are successful in achieving short-term control of disease but infection which is typically associated with drug toxicity, especially
unfortunately patients are left with significant amounts of disease glucocorticoids. A variety of adverse events occur, including weight
burden because of chronic, grumbling disease and risk of relapse. gain, hypertension, osteoporosis, and an increased risk of infec-
In addition, however, the therapies used to treat their condition, tion and cancer. In addition, patients may develop new or wors-
including glucocorticoids and other immunosuppressive agents, ened co-existing co-morbid conditions following the diagnosis of
can result in drug toxicity, which will add to their overall burden. vasculitis. For example, patients with previously stable ischaemic
For these reasons, it is necessary to regularly assess the status of heart disease may find that this becomes unstable after the diag-
the patient; we recommend that this is done in a formal, structured, nosis of vasculitis and they may be at greater risk of future myo-
standardized way, as described in this chapter. We need to determine cardial infarction. Managing each patient requires careful attention
how much of the patient’s current problem is due to active disease, to detail to ensure that each aspect of their condition, that is their
how much damage has occurred, how much co-morbidity (either disease activity, the damage, and the co-morbidity, are separately
directly or indirectly related to vasculitis or completely unrelated to assessed and separately treated. Indeed, severity of disease is partly
it), and how the patient is functioning overall. Measurement of the attributed to disease activity and partly to the subsequent damage
chronic morbidity and damage is sometimes as important as meas- that accumulates. Damage is emerging as an important concept in
uring disease activity because it will assist in the overall manage- chronic diseases such as vasculitis. Both activity and damage con-
ment of patients. A structured approach has facilitated the design of tribute to prognosis but damage probably contributes more pow-
clinical trials of different therapeutic agents in vasculitis; eventually, erfully to long-term outcome, especially when it involves multiple
the results of these trials can be, and in many cases have already organs (Exley et al. 1998). In vasculitis, it is common for multiple
300 SECTION 5 vasculitic diseases and syndromes and related disorders

sites to be involved and therefore an overall structured evaluation there is less certainty about the diagnosis, these instruments can be
is more useful than a focus on a single organ such as the kidney. very valuable in determining a treatment plan.
A patient’s functional outcome from treatment of vasculitis There are problems with generic assessments of disease activity
is very individual in the sense that variable amounts of disease when comparing between different diseases. For example, patients
activity and damage can have vastly different impacts on patients with GPA with multiorgan involvement require many different
in terms of their ability to function in their daily lives. Although aspects of disease to be recorded; therefore the total quantitative
generic instruments such as Short Form 36 can be applied (Walsh score from such patients is likely to be higher than in patients who
et al. 2011), this area of functional outcome is being further evalu- present with giant cell arteritis (GCA), which predominantly affects
ated in vasculitis to develop assessment tools that can accurately the vessels of the head and neck. Although GCA can give rise to
record changes in functional status. All of these aspects (namely systemic features such as weight loss and fever, it is unlikely to be
activity, damage, and patient function) ultimately contribute to the associated with organ-specific features in most cases. Similarly
overall outcome and prognosis. Takayasu’s arteritis affects the great vessels, but the clinical expres-
Measurement of morbidity in vasculitis can be performed using sion of the disease is often limited in terms of recorded items of
clinical assessment as an effective surrogate. Robust clinical instru- disease activity. These patients often present with a long-standing
ments have been developed and can serve as the basis to compare history of ischaemic claudication on movement of limbs and may
development of any new laboratory biomarker or to test existing be found to have lost pulses but other specific organ involvement
potential biomarkers. However, the clinical state of the patient is rare. As a consequence, the range of scores in a patient with
remains a gold standard when deciding therapeutic changes at Takayasu’s arteritis will be much narrower and smaller than the
present, although there is a promise of potential biomarkers to range of scores achieved in patients with active GPA. The Indian
determine prognosis based on more recent findings of changes of Takayasu Arteritis Score (ITAS) has been developed from BVAS,
expression of genes in T-lymphocyte subsets during the course of but with a much greater emphasis on clinical aspects of large-vessel
antineutrophil cytoplasm antibody (ANCA) associated vasculi- abnormalities (Bacon et  al. 2012). The debate continues around
tis (McKinney et al. 2010). Laboratory markers can suggest that a whether disease-specific instruments should be developed for
change in clinical status has occurred, but they can also be very each of the individual vasculitides or whether generic instruments
misleading; for example elevations of ESR and CRP can occur due should be used for all.
to infection as opposed to a flare of vasculitis and therefore cannot One of the major benefits of having a structured disease evalu-
be used as direct indicators of disease activity. In such cases, it is ation in vasculitis is to allow successful implementation of clinical
necessary to re-evaluate patients to assess their clinical state rather trials in this relatively uncommon set of diseases because it requires
than making a judgement that is based entirely on the laboratory collaboration between centres, as well as between countries, in
abnormality. order to test enough patients with the condition. Standardization
across centres is essential and therefore a structured assessment
Benefits of assessment tool for vasculitis also offers a training opportunity for physicians
Prior to starting treatment it is important to perform a detailed in different centres, and also in different specialities, to assess vas-
clinical evaluation of potentially complicated and life-threatening culitis. This can be extrapolated to clinical practice, offering train-
conditions. This can be undertaken even if we are unsure of exactly ing in the diversity of disease activity and damage seen in vasculitis
which type of vasculitis the patient has, because the assessment will and can be used to achieve greater understanding both of the con-
provide essential information on how to manage the patient. There dition and its management. Validated instruments are available
is a lot of overlap between different types of vasculitis so impor- and we strongly recommend their use in any clinical studies of
tant themes in clinical assessment emerge; generic instruments to vasculitis; the Outcome Measures in Rheumatology Clinical Trials
measure disease activity have been developed and are very use- (OMERACT) group has recently endorsed the use of structured
ful as practical tools for evaluating patients. Some tools are more disease assessment in all forms of vasculitis (Merkel et al. 2011).
specific for one disease, such as the Disease Extent Index (DEI) Routine evaluation of patients with structured assessments will
for granulomatosis with polyangiitis (De Groot et  al. 2001)  or be increasingly important in order to justify the use of expensive
the Birmingham Vasculitis Activity Score for Wegener’s granulo- biological agents, which are likely to play a greater role in disease
matosis (BVAS/WG) applied to granulomatosis with polyangiitis management in future.
(Stone et al. 2001). These two instruments have been introduced
for a variety of research reasons. The DEI was mainly developed Uses of disease activity assessment
for long-term outcome studies in GPA in order to document the
granulomatosis manifestations in that condition. BVAS WG was in vasculitis
developed for a particular study of patients with GPA and was vali- The main use of a disease activity index in vasculitis should be to
dated only for that disease; however, they are inherently less valua- provide a detailed description of the state of disease activity. This is
ble in assessing other forms of vasculitis, particularly other forms of based on the premise that the clinician evaluating the patient has
ANCA-associated vasculitis. By comparison, generic instruments the relevant training or experience in vasculitis and the ability to
will allow descriptions of patients where the precise sub-type of accurately document disease activity. It is also possible to use such
vasculitis is not certain or shares features across different sub-types. an instrument to help train individuals in disease assessment. The
For example, many patients with GPA and MPA will have exactly current recommended disease assessment activity tools provide
the same range of manifestations in the lung and other systemic better insight into the spectrum of manifestations occurring across
organs, the exception being the lack of involvement of the upper different body systems to act as an aide memoire for the clinician to
airways in MPA in most cases. Early on in the disease course where enquire about potential disease activity in each patient. Instruments
CHAPTER 22 assessment of disease activity and damage 301

can be used to derive qualitative evaluation but also quantitative deciding if items are present and if they are due to active vasculitis.
measurement of disease activity, which has been used in studies. Points are given for each item with a maximum number of points
When the index is applied to patients, it is necessary to have some per body system and a maximum total score of 63. Items are only
training in the use of the instrument and to understand the con- recorded if they can be attributable to active vasculitis. The more
cepts of the decision making behind each item. Generic instru- specific measures for GPA, namely BVAS WG and DEI, may also be
ments such as the BVAS (Luqmani et al. 1994) and its subsequent used in GPA, and DEI is looking useful as a measure of prognosis.
versions provide an overall assessment. BVAS is based on record- However, for the majority of randomized control trials of therapy,
ing systemic features and symptoms and signs of abnormalities in BVAS is the most widely adopted assessment tool.
eight different organ systems: skin, mucus membrane and eyes, ears
nose and throat, chest, cardiovascular system, gastrointestinal tract, Laboratory tests
kidney, and nervous system (a list of items from BVAS is shown in
Table 22.1). BVAS is based on clinician assessment of patients and There is no gold standard clinical or laboratory test to measure dis-
ease status in vasculitis. Serological tests are sometimes unreliable in
evaluating patients because they can be affected by other conditions,
Table 22.1 Items of activity in systemic vasculitis such as the presence of infection or other inflammatory causes; this
particularly applies to the CRP but also the ESR. Unfortunately,
1. General 6. Cardiovascular patients with vasculitis are more prone to infection, which can
aggravate the situation. In addition, infection can trigger episodes of
Myalgia Loss of pulses
Arthralgia / arthritis Valvular heart disease vasculitis. In patients with ANCA-associated vasculitis the diagno-
Fever >38°C Pericarditis sis can be supported by the presence of ANCA but the variation in
Weight loss >2 kg Ischaemic cardiac pain ANCA during the course of disease is considerable and often does
Cardiomyopathy not relate to clinical manifestations. In up to 40% of cases ANCA
Congestive cardiac failure can vary without clinical manifestations changing. We would there-
2. Cutaneous 7. Abdominal fore strongly argue that changes should not be made on the basis of
Infarct Peritonitis
change in ANCA status, although change should trigger more clini-
Purpura Bloody diarrhoea cal watchfulness especially if patients are going from low titre to high
Ulcer Ischaemic abdominal pain titre or from ANCA negative to ANCA positive. However, patients
Gangrene who present with ANCA initially and who after clinical remission
Other skin vasculitis still have a positive ANCA are more likely to relapse; therefore these
3. Mucous membrane/eyes 8. Renal patients should have their induction therapy continued rather than
switched (Slot et al. 2004). Other laboratory markers of vasculitis are
Mouth ulcers Hypertension
Genital ulcers Proteinuria >1+ being developed and tested but none have so far proven to be reli-
Adnexal inflammation Haematuria ≥10 RBCs/hpf able and effective in determining disease activity.
Significant proptosis Elevated serum creatinine
Scleritis/ episcleritis Significant rise in serum Imaging assessment
Conjunctivitis/ blepharitis/ keratitis creatinine
Blurred vision This will be discussed further in the chapters on large-vessel vasculi-
Sudden visual loss tis, but there is growing use of imaging in evaluation of patients with
Uveitis large-vessel disease. Ultrasound imaging of temporal and auxiliary
Retinal changes (vasculitis/ arteries is a promising non-invasive technique for diagnosis of GCA,
thrombosis/ exudate/ haemorrhage) which may avoid the need for biopsy (Karassa et al. 2005) but still
4. ENT 9. Nervous system needs to be fully evaluated. Whether serial measurement of ultra-
Bloody nasal discharge/ crusts/ ulcers/ Headache sound appearance of temporal arteries following treatment can guide
granulomata Meningitis more effective treatment changes is uncertain. For more widespread
Paranasal sinus involvement Organic confusion aortic involvement, MRI scanning of the great vessels provides evi-
Subglottic stenosis Seizures (not hypertensive) dence of wall abnormalities but does not distinguish active from
Conductive hearing loss Cerebrovascular accident inactive disease. The most effective tool for this is CT PET scanning,
Sensorineural hearing loss Spinal cord lesion which can highlight areas of increased glucose uptake indicating
Cranial nerve palsy vessel wall active inflammation (Luqmani 2012). Unfortunately, the
Sensory peripheral neuropathy
technique involves a lot of radiation (an average of 14 millisieverts
Mononeuritis multiplex
per PET CT scan) and therefore would be limited in use as a serial
5. Chest measure, although it is likely to be helpful (Henes et al. 2011).
Wheeze
Nodules or cavities
Pleural effusion/ pleurisy Disease damage and assessment
Infiltrate Damage is a term we use to mean the accumulation of chronic
Endobronchial involvement scarring as a result of vasculitis. The scarring may have been due
Massive haemoptysis/ alveolar to the vasculitis itself or its treatment or co-morbidity. Overall, we
haemorrhage
acknowledge damage as being an inevitable consequence for the
Respiratory failure
majority of patients with vasculitis. It indirectly measures impact of
302 SECTION 5 vasculitic diseases and syndromes and related disorders

the disease and its treatment and the patient’s physiological status Table 22.2 Items of damage in systemic vasculitis
but it is different from their functional outcome. Conceptually, we
regard the scar in damage as permanent, even if the patients resulting 1. Musculoskeletal 7. Peripheral vascular disease
clinical problem seems to resolve or is compensated for. The concept Significant muscle atrophy or Absent pulses in one limb
of damage is that it represents an accumulation of morbidity that weakness Major vessel stenosis
the patient suffers; it should be regarded as a permanent change in Deforming/ erosive arthritis Claudication >3 months
status. For example, if a patient had a myocardial infarction during Osteoporosis/ vertebral collapse Minor tissue loss
the course of their diagnosis of vasculitis but made a full recovery, Avascular necrosis Major tissue loss
Osteomyelitis Complicated venous thrombosis
there would still be a scar in their heart and we would therefore still
regard that as permanent damage even if they have made a full func- 2. Skin / mucous membranes 8. Gastrointestinal
tional recovery. The vasculitis damage index, the VDI, is the most Alopecia Gut infarction/ resection
widely used measure to document damage in vasculitis (Exley et al. Cutaneous ulcers Mesenteric insufficiency/
1997a). The items in the damage index are listed in Table 22.2. The Mouth ulcers pancreatitis
VDI is validated as a measure of permanent scarring and is based Chronic peritonitis
on multiorgan involvement, collecting information on potentially Oesophageal stricture/ surgery
damage-related items since the diagnosis of vasculitis, or deterio- 3. Ocular 9. Renal
ration as a result of the diagnosis of vasculitis and its treatment Cataract Estimated / measured
(Bhamra and Luqmani 2012). This would include the effects of the Retinal change GFR ≤50%
disease, its treatment, and intercurrent (sometimes coincidental but Optic atrophy Proteinuria ≥0.5 g/24 h
sometimes indirectly related) illness. We do not attempt to discrimi- Visual impairment/ diplopia End-stage renal disease
nate damage due to vasculitis from damage due to any other cause; Blindness
in practice this is often impossible to achieve and there are usually a Orbital wall destruction
number of reasons that explain why damage may have occurred (e.g. 4. ENT 10. Neuropsychiatric
active vasculitis and its treatment and infection and co-morbidity to Hearing loss Cognitive impairment
a greater or lesser extent). Items in the damage index are selected by Nasal blockade/ chronic discharge/ Major psychosis
consensus of expert physicians managing vasculitis and the index is crusting Seizures
a validated means of documenting morbidity in vasculitis. It prob- Nasal bridge collapse/ septal Cerebrovascular accident
ably has a greater prognostic value than the disease activity tool perforation Cranial nerve lesion
(BVAS) and it potentially serves as a surrogate outcome measure in Chronic sinusitis/ Peripheral neuropathy
radiological damage Transverse myelitis
clinical trials. Perhaps we should be considering the use of a dam-
Subglottic stenosis with or
age index as a primary, or at least a secondary, outcome measure in without surgery
more clinical trials, especially given the improvements in managing
disease activity achieved from more recent studies. In turn, it means 5. Pulmonary 11. Other
that we should consider other aspects of patients’ outcome such as Pulmonary hypertension Gonadal failure
damage when judging the relative merits of different drug regimens. Pulmonary fibrosis Marrow failure
Pulmonary infarction Diabetes
Pleural fibrosis Chemical cystitis
Use of the damage index Chronic asthma Malignancy
If we measure the amount of damage serially over the course of a Chronic breathlessness
patient’s illness, damage often occurs early in the disease process Impaired lung function
and it may increase after relapse. Early damage, especially in the first 6. Cardiovascular
6 months, has a significant impact on later mortality. It is unusual for Angina/ angioplasty
patients not to have damage; the vast majority of patients continue to Myocardial infarction
accumulate damage even though they have effective disease control Cardiomyopathy
(Seo et al. 2005). Indeed, in GPA, a number of patients may already Valvular disease
have developed damage before they are diagnosed, because of the long Pericarditis ≥3 months or
duration of features before their condition is recognized. In some stud- pericardectomy
ies over two-thirds of patients had at least two damaged organ systems Diastolic BP ≥95 or
(Exley et al. 1997b); a large study has shown that one third of patients requiring antihypertensives
have at least 5 items of damage, 7 years after diagnosis (Robson et al
2013). Recently, the damage index has been compared with the new
a patient has a good or a bad prognosis so that we can measure
instrument, the combined damage assessment (CDA) tool, which has
the effects of therapy and also consider stratifying patients with
almost double the number of items compared with the original dam-
the worst prognosis to more aggressive therapy. Some trials have
age index; however, the original VDI was found to be more effective
been designed on the basis for prognosis, giving more treatment to
and more reproducible than the CDA (Suppiah et al. 2011a).
patients with worse indicators of poor prognosis. Prognosis tools for
vasculitis are available. The two main ones are the Five Factor Score
Predicting prognosis (Guillevin et al. 1988; Guillevin et al. 1996; Guillevin et al. 2011) and
Patients want to know what their prognosis is when they are diag- the BVAS (Luqmani et al. 1994). The Five Factor Score is a simple
nosed with vasculitis. In clinical trials it is useful to know whether prognostic tool, initially validated in patients with polyarteritis
CHAPTER 22 assessment of disease activity and damage 303

nodosa (PAN) and eosinophilic granulomatosis with polyangiitis, A paediatric version of the VDI is also under development and
based on the extent of serious organ involvement. It has been used as it is likely to be used more widely in paediatric vasculitis in the
the basis of developing trial protocols depending on disease severity, next few years. Variations are applied similar to those applied
that is patients who either have at least one Five Factor Score or no in PVAS in terms of changing items to be more appropriate for
factors have been stratified to either receiving cyclophosphamide in children, for example the creatinine levels, the degree of hyper-
addition to steroids, or steroids alone. BVAS also provides prognos- tension, as well as paediatric damage such as growth failure and
tic information and is effective in a number of studies in vasculitis; delayed puberty.
high BVAS at diagnosis predicts poor subsequent outcome in terms
of mortality but it also predicts responsiveness to therapy (Gayraud Further testing in patients with
et al. 2001; Koldingsnes and Nossent 2002). However, the damage
index, VDI, is probably the most accurate predictor for outcome that
systemic vasculitis
we have. Multiorgan, multisystem involvement in patients in the Most patients with systemic vasculitis can be comprehensively
first 2 years of disease as measured by the VDI is a very strong pre- evaluated by a clinician and a limited set of tests, but on occasion
dictor of risk of death. Patients with more than five items on the VDI further testing is required. This is often in order to determine not
scale by 6 months have a subsequent risk of death of 17 : 1 compared necessarily what the diagnosis is, but whether complications have
to patients with five or less items on the VDI. This is even higher arisen or whether co-morbidities exist. This does not undervalue
when critical items are involved, such as those in the cardiovascular the clinical evaluation, which remains central to the assessment and
or neurological system (Exley et al. 1998). Understandably, patients management of patients; however, supplementary tests will sup-
who accumulate more damage rapidly are more likely to die. port or refine the assessment of disease state and therefore should
A group from Cambridge has looked at gene expression in T cells be included where appropriate. This allows specific tailoring of
from patients with active vasculitis and active lupus. The findings patients’ individual needs, in particular to avoid unnecessary use of
suggest that some genes are over-expressed in patients with vas- immunosuppressive therapy for too long but to introduce it appro-
culitis who will subsequently go on to relapse. These appear to be priately and quickly where necessary.
expressed independently of current disease state; in other words,
they act as true prognostic markers to determine who is going Long-term outcome in vasculitis
to flare or not. Currently, this has only been shown in one centre
The vasculitides have changed from life-threatening short-lived
and does require validation to confirm this finding but potentially
diseases into ones that are characterized by chronic morbid-
holds promise for a biomarker that might predict worse activity and
ity and long-term survival, albeit with shortened life expectancy
could be used as a basis for stratifying therapy in future (McKinney
compared to the general population. The same is true in other
et al. 2010).
inflammatory diseases such as rheumatoid arthritis and SLE. The
long-term risks for such patients are determined by endothe-
Disease states lial dysfunction increasing the risk of cardiovascular disease but
In clinical trials it is essential to have clear definition of disease also the increased risk of infection from therapy and possibly the
states to allow patients to be precisely placed in different catego- increased risk of cancer in patients given toxic regimens. The latter
ries, such as active disease, relapse, remission, or partial remis- is unclear because patients with vasculitis will themselves have an
sion. A further qualification is response to treatment as opposed increased risk of cancer, which may have occurred simultaneously
to achieving remission, because for some resistant forms of vas- with the vasculitis or in some cases prior to it. We know that the
culitis it is often only possible to partially improve rather than risk of cardiovascular events is particularly high in older patients
completely resolve disease activity. Definitions have been inter- who have baseline hypertension and MPO ANCA status (Suppiah
preted using instruments such as BVAS and it is recommended et al. 2011b). Figure 22.1 suggests a schematic relationship between
that a structured scale such as this is used as a basis of these disease the different aspects of vasculitis, their treatment, and the potential
states (Hellmich et al. 2007); indeed, the OMERACT definitions outcomes.
of disease states have ratified the recommendations from EULAR
(Merkel et al. 2011). Summary
We have vastly improved the care of patients’ systemic vasculitis
Paediatric vasculitis with modern therapy. We continue to strive to improve patient
Children with vasculitis are less common than adults but neverthe- outcome, as we improve our treatment strategies from prevent-
less there is a growing interest in developing assessments in pae- ing death to trying to cure disease and minimize sequelae of dis-
diatric vasculitis akin to those published in adults, using a similar ease and its treatment. In order to measure whether or not we
but modified approach. BVAS performs reasonably well in child- are succeeding in these aspirations, it is essential to have a robust
hood vasculitis (Demirkaya et al. 2012). The paediatric vasculitis system to document changes that truly reflect the overall patient
activity score (PVAS) has recently been validated as an instrument experience of disease, so that we can better discriminate between
for childhood vasculitis. It is largely based on the adult BVAS but different treatment regimens. The ideal set of measures would be
individual items have been changed where appropriate for children easy to perform, be robust, and provide an accurate prediction
and new items have been introduced, which are more specific to of future risk. Whilst we are still a long way from these goals,
paediatric vasculitis (Dolezalova et al. 2013). It remains a generic our current imprecise clinical tools remain a major advance in
instrument applied across different forms of vasculitis and will be our ability to support therapeutic decision making for this group
used in clinical trials in paediatric vasculitis in future. of life-threatening and organ-threatening diseases, especially
304 SECTION 5 vasculitic diseases and syndromes and related disorders

Active vasculitis

Death Immunosuppression

Organ failure
Remission

Reduced Function
Damage

Fig. 22.1 Complex relationship between vasculitis, therapy, and outcome. Dashed lines represent speculative relationships.

as we begin to discover the potential benefits of very expensive Churg-Strauss angiitis:  a study in 165 patients. British Journal of
biological therapies. Rheumatology, 27, 258–64.
Guillevin, L., Lhote, F., Gayraud, M., et al. (1996). Prognostic factors in pol-
yarteritis nodosa and Churg-Strauss syndrome. A prospective study in
References 342 patients. Medicine (Baltimore), 75, 17–28.
Bacon, P.A., Sivakumar, R., Danda, D., and Misra, R. (2012). Vasculitis assess- Guillevin, L., Pagnoux, C., Seror, R., et al.; French Vasculitis Study Group
ment and Takayasu aorto-arteritis. Indian Journal of Rheumatology, (FVSG) (2011).The Five-Factor Score revisited: assessment of prognoses
7, 153–8. of systemic necrotizing vasculitides based on the French Vasculitis Study
Bhamra, K., Luqmani, R. (2012). Damage assessment in ANCA-associated Group (FVSG) cohort. Medicine (Baltimore), 90, 19–27.
vasculitis. Current Rheumatology Reports, 14, 494–500. Hellmich, B., Flossmann, O., Gross, W.L., et al. (2007). EULAR recommen-
de Groot, K., Gross, W.L., Herlyn, K., and Reinhold-Keller, E. (2001). dations for conducting clinical studies and/or clinical trials in systemic
Development and validation of a disease extent index for Wegener’s vasculitis: focus on anti-neutrophil cytoplasm antibody-associated vas-
granulomatosis. Clinical Nephrology, 55, 31–8. culitis. Annals of the Rheumatic Diseases, 66, 605–17.
Demirkaya, E., Ozen, S., Pistorio, A., et al.; Paediatric Rheumatology Henes, J.C., Mueller, M., Pfannenberg, C., Kanz, L., and Koetter, I. (2011).
International Trials Organization (PRINTO) (2012). Performance of Cyclophosphamide for large vessel vasculitis: assessment of response by
Birmingham Vasculitis Activity Score and disease extent index in child- PET/CT. Clinical and Experimental Rheumatology, 29 (1 Suppl. 64), S43–8.
hood vasculitides. Clinical and Experimental Rheumatology, 30 (1 Suppl. Karassa, F.B., Matsagas, M.I., Schmidt, W.A., and Ioannidis, J.P. (2005).
70), S162–8. Meta-analysis: test performance of ultrasonography for giant-cell arte-
Dolezalova, P., Price-Kuehne, F.E., Ozen, S., et al. (2013). Disease activity ritis. Annals of Internal Medicine, 142, 359–69.
assessment in childhood vasculitis: development and preliminary vali- Koldingsnes, W. and Nossent, H. (2002). Predictors of survival and organ
dation of the Paediatric Vasculitis Activity Score (PVAS). Annals of the damage in Wegener’s granulomatosis. Rheumatology (Oxford), 41,
Rheumatic Diseases, 72, 1628–33. 572–81.
Exley, A.R., Bacon, P.A., Luqmani, R.A., Kitas, G.D., Carruthers, D.M., and Lapraik, C., Watts, R., Bacon, P., et al.; on behalf of the BSR; BHPR
Moots, R. (1998). Examination of disease severity in systemic vasculitis Standards, Guidelines and Audit Working Group (2007). BSR and
from the novel perspective of damage using the vasculitis damage index BHPR guidelines for the management of adults with ANCA associ-
(VDI). British Journal of Rheumatology, 37, 57–63. ated vasculitis. Rheumatology (Oxford), 46, 1615–6.
Exley, A.R., Bacon, P.A., Luqmani, R.A., et al. (1997a). Development and Initial Luqmani, R. (2012). Large vessel vasculitides:  update for the cardiologist.
Validation of the Vasculitis Damage Index (VDI) for the Standardised Current Opinion in Cardiology, 27, 578–84.
Clinical Assessment of Damage in the Systemic Vasculitides. Arthritis Luqmani, R.A., Bacon, P.A., Moots, R.J., et al. (1994). Birmingham vascu-
and Rheumatism, 40, 371–80. litis activity score (BVAS) in systemic necrotizing vasculitis. Quarterly
Exley, A., Carruthers, D.M., Luqmani, R.A., et al. (1997b). Damage occurs Journal of Medicine, 87, 671–8.
early in systemic vasculitis and is an index of outcome. Quarterly Journal McKinney, E.F., Lyons, P.A. Carr, E.J., et al. (2010). A CD8+ T cell tran-
of Medicine, 90, 391–9. scription signature predicts prognosis in autoimmune disease. Nature
Gayraud, M., Guillevin, L., le Toumelin, P., et al.; French Vasculitis Study Medicine, 16, 586–91.
Group (2001). Long-term followup of polyarteritis nodosa, microscopic Merkel, P.A., Aydin, S.Z., Boers, M., et al. (2011). The OMERACT core set of
polyangiitis, and Churg-Strauss syndrome: analysis of four prospective outcome measures for use in clinical trials of ANCA-associated vasculi-
trials including 278 patients. Arthritis and Rheumatism, 44, 666–75. tis. Journal of Rheumatology, 38, 1480–6.
Guillevin, L., Le Thi Huong, D., Godeau, P., Jais, P., and Wechsler, B. Mukhtyar, C., Guillevin, L., Cid, M.C., et al.; European Vasculitis Study
(1988). Clinical findings and prognosis of polyarteritis nodosa and Group (2009a). EULAR Recommendations for management of primary
CHAPTER 22 assessment of disease activity and damage 305

small and medium vessel vasculitis. Annals of the Rheumatic Diseases, Stone, J.H., Hoffman, G.S., Merkel, P.A., et al. (2001). A Disease-Specific
68, 310–17. Activity Index for Wegener’s Granulomatosis:  Modification of the
Mukhtyar, C., Guillevin, L., Cid, M.C., et al.; European Vasculitis Study Birmingham Vasculitis Activity Score. Arthritis and Rheumatism, 44,
Group (2009b). EULAR Recommendations for the management of large 912–20.
vessel vasculitis. Annals of the Rheumatic Diseases, 68, 318–23. Suppiah, R., Flossmann, O., Mukhtyar, C., et al. (2011a). Measurement of
Robson, J., Doll, H., Suppiah, R. et al. (2013). Damage in the anca-associated damage in systemic vasculitis: a comparison of the Vasculitis Damage
vasculitides: long-term data from the European Vasculitis Study group Index with the Combined Damage Assessment Index. Annals of the
(EUVAS) therapeutic trials. Annals of the Rheumatic Diseases, Nov 15. Rheumatic Diseases, 70, 80–5.
[Epub ahead of print] Suppiah, R., Judge, A., Batra, R., et al. (2011b). A model to predict cardiovas-
Seo, P, Min, Y.-I., Holbrook, J.T., Hoffman, G.S., et al. for the WGET cular events in patients with newly diagnosed Wegener’s granulomatosis
Research Group (2005). Damage from Wegener’s Granulomatosis and and microscopic polyangiitis. Arthritis Care and Research (Hoboken), 63,
its Treatment:  Prospective data from the Wegener’s Granulomatosis 588–96.
Etanercept Trial (WGET). Arthritis and Rheumatism, 52, 2168–78. Walsh, M., Mukhtyar, C., Mahr, A., et al. (2011). Health related quality of
Slot, M.C., Tervaert, J.W., Boomsma, M.M., and Stegeman, C.A. (2004). life in patients with newly diagnosed anti-neutrophil cytoplasm anti-
Positive classic antineutrophil cytoplasmic antibody (C-ANCA) titer body associated vasculitis. Arthritis Care and Research (Hoboken), 63,
at switch to azathioprine therapy associated with relapse in proteinase 1055–61.
3-related vasculitis. Arthritis and Rheumatism, 51, 269–73.
CHAPTER 23

Giant cell arteritis and


polymyalgia rheumatica
Kenneth J. Warrington and Cornelia M. Weyand

Introduction tissue breakdown (Pryshchep et al. 2008). Unique features of the


tissue microenvironment, including DC, shape the course of the
Giant cell arteritis (GCA), also known as temporal arteritis or inflammatory response. The vasculitic process in GCA is therefore
granulomatous arteritis, is an inflammatory vasculopathy that hypothesized to arise from aberrant interactions between the vas-
manifests almost exclusively in medium- and large-size arteries. cular and the adaptive immune systems (Piggott et al. 2009).
GCA occurs in the sixth to eighth decades of life, coinciding with
age-related changes in the immune system and in the vasculature.
The vasculitic process typically involves the extracranial branches
Epidemiology
of the carotid artery and frequently also affects the aorta, and GCA affects people 50 years of age or older and the risk of GCA
aortic arch branches. These vessels are targeted by a transmural, increases with advancing age, with the highest rates observed in
granulomatous, inflammatory process composed of mononuclear individuals between 70 and 79 years of age (Gonzalez-Gay et al.
cells and giant cells. Innate and adaptive immune responses within 2009). Interestingly, the average age at onset of GCA has increased
the wall of arteries lead to vascular damage, resulting in adverse in recent decades. In a population-based study, the mean age at
clinical outcomes that depend on the size of vessel involved. In diagnosis of GCA increased from about 73 years (cases diagnosed
response to vascular injury, medium-sized vessels occlude the 1950–1979), to 76.7 years (cases diagnosed 1980–2004) (Kermani
lumen through intimal hyperplasia, causing end-organ ischaemia. et al. 2010). Since GCA occurs almost exclusively in the elderly, it
The ominous clinical consequences that ensue may include vision is likely that immunosenescence is linked to disease susceptibil-
loss, cerebrovascular ischaemic events, and limb claudication. In ity. However, the mechanisms whereby age-related changes in the
large, elastic arteries such as the aorta, destruction of the wall leads immune system increase the risk for GCA have yet to be defined.
to progressive dilatation, aneurysm formation, and potentially Women are affected by GCA two to three times as often as men,
fatal vascular events (dissection, rupture). The vascular inflamma- suggesting that hormonal and/or reproductive factors may also
tion of GCA is almost always accompanied by a non-specific sys- play a role in disease pathogenesis.
temic inflammatory syndrome. Indeed, elevation of inflammatory GCA is one of the most common forms of vasculitis in adults
biomarkers, which fall into the category of acute phase reactants, is in Western countries, with an incidence that varies by race and
considered a hallmark of GCA. The resultant constitutional symp- geographic location. In the United States, GCA preferentially
toms and proximal myalgias are frequent in patients with GCA, affects white people of Scandinavian descent and is rare among
but may also occur in isolation as a condition termed polymyal- African Americans (Gonzalez-Gay et al. 2009). In Olmsted County,
gia rheumatica (PMR) (Weyand and Goronzy 2003a; Weyand and Minnesota, United States, the average annual incidence is 19 cases
Goronzy 2003b). per 100 000 people 50 years of age or older (Kermani et al. 2010).
While corticosteroids (CS) remain the cornerstone of therapy Using epidemiological data from the Olmsted County population,
for GCA, it is now recognized that current treatment does not the lifetime risk of GCA has been estimated to be about 1% for
readily abrogate vascular inflammation and long-term vascular women and 0.5% for men (Crowson et al. 2011).
complications still arise. Studies in untreated and treated patients The incidence of GCA in northern European countries, such as
have contributed significantly to elucidating the pathogenesis of Norway and Sweden, is similar at about 20–30 cases per year per
GCA, revealing that two separable immune pathways are involved. 100 000 population age ≥50 years. In contrast, GCA is less com-
While the Th17 pathway is treatment responsive, chronic lesions mon in Southern European and Mediterranean countries, where
of vasculitis are maintained by ongoing Th1-driven inflammation the annual incidence is about 7–10 per 100  000 population age
refractory to CS (Weyand et al. 2011). Also, it has been shown that ≥50 years (Gonzalez-Gay et al. 2009). GCA appears to be rare in
blood vessels play an active role in immunosurveillance, as they Asian populations, and Japan has a reported incidence of 1.5 cases
are equipped with specialized cells, so-called vascular dendritic per 100 000. Several population-based epidemiological studies have
cells (vasDC) that have immune-sensing functions. Such vasDC reported that the incidence of GCA has been increasing over the
reside in the wall of medium and large arteries and serve a moni- second half of the twentieth century. However, increased physician
toring function for danger signals resulting from infections or awareness of the disease appears to account for this observation.
308 SECTION 5 vasculitic diseases and syndromes and related disorders

Indeed, the incidence of GCA appears to have stabilized over the (a) (b)
past 10–15 years (Gonzalez-Gay et al. 2009).

Aetiology
The exact cause of GCA remains unknown; however, genetic and
environmental factors are likely contributors to disease pathogen-
esis. Genetic polymorphisms of the HLA class II region are asso-
ciated with susceptibility to GCA. Specifically, HLA-DRB1*0401
and DRB1*0404/8 alleles are over-represented in patients with
GCA, suggesting that mechanisms related to antigen recognition 500 µm
by T cells or T-cell repertoire selection may predispose to GCA
(Weyand et  al. 1992). Several genetic association studies have Fig. 23.1 Histomorphological findings in GCA. (a) Cross-section of the temporal
artery with mononuclear cells infiltrating the adventitia, media, and intima.
implicated genetic variants in key components of immune and
Occlusion of the vascular lumen by hyperplastic intima. (b) Typical positioning of
inflammatory pathways in GCA susceptibility. Polymorphisms multinucleated giant cells at the media-intima border.
in cytokine genes such as tumour necrosis factor alpha (TNF-α),
monocyte chemotactic protein-1 (MCP-1), and interleukin
(IL)-10 have been identified as possible susceptibility factors.
Other polymorphisms, such as those in the intercellular adhe- The internal elastic lamina is fragmented and may be surrounded
sion molecule 1 (ICAM-1), vascular endothelial growth factor by multinucleated giant cells (Figure 23.1). In up to one-half of
(VEGF), and interferon-gamma (IFN-γ) genes, may predispose biopsy specimens with arteritis, giant cells may be absent and
to increased disease severity. However, the overall risk associated are not required for the diagnosis of GCA (Lie 1990). The inner
with non-MHC gene polymorphisms is very low and because layer of the artery undergoes concentric fibrointimal proliferation
most studies included small patient cohorts, the reported genetic resulting in luminal stenosis, and, at times, occlusion of the ves-
associations will need validation in larger, multicentre cohorts sel. The arterial inflammation in GCA is often segmental, both
(Warrington et al. 2009). radially around the circumference of the artery and longitudi-
Several infectious agents have been implicated in disease patho- nally along the course of the vessel. Fibrinoid necrosis is not a
genesis, but conclusive evidence is lacking. Mycoplasma pneu- feature of GCA and its presence may indicate an alternate type
moniae, Chlamydia pneumoniae, parvovirus B19, parainfluenza of systemic vasculitis, such as antineutrophil cytoplasmic anti-
virus, and varicella zoster virus have all been suggested as dis- body (ANCA)-associated vasculitis or a paraneoplastic vasculitis
ease triggers (Weyand and Goronzy 2003b). However, attempts to (Warrington et al. 2003).
identify a single pathogenic organism within arterial specimens In a subset of patients, arterial biopsy may show intimal hyper-
from patients with GCA have yielded inconsistent results. This plasia and fragmentation of the internal elastic lamina without
may also suggest that various organisms could trigger vascular active transmural inflammation. This is often reported as ‘healed
inflammation by non-specific immune activation. Indeed, com- arteritis’ by the examining pathologist. However, these findings
pelling evidence that non-infectious bacterial products can trig- can be non-specific and there is significant interobserver variation
ger vasculitis comes from studies using a mouse model in which when interpreting such biopsy findings. Indeed, fragmentation of
human arteries are engrafted into immunodeficient mice that also the internal elastic lamina may be seen in the temporal arteries of
carry a human immune system (human artery-SCID chimera normal, elderly individuals. True ‘healed arteritis’ can be seen in
model). In this model, vasculitis is induced by bacterial compo- patients who receive a prolonged course of CS prior to undergoing
nents such as lipopolysaccharide and flagellin. These molecules TAB (Lee et al. 2012). In less than 10% of patients with GCA, the
trigger innate immune responses and downstream activation of arterial inflammation is limited to the vasa vasorum (in the adven-
the adaptive immune system (Deng et  al. 2009). In summary, titia), the small vessels surrounding the adventitia, or both. Cranial
GCA is probably triggered by immunostimulatory danger signals signs and symptoms of GCA tend to be less frequent in patients
in a genetically predisposed person. with periadventitial small-vessel vasculitis (SVV) and/or vasa vaso-
rum vasculitis (VVV) than in those with classic transmural GCA.
However, the prevalence of visual loss and PMR in patients with
Histopathology SVV/VVV is similar to that of patients with classic GCA (Restuccia
The vascular inflammation due to GCA typically involves the et al. 2012).
extracranial branches of the aorta and spares the intracranial ves- Histopathological findings in the aorta of patients with GCA
sels. However, widespread involvement of the aorta itself as well differ somewhat from those seen in temporal artery specimens. In
as the upper and lower extremity arteries is also not uncommon the aorta, inflammation tends to be more prominent in the adven-
(Lie, 1995). The superficial temporal arteries are most acces- titia and medial layers, and luminal narrowing does not occur.
sible for sampling, and the diagnosis of GCA is generally made Sometimes, the histological pattern of GCA in the aorta is referred
by histological examination of a temporal artery biopsy (TAB) to as ‘medial laminar necrosis’, which is characterized by intramu-
specimen. The typical histological findings in GCA consist of ral infarction causing loss of smooth muscle cells and collapse of
transmural mononuclear cell infiltrates, involving all three layers the medial elastic layers. Giant cells and macrophage infiltrates are
of the arterial wall (intima, media, and adventitia). In the media, often seen around these areas of necrosis. Vasculitic damage to the
the cellular infiltrate is organized in granulomas, consisting of wall of the aorta leads to mural weakening and potential aneurysm
activated T cells and macrophages (Weyand and Goronzy 2003b). formation (Miller and Maleszewski 2011).
CHAPTER 23 giant cell arteritis and polymyalgia rheumatica 309

Pathogenesis (Ma-Krupa et al. 2004). In GCA, activated DC produce proinflam-


matory cytokines (e.g. IL-1β, IL-6) and chemokines that recruit
Innate immune activation T  cells and macrophages into the arterial wall. DC also provide
The inflammatory process in GCA appears to arise in the adventi- powerful co-stimulatory signals to activate T lymphocytes via
tial layer of the arterial wall. The initial stage of the disease involves expression of surface receptors such as CD80 and CD86, which
activation of resident dendritic cells (vasDC) that are able to attract bind to CD28 on T-cell membranes. Moreover, in GCA, DC fail to
and induce T cells to invade the arterial wall (Figure 23.2). Recently, migrate out of the inflammatory vascular lesions. Under physiolog-
it has been recognized that normal arteries are able to initiate innate ical conditions, activated DC migrate towards lymphoid organs to
immune responses. In normal temporal arteries, DC positioned at present antigen to T cells. However, in GCA-affected arteries DC
the junction of the media and adventitial layer of the artery func- express homing chemokines (CCL18, CCL19, and CCL21) which
tion as surveyors of the environment. DC therefore equip the arte- direct them to remain trapped in the vessel wall. Here, these mature
rial system with an immune-sensing mechanism and enable the DCs are able to then initiate adaptive immune responses and there-
blood vessel to actively communicate with the adaptive immune fore sustain and amplify the inflammatory process (Ma-Krupa
system. Indeed, vessel walls that have been stripped of the adven- et al. 2004). Interestingly, the activation signal for DC contributes
titial layer (therefore lacking DC) lose their pathogen-sensing to shaping the pattern of adaptive immune response in the artery.
ability (Pryshchep et  al. 2008). The normal artery, via vasDC, is In a study utilizing the human–SCID chimera model of GCA, DC
able to sense danger signals such as lipopolysaccharide and other activation induced through either TRL4 or TLR5 ligands elicited
pathogen-associated molecular patterns (PAMPs), as well as distinct patterns of vascular inflammation (Deng et  al. 2009).
molecules from injured or necrotic cells and degraded extracel- DC-sensing TLR5 instruct T cells to arrange in perivascular clus-
lular matrix components (damage-associated molecular patterns, ters, around vasa vasorum. This pattern of vasculitis resembles
DAMPs). These danger signals activate vasDC via pattern recogni- adventitial vasa vasoritis, which is seen in a subset of patients with
tion receptors, including those of the Toll-like receptor (TLR) fam- GCA. Conversely, T cells stimulated by TLR4-activated DC infil-
ily. Interestingly, the pattern of TLR expression varies by vascular trate into all layers of the arterial wall and cause transmural inflam-
territory and each vessel supports selective T-cell responses. In a mation (similar to the classic histological pattern of GCA). These
study of human arteries from six regions (aorta, carotid, subcla- studies suggest that the nature of the initial trigger of vasculitis is
vian, temporal, mesenteric, iliac artery) each had a unique profile critical in shaping the pattern and progression of the disease.
of TLR, and when stimulated with TLR ligands, arteries responded Another important player in the innate immune response in GCA
with a vessel-specific pattern. Therefore, it is hypothesized that is the macrophage. Macrophages are an essential component of the
the immune reactivity of different vascular beds defines the tissue granulomatous reaction in the artery; they induce tissue damage
tropism of GCA and may explain why the disease preferentially and participate in the maladaptive healing response of the vessel
targets certain arteries (e.g. temporal and subclavian) (Pryshchep (Weyand and Goronzy 2003b). Macrophages infiltrating the arte-
et al. 2008). rial wall specialize in different functions, consistent with the partic-
The activation of vasDC via TLR is an early and critical step in the ipation of M1 as well as M2 macrophages in the vasculitic response.
pathogenesis of vasculitis (Han et al. 2008). In the chimera model M1 macrophages are proinflammatory and characterized by the
of GCA it has been shown that depletion of DC promptly abro- release of cytokines and reactive oxygen species whereas M2 mac-
gates T cell and macrophage activation and vascular inflammation rophages support angiogenesis, tissue remodelling, and repair. The

(b)
(a)
Vasa
Vasa vasorum
vasorum IFN-γ-producing
IFN-γ-producing Th1 cell
Th1 cell Adventitial
Adventitia thickening

vascular
vascular dendritic cell
dendritic cell

IFN-γ-producing
IL-17-producing Th1 cell
Th17 cell Medial
Media thinning
multinucleated
macrophage giant cell
Lamina elastica
interna
Intimal
Intima hyperplasia

Fig. 23.2 Scheme of the immunopathogenic events in GCA. (a) Untreated vasculitis: vascular dendritic cells positioned in the adventitia attract T cells and macrophages
into the vessel wall. T cells are activated in situ and differentiate into two distinct functional lineages; IL-17-producing Th17 cells and IFN-γ-producing Th1 cells.
Macrophages join T cells to form granulomatous infiltrates. (b) Chronic vasculitis: corticosteroid treatment effectively suppresses IL-1, IL-6, and IL-23 and thus disrupts
induction of Th17 cells. The vasculitic infiltrates are reorganized and persist as ‘pure’ Th1 responses. IFN-γ promotes differentiation of macrophages into distinct subsets
and fosters giant cell formation. Tissue damage preferentially affects the medial layer. Fragmentation of the lamina elastica interna allows migrating myofibroblasts to
reach the intima and form lumen-stenosing hyperplastic intima.
310 SECTION 5 vasculitic diseases and syndromes and related disorders

arterial microenvironment appears to determine the topographi- are almost tenfold elevated in the blood and these cells accumulate
cal arrangement as well as the functional profile of macrophages in the vascular infiltrates. With CS treatment, however, both cir-
(Weyand et al. 1996). In GCA, adventitial macrophages positioned culating and tissue-infiltrating Th17 cells are depleted and remain
close to T cells primarily secrete proinflammatory cytokines such suppressed during the chronic phase of the vasculitis. Indeed, the
as IL-1 and IL-6 (Wagner et al. 1994). Macrophages in the media Th17 pathway appears to be very sensitive to even low doses of CS.
injure vascular smooth muscle cells and endothelial cells via oxi- CS also abrogate production of IL-17-polarizing cytokines includ-
dative stress mechanisms, including lipid peroxidation and protein ing IL-1β, IL-6, and IL-23. On the other hand, Th1 cells are refrac-
nitration (Rittner et  al. 1999). Medial macrophages also synthe- tory to CS therapy and persist in the lesions of chronic vasculitis.
size matrix metalloproteinases (MMP), which are a major source Even after 6 months of treatment, more than 80% of patients with
of tissue-digestive activity, destroying matrix and fragmenting the GCA still had inflammatory infiltrates in arterial biopsy speci-
internal elastic lamina. On the other hand, macrophages residing mens, dominated by Th1 cells. Forty per cent of patients had posi-
closer to the intima–media junction exhibit a predominantly M2 tive biopsies after 9 months and 20% of the biopsies showed active
phenotype. These macrophages, as well as multinucleated giant vasculitis after 1 full year of CS treatment (Deng et al. 2010). The
cells, provide growth factors and proangiogenic molecules, includ- persistence of Th1 cells despite CS therapy is likely to be respon-
ing vascular endothelial growth factor (VEGF), platelet-derived sible for the chronicity and relapsing nature of GCA. Th1 cells
growth factor (PDGF), and fibroblast growth factor (FGF). These mainly produce INF-γ, which stimulates macrophages and induces
mediators induce migration and proliferation of myofibroblasts, formation of multinucleated giant cells. High INF-γ production
new vessel formation, and marked thickening of the arterial inti- is seen in patients with ischaemic complications, implicating this
mal. The process of intimal expansion and hyperplasia leads to Th1-derived cytokine in the process leading to luminal occlusion
stenosis and potentially occlusion of the vessel lumen, resulting in (Weyand et al. 1997).
tissue ischaemia. Indeed, ischaemic manifestations such as vision A recent study has shown that the NOTCH pathway regulates
loss tend to correlate with the degree of intimal hyperplasia and vasculitic T cells in GCA. The NOTCH signalling pathway is impor-
accumulation of PDGF-producing macrophages at the intima– tant for cell–cell communication and regulation of cell differentia-
media junction (Kaiser et  al. 1998). A  recent study documented tion processes. In acute lymphoblastic leukaemia, gain-of-function
overexpression of IL-33 in the inflamed arteries of patients with mutations of NOTCH receptors lead to uncontrolled T-cell growth.
GCA, and suggested that IL-33 contributes to M2 polarization of Arteries affected by GCA display abundant expression of the
macrophages (Ciccia et al. 2013). NOTCH receptor and its ligands. In a humanized mouse model
of GCA, NOTCH pathway disruption inhibited T-cell activation in
the early and established phases of vascular inflammation. NOTCH
Adaptive immune responses inhibition was particularly effective in down-regulating Th17
The granulomatous reactions in GCA depend on CD4 T cells, responses, but also markedly suppressed Th1 responses (Piggott
which orchestrate the stimulation of macrophages and, probably, et al. 2011).
the fusion of macrophages into giant cells. Sequence analysis of
the T-cell receptor (TCR) has demonstrated that clonal expansion
of T cells occurs within the arterial wall. Moreover, T cells with Clinical features
identical TCR were isolated from distinct inflammatory foci in GCA typically presents as new-onset headache associated with a
the same arterial specimen (Weyand et  al. 1994b). These obser- systemic inflammatory syndrome in an individual over the age of
vations suggest that in situ T-cell activation occurs in response 50 years. However, it should be emphasized that the clinical presen-
to the same antigen at different locations. However, no common tation is highly variable and depends on the distribution of vascular
T-cell receptor families were identified among a series of patients inflammation. GCA should be on the list of differential diagnosis
with GCA, making a single shared GCA-specific antigen less likely in patients with systemic inflammation and in cases of ischaemic
(Martinez-Taboada et  al. 1996). Indeed, it is plausible that more disease related to medium and large arteries. Two major categories
than one disease trigger drives the vasculitic process in GCA. In of GCA have been distinguished: cranial arteritis predominantly
support of this hypothesis, it has recently been shown that at least affecting the branches of the external carotid artery and large-vessel
two separate lineages of CD4 T cells participate in vascular inflam- GCA, involving the aorta and its primary branches (Weyand and
mation. Both the interferon-gamma (INF-γ)-producing Th1 and Goronzy 2003b).
the IL-17-producing Th17 cells, are critical components of the Cranial symptoms of GCA primarily include headache and scalp
adaptive response (Figure 23.2). The Th1 and Th17 pathways are tenderness. Headache due to GCA is generally persistent, refrac-
differentially triggered by independent signals from two types of tory to simple analgesics, and located over the temporal or less
antigen-presenting cells (APC). APC that secrete IL-12 promote commonly, the occipital areas. Headache may be accompanied by
Th1 differentiation, while the Th2 pathway is dependent upon scalp tenderness and thickening or nodularity of the frontal or pari-
IL-1β/IL-6/IL-23-producing APC (Weyand et al. 2011). This dis- etal branches of the superficial temporal arteries (Salvarani et al.
covery derives from the analysis of two consecutive arterial biop- 2008). Clinical abnormalities of the temporal artery (nodularity,
sies from the same patient: one obtained prior to therapy and the swelling, diminished pulse, or tenderness to palpation) are predic-
second obtained after a defined period of CS therapy. Using this tive of a positive temporal artery biopsy (Smetana and Shmerling
study design, it was possible to compare the functional profiles of 2002). Necrosis of the scalp due to lack of tissue perfusion is a
T cells driving early and chronic GCA (Deng et al. 2010). In early, rare complication of GCA (Brick et  al. 2010). Although present
untreated disease both Th1 and Th17 cells are present in the vascu- in only about a third of patients, jaw claudication is characteris-
litic lesions. In patients with untreated GCA, Th17 cell frequencies tic and highly specific for GCA. Patients generally describe this as
CHAPTER 23 giant cell arteritis and polymyalgia rheumatica 311

pain with chewing that is unilateral or bilateral and results from patients, constitutional symptoms may predominate or may be the
vascular insufficiency of the muscles of mastication. Occasionally, only presenting feature, leading clinicians to pursue a thorough
patients report claudication of the tongue with activities such as evaluation for occult malignancy or indolent infectious process
talking or singing. Upper respiratory symptoms occur in about 10% before a diagnosis of GCA is considered. It has been estimated that
of patients, and may include non-productive cough, sore throat, or about 15% of patients with GCA can initially present with fever of
hoarseness (Larson et al. 1984). unknown origin (Salvarani et al. 2008).
Ocular symptoms such as impaired vision, diplopia, and amau- About 40% of patients with GCA experience symptoms of poly-
rosis fugax are common in GCA, and up to 15% of patients myalgia rheumatica (PMR). PMR is a clinical syndrome character-
develop permanent visual loss (Aiello et  al. 1993; Gonzalez-Gay ized by aching and stiffness affecting the neck, shoulders, hips, and
et al. 2000). Anterior ischaemic optic neuropathy (AION) due to proximal extremities. Symptoms are usually bilateral and more pro-
occlusion of the posterior ciliary arteries or the ophthalmic artery nounced in the morning or after periods of inactivity. Clinical eval-
is the predominant ocular manifestation of GCA. Other less fre- uation generally reveals that active range of motion of the shoulders
quent causes of vision loss include central retinal artery occlu- and hips is limited due to pain. Peripheral arthritis of joints such
sion, cilioretinal artery occlusion, and posterior ischaemic optic as the wrists and knees may occur, but is uncommon. There is no
neuropathy (Hayreh et  al. 2002). Partial or complete monocular specific laboratory marker for PMR and several clinical diagnostic
vision loss should be considered an ophthalmological emergency criteria have been proposed. Provisional classification criteria for
as patients may progress to bilateral involvement and complete PMR that incorporate use of diagnostic ultrasound have been pub-
blindness. Fundoscopic examination in a patient with ischaemia lished (Dasgupta et al. 2012) (Table 23.1). Musculoskeletal imaging
of the optic nerve may reveal pallor and oedema of the optic disk. such as ultrasonography and magnetic resonance imaging (MRI)
Occasionally, cotton-wool spots and small haemorrhages are evi- can demonstrate bursitis, tenosynovitis, and/or synovitis involving
dent. Neurological manifestations of GCA are uncommon and may the shoulders and hips. Positron emission tomography (PET) scans
include stroke or transient ischaemic attack, generally due to vascu- in patients with isolated PMR have demonstrated inflammatory
litis involving the posterior circulation (vertebral and basilar arter- activity (increased fluorodeoxyglucose uptake) in the shoulders,
ies). Peripheral neuropathy and multiple mononeuropathies occur hips, and cervical and lumbar interspinous processes (Kermani and
in less than 10% of patients with GCA (Caselli et al. 1988). Warrington 2012).
Patients with large-vessel GCA (LV-GCA) generally present with PMR can occur in the absence of GCA; however, it has been
symptoms of vascular insufficiency affecting the extremities, typi- recognized that a subset of patients with apparently isolated PMR
cally the arms. Symptoms of claudication occur with use, for exam- has subclinical vascular inflammation. PMR and GCA share many
ple vacuuming, and resolve with rest. Patients with LV-GCA tend to similarities including epidemiological and immunological fea-
be younger than those with cranial GCA and cranial symptoms are tures, suggesting that they may represent different spectra of the
often absent, resulting in a delayed diagnosis. Asymmetric upper same disease. Both PMR and GCA are characterized by a chronic
extremity blood pressures or absence of the radial pulse may also
prompt medical evaluation (Brack et al. 1999; Schmidt et al. 2008).
Rarely, vasculitic involvement can extend to the lower extremity Table 23.1 European League against Rheumatism and American
arteries, particularly the superficial femoral arteries (Kermani et al. College of Rheumatology provisional criteria for classification of
2009). Such patients may present with rapidly progressive, bilateral polymyalgia rheumatica (PMR)
leg claudication without classic cranial symptoms. Lower extrem-
Required criteria
ity claudication due to vasculitis may be difficult to distinguish
Age ≥50 years
from atherosclerotic peripheral arterial disease based on clinical Bilateral shoulder pain
assessment and the diagnosis is made based on imaging findings. Abnormal erythrocyte sedimentation rate and/or C-reactive protein
Clinical assessment of patients with LV-GCA may disclose arterial
Criteria for scoring algorithm Points
bruits over the carotid, subclavian, or femoral arteries. Therefore,
physical examination of patients with GCA should include not Clinical criteria
only a careful assessment of the temporal arteries but also a com- Morning stiffness lasting >45 minutes 2
plete cardiovascular examination with particular attention to the Hip pain or limited range of motion 1
peripheral vessels. Asymmetry in upper extremity blood pressures Absence of rheumatoid factor and/or anticitrullinated 2
protein antibody
or absent radial pulses may provide important clues for the diag-
Absence of peripheral joint pain 1
nosis of LV-GCA. The thoracic aorta is also frequently involved by
granulomatous aortitis. This vessel does not occlude but the vascu- Ultrasound criteria
litis causes progressive damage to the arterial wall, which may con- At least 1 shoulder with subdeltoid bursitis and/or biceps 1
sequently lead to aortic dilation. Clinically, an aortic insufficiency tenosynovitis and/or
glenohumeral synovitis and at least 1 hip with synovitis
murmur could indicate the presence of aortic root dilatation and
and/or trochanteric bursitis
ascending aortic aneurysm. Therefore, patients with GCA and aor-
Both shoulders with subdeltoid bursitis, biceps tenosynovitis, 1
tic regurgitation should be evaluated promptly for possible thoracic or, glenohumeral synovitis
aortic aneurysm.
Constitutional symptoms commonly occur in patients with A patient with a score of ≥4 clinical criteria (or ≥5 if ultrasonography findings are
GCA and may include malaise, fatigue, weight loss, and low-grade considered) can be categorized as having PMR.
(Dasgupta, B., Cimmino, M.A., Kremers, H. M., et al. (2012). 2012 Provisional classification
fever. When these occur in the context of classic cranial symptoms, criteria for polymyalgia rheumatica: a European League Against Rheumatism/American
the diagnosis of GCA is readily apparent. However, in a subset of College of Rheumatology collaborative initiative. Arthritis and Rheumatism, 64, 943–54)
312 SECTION 5 vasculitic diseases and syndromes and related disorders

inflammatory state and similar cytokine profiles. TAB specimens Diagnostic approach
from patients with PMR show evidence of cytokine transcrip-
tion even in the absence of histopathological findings of arteritis. The American College of Rheumatology has developed a set of
Indeed, it has been hypothesized that PMR represents GCA with an clinical, laboratory, and histopathology criteria, which are helpful
incompletely developed vascular component (Weyand et al. 1994a). in guiding the clinician in the diagnostic process. Originally devel-
In support of this hypothesis, PET scan imaging shows that many oped to distinguish GCA from other vasculitic disorders, the cri-
patients with isolated PMR have FDG uptake in the aorta and its teria are relatively easily applied in clinical practice (Hunder et al.
proximal branches, consistent with subclinical vasculitis (Moosig 1990) (Table 23.2). Histopathological examination of a superficial
et al. 2004). TAB specimen remains the ‘gold standard’ diagnostic modality
for GCA. Due to the possibility of ‘skip lesions’ (segmental areas
of inflammation), the TAB should be of sufficient length to avoid
Laboratory investigations false-negative results. In one series, vascular inflammation was more
Laboratory studies generally reveal evidence of systemic inflamma- likely to be present in arteries that were ≥ 1 cm in length compared
tion; however, specific diagnostic biomarkers for GCA are lacking. to those that were <1 cm long (74.3% versus 25.7%, respectively)
Complete blood count abnormalities may include anaemia (normo- (Taylor-Gjevre et al. 2005). Other investigators have reported that a
chromic, normocytic) and thrombocytosis. Although non-specific, biopsy length of ≥ 0.5 cm was associated with a sixfold higher like-
the presence of thrombocytosis (platelet count >400  000/µl) is lihood of a positive result when compared with a biopsy of <0.5 cm
associated with increased likelihood of GCA (Walvick and Walvick (Mahr et  al. 2006). In clinical practice, a TAB length of at least
2011). Activation of the innate immune system and excess pro- 1 cm for histopathological evaluation is recommended (Dasgupta
duction of IL-6 leads to the induction of the acute phase response, 2010). TAB should still be considered in patients who are already
which is typically measured by the erythrocyte sedimentation rate receiving CS. It has been consistently shown TAB remains a valu-
(ESR) and C-reactive protein (CRP). able diagnostic procedure even after several weeks of CS treatment
Elevated ESR has been considered a hallmark of GCA and is one (Achkar et al. 1994; Ray-Chaudhuri et al. 2002; Younge et al. 2004).
of the American College of Rheumatology’s classification criteria Indeed, some patients have persistent vascular inflammation of the
for the disease (Hunder et al. 1990). The sensitivity of an elevated temporal arteries for many months in spite of CS therapy (Deng
ESR for GCA has been estimated to be about 96% (Smetana et al. et al. 2010).
2002). However, the ESR lacks specificity and may be elevated in The issue of whether unilateral or simultaneous bilateral tem-
a variety of autoimmune, infectious, and neoplastic conditions. poral artery biopsies should be performed remains debatable. The
A normal ESR does not exclude a diagnosis of GCA, but renders rate of discordance (i.e. one side positive and the contralateral side
the diagnosis less likely. In one series, only 4% of patients with GCA negative) ranges from 1.4% to 12.7% among studies (Boyev et al.
had a normal ESR at diagnosis (Smetana et al. 2002). CRP is often 1999; Breuer et al. 2009; Danesh-Meyer et al. 2000; Pless et al. 2000;
considered to be a more sensitive marker of inflammation than Ponge et al. 1988). In a pooled analysis of four studies with bilateral
the ESR. In a study of 3001 subjects who underwent TAB, elevated TAB results, the rate of discordance was 5.9% (Niederkohr et al.
CRP was associated with significantly increased odds of a posi- 2007). Since TAB is a sensitive test and the concordance rate of
tive TAB (OR 5.3; 95% CI: 3.1–8.9) (Walvick and Walvick 2011).
The prevalence of normal CRP at diagnosis in GCA has only been
evaluated in small studies, some of which also included subjects
Table 23.2 American College of Rheumatology 1990 criteria for the
with PMR (Kyle et al. 1989; Myklebust et al. 1996; Pountain et al.
classification of GCA
1994). In one reported series, only 1.7% had a normal CRP at the
time of GCA diagnosis (Parikh et al. 2006). Therefore, a normal For the diagnosis of GCA, at least 3 of the following 5 criteria must be present.
CRP also has an excellent negative predictive value for GCA. In a The presence of any 3 or more criteria yields a sensitivity of 93.5% and a
study published in 2012, elevated CRP and elevated ESR provided specificity of 91.2% for distinguishing GCA from other forms of vasculitis.
a sensitivity of 86.9% and 84.1%, respectively, for a positive TAB. 1. Age at disease onset ≥50 years
Interestingly, 4% of patients with a positive TAB for GCA had both Development of symptoms or findings beginning at age 50 or older
normal ESR and CRP. Patients with normal biomarkers at diagnosis
2. New headache
frequently had PMR symptoms, whereas constitutional symptoms
New onset of or new type of localized pain in the head
were generally absent (Kermani et al. 2012). In summary, although
non-specific, elevated acute phase markers often prompt the clini- 3. Temporal artery abnormality
cian to pursue further testing for GCA. Conversely, the presence of Temporal artery tenderness to palpation or decreased pulsation,
normal inflammatory markers has a high negative predictive value unrelated to arteriosclerosis of cervical arteries
for GCA, but does not completely rule out the disease. Once the 4. Elevated erythrocyte sedimentation rate
diagnosis of GCA is established, the ESR and CRP are measured Erythrocyte sedimentation rate ≥50 mm/hour by the Westergren method
serially as markers of disease activity. 5. Abnormal artery biopsy
Liver function test abnormalities are frequently observed in GCA, Biopsy specimen with artery showing vasculitis characterized by a
including elevated alkaline phosphatase and transaminases (Kyle, predominance of mononuclear cell infiltration or granulomatous
1991). These changes are reversible with CS treatment. In select inflammation, usually with multinucleated giant cells
patients, screening for autoantibodies may be helpful to exclude
(Hunder, G.G., Bloch, D.A., Michel, B.A., et al. (1990). The American College of
other diseases that can mimic GCA, such as rheumatoid arthritis, Rheumatology 1990 criteria for the classification of giant cell arteritis. Arthritis and
systemic lupus erythematosus, and ANCA-associated vasculitis. Rheumatism, 33, 1122–8.)
CHAPTER 23 giant cell arteritis and polymyalgia rheumatica 313

bilateral biopsies is high, it may not be necessary to routinely biopsy of the aortic arch branches or aneurysmal dilatation of the aorta.
both sides. However, if one side is negative for inflammation and However, the diagnostic accuracy of MRA has not been formally
there is still a strong clinical suspicion for GCA, then a subsequent studied in GCA. In clinical practice, MRA is used for the evalu-
contralateral biopsy can be considered in an attempt to confirm the ation and diagnosis of patients with suspected large-vessel GCA.
diagnosis. In patients with cranial symptoms, negative TAB has an However, the role of serial MRA during the treatment phase and
excellent negative predictive value for GCA. follow-up of these patients is still unclear. Computed tomogra-
Imaging of the superficial temporal arteries with colour duplex phy angiography (CTA) also offers a non-invasive means to image
ultrasonography or MRI may show changes suggestive of vasculi- the aorta and its primary branches and is the method of choice in
tis. On ultrasound examination, the most specific sign of vascular patients who have difficulties tolerating the extended time period
inflammation is the ‘dark halo’, a hypoechoic area surrounding the involved in MRI (Figure 23.3). Evidence of vasculitis on CTA may
lumen of the artery, thought to represent oedema of the arterial include wall thickening, delayed wall enhancement, and/or luminal
wall (Schmidt et al. 1997). In a meta-analysis of 23 studies that eval- changes such as long segments of tapered stenoses. In a prospective
uated the use of ultrasound for suspected GCA, the sensitivity of study, CTA was performed in 40 consecutive patients with newly
an abnormal ultrasound (halo, stenosis, or occlusion) for GCA was diagnosed biopsy-proven GCA. Findings of large-vessel vasculitis,
88% with a specificity of 78% (Karassa et al. 2005), as compared to mainly in the aorta and aortic arch branches, were present in 67.5%
the gold standard TAB. However, the diagnostic accuracy of ultra- of patients. A subset of the cohort (15%) had evidence of aortic dila-
sonography can vary depending on the skill and experience of the tion at disease onset (Prieto-Gonzalez et al. 2012). Conventional
operator or the interpreting physician. Moreover, false-negatives catheter-based angiography is invasive, involves radiation exposure,
may occur in case of patchy mural inflammation. Detection by and only detects luminal changes (stenosis, aneurysm). Therefore,
ultrasound requires full-blown and transmural disease, associated this imaging technique is now reserved for presurgical evaluation
with profound tissue oedema. Therefore, cases of difficult-to-detect and has mostly been replaced by CTA and MRA.
vasculitis are at highest risk to be missed. MRI findings that suggest Positron emission tomography (PET) imaging is now part of
vascular inflammation may include vessel wall thickening, oedema the diagnostic armamentarium available for GCA and is particu-
(on T2-weighted images), and postgadolinium enhancement. Bley larly useful for patients with atypical presentations and/or nega-
and colleagues have studied the diagnostic value of MRI of the tive TAB (Figure 23.3). In a prospective study of PET imaging in
superficial cranial arteries. They reported that contrast-enhanced patients with GCA, vascular FDG uptake was noted in 83% of
high-resolution MRI had a sensitivity of 80.6% and specificity of patients, particularly at the subclavian arteries (74%), but also in
97% for the diagnosis of GCA, as compared to clinical diagnosis the thoracic and abdominal aorta (>50%) and up to the femoral
(Bley et al. 2007). The diagnostic utility of MRI and duplex ultra- arteries (37%) (Blockmans et al. 2009). However, smaller arteries
sonography for GCA appears to be comparable (Bley et al. 2008). such as the superficial temporal arteries are below the resolution of
However, both imaging modalities are not widely available and are PET (Brodmann et al. 2004). A recent meta-analysis of six studies
of least value in the challenging patient who has non-classical pres- evaluating PET scan for the diagnosis of GCA calculated an over-
entation. In clinical practice, therefore, TAB remains the preferred all sensitivity of 80% and specificity of 89%. Moreover, the nega-
diagnostic modality for GCA. tive predictive value of a PET scan for GCA was excellent (88%)
It is important for clinicians to recognize that a subset of patients (Besson et al. 2011). Therefore, the absence of vascular FDG-uptake
with GCA have predominantly extracranial (large-vessel) vas- in a patient with suspected GCA renders this diagnosis unlikely.
culitis, without involvement of the superficial temporal arteries. Conversely, in the right clinical setting, the presence of large-vessel
Indeed, TAB may be negative in up to 42% of cases of GCA with FDG uptake strongly suggests the diagnosis of GCA. However,
large-vessel GCA (Brack et al. 1999). Patients with aortitis and no imaging studies performed in patients who are already receiving
cranial symptoms may also have a low frequency of TAB positivity. CS therapy need to be interpreted with caution as FDG uptake is
Therefore, in this subset of patients, large-vessel imaging becomes likely attenuated with treatment (Blockmans et al. 2009). Once a
an essential component of the diagnostic evaluation. Moreover, diagnosis of GCA is established, there is currently no role for serial
increased utilization of imaging modalities in recent years has led PET imaging as this modality is unable to identify which patients
to the appreciation of the extent of large-vessel inflammation in are at highest risk of disease relapse.
GCA. Ultrasonography can be used to image the proximal upper In view of the morbidity associated with prolonged CS therapy for
extremity arteries, particularly the subclavian, axillary and brachial GCA, every effort should be made to confirm or reasonably exclude
arteries. Similar to findings in the superficial temporal arteries, a diagnosis of GCA by tissue biopsy and/or imaging studies.
ultrasonography of the upper extremity arteries may demonstrate
a ‘halo sign’ due to mural thickening and/or luminal changes (long
segment stenosis or occlusion) (Schmidt et al. 2002; Schmidt et al. Management
2008). In a prospective study using ultrasonographic imaging, The goal of treatment is to improve patients’ symptoms and to
30% of patients with GCA had inflammatory changes in the upper reduce the risk of ischaemic complications, such as vision loss.
extremity arteries, often with bilateral involvement (Schmidt et al. CS are the standard of therapy for GCA and are highly effective
2008). However, ultrasonography cannot be used to assess for the in suppressing signs and symptoms of the disease within 24 to
presence of aortitis. Magnetic resonance angiography (MRA) is a 48 hours (Warrington and Matteson 2007). Although CS have not
particularly useful modality for non-invasive imaging of the aorta been evaluated in placebo-controlled clinical trials, CS therapy has
and its primary branches in GCA. Vessel wall oedema, contrast been shown in clinical practice over decades to be very effective
enhancement, and/or wall thickening are important imaging find- in the treatment of GCA. Indeed, in an early retrospective study
ings of large-vessel GCA. In addition, MRA may reveal stenoses comparing treated patients to a precorticosteroid group, it was
314 SECTION 5 vasculitic diseases and syndromes and related disorders

(b)

(a)

(c)

Fig. 23.3 (a) Whole-body PET scan demonstrates findings consistent with large-vessel vasculitis. There is increased FDG uptake in the thoracic aorta, bilateral subclavian
arteries, bilateral axillary arteries, upper abdominal aorta, and the partially visualized bilateral superficial femoral arteries. (b) CT angiogram of a patient presenting with
bilateral arm claudication due to GCA. Coronal 3D reformatted image shows bilateral stenoses of the axillary and proximal brachial arteries. (c) Axial CT angiogram
image shows aneurysmal dilatation of the thoracic aorta with a type A dissection. The patient had a known history of GCA and present acutely with severe chest pain.
Histopathology of the aortic tissue demonstrated active giant cell aortitis.

demonstrated that cortisone significantly prevented vision loss should receive induction therapy with pulse-dose methylpredni-
(Birkhead et  al. 1957). Because of the risk of ischaemic events, solone intravenously (1  gram daily for 3  days), followed by the
CS therapy should be initiated promptly if a diagnosis of GCA is standard oral CS regimen. Retrospective study data suggest that
strongly suspected or confirmed. There is a substantial window of patients with visual loss who received intravenous CS were more
several weeks during which temporal artery biopsy may still yield likely to improve compared to those receiving oral CS only (Chan
a diagnosis of arteritis, even in treated patients (Achkar et al. 1994; et  al. 2001). However, the benefit of pulse CS over oral therapy
Ray-Chaudhuri et  al. 2002). Therefore the diagnostic evaluation remains controversial and established ischaemic optic neuropathy
should not delay initiation of treatment. is typically irreversible. Initial bolus doses of methylprednisolone
The initial daily dose of oral prednisone (or equivalent) should of 1 gram daily for 3 days followed by an oral CS taper may result
be 1 mg/kg (generally 40–60 mg) for 4 weeks (Pipitone and in lower long-term CS requirements (Mazlumzadeh et al. 2006). In
Salvarani 2008). After 4 weeks of treatment, the daily prednisone another trial, pulse methylprednisolone did not appear to modify
dose should be gradually tapered by about 10% of the daily dose the course of the disease and failed to achieve CS-sparing effects
every 2 weeks. Once a daily dose of 10 mg of prednisone is reached, (Chevalet et al. 2000). Alternate-day CS therapy is generally not
further tapering is usually by 1 mg per month until discontinua- recommended. Alternate-day prednisone has been compared
tion or relapse. However, the disease course is variable and treat- with daily prednisone in a randomized, prospective study and
ment must be individualized according to patient response. it was shown that disease control was inadequate in patients on
Patients who present with impending or recent-onset of visual loss alternate-day therapy (Hunder et al. 1975).
CHAPTER 23 giant cell arteritis and polymyalgia rheumatica 315

CS therapy is associated with adverse effects in more than 80% Most patients do not require long-term immunosuppression. In
of patients with GCA (Kyle and Hazleman 1989; Proven et  al. patients with CS-related toxicity and/or frequent disease relapses
2003). Therefore, every effort needs to be made to make the cor- steroid-sparing agents are considered. Because of familiarity with
rect diagnosis and to avoid excessive use of CS. Also, the clinical the drug, rheumatologists often choose methotrexate as the first-line
assessment of patients with GCA should include careful monitor- immunosuppressive agent for patients with GCA. Randomized
ing for CS-related complications such as weight gain, bone loss, clinical trials evaluating the efficacy of methotrexate in patients
hypertension, hyperglycaemia, dyslipidaemia, and cataract for- with GCA have yielded conflicting results (Hoffman et  al. 2002;
mation. CS-induced bone loss should be prevented and/or man- Jover et al. 2001; Spiera et al. 2001). However, a meta-analysis of
aged with calcium, vitamin D, and bisphosphonates according to three randomized placebo-controlled trials suggested that metho-
guidelines of the American College of Rheumatology (Grossman trexate therapy (7.5–20 mg a week and folate, 1 mg/day) allows
et al. 2010). Since patients with GCA are expected to receive more reduction in the prednisone dosage and reduces the risk of GCA
than 3  months of CS therapy, bisphosphonate therapy is recom- relapses (Mahr et al. 2007). However, a decrease in CS-related tox-
mended. Bone mineral density should be measured at baseline icity was not observed in this analysis.
and every 1 to 2 years. Patients with GCA are at increased risk of For patients who flare on methotrexate and/or have
infections, particularly early in the disease course (Durand and methotrexate-related toxicity/ intolerance, alternate immuno-
Thomas 2012). Pneumocystis jiroveci pneumonia (PJP) occurs suppressive agents can be considered, for example azathioprine,
rarely in patients with GCA (Kermani et al. 2011). However, this mycophenolate mofetil. However, there are very few published
preventable infection is associated with significant morbidity and data to guide the use of these medications for GCA. Azathioprine
mortality. Therefore, unless contraindicated, PJP prophylaxis with for GCA has been evaluated in a placebo-controlled trial and was
trimethoprim–sulfamethoxazole, one single-strength tablet daily is found to be of limited benefit (De Silva and Hazleman 1986).
recommended. For patients with sulfa allergy, alternatives include Ciclosporin does not appear to have a role in the treatment of
atovaquone, dapsone, or inhaled pentamidine. PJP prophylaxis is GCA (Schaufelberger et  al. 2006). Use of dapsone and hydroxy-
typically discontinued when the prednisone dose (or equivalent) is chloroquine for GCA has been reported, but these agents have
less than 20 mg daily. not been studied in randomized clinical trials and are not gener-
Unless aspirin is contraindicated due to co-morbidities, low-dose ally recommended (Doury et  al. 1983; Le Guennec et  al. 1994).
aspirin (81 mg) should be part of the treatment regimen for GCA. Cyclophosphamide is only used in exceptional circumstances for
Retrospective studies as well as experimental models indicate that patients with life- or organ-threatening disease that is refractory to
aspirin is potentially effective in preventing ischaemic complica- CS therapy (de Vita et al. 1992).
tions of GCA, without increasing the risk of bleeding complica- Tumour-necrosis factor inhibitors (TNFi) are generally not rec-
tions (Lee et  al. 2006; Weyand et  al. 2002). Specifically, in one ommended for treatment of GCA. Case reports and small case
study, the risk of visual loss and cerebrovascular events was lower series suggested that infliximab and adalimumab may have a
in patients with GCA receiving aspirin compared to those who therapeutic role in GCA (Ahmed et al. 2007; Cantini et al. 2001).
were not on antiplatelet therapy (Nesher et al. 2004). However, it However, a randomized multicentre clinical trial demonstrated that
should be noted that other observational studies have not dem- infliximab was of no benefit as maintenance therapy in patients
onstrated a reduction in ischaemic complications with the use of with newly diagnosed GCA (Hoffman et al. 2007). There are lim-
antiplatelet therapy (Berger et al. 2009; Salvarani et al. 2009). ited data from case reports and small series on the efficacy of toci-
Patients with GCA should be followed regularly by clinical lizumab (anti-IL-6 receptor) therapy for GCA, and this agent may
examination and laboratory studies, including inflammatory mark- hold promise for the future (Beyer et  al. 2011; Seitz et  al. 2011;
ers (ESR and CRP). Disease relapses (defined as a recurrence of Unizony et al. 2012).
symptoms and/or signs of GCA that requires an increase in CS
dosage) frequently occur during the treatment course (Salvarani
et al. 2002). Elevation of inflammatory markers (i.e. ESR or CRP) Prognosis
in the absence of clinical symptoms should not routinely result Overall, the life expectancy of patients with GCA is similar to
in escalation of therapy. Treatment of GCA relapses depends on that of the general population. However, a late complication from
the clinical context and should be tailored according to severity GCA that can influence survival is development of aortic aneu-
of relapse and level of inflammatory markers. Often, only a small rysms. Patients with GCA have a 17-fold increased risk of tho-
increase in CS dose is necessary to regain disease control. However, racic aneurysm (TAA) and a 2.4-fold increased risk of abdominal
a major relapse with ischaemic complications such as visual loss aneurysm (AAA) when compared to individuals without a history
or new large-vessel involvement should be managed by reinitiat- of GCA (Evans et al. 1995). In a population-based study, 18% of
ing high-dose CS. Biomarkers that can predict risk of GCA relapse patients with GCA developed aortic aneurysm and/or dissection
are lacking. However, it has been shown that patients who have (Nuenninghoff et al. 2003a). Development of aortic aneurysm in
a strong inflammatory response at diagnosis tend to have higher GCA is predominantly a late complication and on average, occurs
and more prolonged CS requirements (Hernandez-Rodriguez et al. about 3 to 5  years after diagnosis (Gonzalez-Gay et  al. 2004;
2002). In most cases, CS can be discontinued after 18–24 months Nuenninghoff et al. 2003a). Although various clinical features have
of treatment. It is not unusual that acute phase reactants, such as been identified as potential risk factors for aneurysm formation,
IL-6 and CRP, begin to rise again after CS have been withdrawn these have not been consistently replicated across epidemiologi-
(Weyand et al. 2000). It is now recognized that a considerable frac- cal studies (Gonzalez-Gay et al. 2004; Nuenninghoff et al. 2003a).
tion of patients requires long-term CS, typically at low doses of 5 Aortic aneurysm may lead to aortic dissection, and patients with
mg of prednisolone or less. GCA who develop aortic dissection have a markedly increased
316 SECTION 5 vasculitic diseases and syndromes and related disorders

risk of mortality (Nuenninghoff et al. 2003b). Long-term monitor- Breuer, G.S., Nesher, G., and Nesher, R. (2009). Rate of discordant findings in
ing for aortic aneurysms is therefore recommended, although the bilateral temporal artery biopsy to diagnose giant cell arteritis. Journal of
optimal frequency and imaging modality has not yet been stud- Rheumatology, 36, 794–6.
Brick, K.E., Cook-Norris, R.H., Wetter, D.A., and Warrington, K.J. (2010).
ied (Bongartz and Matteson 2006). Transthoracic echocardiogram
Scalp necrosis in giant cell arteritis after initiation of therapeutic
(most aneurysms develop in the ascending aorta) appears to be an corticosteroids. Journal of the American Academy of Dermatology,
attractive, non-invasive screening method that avoids the radiation 63, 343–4.
exposure associated with CT scans. Patients with GCA who have Brodmann, M., Lipp, R.W., Passath, A., Seinost, G., Pabst, E., and Pilger, E.
evidence of dilatation of the aorta at any time should be followed (2004). The role of 2-18F-fluoro-2-deoxy-D-glucose positron emission
closely with imaging studies, and aneurysm repair considered tomography in the diagnosis of giant cell arteritis of the temporal arter-
depending on size and rate of progression. ies. Rheumatology (Oxford), 43, 241–2.
Patients with stenosis or occlusion of the upper extremity arteries Cantini, F., Niccoli, L., Salvarani, C., Padula, A., and Olivieri, I. (2001).
may experience symptoms of arm claudication for many months. Treatment of longstanding active giant cell arteritis with inflixi-
mab: report of four cases. Arthritis and Rheumatism, 44, 2933–5.
However, symptoms generally improve gradually as collateral
Caselli, R.J., Daube, J.R., Hunder, G.G., and Whisnant, J.P. (1988). Peripheral
circulation develops and ischaemic limb complications are very neuropathic syndromes in giant cell (temporal) arteritis. Neurology,
rare. In a subset of patients, revascularization procedures such as 38, 685–9.
stenting or vascular bypass surgery can be considered if disabling Chan, C.C., Paine, M., and O’Day, J. (2001). Steroid management in giant cell
symptoms persist. However, optimal control of the vascular inflam- arteritis. British Journal of Ophthalmology, 85, 1061–4.
mation with medical therapy is essential before endovascular pro- Chevalet, P., Barrier, J.H., Pottier, P., et al. (2000). A randomized, multi-
cedures or bypass surgery is performed. center, controlled trial using intravenous pulses of methylpredniso-
lone in the initial treatment of simple forms of giant cell arteritis: a
one year followup study of 164 patients. Journal of Rheumatology, 27,
References 1484–91.
Achkar, A.A., Lie, J.T., Hunder, G.G., O’Fallon, W.M., and Gabriel, S.E. Ciccia, F., Alessandro, R., Rizzo, A., et al. (2013). IL-33 is overexpressed in
(1994). How does previous corticosteroid treatment affect the biopsy the inflamed arteries of patients with giant cell arteritis. Annals of the
findings in giant cell (temporal) arteritis? Annals of Internal Medicine, Rheumatic Diseases, 72, 258–64.
120, 987–92. Crowson, C.S., Matteson, E.L., Myasoedova, E., et al. (2011). The lifetime risk
Ahmed, M.M., Mubashir, E., Hayat, S., Fowler, M., and Berney, S.M. (2007). of adult-onset rheumatoid arthritis and other inflammatory autoim-
Treatment of refractory temporal arteritis with adalimumab. Clinical mune rheumatic diseases. Arthritis andRheumatism, 63, 633–9.
Rheumatology, 26, 1353–5. Danesh-Meyer, H.V., Savino, P.J., Eagle, R.C., Jr., Kubis, K.C., and Sergott,
Aiello, P.D.,Trautmann, J.C., McPhee, T.J., Kunselman, A.R., and Hunder, G.G. R.C. (2000). Low diagnostic yield with second biopsies in suspected
(1993). Visual prognosis in giant cell arteritis. Ophthalmology, 100, 550–5. giant cell arteritis. Journal of Neuro-Ophthalmology, 20, 213–5.
Berger, C.T., Wolbers, M., Meyer, P., Daikeler, T., and Hess, C. (2009). High Dasgupta, B. (2010). Concise guidance: diagnosis and management of giant
incidence of severe ischaemic complications in patients with giant cell cell arteritis. Clinical Medicine, 10, 381–6.
arteritis irrespective of platelet count and size, and platelet inhibition. Dasgupta, B., Cimmino, M.A., Kremers, H.M., et al. (2012). 2012 Provisional
Rheumatology (Oxford), 48, 258–61. classification criteria for polymyalgia rheumatica: a European League
Besson, F.L., Parienti, J.J., Bienvenu, B., et al. (2011). Diagnostic performance Against Rheumatism/American College of Rheumatology collaborative
of (18)F-fluorodeoxyglucose positron emission tomography in giant initiative. Arthritis and Rheumatism, 64, 943–54.
cell arteritis: a systematic review and meta-analysis. European Journal of Deng, J., Ma-Krupa, W., Gewirtz, A.T., Younge, B.R., Goronzy, J.J., and
Nuclear Medicine and Molecular Imaging, 38, 1764–72. Weyand, C.M. (2009). Toll-like receptors 4 and 5 induce distinct types
Beyer, C., Axmann, R., Sahinbegovic, E., et  al. (2011). Anti-interleukin 6 of vasculitis. Circulation Research, 104, 488–95.
receptor therapy as rescue treatment for giant cell arteritis. Annals of the Deng, J., Younge, B.R., Olshen, R.A., Goronzy, J.J., and Weyand, C.M. (2010).
Rheumatic Diseases, 70, 1874–5. Th17 and Th1 T-cell responses in giant cell arteritis. Circulation, 121,
Birkhead, N.C., Wagener, H.P., and Shick, R.M. (1957). Treatment of tem- 906–15.
poral arteritis with adrenal corticosteroids; results in fifty-five cases De Silva, M. and Hazleman, B.L. (1986). Azathioprine in giant cell arteritis/
in which lesion was proved at biopsy. Journal of the American Medical polymyalgia rheumatica: a double-blind study. Annals of the Rheumatic
Association, 163, 821–7. Diseases, 45, 136–8.
Bley, T.A., Reinhard, M., Hauenstein, C., et al. (2008). Comparison of duplex de Vita, S., Tavoni, A., Jeracitano, G., Gemignani, G., Dolcher, M.P., and
sonography and high-resolution magnetic resonance imaging in the Bombardieri, S. (1992). Treatment of giant cell arteritis with cyclophos-
diagnosis of giant cell (temporal) arteritis. Arthritis and Rheumatism, phamide pulses. Journal of Internal Medicine, 232, 373–5.
58, 2574–8. Doury, P., Pattin, S., Eulry, F., and Thabaut, A. (1983). The use of dapsone
Bley, T.A., Uhl, M., Carew, J., et al. (2007). Diagnostic value of high-resolution in the treatment of giant cell arteritis and polymyalgia rheumatica.
MR imaging in giant cell arteritis. American Journal of Neuroradiology, Arthritis and Rheumatism, 26, 689–90.
28, 1722–7. Durand, M. and Thomas, S.L. (2012). Incidence of infections in patients
Blockmans, D., Bley, T., and Schmidt, W. (2009). Imaging for large-vessel vas- with giant cell arteritis: A cohort study. Arthritis Care Res (Hoboken),
culitis. Current Opinion in Rheumatology, 21, 19–28. 64, 581–8.
Bongartz, T. and Matteson, E.L. (2006). Large-vessel involvement in giant cell Evans, J.M., O’Fallon, W.M., and Hunder, G.G. (1995). Increased incidence
arteritis. Current Opinion in Rheumatology, 18, 10–7. of aortic aneurysm and dissection in giant cell (temporal) arteritis.
Boyev, L.R., Miller, N.R., and Green, W.R. (1999). Efficacy of unilateral versus A population-based study. Annals of Internal Medicine, 122, 502–7.
bilateral temporal artery biopsies for the diagnosis of giant cell arteritis. Gonzalez-Gay, M.A., Garcia-Porrua, C., Llorca, J., et al. (2000). Visual mani-
American Journal of Ophthalmology, 128, 211–5. festations of giant cell arteritis. Trends and clinical spectrum in 161
Brack, A., Martinez-Taboada, V., Stanson, A., Goronzy, J.J., and Weyand, patients. Medicine, 79, 283–92.
C.M. (1999). Disease pattern in cranial and large-vessel giant cell arteri- Gonzalez-Gay, M.A., Garcia-Porrua, C., Pineiro, A., Pego-Reigosa, R.,
tis. Arthritis and Rheumatism, 42, 311–7. Llorca,  J., and Hunder, G.G. (2004). Aortic aneurysm and dissection
CHAPTER 23 giant cell arteritis and polymyalgia rheumatica 317

in patients with biopsy-proven giant cell arteritis from northwestern Larson, T.S., Hall, S., Hepper, N.G., and Hunder, G.G. (1984). Respiratory
Spain: a population-based study. Medicine, 83, 335–41. tract symptoms as a clue to giant cell arteritis. Annals of Internal
Gonzalez-Gay, M.A., Vazquez-Rodriguez, T.R., Lopez-Diaz, M.J., et  al. Medicine, 101, 594–7.
(2009). Epidemiology of giant cell arteritis and polymyalgia rheumatica. Lee, M.S., Smith, S.D., Galor, A., and Hoffman, G.S. (2006). Antiplatelet and
Arthritis and Rheumatism, 61, 1454–61. anticoagulant therapy in patients with giant cell arteritis. Arthritis and
Grossman, J.M., Gordon, R., Ranganath, V.K., et  al. (2010). American Rheumatism, 54, 3306–9.
College of Rheumatology 2010 recommendations for the prevention Lee, Y.C., Padera, R.F., Noss, E.H., et al. (2012). Clinical course and manage-
and treatment of glucocorticoid-induced osteoporosis. Arthritis Care ment of a consecutive series of patients with “healed temporal arteritis”.
Res (Hoboken), 62, 1515–26. Journal of Rheumatology, 39, 295–302.
Han, J.W., Shimada, K., Ma-Krupa, W., et al. (2008). Vessel wall-embedded Le Guennec, P., Dromer, C., Sixou, L., Marc, V., Coustals, P., and Fournie, B.
dendritic cells induce T-cell autoreactivity and initiate vascular inflam- (1994). [Treatment of Horton disease. Value of synthetic antimalarials.
mation. Circulation Research, 102, 546–53. Apropos of a retrospective study of 36 patients]. Revue du Rhumatisme
Hayreh, S.S., Zimmerman, B., and Kardon, R.H. (2002). Visual improvement (ed. Française), 61, 485–90.
with corticosteroid therapy in giant cell arteritis. Report of a large study Lie, J.T. (1990). Illustrated histopathologic classification criteria for
and review of literature. Acta Ophthalmologica Scandinavica, 80, 355–67. selected vasculitis syndromes. American College of Rheumatology
Hernandez-Rodriguez, J., Garcia-Martinez, A., Casademont, J., et al. (2002). Subcommittee on Classification of Vasculitis. Arthritis and Rheumatism,
A strong initial systemic inflammatory response is associated with 33, 1074–87.
higher corticosteroid requirements and longer duration of therapy in Lie, J.T. (1995). Aortic and extracranial large vessel giant cell arteritis:  a
patients with giant-cell arteritis. Arthritis and Rheumatism, 47, 29–35. review of 72 cases with histopathologic documentation. Seminars in
Hoffman, G.S., Cid, M.C., Hellmann, D.B., et al. (2002). A multicenter, rand- Arthritis and Rheumatism, 24, 422–31.
omized, double-blind, placebo-controlled trial of adjuvant methotrexate Ma-Krupa, W., Jeon, M.S., Spoerl, S., Tedder, T.F., Goronzy, J.J., and Weyand,
treatment for giant cell arteritis. Arthritis and Rheumatism, 46, 1309–18. C.M. (2004). Activation of arterial wall dendritic cells and breakdown
Hoffman, G.S., Cid, M.C., Rendt-Zagar, K.E., et  al. (2007). Infliximab for of self-tolerance in giant cell arteritis. Journal of Experimental Medicine,
maintenance of glucocorticosteroid-induced remission of giant cell 199, 173–83.
arteritis: a randomized trial. Annals of Internal Medicine, 146, 621–30. Mahr, A., Saba, M., Kambouchner, M., et al. (2006). Temporal artery biopsy
Hunder, G.G., Bloch, D.A., Michel, B.A., et al. (1990). The American College for diagnosing giant cell arteritis: the longer, the better? Annals of the
of Rheumatology 1990 criteria for the classification of giant cell arteritis. Rheumatic Diseases, 65, 826–8.
Arthritis and Rheumatism, 33, 1122–8. Mahr, A.D., Jover, J.A., Spiera, R.F., et al. (2007). Adjunctive methotrexate for
Hunder, G.G., Sheps, S.G., Allen, G.L., and Joyce, J.W. (1975). Daily and treatment of giant cell arteritis: an individual patient data meta-analysis.
alternate-day corticosteroid regimens in treatment of giant cell arte- Arthritis and Rheumatism, 56, 2789–97.
ritis:  comparison in a prospective study. Annals of Internal Medicine, Martinez-Taboada, V., Hunder, N.N., Hunder, G.G., Weyand, C.M., and
82, 613–8. Goronzy, J.J. (1996). Recognition of tissue residing antigen by T cells
Jover, J.A., Hernandez-Garcia, C., Morado, I.C., Vargas, E., Banares, A., and in vasculitic lesions of giant cell arteritis. Journal of Molecular Medicine,
Fernandez-Gutierrez, B. (2001). Combined treatment of giant-cell arte- 74, 695–703.
ritis with methotrexate and prednisone. a randomized, double-blind, Mazlumzadeh, M., Hunder, G.G., Easley, K.A., et al. (2006). Treatment of
placebo-controlled trial. Annals of Internal Medicine, 134, 106–14. giant cell arteritis using induction therapy with high-dose glucocorti-
Kaiser, M., Weyand, C.M., Bjornsson, J., and Goronzy, J.J. (1998). coids: a double-blind, placebo-controlled, randomized prospective clin-
Platelet-derived growth factor, intimal hyperplasia, and ischemic com- ical trial. Arthritis and Rheumatism, 54, 3310–8.
plications in giant cell arteritis. Arthritis and Rheumatism, 41, 623–33. Miller, D.V. and Maleszewski, J.J. (2011). The pathology of large-vessel vascu-
Karassa, F.B., Matsagas, M.I., Schmidt, W.A., and Ioannidis, J.P. (2005). litides. Clinical and Experimental Rheumatology, 29, S92–8.
Meta-analysis: test performance of ultrasonography for giant-cell arte- Moosig, F., Czech, N., Mehl, C., et  al. (2004). Correlation between
ritis. Annals of Internal Medicine, 142, 359–69. 18-fluorodeoxyglucose accumulation in large vessels and serological
Kermani, T.A., Matteson, E.L., Hunder, G.G., and Warrington, K.J. (2009). markers of inflammation in polymyalgia rheumatica: a quantitative PET
Symptomatic lower extremity vasculitis in giant cell arteritis:  a case study. Annals of the Rheumatic Diseases, 63, 870–3.
series. Journal of Rheumatology, 36, 2277–83. Myklebust, G. and Gran, J.T. (1996). A prospective study of 287 patients with
Kermani, T.A., Schafer, V.S., Crowson, C.S., et  al. (2010). Increase in age polymyalgia rheumatica and temporal arteritis: clinical and laboratory
at onset of giant cell arteritis: a population-based study. Annals of the manifestations at onset of disease and at the time of diagnosis. British
Rheumatic Diseases, 69, 780–1. Journal of Rheumatology, 35, 1161–8.
Kermani, T.A., Schmidt, J., Crowson, C.S., et al. (2012). Utility of erythrocyte Nesher, G., Berkun, Y., Mates, M., Baras, M., Rubinow, A., and Sonnenblick,
sedimentation rate and C-reactive protein for the diagnosis of giant cell M. (2004). Low-dose aspirin and prevention of cranial ischemic
arteritis. Seminars in Arthritis and Rheumatism, 41, 866–71. complications in giant cell arteritis. Arthritis and Rheumatism, 50,
Kermani, T.A. and Warrington, K.J. (2012). Polymyalgia rheumatica. Lancet. 1332–7.
Kermani, T.A., Ytterberg, S.R., and Warrington, K.J. (2011). Pneumocystis Niederkohr, R.D. and Levin, L.A. (2007). A Bayesian analysis of the true
jiroveci pneumonia in giant cell arteritis: A case series. Arthritis Care Res sensitivity of a temporal artery biopsy. Investigative Ophthalmology and
(Hoboken), 63, 761–5. Visual Science, 48, 675–80.
Kyle, V. (1991). Laboratory investigations including liver in polymyalgia Nuenninghoff, D.M., Hunder, G.G., Christianson, T.J., McClelland, R.L., and
rheumatica/giant cell arteritis. Bailliere’s Clinical Rheumatology, 5, Matteson, E.L. (2003a). Incidence and predictors of large-artery com-
475–84. plication (aortic aneurysm, aortic dissection, and/or large-artery steno-
Kyle, V., Cawston, T.E., and Hazleman, B.L. (1989). Erythrocyte sedimenta- sis) in patients with giant cell arteritis: a population-based study over
tion rate and C reactive protein in the assessment of polymyalgia rheu- 50 years. Arthritis and Rheumatism, 48, 3522–31.
matica/giant cell arteritis on presentation and during follow up. Annals Nuenninghoff, D.M., Hunder, G.G., Christianson, T.J., McClelland, R.L., and
of the Rheumatic Diseases, 48, 667–71. Matteson, E.L. (2003b). Mortality of large-artery complication (aortic
Kyle, V. and Hazleman, B.L. (1989). Treatment of polymyalgia rheumatica aneurysm, aortic dissection, and/or large-artery stenosis) in patients
and giant cell arteritis. II. Relation between steroid dose and steroid with giant cell arteritis: a population-based study over 50 years. Arthritis
associated side effects. Annals of the Rheumatic Diseases, 48, 662–6. and Rheumatism, 48, 3532–7.
318 SECTION 5 vasculitic diseases and syndromes and related disorders

Parikh, M., Miller, N.R., Lee, A.G., et  al. (2006). Prevalence of a normal Seitz, M., Reichenbach, S., Bonel, H.M., Adler, S., Wermelinger, F., and
C-reactive protein with an elevated erythrocyte sedimentation rate in Villiger, P.M. (2011). Rapid induction of remission in large vessel vascu-
biopsy-proven giant cell arteritis. Ophthalmology, 113, 1842–5. litis by IL-6 blockade. A case series. Swiss Medical Weekly, 141, w13156.
Piggott, K., Biousse, V., Newman, N.J., Goronzy, J.J., and Weyand, C.M. (2009). Smetana, G.W. and Shmerling, R.H. (2002). Does this patient have temporal
Vascular damage in giant cell arteritis. Autoimmunity, 42, 596–604. arteritis? Journal of the American Medical Association, 287, 92–101.
Piggott, K., Deng, J., Warrington, K., et  al. (2011). Blocking the NOTCH Spiera, R.F., Mitnick, H.J., Kupersmith, M., et  al. (2001). A prospective,
pathway inhibits vascular inflammation in large-vessel vasculitis. double-blind, randomized, placebo controlled trial of methotrexate in
Circulation, 123, 309–18. the treatment of giant cell arteritis (GCA). Clinical and Experimental
Pipitone, N. and Salvarani, C. (2008). Improving therapeutic options for Rheumatology, 19, 495–501.
patients with giant cell arteritis. Current Opinion in Rheumatology, Taylor-Gjevre, R., Vo, M., Shukla, D., and Resch, L. (2005). Temporal artery
20, 17–22. biopsy for giant cell arteritis. Journal of Rheumatology, 32, 1279–82.
Pless, M., Rizzo, J.F., 3rd, Lamkin, J.C., and Lessell, S. (2000). Concordance Unizony, S., Arias-Urdaneta, L., Miloslavsky, E., et al. (2012). Tocilizumab
of bilateral temporal artery biopsy in giant cell arteritis. Journal of for the treatment of large-vessel vasculitis (giant cell arteritis, Takayasu
Neuro-Ophthalmology, 20, 216–8. arteritis) and polymyalgia rheumatica. Arthritis Care Res (Hoboken), 64,
Ponge, T., Barrier, J.H., Grolleau, J.Y., Ponge, A., Vlasak, A.M., and Cottin, 1720–9.
S. (1988). The efficacy of selective unilateral temporal artery biopsy Wagner, A.D., Goronzy, J.J., and Weyand, C.M. (1994). Functional profile
versus bilateral biopsies for diagnosis of giant cell arteritis. Journal of of tissue-infiltrating and circulating CD68+ cells in giant cell arteri-
Rheumatology, 15, 997–1000. tis. Evidence for two components of the disease. Journal of Clinical
Pountain, G.D., Calvin, J., and Hazleman, B.L. (1994). Alpha Investigation, 94, 1134–40.
1-antichymotrypsin, C-reactive protein and erythrocyte sedimentation Walvick, M.D. and Walvick, M.P. (2011). Giant cell arteritis: laboratory pre-
rate in polymyalgia rheumatica and giant cell arteritis. British Journal of dictors of a positive temporal artery biopsy. Ophthalmology, 118, 1201–4.
Rheumatology, 33, 550–4. Warrington, K.J. and Matteson, E.L. (2007). Management guidelines and out-
Prieto-Gonzalez, S., Arguis, P., Garcia-Martinez, A., et al. (2012). Large ves- come measures in giant cell arteritis (GCA). Clinical and Experimental
sel involvement in biopsy-proven giant cell arteritis: prospective study Rheumatology, 25, 137–41.
in 40 newly diagnosed patients using CT angiography. Annals of the Warrington, K.J. and Matteson, E.L. (2009). Personalized medicine for the
Rheumatic Diseases, 71, 1170–6. vasculitis patient: hope for the future? International Journal of Clinical
Proven, A., Gabriel, S.E., Orces, C., O’Fallon, W.M., and Hunder, G.G. (2003). Rheumatology, 4, 127–32.
Glucocorticoid therapy in giant cell arteritis: duration and adverse out- Warrington, K.J., Scheithauer, B.W., and Michet, C.J. (2003). Acute myeloid
comes. Arthritis and Rheumatism, 49, 703–8. leukemia associated with necrotizing temporal arteritis. Journal of
Pryshchep, O., Ma-Krupa, W., Younge, B.R., Goronzy, J.J., and Weyand, C.M. Rheumatology, 30, 846–8.
(2008). Vessel-specific Toll-like receptor profiles in human medium and Weyand, C.M., Fulbright, J.W., Hunder, G.G., Evans, J.M., and Goronzy,
large arteries. Circulation, 118, 1276–84. J.J. (2000). Treatment of giant cell arteritis: interleukin-6 as a biologic
Ray-Chaudhuri, N., Kine, D.A., Tijani, S.O., et al. (2002). Effect of prior ster- marker of disease activity. Arthritis and Rheumatism, 43, 1041–8.
oid treatment on temporal artery biopsy findings in giant cell arteritis. Weyand, C.M. and Goronzy, J.J. (2003a). Giant-cell arteritis and polymyalgia
British Journal of Ophthalmology, 86, 530–2. rheumatica. Annals of Internal Medicine, 139, 505–15.
Restuccia, G., Cavazza, A., Boiardi, L., et al. (2012). Small-vessel vasculitis Weyand, C.M. and Goronzy, J.J. (2003b). Medium- and large-vessel vasculi-
surrounding an uninflamed temporal artery and isolated vasa vaso- tis. New England Journal of Medicine, 349, 160–9.
rum vasculitis of the temporal artery: two subsets of giant cell arteritis. Weyand, C.M., Hicok, K.C., Hunder, G.G., and Goronzy, J.J. (1992). The
Arthritis and Rheumatism, 64, 549–56. HLA-DRB1 locus as a genetic component in giant cell arteritis. Mapping
Rittner, H.L., Kaiser, M., Brack, A., Szweda, L.I., Goronzy, J.J., and Weyand, of a disease-linked sequence motif to the antigen binding site of the
C.M. (1999). Tissue-destructive macrophages in giant cell arteritis. HLA-DR molecule. Journal of Clinical Investigation, 90, 2355–61.
Circulation Research, 84, 1050–8. Weyand, C.M., Hicok, K.C., Hunder, G.G., and Goronzy, J.J. (1994a). Tissue
Salvarani, C., Cantini, F., Boiardi, L., and Hunder, G.G. (2002). Polymyalgia cytokine patterns in patients with polymyalgia rheumatica and giant cell
rheumatica and giant-cell arteritis. New England Journal of Medicine, arteritis. Annals of Internal Medicine, 121, 484–91.
347, 261–71. Weyand, C.M., Kaiser, M., Yang, H., Younge, B., and Goronzy, J.J. (2002).
Salvarani, C., Cantini, F. and Hunder, G.G. (2008). Polymyalgia rheumatica Therapeutic effects of acetylsalicylic acid in giant cell arteritis. Arthritis
and giant-cell arteritis. Lancet, 372, 234–45. and Rheumatism, 46, 457–66.
Salvarani, C., Della Bella, C., Cimino, L., et al. (2009). Risk factors for severe Weyand, C.M., Schonberger, J., Oppitz, U., Hunder, N.N., Hicok, K.C., and
cranial ischaemic events in an Italian population-based cohort of Goronzy, J.J. (1994b). Distinct vascular lesions in giant cell arteritis
patients with giant cell arteritis. Rheumatology (Oxford), 48, 250–3. share identical T cell clonotypes. Journal of Experimental Medicine, 179,
Schaufelberger, C., Mollby, H., Uddhammar, A., Bratt, J., and Nordborg, E. 951–60.
(2006). No additional steroid-sparing effect of cyclosporine A in giant Weyand, C.M., Tetzlaff, N., Bjornsson, J., Brack, A., Younge, B., and Goronzy,
cell arteritis. Scandinavian Journal of Rheumatology, 35, 327–9. J.J. (1997). Disease patterns and tissue cytokine profiles in giant cell arte-
Schmidt, W.A., Kraft, H.E., Vorpahl, K., Volker, L., and Gromnica-Ihle, E.J. ritis. Arthritis and Rheumatism, 40, 19–26.
(1997). Color duplex ultrasonography in the diagnosis of temporal arte- Weyand, C.M., Wagner, A.D., Bjornsson, J., and Goronzy, J.J. (1996).
ritis. New England Journal of Medicine, 337, 1336–42. Correlation of the topographical arrangement and the functional pat-
Schmidt, W.A., Natusch, A., Moller, D.E., Vorpahl, K., and Gromnica-Ihle, E. tern of tissue-infiltrating macrophages in giant cell arteritis. Journal of
(2002). Involvement of peripheral arteries in giant cell arteritis: a color Clinical Investigation, 98, 1642–9.
Doppler sonography study. Clinical and Experimental Rheumatology, 20, Weyand, C.M., Younge, B.R., and Goronzy, J.J. (2011). IFN-gamma and
309–18. IL-17: the two faces of T-cell pathology in giant cell arteritis. Current
Schmidt, W.A., Seifert, A., Gromnica-Ihle, E., Krause, A., and Natusch, A. Opinion in Rheumatology, 23, 43–9.
(2008). Ultrasound of proximal upper extremity arteries to increase Younge, B.R., Cook, B.E., Jr., Bartley, G.B., Hodge, D.O., and Hunder, G.G.
the diagnostic yield in large-vessel giant cell arteritis. Rheumatology (2004). Initiation of glucocorticoid therapy:  before or after temporal
(Oxford), 47, 96–101. artery biopsy? Mayo Clinic Proceedings, 79, 483–91.
CHAPTER 24

Takayasu’s arteritis
Yasushi Kobayashi, Tomohiro Ishii, and
Hideo Harigae

Introduction in 2005 was 4.2 per 100 000 persons, and in Sweden and the USA
it was reported as 0.26–0.64 per 100 000 person (Hall et al. 1985;
Takayasu’s arteritis (TA) is a chronic granulomatous vasculitis char- Waern et al. 1983).
acterized by stenosis, occlusion, and sometimes aneurysm of large TA causes different aortic lesions in different countries. TA in
elastic arteries, and mainly affecting the aorta and its main branches Japan, Korea, and China affects mainly the aortic arch. In contrast,
including pulmonary and coronary arteries (Jennette et al. 1994; TA in south Asian countries, such as India and Thailand, and in
Kerr et al. 1994; Numano 2000; Sharma et al. 1996) (Figure 24.1). TA Turkey, Israel, and Middle Eastern countries frequently affects the
generally occurs in young, particularly Asian, women during their abdominal aorta (Yajima et al. 1994).
reproductive period (Cipriano et al. 1977; Numano et al. 2000). Moreover, the female to male ratio of TA appears to decline from
TA was described by Mikito Takayasu, Professor of the Far East toward the West (Yajima et al. 1994). The apparent var-
Ophthalmology of Kanazawa University, Japan, at the 12th Annual iation in prevalence and phenotype of TA in different geographical
Meeting of the Japanese Ophthalmology Society held in 1905 areas may reflect genetic factors as in giant cell arteritis, which is
(Takayasu 1908; Numano and Kakuta 1996). The patient was frequently found in Caucasian populations but not in others.
a 21-year-old woman who had a peculiar, wreath-like, arterio- TA rarely affects very young children (Sharma et al. 1991; Millar
venous anastomosis (described as coronary anastomosis) around et al. 1996; Mitchell and Parisi 1997; Hari et al. 2000; Stanley et al.
the papilla due to ischaemia. The term Takayasu’s arteritis was 2003; Ladhani et al. 2001; D’Souza et al. 1998; Martini 1995; Cloux
first used in 1942 (Shinmi 1942) and subsequently by many other Blasco et al. 1991; Rose et al. 1990; Chiasson et al. 1990; Hong et al.
researchers (Caccamise and Okuda 1954; Azizi and Rafat 1956; 1992; Morales et al. 1991). These juvenile-onset type TA may differ
Jacobsen and Rasch 1956; Roedenbeck and Bauer 1956; Lobato from adult TA in its pathogenesis.
1956; Jimenez Casado and Moncada Moneu 1956). Morgagni
reported a 40-year-old woman in Italy in 1761, whose radial pulses
were not palpable for many years (Altschuler and Wheat 2000). In Aetiology and pathogenesis
1856, Savory described a young woman in whom the main arter-
ies of both upper extremities and of the left side of the neck were The aetiology of TA still remains unknown; however, autoreactive
completely obliterated (Savory 1856). Both these cases were likely lymphocytes that may recognize antigens expressing around vasa
to have been TA. TA has historically been called various names, vasorum are proposed to play a major role in the pathogenesis in
including pulseless disease (Jimenez Casado and Moncada Moneu TA, even though these lymphocytes are not yet proven.
1956), aortitis syndrome (Kawai et al. 1975), aortic arch syndrome HLA studies of TA population in Japan revealed that positive asso-
(Ross and McKusick 1953), long-segment atypical coarctation of ciations between TA and MHC class II alleles HLA-DRB1*1502 and
the aorta (Hatano et al. 1975), Martorell’s syndrome (Planas 1978), DPB1* 0901 and MHC class I alleles HLA-B*5201 and HLA-B*3902
and occlusive thromboaortopathy (Ishikawa 1975). (Isohisa et al. 1978; Naito et al. 1978; Yoshida et al. 1993; Kimura
et al. 2000). DRB1*1502 and DPB1*0901 were shown to be in strong
linkage disequilibrium with HLA-B*5201 in the Japanese popula-
Epidemiology tion (Kimura et al. 2000). Numano et al. reported that TA patients
Although TA has been reported all over the world, there is a wide with HLA-Bw52 (HLA-B*5201) exhibited more severe inflamma-
variation in its prevalence in different geographical regions. TA tion and a higher degree of aortic insufficiency than those without
is believed to be predominantly found in Asia (Koide 1992; Park Bw52 (Moriwaki and Numano 1992). In addition, HLA-B*3902
and Park 1992; Zheng et al. 1992; Sharma et al. 1992; Piyachon and may be associated with renal vascular lesions in the Japanese TA
Suwanwela 1992), the Middle East (Rosenthal et al. 1992; Turkoglu population (Kitamura et  al. 1998). The association of HLA with
et al. 1996; Ureten et al. 2004; el-Reshaid et al. 1995), and South TA was also reported in North and South America: TA in Mexico
America (Canas et  al. 1998; Robles and Reyes 1994; Sato et  al. with HLA-DRB1*1301 (Girona et al. 1996) and HLA-DRB1*1602
1998). Nevertheless, patients with TA have been increasingly recog- and DRB1*1001 with TA in Colombia (Salazar et al. 2000). HLA
nized in Africa (Mwipatayi et al. 2005), Europe (Vanoli et al. 1995), B67 has been identified as an important candidate marker of TA
and North America (Kerr et al. 1994). The incidence of TA in Japan (Takamura et al. 2012).
320 SECTION 5 vasculitic diseases and syndromes and related disorders

the adventitia (Seko et al. 1994). They showed that infiltrating cells,
such as γδ T cells, NK cells, and CD8 T cells, release massive amount
of perforin, which may cause tissue damage. They also showed
that the expression of co-stimulatory molecules 4-1BBL, Fas, and
MICA were induced in the affected aortic tissue in TA (Seko et al.
2004). These data suggested that γδT cells infiltrated aortic tissue
recognizing MICA, and resulting in the induction of MHC anti-
gens and co-stimulatory molecules, and infiltrating αβT cells that
recognized autoantigens presented by MHC antigens, leading to
chronic inflammation. An apoptosis pathway is also involved in the
tissue injury in TA. They showed furthermore that T cells from the
affected aortic tissue of TA had limited repertoires of TCR Vα and
Vβ gene transcripts compared with atherosclerosis, which may sug-
gest that autoreactive lymphocytes are involved in the inflamma-
tion of TA (Seko et al. 1996). Recently, Th17 cells were shown to be
implicated in the pathogenesis of giant cell arteritis; interestingly,
prednisolone suppressed Th17 but not Th1 cells (Deng et al. 2010).
Th17 may play a role also in the development of TA.
The formation of granulomata may ensue under the control of
proinflammatory cytokines such as INF-γ produced by T cells and
TNF-α and IL-6, products of T cells and activated macrophages
(Weyand and Goronzy 2003). The serum proinflammatory
cytokines, IL-12, TNF-α, IL-6, and IL-1, are elevated in the serum
of patients with active TA and may promote granulomatous vas-
cular lesion formation in TA (Park et al. 2006; Tripathy et al. 2004;
Verma et  al. 2005). Recently, anti-TNF-α and anti-IL-6 receptor
Fig. 24.1 Three-dimensional reconstituted enhanced CT showing aneurysm antibodies were reported to control the inflammation in TA clini-
formation in the left common carotid artery and obstruction in the left cally (Hoffman et al. 2004; Schafer and Zwerina 2012; Comarmond
subclavian artery. et al. 2012). The findings clearly suggested that these proinflamma-
tory cytokines should have important roles in the development of
inflammation in TA.
Monozygotic twins cases suggests that a genetic component plays Antiendothelial cell antibodies (AECA) have been reported
a role in the development of TA (Numano et  al. 1978; Sasazuki in the serum of TA patients (Eichhorn et  al. 1996). AECA may
et al. 1979; Kobayashi and Numano 2002). Polymorphisms in the activate endothelial cells and be involved in the pathogenesis of
NFKBIL1 gene, which encodes a putative ankyrin repeat protein, TA (Blank et al. 1999; Kornberg et al. 2000; Tripathy et al. 2001).
have higher NFKBIL1 promoter activity in vitro and appear to be Antineutrophil cytoplasmic antibodies (ANCA), often found in
associated with TA (Shibata et al. 2006). Although the function of many primary vasculitis syndromes, were reported to be associated
NFKBIL1 is unknown, it may confer the proinflammatory environ- with TA; however, the roles of B cells and antibodies in TA are still
ment necessary for the development of TA. controversial.
The external trigger that may break the tolerance of autoreactive Several animal models of aortitis have been described. Virgin and
lymphocytes in TA remains unknown. Tuberculosis was suspected colleagues reported that a g-herpes virus induced aortitis in IFN-γ
for a long time as an aetiological factor of TA in India and Mexico knockout mice (Weck et al. 1997; Dal Canto et al. 2000). A g-herpes
(Lupi-Herrera et al. 1977; Sen et al. 1972). Tubercle bacilli injected virus, gHV68, causes inflammation in the elastic arteries in mice;
into rabbits induced inflammatory changes resembling those of however, autoimmunity was not responsible for the aortitis, which
TA, and the presence of giant cell granulomata resembling tuber- was related to persistent virus replication (Dal Canto and Virgin
culosis follicles in the arterial wall suggested concomitant tuber- 2000). An a-herpes virus was reported to cause large-vessel inflam-
culosis (Sen et al. 1972). However Sharma and colleagues did not mation in chickens (Fabricant 1985), and b-herpes virus and
find an association between TA and tuberculosis and reported that murine CMV also cause aortic inflammation.
the incidence of tuberculosis in TA was not higher than that in the Nicklin and Iwakura have independently studied an interleukin-1
general population (Jain et  al. 1996; Sharma et  al. 1992; Kothari receptor antagonist (IL-1ra) knockout mouse, in which IL-1 sig-
1995). Tuberculosis may be a trigger of TA, but tuberculosis itself nalling is enhanced. IL-1ra-deficient mice developed transmural
is now considered not to be a causative agent of TA. Cases of TA inflammation in elastic arteries at the site of turbulent flow, result-
after hepatitis B vaccination have been reported (Castresana-Isla ing in inflammation at the root of the aorta with massive infiltra-
et al. 1993; Zaas et al. 2001). The host response to infection or other tion of macrophages and monocytes and loss of lamellae in the
external stress may be a trigger of TA. aortic media. Adoptive transfer experiments showed that T cells
Seko et al. showed that innate lymphocytes, γδT cells, predomi- from IL-1ra knockout mice induced aortitis in nu/nu mice. These
nantly infiltrated diseased aortic tissue from TA patients. A 65-kD findings suggested that T cells may play an important role in its
heat shock protein, which may be one of the targets of γδT cells, was pathogenesis. Iwakura also reported that IL-1 and TNF-α are key
expressed at high levels in the media and along the vasa vasorum in regulators in the development of inflammation in the aorta in their
CHAPTER 24 takayasu’s arteritis 321

model (Nicklin et al. 2000; Shepherd and Nicklin 2005; Matsuki


et al. 2005).
TA has been reported to be associated with inflammatory
bowel diseases, including Crohn’s disease (Lenhoff and Mee 1982;
Owyang et  al. 1979; Friedman and Tegtmeyer 1979; Yassinger
et al. 1976; Baqir et al. 2007) and ulcerative colitis (Numano et al.
1996; Sakhuja et al. 1990; Achar and Al-Nakib 1986; Miwa et al.
1979; Chapman et al. 1978; Morita et al. 1996). These findings sug-
gested that TA might share common pathological mechanisms with
inflammatory bowel diseases.
TA rarely affects toddlers. The pathogenesis of juvenile-onset-type
TA may be dependent on genetic defects in innate immune
response. An autoinflammatory disease of very young children,
Blau’s syndrome, is a rare condition typically defined by granu-
lomatous arthritis, skin eruption, and uveitis in the absence of
lung or other visceral involvement (Blau 1985). Some variants of
Blau’s syndrome cause large-vessel arteritis (Wang et  al. 2002).
Mutations in the NOD2 gene found in Blau’s syndrome have been
reported to activate NFκΒ constitutively (Miceli-Richard et  al.
2001). Early-onset TA may partially overlap with Blau’s syndrome
(Rose et al. 1990; Schapiro et al. 1994; Fernandes et al. 2000; Hilario
et al. 1998).

Histopathological findings Fig. 24.2 A section of the aorta showing intimal thickening, moth-eaten
Takayasu’s arteritis is defined as an inflammatory vasculopathy that appearance in the outer media, cellular infiltrate around the vasa vasorum, and
involves mainly large and middle-size vessels. TA may be divided non-cellular thickening adventitia as commonly found in TA.
into an acute, florid inflammatory phase, and a chronic healed
fibrotic phase. Both types may be seen simultaneously, suggesting
recurrent inflammation in TA (Gravanis 2000). et al. 2005). The aorta is thick and often rigid, due to fibrosis of all
In the acute phase, the inflammatory lesions originate in the vasa three arterial layers, notably adventitia and intima. Extension of
vasorum in the aorta and are characterized by perivascular cuffing, adventitial fibrosis and inflammatory cell infiltration to the adjacent
mainly composed of γδT lymphocytes, cytotoxic T lymphocytes, structures is sometimes observed. A characteristic of TA is that the
and T helper cells (Figure  24.2). Luminal stenosis of adventitial aortic lumen is narrowed in a ‘fusion that has skipped’ area. These
small arteries due to intimal thickening is relatively common. An areas of narrowing may alternate with aneurysmal dilatations of the
increase in adventitial thickness due to fibrosis is a histopathologi- aortic wall. Ascending aortitis almost always results in dilatation of
cal feature found in the chronic phase of TA. Inflammatory infil- the aorta but not narrowing, perhaps due to haemodynamic influ-
trates in the arterial wall are typically arranged in granulomas that ences. The gross appearance of the intima may be ‘cobblestoned’
are dependent on T cells regulating the activity and integrity of with round, well-circumscribed, variably-sized, white, gelatinous,
macrophages. Neovascularization, often accompanied by infiltrates smooth patches with intervening normal intima. The thick intima
of lymphocytes, plasma cells, and occasionally Langhans-type giant may have the gross appearance of tree bark, due to longitudinal
cells, is seen in the media. Giant cells of the foreign body type are wrinkling and ridges, and the affected aorta in end-stage TA may
sometimes strewn around degraded elastic tissue and patches of have a ‘lead pipe’ appearance.
medial coagulation necrosis surrounded by a fence-like arrange- Approximately 50% of patients with TA have pulmonary artery
ment of epithelioid cells are occasionally noted at the periphery involvement. The morphological changes in the pulmonary arterial
(Gravanis 2000). Active inflammation may be present in the arte- wall, such as inflammation and fibrosis of the adventitia and media
rial circuit even when the disease appears to be clinically silent. The and thickening of the intima secondary to fibrosis, are similar to
activity of the disease often persists and new lesions in the arter- those in the aorta (Yamada et al. 1992).
ies continue to appear (Bali et al. 1998). The inflammation caused
by TA does not damage the aorta uniformly and it creates patchy
lesions; usually the prominent lesion appears and disappears ran- Natural course
domly (Gravanis 2000; Nasu 1982). Paralleling the histopathological changes, the clinical course of
In the chronic stage of TA, intimal fibrosis is often accompanied TA typically progresses through two basic stages, an acute and a
by well-formed fibrous atherosclerotic plaques and lamellar calci- chronic stage. Early in the acute stage, symptoms are non-specific
fied lesions. Extension of the adventitial fibrosis and mononuclear features of aortic inflammation, and include fever, fatigabil-
cell (Sharma et  al. 1998)  infiltration to adjacent structures may ity, weight loss, arthralgias, haemoptysis, and tachycardia. After
result in retroperitoneal fibrosis, and extension of similar fibrotic weeks, months, and years in some cases, vascular lesions appear
processes to the pericardium may induce a fibrosing pericarditis. and cause ischaemic symptoms due to stenotic lesions or throm-
Inflammation may extend to the heart, causing myocarditis (Takeda bus formation. Almost all the patients have ischaemic disorders of
322 SECTION 5 vasculitic diseases and syndromes and related disorders

the involved vessels, which can present as dizziness, syncope, visual common (Li-Xin et al. 2011). Hypertension may be caused from
disturbances, faint or absent pulses, or differences in systolic blood atypical coarctation of the aorta, loss of vascular compliance, aortic
pressure between arms (Ishikawa and Maetani 1994; Iwai et  al. insufficiency, or renal artery stenosis (Abe et al. 1976; Ask-Upmark
2000). Twenty per cent of patients have a self-limited, monophasic 1961; Sharma et al. 1985). Severe hypertension may carry a poor
inflammatory episode; however, a progressive and relapsing course prognosis.
is the rule. Serial angiographic studies show that new lesions can be Inflammatory bowel syndromes such as Crohn’s disease, ulcer-
found in 61% of patients even when the disease is thought to be in ative colitis, and non-specific inflammatory colitis have been
the chronic phase (Hoffman et al. 1994). Subsequent recurrence of associated with TA (Baqir et  al. 2007), and sometimes gastroin-
TA is found in 20% of Japanese TA patients in remission. Among testinal bleeding is the first symptom in TA. Erythema nodosum
the complications of TA, cardiovascular complications such as aor- is reported as the most common skin lesion in TA in Caucasian
tic insufficiency and hypertension are strongly associated with a populations, and ulcerative, subacute, nodular lesions have been
poor prognosis. described in Mexico and Japan. Pyoderma gangrenosum, papulo-
Ischaemia and thrombosis in TA cause ocular complications. necrotic eruptions, nodular erythematous lesions, facial lupus-like
Emboli may cause sudden blindness in the acute stage. Ischaemic rashes, and panniculitis also occur. Skin lesions tend to occur with-
retinopathy is often classified into three stages: stage I, venous out any relationship to the sites of vascular lesions; however, patho-
dilatation; stage II, microaneurysm formation; and stage III, logical findings include granulomatous features suggesting a shared
arteriovenous anastomosis (Ishikawa et  al. 1983; Vedantham pathogenesis.
et al. 2005; Noel et al. 2013). Hypertensive retinopathy is more Aortic insufficiency is present in almost one-third of TA
commonly seen with TA. Other ocular complications of TA patients in the Japanese population; it may be caused by annu-
include cataracts, optic atrophy, loss of eye reflexes, iris atrophy, loaortic ectasia. Left ventricular hypertrophy in TA is observed
and rubeosis iris. in the presence of aortic insufficiency and hypertension and
TA patients have a higher rate of atherosclerotic plaques, as often these may eventually result in heart failure, and carry a poor
observed among autoimmune disease patients (Seyahi et al. 2006). prognosis. Inflammation of TA involving the ostium of coronary
Activated immune system in TA may exacerbate the development arteries may lead to acute myocardial infarction or exertional
of atherosclerosis. angina (Matsubara et  al. 1992). Pulmonary lesions were found
in 15% of TA patients by angiography; however, clinical symp-
toms such as haemoptysis are rare (Frankel et al. 2006). Isolated
Clinical features pulmonary arteritis has also been reported (Nakabayashi et al.
In the acute stage, the symptoms of TA are usually non-specific, 1996; Lie 1996).
generalized inflammatory symptoms, as mentioned in section
Natural Course. Early diagnosis is not easy because symptoms are
not specific and unique disease markers do not exist. Therefore Laboratory findings
some TA patients have been left with diagnoses such as fever of The most frequent laboratory abnormality is an elevated erythro-
unknown origin or non-specific chronic inflammatory disease. cyte sedimentation rate (ESR), which is considered a good index
Progress with 18F-fluorodeoxyglucose positron emission tomog- of disease activity. C-reactive protein (CRP) is also used as a sur-
raphy/ computed tomography (18F-FDG PET/CT) analyses can rogate marker of the disease. Non-specific leucocytosis, hypergam-
identify aortic inflammation even before a vascular lesion appears maglobulinaemia, mild anaemia, and elevation of complement
(Walter 2007; Umekita et  al. 2006). The disease is characterized components are features, as are coagulopathies and platelet activa-
by a specific predilection for women, though the female-to-male tion in the acute stage. 18F-FDG PET analyses showed that inflam-
ratio varies as follows: Japan 9 : 1, Korea 6.6 : 1, China 2.9 : 1, India matory activity did not always correlate with CRP and ESR levels
1.6 : 1, USA 29.1 : 1, Mexico 5.3 : 1 (Zheng et al. 1992; Hoffman (Kobayashi et  al. 2005). ECG may show signs of left ventricular
et al. 1994; Koide 1992; Lupi-Herrera et al. 1977; Park et al. 1992; hypertrophy (Hashimoto et al. 1992a).
Bali et al. 1998). The age of onset in Japanese and Indian patients
is 15–35 years. A majority of TA patients present symptoms due to
vascular lesions. TA in Far Eastern countries affects the aortic arch Radiological findings
and its tributaries most often, leading to decreased or absent pulses A comprehensive angiogram is usually performed to diagnose
in upper limbs and difference of blood pressure in upper limbs. Low TA and evaluate the extent of disease and guide therapy. Digital
upper-limb blood pressures warrant determination of ankle blood subtraction angiography (DSA), computed tomography (CT), and
pressure, which may be elevated (Hafner et al. 2012). TA in south magnetic resonance angiography (MRA) may also provide valu-
Asian countries may preferentially affect abdominal arteries; there- able information (Sider et  al. 1985; Yamada et  al. 1993; Yamada
fore hypertension may be the first clue to diagnosis of TA. Easy et al. 1998) (Figures 24.3 and 24.4). The angiographic findings of
fatigability of upper limbs, pain, and intermittent claudication due segmental, fine dystrophic calcification with abrupt termination
to vasculopathy are also noticed. Stroke, cerebral vascular accident/ are very suggestive of TA. Similarly, calcification outlining a long
transient ischaemic attack, and sudden blindness may be caused by stenotic segment of the aorta may be seen in young TA patients
thrombosis of cerebral arteries in the acute stage. Asthenia or dizzi- (Figure 24.5). Rib notching due to aortic aneurysm or dilatation of
ness are also sometimes noticed,particularly also in the acute stage. the ascending aorta may also be found.
These symptoms may be due to stroke or in part to overstimula- Based on arteriographic involvement in a cohort of Japanese
tion of the baroreceptor in the aortic arch, leading to hypotension TA patients, Nasu classified the disease into four types, depend-
(Takeshita et al. 1977). Reversible posterior encephalopathy is less ing on which segments of the aorta and its branches were involved
CHAPTER 24 takayasu’s arteritis 323

Fig. 24.3 Angiogram shows obstruction of right and left subclavian arteries and Fig. 24.5 Chest radiograph showing calcification of the aortic arch and the
marked narrowing in the left common carotid artery (arrows). descending aorta (arrow) in a 30-year-old TA patient.

(Nasu 1975). A  new classification proposed by an international involvement of the right pulmonary artery. Coronary artery lesions
co-operative study on Takayasu’s arteritis has been introduced are usually ostial or limited to the proximal segment.
to compare TA internationally, based on the location of vascular Aortic insufficiency and cardiac function can be assessed by
lesions (Hata et al. 1996). Classification of the types of TA accord- echocardiography (Hashimoto et al. 1992b). Ultrasonography may
ing to site of involvement is shown in Table 24.1. Early angiographic show carotid artery lesions and intimal thickness that may reflect
changes include localized narrowing or irregularity of the aortic disease activity (Park et  al. 2001; Schmidt et  al. 2001; Seth et  al.
wall, which may progress to obliterative vascular lesions produc- 2006). Sonography can detect ‘brightening’ of the diffuse circum-
ing narrowing, stenosis, or complete occlusion. The vessel may be ferential thickening of the carotid artery wall, the so called ‘maca-
dilated or aneurysmal, and there may be a combination of obstruc- roni’ sign (Keo et al. 2013). MRI has been reported to detect vessel
tion and aneurysmal dilatation. Segments of narrowing are com- wall oedema and can be used to monitor the disease activity of TA
monly found in angiograms. (Flamm et al. 1998).
The disease involves branches of the aorta at or near the point 18F-FDG PET has been introduced to diagnose TA (Hara
of origin and extends into the branch arteries for a variable dis- et al. 1999; Belhocine 2004; Webb et al. 2004; Meller et al. 2003;
tance. In India and other South Asian countries, the renal arter- Kobayashi et  al. 2005). 18F-FDG PET/CT can identify aortic
ies are affected in the majority of the patients; bilateral disease can inflammation anatomically (Kobayashi et al. 2005) (Figure 24.6)
result in severe renovascular hypertension. The pulmonary arter- (see Chapter 20). The 18F-FDG PET/CT can show signs of inflam-
ies are affected in half of TA patients; there is a predilection for mation and its location in the aorta directly, providing early
diagnosis of TA even before development of vascular lesion
and estimation of the inflammatory activity, particularly during

Table 24.1 Classification of Takayasu’s arteritis

Type Site of involvement


I Branches from aortic arch
IIa Ascending aorta, aortic arch and its branches
IIb Ascending aorta, aortic arch, its branches, and thoracic descending
aorta
III Thoracic descending aorta, abdominal aorta and/or renal arteries
IV Only abdominal aorta and/or renal arteries
V Combination of Type IIb and IV
Fig. 24.4 CT images showing wall thickness in the carotid artery (arrow).
324 SECTION 5 vasculitic diseases and syndromes and related disorders

X: –19.1 mm
Y: 17.0 mm
Z: 116.5 mm

Fig. 24.6 18F-FDG PET image co-registered with enhanced CT showing active inflammation in both vertebral arteries.

medication. 11C-PK11195 PET/CT has also identified vascular inflammatory abdominal aortic aneurysm, vascular Behçet’s syn-
inflammation (Pugliese et al. 2010). drome, syphilitic aortitis, giant cell arteritis, congenital vascular
abnormality, and mycotic aneurysm.
18F-FDG PET/CT analyses may help establish the diagnosis of
Diagnosis
TA at an earlier stage and start medication to prevent development
The diagnosis of TA is based on findings of vascular lesion forma- of serious vascular lesion formation.
tion in large and middle-size vessel by angiogram, CT, or MRI. Age
of onset of vascular lesion formation should also be considered in
making the diagnosis of TA to exclude giant cell arteritis. Usually Management
in the early stages of TA, non-specific inflammatory symptoms The main goal of management in the acute stage in TA is control
are common and a work up for fever of unknown origin (FUO) of inflammation to prevent progression of vascular lesion forma-
may be performed (Meller et  al. 2007; Wagner et  al. 2005). The tion due to inflammation. Medical management of TA in the acute
American College of Rheumatology proposed classification criteria stage consists of glucocorticoids, particularly prednisone or pred-
(Arend et al. 1990; Bloch et al. 1990) consisting of: (1) age of onset nisolone, as a first-line therapy. Patients with active disease are ini-
<40 years old; (2) claudication of an extremity; (3) decreased bra- tially treated with oral prednisolone at a dose of 0.5–1 mg/kg per
chial artery pulse; (4) a difference of more than 10 mmHg systolic day (Kerr et al. 1994; Mukhtyar et al. 2009) or an equivalent dose
pressure between two limbs; (5) a bruit over subclavian arteries or of prednisone. About 60 to 80% of TA patients will go into remis-
the aorta; and (6) angiographic evidence of narrowing or occlusion sion but relapse during taper occurs in more than 50% of patients
of the aorta, its primary branches, or large arteries in the proximal (Hoffman 1995). In Japan, HLA-B*5201-positive TA patients tend
upper or lower extremities. The presence of three of the six crite- to need a larger dose of prednisolone than HLA-B*5201-negative
ria is required for diagnosis among a set of subjects with systemic TA patients (Moriwaki and Numano 1992).
vasculitis. To minimize steroid adverse events, immunosuppressive agents
The Japanese Research Committee on Vasculitis Syndromes pro- such as ciclosporin, methotrexate, azathioprine, and cyclophos-
posed another set of diagnostic criteria: (1) angiographic evidence phamide can induce remission and prevent progression of vascular
of narrowing or occlusion of the aorta, its primary branches, or lesion formation (Hoffman et al. 1994; Mevorach et al. 1992; Sato
large arteries in the proximal upper or lower extremities by angio- et al. 2000; Fullerton et al. 1991).
gram, CT, or MRI; (2) early age of onset; (3) presence of markers Anti-TNF-α therapy and anti-IL-6 receptor therapy have been
of inflammation; and (4) exclusion of patients with atherosclerosis, introduced for refractory TA (Hoffman et  al. 2004; Schafer and
CHAPTER 24 takayasu’s arteritis 325

Zwerina 2012; Comarmond et al. 2012) because of the roles that TNF (1990). The American College of Rheumatology 1990 criteria for the clas-
and IL-6 appear to play in the development of lesions. In one study, sification of Takayasu arteritis. Arthritis and Rheumatism, 33, 1129–34.
treatment with TNF inhibitors induced remission in patients with Ask-Upmark, E. (1961). On the pathogenesis of the hypertension in
refractory TA; 33% of patients relapsed during treatment and 20% Takayashu’s syndrome. Acta Medica Scandinavica, 169, 467–77.
Azizi, S.P. and Rafat, A. (1956). [Observation of a patient with Takayasu’s
stopped treatment because of adverse effects (Schmidt et al. 2012).
disease; pulseless disease]. Acta Medica Iranica, 1, 43–52.
Thrombotic events happen in the acute stage in TA and some-
Bali, H.K., Jain, S., Jain, A. and Sharma, B.K. (1998). Stent supported angio-
times lead to optic complication or stroke. Low-dose aspirin in plasty in Takayasu arteritis. International Journal of Cardiology, 66
addition to glucocorticoids treatment reduces ischaemic complica- (Suppl. 1), S213–7; discussion S219–20.
tions (Numano et al. 1985). Baqir, M., Usman, M.H., Adenwalla, H.N., Aziz, S., Noor, F., Ul- Islam, T.,
Currently, remission of TA is considered when there is nor- Ahmed, M., and Hotiana, M. (2007). Takayasu’s arteritis with skin mani-
malization of general inflammatory markers such as CRP or ESR festations in a patient with inflammatory bowel disease: coincidence or
and no progression of arterial lesion by CT or MRI for more than concurrence? Clinical Rheumatology, 26, 996–8.
2 years. There are no reliable surrogate markers of disease activity Belhocine, T. (2004). 18FDG imaging of giant cell arteritis:  usefulness of
whole-body plus brain PET. European Journal of Nuclear Medicine and
(Mason 2010).
Molecular Imaging, 31, 1055–6.
Surgical intervention may be needed if severe ischaemic organ Blank, M., Krause, I., Goldkorn, T., Praprotnik, S., Livneh, A., Langevitz, P.,
dysfunction is present because established vascular lesions are usu- Kaganovsky, E., Morgenstern, S., Cohen, S., Barak, V., Eldor, A., Weksler,
ally not reversible with medical treatment. Bypass graft may be B., and Shoenfeld, Y. (1999). Monoclonal anti-endothelial cell antibod-
considered for cerebrovascular disease due to: cervicocranial vessel ies from a patient with Takayasu arteritis activate endothelial cells from
stenosis, coronary artery disease, severe to moderate aortic insuf- large vessels. Arthritis and Rheumatism, 42, 1421–32.
ficiency, severe coarctation of the aorta, renovascular hypertension, Blau, E.B. (1985). Familial granulomatous arthritis, iritis, and rash. Journal of
limb claudication, and progressive aneurysm enlargement (Kerr Pediatrics, 107, 689–93.
et al. 1994; Matsuura et al. 2005; Miyata et al. 2003; Taketani et al. Bloch, D.A., Michel, B.A., Hunder, G.G., McShane, D.J., Arend, W.P.,
Calabrese, L.H., Edworthy, S.M., Fauci, A.S., Fries, J.F., Leavitt, R.Y.
2005; Weaver et al. 2004). Control of inflammation is important for
and et al. (1990). The American College of Rheumatology 1990 criteria
successful bypass surgery (Weaver et al. 2004). for the classification of vasculitis. Patients and methods. Arthritis and
Percutaneous transluminal angioplasty of vascular lesions, with Rheumatism, 33, 1068–73.
or without a stent, has had variable results (Takahashi et al. 2002; Caccamise, W.C. and Okuda, K. (1954). Takayasu’s or pulseless disease: an
Sharma et al. 2000; Tyagi et al. 1998; Kerr et al. 1994; Fava et al. unusual syndrome with ocular manifestations. American Journal of
1993; Weaver et al. 1990). Ophthalmology, 37, 784–6.
Women sometimes develop TA during pregnancy and around Canas, C.A., Jimenez, C.A., Ramirez, L.A., Uribe, O., Tobon, I., Torrenegra,
the time of delivery. Numano and Shimamoto found continuous A., Cortina, A., Munoz, M., Gutierrez, O., Restrepo, J.F., Pena, M. and
Iglesias, A. (1998). Takayasu arteritis in Colombia. International Journal
high-level urinary excretion of total oestrogens in TA patients
of Cardiology, 66 (Suppl. 1), S73–9.
(Numano and Shimamoto 1971). Hormonal change or other stress
Castresana-Isla, C.J., Herrera-Martinez, G. and Vega-Molina, J. (1993).
related to pregnancy or delivery may serve as a trigger. Erythema nodosum and Takayasu’s arteritis after immunization with
Fertility is apparently not affected by TA; however, severe car- plasma derived hepatitis B vaccine. Journal of Rheumatology, 20, 1417–8.
diovascular complications due to TA may hamper pregnancy Chapman, R., Dawe, C., Whorwell, P.J., and Wright, R. (1978). Ulcerative
and delivery. Management of labour and the decision for cae- colitis in association with Takayasu’s disease. American Journal of
sarean section are based on the assessment of maternal risk fac- Digestive Diseases, 23, 660–2.
tors. Inflammation during pregnancy may be managed by small Chiasson, D.A., Ipp, M., Benson, L., and Silver, M.M. (1990). Acute heart
amounts of glucocorticoids. failure in an 8-year-old diabetic girl. Journal of Pediatrics, 116, 472–7.
Cipriano, P.R., Silverman, J.F., Perlroth, M.G., Griepp, R.B., and Wexler, L.
(1977). Coronary arterial narrowing in Takayasu’s aortitis. American
Prognosis Journal of Cardiology, 39, 744–50.
Cloux Blasco, J., Lozano De La Torre, M.J., Alvarez Granda, J.L., Vidal
The prognosis of TA has recently much improved due to advances Sampedro, J., and Garcia Fuentes, M. (1991). [Takayasu arteritis in a
in early diagnosis by imaging and improved medication (Ohigashi 5-year-old girl]. Anales Españoles de Pediatría, 35, 134–6.
et  al. 2012). Major features contributing to a poor prognosis of Comarmond, C., Plaisier, E., Dahan, K., Mirault, T., Emmerich, J., Amoura,
TA are cardiovascular complications, such as aortic insufficiency, Z., Cacoub, P., and Saadoun, D. (2012). Anti TNF-alpha in refrac-
hypertension, and ischaemic organ dysfunction. It is important to tory Takayasu’s arteritis:  Cases series and review of the literature.
diagnose TA and begin treatment in its earliest stage. Autoimmunity Reviews, 11, 678–84.
Dal Canto, A.J. and Virgin, H.W.T. (2000). Animal models of infection-mediated
vasculitis:  implications for human disease. International Journal of
References Cardiology, 75 (Suppl. 1), S37–45; discussion S47–52.
Abe, K., Miyazaki, S., Kusaka, T., Irokawa, N., and Aoyagi, H. (1976). Dal Canto, A.J., Virgin, H.W.T., and Speck, S.H. (2000). Ongoing viral repli-
Elevated plasma renin activity in aortitis syndrome. Japanese Heart cation is required for gammaherpesvirus 68-induced vascular damage.
Journal, 17, 1–11. Journal of Virology, 74, 11304–10.
Achar, K.N. and Al-Nakib, B. (1986). Takayasu’s arteritis and ulcerative coli- Deng, J., Younge, B.R., Olshen, R.A., Goronzy, J.J., and Weyand, C.M. (2010).
tis. American Journal of Gastroenterology, 81, 1215–7. Th17 and Th1 T-cell responses in giant cell arteritis. Circulation, 121,
Altschuler, E.L. and Wheat, J. (2000). Morgagni-Takayasu arteritis. Lancet, 906–15.
356, 2013. D’Souza, S.J., Tsai, W.S., Silver, M.M., Chait, P., Benson, L.N., Silverman, E.,
Arend, W.P., Michel, B.A., Bloch, D.A., Hunder, G.G., Calabrese, L.H., Hebert, D., and Balfe, J.W. (1998). Diagnosis and management of sten-
Edworthy, S. M., Fauci, A.S., Leavitt, R.Y., Lie, J.T., Lightfoot, R.W., Jr., et al. otic aorto-arteriopathy in childhood. Journal of Pediatrics, 132, 1016–22.
326 SECTION 5 vasculitic diseases and syndromes and related disorders

Eichhorn, J., Sima, D., Thiele, B., Lindschau, C., Turowski, A., Schmidt, H., Hoffman, G.S., Leavitt, R.Y., Kerr, G.S., Rottem, M., Sneller, M.C., and Fauci,
Schneider, W., Haller, H., and Luft, F.C. (1996). Anti-endothelial cell A.S. (1994). Treatment of glucocorticoid-resistant or relapsing Takayasu
antibodies in Takayasu arteritis. Circulation, 94, 2396–401. arteritis with methotrexate. Arthritis and Rheumatism, 37, 578–82.
El-Reshaid, K., Varro, J., Al-Duwairi, Q., and Anim, J.T. (1995). Takayasu’s Hoffman, G.S., Merkel, P.A., Brasington, R.D., Lenschow, D.J., and Liang,
arteritis in Kuwait. Journal of Tropical Medicine and Hygiene, 98, P. (2004). Anti-tumor necrosis factor therapy in patients with difficult
299–305. to treat Takayasu arteritis. Arthritis and Rheumatism, 50, 2296–304.
Fabricant, C.G. (1985). Atherosclerosis: the consequence of infection with a Hong, C.Y., Yun, Y.S., Choi, J.Y., Sul, J.H., Lee, K.S., Cha, S.H., Hong,
herpesvirus. Advances in Veterinary Science and Comparative Medicine, Y.M., Lee, H.J., Hong, Y.J., and Sohn, K.C. (1992). Takayasu arteritis
30, 39–66. in Korean children:  clinical report of seventy cases. Heart Vessels, 7
Fava, M.P., Foradori, G.B., Garcia, C.B., Cruz, F.O., Aguilar, J.G., Kramer, (Suppl.), 91–6.
A.S., and Valdes, F.E. (1993). Percutaneous transluminal angioplasty in Ishikawa, K. (1975). [The history of occlusive thromboaortopathy
patients with Takayasu arteritis: five-year experience. Journal of Vascular (Takaysu’s and allied diseases) (author’s transl.)]. Kokyu To Junkan,
and Interventional Radiology, 4, 649–52. 23, 699–707.
Fernandes, S.R., Singsen, B.H., and Hoffman, G.S. (2000). Sarcoidosis and Ishikawa, K. and Maetani, S. (1994). Long-term outcome for 120 Japanese
systemic vasculitis. Seminars in Arthritis and Rheumatism, 30, 33–46. patients with Takayasu’s disease. Clinical and statistical analyses of
Flamm, S.D., White, R.D., and Hoffman, G.S. (1998). The clinical application related prognostic factors. Circulation, 90, 1855–60.
of ‘edema-weighted’ magnetic resonance imaging in the assessment of Ishikawa, K., Uyama, M., and Asayama, K. (1983). Occlusive thromboaor-
Takayasu’s arteritis. International Journal of Cardiology, 66 (Suppl. 1), topathy (Takayasu’s disease): cervical arterial stenoses, retinal arterial
S151–9; discussion S161. pressure, retinal microaneurysms and prognosis. Stroke, 14, 730–5.
Frankel, S.K., Cosgrove, G.P., Fischer, A., Meehan, R.T., and Brown, K.K. Isohisa, I., Numano, F., Maezawa, H., and Sasazuki, T. (1978). HLA-Bw52 in
(2006). Update in the diagnosis and management of pulmonary vascu- Takayasu disease. Tissue Antigens, 12, 246–8.
litis. Chest, 129, 452–65. Iwai, T., Inoue, Y., Matsukura, I., Sugano, N., and Numano, F. (2000). Surgical
Friedman, C.J. and Tegtmeyer, C.J. (1979). Crohn’s disease associated with technique for management of Takayasu’s arteritis. International Journal
Takayasu’s arteritis. Digestive Diseases and Sciences, 24, 954–8. of Cardiology, 75 (Suppl. 1), S135–40.
Fullerton, S.H., Abel, E.A., Getz, K., and El-Ramahi, K. (1991). Cyclosporine Jacobsen, H.H. and Rasch, P.J. (1956). [Takayasu’s syndrome, pulseless dis-
treatment of severe recalcitrant pyoderma gangrenosum in a patient ease, or brachiocephalic arteritis]. Nordisk Medicine, 56, 1328–31.
with Takayasu’s arteritis. Archives of Dermatology, 127, 1731–2. Jain, S., Kumari, S., Ganguly, N.K., and Sharma, B.K. (1996). Current sta-
Girona, E., Yamamoto-Furusho, J.K., Cutino, T., Reyes, P., Vargas-Alarcon, tus of Takayasu arteritis in India. International Journal of Cardiology, 54
G., Granados, J., and Alarcon-Segovia, D. (1996). HLA-DR6 (possibly (Suppl.), S111–6.
DRB1*1301). is associated with susceptibility to Takayasu arteritis in Jennette, J.C., Falk, R.J., Andrassy, K., Bacon, P.A., Churg, J., Gross, W.L.,
Mexicans. Heart Vessels, 11, 277–80. Hagen, E.C., Hoffman, G.S., Hunder, G.G., Kallenberg, C.G., et  al.
Gravanis, M.B. (2000). Giant cell arteritis and Takayasu aortitis:  mor- (1994). Nomenclature of systemic vasculitides. Proposal of an interna-
phologic, pathogenetic and etiologic factors. International Journal of tional consensus conference. Arthritis and Rheumatism, 37, 187–92.
Cardiology, 75 (Suppl. 1), S21–33; discussion S35–6. Jimenez Casado, M. and Moncada Moneu, A. (1956). [Pulseless disease;
Hafner, F., Froehlich, H., Gar, T., Tiesenhausen, K., Scarpatetti, M., and Takayasu disease]. Revista Clínica Española, 63, 166–8.
Brodmann, M. (2012). Blood pressure measurements in patients with Kawai, T., Yamada, Y., Tsuneda, J., Aoyagi, T., and Mikata, A. (1975). Pleural
Takayasu arteriktis: a work of caution. Annals of Thoracic Surgery, 93, effusion associated with aortitis syndrome. Chest, 68, 826–8.
1299–301 Keo, H.H., Caliezi, G., Baumgartner, I., Diehm, N., and Willenberg, T. (2013).
Hall, S., Barr, W., Lie, J.T., Stanson, A.W., Kazmier, F.J., and Hunder, G.G. Increasing echogenicity of diffuse circumferential thickening (‘macaroni
(1985). Takayasu arteritis. A  study of 32 North American patients. sign’). of the carotid artery wall with decreasing inflammatory activity of
Medicine (Baltimore), 64, 89–99. Takayasu arteritis. Journal of Clinical Ultrasound, 41, 59–62.
Hara, M., Goodman, P.C., and Leder, R.A. (1999). FDG-PET finding in Kerr, G.S., Hallahan, C.W., Giordano, J., Leavitt, R.Y., Fauci, A.S., Rottem,
early-phase Takayasu arteritis. Journal of Computer Assisted Tomography, M., and Hoffman, G.S. (1994). Takayasu arteritis. Annals of Internal
23, 16–8. Medicine, 120, 919–29.
Hari, P., Bagga, A., and Srivastava, R.N. (2000). Sustained hypertension in Kimura, A., Ota, M., Katsuyama, Y., Ohbuchi, N., Takahashi, M., Kobayashi,
children. Indian Pediatrics, 37, 268–74. Y., Inoko, H., and Numano, F. (2000). Mapping of the HLA-linked genes
Hashimoto, Y., Numano, F., Oniki, T., and Shimizu, S. (1992a). Left ventricu- controlling the susceptibility to Takayasu’s arteritis. International Journal
lar geometry in Takayasu arteritis complicated by severe aortic regurgi- of Cardiology, 75 (Suppl 1.), S105–10; discussion S111–2.
tation. Cardiology, 80, 180–3. Kitamura, H., Kobayashi, Y., Kimura, A., and Numano, F. (1998). Association
Hashimoto, Y., Oniki, T., Aerbajinai, W., and Numano, F. (1992b). Aortic of clinical manifestations with HLA-B alleles in Takayasu arteritis.
regurgitation in patients with Takayasu arteritis:  assessment by color International Journal of Cardiology, 66 (Suppl. 1), S121–6.
Doppler echocardiography. Heart Vessels, 7 (Suppl.), 111–5. Kobayashi, Y., Ishii, K., Oda, K., Nariai, T., Tanaka, Y., Ishiwata, K., and
Hata, A., Noda, M., Moriwaki, R., and Numano, F. (1996). Angiographic Numano, F. (2005). Aortic wall inflammation due to Takayasu arteritis
findings of Takayasu arteritis: new classification. International Journal of imaged with 18F-FDG PET coregistered with enhanced CT. Journal of
Cardiology, 54 (Suppl.), S155–63. Nuclear Medicine, 46, 917–22.
Hatano, R., Yamada, T., Sunamori, M., Tsukuura, T., and Sakamoto, T. (1975). Kobayashi, Y. and Numano, F. (2002). 3. Takayasu arteritis. Internal Medicine,
Simplified operative technique for the long-segment atypical coarctation 41, 44–6.
of the aorta. Japanese Journal of Surgery, 5, 246–54. Koide, K. (1992). Takayasu arteritis in Japan. Heart Vessels, 7 (Suppl.), 48–54.
Hilario, M.O., Terreri, M.T., Prismich, G., Len, C., Kihara, E.N., Goldenberg, Kornberg, A., Renaudineau, Y., Blank, M., Youinou, P., and Shoenfeld, Y.
J., and Sole, D. (1998). Association of ankylosing spondylitis, Crohn’s (2000). Anti-beta 2-glycoprotein I  antibodies and anti-endothelial
disease and Takayasu’s arteritis in a child. Clinical and Experimental cell antibodies induce tissue factor in endothelial cells. Israel Medical
Rheumatology, 16, 92–4. Association Journal, 2 (Suppl.), 27–31.
Hoffman, G.S. (1995). Treatment of resistant Takayasu’s arteritis. Rheumatic Kothari, S.S. (1995). Aetiopathogenesis of Takayasu’s arteritis and BCG vac-
Diseases Clinics of North America, 21, 73–80. cination: the missing link? Medical Hypotheses, 45, 227–30.
CHAPTER 24 takayasu’s arteritis 327

Ladhani, S., Tulloh, R., and Anderson, D. (2001). Takayasu disease masquar- H., Salvarani, C., Scott, D.G., Stegeman, C., Watts, R., Westman, K.,
ading as interruption of the aortic arch in a 2-year-old child. Cardiology Witter, J., Yazici, H., and Luqmani, R. (2009). EULAR recommendations
in the Young, 11, 244–6. for the management of large vessel vasculitis. Annals of the Rheumatic
Lenhoff, S.J. and Mee, A.S. (1982). Crohn’s disease of the colon with takayasu’s Diseases, 68, 318–23.
arteritis. Postgraduate Medical Journal, 58, 386–9. Mwipatayi, B.P., Jeffery, P.C., Beningfield, S.J., Matley, P.J., Naidoo, N.G.,
Lie, J.T. (1996). Isolated pulmonary Takayasu arteritis:  clinicopathologic Kalla, A.A., and Kahn, D. (2005). Takayasu arteritis:  clinical fea-
characteristics. Modern Pathology, 9, 469–74. tures and management: report of 272 cases. ANZ Journal of Surgery,
Li-Xin, Z., Jun, N., Shan, G., Bin, P., and Li-Ying, C. (2011). Neurological 75, 110–7.
manifestations of Takayasu Arteritis. Chinese Medical Sciences Journal, Naito, S., Arakawa, K., Saito, S., Toyoda, K., and Takeshita, A. (1978).
26,227–30. Takayasu’s disease: association with HLA-B5. Tissue Antigens, 12, 143–5.
Lobato, O. (1956). [Takayasu disease; case report]. Arquivos Brasileiros de Nakabayashi, K., Kurata, N., Nangi, N., Miyake, H., and Nagasawa, T. (1996).
Cardiologia, 9, 277–88. Pulmonary artery involvement as first manifestation in three cases of
Lupi-Herrera, E., Sanchez-Torres, G., Marcushamer, J., Mispireta, J., Horwitz, Takayasu arteritis. International Journal of Cardiology, 54 (Suppl.), S177–83.
S., and Vela, J.E. (1977). Takayasu’s arteritis. Clinical study of 107 cases. Nasu, T. (1975). Takayasu’s truncoarteritis in Japan. A statistical observation
American Heart Journal, 93, 94–103. of 76 autopsy cases. Pathologia et Microbiologia (Basel), 43, 140–6.
Martini, A. (1995). Behçet’s disease and Takayasu’s disease in children. Nasu, T. (1982). Takayasu’s truncoarteritis. Pulseless disease or aortitis syn-
Current Opinion in Rheumatology, 7, 449–54. drome. Acta Pathologica Japonica, 32 (Suppl.) 1, 117–31.
Mason, J.C. (2010). Takayasu arteritis—advances in diagnosis and manage- Nicklin, M.J., Hughes, D.E., Barton, J.L., Ure, J.M., Duff, G.W. (2000). Arterial
ment. Nature Reviews Rheumatology, 6, 406–15. inflammation in mice lacking the interleukin 1 receptor antagonist gene.
Matsubara, O., Kuwata, T., Nemoto, T., Kasuga, T., and Numano, F. (1992). J Exp Med, 191, 303–12.
Coronary artery lesions in Takayasu arteritis: pathological considera- Noel, N., Butel, N., Le Hoang, P., Koskas, F., Costedoat-Chalumeau, N.,
tions. Heart Vessels (Suppl.), 7, 26–31. Wechsler, B., Amoura, Z., Bodaghi, B., Cacoub, P., and Saadoun, D.
Matsuki, T., Isoda, K., Horai, R., Nakajima, A., Aizawa, Y., Suzuki, K., Ohsuzu, (2013). Small vessel involvement in Takayasu’s arteritis. Autoimmunity
F., and Iwakura, Y. (2005). Involvement of tumor necrosis factor-alpha Reviews, 12, 355–62.
in the development of T cell-dependent aortitis in interleukin-1 receptor Numano, F. (2000). Vasa vasoritis, vasculitis and atherosclerosis. International
antagonist-deficient mice. Circulation, 112, 1323–31. Journal of Cardiology, 75 (Suppl. 1), S1–8; discussion S17-9.
Matsuura, K., Ogino, H., Kobayashi, J., Ishibashi-Ueda, H., Matsuda, H., Numano, F., Isohisa, I., Kishi, U., Arita, M., and Maezawa, H. (1978).
Minatoya, K., Sasaki, H., Bando, K., Niwaya, K., Tagusari, O., Nakajima, Takayasu’s disease in twin sisters. Possible genetic factors. Circulation,
H., Yagihara, T., and Kitamura, S. (2005). Surgical treatment of aortic 58, 173–7.
regurgitation due to Takayasu arteritis: long-term morbidity and mor- Numano, F. and Kakuta, T. (1996). Takayasu arteritis—five doctors in the
tality. Circulation, 112, 3707–12. history of Takayasu arteritis. International Journal of Cardiology, 54
Meller, J., Grabbe, E., Becker, W., and Vosshenrich, R. (2003). Value of F-18 (Suppl.), S1–10.
FDG hybrid camera PET and MRI in early takayasu aortitis. European Numano, F., Miyata, T., and Nakajima, T. (1996). Ulcerative colitis, Takayasu
Radiology, 13, 400–5. arteritis and HLA. Internal Medicine, 35, 521–2.
Meller, J., Sahlmann, C.O., and Scheel, A.K. (2007). 18F-FDG PET and PET/ Numano, F., Okawara, M., Inomata, H., and Kobayashi, Y. (2000). Takayasu’s
CT in fever of unknown origin. Journal of Nuclear Medicine, 48, 35–45. arteritis. Lancet, 356, 1023–5.
Mevorach, D., Leibowitz, G., Brezis, M., and Raz, E. (1992). Induction of Numano, F. and Shimamoto, T. (1971). Hypersecretion of estrogen in
remission in a patient with Takayasu’s arteritis by low dose pulses of Takayasu’s disease. American Heart Journal, 81, 591–6.
methotrexate. Annals of the Rheumatic Diseases, 51, 904–5. Numano, F., Yajima, M., Shimokado, K., and Maruyama, Y. (1985). Aspirin
Miceli-Richard, C., Lesage, S., Rybojad, M., Prieur, A.M., Manouvrier-Hanu, treatment for patients with cardiovascular disease. Advances in
S., Hafner, R., Chamaillard, M., Zouali, H., Thomas, G., and Hugot, Prostaglandin Thromboxane and Leukotriene Research, 15, 543–6.
J.P. (2001). CARD15 mutations in Blau syndrome. Nature Genetics, Ohigashi, H., Haraguchi, G., Konishi, M., Tezuka, D., Kamiishi, T., Ishihara,
29, 19–20. T., and Isobe, M.E. (2012). Improved prognosis of Takayasu arteritis over
Millar, A.J., Gilbert, R.D., Brown, R.A., Immelman, E.J., Burkimsher, D.A., the past decade—comprehensive analysis of 106 patients. Circulation
and Cywes, S. (1996). Abdominal aortic aneurysms in children. Journal Journal, 76, 1044–11.
of Pediatric Surgery, 31, 1624–8. Owyang, C., Miller, L.J., Lie, J.T., and Fleming, C.R. (1979). Takayasu’s arteri-
Mitchell, C.S. and Parisi, M.T. (1997). Magnetic resonance imaging of tis in Crohn’s disease. Gastroenterology, 76, 825–8.
Takayasu’s aortitis in an infant. Journal of the American Osteopathic Park, M.C., Lee, S.W., Park, Y.B., and Lee, S.K. (2006). Serum cytokine pro-
Association, 97, 607–9. files and their correlations with disease activity in Takayasu’s arteritis.
Miwa, Y., Nagasako, K., Sasaki, H., Ichikawa, K., Yaguchi, T., Hasegawa, K., Rheumatology (Oxford), 45, 545–8.
and Kubokura, T. (1979). Aortitis syndrome associated with ulcerative Park, M.H. and Park, Y.B. (1992). HLA typing of Takayasu arteritis in Korea.
colitis: report of a case. Gastroenterologia Japonica, 14, 492–5. Heart Vessels (Suppl.), 7, 81–4.
Miyata, T., Sato, O., Koyama, H., Shigematsu, H., and Tada, Y. (2003). Park, S.H., Chung, J.W., Lee, J.W., Han, M.H., and Park, J.H. (2001). Carotid
Long-term survival after surgical treatment of patients with Takayasu’s artery involvement in Takayasu’s arteritis: evaluation of the activity by
arteritis. Circulation, 108, 1474–80. ultrasonography. Journal of Ultrasound in Medicine, 20, 371–8.
Morales, E., Pineda, C., and Martinez-Lavin, M. (1991). Takayasu’s arteritis Park, Y.B., Hong, S.K., Choi, K.J., Sohn, D.W., Oh, B.H., Lee, M.M., Choi,
in children. Journal of Rheumatology, 18, 1081–4. Y.S., Seo, J.D., Lee, Y.W., and Park, J.H. (1992). Takayasu arteritis in
Morita, Y., Yamamura, M., Suwaki, K., Mima, A., Ishizu, T., Hirohata, M., Korea: clinical and angiographic features. Heart Vessels, 7 (Suppl.), 55–9.
Kashihara, N., Makino, H., and Ota, Z. (1996). Takayasu’s arteritis asso- Piyachon, C. and Suwanwela, N. (1992). Takayasu arteritis in Thailand. Heart
ciated with ulcerative colitis; genetic factors in this association. Internal Vessels, 7 (Suppl.), 60–7.
Medicine, 35, 574–8. Planas, E. (1978). [Martorell’s syndrome and Takayasu arteritis]. Angiologia,
Moriwaki, R. and Numano, F. (1992). Takayasu arteritis: follow-up studies for 30, 134–6.
20 years. Heart Vessels, 7 (Suppl.), 138–45. Pugliese, F., Gaemperli, O., Kinderlerer, A.R., Lamare, F., Shalhoub, J.,
Mukhtyar, C., Guillevin, L., Cid, M.C., Dasgupta, B., De Groot, K., Gross, W., Davies, A.H., Rimoldi, O.E., Mason, J.C., and Camici, P.G. (2010).
Hauser, T., Hellmich, B., Jayne, D., Kallenberg, C.G., Merkel, P.A., Raspe, Imaging of vascular inflammation with [11C]-PK11195 and positron
328 SECTION 5 vasculitic diseases and syndromes and related disorders

emission tomography/computed tomography angiography. Journal of Sen, P.K., Kinare, S.G., Kelkar, M.D., and Parulkar, G.B. (1972). Non-specific
the American College of Cardiology, 56, 653–61. aortoarteritis. Tata McGraw Hill, Bombay.
Robles, M. and Reyes, P.A. (1994). Takayasu’s arteritis in Mexico: a clinical Seth, S., Goyal, N.K., Jagia, P., Gulati, G., Karthikeyan, G., Sharma, S., and
review of 44 consecutive cases. Clinical and Experimental Rheumatology, Talwar, K.K. (2006). Carotid intima-medial thickness as a marker of dis-
12, 381–8. ease activity in Takayasu’s arteritis. International Journal of Cardiology,
Roedenbeck, S.D. and Bauer, J. (1956). [Takayasu’s disease; neurological 108, 385–90.
aspect of a case of pulseless disease]. Anales de la Facultad de Medicina Seyahi, E., Ugurlu, S., Cumali, R., Balci, H., Seyahi, N., Yurdakul, S., and
Lima, 39, 1407–16. Yazici, H. (2006). Atherosclerosis in Takayasu arteritis. Annals of the
Rose, C.D., Eichenfield, A.H., Goldsmith, D.P., and Athreya, B.H. Rheumatic Diseases, 65, 1202–7.
(1990). Early onset sarcoidosis with aortitis—‘juvenile systemic Sharma, B.K., Jain, S., Bali, H.K., Jain, A., and Kumari, S. (2000). A follow-up
granulomatosis?’Journal of Rheumatology, 17, 102–6. study of balloon angioplasty and de-novo stenting in Takayasu arteritis.
Rosenthal, T., Morag, B., and Itzchak, Y. (1992). Takayasu arteritis in Israel. International Journal of Cardiology, 75 (Suppl. 1), S147–52.
Heart Vessels, 7 (Suppl.), 44–7. Sharma, B.K., Jain, S., Suri, S., and Numano, F. (1996). Diagnostic criteria
Ross, R.S. and McKusick, V.A. (1953). Aortic arch syndromes; diminished or for Takayasu arteritis. International Journal of Cardiology, 54 (Suppl.),
absent pulses in arteries arising from arch of aorta. Archives of Internal S141–7.
Medicine, 92, 701–40. Sharma, B.K., Sagar, S., Chugh, K.S., Sakhuja, V., Rajachandran, A., and
Sakhuja, V., Gupta, K.L., Bhasin, D.K., Malik, N., and Chugh, K.S. (1990). Malik, N. (1985). Spectrum of renovascular hypertension in the young
Takayasu’s arteritis associated with idiopathic ulcerative colitis. Gut, in north India: a hospital based study on occurrence and clinical fea-
31, 831–3. tures. Angiology, 36, 370–8.
Salazar, M., Varela, A., Ramirez, L.A., Uribe, O., Vasquez, G., Egea, E., Yunis, Sharma, B.K., Sagar, S., Singh, A.P., and Suri, S. (1992). Takayasu arteritis in
E.J., and Iglesias-Gamarra, A. (2000). Association of HLA-DRB1*1602 India. Heart Vessels, 7 (Suppl.), 37–43.
and DRB1*1001 with Takayasu arteritis in Colombian mestizos as Sharma, S., Rajani, M., Kamalakar, T., Talwar, K.K., and Sunderam, K.R.
markers of Amerindian ancestry. International Journal of Cardiology, 75 (1991). Clinical and angiographic features of nonspecific aorto-arteritis
(Suppl. 1), S113–6. in children and adults. Acta Radiologica, 32, 485–7.
Sasazuki, T., Ohta, N., Isohisa, I., Numano, F., and Maezawa, H. (1979). Sharma, S., Taneja, K., Bahl, V.K., and Rajani, M. (1998). Morphological
Association between Takayasu disease and HLA-DHO. Tissue Antigens, mural changes in the aorta in non-specific aortoarteritis (Takayasu’s
14, 177–8. arteritis):  assessment by intravascular ultrasound imaging. Clinical
Sato, E.I., Hatta, F.S., Levy-Neto, M., and Fernandes, S. (1998). Demographic, Radiology, 53, 37–43.
clinical, and angiographic data of patients with Takayasu arteritis Shepherd, J. and Nicklin, M.J. (2005). Elastic-vessel arteritis in interleu-
in Brazil. International Journal of Cardiology, 66 (Suppl. 1), S67–70; kin-1 receptor antagonist-deficient mice involves effector Th1 cells and
discussion S71. requires interleukin-1 receptor. Circulation, 111, 3135–40.
Sato, E.I., Lima, D.N., Espirito Santo, B., and Hata, F. (2000). Takayasu arteri- Shibata, H., Yasunami, M., Obuchi, N., Takahashi, M., Kobayashi, Y., Numano,
tis. Treatment and prognosis in a university center in Brazil. International F., and Kimura, A. (2006). Direct determination of single nucleotide pol-
Journal of Cardiology, 75 (Suppl. 1), S163–6. ymorphism haplotype of NFKBIL1 promoter polymorphism by DNA
Savory, W. (1856). Royal Medical and Chirurgical Society. On a case of young conformation analysis and its application to association study of chronic
woman in whom the main arteries of both upper extremities and of the left inflammatory diseases. Human Immunology, 67, 363–73.
side of the neck were throughout completely obliterated. Lancet, 67, 373. Shinmi, Y. (1942). A case of Takayasu’s arteritis.. Sogo Gannka (Tokyo), 36,
Schafer, V.S. and Zwerina, J. (2012). Biologic treatment of large-vessel vascu- 1404–10.
litides. Current Opinion in Rheumatology, 24, 31–7. Sider, L., Mintzer, R.A., Vrla, R.F., and Mendelson, E.B. (1985). Use of DSA
Schapiro, J.M., Shpitzer, S., Pinkhas, J., Sidi, Y., and Arber, N. (1994). for diagnosis of Takayasu’s arteritis. Illinois Medical Journal, 167, 53–5.
Sarcoidosis as the initial manifestation of Takayasu’s arteritis. Journal of Stanley, P., Roebuck, D., and Barboza, A. (2003). Takayusu’s arteritis in chil-
Medicine, 25, 121–8. dren. Techniques in Vascular and Interventional Radiology, 6, 158–68.
Schmidt, J., Kermani, T.A., Kirstin Bacani, A., Crowson, C.S., Matteson, Takahashi, J.C., Sakai, N., Manaka, H., Iihara, K., Sakai, H., Sakaida, H.,
E.L., and Warrington, K.J. (2012). Tumor necrosis factor inhibitors in Higashi, T., Ishibashi, T., and Nagata, I. (2002). Multiple supra-aortic
patients with Takayasu arteritis: Experience from a referral center with stenting for Takayasu arteritis: extensive revascularization and two-year
long-term follow-up. Arthritis Care Res (Hoboken), 64, 1079–83 follow-up. American Journal of Neuroradiology, 23, 790–3.
Schmidt, W.A., Seipelt, E., Molsen, H.P., Poehls, C., and Gromnica-Ihle, Takamura, C., Ohhigashi, H., Ebana, Y., and Isobe, M. (2012). New Human
E.J. (2001). Vasculitis of the internal carotid artery in Wegener’s gran- Leukocyte Antigen Risk Allele in Japanese Patients with Takayasu
ulomatosis:  comparison of ultrasonography, angiography, and MRI. Arteritis. Circulation Journal, 76, 1697–702.
Scandinavian Journal of Rheumatology, 30, 48–50. Takayasu, M. (1908). A case with peculiar changes of the retinal central ves-
Seko, Y., Minota, S., Kawasaki, A., Shinkai, Y., Maeda, K., Yagita, H., Okumura, sels. Acta Society of Ophtalmology of Japan, 2, 554–5.
K., Sato, O., Takagi, A., Tada, Y., et al. (1994). Perforin-secreting killer cell Takeda, N., Takahashi, T., Seko, Y., Maemura, K., Nakasone, H., Sakamoto,
infiltration and expression of a 65-kD heat-shock protein in aortic tis- K., Hirata, Y., and Nagai, R. (2005). Takayasu myocarditis mediated by
sue of patients with Takayasu’s arteritis. Journal of Clinical Investigation, cytotoxic T lymphocytes. Internal Medicine, 44, 256–60.
93, 750–8. Takeshita, A., Tanaka, S., Orita, Y., Kanaide, H., and Nakamura, M. (1977).
Seko, Y., Sato, O., Takagi, A., Tada, Y., Matsuo, H., Yagita, H., Okumura, K., Baroreflex sensitivity in patients with Takayasu’s aortitis. Circulation,
and Yazaki, Y. (1996). Restricted usage of T-cell receptor Valpha-Vbeta 55, 803–6.
genes in infiltrating cells in aortic tissue of patients with Takayasu’s arte- Taketani, T., Miyata, T., Morota, T., and Takamoto, S. (2005). Surgical treat-
ritis. Circulation, 93, 1788–90. ment of atypical aortic coarctation complicating Takayasu’s arteritis—
Seko, Y., Sugishita, K., Sato, O., Takagi, A., Tada, Y., Matsuo, H., Yagita, H., experience with 33 cases over 44 years. Journal of Vascular Surgery, 41,
Okumura, K., and Nagai, R. (2004). Expression of costimulatory mole- 597–601.
cules (4-1BBL and Fas) and major histocompatibility class I chain-related Tripathy, N.K., Sinha, N., and Nityanand, S. (2004). Interleukin-8 in
A (MICA) in aortic tissue with Takayasu’s arteritis. Journal of Vascular Takayasu’s arteritis: plasma levels and relationship with disease activity.
Research, 41, 84–90. Clinical and Experimental Rheumatology, 22, S27–30.
CHAPTER 24 takayasu’s arteritis 329

Tripathy, N.K., Upadhyaya, S., Sinha, N., and Nityanand, S. (2001). Weaver, F.A., Kumar, S.R., Yellin, A.E., Anderson, S., Hood, D.B., Rowe, V.L.,
Complement and cell mediated cytotoxicity by antiendothelial cell Kitridou, R.C., Kohl, R.D., and Alexander, J. (2004). Renal revascu-
antibodies in Takayasu’s arteritis. Journal of Rheumatology, 28, 805–8. larization in Takayasu arteritis-induced renal artery stenosis. Journal of
Turkoglu, C., Memis, A., Payzin, S., Akin, M., Kultusay, H., Akilli, A., Can, Vascular Surgery, 39, 749–57.
L., and Altintig, A. (1996). Takayasu arteritis in Turkey. International Weaver, F.A., Yellin, A.E., Campen, D.H., Oberg, J., Foran, J., Kitridou, R.C.,
Journal of Cardiology, 54 (Suppl.), S135–6. Lee, S.E., and Kohl, R.D. (1990). Surgical procedures in the manage-
Tyagi, S., Verma, P.K., Gambhir, D.S., Kaul, U.A., Saha, R., and Arora, ment of Takayasu’s arteritis. Journal of Vascular Surgery, 12, 429–37;
R. (1998). Early and long-term results of subclavian angioplasty in discussion 438–9.
aortoarteritis (Takayasu disease):  comparison with atherosclerosis. Webb, M., Chambers, A., Al Nahhas, A., Mason, J.C., Maudlin, L., Rahman,
Cardiovascular and Interventional Radiology, 21, 219–24. L., and Frank, J. (2004). The role of 18F-FDG PET in characterising dis-
Umekita, K., Takajo, I., Miyauchi, S., Tsurumura, K., Ueno, S., Kusumoto, N., ease activity in Takayasu arteritis. European Journal of Nuclear Medicine
Kai, Y., Kuroki, M., Sasaki, T., and Okayama, A. (2006). [18F]fluorode- and Molecular Imaging, 31, 627–34.
oxyglucose positron emission tomography is a useful tool to diagnose Weck, K.E., Dal Canto, A.J., Gould, J.D., O’Guin, A.K., Roth, K.A., Saffitz,
the early stage of Takayasu’s arteritis and to evaluate the activity of the J.E., Speck, S.H., and Virgin, H.W. (1997). Murine gamma-herpesvirus
disease. Modern Rheumatology, 16, 243–7. 68 causes severe large-vessel arteritis in mice lacking interferon-gamma
Ureten, K., Ozturk, M.A., Onat, A.M., Ozturk, M.H., Ozbalkan, Z., Guvener, responsiveness: a new model for virus-induced vascular disease. Nature
M., Kiraz, S., Ertenli, I., and Calguneri, M. (2004). Takayasu’s arteri- Medicine, 3, 1346–53.
tis: results of a university hospital of 45 patients in Turkey. International Weyand, C.M. and Goronzy, J.J. (2003). Medium- and large-vessel vasculitis.
Journal of Cardiology, 96, 259–64. New England Journal of Medicine, 349, 160–9.
Vanoli, M., Miani, S., Amft, N., Bacchiani, G., Radelli, L., and Scorza, R. Yajima, M., Numano, F., Park, Y.B., and Sagar, S. (1994). Comparative stud-
(1995). Takayasu’s arteritis in Italian patients. Clinical and Experimental ies of patients with Takayasu arteritis in Japan, Korea and India—com-
Rheumatology, 13, 45–50. parison of clinical manifestations, angiography and HLA-B antigen.
Vedantham, V., Ratnagiri, P.K., and Ramasamy, K. (2005). Hypotensive retin- Japanese Circulation Journal, 58, 9–14.
opathy in Takayasu’s arteritis. Ophthalmic Surgery Lasers and Imaging, Yamada, I., Nakagawa, T., Himeno, Y., Numano, F., and Shibuya, H. (1998).
36, 240–4. Takayasu arteritis: evaluation of the thoracic aorta with CT angiography.
Verma, D.K., Tripathy, N.K., Verma, N.S., and Tiwari, S. (2005). Interleukin Radiology, 209, 103–9.
12 in Takayasu’s arteritis: plasma concentrations and relationship with Yamada, I., Numano, F., and Suzuki, S. (1993). Takayasu arteritis: evaluation
disease activity. Journal of Rheumatology, 32, 2361–3. with MR imaging. Radiology, 188, 89–94.
Waern, A.U., Andersson, P., and Hemmingsson, A. (1983). Takayasu’s arteri- Yamada, I., Shibuya, H., Matsubara, O., Umehara, I., Makino, T., Numano,
tis: a hospital-region based study on occurrence, treatment and progno- F., and Suzuki, S. (1992). Pulmonary artery disease in Takayasu’s arteri-
sis. Angiology, 34, 311–20. tis: angiographic findings. American Journal of Roentgenology, 159, 263–9.
Wagner, A.D., Andresen, J., Raum, E., Lotz, J., Zeidler, H., Kuipers, J.G., and Yassinger, S., Adelman, R., Cantor, D., Halsted, C.H., and Bolt, R.J. (1976).
Jendro, M.C. (2005). Standardised work-up programme for fever of Association of inflammatory bowel disease and large vascular lesions.
unknown origin and contribution of magnetic resonance imaging for Gastroenterology, 71, 844–6.
the diagnosis of hidden systemic vasculitis. Annals of the Rheumatic Yoshida, M., Kimura, A., Katsuragi, K., Numano, F., and Sasazuki, T. (1993).
Diseases, 64, 105–10. DNA typing of HLA-B gene in Takayasu’s arteritis. Tissue Antigens,
Walter, M.A. (2007). [(18)F]fluorodeoxyglucose PET in large vessel vasculi- 42, 87–90.
tis. Radiology Clinics of North America, 45, 735–44, viii. Zaas, A., Scheel, P., Venbrux, A., and Hellmann, D.B. (2001). Large artery
Wang, X., Kuivaniemi, H., Bonavita, G., Mutkus, L., Mau, U., Blau, E., vasculitis following recombinant hepatitis B vaccination: 2 cases. Journal
Inohara, N., Nunez, G., Tromp, G., and Williams, C.J. (2002). CARD15 of Rheumatology, 28, 1116–20.
mutations in familial granulomatosis syndromes: a study of the original Zheng, D., Fan, D., and Liu, L. (1992). Takayasu arteritis in China: a report of
Blau syndrome kindred and other families with large-vessel arteritis and 530 cases. Heart Vessels, 7 (Suppl.), 32–6.
cranial neuropathy. Arthritis and Rheumatism, 46, 3041–5.
CHAPTER 25

Polyarteritis nodosa
Loïc Guillevin and Benjamin Terrier

Introduction arteritis. All the subsequent classifications of vasculitides have


been more-or-less derived from Zeek’s endeavour. In Fauci’s clas-
Polyarteritis nodosa (PAN) was first described by Küssmaul and sification (Fauci et al. 1978), systemic vasculitides comprised PAN,
Maier (1866). This necrotizing angiitis predominantly involves eosinophilic granulomatosis with polyangiitis (EGPA, Churg–
medium-sized arteries and can affect the majority of organs in the Strauss syndrome), and overlap angiitis. In 1990, the American
body. Considering the aetiologies of PAN, primary and secondary College of Rheumatology (ACR) established criteria for PAN clas-
forms can be distinguished, because PAN can be the consequence of sification (Lightfoot et al. 1990) but did not distinguish between
hepatitis B virus (HBV) infection (Prince and Trépo 1971; Trépo and PAN and microscopic polyangiitis (MPA) (Table 25.1). In the past,
Thivolet 1970a) or other aetiological agents (Corman and Dolson PAN and MPA were considered different forms of the same dis-
1992; Calabrese 1991; Mader and Keystone 1992). Among vascu- ease because their main clinical manifestations (i.e. neurological,
litides, PAN is now less frequent than in the past for reasons dis- gastrointestinal (GI), constitutional symptoms) are very similar,
cussed in Section Aetiology and Precipitating Factors. In this chapter, thereby explaining why they were grouped together in the ACR
we review the classification criteria for PAN, the main characteristics classification criteria.
of the disease, and its pathogenesis, outcome, and treatment. However, major differences exist between these two entities,
as clarified and established in the first Chapel Hill Nomenclature
(Jennette et al. 1994) and further refined in the revised nomencla-
Classification criteria ture in 2012 (Jennette et al. 2013) (Table 25.2). PAN is defined as a
The first attempt to classify vasculitides was published in 1952, necrotizing arteritis of medium- or small-sized arteries without glo-
when Zeek (1952) proposed the generic term ‘necrotizing vasculi- merulonephritis, or vasculitis in arterioles, capillaries or venules, and
tis’ to identify five distinct types of systemic vasculitides defined by not associated with antineutrophil cytoplasm antibodies (ANCA).
their clinical and histological signs: hypersensitivity angiitis, aller- MPA is responsible for glomerulonephritis and lung capillaritis,
gic granulomatous angiitis, rheumatic arteritis, PAN, and temporal while PAN is characterized by vascular nephropathy and never

Table 25.1 1990 ACR criteria for the classification of polyarteritis nodosa

Criterion Definition
1. Weight loss >4 kg Loss of ≥4 kg of body weight since illness began, not due to dieting or other factors
2. Livedo reticularis Mottled reticular pattern over the skin of portions of the extremities or torso
3. Testicular pain or tenderness Pain or tenderness of the testicles, not due to infection, trauma or other causes
4. Myalgias, weakness or polyneuropathy Diffuse myalgias (excluding shoulder and hip girdles), or weakness of muscles or tenderness of leg muscles
5. Mononeuropathy or polyneuropathy Development of mononeuropathy, multiple mononeuropathies, or polyneuropathy
6. Diastolic BP >90 mmHg Development of hypertension with the diastolic BP >90 mmHg
7. Elevated BUN or creatinine Elevation of BUN >40 mg/dl (14.3 µmol/l) or creatinine >1.5 mg/dl (132 µmol/l), not due to dehydration
or obstruction
8. Hepatitis B virus Presence of hepatitis B surface antigen or antibodies in serum
9. Arteriographic abnormality Arteriogram showing microaneurysms or occlusions of the visceral arteries, not due to arteriosclerosis,
fibromuscular dysplasia, or non-inflammatory causes
10. Biopsy of small- or medium-sized artery Histological changes showing the presence of granulocytes or granulocytes and mononuclear leukocytes in
containing neutrophils the artery wall

For classification purposes, a patient with vasculitis shall be said to have PAN if at least 3 of these 10 criteria are present. The presence of any 3 or more criteria yields a sensitivity of 82.2%
and a specificity of 86.6%. BP, blood pressure; BUN, blood urea nitrogen.
(Lightfoot, R.W. Jr, Michel, B.A., Bloch, D.A., et al. (1990). The American College of Rheumatology 1990 criteria for the classification of polyarteritis nodosa. Arthritis Rheum, 33, 1088–93.)
Table 25.2 Definitions for vasculitides adopted by the 2011–2012 International Chapel Hill Consensus Conference (CHCC) Nomenclature
of Vasculitides

CHCC 2012 name CHCC 2012 definition


Large-vessel vasculitis (LVV) Vasculitis affecting large arteries more often than other vasculitides. Large arteries are the aorta and its major branches. Any
size artery may be affected.
Takayasu’s arteritis (TAK) Arteritis, often granulomatous, predominantly affecting the aorta and/or its major branches. Onset usually in patients younger
than 50.
Giant cell arteritis (GCA) Arteritis, often granulomatous, usually affecting the aorta and/or its major branches, with a predilection for the branches
of the carotid and vertebral arteries. Often involves the temporal artery. Onset usually in patients older than 50 and often
associated with polymyalgia rheumatica.
Medium-vessel vasculitis (MVV) Vasculitis predominantly affecting medium arteries defined as the main visceral arteries and their branches. Any size artery
may be affected. Inflammatory aneurysms and stenoses are common.
Polyarteritis nodosa (PAN) Necrotizing arteritis of medium or small arteries without glomerulonephritis or vasculitis in arterioles, capillaries, or venules;
and not associated with ANCA.
Kawasaki’s disease (KD) Arteritis associated with the mucocutaneous lymph node syndrome and predominantly affecting medium and small arteries.
Coronary arteries are often involved. Aorta and large arteries may be involved. Usually occurs in infants and young children.
Small-vessel vasculitis (SVV) Vasculitis predominantly affecting small vessels, defined as small intraparenchymal arteries, arterioles, capillaries and venules.
Medium arteries and veins may be affected.
ANCA-associated vasculitis Necrotizing vasculitis, with few or no immune deposits, predominantly affecting small vessels (i.e. capillaries, venules, arterioles
(AAV) and small arteries), associated with MPO-ANCA or PR3-ANCA. Not all patients have ANCA. Add a prefix indicating ANCA
reactivity, e.g. PR3-ANCA, MPO-ANCA, ANCA-negative.
Microscopic polyangiitis (MPA) Necrotizing vasculitis, with few or no immune deposits, predominantly affecting small vessels (i.e. capillaries, venules, or
arterioles). Necrotizing arteritis involving small and medium arteries may be present. Necrotizing glomerulonephritis is very
common. Pulmonary capillaritis often occurs. Granulomatous inflammation is absent.
Granulomatosis with polyangiitis Necrotizing granulomatous inflammation usually involving the upper and lower respiratory tract, and necrotizing
(Wegener’s) (GPA) vasculitis affecting predominantly small to vessels (e.g. capillaries, venules, arterioles, arteries, and veins). Necrotizing
glomerulonephritis is common.
Eosinophilic granulomatosis Eosinophil-rich and necrotizing granulomatous inflammation often involving the respiratory tract, and necrotizing vasculitis
with polyangiitis (Churg– predominantly affecting small to medium vessels, and associated with asthma and eosinophilia. ANCA is more frequent
Strauss) (EGPA) when glomerulonephritis is present.
Immune complex vasculitis Vasculitis with moderate to marked vessel wall deposits of immunoglobulin and/or complement components predominantly
affecting small vessels (i.e. capillaries, venules, arterioles, and small arteries). Glomerulonephritis is frequent.
Anti-GBM disease Vasculitis affecting glomerular capillaries, pulmonary capillaries, or both, with basement membrane deposition of
antibasement membrane autoantibodies. Lung involvement causes pulmonary haemorrhage, and renal involvement causes
glomerulonephritis with necrosis and crescents.
Cryoglobulinaemic Vasculitis with cryoglobulin immune deposits affecting small vessels (predominantly capillaries, venules, or arterioles) and
vasculitis (CV) associated with cryoglobulins in serum. Skin, glomeruli, and peripheral nerves are often involved.
IgA vasculitis (IgAV) Vasculitis, with IgA1-dominant immune deposits, affecting small vessels (predominantly capillaries, venules, or arterioles). Often
(Henoch–Schönlein) involves skin and gut, and frequently causes arthritis. Glomerulonephritis indistinguishable from IgA nephropathy may occur.
Hypocomplementaemic Vasculitis accompanied by urticaria and hypocomplementaemia affecting small vessels (i.e. capillaries, venules, or arterioles), and
urticarial vasculitis (HUV) associated with anti-C1q antibodies. Glomerulonephritis, arthritis, obstructive pulmonary disease, and ocular inflammation are
(anti-C1q vasculitis) common.
Variable-vessel vasculitis (VVV) Vasculitis with no predominant type of vessel involved that can affect vessels of any size (small, medium, and large) and type
(arteries, veins, and capillaries).
Behçet's disease (BD) Vasculitis occurring in patients with Behçet's disease that can affect arteries or veins. Behçet's disease is characterized by recurrent
oral and/or genital aphthous ulcers accompanied by cutaneous, ocular, articular, gastrointestinal, and/or central nervous system
inflammatory lesions. Small vessel vasculitis, thromboangiitis, thrombosis, arteritis and arterial aneurysms may occur.
Cogan’s syndrome (CS) Vasculitis occurring in patients with Cogan's syndrome. Cogan’s syndrome is characterized by ocular inflammatory lesions,
including interstitial keratitis, uveitis, and episcleritis, and inner ear disease, including sensorineural hearing loss and vestibular
dysfunction. Vasculitic manifestations may include arteritis (affecting small, medium, or large arteries), aortitis, aortic aneurysms,
and aortic and mitral valvulitis.
Single-organ vasculitis (SOV) Vasculitis in arteries or veins of any size in a single organ that has no features that indicate that it is a limited expression of a
systemic vasculitis. The involved organ and vessel type should be included in the name (e.g. cutaneous small vessel vasculitis,
testicular arteritis, central nervous system vasculitis). Vasculitis distribution may be unifocal or multifocal (diffuse) within an
organ. Some patients originally diagnosed with SOV will develop additional disease manifestations that warrant re-defining
the case as one of the systemic vasculitides (e.g. cutaneous arteritis later becoming systemic polyarteritis nodosa, etc.).
Vasculitis associated with Vasculitis that is associated with and may be secondary to (caused by) a systemic disease. The name (diagnosis) should have a
systemic disease prefix term specifying the systemic disease (e.g. rheumatoid vasculitis, lupus vasculitis, etc.).
Vasculitis associated with Vasculitis that is associated with a probable specific aetiology. The name (diagnosis) should have a prefix term specifying
probable aetiology the association (e.g. hydralazine-associated microscopic polyangiitis, hepatitis B virus-associated vasculitis, hepatitis C
virus-associated cryoglobulinaemic vasculitis, etc.).
CHAPTER 25 polyarteritis nodosa 333

Table 25.3 Proposed diagnostic criteria for PANa 1.0

Criteria Odds ratio 95% CI R²


0.8
Positive for PAN      
HBV infection 16.85 6.30–45.08 0.320
Myalgias 1.93 1.06–3.53 0.517 0.6

Sensitivity
Mononeuropathy or 3.36 1.93–5.86 0.619
polyneuropathy
0.4
Arteriographic abnormalities 20.40 7.30–56.99 0.640
Testicular pain or tenderness 5.27 1.98–28.26 0.661
Proposed diagnostic criteria
Negative (exclusion)       0.2 1990 ACR classification criteria
for PAN
ANCA positivity 0.11 0.05–0.23 0.668
Glomerulonephritis 0.07 0.02–0.29 0.674 0.0
0.0 0.2 0.4 0.6 0.8 1.0
Recent asthma onset 0.01 0.01–0.06 0.433
1 - Specificity
a Based on the analysis of 582 systemic vasculitis patients with all data available in the

French Vasculitis Study Group's database: 194 PAN (among whom 117 had HBV-related Fig. 25.1 Receiver operating-characteristics curves illustrating discriminatory
PAN) and 388 other systemic vasculitides (GPA, n=144; EGPA, n=115; MPA, n=101; performances of the proposed clinical diagnostic set of criteria for PAN
cryoglobulinaemia, n=28). (Table 25.3) compared to the 1990 ACR classification criteria (Table 25.1).
(Henegar, C., Puéchal, X., Pagnoux, C., et al. (2008). A paradigm of diagnostic criteria for
polyarteritis nodosa: analysis of a series of 949 patients with vasculitides. Arthritis Rheum,
52 (Suppl.), S225.)
per 1 000 000 inhabitants in Seine–Saint-Denis, a northern suburb
of Paris (Mahr et al. 2004). Its prevalence was the highest among
affects the lungs. Specifying that ANCA are not associated with PAN the vasculitides that were studied (MPA, PAN, granulomatosis with
and the addition of a category for ANCA-associated vasculitis in this polyangiitis (GPA, Wegener’s granulomatosis), and EGPA) but
new nomenclature reflect advances in knowledge about ANCA since its incidence has declined in parallel with that of HBV infection
the 1994 Chapel Hill Consensus Conference nomenclature. (Guillevin et al. 2005).
These criteria are only satisfied in the patients presenting with
the most severe forms of MPA or PAN who have nephropathy with Pathogenesis
or without lung involvement, respectively. Other patients may not
be classified so easily when clinical manifestations do not include The immunopathogenic mechanisms leading to vascular injury
kidney and/or lung involvement. In that setting, the Chapel Hill in PAN are probably heterogeneous. The mechanism of vascular
Nomenclature, such as the ACR classification of PAN, does not inflammation implicated most often, based on animal models, is
contain enough items to clarify the distinction. Even biopsies are an immune-complex-induced lesion (Trépo et al. 1974; Fye et al.
sometimes unable to contribute to the diagnosis because vessel type 1977; Guillevin et al. 1990). In some cases, PAN is the consequence
and size are not easy to describe. of HBV infection and there is evidence of immune-complex dis-
Diagnostic criteria based on clinical manifestations and biologi- ease with hepatitis B surface antigen (HBsAg) being the trigger-
cal or immunological signs can guide clinicians. Because ANCA ing factor (Trépo and Thivolet 1970b; Prince and Trépo 1971;
are present in the majority of MPA patients, they might be a major Guillevin et al. 1995b). Almost all cases of HBV-related PAN are
indicator for classification and their presence should exclude the associated with wild-type HBV, HBe antigenaemia, and high HBV
diagnosis of PAN. Diagnostic criteria are needed but have not replication, supporting the concept that lesions can result from the
yet been established and we propose herein a list of clinical items deposition of soluble viral antigen–antibody complexes in anti-
able to help diagnose PAN (Table 25.3 and Figure 25.1) (Henegar gen excess, possibly involving HBeAg. According to this hypoth-
et al. 2008). esis, immune complexes would activate the complement cascade,
whose activated products in turn attract and activate neutrophils.
However, the possible association of PAN with a precore muta-
Epidemiology tion would abrogate the formation of HBeAg and counter that
PAN is a rare disease that affects all racial groups. Estimates of the postulate. It might suggest that other, still undefined, circulating
annual incidence of PAN-type systemic vasculitides in the general HBV-related antigens distinct from HBeAg could be involved.
population range from 4.6 per 1 000 000 inhabitants in England Another mechanism could be direct injury of endothelial cells
(Scott et al. 1982), and 9.0 per 1 000 000 inhabitants in Olmsted caused by viral replication (Mason et al. 2005). However, no recent
County, Minnesota, to 77 per 1 000 000 inhabitants in a hepati- study has explored the pathogenic mechanisms of PAN, which
tis B-hyperendemic Alaskan Eskimo population (McMahon et al. remain largely unexplained.
1989). In a German study (Reinhold-Keller et al. 2002), the PAN ANCA were rarely found in earlier studies (Guillevin et  al.
incidence was extremely low (0.3–0.4/1 000 000 inhabitants accord- 1993b) and re-examination of those data showed that the major-
ing to the year and part of the country). A comparison of the PAN ity of ANCA-positive patients would now be diagnosed as hav-
incidence in two European areas (Lugo, Spain, and Norwich, UK) ing MPA (Guillevin et al. 1995a). Our current opinion now is that
found no significant differences: respectively, 6.2 and 9.7/1 000 000 ANCA positivity should be considered a criterion for excluding the
inhabitants (Watts et al. 2001). In France, PAN prevalence was 34 diagnosis of PAN.
334 SECTION 5 vasculitic diseases and syndromes and related disorders

Antiendothelial cell autoantibodies (AECA) are directed patients a year have been diagnosed throughout France. Moreover,
against antigens expressed on the surface of those cells and have the frequency of PAN, due to HBV infection or not, has also
been implicated as a participating pathogenic factor in vascu- decreased in parallel.
litis (Cid et  al. 1996), by causing direct endothelial damage.
However, little is known about vasculitis target autoantigens or Other aetiologies
whether AECA are really of pathogenic significance. Indeed, Some other viruses have also been associated with PAN but can
AECA are not disease-specific, because they are detectable in only explain a few cases. The prevalence of HCV in our patients
various types of vasculitis including GPA, MPA, and Kawasaki’s was anecdotal. Some patients were HBV–HCV co-infected. HCV
disease, but also in PAN (Le Tonqueze et al. 1996; Salojin et al. does not seem to be an important aetiological factor for PAN. GB
1996; Chanseaud et  al. 2005; Tamby et  al. 2005). The 60-kDa virus-C, a virus searching for a disease, has been sought in patients
heat-shock protein (Hsp60) was shown to be targeted by some with PAN, but has not been considered a potential agent respon-
AECA in PAN patients’ sera (Le Tonqueze et  al. 1996; Salojin sible for the disease (Servant et al. 1998). A few cases associated
et al. 1996; Jamin et al. 2005; Dugué et al. 2004). Some authors with parvovirus B19 infections have been described (Corman and
(Chanseaud et al. 2005; Tamby et al. 2005) found that IgM and Dolson 1992; Leruez et  al. 1994)  but systematic testing of PAN
IgG from PAN patients reacted specifically with tissue antigens patients did not show them to have a higher frequency of parvovi-
in artery and kidney extracts. rus B19 infections than the control population (Leruez et al. 1994).
Cytokines may be involved in the pathogenesis of vasculitides. Other viruses have been incriminated in PAN and anecdotal human
Marked increases of serum interferon-α and interleukin-2 levels, immunodeficiency virus infections have been reported (Calabrese
and moderately elevated levels of tumour necrosis factor-α and 1991; Gherardi et al. 1993; Gisselbrecht et al. 1998). Herpes zoster
interleukin-1β have been detected in PAN and EGPA patients might also be responsible for vasculitis (Rodríguez Pereira et  al.
(Grau et  al. 1989). Immunohistochemical studies performed on 1997) and some members of the Herpesviridae family, for exam-
biopsied perineurial and muscle vessels from homogeneous pop- ple herpesvirus 68, has been considered to be possibly involved in
ulations of PAN patients showed that inflammatory infiltrates the pathogenesis of large-vessel vasculitis in experimental models
consisted mainly of macrophages and T lymphocytes, particu- (Weck et al. 1997).
larly of the CD8+ subset (Panegyres et al. 1990). A homogeneous In addition to infectious causes, PAN has been described in
immunofluorescent-labelling pattern of nerve and muscle vas- association with cancers and haematological diseases, mainly
culitic lesions suggests that some circulating factor, such as an malignant haemopathies. The closest relationship has been estab-
antibody or complement component binding to a common deter- lished with hairy-cell leukaemia (Elkon et al. 1979; Krol et al. 1983;
minant expressed by muscle and nerve, might accumulate around Hasler et al. 1995). Malignancies are, for the most part, associated
epineurial and muscle endothelial cells (Panegyres et  al. 1990). with small-size-vessel vasculitides and very few cases associated
Those observations suggest that T-cell mediated immune mecha- with PAN have been reported. In a retrospective study from the
nisms might play a role in the development and perpetuation of Cleveland Clinic Foundation (Hutson and Hoffman 2000), only 69
PAN lesions. patients had both a malignancy and a systemic vasculitis over an
To date, there is no reliable animal model of the disease. In fact, 18-year period. Among them, only 12 had both vasculitis and can-
the PAN-like disease in cynomolgus macaques (Porter et al. 2003; cer occurring within the same 12-month period, and only two of
Colmegna and Maldonado-Cocco 2005), which is very similar to the latter had PAN.
the human disease, occurs only sporadically.
Pathology
Aetiology and precipitating factors The histological lesion defining PAN is focal segmental necrotiz-
For the majority of patients, the search for an aetiology remains ing vasculitis of medium-sized arteries, less commonly arterioles,
elusive. Nevertheless, in a few cases, infections, mainly viral, have and only rarely capillaries and venules. The lesion may occur in any
been identified as being responsible for PAN, as in the case of HBV artery of the body, but involvement of the aorta and/or other large
infection. elastic arteries and/or pulmonary arteries has been described only
rarely. The acute phase of arterial wall inflammation is character-
HBV-related PAN ized by fibrinoid necrosis of the media and an intense pleomorphic
Although viral antigens or immune complexes have rarely been cellular infiltration, with predominantly neutrophils and variable
found in the vessel walls of PAN patients, a close relationship numbers of lymphocytes and eosinophils.
has been demonstrated between PAN and HBV infection (Trépo The normal architecture of the vessel wall, including the elastic
and Thivolet 1970a). In France, HBV infection transmitted by laminae, appears to be completely destroyed and replaced by a band
contaminated blood transfusion has now disappeared; the last of amorphous eosinophilic material, resembling fibrin when stained.
proven case occurred in 1987 (Guillevin et al. 2005), but intrave- Arterial aneurysms and thromboses can occur at the site of the lesion.
nous drug abuse has rapidly become a major cause of HBV-related Arterial healing is characterized by fibrotic endarteritis that may lead
PAN (Guillevin et al. 2005) as is sexual transmission of HBV to to aneurysm regression or, when too abundant, to vessel occlusion.
non-vaccinated individuals at risk. The development of anti-HBV One of the characteristic histological features of PAN is the coexist-
vaccines and their administration to people at risk also explain the ence of necrotizing vasculitis and a healed lesion or normal arteries
dramatically decreased numbers of new cases since 1989. Over the in different tissues or different parts of the same tissues.
past few years, the frequency of HBV-related PAN has declined Biopsies from several sites can be diagnostic for PAN. In
to 20% (Guillevin et al. 2005). For the last 3 years, fewer than ten patients complaining of myalgias with or without concomitant
CHAPTER 25 polyarteritis nodosa 335

mononeuropathy multiplex, or presenting with constitutional symptoms occur early in the course of the disease, may be present
symptoms, muscle biopsy is useful for diagnosis. In general, muscle at its onset, and isolated. The diagnosis of PAN is made only when
biopsy should be performed at the site of myalgias or in the gastroc- other systemic manifestations occur. The characteristics of fever
nemius or peroneal muscles. The diagnostic sensitivity of biopsies vary from one patient to another:  high, remittent, with chills or
performed in proximal muscles (deltoid or quadriceps) is lower intermittent.
than that of those performed in distal muscles. Nerve biopsy in a
sensory branch of the sciatic or peroneal nerve can also be taken Myalgias and arthralgias
when the patient suffers from distal mononeuropathy multiplex Half of the patients suffer from myalgias. They may be intense, dif-
in its sensory or sensorimotor form. When patients do not have fuse, and occur spontaneously, or only after applying pressure to
sensory signs in a candidate region for biopsy, the results have low the muscle. The diagnosis of an inflammatory myopathy can be
sensitivity. Nerve biopsy can often prove the presence of vasculitis considered at that point but muscle enzymes are usually normal or
but cannot easily give the diagnosis of PAN because nerve arteries only slightly elevated. Conversely, amyotrophy can be marked, but
are usually small. Furthermore, the biopsy itself can cause a perma- mostly reflects weight loss, sometimes of more than 20 kg. Some
nent sensory deficit. A deep skin biopsy is easy to obtain and can patients are bedridden due to the intensity of pain and amyotrophy.
show medium-size-vessel involvement in patients with infiltrated Magnetic resonance imaging (MRI) of painful muscles may show
necrotic purpura. In patients with visceral involvement, surgical some inflammation, with hyperintensity on T2-weighted sequences
specimens, such as small intestine, can also provide the diagnosis. (Figure 25.2), usually enhanced after gadolinium injection. Muscle
Because renal involvement results from ischaemia and the risk of biopsy can contribute decisively to the diagnosis of PAN when per-
haematoma due to microaneurysm rupture, renal biopsy is not rel- formed at this stage of illness. Forms of necrotizing vasculitis lim-
evant or recommended. Temporal artery biopsy can show necrotiz- ited to one muscle or group of muscles with histological features of
ing vasculitis of the main artery or one of its branches, even in the PAN but no other organ involvement are described with increasing
absence of clinical symptoms (Haugeberg et  al. 1997; Généreau frequency (Nakamura et al. 2003; Lupoli et al. 2004).
et al. 1999; Hamidou et al. 2003). Arthralgias predominate in knees, ankles, elbows, and wrists,
rather than shoulders or hips. Synovitis is rarely observed and joints
Clinical features are usually not deformed. Symptoms differ from those of rheuma-
toid arthritis, a disease that can also be associated with vasculitis as
Age and sex
a secondary phenomenon.
PAN can be observed in patients of all ages, including children and
the elderly, but predominates between 40 and 60 years, with no sex Neurological manifestations
predominance. In our patients, the mean age was 47.5 (range 4–92)
Peripheral neurological symptoms are frequent; central nervous
years (Table 25.4).
system (CNS) involvement has been reported, but is rare.
General (constitutional) symptoms Peripheral neuropathy
Poor general condition is common among patients with PAN, with Peripheral neuropathy is the most frequent finding in PAN patients
weight loss and fever being present in two-thirds. Non-specific (50–75%) (Frohnert and Sheps 1967; Moore and Fauci 1981;

Table 25.4 Percentages of main organ/ system involvements in or manifestation of PAN according to main published series

Reference Patients (n) Mean age Organ/ system involved or manifestation (%)

Heart Hypertension Skin CNS PNS Kidney GI


Pagnoux et al. 2010 348 51 22 35 50 5 74 51 38
Gunal et al. 1997 15 (children) 10 67 20 20 0 0 13 47
Travers et al. 1979a 17 41 89 29 (mild) 65 41 59 77 65
41 (severe)
Schrader et al. 1985a 36 – 61 72 – – – 76 –
Fortin et al. 1995 45 54 18 – 44 24 51 44 53
Cohen et al. 1980a 53 54 4 14 58 – 60 66 25
Leib et al. 1979a 64 47 30 25 28 25 72 63 42
Frohnert and Sheps 1967a 130 – 10 – 58 3 52 8 14
Guillevin et al. 1988 165 48 23 31 46 17 67 29 31
Mowrey et al. 1954a 607 – – 58 25 – 66 83 48
a These older studies may have included PAN patients who would now be diagnosed as having MPA.

CNS, central nervous system; GI, gastrointestinal; PNS, peripheral nervous system.
336 SECTION 5 vasculitic diseases and syndromes and related disorders

12–18 months are often necessary to obtain and evaluate maxi-


mum recovery. The degree of recovery is variable and unpredict-
able because, in some patients, severe neuropathy with extensive
palsies may regress completely, whereas in others, minor pal-
sies or sensory symptoms may never totally disappear. Sensory
symptoms, usually paraesthesias, persist longer and sometimes
indefinitely, and flares of peripheral neuropathy cannot be pre-
dicted. Electromyography can be used during follow-up to
evaluate neuropathy regression or progression. Physical rehabili-
tation is indicated to prevent deformations and accelerate clinical
improvement.

Central nervous system involvement


CNS involvement is much less common than peripheral neuropa-
thy, and is noted in 3–38% of PAN patients (Frohnert and Sheps
Fig. 25.2 Peroneal muscle hypersignal on T2-weighted MR images in a PAN 1967; Sack et  al. 1975). It is usually a late manifestation in the
patient with muscle involvement. course of the disease and is considered a poor-prognosis factor
(Guillevin et al. 1996; Castaigne et al. 1970). Its common presen-
tations include encephalopathy in 8–20% (Moore and Fauci 1981;
Scott et  al. 1982), affecting cognitive function and character-
Guillevin et al. 1992b) and is the earliest symptom of the disease in ized by disorientation, psychosis and hallucinations, delusion or
23–33% (Frohnert and Sheps 1967; Guillevin et al. 1988). Indeed, diminished consciousness, and focal or multifocal disturbances
a major variant of PAN exists that exclusively involves peripheral of the brain and spinal cord, with seizures, strokes and/or suba-
nerve and muscle (Abgrall et al. 2001). Onset is usually acute, but rachnoid haemorrhages (Moore and Calabrese 1994), resulting
may be more indolent, particularly in the elderly. Sensory signs either from vasculitis of a cerebral artery (i.e. arteritis, rupture
are responsible for hypo- or hyperaesthesia, dysaesthesia or frank of cerebral artery aneurysm, haematoma), or as a consequence
pain as the prominent and earliest features (Moore and Calabrese of malignant hypertension. A case of extrapyramidal syndrome
1994). Usually, motor deficits start later, also with sudden onset, was also described (Mayo et al. 1986). Distal occlusion of spinal
but may sometimes precede the sensory sign(s). The first mani- vessels can be responsible for spinal cord and/or cauda equina
festations often affect the lower limbs, with one particular nerve involvement (Castaigne et  al. 1970), which, in turn, engenders
involved. Later, other nerves become affected, with this pattern sphincter dysfunction (Amarenco et al. 1988).
being referred to as mononeuritis (or mononeuropathy) multi- Computed tomography (CT) scans of the brain are usually nor-
plex. Indeed, mononeuropathy multiplex affects 56.5–61.5% of the mal but MRI may show T2-weighed hyperintensities, localized to
patients (Moore and Fauci 1981; Said and Lacroix 2005). The main the white matter in various parts of the brain, which are suggestive
localizations are distal and asymmetric. The following nerves are but not specific. MR angiography and conventional cerebral angi-
preferentially involved: superficial peroneal, sural, radial, cubital, ography (which is performed less frequently than in the past) are
and/or median. In its late stage, so many nerves can be involved often normal, but non-specific focal or diffuse segmental narrow-
that mononeuropathy multiplex can be mistaken for a symmetri- ing of the intracranial vessels may be seen.
cal process. Segmental oedema may precede the development
of palsies, reflecting capillary permeability dysfunction, which Cranial nerve involvement
regresses spontaneously or with treatment. Amyotrophy quickly Cranial nerve palsies are found in approximately 1% of PAN
ensues. Cerebrospinal fluid, when analysed, is generally normal. patients (Guillevin et al. 1988). Oculomotor (III), trochlear (IV),
Electromyography typically shows axonal neuropathy. Motor and abducens (VI), facial (VII), and/or acoustic (VIII) nerves (Ford
sensory nerve action-potential amplitudes are markedly decreased, and Sieckert 1965) are most often affected. Partial or full recov-
or may even be absent, in the most severely affected nerves, while ery can be achieved with treatment, but is unlikely to be complete
motor nerve-conduction velocities are normal or only slightly for the VIIIth vestibulocochlear nerve(s) (Vathenen et al. 1988).
diminished. The extent of the neuropathy on electromyography Vasculitis of the optic nerve, optic chiasm, and/or occipital cor-
may be greater than expected based on clinical manifestations tex has also been thoroughly described (Kinyoun et  al. 1987).
(Bouche et  al. 1986; Said 1997). Muscle and nerve biopsies may Blurred vision or visual loss may also occur as a result of cho-
contain characteristic muscle and epineurial artery lesions. roiditis (see Section Ophthalmological Signs), retinitis, or brain
Mononeuropathy (simplex) is less frequently observed, in 16.5% of parenchymal arteritis (Moore and Calabrese 1994). Cerebrospinal
the patients, and distal symmetrical sensory, often patchy, or sensori- fluid is usually normal but, in a few cases, an elevated protein
motor neuropathy in 25%. Very few cases of brachial plexus neurop- concentration, without increased cellularity, is present (Hirohata
athy have been reported (Jamieson et al. 1991). Conduction block, et al. 1993).
suggestive of demyelination, as in Guillain–Barré syndrome, nerve
entrapment, or other acquired demyelinating neuropathies, has been Skin manifestations
described in rare cases (Allan et al. 1982; Ropert and Metral 1990). Cutaneous lesions have been reported in 20–60% of patients with
Under treatment, mononeuropathy multiplex in PAN regresses systemic PAN, but less frequently in those over 65 years old (Cohen
slowly and patients may recover without sequelae although et al. 1980; Puisieux et al. 1997). It must be remembered that, in
CHAPTER 25 polyarteritis nodosa 337

the majority of series, PAN and MPA were not differentiated, and symptoms, such as arthralgias, hypertension, GI manifestations,
that, today, most cutaneous manifestations would probably to be and/or peripheral neuropathy. However, PAN may at first be
attributed to MPA. Theoretically, cutaneous or subcutaneous nod- limited to the skin in a few patients, with systemic PAN develop-
ules are hallmarks of PAN, occurring in 8–27% of patients. They ing later, after a variable period ranging from 1 to 20 years after
occur in clusters along the trajectories of superficial arteries and the first cutaneous signs (Diaz-Perez and Winkelmann 1974).
often disappear spontaneously within a few days, before new ones Conversely, PAN may remain strictly confined to the skin, as a
develop. They measure 5–20 mm in diameter and are dermal and/or chronic disease, often associated with multiple cutaneous relapses
hypodermal, mainly located in the lower legs, especially the knees (see Chapter 27).
and feet. Indeed, the most common cutaneous finding is palpable
purpura, often necrotic (Figure 25.3), corresponding to subcuta- Renal manifestations
neous small-vessel vasculitis, in association with the involvement
Renal artery involvement can be responsible for mild-to-severe
of medium-sized vessels. Although the Chapel Hill Nomenclature
and malignant arterial hypertension and/or vascular ischaemic
distinguishes large, medium-size and small-size-vessel vascu-
nephropathy with renal insufficiency (Cohen et  al. 1986), which
litides, it also recognizes some overlap forms, that is PAN with
has been identified as a poor-prognosis factor (Guillevin et  al.
some, but not predominant, involvement of small vessels, espe-
1996). Angiography (when performed despite renal involvement)
cially in skin biopsies (ANCA Workshop, Birmingham, UK, 1998,
may show renal parenchymal infarcts, and characteristic multiple
unpublished revised version of the Nomenclature). A convenient
stenoses and microaneurysms of branches of celiac, mesenteric,
threshold defining small-size-vessel vasculitis has been set, pri-
and renal arteries (Figure 25.5), in 90% and 40–62% of the cases
marily for nerve biopsies, at around 50–70  µm in diameter for
with or without GI symptoms, respectively (Camilleri et al. 1983;
affected epineurial arteries and vasa nervorum (Gherardi et  al.
Ha et al. 2000; Miller 2000). Microaneurysms can occasionally rup-
1993; Moore 1995).
ture spontaneously or after renal biopsy, which is strongly contrain-
Ulcerations and livedo are less frequent (Leib et al. 1979). Livedo
dicated in the presence of microaneurysms (Cornfield et al. 1988;
reticularis may precede, follow, or occur concomitantly with the
Tasdemir et al. 1988; Akcicek et al. 1994). Renal haematoma can be
onset of nodules. Livedo reticularis in PAN is typically localized
extensive and requires embolization (Hachulla et al. 1993) or even
on the lower limbs, the backs of the arms and, sometimes, on the
nephrectomy (San et al. 1990).
trunk. It has a fishnet reticular pattern, with discontinuous circles,
Acute renal insufficiency usually occurs early during the disease
and often with some infiltrated areas.
course or a flare. Plasma exchanges might be required initially,
Local rupture of superficial arteries may lead to cutaneous
but renal function outcome remains unpredictable. Some patients
haematoma or ecchymosis. A biopsy of infiltrated and/or cen-
may come off dialysis but end-stage renal disease, resulting from
tral lesion zones can show vasculitis. Painful ulcerations may
chronic renal ischaemia, may develop later, sometimes years after
develop, frequently associated with indurated plaques result-
the first manifestations of PAN.
ing from the coalescence of nodules. Peripheral embolization
of thrombi may cause infarction of the extremities (toes, fin-
gers; Figure 25.4) or some cutaneous areas, often proximal, with Arterial hypertension
cholesterol or atherosclerotic emboli being possible differential Hypertension is present in a mean of 40% of PAN patients and is
diagnoses. usually mild; however, it can be triggered or exacerbated by corti-
Other manifestations have been reported, such as bullous costeroids. Severe, malignant hypertension was detected in 7/165
purpura and vesicles that may precede necrosis, urticaria, tran- (4%) of our patients (Guillevin et al. 1988) and 15% of those with
sient erythema, or erythema annulare fugax, superficial phlebi- HBV-related PAN (Guillevin et  al. 1993a). Hypertension can be
tis, Raynaud’s phenomenon, and splinter haemorrhages. Most attenuated with angiotensin-converting enzyme inhibitors, which
patients’ cutaneous signs are associated with other PAN-related have been proven beneficial in this context.

Fig. 25.3 Necrotic purpura in the legs and ankles of a PAN patient. Fig. 25.4 Toe ischaemia in a PAN patient.
338 SECTION 5 vasculitic diseases and syndromes and related disorders

but severe because it can be responsible for haemopericardium.


Most of the cases have been described in infants (Tang and Segal
1971; Holt and Jackson 1975), with some of them possibly being
Kawasaki’s disease rather than PAN. To date, no guidelines regard-
ing the indications, modes of exploration, or frequency of coronary
artery involvement have been formulated for PAN patients without
cardiac symptoms. Cardiac MRI seems to be promising but needs
to be further evaluated.
The right ventricle may also be involved, as reported in a small
series for six of eight patients with heart involvement (Blétry et al.
1980). Finally, interstitial myocarditis was seen in 14% of autopsied
patients (Schrader et al. 1985).
Murmurs are relatively frequent, having been noted in 39%
(Holsinger et al. 1962) of vasculitis patients. They are usually innoc-
uous, favoured by anaemia, underlining the rarity of valve involve-
ment in PAN. Indeed, endocarditis is rarely observed in PAN and
the presence of endocardial manifestations should orient the diag-
nosis towards another entity. However, some patients with mitral
and tricuspid regurgitation, supposedly specific to PAN, have been
reported (Blétry et al. 1980; Gunal et al. 1997).
The pericardium is implicated even more rarely, in only 0–5% of
PAN patients (Hu et al. 1997), rising to 19% (Gunal et al. 1997) to
one-third (Holsinger et al. 1962) in autopsy series; usually pericar-
Fig. 25.5 Multiple renal artery stenoses and microaneurysms (arrows) with an
inferior kidney pole infarction on arteriography (paler, less well irrigated pyramidal ditis is non-specific and occurs secondarily to myocardial involve-
corticoparenchymal area) in a PAN patient. ment, when not attributable to other cause(s). About half of the
first reported cases of pericardial effusion were indeed secondary
to uraemia, which is much less frequent now that the diagnosis and
overall therapeutic management of patients have improved.
Cardiac manifestations In patients with cardiac involvement, tachycardia is very com-
Cardiac involvement was mentioned in the first publication on mon and often precedes other cardiac symptoms. Sinus tachycardia
PAN (Küssmaul and Maier 1866), which described a case of ‘nodu- is usual and non-specific. Arrhythmias and conduction disorders,
lar coronaritis’. It was subsequently reported with frequencies rang- mainly supraventricular, can occur in 2–19% of PAN patients
ing from 10% in clinical investigations of PAN patients (Frohnert (Holsinger et al. 1962; Blétry et al. 1980) because of arteritis of the
and Sheps 1967), to 40% when considering radiological and/or sinus node or neighbouring nerve fibres.
electrocardiographic anomalies, and to 78% in a histopathological
study (Holsinger et al. 1962).
Congestive heart failure is the main clinical feature, occur-
Aortic dissection and peripheral
ring in 6–57% of PAN patients, but less frequently than in EGPA. vascular manifestations
It is specific, due to vasculitis of the coronary arteries or their Aortic dissection is a rare complication attributed to diffuse vas-
branches, with myocardial arteriolar infarcts, or can result from culitis of the aorta vasa vasorum. It has been reported to evolve to
other vasculitis-related organ involvements or disorders, mainly fatal tamponade (Iino et al. 1992). Dissections of proximal aortic
hypertension and/or renal involvement (Blétry et al. 1980). Specific branches have also been reported, but some were attributed to other
cardiomyopathy can occur as early as 3–4  months after PAN causes, such as atherosclerotic aneurysms (Bookman et al. 1983;
onset. However, despite coronary artery vasculitis, clinical angina Lomeo et  al. 1989; Hautekeete et  al. 1990; Hachulla et  al. 1993),
is rare, reported in ‘only’ 2–18% of patients (Frohnert and Sheps syphilis, cystic medial necrosis, trauma, sepsis, or hypertension.
1967; Blétry et al. 1980), as are myocardial infarctions diagnosed Peripheral arterial occlusions may be responsible for distal gan-
throughout patients’ lives, in 1–12%, whereas the latter were a grene of the toes or fingers. Angiography can demonstrate the pres-
common finding at autopsy (Frohnert and Sheps 1967). Among ence of stenoses and/or microaneurysms (Figure 25.6) (Héron et al.
66 autopsied patients, 41 had features of myocardial infarction, but 2003). Raynaud’s phenomenon, when present, can remain isolated
only three of them had clinical symptoms and three had coronary or be complicated by necrosis. In some patients, type II or III cry-
atherosclerosis (Holsinger et al. 1962). Schrader et al. (1985) autop- oglobulinaemia can be found. It is not known whether cryoglob-
sied 36 PAN patients, 50% of whom had evidence of coronary arte- ulinaemia may accompany PAN, or if its detection should rather
ritis, with severe lesions involving small subepicardial vessels just as exclude the diagnosis of PAN; this latter position is probably more
they entered the myocardium and 8% had large infarcts. rigorous, at least for clinical trials.
Angiography can prove coronary involvement in 85% of the
patients with clinical signs of infarction. In the remaining 15%, Gastrointestinal manifestations
infarction may be due to arteritis in small coronary vessels or GI tract involvement represents one of the most severe manifes-
spasms (Rajani et  al. 1991). Coronary aneurysms were found in tations of PAN, reportedly affecting 40–60% of the patients, more
9% of children with PAN (Takahashi 1993). Their rupture is rare often in HBV-related PAN. GI manifestations are usually associated
CHAPTER 25 polyarteritis nodosa 339

Fig. 25.7 Splenic infarct (arrow) seen on a PAN patient’s abdominal computed


tomography scan.

the gallbladder (2–17% of PAN patients) or the appendix (Chen


1989; Nohr et al. 1989; Ito et al. 1991; Parangi et al. 1991; Blidi et al.
1996) can be the first sign of PAN, sometimes isolated, hence rep-
resenting another possible localized form of PAN. However, it was
reported that 25% of the patients with isolated and histologically
proven vasculitic appendicitis further evolved to systemic PAN,
within the following 5 years (Godeau et al. 1979). Indeed, except
for isolated appendicitis, in which vasculitic aetiology might be
perceived as a pathological curiosity, medical therapy alone for
GI-localized manifestations appears to be overconfident. However,
the prognosis of such limited forms remains good, owing to
prompt combined medical and surgical treatment. Likewise, acute
necrotizing or, less commonly, chronic pancreatitis, sometimes
with pseudocysts, has been diagnosed in approximately 2–3% of
the patients (Cacoub et al. 1988), whose prognoses are extremely
Fig. 25.6 Tibial artery stenoses and microaneurysms (arrow) in a PAN patient. dismal because of the regular association with severe small intes-
tine ischaemia and/or perforations.
Digestive malabsorption and exudative enteropathy have also
been reported on rare occasions. The liver and spleen can be
with other systemic signs of PAN, but they can be the first symp- involved, even in the absence of HBV infection, with infarct(s)
toms of PAN in 2–16%, more often in HBV-related PAN (Zizic (Figure 25.7) and/or haematoma(s) (Li et al. 1979; Nakazawa et al.
et al. 1982; Camilleri et al. 1983; Le Thi Huong et al. 1985). They 1992) that are usually clinically silent. One case of fibrinoid necrosis
are the major cause of deaths within the first year after PAN onset, of the splenic artery with subsequent splenic rupture was described
and thereafter the third, just after infections and heart disease (Fortin et al. 1995).
(Bourgarit et al. 2005). When abdominal pain is present, abdominal CT scans can be
The spectrum of GI symptoms is broad and usually non-specific, useful to detect plain organ infarcts or pancreatitis, but it has only
with abdominal pain being the most frequent, recorded in 75% sensitivity and vasculitis has no specific features, even though
30–40% of PAN patients, and in 97% of those with GI involve- bowel wall inflammation and thickening may be suggestive of
ment. It is always difficult to establish the exact origin of this pain more severe GI involvement. Gastric and/or colon endoscopy can
but its presence can be the symptom signalling more severe and detect ischaemic areas and ulcers that may precede perforation, but
life-threatening GI vasculitis, and, hence, should always be taken can also easily cause iatrogenic perforations, mostly during colo-
seriously. noscopy, in these patients with fragile and inflamed GI mucosae.
GI haemorrhages and small intestine perforations are the most Furthermore, GI biopsies are rarely contributory. Angiography
feared manifestations (Guillevin et al. 1995c), with reported fre- may detect infarcts, haematomas, or more diagnostic arterial sten-
quencies of 20–50% and 2–40% of PAN patients, respectively. oses and microaneurysms, 1–5 mm in diameter or more, in 90% of
When present, ischaemic vasculitis mainly affects the small bowel the patients with GI symptoms. These occur mainly in renal (54%),
then, more rarely, the colon or stomach. Oesophageal perforations celiac (24%), mesenteric (14%), hepatic, or, more rarely, splenic
are rare (Lee and Kay 1958; Gourgoutis et al. 1971). Vasculitis of arteries (Camilleri et  al. 1983; Ewald et  al. 1987; Miller 2000),
340 SECTION 5 vasculitic diseases and syndromes and related disorders

which have no prognostic value. Intraperitoneal ruptures of these Bone manifestations


aneurysms have been reported that, when not rapidly fatal, might
Periosteal modifications can be seen (Short and Webley 1984;
be treatable in some specialized interventional radiology centres,
Esteva-Lorenzo et  al. 1994; MacDonald and Blake 2004). They
with selective arterial embolization (Bookman et al. 1983: Hachulla
develop mainly in the legs. Localized oedema and pain are com-
et al. 1993).
mon symptoms. When biopsies are performed, necrotizing vascu-
Severe GI manifestations, such as bowel perforations and/
litis can be observed. A few cases of bone necrosis have also been
or ischaemia, peritonitis, intestinal occlusion (and pancreati-
reported (Wang et al. 1988).
tis because of its frequent association with bowel perforations)
have poor prognoses. Treatment must combine prompt surgical Ophthalmological signs
intervention and medical therapy with corticosteroids (CS) and
The eye can be affected in PAN, sometimes severely, as with unilat-
immunosuppressants. The 5-year survival rate of the patients with
eral or bilateral choroiditis (Kinyoun et al. 1987), iritis, iridocycli-
these severe GI manifestations was indeed only 56%, compared to
tis, retinal detachment, and/or retinal vasculitis (Akova et al. 1993;
82% for those with less severe GI involvement (Levine et al. 2002;
Horgan et al. 1986; Malan et al. 1986). Cotton-wool nodules and/or
Pagnoux et al. 2005).
uveitis have also been observed.
Orchitis Miscellaneous
Orchitis is one of the most characteristic manifestations of PAN Rare anecdotal descriptions of specific gingivitis (Cowpe and
(Teichman et al. 1993; Warfield et al. 1994; Guillevin et al. 1995b) Hislop 1983; Thone 1988) and breast (Levy et al. 1986; Yamashina
and it was retained as one of the ACR classification criteria and Wilson 1985) involvement have been published.
(Lightfoot et al. 1990). Caused by testicular artery ischaemia, it is
rarely the first manifestation of the disease and is unilateral. When Specific manifestations of HBV-PAN
treated immediately, orchitis may regress under CS. In other cases,
the ischaemia can be irreversible. No infectious aetiology has ever The immunological process responsible for PAN occurs early dur-
been found. Fever and poor general condition are usual. In the ing the course of HBV infection. When it was possible to date
case of isolated testicular involvement, a tumour or testicular tor- the HBV contamination, in most cases PAN developed within
sion comes to mind and histological examination is able to provide less than 6 months after infection. Hepatitis usually remains silent
the diagnosis (Warfield et al. 1994). We and others have observed before PAN onset, which can be the first manifestation of HBV
that orchitis was often present in HBV-related PAN (Guillevin et al. infection. Hepatic cytolysis is usually moderate and cholestasis is
1995b) but no close relationship could be demonstrated between minor or absent. When liver biopsies were taken, they frequently
the viral infection and testicular manifestations. exhibited signs of chronic hepatitis, even when PAN became
manifest only a few months after HBV infection. In the group
of patients with HBV-related PAN, clinical data were roughly
Ureteral and urogenital manifestations
the same as those commonly observed for PAN (Table  25.5)
Ureteral manifestations are rare. They are observed in several (Guillevin et al. 2005), but some differences were found: patients
types of vasculitides and are not specific to PAN. They more often were usually under 40  years old; malignant hypertension (5%),
have been considered characteristic of small-vessel vasculitides. renal infarction, and orchiepididymitis (25%) were more frequent
However, unilateral or bilateral ureteral stricture (Gargollo et al. (Guillevin et  al. 1995b; Guillevin et  al. 2005). Other frequently
2004), ureteral extrinsic compression by retroperitoneal involve- observed symptoms arose from the abdomen (53%), resulting
ment (Lie 1992), and hydronephrosis due to ureteral myoneuropa- at times in surgical emergencies. In the study by Sergent et  al.
thy (Casserly et al. 1999) have been observed in PAN patients. (1976), two of the three deaths (among nine patients) were attrib-
In patients with difficulty urinating, urodynamic and electro- uted to colon vasculitis. Among the Eskimo patients described
physiological examinations can demonstrate hypoactivity of the by McMahon et  al. (1989), 31% of them died and one of the
detrusor, with loss of the sensation to urinate. The latter often four early deaths was the consequence of small-bowel perfora-
reflects major hypoaesthesia of the bladder due to vasculitis of the tion. Digestive and renal manifestations resulted from ischaemia
vasa nervosum of the vessels supplying the bladder rather than and angiography demonstrated the presence of microaneurysms
mucosal involvement. Spinal cord involvement or cauda equina and infarctions. It should be noted that subsequent angiograms
syndrome should be considered, but are extremely rare. showed that the aneurysms had disappeared when PAN had
Involvement of the glans penis resembling cancer has been been effectively treated (Darras-Joly et al. 1995). This disappear-
described (Casserly et al. 1999). Cervical (Abu-Farsakh et al. 1994; ance could result from aneurysms thrombosis with evolution to
Ganesan et al. 2000), ovarian (Kaya et al. 1994), and uterine artery fibrosis. HBV-related PAN is acute and initially severe but the
or body (Piette et al. 1987; Grasland et al. 1996) involvement have outcome is excellent for most patients when adequate treatment
been published. is prescribed. Seroconversion usually leads to recovery. Sequelae
are the consequence of vascular nephropathy but, even in patients
Pulmonary manifestations with renal insufficiency, it is possible to obtain recovery with little
The lungs are spared in PAN, unlike MPA, GPA, or EGPA. Although residual impairment of renal function.
autopsy studies had detected vasculitis of bronchial arteries, it
had been clinically asymptomatic. When pulmonary symptoms Localized forms of PAN
occur, infection should be looked for first, and is usually identified Localized PAN is rare except for the limited cutaneous forms that
(Godeau et al. 1994). represent 10% of all PAN cases (Moreland and Ball 1990). Isolated
CHAPTER 25 polyarteritis nodosa 341

Table 25.5 Relevant clinical and biological symptoms in 115 patients Table 25.6 EULAR/PReS proposed classification criteria for
with HBV-related PAN childhood PANa

Clinical symptoms Valuesa A systemic illness characterized by the presence of at least 2 of the following
7 criteria
Age, mean ± SD (years) 51.1 ± 17
1. Skin involvement (livedo reticularis, tender subcutaneous nodules, other
Sex ratio, M/F 74/41 vasculitic lesions)
General symptoms 97
2. Myalgias or muscle tenderness
Fever 69
Weight loss 87 3. Systemic hypertension, relative to normal childhood values
Arthralgias 56 4. Mononeuropathy or polyneuropathy
Myalgias 47 5. Abnormal urine analysis and/or impaired renal function
Mononeuritis multiplex 84 6. Testicular pain or tenderness
GI-tract involvement 53 7. Signs or symptoms suggesting vasculitis of any other major organ
Abdominal pain 51 system (GI, cardiac, respiratory, or CNS)
Bleeding 3
Appendicitis 2 In the presence of one, at least, of the following as a mandatory criterion
Small intestine perforation 6 1. Biopsy showing small and medium-sized artery necrotizing vasculitis
Cholecystitis 5
Pancreatitis 6 2. Angiographic abnormalitiesb (aneurysms or occlusions)

Renal and/or urogenital involvement 38 a Proposed criteria are currently being reviewed by the ACR.
b Should include angiography if MR angiography is negative.
Creatininaemia, mean ± SD, (mg/dl) 1.52 ± 1.39
Orchitis 25 (Reproduced from Annals of the Rheumatic Diseases, Ozen, S., Ruperto, N., Dillon M.J., et al.,
65, 936-41, 2006, with permission from BMJ Publishing Group Ltd.)
Microaneurysmsb 69
Renal infarctsb,c 28
Skin involvement 31 (cholecystectomy or appendectomy). In PAN limited to the skin
Purpura 17 or muscle, however, relapses are frequent. Among patients with
Infiltrated purpura 11 appendicitis in whom PAN was found during histological examina-
Livedo 10 tion, approximately 25% may subsequently develop systemic PAN
Nodules 9
(Godeau et al. 1979; Le Thi Huong et al. 1985).
Oedema (ankles) 16
Vascular manifestations 18
Hypertension 31 PAN in childhood
Malignant hypertension 5 After Kawasaki’s disease and Henoch–Schönlein purpura, PAN is the
Cardiac insufficiency 12 most common systemic necrotizing vasculitis in children (Ozen et al.
Raynaud’s phenomenon 3 2004). Neither the ACR criteria nor the Chapel Hill Nomenclature
Pericarditis 5
is validated for children. European League against Rheumatism/
Digital ischaemia 4
Myocardial infarction 1 Paediatric Rheumatology European Society (EULAR/PReS) have
proposed specific classification criteria for juvenile vasculitides
CNS involvement 10 that are currently being reviewed by the ACR (Table  25.6) (Ozen
Retinal vasculitis 2 et al. 2006). Those criteria were mainly based on a literature review
Erythrocyte sedimentation rate >30 mm/1st h 78 and a consensus-based process. In 2008, the Ankara Consensus
Conference had as its primary objective the validation of the EULAR
ANCAd 0
criteria for paediatric vasculitides. The final classification criteria of
a Values are %, unless otherwise indicated. classic PAN required a systemic inflammatory disease with evidence
b 66 angiographies.
of necrotizing vasculitis or angiographic abnormalities of medium-/
c Not related to vasculitis.
d ANCA were sought in the sera of 66 patients. small-sized arteries (mandatory criterion) plus one of the following
(Guillevin, L., Mahr, A., Callard, P., et al. (2005). Hepatitis B virus-associated polyarteritis five criteria: skin involvement, myalgia/ muscle tenderness, hyper-
nodosa: clinical characteristics, outcome, and impact of treatment in 115 patients. Medicine tension, peripheral neuropathy, renal involvement (Ozen et al. 2010).
(Baltimore), 84, 313–22.) The sensitivity and specificity of the final EULAR/PReS childhood
PAN classification criteria were 89.6% and 99.6%, respectively.
involvement of one skeletal muscle or a muscle group, and isolated The mean age at juvenile PAN diagnosis is 9 ± 4 years old, with
neuropathy (mononeuritis multiplex or simplex) without systemic equal numbers of boys and girls. Clinical manifestations fall within
symptoms have been described (Dyck et  al. 1987; Gallien et  al. the same spectrum as for adults, but with more frequent cutane-
2002), such as occasional cases involving only one organ, in order ous forms. Indeed, 80% of the affected children have skin involve-
of decreasing frequency: appendix, gallbladder (Blidi et al. 1996; ment and limited cutaneous forms may represent up to one-third
Pagnoux et  al. 2005), or uterus (Piette et  al. 1987). These forms of the cases.
usually carry good prognoses, often remitting spontaneously, with Prognosis appears to be better for children than adults, with an
local therapy (such as topical CS for skin lesions) or after surgery overall mortality rate of 1–16%. Relapses are more frequent, may
342 SECTION 5 vasculitic diseases and syndromes and related disorders

occur many years after diagnosis, and are sometimes preceded by present. Although the Chapel Hill consensus conference (Jennette
ear, nose, and throat infections. Evolution from limited cutane- et al. 1994; Jennette et al. 2013) did not consider microaneurysms,
ous disease to a systemic form is rare. Antistreptolysin antibodies we think their presence should be taken into consideration for
should be systematically sought, with serial determinations when the classification of PAN, and represent an exclusionary criterion
initially positive, because penicillin may help to cure the disease for MPA.
and has been shown to lower the relapse rate (Till and Amos
1997), but the optimal antibiotic regimen is unknown, because
relapses have been described up to 20 years after the first episode.
Outcome and prognosis of PAN
HBV-related PAN in children is rare (<5%), especially after large In its systemic form, PAN is an acute disease which can be severe
campaigns to vaccinate children against HBV. Conversely, a link and responsible for death, if not treated adequately. In historical
between PAN and streptococcal infections has been identified, series, only about 12% (Leib et al. 1979) or 13% (Frohnert and
which may be relevant for as many as 75% of paediatric cases (Till Sheps 1967) of untreated patients survived. Since the introduction
and Amos 1997). of CS, and their later combination with immunosuppressant(s),
antiviral therapy for HBV-related PAN, and plasma exchanges
when appropriate, the prognosis of PAN improved and overall
Laboratory tests survival rates increased to more than 76–89% for PAN (Gayraud
Indicators of inflammation are found in the majority of patients. et  al. 2001)  and 64–70% for HBV-related PAN (Guillevin
An erythrocyte sedimentation rate >60  mm/h (78–89% of the et al. 2005).
patients), elevated C-reactive protein, alpha-2 globulin levels, A systematic retrospective study was performed on 348 patients
and white blood cell counts (45–75% of the patients), sometimes diagnosed with PAN, registered in the French Vasculitis Study
eosinophilia >1500/mm3, and normochromic anaemia (34–79%) Group database and satisfying ACR criteria and Chapel Hill
are common laboratory findings. The presence of HBsAg should nomenclature (Pagnoux et  al. 2010). During a mean follow-up
be systematically sought. The frequency of HBV infection fell from of 68.3  months, 76 (21.8%) patients relapsed (63 [28%] with
38.5% during 1972–1976 to 17.4% in 1997–2002, with a peak of non-HBV-related PAN versus 13 [10.6%] with HBV-related
48.8% between 1982 and 1986. HBV is now rarely found because PAN; P <0.001); 86 (24.7%) patients died (44 [19.6%] with
of vaccination campaigns and increased safety of blood transfu- non-HBV-related PAN versus 42 [34.1%] with HBV-related PAN;
sions. We hypothesize that all PAN are the consequence of infec- P=0.003). Their respective 5-year relapse-free survival rates were
tion, even if a micro-organism has not been isolated from all the 59.4% (95% confidence interval [95% CI] 52.6–67.0) versus 67.0%
patients. The general measures to improve transfusion safety and (95% CI 58.5–76.8). Although treatment can now ensure favour-
patient care could explain the declining overall frequency of PAN. able outcomes for the majority of patients, some relapse or die of
We also observed that the majority of HBV-infected patients come disease- or treatment-related causes.
from northern or sub-Saharan African countries that have no vac-
cination policies, are intravenous drug addicts, or patients at risk Relapses
for sexually transmitted infections. Once remission has been obtained, PAN tends not to recur,
ANCA are rare in PAN, unlike MPA, GPA, and EGPA. In the unlike GPA or MPA. In an earlier study on 278 patients, 8% of the
context of systemic vasculitides, the presence of ANCA giving a HBV-related PAN and 19% of the non-HBV-related PAN patients
perinuclear labelling pattern (p), primarily directed against mye- relapsed, with a mean time to first relapse of 37 and 29 months,
loperoxidase (anti-MPO) in enzyme-linked immunosorbent assay, respectively (Gayraud et al. 2001). In a more recent investigation,
should be considered exclusionary for PAN and be taken as an 10% of the patients with HBV-related PAN relapsed (Guillevin
argument in favour of MPA. In some patients, especially in those et al. 2005). In HBV-PAN, relapses occur in those who have per-
without renal involvement, it is difficult, even when biopsies have sistent, active virus replication after treatment. Independent pre-
been examined, to distinguish between PAN and MPA. This dif- dictors of relapse retained in the stepwise multivariate analysis
ficulty most probably explains why pANCA with MPO reactivity were non-HBV-related PAN, with a corresponding hazard ratio
has been found in a small number of patients diagnosed with PAN (HR) of 2.27 (95% CI 1.11–4.63), and cutaneous manifestations
(Gaskin et al. 1991). Hauschild et al. (1994) reported an ANCA at diagnosis (HR 1.85 [95% CI 1.08–3.23]), especially when only
frequency of 19.4% in 36 patients with PAN (pANCA in three, nodules were considered (HR 2.21 [95% CI 1.30–3.78]) (Pagnoux
cytoplasm-labelling pattern (c) ANCA in four). et  al. 2010). The clinical pattern of relapse does not necessarily
repeat the original presentation, in that entirely new organs can
be involved at relapse. Although the severity of relapses cannot be
Angiography predicted, the clinical features at relapse are rash and arthralgias in
Bron et al. (1965) showed that angiography could visualize microa- most patients, and are generally less severe than during the initial
neurysms and stenoses in medium-sized vessels. Although microa- presentation.
neurysms are not pathognomonic, they are commonly present in
PAN and rare in GPA, EGPA, and MPA. Arterial saccular or fusi- Deaths
form aneurysms range in size from 1 to 5 mm and are predomi- Although the causes of death vary from one vasculitis to the other,
nantly seen in the kidneys, mesentery, and liver. The lesions may they can be divided into two categories: deaths attributable to the
disappear under effective vasculitis therapy (Darras-Joly et al. 1995). vasculitic process and those due to treatment side-effects.
Angiography is a useful tool when other diagnostic examinations Lethal complications may occur in any form of vasculitis that
are negative, especially when abdominal pain and nephropathy are involves major organs. A few patients die during the first months of
CHAPTER 25 polyarteritis nodosa 343

the disease from multivisceral involvement unresponsive to treat- also occur in PAN and MPA. Lowering doses and shortening
ment (Bourgarit et al. 2005). Death most often occurs accompanied treatment duration will probably lead to fewer infectious compli-
by fever, rapid weight loss, and one or several major organ involve- cations, but treatment intensity and duration need to be further
ments. For PAN, deaths are often the consequence of GI involve- refined, based on prospective studies.
ment. The major causes of death of PAN patients are summarized As in other vasculitides, a small number of patients died within the
in Table 25.7. first weeks or months following PAN diagnosis, despite institution
While the deaths occurring during the first few months of the of adequate treatment. Bourgarit et al. (2005) showed that 38/309
disease are often caused by uncontrolled vasculitis, those occur- (12%) patients died during the first year, predominantly from vascu-
ring during the following years may be the consequence of treat- litis (58%), with the other causes being treatment side-effects inde-
ment side-effects and are not rare. These adverse events emphasize pendent of vasculitis. The primary cause of early death in PAN is
the importance of tailoring the therapeutic regimen after care- severe GI involvement, with perforations or haemorrhage, included
ful analysis of parameters available to predict the outcome (see in the Five Factor Score (FFS) (Guillevin et al. 1996). The presence of
below). Infections represent the primary cause of death and are HBV infection was not identified as a factor of severity.
favoured by CS and/or cytotoxic agents. Septicaemia occurring When the patients were hospitalized in intensive care units, the
in the first months of treatment is the consequence of the intense main prognostic factor for early deaths was the APACHE (Acute
initial therapy with CS and cytotoxic agents. Viral infections usu- Physiology and Chronic Health Evaluation) score, which was not
ally occur later because of the profound immunosuppression specifically devised for vasculitis. In contrast, the specific scores
induced by drugs prescribed to control the vasculitis. Rare cases for vasculitides, for example Birmingham Vasculitis Activity Score
of Pneumocystis jiroveci pneumonia, which have mainly been (BVAS) (Luqmani et al. 1994) and FFS, did not correlate with death
described in GPA (Jarrousse et al. 1993; Godeau et al. 1995), can in intensive care units (Cruz et al. 2003).

Table 25.7 Causes of deaths of patients with PAN, HBV-related or not

Number of patients Gayraud Cruz Cacoub Leib Cohen Guillevin


Type of vasculitis (n) et al. 2001 et al. 2003 et al. 1988 et al. 1979 et al. 1980 et al. 2005
   278 26 165 64 53 115
  HBV-PAN (63), PAN Patients admitted PAN PAN PAN HBV-related PAN
(93), MPA (58), to ICUa, PAN (5), MPA
and CSS (64) (4), CSS (7), and other (10)
Deaths (%) 85 (31) 2/5 PAN (20) 61 (37)b 32 (50) 22 (42) 41 (36)
Cause of death (%)            
Progressive vasculitis 22 (26) 0 21 (43)b 20 (63) – 11 (27)
Bowel infarction 10 – 8 – 6 (27) 7
Cardiac failure 5 – 4 – – 1
Multivisceral 3 – 2 – –
involvement
Renal failure 3 – 5 – 6 (27)
Stroke 1 – 2 – –
Infectious side-effects 11 (13) 2 (100) 13 (27) 2 (6) - 4 (10)
of treatment
Bacterial pneumonia 5 1 11 – – –
Septicaemia 6 1 6/11 – – 3/4
Sudden deaths 9 (11) 0 3 (6) – – 1 (2)
Heart disease 8 (9) 0 6(12)b 5 (16) 8 (36)c 5 (12)c
Cancer 13 (15) 0 5 (10) 1 (3) 1 (5) 3 (7)
Pulmonary embolism 3 (3.5) 0 2 (4) – –
Chronic respiratory disease 3 (3.5) 0 – – –
End-stage renal disease – – – 3 (9) –
Fulminant viral hepatitis 2 (2) 0 – – – 1 (2)
  Miscellaneous 14 (17) 0 2 (4) 1 (3) 1 (5) 16 (39)
a ICU, intensive care unit.
b Some patients may have died, e.g. from heart disease due to vasculitis alone but also to treatments or other associated conditions, not always specified, so that total of listed causes
is >100%.
c Includes cerebrovascular and cardiovascular complications.
344 SECTION 5 vasculitic diseases and syndromes and related disorders

Treatment of PAN In the presence of angina or cardiac insufficiency, echocardi-


ography, cardiac catheterization, and coronary angiography can
Severity of vasculitis document PAN involvement and detect co-morbid conditions, for
The FFS has significant prognostic value and can guide the phy- example atherosclerosis, which can be treated.
sicians’ choices of therapeutic agents and avoid overtreatment A vasculitis damage index (VDI) (Exley et  al. 1997)  has been
(Guillevin et  al. 1996). The following parameters are associated devised to measure the cumulative impact of scars and sequelae
with higher mortality:  proteinuria >1 g/day, renal insufficiency caused by the disease and its therapy.
(creatininaemia >140 µmol/l or 1.6 mg/dl), specific cardiomyopa-
thy, GI manifestations and CNS involvement. In a study of 342 How should PAN be treated?
patients with PAN and EGPA, when FFS equalled 0, 1, or 2, mor- Nearly all the reports on the treatment and outcome of PAN series
tality at 5 years was respectively 12%, 26%, or 46%. Although it has included patients with MPA (Frohnert and Sheps 1967; Leib et al.
not yet been demonstrated that treatment should be chosen based 1979; Cohen et al. 1980), and sometimes EGPA as well (Guillevin
on these criteria, they should probably be considered in deciding et al. 1999); thus their results concerned at least two distinct enti-
the therapeutic strategy. For PAN forms without symptoms of poor ties. Today, treatments for MPA and GPA are considered together
prognosis (FFS=0), we treat patients with CS alone to lower the (Rasmussen et al. 1995), based on their shared pathogenic mecha-
number of side-effects. This strategy is effective, with a few minor nisms, such as ANCA positivity, but it seems more reasonable to
relapses necessitating transient intensification of the CS dose or consider the therapeutic strategy for PAN independently from that
addition of an immunosuppressant agent (personal observations). of MPA.
The 1996 FFS for systemic necrotizing vasculitides was revisited, Since the initial use of CS in 1950 to treat PAN (Bagenstoss et al.
including GPA (Guillevin et al. 2011). The following factors were 1950), their use has increased the 5-year survival rate from 10% for
significantly associated with higher 5-year mortality: age >65 years, untreated patients to about 55% in the mid-to-late 1970s (Frohnert
cardiac symptoms, GI involvement, and renal insufficiency (stabi- and Sheps 1967; Leib et al. 1979; Cohen et al. 1980). Survival was
lized peak creatinine ≥150 μmol/l or 1.7 mg/dl). The presence of further prolonged by adding immunosuppressants, either azathi-
each was accorded +1 point. Because ear, nose, and throat symp- oprine or cyclophosphamide (CYC) (Fauci et al. 1979; Leib et al.
toms are associated with a better prognosis, their absence is score 1979), to the treatment regimen, attaining a 5-year survival rate of
+1 point. Respective 5-year mortality rates for FFS of 0, 1, or ≥2 82% for patients given CS and CYC (Leib et al. 1979). Those rates
were 9%, 21%, or 40%. The same strategy as previously described were obtained in retrospective studies and have not always been
for the 1996 FFS is now applied for PAN patients. confirmed by prospective studies (Cohen et al. 1980). Fauci et al.
Other criteria to determine the intensity of treatment, such as (1979), who advocated the use of CYC, found that the mean dura-
the BVAS (Luqmani et al. 1994), are being tested in the prospective tion of CS treatment at the time of entry into the study had been
trials proposed by the European Vasculitis Study Group (EUVAS). 22 (range 2–48) months. Studies that included such patients with
longstanding vasculitis might have introduced some bias, in that
most deaths directly attributable to uncontrolled vasculitis (Fauci
Recommendations for specific conditions et al. 1979) usually occurred within the first 6 months of treatment.
Supportive care represents an important part of the therapeu-
tic regimen for patients with potentially fatal vasculitis flare. Corticosteroids
Since maximal immunosuppression is given at the beginning Corticosteroids (CS) are given to all patients with PAN. In the case
of treatment, prevention of opportunistic infections, such as of HBV-related PAN, CS should be administered for only a few days
Pneumocystis jiroveci pneumonia, may be necessary and prophy- (see below in this section). For the other PAN forms, treatment is
laxis should be prescribed on an individual basis (Jarrousse et al. prolonged, lasting around 12 months. High doses may be useful
1993; Godeau et al. 1995). Pain control, prevention of pressure initially. The administration of methylprednisolone pulses (usually
sores, and physical therapy are needed for mononeuritis multi- 7.5–15 mg/kg i.v. over 60 min repeated at 24-h intervals for 1–3 days)
plex. Introduction of angiotensin-converting-enzyme inhibitors has become widely used at the beginning of therapy for severe sys-
is effective against severe hypertension caused by renal vascu- temic vasculitis, especially in the presence of life-threatening organ
litis and has a beneficial effect on renal function. Fulminating involvement or the extension phase of mononeuropathy multiplex.
vasculitis usually presents as GI involvement, renal failure, This regimen has a rapid onset of action and is relatively safe. The
pulmonary haemorrhage, and/or, more rarely, cerebral involve- dose of pulse methylprednisolone is empirical and doses under
ment. Some patients do not respond to treatment and die within 1000 mg may be as effective. Side-effects of pulse methylpredni-
a few months after diagnosis. solone are usually mild and transient and include a bitter taste,
When GI involvement causes persistent abdominal pain despite facial flushing, headache, asthenia, marked blood pressure rise, and
medical treatment, an exploratory laparotomy should be per- temporary glucose intolerance. Severe side-effects, fortunately rare,
formed to identify and treat possible bowel perforations. For the include sudden death, cardiac arrhythmia, myocardial infarction,
same group of patients, it seems reasonable to administer drugs GI bleeding, and seizure. Oral CS are given at the dose of 1 mg/kg/
intravenously to circumvent impaired drug absorption. Rapid and day of prednisone or its equivalent of methylprednisolone. Therapy
severe weight loss due to severe GI involvement must be countered should be consolidated into a single morning dose. As the patient’s
with parenteral nutrition. Despite the fact that weight loss has not clinical status improves and the biological markers of inflammation
been demonstrated to be a factor of poor prognosis (Guillevin et al. (C-reactive protein, erythrocyte sedimentation rate) return to nor-
1996), good general condition is always preferable, because it can mal, usually within 3 weeks to 1 month, tapering of the prednisone
also help reduce the infection rate under cytotoxic agents. dose can begin.
CHAPTER 25 polyarteritis nodosa 345

In the therapeutic trials conducted by the French Vasculitis Study In our opinion, CYC should not be prescribed systematically as
Group (Guillevin et al. 1992b), the timetable for prednisone taper- first-line treatment to all PAN patients and the management deci-
ing was long and drawn out. The daily dose was decreased by 2.5 sion must consider the anatomical location of the involvement and
mg every 10 days for 1 month, and then by 2.5 mg every week until its severity. When patients fail to respond to pulse CYC or when
a level equivalent to half the initial dose was reached. This dose relapse occurred with the first 6  months of treatment, oral CYC
was maintained for 3 weeks and then further decreased by 2.5 mg has been successfully used to control disease activity (Généreau
every week, to approximately 20 mg/day. A  more careful taper- et al. 1994).
ing schedule was followed for doses below 20 mg/day. The daily
dose was decreased by 1 mg every 1–2 weeks, to a dose of 10 mg/ Other cytotoxic agents
day. This level was maintained for 3 weeks and was then further Azathioprine, methotrexate, and several other cytotoxic agents
decreased by 1 mg every month until it was completely withdrawn. have been tried in PAN patients. They are reserved for patients with
This regimen is effective and may control the disease without the contraindications to CYC, or as maintenance therapy for patients
addition of cytotoxic agents. When prednisone is combined with with the more severe forms after stopping CYC, and for a recom-
CYC, the prednisone dose should be tapered more rapidly to avoid mended duration of 12–18 months.
complications. The efficacy and safety of azathioprine versus pulse CYC as
adjunctive immunosuppressive therapy for PAN or MPA patients
Cyclophosphamide without poor-prognosis factors and experiencing treatment failure
CYC is prescribed at a dose of 2 mg/kg/day or less for 1 year and, or relapse after initial therapy with CS alone were assessed (Ribi
in combination with CS, represented the conventional treatment of et al. 2010). Patients were randomized to receive 6 months of ther-
systemic necrotizing vasculitides. Although this regimen is effective, apy with oral azathioprine or six CYC pulses. In that study, first-line
it has a low therapeutic/ toxic index. Side-effects have been exten- CS were able to achieve and maintain remission in only about half
sively described in GPA patients, but are not disease-specific. Major of the patients, and 40% of the patients required additional immu-
side-effects associated with daily CYC administration include haem- nosuppressive therapy. At the time of treatment failure or disease
orrhagic cystitis, bladder fibrosis, bone-marrow suppression, ovarian relapse, azathioprine or CYC pulse was fairly effective for treating
failure, and cancer (bladder cancer and haematological malignan- CS-resistant disease or major relapses.
cies) (Stillwell et al. 1988; Hoffman et al. 1992; Sneller 2000). Severe
infections represent a major cause of mortality of patients with sys- Plasma exchanges
temic necrotizing vasculitis, especially while they are receiving high To date, no argument supports the systematic prescription of plasma
CS doses with adjunctive immunosuppressive drugs (Gayraud et al. exchanges at the time of diagnosis of PAN without HBV infection
1997; Gayraud et al. 2001; Bourgarit et al. 2005). (Guillevin et al. 1992a), even for patients with poor-prognosis fac-
In an attempt to limit the morbidity associated with daily CYC tors (Guillevin et al. 1995d). Plasma exchanges might be useful as
administration, protocols using intermittent treatment with higher second-line treatment of PAN refractory to conventional therapy
doses have been developed (Gayraud et al. 1997; Guillevin et al. and might be able to limit disease sequelae, as shown for severe
2003). Pulse CYC is now being used increasingly to treat systemic renal disease in ANCA-associated vasculitides.
necrotizing vasculitides and might be preferred to oral CYC. The
CYC content of each pulse, and both the total number and frequency
Treatment duration
of the pulses, has to be adjusted according to the patient’s condition, Because of its low relapse rate, PAN may be treated for a shorter
renal function, haematological data, and the disease’s response to time than other systemic necrotizing vasculitides. Indeed, treat-
previous therapies, including previous CYC pulses. High-dose i.v. ment often cures PAN and no maintenance regimen is needed.
CYC may be dangerous in patients with renal insufficiency, sug- For PAN patients with poor-prognosis factors, CS and CYC are
gesting that dose reduction according to renal function is prudent. given for about 12 months. Alternatively, CS and CYC may be used
Intense hydration is recommended during pulse therapy com- together for 6 months, followed by CS and a less toxic drug, for
bined with administration of sodium 2-mercaptoethanesulfonate example azathioprine or methotrexate.
(mesna), as for oral CYC. However, pulse CYC therapy allows a Ongoing trials are comparing doses and intensities of treatment
lower cumulative dose to be given and exposes the patient to less with conventional drugs, and evaluating the efficacy of autologous
potential toxicity for shorter periods than oral CYC. In the proto- bone-marrow transplantation (Bacon et al. 1998).
cols of the French Vasculitis Study Group, the CYC-pulse dose was
0.6 g/m2 given once monthly. When the results of six CYC pulses
Therapeutic specificities
were compared to 12 pulses administered every 2 weeks for the HBV-Related PAN
three first pulses, then every 4 weeks until reaching the number of For HBV-related PAN, conventional treatment with CS and CYC
pulses scheduled, with no subsequent maintenance treatment for allows the virus to replicate, thereby facilitating evolution towards
PAN patients with FFS ≥ 1, the event-free survival rate was much chronic hepatitis and liver cirrhosis. Thus, CYC and prolonged CS
higher in the 12-pulse group than in the six-pulse group (68% ver- are contraindicated. The preferred initial treatment approach is to
sus 19%, respectively) (Guillevin et  al. 2003). It may be feasible combine plasma exchanges and an antiviral drug with CS to rap-
to treat severe PAN patients with CYC until remission has been idly control the most severe life-threatening PAN manifestations,
achieved and then to maintain remission with another immuno- which are common during the first weeks of the disease. CS are
suppressant (such as azathioprine) for 12 months, as reported for then abruptly discontinued to enhance immunological clearance of
systemic GPA (Jayne et al. 2003), but this approach has not been HBV-infected hepatocytes and favour seroconversion from a posi-
validated in a prospective trial on PAN. tive HBeAg to a positive anti-HBeAb.
346 SECTION 5 vasculitic diseases and syndromes and related disorders

The combination of antiviral agents (vidarabine or interferon Bourgarit, A., Le Toumelin, P., Pagnoux, C., et  al. (2005). Deaths occur-
alpha-2b) gave excellent overall therapeutic results (Guillevin ring during the first year after treatment onset for polyarteritis nodosa,
et al. 1993a; Guillevin et al. 1994) and should be preferred to con- microscopic polyangiitis or Churg–Strauss syndrome. A  retrospec-
tive analysis of causes and factors predictive of mortality based on 595
ventional regimens that jeopardize the outcome. The efficacy of
patients. Medicine (Baltimore), 84, 323–30.
this strategy was confirmed in a series of 41 patients: 23 (56.1%) Bron, K., Strott, C., and Shapiro, A. (1965). The diagnostic value of
patients no longer exhibit serological evidence of replication and angiographic observations in polyarteritis nodosa. A case of multi-
80.5% were cured (Guillevin et  al. 1995b). New antiviral agents, ple aneurysms in the visceral organs. Archives of Internal Medicine,
such as lamivudine, have been shown to increase the seroconver- 116, 454–9.
sion rate up to 60% (Guillevin et al. 2004; Guillevin et al. 2005), Cacoub, P., Le Thi Huong, D., Guillevin, L., and Godeau, P. (1988). Causes of
even if they were not able to improve the survival rate at 18 months death in systemic vasculitis of polyarteritis nodosa. Analysis of a series
when compared to conventional CS with or without CYC and with- of 165 patients [in French]. Annales de Médecine Interne (Paris), 139,
381–90.
out antiviral drugs (mortality rate at 18 months, 27.5% vs. 17.5%
Calabrese, L.H. (1991). Vasculitis and infection with the human immuno-
with combined antiviral therapy, P=0.46). The benefits of adjunc-
deficiency virus. Rheumatology Disease Clinics of North America, 17,
tive antiviral therapy probably appear after prolonged follow-up, 131–47.
since these drugs lower the risk of cirrhosis or liver failure. Camilleri, M., Pusey, C., Chadwick, V., and Rees, A. (1983). Gastrointestinal
Localized forms manifestations of systemic vasculitis. QJM, 52, 141–9.
Casserly, L.F., Reddy, S.M., Rennke, H.G., Carpinito, G.A., and Levine, J.S.
As for PAN without poor-prognosis factors, localized forms of (1999). Reversible bilateral hydronephrosis without obstruction in
PAN can be treated initially without immunosuppressants, holding hepatitis B-associated polyarteritis nodosa. American Journal of Kidney
the latter in reserve for refractory or relapsing disease. It should be Diseases, 34, e11.
noted that isolated appendix or gallbladder involvement has a good Castaigne, P., Cambier, J., Escourolle, R., et  al. (1970). Les manifestations
prognosis—after surgical removal. Cutaneous PAN, particularly nerveuses centrales de la périartérite noueuse. A propos d’une observa-
in children, may be successfully treated with colchicine or disulone tion anatomo-clinique [in French]. Annales de Médecine Interne (Paris),
(dapsone), often in combination with topical CS. Similarly, colchicine 121, 375–6.
or dapsone may be effective against PAN restricted to skeletal muscles. Chanseaud, Y., Tamby, M.C., Guilpain, P., et al. (2005). Analysis of autoan-
tibody repertoires in small- and medium-sized vessel vasculitides.
Evidence for specific perturbations in polyarteritis nodosa, microscopic
References polyangiitis, Churg–Strauss syndrome and Wegener’s granulomatosis.
Abgrall, S., Mouthon, L., Cohen, P., et  al. (2001). Localized neurological Journal of Autoimmunity, 24, 169–79.
necrotizing vasculitides. Three cases with isolated mononeuritis multi- Chen, K.T. (1989). Gallbladder vasculitis. Journal of Clinical Gastroenterology,
plex. Journal of Rheumatology, 28, 631–3. 11, 537–40.
Abu-Farsakh, H., Mody, D., Brown, R.W., and Truong, L.D. (1994). Isolated Cid, M. (1996). New developments in the pathogenesis of systemic vasculitis.
vasculitis involving the female genital tract: clinicopathologic spectrum Current Opinion in Rheumatology, 9, 1–11.
and phenotyping of inflammatory cells. Modern Pathology, 7, 610–5. Cohen, L., Guillevin, L., Meyrier, A., Bironne, P., Blétry, O., and Godeau,
Akcicek, F., Dilber, S., Ozgen, G., et al. (1994). Spontaneous perirenal hema- P. (1986). Malignant arterial hypertension in periarteritis nodosa.
toma due to periarteritis nodosa. Nephron, 68, 396. Incidence, clinicobiologic parameters and prognosis based on a series of
Akova, Y.A., Jabbur, N.S., and Foster, C.S. (1993). Ocular presentation of 165 cases [in French]. Archives des Maladies du Coeur et des Vaisseaux,
polyarteritis nodosa. Clinical course and management with steroid and 79, 773–8.
cytotoxic therapy. Ophthalmology, 100, 1775–81. Cohen, R.D., Conn, D.L., and Ilstrup, D.M. (1980). Clinical features, progno-
Allan, S., Towla, H., Smith, C., et al.(1982). Painful brachial plexopathy: an sis, and response to treatment in polyarteritis. Mayo Clinic Proceedings,
unusual presentation of polyarteritis nodosa. Postgraduate Medical 55, 146–55.
Journal, 58, 311–3. Colmegna, I. and Maldonado-Cocco, J.A. (2005). Polyarteritis nodosa revis-
Amarenco, P., Amarenco, G., Guillevin, L., et al. (1988). Neuropathie vésicale ited. Current Rheumatology Reports, 7, 288–96.
au cours des vascularites systémiques [in French]. Annales de Médecine Corman, L.C. and Dolson, D.J. (1992). Polyarteritis nodosa and parvovirus
Interne (Paris), 139, 183–5. B19 infection. Lancet, 339, 491.
Bacon, P., Exley, A., Carruthers, D., et  al. (1998). Immune ablation with Cornfield, J.Z., Johnson, M.L., Dolehide, J., and Fowler, J.J. (1988). Massive
peripheral stem cell rescue in Wegener’s: systemic vasculitis. Clinical and renal hemorrhage owing to polyarteritis nodosa. Journal of Urology,
Experimental Immunology, 112, S156. 140, 808–9.
Bagenstoss, A., Shick, R., and Polley, H. (1950). The effect of cortisone on the Cowpe, J. and Hislop, W. (1983). Oral presentation of polyarteritis nodosa.
lesions of periarteritis nodosa. American Journal of Pathology, 26, 709. Oral Surgery, 56, 597.
Blétry, O., Godeau, P., Charpentier, G., et al. (1980). Manifestations cardi- Cruz, B.A., Ramanoelina, J., Mahr, A., et al. (2003). Prognosis and outcome of
aques de la périartérite noueuse. Fréquence de la cardiomyopathie 26 patients with systemic necrotizing vasculitis admitted to the intensive
non hypertensive [in French]. Archives des Maladies du Coeur et des care unit. Rheumatology (Oxford), 42, 1183–8.
Vaisseaux, 73, 1027–35. Darras-Joly, C., Lortholary, O., Cohen, P., Brauner, M., and Guillevin, L.
Blidi, M., Quang, T.N., Cassan, P., and Guillevin, L. (1996). Cholécystites (1995). Regressing microaneurysms in 5 cases of hepatitis B virus-related
aigues de la périartérite noueuse. Huit observations [in French]. Annales polyarteritis nodosa. Journal of Rheumatology, 22, 876–80.
de Médecine Interne (Paris), 147, 304–12. Diaz-Perez, J.L. and Winkelmann, R.K. (1974). Cutaneous periarteritis
Bookman, A., Goode, E., McLoughlin, M., and Cohen, Z. (1983). Polyarteritis nodosa. Archives of Dermatology, 110, 407–14.
nodosa complicated by a ruptured intrahepatic aneurysm. Arthritis and Dugué, C., Renaudineau, Y., Jamin, C., and Youinou, P. (2004). Diagnostic
Rheumatism, 26, 106–8. and pathogenic implications of the heterogeneity of antiendothelial cell
Bouche, P., Léger, J.M., Travers, M.A., Cathala, H.P., and Castaigne, P. (1986). antibodies. Autoimmunity Reviews, 3 (Suppl. 1), 11–3.
Peripheral neuropathy in systemic vasculitis: clinical and electrophysi- Dyck, P., Benstead, T., Conn, D., Stevens, J., and Windebandk, A. (1987). Non
ologic study of 22 patients. Neurology, 36, 1598–602. systemic vasculitic neuropathy. Brain, 110, 843–54.
CHAPTER 25 polyarteritis nodosa 347

Elkon, K.B., Hughes, G.R., Catovsky, D., et al. (1979). Hairy-cell leukaemia Godeau, B., Coutant-Perronne, V., Le Thi Huong, D., et  al. (1994).
with polyarteritis nodosa. Lancet, 2, 280–2. Pneumocystis carinii pneumonia in the course of connective tissue dis-
Esteva-Lorenzo, F.J., Ferreiro, J.L., Tardaguila, F., de la Fuente, A., Falasca, G., ease: report of 34 cases. Journal of Rheumatology, 21, 246–51.
and Reginato, A.J. (1994). Case report 866. Pseudotumor of the muscle Godeau, B., Mainardi, J.L., Roudot, T.F., et al. (1995). Factors associated with
associated with necrotizing vasculitis of medium- and small-sized arter- Pneumocystis carinii pneumonia in Wegener’s granulomatosis. Annals
ies and chronic myositis. Skeletal Radiology, 23, 572–6. of the Rheumatic Diseases, 54, 991–4.
Ewald, E.A., Griffin, D., and McCune, W.J. (1987). Correlation of angio- Godeau, P., Guillevin, L., Tucat, G., et al. (1979). Périartérite noueuse avec
graphic abnormalities with disease manifestations and disease severity pancréatite aiguë. Deux nouvelles observations [in French]. Lyon
in polyarteritis nodosa. Journal of Rheumatology, 14, 952–6. Médical, 242, 279–81.
Exley, A., Carruthers, D., Luqmani, R., et al. (1997). Damage occurs early in Gourgoutis, G.D., Paguirigan, A.A., and Berzins, T. (1971). Gastric perfora-
systemic vasculitis and is an index of outcome. QJM, 90, 391–9. tion in polyarteritis nodosa. Report of a patient. American Journal of
Fauci, A.S., Haynes, B., and Katz, P. (1978). The spectrum of vasculitis: clini- Digestive Diseases, 16, 171–7.
cal, pathologic, immunologic and therapeutic considerations. Annals of Grasland, A., Pouchot, J., Damade, R., and Vinceneux, P. (1996). Uterine
Internal Medicine, 89, 660–76. localization of periarteritis nodosa disclosed by fever of long duration
Fauci, A.S., Katz, P., Haynes, B.F., and Wolff, S.M. (1979). Cyclophosphamide [in French]. Revue de Médecine Interne, 17, 58–60.
therapy of severe systemic necrotizing vasculitis. New England Journal of Grau, G.E., Roux, L.P., Gysler, C., et al. (1989). Serum cytokine changes in
Medicine, 301, 235–8. systemic vasculitis. Immunology, 68, 196–8.
Ford, R. and Sieckert, R. (1965). Central nervous system manifestations of Guillevin, L., Cohen, P., Gayraud, M., Lhote, F., Jarrousse, B., and Casassus,
periarteritis nodosa. Neurology, 15, 114–22. P. (1999). Churg–Strauss syndrome. Clinical study and long-term
Fortin, P.R., Larson, M.G., Watters, A.K., Yeadon, C.A., Choquette, D., and follow-up of 96 patients. Medicine (Baltimore), 78, 26–37.
Esdaile, J.M. (1995). Prognostic factors in systemic necrotizing vascu- Guillevin, L., Cohen, P., Mahr, A., et al. (2003). Treatment of polyarteritis
litis of the polyarteritis nodosa group: a review of 45 cases. Journal of nodosa and microscopic polyangiitis with poor prognosis factors: a pro-
Rheumatology, 22, 78–84. spective trial comparing glucocorticoids and six or twelve cyclophos-
Frohnert, P. and Sheps, S. (1967). Long term follow-up study of periarteritis phamide pulses in sixty-five patients. Arthritis and Rheumatism, 49,
nodosa. American Journal of Medicine, 43, 8–14. 93–100.
Fye, K., Becker, M., Theofilopoulos, A., Moutsopoulos, H., Feldman, J., and Guillevin, L., Fain, O., Lhote, F., et al. (1992a). Lack of superiority of steroids
Talal, N. (1977). Immune complexes in hepatitis B antigen-associated per- plus plasma exchange to steroids alone in the treatment of polyarteritis
iarteritis nodosa: detection by antibody independent cell-mediated cyto- nodosa and Churg–Strauss syndrome. A prospective, randomized trial
toxicity and the Raji cell assay. American Journal of Medicine, 62, 783–91. in 78 patients. Arthritis and Rheumatism, 35, 208–15.
Gallien, S., Mahr, A., Réty, F., et al. (2002). Magnetic resonance imaging of Guillevin, L., Le Thi Huong, D., Godeau, P., Jais, P., and Wechsler, B. (1988).
skeletal muscle involvement in limb restricted vasculitis. Annals of the Clinical findings and prognosis of polyarteritis nodosa and Churg–
Rheumatic Diseases, 61, 1107–9. Strauss angiitis: a study in 165 patients. British Journal of Rheumatology,
Ganesan, R., Ferryman, S.R., Meier, L., and Rollason, T.P. (2000). Vasculitis of 27, 258–64.
the female genital tract with clinicopathologic correlation: a study of 46 Guillevin, L., Lhote, F., Brauner, M., and Casassus, P. (1995a). Antineutrophil
cases with follow-up. International Journal of Gynecological Pathology, cytoplasmic antibodies (ANCA) and abnormal angiograms in polyarte-
19, 258–65. ritis nodosa and Churg–Strauss syndrome: indications for the diagnosis
Gargollo, P.C., Barnewolt, C.E., and Diamond, D.A. (2004). Calcified ureteral of microscopic polyangiitis. Annales de Médecine Interne (Paris), 146,
stricture in a child with polyarteritis nodosa. Journal of Urology, 171, 548–50.
1254–5. Guillevin, L., Lhote, F., Cohen, P., et al. (1995b). Polyarteritis nodosa related
Gaskin, G., Savage, C., Ryan, J., et al. (1991). Antineutrophil cytoplasmic anti- to hepatitis B virus. A prospective study with long-term observation of
bodies and disease activity during long-term follow-up of 70 patients 41 patients. Medicine (Baltimore), 74, 238–53.
with systemic vasculitis. Nephrology Dialysis Transplantation, 6, 689–94. Guillevin, L., Lhote, F., Cohen, P., et al. (1995d). Corticosteroids plus pulse
Gayraud, M., Guillevin, L., Cohen, P., et  al. (1997). Treatment of cyclophosphamide and plasma exchanges versus corticosteroids
good-prognosis polyarteritis nodosa and Churg–Strauss syn- plus pulse cyclophosphamide alone in the treatment of polyarteritis
drome: comparison of steroids and oral or pulse cyclophosphamide in nodosa and Churg–Strauss syndrome patients with factors predicting
25 patients. French Cooperative Study Group for Vasculitides. British poor prognosis. A prospective, randomized trial in sixty-two patients.
Journal of Rheumatology, 36, 1290–7. Arthritis and Rheumatism, 38, 1638–45.
Gayraud, M., Guillevin, L., le Toumelin, P., et al. (2001). Long-term follow-up Guillevin, L., Lhote, F., Gallais, V., et  al. (1995c). Gastrointestinal tract
of polyarteritis nodosa, microscopic polyangiitis, and Churg–Strauss involvement in polyarteritis nodosa and Churg- Strauss syndrome.
syndrome:  analysis of four prospective trials including 278 patients. Annales de Médecine Interne (Paris), 146, 260–7.
Arthritis and Rheumatism, 44, 666–75. Guillevin, L., Lhote, F., Gayraud, M., et al. (1996). Prognostic factors in pol-
Généreau, T., Lortholary, O., Leclerq, P., et al. (1994). Treatment of systemic yarteritis nodosa and Churg–Strauss syndrome. A prospective study in
vasculitis with cyclophosphamide and steroids: daily oral low-dose cyclo- 342 patients. Medicine (Baltimore), 75, 17–28.
phosphamide administration after failure of a pulse intravenous high-dose Guillevin, L., Lhote, F., Jarrousse, B., and Fain, O. (1992b). Treatment of
regimen in four patients. British Journal of Rheumatology, 33, 959–62. polyarteritis nodosa and Churg–Strauss syndrome. A  meta- analysis
Généreau, T., Lortholary, O., Pottier, M.A., et  al. (1999). Temporal artery of 3 prospective controlled trials including 182 patients over 12 years.
biopsy:  a diagnostic tool for systemic necrotizing vasculitis. French Annales de Médecine Interne (Paris), 143, 405–16.
Vasculitis Study Group. Arthritis and Rheumatism, 42, 2674–81. Guillevin, L., Lhote, F., Leon, A., Fauvelle, F., Vivitski, L., and Trépo, C.
Gherardi, R., Belec, L., Mhiri, C., et al. (1993). The spectrum of vasculitis in (1993a). Treatment of polyarteritis nodosa related to hepatitis B
human immunodeficiency virus-infected patients. A clinicopathologic virus with short term steroid therapy associated with antiviral agents
evaluation. Arthritis and Rheumatism, 36, 1164–74. and plasma exchanges. A  prospective trial in 33 patients. Journal of
Gisselbrecht, M., Cohen, P., Lortholary, O., et al. (1998). Human immunode- Rheumatology, 20, 289–98.
ficiency virus-related vasculitis. Clinical presentation of and therapeutic Guillevin, L., Lhote, F., Sauvaget, F., et al. (1994). Treatment of polyarteri-
approach to eight cases [in French]. Annales de Médecine Interne (Paris), tis nodosa related to hepatitis B virus with interferon-alpha and plasma
149, 398–405. exchanges. Annals of the Rheumatic Diseases, 53, 334–7.
348 SECTION 5 vasculitic diseases and syndromes and related disorders

Guillevin, L., Mahr, A., Callard, P., et al. (2005). Hepatitis B virus-associated Iino, T., Eguchi, K., Sakai, M., Nagataki, S., Ishijima, M., and Toriyama, K.
polyarteritis nodosa:  clinical characteristics, outcome, and impact of (1992). Polyarteritis nodosa with aortic dissection: necrotizing vasculitis
treatment in 115 patients. Medicine (Baltimore), 84, 313–22. of the vasa vasorum. Journal of Rheumatology, 19, 1632–6.
Guillevin, L., Mahr, A., Cohen, P., et al. (2004). Short-term corticosteroids Ito, M., Sano, K., Inaba, H., and Hotchi, M. (1991). Localized necrotizing
then lamivudine and plasma exchanges to treat hepatitis B virus-related arteritis. A report of two cases involving the gallbladder and pancreas.
polyarteritis nodosa. Arthritis and Rheumatism, 51, 482–7. Archives of Pathology and Laboratory Medicine, 115, 780–3.
Guillevin, L., Pagnoux, C., Seror, R., et  al. (2011). The Five-Factor Score Jamieson, P., Giuliani, M., and Martinez, A. (1991). Necrotizing angi-
revisited: assessment of prognoses of systemic necrotizing vasculitides opathy presenting with multifocal conduction blocks. Neurology,
based on the French Vasculitis Study Group (FVSG) cohort. Medicine 41, 442–4.
(Baltimore), 90, 19–27. Jamin, C., Dugué, C., Alard, J.E., et al. (2005). Induction of endothelial cell
Guillevin, L., Ronco, P., and Verroust, P. (1990). Circulating immune com- apoptosis by the binding of anti-endothelial cell antibodies to Hsp60
plexes in systemic necrotizing vasculitis of the polyarteritis nodosa in vasculitis-associated systemic autoimmune diseases. Arthritis and
group. Comparison of HBV-related polyarteritis nodosa and Churg Rheumatism, 52, 4028–38.
Strauss Angiitis. Journal of Autoimmunity, 3, 789–92. Jarrousse, B., Guillevin, L., Bindi, P., et  al. (1993). Increased risk of
Guillevin, L., Visser, H., Noël, L.H., et al. (1993b). Antineutrophil cytoplasm Pneumocystis carinii pneumonia in patients with Wegener’s granulo-
antibodies in systemic polyarteritis nodosa with and without hepatitis matosis. Clinical and Experimental Rheumatology, 11, 615–21.
B virus infection and Churg–Strauss syndrome- -62 patients. Journal of Jayne, D., Rasmussen, N., Andrassy, K., Bacon, P., et  al. (2003). A rand-
Rheumatology, 20, 1345–9. omized trial of maintenance therapy for vasculitis-associated with anti-
Gunal, N., Kara, N., Cakar, N., Kocak, H., Kahramanyol, O., and Cetinkaya, neutrophil cytoplasmic antibodies. New England Journal of Medicine,
E. (1997). Cardiac involvement in childhood polyarteritis nodosa. 349, 36–44.
International Journal of Cardiology, 60, 257–62. Jennette, J.C., Falk, R.J., Andrassy, K., et  al. (1994). Nomenclature of sys-
Ha, H.K., Lee, S.H., Rha, S.E., et al. (2000). Radiologic features of vasculitis temic vasculitides. Proposal of an international consensus conference.
involving the gastrointestinal tract. Radiographics, 20, 779–94. Arthritis and Rheumatism, 37, 187–92.
Hachulla, E., Bourdon, F., Taieb, S., et al. (1993). Embolization of two bleed- Jennette J.C., Falk R.J., Bacon P.A., et al. (2013). 2012 revised International
ing aneurysms with platinum coils in a patient with polyarteritis nodosa. Chapel Hill Consensus Conference Nomenclature of Vasculitides.
Journal of Rheumatology, 20, 158–61. Arthritis Rheum, 65, 1–11.
Hamidou, M.A., Moreau, A., Toquet, C., El Kouri, D., de Faucal, P., and Falk, R.J., Bacon, P.A., Basu, N., et al. (2013). 2012 Revised International
Grolleau, J.Y. (2003). Temporal arteritis associated with systemic Chapel Hill Consensus Conference Nomenclature of Vasculitides.
necrotizing vasculitis. Journal of Rheumatology, 30, 2165–9. Arthritis and Rheumatism, 65, 1–11.
Hasler, P., Kistler, H., and Gerber, H. (1995). Vasculitides in hairy cell leuke- Kaya, E., Utas, C., Balkanli, S., Basbug, M., and Onursever, A. (1994).
mia. Seminars in Arthritis and Rheumatism, 25, 134–42. Isolated ovarian polyarteritis nodosa. Acta Obstetricia et Gynecologica
Haugeberg, G., Bie, R., and Johnsen, V. (1997). Vasculitic changes in the Scandinavica, 73, 736–8.
temporal artery in polyarteritis nodosa. Scandinavian Journal of Kinyoun, J., Kalina, R., and Klein, M. (1987). Choroidal involvement in sys-
Rheumatology, 26, 383–5. temic necrotizing vasculitis. Archives d'Ophtalmologie, 105, 939.
Hauschild, S., Csernok, E., Schmitt, W.H., and Gross, W.L. (1994). Krol, T., Robinson, J., Bekeris, L., and Messmore, H. (1983). Hairy cell leuke-
Antineutrophil cytoplasmic antibodies in systemic polyarteritis nodosa mia and a fatal periarteritis nodosa-like syndrome. Archives of Pathology
with and without hepatitis B virus infection and Churg–Strauss syn- and Laboratory Medicine, 107, 583–5.
drome–62 patients. Journal of Rheumatology, 21, 1173–4. Küssmaul, A. and Maier, R. (1866). Ueber eine bisher nicht beschriebene
Hautekeete, M.L., Babany, G., Marcellin, P., et  al. (1990). Retroperitoneal eigenthümliche Arterienerkrankung (Periarteritis nodosa), die mit
fibrosis after surgery for aortic aneurysm in a patient with periarteri- Morbus Brightii und rapid fortschreitender allgemeiner Muskellähmung
tis nodosa: successful treatment with corticosteroids. Journal of Internal einhergeht. Deutsches Archiv für Klinische Medizin, 1, 484–518.
Medicine, 228, 533–6. Lee, C. and Kay, S. (1958). Primary polyarteritis nodosa of the stomach and
Henegar, C., Puéchal, X., Pagnoux, C., et al. (2008). A paradigm of diagnostic small intestine as a cause of gastro-intestinal hemorrhage. Annals of
criteria for polyarteritis nodosa: analysis of a series of 949 patients with Surgery, 147, 714–25.
vasculitides. Arthritis and Rheumatism, 52 (Suppl.), S225. Leib, E.S., Restivo, C., and Paulus, H.E. (1979). Immunosuppressive and
Héron, E., Fiessinger, J.N., and Guillevin, L. (2003). Polyarteritis nodosa pre- corticosteroid therapy of polyarteritis nodosa. American Journal of
senting as acute leg ischemia. Journal of Rheumatology, 30, 1344–6. Medicine, 67, 941–7.
Hirohata, S., Tanimoto, K., and Ito, K. (1993). Elevation of cerebrospinal fluid Leruez, V.M., Lauge, A., Morinet, F., Guillevin, L., and Dény, P. (1994).
interleukin-6 activity in patients with vasculitides and central nervous Polyarteritis nodosa and parvovirus B19. Lancet, 344, 263–4.
system involvement. Clinical Immunology and Immunopathology, Le Thi Huong, D., Wechsler, B., Guillevin, L., et al. (1985). Digestive mani-
66, 225–9. festations of periarteritis nodosa in a series of 120 cases [in French].
Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., et al. (1992). Wegener granulomato- Gastroentérologie Clinique et Biologique, 9, 697–703.
sis: an analysis of 158 patients. Annals of Internal Medicine, 116, 488–98. Le Tonqueze, M., Jamin, C., Bohme, M., Le Corre, R., Dueymes, M., and
Holsinger, D., Osmundson, P., and Edward, J. (1962). The heart in polyarteri- Youinou, P. (1996). Establishment and characterization of permanent
tis nodosa. Circulation, 25, 610–8. human endothelial cell clones. Lupus, 5, 103–12.
Holt, S. and Jackson, P. (1975). Ruptured coronary aneurysm and val- Levine, S.M., Hellmann, D.B., and Stone, J.H. (2002). Gastrointestinal
vulitis in an infant with polyarteritis nodosa. Journal of Pathology, involvement in polyarteritis nodosa (1986-2000): presentation and out-
117, 83–7. comes in 24 patients. American Journal of Medicine, 112, 386–91.
Horgan, C., Foster, C., D’Amico, D., et al. (1986). Retinal vasculitis in pol- Levy, A., Weinberger, A., Mor, C., and Pinkhas, J. (1986). Localized pol-
yarteritis nodosa. Retina, 6, 205. yarteritis nodosa: cases involving the lower extremities and the breast.
Hu, P.J., Shih, I.M., Hutchins, G.M., and Hellmann, D.B. (1997). Polyarteritis Rheumatology International, 6, 43–4.
nodosa of the pericardium: antemortem diagnosis in a pericardiectomy Li, A., Rhodes, J., and Valentine, A. (1979). Spontaneous liver rupture in pol-
specimen. Journal of Rheumatology, 24, 2042–4. yarteritis nodosa. British Journal of Surgery, 66, 251–2.
Hutson, T.E. and Hoffman, G.S. (2000). Temporal concurrence of vasculitis Lie, J.T. (1992). Retroperitoneal polyarteritis nodosa presenting as ureteral
and cancer: a report of 12 cases. Arthritis Care and Research, 13, 417–23. obstruction. Journal of Rheumatology, 19, 1628–31.
CHAPTER 25 polyarteritis nodosa 349

Lightfoot, R.W. Jr, Michel, B.A., Bloch, D.A., et  al. (1990). The American Pagnoux, C., Mahr, A., Cohen, P., and Guillevin, L. (2005). Presentation and
College of Rheumatology 1990 criteria for the classification of polyarte- outcome of gastrointestinal involvement in systemic necrotizing vas-
ritis nodosa. Arthritis and Rheumatism, 33, 1088–93. culitides: analysis of 62 patients with polyarteritis nodosa, microscopic
Lomeo, R.M., Silver, R.M., and Brothers, M. (1989). Spontaneous dissec- polyangiitis, Wegener granulomatosis, Churg–Strauss syndrome, or
tion of the internal carotid artery in a patient with polyarteritis nodosa. rheumatoid arthritis-associated vasculitis. Medicine (Baltimore), 84,
Arthritis and Rheumatism, 32, 1625–6. 115–28.
Lupoli, S., Gargiulo, A., Rossi, R., and Ames, P.R. (2004). Myositis as a Pagnoux, C., Seror, R., Henegar, C., et al. (2010). Clinical features and out-
presenting feature of polyarteritis nodosa. Clinical and Experimental comes in 348 patients with polyarteritis nodosa: a systematic retrospec-
Rheumatology, 22, 507–8. tive study of patients diagnosed between 1963 and 2005 and entered into
Luqmani, R., Bacon, P., Moots, R., et  al. (1994). Birmingham Vasculitis the French Vasculitis Study Group Database. Arthritis and Rheumatism,
Activity Score (BVAS) in systemic necrotizing vasculitis. QJM, 87, 671–8. 62, 616–26.
MacDonald, W.B. and Blake, M.P. (2004). Periostitis and localized myositis in Panegyres, P., Blumberg, P., Leong, A., and Bourne, A. (1990). Vasculitis of
polyarteritis nodosa. Clinical Nuclear Medicine, 29, 703–5. peripheral nerve and skeletal muscle:  clinicopathological correlation
Mader, R. and Keystone, E.C. (1992). Infections that cause vasculitis. Current and immunopathic mechanisms. Journal of the Neurological Sciences,
Opinion in Rheumatology, 4, 35–8. 100, 193–202.
Mahr, A., Guillevin, L., Poissonnet, M., and Aymé, S. (2004). Prevalences of Parangi, S., Oz, M.C., Blume, R.S., et al. (1991). Hepatobiliary complications
polyarteritis nodosa, microscopic polyangiitis, Wegener’s granulomatosis, of polyarteritis nodosa. Archives of Surgery, 126, 909–12.
and Churg–Strauss syndrome in a French urban multiethnic population Piette, J.C., Bourgault, I., Legrain, S., et  al. (1987). Systemic polyarteri-
in 2000: a capture-recapture estimate. Arthritis and Rheumatism, 51, 92–9. tis nodosa diagnosed at hysterectomy. American Journal of Medicine,
Malan, P., Ciurana, A., and Boudet, C. (1986). Artérite rétinienne bilatérale 82, 836–8.
avec dilatations anévrysmales multiples [in French]. Journal Français Porter, B.F., Frost, P., and Hubbard, G.B. (2003). Polyarteritis nodosa in a
d'Ophtalmologie, 9, 23–8. cynomolgus macaque (Macaca fascicularis). Veterinary Pathology,
Mason, A., Theal, J., Bain, V., Adams, E., and Perrillo, R. (2005). Hepatitis B 40, 570–3.
virus replication in damaged endothelial tissues of patients with extra- Prince, A.M. and Trépo, C. (1971). Role of immune complexes involving
hepatic disease. American Journal of Gastroenterology, 100, 972–6. SH antigen in pathogenesis of chronic active hepatitis and polyarteritis
Mayo, J., Arias, M., Leno, C., and Berciano, J. (1986). Vascular parkinsonism nodosa. Lancet, 1, 1309–12.
and periarteritis nodosa. Neurology, 36, 874–5. Puisieux, F., Woesteland, H., Hachulla, E., Hatron, P.Y., Dewailly, P., and
McMahon, B.J., Heyward, W.L., Templin, D.W., Clement, D., and Lanier, Devulder, B. (1997). Clinical symptomatology and prognosis of peri-
A.P. (1989). Hepatitis B-associated polyarteritis nodosa in Alaskan arteritis nodosa in the elderly. Retrospective study of 25 periarteritis
Eskimos: clinical and epidemiologic features and long-term follow-up. nodosa cases in young adults and 22 cases in aged patients [in French].
Hepatology, 9, 97–101. Revue de Médecine Interne, 18, 195–200.
Miller, D.L. (2000). Angiography in polyarteritis nodosa. American Journal of Rajani, R.M., Dalvi, B.V., D’Silva, S.A., Lokhandwala, Y.Y., and Kale, P.A.
Roentgenology, 175, 1747–8. (1991). Acute myocardial infarction with normal coronary arteries in
Moore, P. and Fauci, A. (1981). Neurologic manifestations of systemic vas- a case of polyarteritis nodosa: possible role of coronary artery spasm.
culitis: a retrospective and prospective study of the clinico-pathologic Postgraduate Medical Journal, 67, 78–80.
features and responses to therapy in 25 patients. American Journal of Rasmussen, N., Jayne, D., Abramowicz, D., et al. (1995). European therapeu-
Medicine, 71, 517–24. tic trials in ANCA-associated systemic vasculitis: disease scoring, con-
Moore, P.M. (1995). Neurological manifestation of vasculitis:  update on sensus regimens and proposed clinical trials. Clinical and Experimental
immunopathogenic mechanisms and clinical features. Annals of Immunology, 101, 29–34.
Neurology, 37 (Suppl. 1), 131–41. Reinhold-Keller, E., Herlyn, K., Wagner-Bastmeyer, R., et  al. (2002). No
Moore, P.M. and Calabrese, L.H. (1994). Neurologic manifestations of sys- difference in the incidences of vasculitides between north and south
temic vasculitides. Seminars in Neurology, 14, 300–6. Germany: first results of the German vasculitis register. Rheumatology
Moreland, L.W. and Ball, G.V. (1990). Cutaneous polyarteritis nodosa. (Oxford), 41, 540–9.
American Journal of Medicine, 88, 426–30. Ribi, C., Cohen, P., Pagnoux, C., et  al. (2010). Treatment of polyarteritis
Mowrey, F.H. and Lundberg, E.A. (1954). The clinical manifestations of nodosa and microscopic polyangiitis without poor-prognosis factors: a
essential polyangiitis (periarteritis nodosa), with emphasis on the prospective randomized study of one hundred twenty-four patients.
hepatic manifestations. Annals of Internal Medicine, 40, 1145–64. Arthritis and Rheumatism, 62, 1186–97.
Nakamura, T., Tomoda, K., Yamamura, Y., Tsukano, M., Honda, I., and Rodríguez Pereira, C., Suárez Peñaranda, J.M., del Río, E., and Forteza Vila, J.
Iyama, K. (2003). Polyarteritis nodosa limited to calf muscles:  a case (1997). Cutaneous granulomatous vasculitis after herpes zoster infection
report and review of the literature. Clinical Rheumatology, 22, 149–53. showing polyarteritis nodosa-like features. Clinical and Experimental
Nakazawa, K., Itoh, N., Duan, H.J., Komiyama, Y., and Shigematsu, H. Dermatology, 22, 274–6.
(1992). Polyarteritis nodosa with atrophy of the left hepatic lobe. Acta Ropert, A. and Metral, S. (1990). Conduction block in neuropathies with
Pathologica Japonica, 42, 662–6. necrotizing vasculitis. Muscle Nerve, 13, 102–5.
Nohr, M., Laustsen, J., and Falk, E. (1989). Isolated necrotizing panarteritis of Sack, M., Cassidy, J.T., and Bole, G.G. (1975). Prognostic factors in polyarte-
the gallbladder. Case report. Acta Chirurgica Scandinavica, 155, 485–7. ritis. Journal of Rheumatology, 2, 411–20.
Ozen, S., Anton, J., Arisoy, N., et al. (2004). Juvenile polyarteritis: results of Said, G. (1997). Necrotizing peripheral nerve vasculitis. Neurologic Clinics,
a multicenter survey of 110 children. Journal of Pediatrics, 145, 517–22. 15, 835–48.
Ozen, S., Pistorio, A., Iusan, S.M., et al. (2010). EULAR/PRINTO/PReS cri- Said, G. and Lacroix, C. (2005). Primary and secondary vasculitic neuropa-
teria for Henoch-Schönlein purpura, childhood polyarteritis nodosa, thy. Journal of Neurology, 252, 633–41.
childhood Wegener granulomatosis and childhood Takayasu arte- Salojin, K.V., Le Tonqueze, M., Nassovov, E.L., et al. (1996). Anti-endothelial
ritis:  Ankara 2008. Part II:  Final classification criteria. Annals of the cell antibodies in patients with various forms of vasculitis. Clinical and
Rheumatic Diseases, 69,798–806. Experimental Rheumatology, 14, 163–9.
Ozen, S., Ruperto, N., Dillon, M., et al. (2006). EULAR/PReS endorsed con- San, A., Aydin, N.E., Akcay, G., Selcuk, Y., and Okyar, G. (1990). Spontaneous
sensus criteria for the classification of childhood vasculitides. Annals of bilateral rupture of kidneys in a patient with polyarteritis nodosa. A case
the Rheumatic Diseases, 65, 936–41. report. Scandinavian Journal of Urology and Nephrology, 24, 319–21.
350 SECTION 5 vasculitic diseases and syndromes and related disorders

Schrader, M.L., Hochman, J.S., and Bulkley, B.H. (1985). The heart in pol- Till, S.H. and Amos, R.S. (1997). Long-term follow-up of juvenile-onset
yarteritis nodosa:  a clinicopathologic study. American Heart Journal, cutaneous polyarteritis nodosa associated with streptococcal infection.
109, 1353–9. British Journal of Rheumatology, 36, 909–11.
Scott, D., Bacon, P., Elliott, P., Tribe, C., and Wallington, T. (1982). Systemic Travers, R.L., Allison, D.J., Brettle, R.P., and Hughes, G.R. (1979). Polyarteritis
vasculitis in a district general hospital 1972-1980: clinical and labora- nodosa:  a clinical and angiographic analysis of 17 cases. Seminars in
tory features, classification and prognosis in 80 cases. QJM, 51, 292–311. Arthritis and Rheumatism, 8, 184–99.
Sergent, J., Lockshin, M., Christian, C., and Gocke, D. (1976). Vasculitis Trépo, C. and Thivolet, J. (1970a). Hepatitis associated antigen and periarte-
with hepatitis B antignemia: long term observations in nine patients. ritis nodosa. Vox Sanguinis, 19, 410–1.
Medicine (Baltimore), 55, 1–18. Trépo, C. and Thivolet, J. (1970b). Antigène Australia, virus de l’hépatite et
Servant, A., Bogard, M., Delaugerre, C., Cohen, P., Dény, P., and Guillevin, périartérite noueuse. Presse Médicale, 78, 1575.
L. (1998). GB virus C in systemic medium- and small-vessel necrotizing Trépo, C., Zuckerman, A., Bird, R., and Prince, A. (1974). The role of circulat-
vasculitides. British Journal of Rheumatology, 37, 1292–4. ing hepatitis B antigen/antibody immune complexes in the pathogenesis
Short, D.J. and Webley, M. (1984). Periosteal new bone formation com- of vascular and hepatic manifestations in polyarteritis nodosa. Journal of
plicating juvenile polyarteritis nodosa. Journal of the Royal Society of Clinical Pathology, 27, 863–8.
Medicine, 77, 325–7. Vathenen, A., Skinner, D., and Shale, D. (1988). Treatment response with
Sneller, M.C. (2000). Cystitis, bladder cancer, and myelodysplasia in bilateral mixed deafness and facial palsy in polyarteritis nodosa.
patients with Wegener’s granulomatosis:  comment on the article by American Journal of Medicine, 84, 1081–2.
Reinhold-Keller et al. Arthritis and Rheumatism, 43, 2853–5. Wang, T., Avlonitis, E., and Relkin, R. (1988). Systemic necrotizing vasculitis
Stillwell, T., Benson, R., DeRemee, R., et al. (1988). Cyclophosphamide’induced causing bone necrosis. American Journal of Medicine, 84, 1085–6.
bladder toxicity in Wegener’s granulomatosis. Arthritis and Rheumatism, Warfield, A.T., Lee, S.J., Phillips, S.M., and Pall, A.A. (1994). Isolated tes-
31, 465–70. ticular vasculitis mimicking a testicular neoplasm. Journal of Clinical
Takahashi, M. (1993). Inflammatory diseases of the coronary artery in chil- Pathology, 47, 1121–3.
dren. Coronary Artery Disease, 4, 133–8. Watts, R., Gonzalez-Gay, M., Lane, S., et al. (2001). Geoepidemiology of sys-
Tamby, M.C., Chanseaud, Y., Humbert, M., et al. (2005). Anti-endothelial temic vasculitis: comparison of the incidence in two regions of Europe.
cell antibodies in idiopathic and systemic sclerosis associated pulmo- Annals of the Rheumatic Diseases, 60, 170–2.
nary arterial hypertension. Thorax, 60, 765–72. Weck, K.E., Dal Canto, A.J., Gould, J.D., et  al. (1997). Murine
Tang, P.H. and Segal, A.J. (1971). Polyarteritis nodosa of infancy. Fatal late gamma-herpesvirus 68 causes severe large-vessel arteritis in mice lack-
complication. JAMA, 217, 1666–70. ing interferon-gamma responsiveness: a new model for virus-induced
Tasdemir, I., Turgan, C., Emri, S., et al. (1988). Spontaneous perirenal hae- vascular disease. Nature Medicine, 3, 1346–53.
matoma secondary to polyarteritis nodosa. British Journal of Urology, Yamashina, M. and Wilson, T.K. (1985). A mammographic finding in focal
62, 219–22. polyarteritis nodosa. British Journal of Radiology, 58, 91–2.
Teichman, J.M., Mattrey, R.F., Demby, A.M., and Schmidt, J.D. (1993). Zeek, P. (1952). Periarteritis nodosa. A critical review. American Journal of
Polyarteritis nodosa presenting as acute orchitis:  a case report and Pathology, 22, 277–90.
review of the literature. Journal of Urology, 149, 1139–40. Zizic, T.M., Classen, J.N., and Stevens, M.B. (1982). Acute abdominal com-
Thone, M. (1988). Buccal ulceration in a case of periarteritis nodosa [in plications of systemic lupus erythematosus and polyarteritis nodosa.
French]. Acta Stomatologica Belgica, 85, 69–75. American Journal of Medicine, 73, 525–31.
CHAPTER 26

Microscopic polyangiitis
Loïc Guillevin and Benjamin Terrier

Introduction and/or lung capillaritis. However, it recognizes that MPA may also,
but not predominantly, affect medium-sized vessels, which may
Initially considered a ‘microscopic’ form of polyarteritis nodosa raise a real diagnostic dilemma for clinicians and pathologists con-
(PAN), microscopic polyangiitis (MPA) was considered a distinct fronted with such an overlapping clinical picture.
entity by Wohlwill (1923), and then redefined by Davson et  al. Another International Chapel Hill Consensus Conference, held
(1948). Its unanimous recognition as a clearly different disease in 2012, was convened to improve the 1994 nomenclature, but
among systemic vasculitides occurred only years later (Jennette the definition of MPA was not modified (see Table 25.2) (Jennette
et al. 1994). MPA is a small-size-vessel systemic necrotizing vas- et al. 2013).
culitis whose several typical clinical extrarenal and pulmonary
manifestations are similar to those encountered in PAN, thereby
explaining decades of confusion. In further contrast to PAN, MPA
Epidemiology
belongs to the group of vasculitides associated with antineutrophil Like all other vasculitides, MPA is a rare disease. Furthermore,
cytoplasm antibodies (ANCA), such as granulomatosis with poly- because PAN and MPA were long grouped together in published
angiitis (Wegener’s granulomatosis, GPA) and eosinophilic granu- studies valid epidemiological data are sparse; therefore only the
lomatosis with polyangiitis (Churg–Strauss syndrome, EGPA). more recent studies can be considered reliable for MPA frequen-
Recognition of MPA as a separate entity also implied different cies. MPA has been reported world-wide and affects all racial/ eth-
therapeutic strategies than those adopted for PAN and closer to the nic groups, but with a probable predominance in Caucasians (Falk
treatment of GPA. However, MPA has some unique features, which et al. 1990b; Handa et al. 2001). Males are affected slightly more fre-
are detailed in this chapter. quently with a male:female ratio ranging from 1.1 to 1.8 (Adu et al.
1987; Savage et al. 1985). The average age at onset is about 50 years.
Soon after the introduction of ANCA-testing and the first Chapel
Classification criteria Hill Consensus Conference, total annual incidence was estimated
MPA did not appear in the classification criteria established in at 3.6 per 1 000 000 inhabitants in England (Watts et al. 1996). As
1990 by the American College of Rheumatology (ACR) (Bloch for GPA, some authors reported a possible North–South gradient,
et al. 1990; Lightfoot et al. 1990). In the past, PAN and MPA were with the highest annual incidence being observed in Northern
thought to be different forms of the same disease because of their European countries, at 16 per 1 000 000 inhabitants in Sweden, for
shared main clinical manifestations (constitutional symptoms, example (Tidman et al. 1998). In France, MPA prevalence was 25.1
neurological, gastrointestinal manifestations, etc.), and therefore per 1 000 000 inhabitants in Seine–Saint-Denis, a northern suburb
they were grouped together in the ACR classification criteria. of Paris (Mahr et al. 2004), suggesting a possible increasing MPA
However, major differences exist between these two entities, and frequency over the last few decades. Its recognition as a distinct
this divergence needed to be clarified. MPA is a small-size-vessel clinical entity and the use of ANCA-testing might also explain (at
vasculitis, which explains some of its clinical characteristics, such least in part) these findings, which must be confirmed by additional
as glomerular involvement, with frequent presence of rapidly pro- studies.
gressive glomerulonephritis (RPGN), and sometimes pulmonary
capillaritis, manifesting as alveolar haemorrhage, signs of which
are not found in PAN (Savage et al. 1985). Since the discovery of Pathogenesis
ANCA (van der Woude et al. 1985), which are only present in some MPA is an ANCA-associated vasculitis. Perinuclear-labelling
small-size-vessel vasculitides, but not in PAN, MPA has been fur- pANCA with antimyeloperoxidase (MPO) specificity (Jennette
ther differentiated from PAN. et al. 1989) have been found in nearly 75% of the patients (Guillevin
The Chapel Hill International Consensus Conference estab- et al. 1999). MPO is a 140-kDa protein composed of two identi-
lished a new nomenclature for systemic vasculitides that defines cal heterodimers encoded by a single gene, located in chromo-
and clearly separates MPA and PAN (Jennette et al. 1994). In this some 17q21.3. MPO constitutes about 5% of all neutrophil proteins
nomenclature, intended for classification and not for diagnosis, and is present in the cytoplasmic granules at very high concen-
MPA is defined as a necrotizing vasculitis, with few or no immune trations. Investigators have thought that this enzyme catalysed
deposits, that affects small-sized vessels, especially arterioles, capil- the H2O2-dependent oxidation of halides that can react with and
laries and venules, and is often responsible for glomerulonephritis kill microbes. Another possible MPO function is to protect the
352 SECTION 5 vasculitic diseases and syndromes and related disorders

digestive enzymes from oxidative denaturation by removing H2O2 systemic vasculitis with lung, spleen, and lymph-node involve-
from the phagocyte vacuole, and, in fact, MPO-knockout mice are ment. These diseases were reproduced by passive transfer of puri-
abnormally susceptible to bacterial and mainly fungal infections. fied IgG anti-MPO from MPO−/− mice into rag2−/− and wild-type
A few MPA patients have cytoplasm-labelling cANCA with mice, thereby demonstrating their pathogenicity. Subsequent stud-
antiproteinase 3 (PR3) specificity. PR3 is a 29-kDa neutrophil ser- ies using this experimental model focused on the crucial role of
ine protease, encoded by a single gene, located in chromosome neutrophils in glomerulonephritis (Xiao et al. 2005). The Chapel
19p13.3. This protein is stored in granules of human neutrophils Hill group showed that selective neutrophil depletion in these mice
and monocytes. PR3 is mostly involved in GPA pathogenesis. with a monoclonal antibody protected them against vasculitis.
ANCA are now considered a useful immunological marker for These observations raised the question of whether a therapeutic
the diagnosis of vasculitides. Over the last decade, numerous in strategy should, or even could, be devised to target neutrophils.
vitro data and two animal models (Heeringa et  al. 1998; Pfister The same group demonstrated the role of TNF-α in this experi-
et  al. 2004; Xiao et  al. 2002)  supported a direct pathogenic role mental MPA model, when pretreatment with anti-TNF-α antibod-
of ANCA in systemic vasculitides. Falk et al. (1990a) showed that ies attenuated the glomerulonephritis induced by MPO-ANCA and
IgG ANCA can activate tumour necrosis factor (TNF)-α-primed bacterial lipopolysaccharides (Huugen et al. 2005).
neutrophils, thereby inducing the production of reactive oxygen Little et al. (2006) used an experimental rat model to demon-
metabolites and the release of lysosomal proteolytic enzymes, strate that MPO-ANCA are pathogenic. Immunization of that
including ANCA-target antigens. They demonstrated that ANCA rat strain with human MPO led to the synthesis of antibodies
promote neutrophil adhesion to endothelial cells and their lysis. recognizing murine MPO, and to the development of systemic
The mechanisms involved in ANCA-mediated neutrophil activa- vasculitis with pauci-immune glomerulonephritis, closely resem-
tion are not clearly understood. After TNF-α-priming, neutrophils bling human disease. In addition, passive transfer of purified
express the ANCA-target antigen on their cell membranes, mak- IgG anti-MPO from immunized rats into non-immunized rats
ing it accessible for interaction with ANCA (Csernok et al. 1999). also induced systemic vasculitis. Intravital microscopy of these
These target antigens are also expressed on the surface membranes rats showed dynamic leukocyte interaction with vessel walls after
of apoptotic non-primed neutrophils and recognized by ANCA injection of purified MPO-ANCA. Furthermore, these modifi-
(Falk et al. 1990a; Gilligan et al. 1996). cations of leukocyte adhesion and vessel wall interactions were
It has been shown that ANCA-induced activation is depend- also found in vitro for this animal model. Notably, transplacental
ent on the cross-linking of surface molecules. For example IgG MPO-ANCA transfer resulting in neonatal MPA was reported,
ANCA and F(ab)2 fragments, but not Fab fragments, were able to supporting MPO-ANCA pathogenicity in humans (Bansal and
stimulate the production of oxygen radicals by primed neutrophils Tobin 2004; Schlieben et al. 2005).
(Kettritz et  al. 1997). Because neither PR3 nor MPO contains a More recently, Guilpain et  al. (2007) investigated whether
transmembrane domain and a cytoplasmic tail for signal transduc- anti-MPO antibodies could activate MPO to generate oxidative
tion, other molecules or receptors must perform this function, for stress potentially deleterious to the endothelium. Those antibodies
example FcγRIIa receptors, which are extensively involved in neu- were indeed able to activate MPO in vitro and to generate hypochlo-
trophil activation (Mulder et al. 1994). In addition, β2-integrins rous acid. Moreover, the by-products of MPO activation by MPA
have been shown to bind MPO (Reumaux et al. 1995) and are a sera exerted strong cytolytic activity against endothelial cells in
third factor required for ANCA activation of neutrophils. Indeed, culture, and both hypochlorous acid production and endothelial
ANCA-mediated neutrophil activation does not occur when neu- lysis were abrogated by N-acetylcysteine. In addition to the severe
trophil adhesion is prevented by either continuous stirring of endothelial damage, the interaction of MPO with anti-MPO anti-
the cell suspension or the addition of antibodies directed against bodies could trigger the fibrotic process observed in MPA, in par-
CD18, a subunit of β2-integrins. Importantly, ANCA can also acti- ticular lung fibrosis (Guilpain et al. 2011).
vate monocytes, which results in enhanced production of reactive The role of complement as an important mediator in
oxygen species (Weidner et  al. 2001), interleukin-8, and mono- anti-MPO-associated vasculitis was also investigated. First, the
cyte chemoattractant protein-1 (MCP-1) (Casselman et al. 1995; glomerulonephritis induced by the transfer of anti-MPO IgG into
Ralston et al. 1997). wild-type mice or anti-MPO splenocytes into immune-deficient
Animal models of ANCA-associated vasculitis with glomerulo- mice could be completely blocked by complement deple-
nephritis have failed to implicate ANCA as an independent cause tion. The same model was used in mice with knocked-out
of the disease (reviewed by Heeringa et al. 1998). Convincing evi- complement-pathway components, showing that C5−/− and factor
dence came from MPO-deficient mice developed by Xiao et  al. B−/− mice developed no disease. Those observations suggest that
(2002). Their experimental model is based on immunization of ANCA stimulation of neutrophils causes the release of factors that
MPO-deficient mice to circumvent MPO tolerance. First, the activate complement via the alternative pathway and initiate an
immunization with murine MPO generated antimurine MPO anti- inflammatory amplification loop that mediates the severe necrotiz-
bodies. Second, another immune-deficient mouse, lacking T and ing inflammation of ANCA vasculitis (Xiao et al. 2007).
B cells, was created by inactivating recombinase-activating gene-2 The general concept derived from all those studies is that
(rag2) to obtain rag2-deficient animals to study the effect of adoptive ANCA-induced vasculitis may be a two-hit process, in which
transfer of foreign antibodies or graft immunity. When splenocytes ANCA, in conjunction with proinflammatory stimuli, most
from MPO−/− immunized mice were transferred into rag2−/− likely infectious, are required for the development of full-blown
mice, anti-MPO antibodies were detected a few days later and all disease.
mice developed severe necrotizing crescentic glomerulonephritis, Antiendothelial cell antibodies (AECA) constitute a hetero-
closely resembling human disease. Other rag2−/− mice developed geneous group of autoantibodies distinct from ANCA. AECA
CHAPTER 26 microscopic polyangiitis 353

have been detected in systemic lupus erythematosus, systemic (MHC) and non-MHC associations with ANCA-associated vascu-
sclerosis, and primary and secondary systemic vasculitides litides, and also demonstrated that GPA and MPA are genetically
(Chan and Cheng 1996; Salojin et  al. 1997; Praprotnik et  al. distinct. The strongest genetic associations were with the anti-
2000). AECA presence has been well documented in idiopathic genic specificity of ANCA, not with the clinical syndrome. Along
retinal vasculitis, GPA, MPA, and Kawasaki disease, and their this line, anti-PR3 antibody was associated with HLA-DP and the
titres paralleled disease activity. A  growing body of evidence genes encoding α1-antitrypsin and proteinase 3 (P=6.2×10−89,
suggests that AECA might be pathogenic, inducing systemic P=5.6×10−12, and P=2.6×10−7, respectively), while anti-MPO anti-
vasculitis by enhancing leukocyte adhesion to endothelial cells. body was associated with HLA-DQ (P=2.1×10−8).
AECA targets comprise a wide range of extracellular matrix
proteins, DNA, or phospholipids, but their precise nature in Clinical features
patients with small- and medium-size-vessel systemic vascu-
litides remains unknown. Using a quantitative immunoblot- Clinical features of MPA are summarized in Table 26.1. It should
ting technique, Chanseaud et al. (2003) showed that IgM and, be kept in mind that, in addition to failure to distinguish between
to a lesser degree, IgG from MPA patients recognized multi- PAN and MPA in earlier series, the first reported studies on patients
ple endothelial cell antigens, but their precise identity remains who satisfied MPA classification criteria were primarily from neph-
unknown. rology departments and only later from internal medicine depart-
The role of T cells in MPA, as opposed to GPA, is poorly estab- ments as well. Hence, the reported frequencies of involvement of
lished; however, T-cell responses to MPO have been observed each organ in MPA should always be interpreted after considering
(King et  al. 1998; Popa et  al. 2002). The lack of substantial lym- the patients’ characteristics.
phocyte participation in the early stages of MPA is consistent with
the observation that, in the mouse model of Xiao et al. (2002), glo- General symptoms and clinical
merulonephritis can be induced by injecting IgG anti-MPO into picture at disease onset
rag2−/− mice that lack functional T cells. Systemic symptoms (e.g. myalgias, arthralgias, or arthritis) are pre-
Finally, the role of predisposing genetic factors was demonstrated sent in 56–76% of patients before MPA is diagnosed. There can be
in a genome-wide association study (Lyons et al. 2012), performed an indolent course of several months or even years before diagno-
on a UK discovery cohort and was replicated in northern European sis, with general symptoms or even episodic, mildly bloody sputum
patients. That study revealed both major histocompatibility complex which is ignored by the patient.

Table 26.1 Microscopic polyangiitis: clinical features and organ or system involvement, expressed as percentages of the studied population

Serra et al. 1984 Savage et al. 1985 D'Agati et al. 1986 Adu et al. 1987 Guillevin et al. 1999
No. of patients 53 34 20 43 85
Mean age (year) 53 50 50 – 57
Sex ratio M/F 1.5 1.8 1 1.7 1.2
General signs (fever, asthenia, myalgias, 79 76 – – 73
arthralgias)
Hypertension 26 29 35 21 34
Kidney 100 100 100 100 79
Skin 60 – 35 53 62
Purpura 40 44 – – 41
Lung 55 – 55 34 25
Haemoptysis 23 32 – – –
Infiltrates 30 – – – 10
Pleural effusion 19 15 – – 6
Gastrointestinal tract 51 – – 56 30
Ear, nose, throat 30 – – 20 –
Sinusitis 6 9 – – 11
Eyes 30 – – 28 –
Nervous system 28 – – – –
Peripheral 19 18 15 14 58
Central 15 18 40 0 12
Heart 15 – – 9 –
354 SECTION 5 vasculitic diseases and syndromes and related disorders

Renal and urogenital involvement


Renal involvement, predominantly RPGN, is a major feature of
MPA. It appeared to be nearly constant in the first series of MPA
patients, but they were reported by nephrologists, leading to refer-
ral bias in the incidence of renal disease. Most of the patients have
renal impairment at diagnosis and renal function that rapidly dete-
riorates if not treated. Eight of the 34 patients reported by Savage
et al. (1985) required dialysis for oliguric or anuric renal disease, as
did 20 of the 43 patients described by Adu et al. (1987). Initial renal
manifestations are often silent, but detection of microscopic haema-
turia with or without proteinuria usually precedes deterioration of
renal function. Several MPA flares with very few clinical symptoms
may occur before renal manifestations become evident, as shown
by the results of renal biopsies, which can reveal the co-existence of
acute glomerular lesions and glomerular scars.
Ureteral involvement, which is much more suggestive of GPA, is
rare in MPA. It has been described rarely in PAN, but some of those Fig. 26.1 Computed tomography scan showing microcystic fibrosis of the lung in
cases might have been MPA rather than PAN. Uni- or bilateral ure- a patient with anti-MPO ANCA.
teral stenoses may occur, and are usually localized to the lower part
of ureter(s) or more rarely at the ureteropelvic junction. Abdominal
fibrosis associated with other collagen vascular diseases in which
pain and/or anuria are the most common, but non-specific, symp-
anti-MPO pANCA are not found (Homma et al. 2004).
toms of this complication. Indeed, some patients have no clinical
urinary warning symptoms and renal destruction may progress Cutaneous manifestations
silently. Diagnosis of ureteral stenoses can be confirmed by intra-
venous (i.v.) urography, when renal impairment does not firmly Skin lesions are found in 30–60% of the patients (Penas et al. 1996;
contraindicate iodinated contrast-medium injection, or ultra- Lhote et  al. 1998). Maculopapular purpuric lesions of the lower
sonography or magnetic resonance imaging can be done, as both limbs are the most frequent skin manifestations. Other lesions have
these techniques are less toxic. Vasculitis may be detected in the been described, such as mouth ulcers, vesicles, necrosis, ulcera-
ureteral wall or the periureteral fat. Retroperitoneal involvement tions, nodules, splinter haemorrhages, livedo reticularis, hand and/
might also be responsible for ureteral obstruction, as was described or finger erythema, and facial oedema (Homas et al. 1992; Seishima
for PAN (Lie 1992; van Bommel et al. 2002). et al. 2004). Leukocytoclastic vasculitis of the small vessels of the
dermis is usually observed. Sometimes, arterioles or smaller vessels
of the deep dermis and subcutis are also involved, thereby explain-
Pulmonary involvement ing the nodular appearance of some skin lesions. In one patient,
Alveolar haemorrhage can be observed in MPA. Moreover, as in vasculitis was associated with eosinophilic panniculitis (Penas et al.
GPA or Goodpasture’s syndrome, MPA can also be responsible 1996). All these cutaneous lesions usually disappear rapidly under
for a pulmonary–renal syndrome. Haemoptysis, or even moder- treatment, but relapses may occur.
ately bloody expectorations, may precede severe alveolar haem-
orrhage, which is characterized by dyspnoea and anaemia, and Other clinical manifestations
may progress to diffuse alveolar damage and respiratory distress Not surprisingly, clinical features are similar to those of PAN.
due partially or completely to capillaritis (Savage et  al. 1985). Peripheral neuropathy is found in 10–58% of the patients, much
Bronchial arteritis with intimal thickening and focal destruction less frequently than in PAN (Savage et al. 1985; Rodgers et al. 1989;
of the internal elastic lamina, associated with subintimal fibrous Guillevin et al. 1999). Mononeuropathy multiplex is, as in all other
scarring in the media, may be found during histological examina- vasculitides, the main clinical feature, and is sometimes isolated.
tion of a lung biopsy. Among the patients with peripheral nervous system involvement,
Diffuse alveolar damage and interstitial lung fibrosis (Figure 26.1) 69% have mononeuropathy multiplex, 12% have symmetrical poly-
can also be complications of the vasculitis, mainly in patients with neuropathy, and 19% have asymmetrical polyneuropathy (Hattori
anti-MPO pANCA, and the fibrosis and vasculitis can evolve inde- et al. 2002). Necrotizing vasculitis has been reported in up to 81% of
pendently (Souid et al. 2001; Hervier et al. 2009). However, lung sural nerve biopsies. Central nervous system involvement (mostly
fibrosis has also been described as an isolated process confined to encephalopathy and stroke) and cranial neuropathies have been
the lungs in some patients with anti-MPO pANCA, but no evidence reported in 12–18% of patients (Savage et al. 1985; Généreau et al.
of vasculitis. Hence, lung fibrosis with anti-MPO ANCA might be 1999), reflecting small-vessel vasculitis. Pachymeningitis, thought
a separate entity from MPA (Hiromura et al. 2000; Homma et al. to possibly result from dural necrotizing vasculitis, has also been
2004). Chest computed-tomography (CT) scans usually show hon- described (Kono et al. 2000).
eycombing fibrosis in the bases of the lungs, and histological fea- Gastrointestinal symptoms occur in 30–60% of the patients,
tures are compatible with the usual interstitial pneumonia pattern characterized by abdominal pain (32–58%) and/or bleeding (29%).
and vasculitis, in less than half the patients. The mortality rate of Severe small intestine or large bowel ischaemia, ulcerations, and/or
patients with lung fibrosis and anti-MPO pANCA was similar to perforations occur as in PAN, but published case reports indicate
that of those with idiopathic alveolar fibrosis, but worse than for it is clearly less frequent in MPA (Ueda et al. 2001; Tsai et al. 2004).
CHAPTER 26 microscopic polyangiitis 355

Reports of cardiovascular complications in MPA are rare. Among the presence of glomerular sclerosis and frequent severe tubu-
the 85 MPA patients described by Guillevin et al. (1999), heart fail- lar damage, more than to active glomerular disease and crescents
ure and pericarditis occurred at respective frequencies of 17.6% (Hogan et al. 1996; Zauner et al. 2002; Kapitsinou et al. 2003).
and 10%. Severe acute congestive heart failure has been reported,
but rarely with documented myocardial infarction due to myocar-
dial or coronary small-vessel arteritis (Wang et al. 2002); however,
Outcome and prognosis
subclinical myocardial infarctions may be more frequent, as in PAN Relapses
and other small-vessel vasculitides. MPA lung and kidney involve- Relapses are frequent in MPA. Savage et al. (1985) reported that
ment appear to be associated with pANCA, whereas heart involve- 12/33 (36%) MPA patients had relapses. In the study by Gordon
ment may be more frequent in ANCA-negative patients, as in those et al. (1993), relapses occurred less frequently (25%), with a median
with EGPA (Sablé-Fourtassou et al. 2005; Sinico et al. 2005). time to MPA relapse of 24 months. In our MPA population, 34%
Ocular manifestations, such as eyelid inflammation, iridocyclitis, relapsed (Guillevin et  al. 1999). At present, it is not possible to
scleritis, retinal cotton-wool spots, retinal vasculitis, and/or cho- identify the subgroup of patients who will relapse or to predict the
roiditis, may be seen more frequently in MPA than PAN, but still in severity of those relapses. The clinical pattern of relapse does not
less than 30% of the patients, and they often remain clinically silent necessarily repeat the original presentation and organs that were
(Caster et al. 1996; Mihara et al. 2005). not previously affected can be involved. Nevertheless, relapses are
Some controversy exists regarding ear, nose, and throat involve- usually less severe than initial manifestations, most often consisting
ment, which is more characteristic of GPA and is sometimes con- of rash and arthralgias.
sidered by some authors to be an exclusionary symptom for MPA
diagnosis, which is probably a too restrictive point of view. Mild Deaths
non-granulomatous, non-erosive sinus inflammation may be noted
in up to 29% of MPA patients (Serra et al. 1984; Lane et al. 2005). When vital organs are involved, death may occur. A few patients die
during the first months of the disease from multiorgan involvement
because treatment is unable to control the disease, which is charac-
Laboratory findings terized by fever, rapid weight loss, diffuse pain, and one or multiple
Non-specific tests reflect the systemic inflammatory nature of MPA. major organ involvement. The remaining deaths are mainly attrib-
The major abnormalities are elevated erythrocyte sedimentation uted to renal failure and lung haemorrhage.
rate, C-reactive protein concentration, platelet and white blood cell Iatrogenic deaths are not rare. Those occurring during the first
counts and low haemoglobin (normochromic normocytic anae- few months of the disease are often caused by uncontrolled vascu-
mia), and serum albumin levels. All patients reported by Savage litis; deaths during the following years may be the consequence of
et  al. (1985) had impaired renal function with creatininaemia treatment side-effects. These adverse events emphasize the impor-
>120 µmol/l (1.4 mg/dl) and only 15% of those described by Serra tance of an individually tailored regimen, established after careful
et al. (1984) had normal plasma creatinine levels. The mean plasma analysis of parameters susceptible to ‘predict’ outcome. Infections
creatinine was 574  µmol/l (6.5 mg/dl) (range:  147–1405  µmol/l, are the primary cause of such deaths, influenced by corticosteroids
1.7–15.9 mg/dl) in the Hammersmith series (Savage et al. 1985). and cytotoxic agents. Lowering doses and shortening treatment
Microscopic haematuria is constant and proteinuria is found in duration can limit these complications, but treatment intensity and
more than 90% of patients, often >3 g/24 h. duration need to be further refined based on prospective studies.
ANCA are frequently detected in MPA patients. For example, Septicaemia can develop during the first months of treatment as
75% of the 33 MPA patients reported by Hauschild et al. (1994) a consequence of the intense initial therapy. Viral infections usu-
had ANCA: 61% pANCA and 15% cANCA. Most ANCA detected ally arise later and result from the profound immunosuppression
in MPA are anti-MPO pANCA, although anti-PR3 can also be induced by the drugs prescribed to control the vasculitis. We also
found. Among our 85 patients selected according to the Chapel Hill want to emphasize that rare cases of Pneumocystis jiroveci pneu-
nomenclature, 75% were ANCA positive (Guillevin et  al. 1999). monia, which have mainly been described in GPA (Jarrousse et al.
The usefulness of anti-MPO ANCA to diagnose MPA is now well 1993; Godeau et al. 1994), can also occur in MPA.
established (Jennette et al. 1989). However, despite the close con-
nection between those autoantibodies and MPA, the diagnostic Treatment
sensitivity of this anti-MPO ANCA specificity is not as high as that
for anti-PR3 ANCA in GPA, partly because pANCA are found in a MPA severity
variety of different inflammatory disorders and are not specific to To help the clinician choose the most effective therapy and avoid
vasculitis and/or glomerulonephritis. overtreatment, we devised a prognostic score, the Five Factor Score
Visceral angiography is usually normal, without the stenoses (FFS) (Guillevin et al. 1996b), whose parameters, defined as fol-
and/or microaneurysms characteristic of PAN. ANCA positivity lows, are responsible for higher mortality (Table 26.2): proteinu-
with abnormal angiograms are the exception. Therefore, it does not ria >1 g/day, renal insufficiency (creatininaemia >140  µmol/l or
seem informative to perform angiography before renal biopsy in 1.6  mg/dl), specific cardiomyopathy, gastrointestinal manifesta-
ANCA-positive patients (Guillevin et al. 1996a). tions, and/or central nervous system involvement. Treatment
Renal histology is characterized by focal segmental thrombosing should be chosen as a function of these criteria.
and necrotizing glomerulonephritis. Extracapillary crescents are The 1996 FFS for systemic necrotizing vasculitides was revis-
present in nearly all renal biopsies and often involve more than 60% ited, including GPA (Guillevin et  al. 2011). The following fac-
of the glomeruli. The severity of renal impairment corresponds to tors, significantly associated with higher 5-year mortality, were
356 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 26.2 The Five Factor Score (FFS), as established based on 342 patients with PAN or CSS (Guillevin et al. 1996b) and further validated
for patients with MPA (Gayraud et al. 2001)

FFS 5-year survival rate (%) Relative risk

Proteinuria > 1 g/24 h


Creatininaemia > 140 µmol/l or 1.6 mg/dl 0 88.1 0.62
Specific gastrointestinal involvement 1 74.1* 1.35
Specific cardiomyopathy ≥2 54.1** 2.40
Specific central nervous system involvement *P < 0.005 and **P < 0.0001 as compared to patients with FFS=0.
1 point accorded for each of these 5 items when present

scored +1 point: age >65 years, cardiac symptoms, gastrointestinal cyclophosphamide (CYC) or other cytotoxic agents in addition to
involvement, and renal insufficiency (stabilized peak creatinine corticosteroids (CS). Most MPA patients who relapse do so when
≥150 μmol/l or 1.7 mg/dl). Because ear, nose, and throat symptoms treatment is discontinued (33%), but relapses on treatment are
are associated with a better prognosis, their absence was scored +1 frequent while tapering the CS dose (Gordon et al. 1993). Gaskin
point. Five-year mortality rates for FFS=0, 1, or ≥2 were 9%, 21%, et al. (1991) analysed the relationship between ANCA detection by
or 40%, respectively. The same strategy as previously described for immunofluorescence and clinical evidence of relapse in 70 patients
the 1996 FFS is now used for PAN patients. (GPA and MPA). They found that patients who relapsed invariably
Other criteria, such as those included in the Birmingham had detectable ANCA, that some relapses were preceded by the
Vasculitis Activity Score (BVAS) (Luqmani et  al. 1994; Luqmani reappearance of ANCA, but that many ANCA-positive patients did
et al. 1997), are also used to determine the intensity of treatment to not relapse during the study. Relapses during long-term follow-up
be prescribed, but BVAS has not yet been validated for that purpose were generally associated with high or rising ANCA titres, although
(Bacon et al. 1995). the temporal relationship between clinical relapse and ANCA
changes was not constant. Relapses were very rare in patients with
Recommendations for low or undetectable ANCA.
specific manifestations
Supportive care, prevention of opportunistic infections, such as Corticosteroids
Pneumocystis jiroveci pneumonia, and physical therapy are all impor- Corticosteroids (CS) are prescribed to all patients with MPA, and
tant parts of the therapeutic regimen for patients with MPA, as for high doses may be useful initially. Because of its rapid action and rel-
those with PAN (see Chapter 25). The clinical presentation of ful- ative safety, methylprednisolone pulses (usually 15 mg/kg i.v. over 60
minating MPA is usually that of pulmonary–renal failure and, more min repeated at 24-h intervals for 1–3 days) has become widely used
rarely, cerebral or gastrointestinal involvement. Deterioration of at treatment onset for severe disease (Guillevin et al. 1990), especially
renal function often necessitates haemodialysis. Treatment of mas- in the presence of life-threatening organ involvement or the exten-
sive alveolar haemorrhage requires immediate fluid resuscitation, sion phase of mononeuropathy multiplex. The dose of pulse methyl-
with haemodynamic and respiratory support. In the case of gastroin- prednisolone is empirical and doses lower than 1000 mg may be as
testinal involvement with constant abdominal pain despite medical effective. Oral CS are given at the dose of 1 mg/kg/day of prednisone
treatment, an exploratory surgical procedure should be performed or its equivalent of methylprednisolone, with dose tapering accord-
to identify and treat possible bowel perforation. For the same groups ing to the European vasculitis (EUVAS) study group (Table 26.3).
of patients, it seems reasonable to administer drugs i.v. to circum- This regimen is effective and may control MPA, without the addition
vent possible impaired drug absorption. The prognosis of fulminat- of cytotoxic agents, in patients without poor-prognosis factors.
ing MPA is poor. Savage et al. (1985) reported on 34 MPA patients,
whose actuarial survival and kidney survival at 5-year follow-up were, Cyclophosphamide
respectively, 65 and 55%. Two-thirds of the deaths were due to active CYC should not be systematically prescribed as first-line treatment
vasculitis complicated by renal failure, lung haemorrhage, or to treat- to all MPA patients. Factors influencing the decision to use this drug
ment side-effects. Age over 50 and plasma creatinine >500 µmol/l include the anatomical location of the involvement, its severity, and
(5.7 mg/dl) portend a poor prognosis (Gordon et al. 1993). the intensity of disease activity, as outlined in Section Treatment
principles. Although oral CYC (2 mg/kg/day or less), in combina-
Treatment principles tion with CS is effective for the treatment of vasculitis, it has a low
There is a tendency to consider the treatment of MPA with therapeutic/toxic index. Major side-effects associated with CYC,
poor-prognosis factor(s) and GPA together, based on their shared such as infections, and attempts to decrease their related morbidity,
pathogenic mechanisms and the presence of ANCA, and their such as protocols using intermittent i.v. pulses, are detailed in our
risks of relapses. When no factors of poor prognosis are present, accompanying PAN chapter (Chapter 25).
therphy for MPA is similar to that for PAN (Hoffman et al. 1992; The other recommended regimen is based on initial CYC doses,
Gayraud et al. 1997). Indeed, it has become obvious that MPA with- varying from 0.5 to 0.7 g/m2 at 2-week intervals initially (days 1,
out poor-prognosis criteria does not require the systematic use of 15, and 30), then every 3 weeks, as for GPA, or monthly, depending
CHAPTER 26 microscopic polyangiitis 357

Table 26.3 EUVAS oral corticosteroid-dosing regimen for proved to be even more effective at inducing disease remission for
ANCA-associated vasculitides (in this setting, corticosteroids are those patients enrolled at the time of a relapse (Stone et al. 2010).
combined with an immunosuppressant) Recommendations for RTX use to treat ANCA-associated vascu-
litides were elaborated by some authors (Guerry et al. 2012). First,
Time from treatment onset Prednisone dose RTX was proposed as an alternative to CYC remission-induction
onset 1 mg/kg/day therapy for previously untreated ANCA-associated vasculitides.
Moreover, the authors considered that, in this context, RTX should
1st week 0.75 mg/kg/day be preferred when it would be advisable to avoid CYC because of its
2nd week 0.5 mg/kg/day high gonadal toxicity and carcinogenicity, or an ongoing infection.
4th week 0.4 mg/kg/day Second, RTX was considered effective therapy against refractory/
relapsing ANCA-associated vasculitides.
7th week 0.3 mg/kg/day
10th week 0.28 mg/kg/daya Maintenance therapy for MPA with
poor-prognosis factor(s)
13th week 0.25 mg/kg/daya
Azathioprine (2 mg/kg/day) (Jayne et  al. 2003), methotrexate
3rd month 15 mg/daya
(0.3 mg/kg/week) (de Groot et al. 2005) and several other cyto-
4th month 12.5 mg/daya toxic agents, such as mycophenolate mofetil (starting at 2000
5th month 10 mg/day mg/day), have been tried in MPA (Nowack et al. 1999). They are
now recommended for maintenance therapy, after remission has
6th month 7.5 mg/day
been achieved in MPA patients with poor-prognosis factors, as
12th month 5 mg/day for GPA.
18th month 2.5 mg/day The safety and efficacy of azathioprine and methotrexate for
12 months were compared in the controlled randomized WEGENT
24th month 0
trial on MPA or GPA patients who entered remission with intrave-
a For the thinner patients, minimum dose for the first 3 months is 10 mg/day, which should nous CYC and CS (Pagnoux et al. 2008). In that trial, the two agents
then be maintained until the 5th month. appeared to be comparable alternatives for maintenance therapy in
EUVAS, European Vasculitis Society; ANCA, antineutrophil cytoplasm antibody. MPA or GPA patients after initial remission. In contrast, another
randomized trial compared the effects of mycophenolate mofetil
versus azathioprine for relapse prevention (Hiemstra et al. 2010).
on patient’s general condition, until remission is obtained, followed Relapses were more common in the mycophenolate mofetil group
by maintenance therapy (Nachman et al. 1996; Jayne et al. 2003). (42/76 patients) than the azathioprine group (30/80 patients), with
The inability of CYC to prevent relapse has been disappointing. an unadjusted hazard ratio for mycophenolate mofetil of 1.69 (95%
The duration of CYC treatment and the total amount received did confidence interval (CI), 1.06–2.70; P=0.03). Severe adverse events
not differ between patients with vasculitis who relapsed and those did not differ significantly between groups. The results of that trial
who did not. Serra et al. (1984) described 18 MPA patients who had demonstrated that, among MPA or GPA patients, mycophenolate
chronically active disease, which they called smouldering vasculitis. mofetil was less effective than azathioprine for maintaining disease
When patients fail to respond to pulse CYC, oral CYC has been remission.
successfully introduced to control disease activity or treat relapses Use of these cytotoxic agents is not recommended as first-line
within the first 6 months of treatment (Généreau et al. 1994); how- therapy for MPA with poor-prognosis factor(s), for which CYC is
ever, combined CS and CYC therapy should not exceed 1 year. mandatory.
The randomized CYCLOPS trial compared pulse versus daily oral Systematic RTX is also being evaluated as maintenance therapy
CYC for remission induction in patients with ANCA-associated for MPA or GPA patients, who entered remission taking conven-
vasculitis (de Groot et  al. 2009). Their results showed that pulse tional immunosuppressants or RTX. The results of retrospective
CYC induced remission of ANCA-associated vasculitis as well as studies suggested that RTX maintenance therapy was well tolerated
daily oral intake with a lower cumulative CYC dose and caused and effective, but did not completely prevent relapses or persistent
fewer cases of leukopenia. ‘grumbling’ disease (Roubaud-Baudron et al. 2012). The results of a
prospective, randomized, controlled trial of RTX versus azathioprine
Rituximab to maintain ANCA-associated vasculitis remission were recently
Since 2001, numerous uncontrolled observations reporting the effi- reported (MAINRITSAN trial). Once remission was obtained with
cacy of the chimeric monoclonal anti-CD20 antibody, rituximab a conventional regimen, patients with newly diagnosed or relaps-
(RTX), against MPA, GPA, and EGPA have sparked enthusiasm ing ANCA-associated vasculitis were randomly assigned to receive
and hope that targeted B-cell therapy might cure ANCA-associated a 500-mg RTX infusion on days 1 and 15, then every 6 months for a
vasculitides. In 2010, two randomized clinical trials, RTX in total of five infusions over 18 months, or azathioprine for 22 months
ANCA-associated vasculitis (RAVE) (Stone et al. 2010) and RTX at the initial dose of 2 mg/kg/day. Among the 114 patients participat-
versus CYC for ANCA-associated vasculitis (RITUXVAS) (Jones ing in the study (59 in the azathioprine arm, 55 in the RTX arm), 86
et  al. 2010), provided the first controlled evidence that, at 6 or had GPA, 23 MPA, and five kidney-limited diseases. For the patients
12  months of follow-up, respectively, RTX was as effective and who had completed their follow-up, major relapses had occurred
safe as conventional immunosuppressive therapy (CYC) to control in 15.7% patients, including 3.6% in the RTX arm and 27.1% in
active MPA and GPA. In a subgroup analysis of RAVE data, RTX the azathioprine arm. The results of that study demonstrated that
358 SECTION 5 vasculitic diseases and syndromes and related disorders

500 mg of RTX every 6 months were superior to azathioprine to renal involvement. Annals of Allergy Asthma and Immunology, 93,
maintain ANCA-associated vasculitis remission. 398–401.
Bartolucci, P., Ramanoelina, J., Cohen, P., et  al. (2002). Efficacy of the
Plasma exchanges anti-TNF-alpha antibody infliximab against refractory systemic vascu-
litides: an open pilot study on 10 patients. Rheumatology (Oxford), 41,
Plasma exchanges can improve renal outcome, but not patient 1126–32.
survival, when kidney involvement is severe, defined as oliguria Bloch, D.A., Michel, B.A., Hunder, G.G., et al. (1990). The American College
(<400 ml/24 h) or requiring dialysis or with serum creatinine > of Rheumatology 1990 criteria for the classification of vasculitis. Patients
500 µmol/l (5.7 mg/dl). A typical regimen includes exchanges of and methods. Arthritis and Rheumatism, 33, 1068–73.
60 ml/kg of body weight with 4% albumin-replacement fluid three Booth, A.D., Jefferson, H.J., Ayliffe, W., Andrews, P.A., and Jayne, D.R. (2002).
times per week for 2–3 weeks, then twice per week for 3 weeks, then Safety and efficacy of TNFalpha blockade in relapsing vasculitis. Annals
once per week for 1–3 weeks. The efficacy of this regimen was dem- of the Rheumatic Diseases, 61, 559.
onstrated by the definitive results of the EUVAS MEPEX (methyl- Casselman, B.L., Kilgore, K.S., Miller, B.F., and Warren, J.S. (1995). Antibodies
to neutrophil cytoplasmic antigens induce monocyte chemoattractant
prednisolone or plasma exchange for severe renal vasculitis) (Jayne protein-1 secretion from human monocytes. Journal of Laboratory and
et al. 2007). Plasma exchanges may also be beneficial for patients Clinical Medicine, 126, 495–502.
with alveolar haemorrhage, as for Goodpasture’s syndrome, which Caster, J.C., Shetlar, D..J, Pappolla, M.A., and Yee, R.W. (1996). Microscopic pol-
is also due to pulmonary capillaritis (Simpson et al. 1982; Levy et al. yangiitis with ocular involvement. Archives of Ophthalmology, 114, 346–8.
2001; Guillevin and Pagnoux 2003; Tesar et al. 2003). Chan, T.M. and Cheng, I.K. (1996). A prospective study on anti-endothelial
cell antibodies in patients with systemic lupus erythematosus. Clinical
Other treatments Immunology and Immunopathology, 78, 41–6.
The use of i.v. immunoglobulins (IVIg) to treat systemic vasculi- Chanseaud, Y., Pena-Lefebvre, P.G., Guilpain, P., et al. (2003). IgM and IgG
autoantibodies from microscopic polyangiitis patients but not those
tis has been stimulated by their success in Kawasaki’s disease, in
with other small- and medium-sized vessel vasculitides recognize multi-
which IVIg prevented the development of coronary artery aneu- ple endothelial cell antigens. Clinical Immunology, 109, 165–78.
rysms (Newburger et al. 1991). IVIg have been used to treat GPA Csernok, E., Muller, A., and Gross, W.L. (1999). Immunopathology of
and MPA, with some good results, often with sustained benefit and ANCA-associated vasculitis. Internal Medicine, 38, 759–65.
reduced requirement for immunosuppression (Jayne et  al. 1991; D’Agati, V., Chander, P., Nash, M., and Mancilla-Jimenez, R. (1986).
Jayne and Lockwood 1996). However, there are some discrepancies Idiopathic microscopic polyarteritis nodosa:  ultrastructural observa-
in published studies. IVIg were administered to 15 patients with tions on the renal vascular and glomerular lesions. American Journal of
ANCA-associated systemic vasculitides who were poor responders Kidney Diseases, 7, 95–110.
to conventional therapy (Jayne and Lockwood 1996) and their out- Davson, J., Ball, J., and Platt, R. (1948). Kidney in periarteritis nodosa.
Quarterly Journal of Medicine, 17, 175–202.
comes were less promising: only 40% of them benefited from IVIg
de Groot, K., Harper, L., Jayne, D.R., et  al. (2009). Pulse versus daily oral
and complete remission of disease activity was not obtained. cyclophosphamide for induction of remission in antineutrophil cyto-
In contrast, IVIg were highly effective in our prospective trial on plasmic antibody-associated vasculitis:  a randomized trial. Annals of
relapsing ANCA-associated vasculitis patients. All 20 patients were Internal Medicine, 150, 670–80.
initial responders to IVIg therapy, but IVIg had induced complete de Groot, K., Rasmussen, N., Bacon, P.A., et al. (2005). Randomized trial of
remissions of relapsed ANCA-associated vasculitides in only 13/20 cyclophosphamide versus methotrexate for induction of remission in
patients at month 9, the endpoint evaluation. Because of their good early systemic antineutrophil cytoplasmic antibody-associated vasculi-
safety and tolerance profiles, IVIg were proposed as another option tis. Arthritis and Rheumatism, 52, 2461–9.
in the therapeutic strategy, along with other drugs used to treat Falk, R.J., Terrell, R.S., Charles, L..A, and Jennette, J.C. (1990a).
Anti-neutrophil cytoplasmic autoantibodies induce neutrophils to
relapses of GPA or MPA (Martinez et al. 2008).
degranulate and produce oxygen radicals in vitro. Proceedings of the
Monoclonal antibodies that target proinflammatory cytokine(s), National Academy of Science U S A, 87, 4115–9.
such as anti-TNF-α (e.g. infliximab, etanercept) (Bartolucci et al. Falk, R.J., Hogan, S., Carey, T.S., and Jennette, J.C. (1990b). Clinical course
2002; Booth et  al. 2002), or T cells (Lockwood et  al. 1993)  offer of anti-neutrophil cytoplasmic autoantibody-associated glomerulone-
alternatives to conventional immunosuppressive drugs and have phritis and systemic vasculitis. The Glomerular Disease Collaborative
been used to treat systemic vasculitis patients, who obtained sub- Network. Annals of Internal Medicine, 113, 656–63.
stantial and sustained benefit. Prospective studies are ongoing to Gaskin, G., Clutterbuck, E.J., and Pusey, C.D. (1991). Renal disease in the
define their exact places in the armamentarium for treating MPA. Churg–Strauss syndrome. Diagnosis, management and outcome.
Contributions to Nephrology, 94, 58–65.
At present, all of these biologicals are mostly prescribed for refrac-
Gayraud, M. et al. (2001). Arthritis Rheum. 44, 666–75.
tory forms of the disease or to patients who had suffered multiple
Gayraud, M., Guillevin, L., Cohen, P., et  al. (1997). Treatment of
relapses despite conventional therapies. good-prognosis polyarteritis nodosa and Churg–Strauss syn-
drome: comparison of steroids and oral or pulse cyclophosphamide in
References 25 patients. French Cooperative Study Group for Vasculitides. British
Adu, D., Howie, A.J., Scott, D.G., Bacon, P.A., McGonigle, R.J., and Micheal, Journal of Rheumatology, 36, 1290–7.
J. (1987). Polyarteritis and the kidney. QJM, 62, 221–37. Généreau, T., Lortholary, O., Leclerq, P., et al. (1994). Treatment of systemic
Bacon, P.A., Moots, R.J., Exley, A., Luqmani, R., and Rasmussen, N. (1995). vasculitis with cyclophosphamide and steroids: daily oral low-dose cyclo-
VITAL (Vasculitis Integrated Assessment Log) assessment of vasculitis. phosphamide administration after failure of a pulse intravenous high-dose
Clinical and Experimental Rheumatology, 13, 275–8. regimen in four patients. British Journal of Rheumatology, 33, 959–62.
Bansal, P.J. and Tobin, M.C. (2004). Neonatal microscopic polyangiitis sec- Généreau, T., Lortholary, O., Pottier, M.A., et  al. (1999). Temporal artery
ondary to transfer of maternal myeloperoxidase-antineutrophil cyto- biopsy:  a diagnostic tool for systemic necrotizing vasculitis. French
plasmic antibody resulting in neonatal pulmonary hemorrhage and Vasculitis Study Group. Arthritis and Rheumatism, 42, 2674–81.
CHAPTER 26 microscopic polyangiitis 359

Gilligan, H.M., Bredy, B., Brady, H.R., et al. (1996). Antineutrophil cytoplas- Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., et  al. (1992). Wegener granulo-
mic autoantibodies interact with primary granule constituents on the matosis: an analysis of 158 patients. Annals of Internal Medicine, 116,
surface of apoptotic neutrophils in the absence of neutrophil priming. 488–98.
Journal of Experimental Medicine, 184, 2231–41. Hogan, S.L., Nachman, P.H., Wilkman, A.S., Jennette, J.C., and Falk, R.J.
Godeau, B., Coutant-Perronne, V., Lê Thi Huong, D., et  al. (1994). (1996). Prognostic markers in patients with antineutrophil cytoplasmic
Pneumocystis carinii pneumonia in the course of connective tissue dis- autoantibody-associated microscopic polyangiitis and glomerulone-
ease: report of 34 cases. Journal of Rheumatology, 21, 246–51. phritis. Journal of the American Society of Nephrology, 7, 23–32.
Gordon, M., Luqmani, R.A., Adu, D., et al. (1993). Relapses in patients with a Homas, P.B., David-Bajar, K.M., Fitzpatrick, J.E., West, S.G., and Tribelhorn,
systemic vasculitis. QJM, 86, 779–89. D.R. (1992). Microscopic polyarteritis. Report of a case with cutane-
Guerry, M.J., Brogan, P., Bruce, I.N., et al. (2012). Recommendations for the ous involvement and antimyeloperoxidase antibodies. Archives of
use of rituximab in anti-neutrophil cytoplasm antibody-associated vas- Dermatology, 128, 1223–8.
culitis. Rheumatology (Oxford), 51, 634–43. Homma, S., Matsushita, H., and Nakata, K. (2004). Pulmonary fibrosis in
Guillevin, L., Durand-Gasselin, B., Cevallos, R., et al. (1999). Microscopic myeloperoxidase antineutrophil cytoplasmic antibody-associated vas-
polyangiitis:  clinical and laboratory findings in eighty-five patients. culitides. Respirology, 9, 190–6.
Arthritis and Rheumatism, 42, 421–30. Huugen, D., Xiao, H., van Esch, A., et  al. (2005). Aggravation of
Guillevin, L., Lhote, F., Amouroux, J., Ghérardi, R., Callard, P., and Casassus, anti-myeloperoxidase antibody-induced glomerulonephritis by bacte-
P. (1996a). Antineutrophil cytoplasmic antibodies, abnormal angio- rial lipopolysaccharide: role of tumor necrosis factor-alpha. American
grams and pathological findings in polyarteritis nodosa and Churg– Journal of Pathology, 167, 47–58.
Strauss syndrome: indications for the classification of vasculitides of Jarrousse, B., Guillevin, L., Bindi, P., et  al. (1993). Increased risk of
the polyarteritis nodosa group. British Journal of Rheumatology, 35, Pneumocystis carinii pneumonia in patients with Wegener’s granulo-
958–64. matosis. Clinical and Experimental Rheumatology, 11, 615–21.
Guillevin, L., Lhote, F., Gayraud, M., et al. (1996b). Prognostic factors in pol- Jayne, D.R., Davies, M.J., Fox, C.J., Black, C.M., and Lockwood, C.M. (1991).
yarteritis nodosa and Churg–Strauss syndrome. A prospective study in Treatment of systemic vasculitis with pooled intravenous immunoglob-
342 patients. Medicine (Baltimore), 75, 17–28. ulin. Lancet, 337, 1137–9.
Guillevin, L. and Pagnoux, C. (2003). Indications of plasma exchanges for Jayne, D.R., Gaskin, G., Rasmussen, N., et al. (2007). Randomized trial of
systemic vasculitides. Therapeutic Apheresis and Dialysis, 7, 155–60. plasma exchange or high-dosage methylprednisolone as adjunctive
Guillevin, L., Pagnoux, C., Seror, R., et  al. (2011). The Five-Factor Score therapy for severe renal vasculitis. Journal of the American Society of
revisited: assessment of prognoses of systemic necrotizing vasculitides Nephrology, 18, 2180–8.
based on the French Vasculitis Study Group (FVSG) cohort. Medicine Jayne, D.R. and Lockwood, C.M. (1996). Intravenous immunoglobulin as
(Baltimore), 90, 19–27. sole therapy for systemic vasculitis. British Journal of Rheumatology, 35,
Guillevin, L., Rosser, J., Cacoub, P., Mousson, C., and Jarrousse, B. (1990). 1150–3.
Methylprednisolone in the treatment of Wegener’s granulomatosis, pol- Jayne, D., Rasmussen, N., Andrassy, K., et al. (2003). A randomized trial
yarteritis nodosa and Churg-Strauss angiitis. APMIS Supplementum, of maintenance therapy for vasculitis associated with antineutro-
19, 52–3. phil cytoplasmic autoantibodies. New England Journal of Medicine,
Guilpain, P., Chéreau, C., Goulvestre, C., et al. (2011). The oxidation induced 349, 36–44.
by antimyeloperoxidase antibodies triggers fibrosis in microscopic poly- Jennette, J.C., Falk, R.J., Andrassy, K., et  al. (1994). Nomenclature of sys-
angiitis. European Respiratory Journal, 37, 1503–13. temic vasculitides. Proposal of an international consensus conference.
Guilpain, P., Servettaz, A., Goulvestre, C., et al. (2007). Pathogenic effects of Arthritis and Rheumatism, 37, 187–92.
antimyeloperoxidase antibodies in patients with microscopic polyangii- Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F.,
tis. Arthritis and Rheumatism, 56, 2455–63. Flores-Suarez, L.F., Gross, W.L., Guillevin, L., Hagen, E.C., Hoffman,
Handa, R., Wali, J.P., Gupta, S.D., et al. (2001). Classical polyarteritis nodosa G.S., Jayne, D.R., Kallenberg, C.G.M., Lamprecht, P., Langford, C.A.,
and microscopic polyangiitis: a clinicopathologic study. Journal of the Luqmani, R.A., Mahr, A.D., Matteson, E.L., Merkel, P.A., Ozen, S.,
Association of Physicians of India, 49, 314–9. Pusey, C.D., Rasmussen, N., Rees, A.J., Scott, D.G.I., Specks, U., Stone,
Hattori, N., Mori, K., Misu, K., Koike, H., Ichimura, M., and Sobue, G. J.H., Takahashi, K., and Watts, R.A. (2013). 2012 Revised International
(2002). Mortality and morbidity in peripheral neuropathy-associated Chapel Hill Consensus Conference Nomenclature of Vasculitides.
Churg–Strauss syndrome and microscopic polyangiitis. Journal of Arthritis and Rheumatism, 65, 1–11.
Rheumatology, 29, 1408–14. Jennette, J.C., Wilkman, A.S., and Falk, R.J. (1989). Anti-neutrophil cyto-
Hauschild, S., Csernok, E., Schmitt, W.H., and Gross, W.L. (1994). plasmic autoantibody-associated glomerulonephritis and vasculitis.
Antineutrophil cytoplasmic antibodies in systemic polyarteritis nodosa American Journal of Pathology, 135, 921–30.
with and without hepatitis B virus infection and Churg–Strauss syn- Jones, R.B., Tervaert, J.W., Hauser, T., et al. (2010). Rituximab versus cyclo-
drome. Journal of Rheumatology, 21, 1173–4. phosphamide in ANCA-associated renal vasculitis. New England Journal
Heeringa, P., Brouwer, E., Tervaert, J.W., Weening, J.J., and Kallenberg, C.G. of Medicine, 363, 211–20.
(1998). Animal models of anti-neutrophil cytoplasmic antibody associ- Kapitsinou, P.P., Ioannidis, J.P., Boletis, J.N., et al. (2003). Clinicopathologic
ated vasculitis. Kidney International, 53, 253–63. predictors of death and ESRD in patients with pauci-immune necrotiz-
Hervier, B., Pagnoux, C., Agard, C., et al. (2009). Pulmonary fibrosis associ- ing glomerulonephritis. American Journal of Kidney Diseases, 41, 29–37.
ated with ANCA-positive vasculitides. Retrospective study of 12 cases Kettritz, R., Jennette, J.C., and Falk, R.J. (1997). Crosslinking of
and review of the literature. Annals of the Rheumatic Diseases, 68, 404–7. ANCA-antigens stimulates superoxide release by human neutrophils.
Hiemstra, T.F., Walsh, M., Mahr, A., et al. (2010). Mycophenolate mofetil vs Journal of the American Society of Nephrology, 8, 386–94.
azathioprine for remission maintenance in antineutrophil cytoplasmic King, W.J., Brooks, C.J., Holder, R., Hughes, P., Adu, D., and Savage, C.O.
antibody-associated vasculitis:  a randomized controlled trial. JAMA, (1998). T lymphocyte responses to anti-neutrophil cytoplasmic autoan-
304, 2381–8. tibody (ANCA) antigens are present in patients with ANCA-associated
Hiromura, K., Nojima, Y., Kitahara, T., et  al. (2000). Four cases of systemic vasculitis and persist during disease remission. Clinical and
anti-myeloperoxidase antibody-related rapidly progressive glomerulo- Experimental Immunology, 112, 539–46.
nephritis during the course of idiopathic pulmonary fibrosis. Clinical Kono, H., Inokuma, S., Nakayama, H., and Yamazaki, J. (2000).
Nephrology, 53, 384–9. Pachymeningitis in microscopic polyangiitis (MPA): a case report and
360 SECTION 5 vasculitic diseases and syndromes and related disorders

a review of central nervous system involvement in MPA. Clinical and Pfister, H., Ollert, M., Frohlich, L.F., et al. (2004). Antineutrophil cytoplasmic
Experimental Rheumatology, 18, 397–400. autoantibodies against the murine homolog of proteinase 3 (Wegener
Lane, S.E., Watts, R.A., Shepstone, L., and Scott, D.G. (2005). Primary sys- autoantigen) are pathogenic in vivo. Blood, 104, 1411–8.
temic vasculitis: clinical features and mortality. QJM, 98, 97–111. Popa, E.R., Franssen, C.F., Limburg, P.C., Huitema, M.G., Kallenberg,
Levy, J.B., Turner, A.N., Rees, A.J., and Pusey, C.D. (2001). Long-term out- C.G., and Tervaert, J.W. (2002). In vitro cytokine production and
come of anti-glomerular basement membrane antibody disease treated proliferation of T cells from patients with antiproteinase 3- and
with plasma exchange and immunosuppression. Annals of Internal antimyeloperoxidase-associated vasculitis, in response to proteinase 3
Medicine, 134, 1033–42. and myeloperoxidase. Arthritis and Rheumatism, 46, 1894–904.
Lhote, F., Cohen, P., and Guillevin, L. (1998). Polyarteritis nodosa, micro- Praprotnik, S., Rozman, B., Blank, M., and Shoenfeld, Y. (2000). Pathogenic
scopic polyangiitis and Churg–Strauss syndrome. Lupus, 7, 238–58. role of anti-endothelial cell antibodies in systemic vasculitis. Wiener
Lie, J.T. (1992). Retroperitoneal polyarteritis nodosa presenting as ureteral Klinische Wochenschrift, 112, 660–4.
obstruction. Journal of Rheumatology, 19, 1628–31. Ralston, D.R., Marsh, C.B., Lowe, M.P., and Wewers, M.D. (1997).
Lightfoot, R.W. Jr, Michel, B.A., Bloch, D.A., et  al. (1990). The American Antineutrophil cytoplasmic antibodies induce monocyte IL-8 release.
College of Rheumatology 1990 criteria for the classification of polyarte- Role of surface proteinase-3, alpha1-antitrypsin, and Fcgamma recep-
ritis nodosa. Arthritis and Rheumatism, 33, 1088–93. tors. Journal of Clinical Investigation, 100, 1416–24.
Little, M.A., Bhangal, G., Smyth, C.L., et  al. (2006). Therapeutic effect of Reumaux, D., Vossebeld, P.J., Roos, D., and Verhoeven, A.J. (1995). Effect of
anti-TNF-alpha antibodies in an experimental model of anti-neutrophil tumor necrosis factor-induced integrin activation on Fc gamma recep-
cytoplasm antibody-associated systemic vasculitis. Journal of the tor II-mediated signal transduction: relevance for activation of neutro-
American Society of Nephrology, 17, 160–9. phils by anti-proteinase 3 or anti-myeloperoxidase antibodies. Blood, 86,
Lockwood, C.M., Thiru, S., Isaacs, J.D., Hale, G., and Waldmann, H. (1993). 3189–95.
Long-term remission of intractable systemic vasculitis with monoclonal Rodgers, H., Guthrie, J.A., Brownjohn, A.M., and Turney, J.H. (1989).
antibody therapy. Lancet, 341, 1620–2. Microscopic polyarteritis: clinical features and treatment. Postgraduate
Luqmani, R.A., Bacon, P.A., Moots, R.J., et al. (1994). Birmingham Vasculitis Medical Journal, 65, 515–8.
Activity Score (BVAS) in systemic necrotizing vasculitis. QJM, 87, 671–8. Roubaud-Baudron, C., Pagnoux, C., Méaux-Ruault, N., et  al. (2012).
Luqmani, R.A., Exley, A.R., Kitas, G.D., and Bacon, P.A. (1997). Disease Rituximab maintenance therapy for granulomatosis with polyangiitis
assessment and management of the vasculitides. Baillière's Clinical and microscopic polyangiitis. Journal of Rheumatology, 39, 125–30.
Rheumatology, 11, 423–46. Sablé-Fourtassou, R., Cohen, P., Mahr, A., et al. (2005). Antineutrophil cyto-
Lyons, P.A., Rayner, T.F., Trivedi, S., et al. (2012). Genetically distinct subsets plasmic antibodies and the Churg–Strauss syndrome. Annals of Internal
within ANCA-associated vasculitis. New England Journal of Medicine, Medicine, 143, 632–8.
19, 214–23. Salojin, K.V., Le Tonqueze, M., Saraux, A., et al. (1997). Antiendothelial cell
Mahr, A., Guillevin, L., Poissonnet, M., and Aymé, S. (2004). Prevalences of antibodies:  useful markers of systemic sclerosis. American Journal of
polyarteritis nodosa, microscopic polyangiitis, Wegener’s granulomato- Medicine, 102, 178–85.
sis, and Churg-Strauss syndrome in a French urban multiethnic popula- Savage, C.O., Winearls, C.G., Evans, D.J., Rees, A.J., and Lockwood, C.M.
tion in 2000: a capture-recapture estimate. Arthritis and Rheumatism, (1985). Microscopic polyarteritis: presentation, pathology and progno-
51, 92–9. sis. QJM, 56, 467–83.
Martinez, V., Cohen, P., Pagnoux, C., et al. (2008). Intravenous immunoglobulins Schlieben, D.J., Korbet, S.M., Kimura, R.E., Schwartz, M.M., and Lewis,
for relapses of systemic vasculitides associated with antineutrophil cyto- E.J. (2005). Pulmonary-renal syndrome in a newborn with placen-
plasmic autoantibodies: results of a multicenter, prospective, open-label tal transmission of ANCAs. American Journal of Kidney Diseases, 45,
study of twenty-two patients. Arthritis and Rheumatism, 58, 308–17. 758–61.
Mihara, M., Hayasaka, S., Watanabe, K., Kitagawa, K., and Hayasaka, Y. Seishima, M., Oyama, Z., and Oda, M. (2004). Skin eruptions associated with
(2005). Ocular manifestations in patients with microscopic polyangiitis. microscopic polyangiitis. European Journal ofDermatology, 14, 255–8.
European Journal of Ophthalmology, 15, 138–42. Serra, A., Cameron, J.S., Turner, D.R., et al. (1984). Vasculitis affecting the
Mulder, A.H., Broekroelofs, J., Horst, G., Limburg, P.C., Nelis, G.F., and kidney: presentation, histopathology and long-term outcome. QJM, 53,
Kallenberg, C.G. (1994). Anti-neutrophil cytoplasmic antibodies 181–207.
(ANCA) in inflammatory bowel disease: characterization and clinical Simpson, I.J., Doak, P.B., Williams, L.C., et al. (1982). Plasma exchange in
correlates. Clinical and Experimental Immunology, 95, 490–7. Goodpasture’s syndrome. American Journal of Nephrology, 2, 301–11.
Nachman, P.H., Hogan, S.L., Jennette, J.C., and Falk, R.J. (1996). Sinico, R.A., Di Toma, L., Maggiore, U., et al. (2005). Prevalence and clinical
Treatment response and relapse in antineutrophil cytoplasmic significance of antineutrophil cytoplasmic antibodies in Churg–Strauss
autoantibody-associated microscopic polyangiitis and glomerulone- syndrome. Arthritis and Rheumatism, 52, 2926–35.
phritis. Journal of the American Society of Nephrology, 7, 33–9. Souid, M., Terki, N.H., Nochy, D., and Hillion, D. (2001). Myeloperoxidase
Newburger, J.W., Takahashi, M., Beiser, A.S., et al. (1991). A single intrave- anti-neutrophil cytoplasmic autoantibodies (MPO-ANCA)-related rap-
nous infusion of gamma globulin as compared with four infusions in idly progressive glomerulonephritis (RPGN) and pulmonary fibrosis
the treatment of acute Kawasaki syndrome. New England Journal of (PF) with dissociated evolution. Clinical Nephrology, 55, 337–8.
Medicine, 324, 1633–9. Stone, J.H., Merkel, P.A., Spiera, R., et al. (2010). Rituximab versus cyclo-
Nowack, R., Gobel, U., Klooker, P., Hergesell, O., Andrassy, K., and van der phosphamide for ANCA-associated vasculitis. New England Journal of
Woude, F.J. (1999). Mycophenolate mofetil for maintenance therapy of Medicine, 363, 221–32.
Wegener’s granulomatosis and microscopic polyangiitis: a pilot study in Tesar, V., Rihova, Z., Jancova, E., Rysava, R., and Merta, M. (2003). Current
11 patients with renal involvement. Journal of the American Society of treatment strategies in ANCA-positive renal vasculitis-lessons from
Nephrology, 10, 1965–71. European randomized trials. Nephrology Dialysis Transplantation, 18
Pagnoux, C., Mahr, A., Hamidou, M.A., et al. (2008). Azathioprine or meth- (Suppl. 5), 2–4.
otrexate maintenance for ANCA-associated vasculitis. New England Tidman, M., Olander, R., Svalander, C., and Danielsson, D. (1998). Patients
Journal of Medicine, 359, 2790–803. hospitalized because of small vessel vasculitides with renal involvement
Penas, P.F., Porras, J.I., Fraga, J., Bernis, C., Sarria, C., and Dauden, E. (1996). in the period 1975-95: organ involvement, anti-neutrophil cytoplasmic
Microscopic polyangiitis. A systemic vasculitis with a positive P-ANCA. antibodies patterns, seasonal attack rates and fluctuation of annual fre-
British Journal of Dermatology, 134, 542–7. quencies. Journal of Internal Medicine, 244, 133–41.
CHAPTER 26 microscopic polyangiitis 361

Tsai, C.N., Chang, C.M., Chuang, C.H., Jin, Y.T., Liu, M.F., and Wang, C.R. Weidner, S., Neupert, W., Goppelt-Struebe, M., and Rupprecht, H.D. (2001).
(2004). Extended colonic ulcerations in a patient with microscopic poly- Antineutrophil cytoplasmic antibodies induce human monocytes to pro-
angiitis. Annals of the Rheumatic Diseases, 63, 1521–2. duce oxygen radicals in vitro. Arthritis and Rheumatism, 44, 1698–706.
Ueda, S., Matsumoto, M., Ahn, T., et  al. (2001). Microscopic polyangiitis Wohlwill, F. (1923). Ueber die nur mikroskopisch erkennbare Form der
complicated with massive intestinal bleeding. Journal ofGastroenterol- Periarteritis nodosa. Virchows Archiv für Pathologische Anatomie und
ogy, 36, 264–70. Physiologie, 246, 377–411.
van Bommel, E., Brouwers, A., Makkus, A., and van Vliet, A. (2002). Xiao, H., Heeringa, P., Hu, P., et  al. (2002). Antineutrophil cytoplasmic
Retroperitoneal fibrosis and p-ANCA-associated polyarteritis autoantibodies specific for myeloperoxidase cause glomerulonephritis
nodosa: coincidental or common etiology? European Journal ofInternal and vasculitis in mice. Journal of Clinical Investigation, 110, 955–63.
Medicine, 13, 392. Xiao, H., Heeringa, P., Liu, Z., et al. (2005). The role of neutrophils in the
van der Woude, F.J., Rasmussen, N., Lobatto, S., et al. (1985). Autoantibodies induction of glomerulonephritis by anti-myeloperoxidase antibodies.
against neutrophils and monocytes: tool for diagnosis and marker of dis- American Journal of Pathology, 167, 39–45.
ease activity in Wegener’s granulomatosis. Lancet, 1, 425–9. Xiao, H., Schreiber, A., Heeringa, P., et al. (2007). Alternative complement
Wang, L., Thelmo, W.L., and Axiotis, C.A. (2002). Microscopic polyangiitis pathway in the pathogenesis of disease mediated by anti-neutrophil
with massive myocardial necrosis and diffuse pulmonary hemorrhage. cytoplasmic autoantibodies. American Journal of Pathology, 170, 52–64.
Virchows Archiv, 441, 202–4. Zauner, I., Bach, D., Braun, N., et al. (2002). Predictive value of initial his-
Watts, R.A., Jolliffe, V.A., Carruthers, D.M., Lockwood, M., and Scott, D.G. tology and effect of plasmapheresis on long-term prognosis of rapidly
(1996). Effect of classification on the incidence of polyarteritis nodosa progressive glomerulonephritis. American Journal of Kidney Diseases,
and microscopic polyangiitis. Arthritis and Rheumatism, 39, 1208–12. 39, 28–35.
CHAPTER 27

Cutaneous polyarteritis
Angelo L. Gaffo

Introduction published. The condition seems to be less common than its sys-
temic counterpart PAN, which has an incidence in non-endemic
Cutaneous polyarteritis nodosa (CPAN) is a chronic, relapsing, and hepatitis B areas of 6–10 cases per million people per year (Watts
usually benign necrotizing vasculitis of the small and medium-sized et al. 2001). Early European studies describe a relatively equal fre-
arteries of the deep dermis and subcutaneous layers of the skin. In quency between men and women (Borrie 1972; Daoud et al. 1997),
contrast to the better-known and better-understood polyarteritis while those from Japan present a clear female predominance (8:1).
nodosa (PAN), CPAN spares visceral organs. Nevertheless, CPAN CPAN has been well described in children, and even in neonates.
can have systemic manifestations in the form of malaise, fevers, and Adult series describe patients ranging from ages 16 to 81 with
peripheral neuropathy. mean ages at presentation between 36 and 50 years (Borrie 1972;
The first description of polyarteritis nodosa as an independ- Daoud et al. 1997; Ishiguro and Kawashima 2010; Nakamura et al.
ent clinical entity was published by Adolf Kussmaul and Rudolf 2009; Chen 1989).
Maier in 1866, although the condition was first named ‘periarte-
ritis nodosa’(Kussmaul and Maier 1866). In 1903, Ferrari (1903)
and, in 1908, Dickson (1908) proposed a change in the name to Clinical features
‘polyarteritis nodosa’, to emphasize a distinction from conditions The most common cutaneous manifestations of CPAN are subcu-
that affect only the outer layers of the blood vessels, mainly ter- taneous nodules, purpura, inflammatory plaques, cutaneous ulcers,
tiary syphilis. In 1931, Kaj Lindberg described a form of PAN and livedo (Table 27.1). Subcutaneous nodules tend to be the first
limited to the skin with a benign clinical course in two adoles- manifestation of the disease and appear in clusters ranging from a
cent patients, this being recognized as the first report of CPAN few to several dozen (Borrie 1972; Daoud et al. 1997; Diaz-Perez
(Lindberg 1931). Since then, the recognition of CPAN as a dis-
tinct clinical entity remained uncertain for four decades until
Peter Borrie’s report in 1972 of 17 cases of cutaneous polyarteritis
without systemic manifestations even after prolonged follow-up Table 27.1 Clinical and laboratory manifestations of cutaneous
(Borrie 1972). Previous series describing presumed CPAN had polyarteritis nodosa
included cases of PAN with visceral and cutaneous involvement
(Melcer and Venkei 1947), or they had not clearly specified the Frequency (%)
histological diagnosis, leading to misclassification with other Clinical features
cutaneous vasculitides (Miescher 1946). Some reports failed to Cutaneous
mention whether there was systemic involvement as well as the Nodules 60–100
described cutaneous disease (Ruiter 1958). There were also other
Inflammatory plaques 6–12
small clinical series in which authors did not classify CPAN as
Livedo 45–56
an independent condition, but as a subset of PAN with limited
Ulcers 18–48
involvement and benign prognosis (Lyell and Church 1954; Fisher
and Orkin 1964). Since Borrie’s report, other authors have pub- Extracutaneous
lished cases of CPAN with thorough investigations for systemic Peripheral neuropathy 30–65
disease and prolonged follow-up (Diaz-Perez and Winkelmann Myalgias and myositis 27–40
1974; Daoud et al. 1997; Ishiguro and Kawashima 2010; Moreland Arthralgias and arthritis 18–69
and Ball 1990; Chen 1989), reinforcing the concept of CPAN as Fever 20
an entity distinct from PAN. It is important to note, however, that Laboratory
opinions have differed as to whether CPAN is actually an inde- Elevated erythrocyte sedimentation rate 60
pendent entity (Thomas and Black 1983). Elevated C-reactive protein 60
Anaemia 32
Epidemiology Positive lupus anticoagulant 62
Positive titre of IgM antiphosphatidylserine- 72
There is no certain way to establish disease frequencies for CPAN
prothrombin complex (anti-PS/PT)
as no data pertaining to incidence or prevalence have been
364 SECTION 5 vasculitic diseases and syndromes and related disorders

Fig. 27.1 Erythematous subcutaneous nodules in the lower extremity Fig. 27.2 Livedo racemosa rash in the lower extremities of patient with
of a patient with cutaneous polyarteritis nodosa (Courtesy of Marcin cutaneous polyarteritis nodosa. Reprinted from Journal of the American Academy
Trojanowski, MD). of Dermatology, 63; 4. Kawakami, T, Soma, Y, Use of warfarin therapy at a target
international normalized ratio of 3.0 for cutaneous polyarteritis nodosa, 602.
Copyright (2010), with permission from Elsevier.

and Winkelmann 1974). They are often not visible, but when they and vasculitides (Ehrmann 1907). The term livedo racemosa was
are apparent their colour varies from red to purple and their size mostly forgotten from medical literature until a few years back,
ranges from 0.5 to 3 centimetres; the most common location is the with all livedo rashes being called livedo reticularis without dis-
lower extremities (Figure 27.1). A majority of cases have a sym- tinction. This was also true for livedo rashes associated with PAN
metric distribution, but unilateral bouts of nodular lesions are not and CPAN, even though the livedo rash of CPAN is largely consist-
uncommon (Diaz-Perez and Winkelmann 1974). The trunk, upper ent with livedo racemosa. In 40–56% of cases there is a marked
extremities, and mucosal surfaces can also be involved. Small nod- preference for the legs and lower trunk, although it can also involve
ules resolve in a few days, but larger ones may persist for weeks the arms (Figure 27.2) (Borrie 1972; Daoud et al. 1997; Diaz-Perez
(Borrie 1972). Recurrences tend to involve previously affected areas and Winkelmann 1974; Ishiguro and Kawashima 2010; Nakamura
and nodular lesions are often exquisitely painful. Subcutaneous et al. 2009). Livedo almost always occurs in areas where there is
nodules can precede or accompany other manifestations of CPAN, also nodulation (Borrie 1972), but nodulation may be seen inde-
such as inflammatory plaques or lower-extremity ulcerations pendently of livedo. Other less-common cutaneous manifestations
(Chan et al. 2009; Ishiguro and Kawashima 2010). Purpuric lesions include petechiae and gangrenous changes in the skin or distal
are well described, often appearing alongside areas of subcutaneous extremities.
nodules. Inflammatory plaques with surrounding nodularity are Extracutaneous manifestations of CPAN include fever, fatigue,
more common in the trunk and proximal lower extremities than in muscle and joint pain, and peripheral neuropathy (Borrie 1972;
distal extremities (Chan et al. 2009). Ulcers are universally painful Daoud et  al. 1997; Ishiguro and Kawashima 2010). Fever is
and can be deep or superficial, and always appear in association low-grade, intermittent, and seen in approximately 20% of patients
with pre-existing nodules, inflammatory plaques, or livedo (Daoud (Daoud et al. 1997; Diaz-Perez and Winkelmann 1974; Morgan and
et al. 1997). Schwartz 2010). Fatigue is usually mild and self-limited. Arthralgias
In general, livedo rashes have been described in two distinct pat- have been described in 18–69% of patients (Daoud et  al. 1997;
terns: livedo reticularis, a macular, violaceous, net-like pattern in Ishiguro and Kawashima 2010; Nakamura et al. 2009). Severe but
the skin; and livedo racemosa, that differs from livedo reticularis non-erosive, albeit progressive, arthritis with marked disability
in its shape (irregular, broken, coarse, circular segments) and its has been reported (Gottlieb 1978; Smukler and Schumacher 1977;
location (more widespread, involving limbs as well as the trunk Mekori et al. 1984). Joint disease of greater severity and frequency
and buttocks) (Uthman and Khamashta 2006). Livedo reticularis has been reported more often in older, western publications than
is associated with physiological skin changes on exposure to cold in contemporary Japanese series. Myalgias or frank myositis have
while livedo racemosa is mostly associated with ischaemic and been described in 27–40% of patients (Borrie 1972; Diaz-Perez
inflammatory conditions, such as the antiphospholipid syndrome, and Winkelmann 1974; Ishiguro and Kawashima 2010; Nakamura
CHAPTER 27 cutaneous polyarteritis 365

et al. 2009). The severity of the muscle involvement ranges from them, while titres of IgG anti-PS/PT were higher in the group with
mild and transient to severe and disabling and it may persist for inflammatory plaques (Kawakami and Soma 2011a). Both IgM
years. The most common location is the calves, although it also and IgG anti-PS/PT have been reported in larger proportions of
involves the lower back. The histopathology of the muscle involve- patients with livedo racemosa compared with livedo reticularis
ment is similar to that of PAN (Borrie 1972). Resolution of the (Kawakami et al. 2009) and it is possible that most cutaneous vas-
muscle involvement takes months or years. Peripheral neuropathy culitis associated with livedo racemosa indicates the presence of
is a common and well-documented complication of CPAN, which PS/PT antibodies.
has been reported in 30–65% of patients (Borrie 1972; Daoud et al.
1997; Diaz-Perez and Winkelmann 1974; Ishiguro and Kawashima
2010; Nakamura et  al. 2009). The most common location is the Histopathology
lower extremities, although the upper extremities may also be In 1930, Arkin published the histopathological PAN classification
affected. Involvement should correspond to areas affected by skin that is still accepted (Arkin 1930) and his description of histological
lesions; if not, PAN should be suspected (Nakamura et al. 2009). changes in PAN has been widely applied to CPAN, despite the obvi-
Symptoms of peripheral neuropathy in CPAN are numbness, par- ous caveat that none of Arkin’s originally reviewed cases described
esthesias, burning pain, weakness, foot drop, and absent reflexes. cutaneous lesions. It has been observed that CPAN arterial changes
Electrophysiological studies most often reveal axonal sensorimo- occur in smaller-calibre vessels that those in PAN (200–400  µm
tor neuropathies with a mononeuritis multiplex pattern (Chen in diameter small arteries and arterioles in the former versus
1989). Its clinical course ranges from complete recovery to perma- medium-sized arteries in the latter), which might limit the appli-
nent damage with wasting and contractures (Borrie 1972). Other cability of Arkin’s original descriptions (Ishibashi and Chen 2008).
uncommon reported manifestations of CPAN include digital or The main histopathological feature of CPAN is that of a fibrinoid
toe gangrene, Raynaud’s phenomenon, nail lesions, and vascular necrotizing vasculitis of small and medium-sized cutaneous vessels,
occlusions or thrombosis (Bastian 2008). The latter, along with the often with inflammatory infiltrates at the dermal–subdermal junc-
more severe cutaneous manifestations such as severe, unresolving tion, and leukocytoclasis. The stages currently described for CPAN,
or recurrent ulcers should prompt a reconsideration of the diag- which correspond to Arkin’s classical PAN description, include
nosis of CPAN in favour of PAN or other vasculitides, and careful (Ishibashi and Chen 2008): (1) a first acute stage with neutrophilic
follow-up. infiltration and leukocytoclasis around small muscular arteries in
the dermal–subdermal junction but no disruption of the internal
elastic lamina or fibrinoid necrosis (Figure 27.3); (2) a second suba-
Laboratory findings cute stage with fibrinoid necrosis in the intima and fibrin thrombi
Non-specific laboratory abnormalities occur often and include ele- in the vascular lumen. Inflammatory infiltrates are clearly located
vated erythrocyte sedimentation rate (ESR), anaemia, and mild leu- in and around the vessel walls (Figure 27.4); (3) a third reparative
kocytosis (Table 27.1). As opposed to PAN, few cases are reported stage with a decrease in inflammatory infiltrates, occlusion in vas-
in association with positive hepatitis serologies. Rheumatoid fac- cular lumens by fibrin thrombi and intimal proliferation, and neo-
tors and antinuclear antibodies are detected infrequently; the lat- vascularization and (Figure 27.5); and (4) a final, healed stage with
ter are found more often in patients with ulcerative lesions (Daoud replacement of the vascular wall by an acellular scar tissue, very few
et  al. 1997). Persons who are positive for rheumatoid factor and inflammatory infiltrates, and more mature neovascularization. The
antinuclear antibodies should also be followed closely for the devel- inflammatory infiltrates in CPAN are eosinophilic in one-third of
opment of PAN (Chen 1989). Neutrophil cytoplasmic antibodies cases (Daoud et al. 1997) and different histopathological stages can
(ANCA) are not associated with CPAN, although they have been coexist in 50% of cases (Ishibashi and Chen 2008).
detected in a few instances. Despite complement and IgM deposits Direct immune-fluorescence studies (DIF) have been described
in the vessel walls of patients with CPAN (Kawakami and Soma in a group of 33 patients with CPAN (Kawakami and Soma 2011a).
2011a), serum levels of C3 and other complement components Sixty-seven per cent were positive for C3 deposition while 58%
are usually normal (Chen 1989; Daoud et al. 1997; Diaz-Perez and showed IgM deposits. A prior report by the same group described IgG
Winkelmann 1974). deposition in 1/10 cases studied (Kawakami et al. 2007). Complement
An association between CPAN and a specific variant of deposits in vessel walls are hypothesized to play a role in the damage
antiphospholipid antibody has generated interest. IgM antibodies induced by antiphospholipid antibodies, which may play a pathogenic
directed against the antiphosphatidylserine–prothrombin com- role in CPAN (Holers et al. 2002; Kawakami et al. 2007).
plex (anti-PS/PT) were found in 15 of 16 patients with CPAN in In cases of suspected CPAN, tissue biopsies should be deep to
Japan. Titres for IgM anti-PS/PT were significantly higher than in include the dermal–subdermal layers of the skin. An independent
patients with systemic lupus erythematosus or normal controls. sample will be required to perform DIF, which might be critical in
Other antiphospholipid antibodies were absent (antibodies against differentiating cases of CPAN from other cutaneous vasculitides.
beta-2-glycoprotein) or present in smaller amounts. In all patients Finally, involving an experienced pathologist with expertise in vas-
studied, either anti-PS/PT or lupus anticoagulant was present culitis is important as misdiagnosis in reported cases of CPAN has
(Kawakami et al. 2007). More recently, it has been reported that dif- been recognized retrospectively (Chen 2010).
ferent antiphospholipid antibodies appeared to be associated with
different presentations of the disease. For example, IgM anti-PS/PT
was found at higher titres in patients with livedo racemosa than in Differential diagnosis
those without. On the other hand, titres of IgM anti-PS/PT were The differential diagnosis of CPAN includes other vasculitides,
lower in patients with inflammatory plaques than in those without autoimmune diseases, inflammatory conditions, neoplastic
366 SECTION 5 vasculitic diseases and syndromes and related disorders

(a) (b)

Fig. 27.3 First phase of histopathological progression in cutaneous polyarteritis nodosa. (a) Small muscular artery surrounded by an inflammatory infiltrate composed
mainly of neutrophils with evident leukocytoclasis. (b) Special stain shows a preserved internal elastic lamina. Magnification for both images is 40×. Reproduced with
permission from The American Journal of Dermatopathology, 30; 4. Ishibashi, M, Chen, Ko-Ron, A Morphological Study of Evolution of Cutaneous Polyarteritis Nodosa.
© Lippincott Williams and Wilkins, 2008.

manifestations, and infections (Table 27.2). Systemic PAN should and IgA vasculitis (Henoch–Schönlein purpura) can be initially
be strongly considered after a diagnosis of CPAN. Evidence of dis- differentiated from CPAN on clinical grounds given that these
ease in visceral organs, including intestines, kidney, liver, and heart, conditions are less likely to cause subcutaneous nodules or livedo.
should be investigated. Absence of abdominal pain, hypertension, Histopathology might be the final determinant in cases in which
liver test abnormalities, and proteinuria or haematuria favour there is clinical overlap. Antiphospholipid antibody syndrome
CPAN. Vasculitides associated with ANCA, cryoglobulinaemia, might cause livedo, but it lacks nodular lesions or peripheral

(a) (b)

Fig. 27.4 Second phase of histopathological progression in cutaneous polyarteritis nodosa. (a) Inflammatory infiltrates with fibrinoid necrosis (arrows). (b) Special stain
shows fibrinoid necrosis causing elastic lamina disruption (arrows). Magnification for both images is 100×. Reproduced with permission from The American Journal of
Dermatopathology, 30; 4. Ishibashi, M, Chen, Ko-Ron, A Morphological Study of Evolution of Cutaneous Polyarteritis Nodosa. © Lippincott Williams and Wilkins, 2008..
CHAPTER 27 cutaneous polyarteritis 367

(a) (b)

Fig. 27.5 Third phase of histopathological progression in cutaneous polyarteritis nodosa. (a) Marked intimal proliferation, abundant fibronoid necrosis, and luminal
occlusion in affected artery. (b) Special stain shows more advanced elastic lamina disruption with abundant fibrinoid necrosis. Neovascularization is evident in the
periphery of the slide. Magnification for both images is 40×. Reproduced with permission from The American Journal of Dermatopathology, 30; 4. Ishibashi, M, Chen,
Ko-Ron, A Morphological Study of Evolution of Cutaneous Polyarteritis Nodosa. © Lippincott Williams and Wilkins, 2008.

neuropathy. Erythema nodosum, depending on the underlying If the diagnosis of CPAN is supported by histopathology it is
aetiology, can resemble CPAN. Biopsies should confirm the pres- important to exclude PAN. This distinction should be continu-
ence of septal paniculitis without vasculitis in erythema nodosum ously queried on follow-up. CPAN is uncommonly described in the
(Ter Poorten and Thiers 2002). Systemic lupus erythematosus context of hepatitis B, as opposed to PAN and positive hepatitis B
can present with lobular panniculitis resembling nodular lesions serologies should engender suspicion of PAN (Bauza et al. 2002).
and associated antiphospholipid antibody syndrome with livedo Diagnostic criteria have been proposed by a group of investigators
(Sontheimer and McCauliffe 2007). Cutaneous nodular lesions can from Japan’s Ministry of Health, Labour, and Welfare (Nakamura
be a manifestation of leukaemias, cutaneous lymphomas, or myco- et al. 2009). These include presence of any of the four character-
bacterial infections (erythema induratum of Bazin) (Canueto et al. istic cutaneous lesions (nodules, livedo, ulcers, or purpura) and
2011; Kiniwa et al. 2011; Segura et al. 2008) and thrombophlebitis histopathological confirmation of fibrinoid necrosis of a cutaneous
can mimic CPAN clinically and on biopsy (Chen 2010). small or medium-size artery, along with criteria to exclude systemic
vasculitis and other immunological diseases (Table  27.3). These
diagnostic criteria have not been validated but can serve as a rough
Diagnostic approach guide for an approach to CPAN
CPAN should be considered in patients with cutaneous nodular
or ulcerative lesions associated with livedo in the lower extremi- Disease associations and exposures
ties and the diagnosis is also suggested by the presence of malaise,
fatigue, or sensory manifestations (paresthesias, dysaesthesias) Medications, autoimmune diseases, infections, and preg-
in the involved area (Nakamura et al. 2009). Laboratory workup nancy have been described in association with CPAN. Notably,
should initially be aimed at screening for PAN and might include
a complete blood count, creatinine, liver chemistry tests, sedimen-
tation rate, and hepatitis serologies. Antiphospholipid antibod- Table 27.3 Proposed diagnostic criteria for cutaneous
ies, including anti-PS/PT, would be compatible with diagnosis of polyarteritis nodosa
CPAN and might be ordered (Kawakami et  al. 2007)  along with
tests to exclude AAV, SLE, cryoglobulinaemia, and other rheumatic 1. Any cutaneous manifestation: nodules, livedo, ulcers, or purpura
diseases. A deep incisional cutaneous biopsy of a nodular lesion or 2. Histopathological findings: fibrinoid necrosis of cutaneous small or
the edge of an ulcerative lesion should be performed, ideally with a medium-sized artery
separate sample for DIF (Carlson 2010). 3. Exclusion of:
◆ Persistent fever (38°C for more than 2 weeks) or significant weight loss
(6 kg in 6 months)
Table 27.2 Differential diagnosis of cutaneous polyarteritis nodosa ◆ Hypertension
◆ Rapidly progressive renal failure or renal infarction
Systemic polyarteritis nodosa ◆ Cerebral haemorrhage or cerebral infarction
Small vessel vasculitides: ANCA-associated, cryoglobulinaemia, ◆ Myocardial infarction, ischemic heart disease, pericardial disease, or
Henoch-Schönlein purpura, leukocytoclastic vasculitides heart failure
◆ Pleuritis
Erythema nodosum
◆ Intestinal haemorrhage or infarction
Antiphospholipid antibody syndrome ◆ Peripheral neuropathy unrelated to area of cutaneous involvement

Systemic lupus erythematosus ◆ Joint or muscle involvement unrelated to area of cutaneous involvement
◆ Abnormal angiogram, with microaneurysms or vascular occlusions
Pyoderma gangrenosum
Tuberculosis and other mycobacterial infections In order to be considered for a diagnosis of cutaneous polyarteritis nodosa all 3 criteria
need to be satisfied.
Cutaneous leukemias and lymphomas (Adapted with permission from Archives of Dermatological Research, 301; 1. Nakamura,
Thrombophlebitis T. Cutaneous polyarteritis nodosa: revisiting its definition and diagnostic criteria. ©
Springer, 2009.)
368 SECTION 5 vasculitic diseases and syndromes and related disorders

minocycline exposure has been reported to induce CPAN. Most reaction. This process might be analogous to that described in other
minocycline-induced cases improved with discontinuation of the conditions characterized by livedo, mainly antiphospholipid anti-
medication and short courses of oral glucocorticoids (Pelletier body syndrome (Salmon et al. 2002). Thrombosis and endothelial
et al. 2003; Schrodt and Callen 1999; Szwarc and Kaminski 1971; proliferation leading to vessel-wall occlusion are later occurrences
Tehrani et  al. 2007). There are also reports of PAN associated in the disease process (Ishibashi and Chen 2008). No genetic asso-
with minocycline use (Kermani et al. 2012). Other drugs alleged ciations have been described for CPAN.
to induce CPAN include penicillin (Borrie 1972), sulphona-
mides (Bastian 2008), oestrogen (Keyloun et  al. 1967), metola- Treatment
zone (Weinrauch et al. 1982), and antituberculous drugs (Szwarc
and Kaminski 1971). Both forms of inflammatory bowel disease The therapeutic approach to CPAN is empiric as no clinical trials
(Crohn’s disease and ulcerative colitis) have been described in exist and is based on disease severity (Table 27.4). In most instances,
association with CPAN (Chiu and Rajapakse 1991; Gudbjornsson therapy should be less intense than for systemic PAN. Mild disease,
and Hallgren 1990; Komatsuda et al. 2008; Matsumara et al. 2000). which presents mainly with livedo and nodules, can be treated with
There was no clear temporal or severity correlation between CPAN topical glucocorticoids, non-steroidal anti-inflammatory drugs
and inflammatory bowel disease. Other vasculitides, including (NSAIDs), oral colchicine, or low-doses of oral glucocorticoids
Behçet’s disease (Heller 2005; Vikas et al. 2003), Takayasu’s arte- (10–20 mg/day of oral prednisone or its glucocorticoid equivalent).
ritis (Cajigas et al. 1987), and leukocytoclastic vasculitis (Pischel There is a consensus that the prognosis for these cases is benign and
and Zvaifler 1984)  have been concurrent with CPAN lesions in the disease runs a self-limited course with improvement expected
case reports. by 6  months. Severe cases presenting with ulcers are often asso-
Among the infections reported in association with CPAN, myco- ciated with peripheral neuropathy. Oral glucocorticoids in higher
bacterial infections are the better described, including both tuber- doses (0.5–1 mg/kg/day of oral prednisone or its glucocorticoid
culosis and non-tuberculous mycobacterial infections (Chen et al. equivalent) should be considered as the first-line approach in these
2004; Imanishi et  al. 2012). Treatment of the underlying myco- cases. Disease control is usually effective but recurrences are com-
bacterial infection along with anti-inflammatory therapy leads to mon after the glucocorticoid dosage is tapered. Multiple immune
resolution of most cases. Other concurrent infections, including suppressors, immune modulators, miscellaneous agents, and other
meliodosis (Choonhakarn and Jirarattanapochai 1998), parvovi- therapies have been anecdotally reported to be effective in con-
rus (Durst et al. 2002), and HIV (Lai-Cheong et al. 2007; Peraire trolling disease activity and allowing for glucocorticoid tapering.
et  al. 1993)  have also been described. Hepatitis B infection has Examples include methotrexate (Schartz et al. 2001; Jorizzo et al.
been rarely reported in association with CPAN (Van de Pette et al. 1991), sulfapyridine (Diaz-Perez and Winkelmann 1974; Moreland
1984; Whittaker et al. 1986), as opposed to the well-known associa- and Ball 1990), azathioprine (Ozen et al. 1992), dapsone (Maillard
tion with PAN. Hepatitis B vaccination has also been described to et al. 1999), pentoxifylline (Calderon et al. 1993), cyclophospha-
induce CPAN (Bourgeais et  al. 2003; Ventura et al. 2009). Cases mide (Fathalla et al. 2005), colchicine (Maillard et al. 1999), miz-
have also been reported in association with hepatitis C infection oribine (Kawakami and Soma 2011b), tamoxifen (Cvancara et al.
(Soufir et  al. 1999). In children, and also sporadically in adults, 1998), granulocyte-colony stimulating factor (Tursen et al. 2006),
a preceding infection by group A  beta-haemolytic Streptococcus and hyperbaric oxygen (Mazokopakis et  al. 2009). Intravenous
infection has been documented (Albornoz et al. 1998; Sasamoto immune globulin (IVIg) has been reported to be helpful when
et al. 1999) and cases have been reported after necrotizing fascii- CPAN is refractory to oral glucocorticoids and other secondary
tis caused by Streptococcus spp. (Stein et al. 2001). Onset of CPAN agents (Lobo et  al. 2008; Marie et  al. 2012). The mechanism of
has been reported in association with pregnancy (Janin-Mercier action of IVIg in autoimmune and inflammatory disorders is not
et al. 1982). completely understood but may involve binding of immune com-
plexes containing pathogenic antibodies and complement compo-
Pathogenesis nents (Baerenwaldt et al. 2010). Both of these components of the
Recent progress in the understanding of the histopathology of
CPAN, along with the putative role of antiphospholipid anti- Table 27.4 Therapeutic approach to cutaneous polyarteritis nodosa
bodies (Kawakami et  al. 2007), have lead to interesting hypoth-
eses for the cause of CPAN. The case for an antigen exposure is Mild forms: cutaneous nodules, Severe forms: ulcers, severe
clearly supported by the well-documented cases after minocycline livedo, and mild or transient or persistent extracutaneous
use (Kermani et al. 2012; Pelletier et al. 2003; Schaffer et al. 2001; extracutaneous manifestations manifestations
Schrodt and Callen 1999; Schrodt et al. 1999; Tehrani et al. 2007).
Topical glucocorticoids High dose oral glucocorticoids
The theory that antibodies play a role in the disease is also sup-
ported by CPAN in newborn infants of mothers with the disease. Non-steroidal anti-inflammatory Immune-modulators: methotrexate,
The description of antiphospholipid antibodies (mainly anti-PS/ drugs azathioprine, dapsone, pentoxifylline,
PT) along with C3 and IgM deposits in the vessel walls (Kawakami cyclophosphamide, mizoribine
and Soma 2011a) support the theory that the endothelium is the Colchicine Intravenous immune-globulin
initial target of the disease. This is also described in histopatho- Low-dose oral glucocorticoids Clopidogrel and warfarin
logical reports that identify the endothelium as the initially affected
area (Ishibashi and Chen 2008); an antigen present in the endothe- Miscellaneous: tamoxifen,
granulocyte-colony stimulating factor,
lium presumably induces an antibody response, leading to comple-
hyperbaric oxygen
ment and immune complex deposition and posterior inflammatory
CHAPTER 27 cutaneous polyarteritis 369

immune system have been found in the vessel wall of patients with Bauza, A., Espana, A., and Idoate, M. (2002). Cutaneous polyarteritis nodosa.
CPAN, which provides some rationale for its use (Kawakami and British Journal of Dermatology, 146, 694–9.
Soma 2011a). In addition, IVIg could provide toxin or microbial Borrie, P. (1972). Cutaneous polyarteritis nodosa. British Journal of
Dermatology, 87, 87–95.
component neutralization in cases associated with a specific anti-
Bourgeais, A.M., Dore, M.X., Croue, A., Leclech, C., and Verret, J.L. (2003).
gen. High cost and the logistical details surrounding IVIg infusions
[Cutaneous polyarteritis nodosa following hepatitis B vaccination].
are important limitations on its use. Annales de Dermatologie et de Vénéréologie, 130, 205–7.
In addition, anticoagulant and antiplatelet agents, including war- Cajigas, J.C., Amigo, M.C., Pineda, C., Herrera, R., Sanchez-Torres, G., and
farin (at a target international normalized ratio of 3.0) (Kawakami Martinez-Lavin, M. (1987). Association between Takayasu’s arteri-
and Soma 2010b) and clopidogrel (Kawakami and Soma 2010a), tis and cutaneous polyarteritis nodosa. American Journal of Medicine,
have been described as useful in refractory cases of CPAN. The 82, 382–4.
interesting aspect of these treatment approaches is that serum levels Calderon, M.J., Landa, N., Aguirre, A., and Diaz-Perez, J.L. (1993). Successful
of anti-PS/PT and lupus anticoagulant decreased along with thera- treatment of cutaneous PAN with pentoxifylline. British Journal of
Dermatology, 128, 706–8.
peutic improvement (Kawakami and Soma 2010b). In both cases it
Canueto, J., Meseguer-Yebra, C., Roman-Curto, C., Santos-Briz, A.,
is believed that the agents led to an attenuation of the thrombotic
Fernandez-Lopez, E., Fraile, C., and Unamuno, P. (2011). Leukemic vas-
component of the vasculitis. culitis: a rare pattern of leukemia cutis. Journal of Cutaneous Pathology,
The prognosis of CPAN is in general benign, with no mortality 38, 360–4.
specifically associated with the disease, which usually runs a relaps- Carlson, J.A. (2010). The histological assessment of cutaneous vasculitis.
ing and remitting course with ultimate resolution after some years. Histopathology, 56, 3–23.
Cases with more severe cutaneous manifestations should be fol- Chan, P.T., Ishiko, A., Wada, N., Yamamoto, N., and Amagai, M. (2009).
lowed more closely for development of PAN, which might appear Inflammatory plaque with peripheral nodules:  a new specific finding
several years after the disease onset (Chen 1989). of cutaneous polyarteritis nodosa. Journal of the American Academy of
Dermatology, 60, 320–5.
Chen, H.H., Hsiao, C.H., and Chiu, H.C. (2004). Successive develop-
Cutaneous polyarteritis nodosa in children ment of cutaneous polyarteritis nodosa, leucocytoclastic vasculitis
CPAN is an uncommon but well-described illness in children. and Sweet’s syndrome in a patient with cervical lymphadenitis caused
by Mycobacterium fortuitum. British Journal of Dermatology, 151,
Examples can be seen as early as a few days after birth, resulting 1096–100.
from (presumed) passive antibody transfer from mothers with Chen, K.R. (1989). Cutaneous polyarteritis nodosa: a clinical and histopatho-
active CPAN (Stone et al. 1993; Miller and Fries 1975). Discounting logical study of 20 cases. Journal of Dermatology, 16, 429–42.
very early presentations, cases occur on average at age 9  years Chen, K.R. (2010). The misdiagnosis of superficial thrombophlebitis as cuta-
(Ozen et al. 2004), but can start as early as age 1–2 years (Kumar neous polyarteritis nodosa: features of the internal elastic lamina and
et  al. 1995). The clinical presentation is like that of adults, with the compact concentric muscular layer as diagnostic pitfalls. American
fatigue, arthralgias, and myalgias. Skin lesions can be more severe, Journal of Dermatopathology, 32, 688–93.
with more frequent gangrene and autoamputations because of the Chiu, G. and Rajapakse, C.N. (1991). Cutaneous polyarteritis nodosa and
small calibre of involved vessels (Stone et al. 1993). Preceding infec- ulcerative colitis. Journal of Rheumatology, 18, 769–70.
Choonhakarn, C. and Jirarattanapochai, K. (1998). Cutaneous polyarteritis
tious exposures, mainly upper respiratory tract infections often
nodosa:  a report of a case associated with melioidosis (Burkholderia
caused by Streptococcus spp., are described more frequently than pseudomallei). International Journal of Dermatology, 37, 433–6.
in adults (David et al. 1993; Ozen 2004). Immunizations against Cvancara, J.L., Meffert, J.J., and Elston, D.M. (1998). Estrogen-sensitive
hepatitis B and diphtheria–pertussis–tetanus have also been cutaneous polyarteritis nodosa:  response to tamoxifen. Journal of the
described to precede CPAN in children (Ventura et al. 2009; Kumar American Academy of Dermatology, 39, 643–6.
et al. 1995). Treatment is usually less intense than in cases of adult Daoud, M.S., Hutton, K.P., and Gibson, L.E. (1997). Cutaneous periarte-
CPAN, with less need for utilization of second-line agents. Another ritis nodosa: a clinicopathological study of 79 cases. British Journal of
important difference is that in cases associated with Streptococcus Dermatology, 136, 706–13.
spp. infection, penicillin prophylaxis and follow-up of antistrep- David, J., Ansell, B.M., and Woo, P. (1993). Polyarteritis nodosa associated
with streptococcus. Archives of Disease in Childhood, 69, 685–8.
tolysin antibodies is recommended for relapses (Till and Amos
Diaz-Perez, J.L. and Winkelmann, R.K. (1974). Cutaneous periarteritis
1997). Childhood CPAN can run a relapsing course, but in gen-
nodosa. Archives of Dermatology, 110, 407–14.
eral its prognosis is good with most patients achieving remission Dickson, W. (1908). Polyarteritis acuta nodosa and periarteritis nodosa.
in adolescence. Journal of Pathology and Bacteriology, 12, 31–57.
Durst, R., Goldschmidt, N., and Ben Yehuda, A. (2002). Parvovirus B19
References infection associated with myelosuppression and cutaneous polyarteritis
Albornoz, M.A., Benedetto, A.V., Korman, M., McFall, S., Tourtellotte, C.D., nodosa. Rheumatology (Oxford), 41, 1210–2.
and Myers, A.R. (1998). Relapsing cutaneous polyarteritis nodosa associ- Ehrmann, S. (1907). Ein Gefaessprozess Bei Lues. Wiener Medizinische
ated with streptococcal infections. International Journal of Dermatology, Wochenschrift, 57, 777–782.
37, 664–6. Fathalla, B.M., Miller, L., Brady, S., and Schaller, J.G. (2005). Cutaneous
Arkin, A. (1930). A Clinical and pathological study of periarteritis nodosa: a polyarteritis nodosa in children. Journal of the American Academy of
report of five cases, one histologically healed. American Journal of Dermatology, 53, 724–8.
Pathology, 6, 401–426 5. Ferrari, E. (1903). Ueber Polyarteritis acuta nodosa (sogenannte Periateritis
Baerenwaldt, A., Biburger, M., and Nimmerjahn, F. (2010). Mechanisms nodosa), und ihre Beziehungen zur Polymyositis und Polyneuritis acuta.
of action of intravenous immunoglobulins. Expert Review of Clinical Beiträge zur Pathologischen Anatomie und zur Allgemeinen Pathologie,
Immunology, 6, 425–34. 34, 350–86.
Bastian, H. (2008). Cutaneous polyarteritis. In Vasculitis, 2nd edn (ed. G.V. Fisher, I. and Orkin, M. (1964). Cutaneous form of periarteritis nodosa—an
Ball and S. L. Bridges), 365–72. Oxford University Press, Oxford, UK. entity? Archives of Dermatology, 89, 180–9.
370 SECTION 5 vasculitic diseases and syndromes and related disorders

Gottlieb, N.L. (1978). Arthropathy associated with cutaneous polyarteritis. Kussmaul, A. and Maier, R. (1866). Ueber eine nicht bisher beschriebene
Arthritis and Rheumatism, 21, 281–2. eigenhümliche Arterienerkrankung (Periarteritis Nodosa), die mit
Gudbjornsson, B. and Hallgren, R. (1990). Cutaneous polyarteritis nodosa Morbus Brightii und rapid fortschreitender allgemeiner Muskellähmung
associated with Crohn’s disease. Report and review of the literature. einhergeht. Deutsches Archiv für Klinische Medizin, 1, 484–518.
Journal of Rheumatology, 17, 386–90. Lai-Cheong, J.E., Tang, V., Mazhude, C., Baker, L., and Menage Hdu, P.
Heller, M. (2005). Behçet’s disease (incomplete) and cutaneous polyarteritis (2007). Highly active antiretroviral therapy: a treatment for cutaneous
nosa. Dermatology Online Journal, 11, 25. polyarteritis nodosa-like syndrome in a HIV positive patient? Journal
Holers, V.M., Girardi, G., Mo, L., Guthridge, J.M., Molina, H., Pierangeli, of the European Academy of Dermatology and Venereology, 21, 1138–40.
S.S., Espinola, R., Xiaowei, L.E., Mao, D., Vialpando, C.G., and Salmon, Lindberg, K. (1931). Ein beitrag zur kenntnis der periarteritis nodosa. Acta
J.E. (2002). Complement C3 activation is required for antiphospholipid Medica Scandinavica, 76, 183–225.
antibody-induced fetal loss. Journal of Experimental Medicine, 195, Lobo, I., Ferreira, M., Silva, E., Alves, R., and Selores, M. (2008). Cutaneous
211–20. polyarteritis nodosa treated with intravenous immunoglobulins. Journal
Imanishi, H., Tsuruta, D., Oshimo, T., Sowa, J., Mizuno, N., Nakagawa, of the European Academy of Dermatology and Venereology, 22, 880–2.
K., and Ishii, M. (2012). Cutaneous polyarteritis nodosa induced by Lyell, A. and Church, R. (1954). The cutaneous manifestations of polyarteritis
Mycobacterium tuberculosis. Journal of Dermatology, 39, 738–9. nodosa. British Journal of Dermatology, 66, 335–43.
Ishibashi, M. and Chen, K.R. (2008). A morphological study of evolution of Maillard, H., Szczesniak, S., Martin, L., Garot, D., Machet, M.C., Machet, L.,
cutaneous polyarteritis nodosa. American Journal of Dermatopathology, Lorette, G., and Vaillant, L. (1999). [Cutaneous periarteritis nodosa: diag-
30, 319–26. nostic and therapeutic aspects of 9 cases]. Annales de Dermatologie et de
Ishiguro, N. and Kawashima, M. (2010). Cutaneous polyarteritis nodosa: a Vénéréologie, 126, 125–9.
report of 16 cases with clinical and histopathological analysis and a Marie, I., Miranda, S., Girszyn, N., Soubrane, J.C., Vandhuick, T., and
review of the published work. Journal of Dermatology, 37, 85–93. Levesque, H. (2012). Intravenous immunoglobulins as treatment of
Janin-Mercier, A., Beyvin, A.J., Pablo, M., and Jacquetin, B. (1982). Cutaneous severe cutaneous polyarteritis nodosa. Internal Medicine Journal, 42,
periarteritis nodosa occurring during pregnancy. Acta Dermato- 459–62.
Venereologica, 62, 256–8. Matsumara, Y., Mizuno, K., Okamoto, H., and Imamura, S. (2000). A case of
Jorizzo, J.L., White, W.L., Wise, C.M., Zanolli, M.D., and Sherertz, E.F. (1991). cutaneous polyarteritis nodosa associated with ulcerative colitis. British
Low-dose weekly methotrexate for unusual neutrophilic vascular reac- Journal of Dermatology, 142, 561–2.
tions: cutaneous polyarteritis nodosa and Behçet’s disease. Journal of the Mazokopakis, E.E., Milkas, A.N., Tsartsalis, A.N., Karefilakis, C.M., and
American Academy of Dermatology, 24, 973–8. Besmertis, L.P. (2009). Improvement of cutaneous polyarteritis nodosa
Kawakami, T. and Soma, Y. (2010a). Therapeutic effect of clopidogrel on lesions with hyperbaric oxygen. International Journal of Dermatology,
cutaneous polyarteritis nodosa. Archives of Dermatology, 146, 100–1. 48, 1023–5.
Kawakami, T. and Soma, Y. (2010b). Use of warfarin therapy at a target inter- Mekori, Y.A., Awai, L.E., Wiedel, J.D., and Kohler, P.F. (1984). Cutaneous pol-
national normalized ratio of 3.0 for cutaneous polyarteritis nodosa. yarteritis nodosa associated with rapidly progressive arthritis. Arthritis
Journal of the American Academy of Dermatology, 63, 602–6. and Rheumatism, 27, 574–8.
Kawakami, T. and Soma, Y. (2011a). Correlation of livedo racemosa, cuta- Melcer, N. and Venkei, T. (1947). Uber die Hautformen der Periarteritis
neous inflammatory plaques, and antiphospholipid antibodies in nodosa. Dermatologica, 94, 214.
patients with cutaneous polyarteritis nodosa. Medicine (Baltimore), 90, Miescher, V.G. (1946). Uber kutane formen der Periarteritis Nodosa.
119–24. Dermatologica, 92, 225–245.
Kawakami, T. and Soma, Y. (2011b). Use of mizoribine in two patients with Miller, J.J. 3rd and Fries, J.F. (1975). Simultaneous vasculitis in a mother and
recalcitrant cutaneous polyarteritis nodosa. Journal of the American newborn infant. Journal of Pediatrics, 87, 443–5.
Academy of Dermatology, 64, 1213–4. Moreland, L.W. and Ball, G.V. (1990). Cutaneous polyarteritis nodosa.
Kawakami, T., Yamazaki, M., Mizoguchi, M., and Soma, Y. (2007). High titer American Journal of Medicine, 88, 426–30.
of anti-phosphatidylserine-prothrombin complex antibodies in patients Morgan, A.J. and Schwartz, R.A. (2010). Cutaneous polyarteritis nodosa: a
with cutaneous polyarteritis nodosa. Arthritis and Rheumatism, 57, comprehensive review. International Journal of Dermatology, 49, 750–6.
1507–13. Nakamura, T., Kanazawa, N., Ikeda, T., Yamamoto, Y., Nakabayashi, K., Ozaki,
Kawakami, T., Yamazaki, M., Mizoguchi, M., and Soma, Y. (2009). Differences S., and Furukawa, F. (2009). Cutaneous polyarteritis nodosa:  revisit-
in anti-phosphatidylserine-prothrombin complex antibodies and cuta- ing its definition and diagnostic criteria. Archives for Dermatological
neous vasculitis between regular livedo reticularis and livedo racemosa. Research, 301, 117–21.
Rheumatology (Oxford), 48, 508–12. Ozen, S. (2004). Juvenile polyarteritis:  is it a different disease? Journal of
Kermani, T.A., Ham, E.K., Camilleri, M.J., and Warrington, K.J. (2012). Rheumatology, 31, 831–2.
Polyarteritis nodosa-like vasculitis in association with minocycline Ozen, S., Anton, J., Arisoy, N., Bakkaloglu, A., Besbas, N., Brogan, P.,
use: a single-center case series. Seminars in Arthritis and Rheumatism, Garcia-Consuegra, J., Dolezalova, P., Dressler, F., Duzova, A., Ferriani,
42, 213–21. V.P., Hilario, M.O., Ibanez-Rubio, M., Kasapcopur, O., Kuis, W., Lehman,
Keyloun, V., Halperin, I., and Grace, W.J. (1967). Periarteritis nodosa caused T.J., Nemcova, D., Nielsen, S., Oliveira, S.K., Schikler, K., Sztajnbok,
by hypersensitivity to estrogens. Vascular Diseases, 4, 21–6. F., Terreri, M.T., Zulian, F., and Woo, P. (2004). Juvenile polyarteri-
Kiniwa, Y., Ide, Y., Fukushima, M., Asano, N., and Saida, T. (2011). A case of tis: results of a multicenter survey of 110 children. Journal of Pediatrics,
systemic anaplastic large cell lymphoma with ‘Hodgkin-like’ appearance 145, 517–22.
and skin involvement mimicking lymphomatoid papulosis. Journal of Ozen, S., Besbas, N., Saatci, U., and Bakkaloglu, A. (1992). Diagnostic criteria
Cutaneous Pathology, 38, 38–42. for polyarteritis nodosa in childhood. Journal of Pediatrics, 120, 206–9.
Komatsuda, A., Kinoshita, K., Togashi, M., Maki, N., Masai, R., Niitsu, H., Pelletier, F., Puzenat, E., Blanc, D., Faivre, B., Humbert, P., and Aubin, F.
Ohtani, S., Sageshima, M., Sawada, K., and Wakui, H. (2008). Cutaneous (2003). Minocycline-induced cutaneous polyarteritis nodosa with anti-
polyarteritis nodosa in a patient with Crohn’s disease. Modern neutrophil cytoplasmic antibodies. European Journal of Dermatology,
Rheumatology, 18, 639–42. 13, 396–8.
Kumar, L., Thapa, B.R., Sarkar, B., and Walia, B.N. (1995). Benign cutaneous Peraire, J., Vidal, F., Mayayo, E., Torre, L., and Richart, C. (1993). Cutaneous
polyarteritis nodosa in children below 10 years of age—a clinical experi- polyarteritis nodosa in human immunodeficiency virus infection.
ence. Annals of the Rheumatic Diseases, 54, 134–6. British Journal of Rheumatology, 32, 937–8.
CHAPTER 27 cutaneous polyarteritis 371

Pischel, K.D. and Zvaifler, N.J. (1984). Simultaneous occurrence of polyarte- Stone, M.S., Olson, R.R., Weismann, D.N., Giller, R.H., and Goeken, J.A.
ritis nodosa and leukocytoclastic vasculitis. Journal of Rheumatology, (1993). Cutaneous vasculitis in the newborn of a mother with cutaneous
11, 542–4. polyarteritis nodosa. Journal of the American Academy of Dermatology,
Ruiter, M. (1958). The so-called cutaneous type of periarteritis nodosa. 28, 101–5.
British Journal of Dermatology, 70, 102–6. Szwarc, M. and Kaminski, Z. (1971). [Polyarteritis nodosa probably caused
Salmon, J.E., Girardi, G., and Holers, V.M. (2002). Complement activation by hypersensitivity to antitubercular drugs]. Gruzlica, 39, 135–42.
as a mediator of antiphospholipid antibody induced pregnancy loss and Tehrani, R., Nash-Goelitz, A., Adams, E., Dahiya, M., and Eilers, D. (2007).
thrombosis. Annals of the Rheumatic Diseases, 61 (Suppl. 2), ii46–50. Minocycline-induced cutaneous polyarteritis nodosa. Journal ofClinical
Sasamoto, M., Shigeta, Y., Hisashi, K., Kashiwagi, Y., Koishi, H., Kobayashi, Rheumatology, 13, 146–9.
K., Takekuma, K., and Hoshika, A. (1999). [Cutaneous polyarteritis Ter Poorten, M.C. and Thiers, B.H. (2002). Panniculitis. Dermatology Clinics,
nodosa associated with streptococcal infection in a child and a review of 20, 421–33, vi.
the literature]. Nihon Rinsho Meneki Gakkai Kaishi, 22, 144–50. Thomas, R.H. and Black, M.M. (1983). The wide clinical spectrum of pol-
Schaffer, J.V., Davidson, D.M., Mcniff, J.M., and Bolognia, J.L. (2001). yarteritis nodosa with cutaneous involvement. Clinical and Experimental
Perinuclear antineutrophilic cytoplasmic antibody-positive cutaneous Dermatology, 8, 47–59.
polyarteritis nodosa associated with minocycline therapy for acne vul- Till, S.H. and Amos, R.S. (1997). Long-term follow-up of juvenile-onset
garis. Journal of the American Academy of Dermatology, 44, 198–206. cutaneous polyarteritis nodosa associated with streptococcal infection.
Schartz, N.E., Alaoui, S., Vignon-Pennamen, M.D., Cordoliani, F., Fermand, British Journal of Dermatology, 36, 909–11.
J.P., Morel, P., and Rybojad, M. (2001). Successful treatment in two cases Tursen, U., Api, H., Kaya, T.I., Cinel, L., and Ikizoglu, G. (2006). Rapid
of steroid-dependent cutaneous polyarteritis nodosa with low-dose healing of chronic leg ulcers during perilesional injections of
methotrexate. Dermatology, 203, 336–8. granulocyte-macrophage colony-stimulating factor therapy in a patient
Schrodt, B.J. and Callen, J.P. (1999). Polyarteritis nodosa attributable to with cutaneous polyarteritis nodosa. Journal of the European Academy of
minocycline treatment for acne vulgaris. Pediatrics, 103, 503–4. Dermatology and Venereology, 20, 1341–3.
Schrodt, B.J., Kulp-Shorten, C.L., and Callen, J.P. (1999). Necrotizing vascu- Uthman, I.W. and Khamashta, M.A. (2006). Livedo racemosa:  a striking
litis of the skin and uterine cervix associated with minocycline therapy dermatological sign for the antiphospholipid syndrome. Journal of
for acne vulgaris. Southern Medical Journal, 92, 502–4. Rheumatology, 33, 2379–82.
Segura, S., Pujol, R.M., Trindade, F., and Requena, L. (2008). Vasculitis in Van De Pette, J.E., Jarvis, J.M., Wilton, J.M., and Macdonald, D.M. (1984).
erythema induratum of Bazin:  a histopathologic study of 101 biopsy Cutaneous periarteritis nodosa. Hepatitis B surface antigen-containing
specimens from 86 patients. Journal of the American Academy of immunocomplexes and polymorphonuclear-leukocyte lysosomal
Dermatology, 59, 839–51. enzyme release. Archives of Dermatology, 120, 109–11.
Smukler, N.M. and Schumacher, H.R. Jr. (1977). Chronic nondestruc- Ventura, F., Antunes, H., Brito, C., Pardal, F., Pereira, T., and Vieira, A.P.
tive arthritis associated with cutaneous polyarteritis. Arthritis and (2009). Cutaneous polyarteritis nodosa in a child following hepatitis B
Rheumatism, 20, 1114–20. vaccination. European Journal of Dermatology, 19, 400–1.
Sontheimer, R.D. and McCauliffe, D.P. (2007). Lupus-specific skin dis- Vikas, A., Atul, S., Singh, R., Sarbmeet, L., and Mohan, H. (2003). Behçet’s dis-
ease (cutaneous LE). In Dubois’ lupus erythematosus, 7th edn (ed. D.J. ease with relapsing cutaneous polyarteritis-nodosa-like lesions, respon-
Wallace and B.H. Hahn). Lippincott Williams & Wilkins, Philadelphia, sive to oral cyclosporine therapy. Dermatology Online Journal, 9, 9.
PA, pp. 576–620. Watts, R.A., Gonzalez-Gay, M.A., Lane, S.E., Garcia-Porrua, C., Bentham,
Soufir, N., Descamps, V., Crickx, B., Thibault, V., Cosnes, A., Becherel, P.A., G., and Scott, D.G. (2001). Geoepidemiology of systemic vasculi-
Wolkenstein, P., Bournerias, I., De La Salmoniere, P., Picard, C., Roujeau, tis: comparison of the incidence in two regions of Europe. Annals of the
J.C., Piette, J.C., Belaich, S., Revuz, J., and Frances, C. (1999). Hepatitis C Rheumatic Diseases, 60, 170–2.
virus infection in cutaneous polyarteritis nodosa: a retrospective study Weinrauch, L.A., Belok, S.S., Gauvin, G.P., and D’Elia, J.A. (1982). Palpable
of 16 cases. Archives of Dermatology, 135, 1001–2. acute necrotizing arteritis secondary to metolazone. Cutis, 30, 83–4.
Stein, R.H., Phelps, R.G., and Sapadin, A.N. (2001). Cutaneous polyarteri- Whittaker, S.J., Dover, J.S., and Greaves, M.W. (1986). Cutaneous polyarte-
tis nodosa after streptococcal necrotizing fasciitis. Mt Sinai Journal of ritis nodosa associated with hepatitis B surface antigen. Journal of the
Medicine, 68, 336–8. American Academy of Dermatology, 15, 1142–5.
CHAPTER 28

Kawasaki’s disease
Jane C. Burns

Kawasaki’s disease (KD) is an acute vasculitis predominantly in (Burns et  al. 2005a). The recurrence rate during 2009–2012 was
medium-sized, extraparenchymal arteries, most notably the coro- 3.6%, 1.6% had an affected sibling, and 0.7% had at least one parent
nary arteries (Kawasaki et al. 1974). While the acute, febrile phase with a history of KD (Nakamura et al. 2012). Time–space cluster-
of the illness is self-limited, an intense inflammatory response in ing and outbreaks in communities are recognized; however, there is
the coronary arterial wall can lead to aneurysms with the attendant no evidence of person-to-person transmission. In 1979, 1982, and
risk of thrombotic occlusion, late stenosis, and myocardial infarc- 1986 there were large outbreaks in Japan, but there have been no
tion (Kato et al. 1996). such epidemics over the last 25 years.
KD was first described by Dr Tomisaku Kawasaki in Japan, KD is now widely recognized across the globe but is most prevalent
who recognized his first case in 1961 (Burns et  al. 2000; Shike in Japan and eastern Asia (Straface et al. 2012). In the USA, there is
et  al. 2002). Initially, the condition was thought to be a benign, no active surveillance programme as in Japan, but using an admin-
self-limited illness with a good prognosis, because the acute symp- istrative database, the KD-associated hospitalization rate was 20.8
toms disappeared within 2 to 3 weeks. It was not until 1970 that the per 100 000 children less than 5 years in 2006 (Holman et al. 2010).
first nation-wide survey in Japan reported that 1.7% of the patients The hospitalization rate per 100 000 children less than 5 years old in
died from acute myocardial infarction, and four autopsies revealed 2006 was 30.3 among those of Asian and Pacific Island descent, 17.5
coronary arteritis accompanied by aneurysms and thrombotic in non-Hispanic African-Americans, 15.7 in Hispanics, and 12.0 in
occlusion. In 1973, Kato performed coronary angiography in 20 children of European descent. The fatality rate was approximately
infants and children after resolution of their symptoms. Twelve had 2% in the 1970s and had declined to 0.1% in the 1990s in Japan. In
multiple coronary aneurysms in both the right and left coronary the USA, the in-hospital mortality rate is 0.17%, with peak mortality
arteries, which was the first recognition that coronary aneurysms occurring 15 to 45 days after the onset of illness (Chang 2002).
are present not only in fatal cases, but also in survivors free from
cardiac symptoms (Kato et al. 1975). Pathology
In the past, KD may have been misdiagnosed as scarlet fever,
KD is classified among the systemic vasculitides involving the
enterovirus infection, measles, rheumatic fever, juvenile rheu-
small or medium-sized arteries, particularly the coronary arteries
matoid arthritis, Stevens–Johnson syndrome, and polyarteritis
(Jennette et al. 1994). At autopsy, myocarditis (interstitial myocar-
nodosa, which are included in its differential diagnosis (Kushner
ditis with mild necrosis), pericarditis, endocarditis, and inflam-
et al. 2004; Newburger et al. 2004). Landing reported that patho-
mation of medium-sized arteries throughout the body are seen
logical findings are identical between fatal KD and infantile pol-
(Amano et al. 1980) (Figure 28.1). Three distinct vasculopathic
yarteritis nodosa, thus unifying the two entities (Landing and
processes have been identified and include an acute, self-limited
Larson 1977).
necrotizing arteritis, a subacute/ chronic vasculitis, and a luminal
myofibroblastic proliferation (Orenstein et al. 2012).
Epidemiology
The epidemiology of KD in Japan, where it is most prevalent, has Coronary artery lesions
been well documented by successive nation-wide surveys con- Coronary arteritis begins with neutrophilic infiltration into the
ducted every 2 years in Japan. The number of patients has increased arterial wall from the luminal and adventitial sides, leading imme-
since the 1960s and now totals over 250 000 as of 2012. KD affects diately to inflammation of all layers of the artery (Figure 28.2). The
over 12 000 children each year in Japan and the incidence has been internal elastic lamina, smooth muscle cells of the media, and other
increasing yearly to 240 per 100 000 children less than 5 years of age structural components of the artery are subjected to intense dam-
according to the 2010 survey (Nakamura et al. 2012). Onset may be age; the artery then begins to dilate. When the damage is severe,
as early as 1 month of age and peaks at 1 year of age. Fifty per cent saccular aneurysms may develop on about the 12th day after
of children with KD are less than 2 years of age; those over 10 years disease onset. Myofibroblasts in the media secrete matrix metal-
of age are uncommon. Boys are affected more than girls with a loproteinases and proinflammatory cytokines including IL-17,
male:female ratio of 1.5:1. KD is more prevalent in Japanese and which may recruit inflammatory cells into the vessel wall (Shimizu
other Asians than in children of European descent. Seasonal varia- et al. 2012). Transforming growth factor (TGF)-β has been impli-
tion has been clearly documented in Japan with two annual peaks, cated in the process of epithelial and endothelial to mesenchymal
one in December through February and the second in mid-summer transition and formation of myofibroblasts (Shimizu et al. 2011).
374 SECTION 5 vasculitic diseases and syndromes and related disorders

Fig. 28.3 Coronary artery aneurysm filled in with a fresh thrombus in patient


Fig. 28.1 Gross appearance of the heart with spherical coronary aneurysm who died 18 days after the onset of KD.
(arrow). (Courtesy of Dr A.E. Becker, Amsterdam, the Netherlands.)

A second process begins in the first 2 weeks but can be observed


Rupture of saccular aneurysms is possible at this stage although months to years after KD onset and can involve virtually all muscu-
very rare. Altered haemodynamics in the aneurysm with reduced loelastic arteries except those in the cerebral circulation (Orenstein
wall sheer stress, decreased velocity, and increased particle resi- et al. 2012). The cellular infiltrate in these lesions are lymphocytes
dence time may lead to thrombotic occlusion, which is found in with a predominance of CD8+ T cells and macrophages (Brown
the coronary artery aneurysms of many autopsy cases from the et al. 2001).
first months after KD onset (Takahashi et al. 2005; Sengupta et al. A third process results in luminal narrowing and is medi-
2012) (Figure 28.3). ated exclusively by myofibroblasts that may derive from vascular
smooth muscle cells. Myofibroblastic proliferation can lead to nar-
rowing of the lumen and even complete occlusion (Figure 28.4).
(a) (b)

Fig. 28.2 Panarteritis, inflammation of all layers of the artery, of a coronary


artery in a patient who died 10 days after the onset of Kawasaki’s disease. Fig. 28.4 Coronary artery of a patient who died of meningitis 14 years after the
(a) Hematoxylin and eosin stain; (b) immunohistochemical study using anti-CD68 onset of Kawasaki’s disease. This patient had no coronary aneurysms but coronary
antibody. arteries showed circumferential intimal thickening.
CHAPTER 28 kawasaki’s disease 375

It is common for the wall of an aneurysm to be accompanied by Table 28.1 Validated genetic associations with susceptibility or
laminar calcification surrounding it, with an organized thrombus coronary artery aneurysms (CAA) in KD
on the inner side; plugging of the lumen by a fresh thrombus may
then be lethal. Some lesions suggest that the vessel has been recana- Gene Chr. References for susceptibility or CAA association
lized due to partial thrombolysis (Fujiwara et al. 1986). Multiple
FCGR2A 1q23 Khor et al. 2011a; Onouchi et al. 2012
channels may be opened within the aneurysm giving the classic
‘lotus root appearance’(Naoe et al. 1991). TGFB2 1q41 Shimizu et al. 2011
CCR5 3p21 Burns et al. 2005b; Breunis et al. 2007; Jhang et al.
Myocarditis 2009
Myocarditis on all histological sections was documented by right TGFBR2 3p22 Shimizu et al. 2011
ventricular endomyocardial biopsy from series of hundreds of KD
CASP3 4q35 Onouchi et al. 2010; Onouchi et al. 2012
patients during the acute and subacute phase of the illness (Yutani
et al. 1980; Yonesaka et al. 1989). Late findings may include bridg- IL-4 5q31.1 Burns et al. 2005c
ing fibrosis not in the watershed distribution of the epicardial coro- VEGFA 6p12 Kariyazono et al. 2004; Hsueh et al. 2008; Breunis
nary arteries (Gordon et al. 2009). et al. 2012
HLA 6p21.3 Onouchi et al. 2012
Relationships between kawasaki’s disease
arteritis versus classical polyarteritis nodosa BLK 8p22-23 Lee et al. 2012; Onouchi et al. 2012
and infantile polyarteritis nodosa MMP3 11q22.3 Hong et al. 2008; Shimizu et al. 2010
Kussmaul–Maier-type classical polyarteritis nodosa (PAN) is char- ABCC4 13q32 Khor et al. 2011b
acterized by beaded aneurysms in medium- and small-sized arter-
SMAD3 15q22.33 Kuo et al. 2011a; Shimizu et al. 2011
ies. One of the histological characteristics of PAN is the presence
of fibrinoid necrosis in the arterial lesions, which is usually not a ACE 17q23 Takeuchi et al. 1997; Wu et al. 2004; Shim et al.
prominent feature of KD arteritis. It is difficult to make a simple 2006
comparison between KD and infantile polyarteritis nodosa (IPN), ITPKC 19q13.2 Onouchiet al. 2008; Khor et al. 2011; Kuo et al.
because the former is a clinical entity whereas IPN is a pathologi- 2011; Onouchi et al. 2012; Peng et al. 2012
cal entity. Many patients diagnosed with IPN cannot be histologi- CD40 20q13 Kuo et al. 2012; Lee et al. 2012; Onouchi et al. 2012
cally differentiated from those with KD (Tanaka 1975; Landing and
Larson 1977). The possibility that patients who are diagnosed at
autopsy as IPN may include patients with KD or other similar dis-
eases has been investigated through historical analysis of records European descent populations (Onouchi et al. 2008; Onouchi et al.
(Kushner et al. 2003; Kushner et al. 2004). 2010). Genetic variation in the TGF-β pathway has been associ-
ated with aneurysm formation (Kuo et  al. 2011a; Shimizu et  al.
2011). Genetic determinants unique to Asian populations have
Pathogenesis and aetiology been identified that may explain the unique susceptibility to KD
The aetiology of KD remains unknown despite extensive investi- in children of Asian descent (Onouchi et al. 2012). Despite several
gation. Based on clinical features and genetic and epidemiological genome-wide association studies, the percentage of KD suscepti-
data, KD is now considered to be triggered by an agent or antigen bility that can be explained by genetic variants is low and this will
that enters through the upper airway and causes a severe immune probably be a rapidly evolving field in the future.
response in genetically predisposed children. Work by Rowley
and colleagues has identified intracellular virus-like particles in Clinical presentations
bronchial epithelial cells from KD autopsies (Rowley et al. 2011). Symptoms and diagnosis of kawasaki’s disease
Climate scientists have reported a link between fluctuations in KD
case numbers and large-scale tropospheric wind patterns and have Common clinical findings in KD are shown in Figure  28.5. In
proposed that the trigger of KD could be associated with aerosols the absence of a specific laboratory test, the diagnosis of KD is
carried by these winds (Rodo et al. 2011). made according to diagnostic guidelines (Newburger et al. 2004).
The acute phase of KD is associated with markedly increased pro- (Table  28.2) The principal diagnostic criteria of KD are persis-
duction of proinflammatory cytokines, and high levels of TNF-α, tent fever associated with conjunctival injection, changes in the
IL-1, and IFN-γ are detected in the circulation (Matsubara et al. mucosa of the oropharynx, changes in the peripheral extremities,
1990). The self-limited nature of the illness is due to expansion of erythematous rash, and cervical lymphadenopathy. At least four
both the natural and peripherally induced regulatory T-cell popula- of five principal symptoms should be met for diagnosis of com-
tion with increased secretion of IL-10 (Franco et al. 2010). plete KD (Table  28.2). Patients may have incomplete KD when
fewer than four criteria are present in association with fever of at
least 5 days and laboratory findings suggesting marked systemic
Genetics inflammation (Newburger et al. 2004; Tremoulet et al. 2011). In
A genetic influence on KD susceptibility, response to treatment, patients with fewer than four clinical criteria, the diagnosis can
and development of coronary artery aneurysms is clear (Onouchi be established when coronary abnormalities such as dilatation (Z
2012) (Table 28.1). Single nucleotide polymorphisms in pathways score ≥2.5) (see Section Spectrum of Cardiovascular Disease) or
involving calcium signalling have been validated in both Asian and non-uniformity of arterial lumen, or enhanced brightness of the
376 SECTION 5 vasculitic diseases and syndromes and related disorders

(a) (b) (e)

(f)
(d)

(c)

(g)

(h)

Fig. 28.5 Clinical manifestations of KD. (a) Typical appearance of Kawasaki face, bilateral conjunctival congestion, and reddening and fissuring of lips. (b) Strawberry
tongue. (c and d) Polymorphous exanthema. (e) Indurative oedema of the hands. (f) Redness and swelling of the sole. (g and h) Desquamation of fingers and toes.

arterial wall, are recognized on two-dimensional echocardiogra- and juvenile idiopathic arthritis (Komatsu and Tateno 2007; Kumar
phy (Bratincsak et al. 2012; Muniz et al. 2013). Patients less than et al. 2013; Jaggi et al. 2013).
6  months of age or those over 7  years of age may present with
incomplete symptoms (Burns et  al. 1986; Pannaraj et  al. 2004). Spectrum of cardiovascular disease
Incomplete KD should be considered in infants with unexplained Current rates of aneurysms in patients treated with intravenous
fever for more than 7 days even in the absence of the other princi- immunoglobulin (IVIg) and aspirin within the first 10 days after
pal signs and echocardiography should be performed. In the eval- fever onset range from 3 to 5% (Newburger et  al. 1991). The
uation of suspected incomplete KD associated with a C-reactive coronary arteries are the most common sites of aneurysms in
protein ≥3.0 mg/dl or erythrocyte sedimentation rate ≥40 mm/h, KD; however, aneurysms in other arteries such as in the axil-
the diagnosis is considered confirmed in the presence of at least lary, iliac, or renal arteries, may occur. Valvular heart disease is
three of the following laboratory findings:  albumin ≤3.0 g/dl, seen in 1.5% of the patients (Akagi et al. 1990). Current recom-
anaemia for age, elevated alanine aminotransferase, platelet count mendations are to express the internal diameter of the coronary
≥450  000/μl, white blood cell count ≥15  000, and urine white arteries as standard deviation units from the mean normalized
blood cell count ≥10 cells/ high power field (Yellen et al. 2010). for body surface area (Z score) and to consider arteries with a
Erythema and induration at the site of previous vaccination Z score ≤ 2.5 SD units to be normal (McCrindle et  al. 2007).
with BCG (Bacille Calmette–Guérin) is also helpful in diagnos- Regression equations for this calculation have been published
ing incomplete KD. Complications in KD during the acute phase (Dallaire et al. 2012).
may include disseminated intravascular coagulation, macrophage The main cause of death in KD is acute myocardial infarction
activation syndrome, and a KD shock syndrome (Kanegaye et al. (MI). From a nation-wide survey in Japan, 195 KD patients with
2009; Latino et al. 2010). MI were analysed (Kato et al. 1986): MI was most likely to occur
The differential diagnosis of KD includes viral infections, within 1  year of illness onset. Not surprisingly, coronary angio-
especially adenovirus, enterovirus, and measles, bacterial graphic studies showed that most patients who died had obstruc-
toxin-mediated diseases including scarlet fever and streptococcal tions in the left main coronary artery or in both the right main
and staphylococcal toxic shock, systemic allergic drug reactions, coronary artery and the anterior descending artery while survivors
CHAPTER 28 kawasaki’s disease 377

Table 28.2 Diagnostic guidelines for KD in a patient having a renal artery lesion, and lesions in intratho-
racic arteries may complicate coronary bypass surgery. Gangrene
A. Principal criteria due to peripheral artery stenosis has been reported (Tomita
1. Fever persisting for at least 3 days associated with four of the five clinical et al. 1992).
criteria:
Valvular heart disease, myocarditis, and
2. Bilateral conjunctival injection with perilimbal sparing
pericarditis
3. Changes of lips and oral cavity: Valvular heart disease appears in about 1% of patients, mostly in the
Reddening and fissuring of lips
mitral valve and rarely the aortic valve. In one series, acute mitral
Strawberry tongue
regurgitation was noted in 28 of 2180 cases (1.3%), and resolved
Diffuse injection of oral and pharyngeal mucosa
in one-half of patients after months to several years. The aetiology
4. Polymorphous exanthema may be valvulitis or papillary muscle dysfunction caused by ischae-
5. Changes of peripheral extremities: mia (Akagi et al. 1990). There were five patients with aortic regur-
Initial stage: Reddening of palms and soles gitation (0.2%). It is noteworthy that aortic regurgitation appeared
Oedema of dorsa of hand and feet after the acute or subacute stage of illness and slowly progressed
Convalescent stage: Periungual desquamation of fingers and toes to severe regurgitation in some patients (Gidding et  al. 1986).
6. Cervical lymphadenopathy, usually unilateral with lymph node mass Subclinical pericarditis or pericardial effusion appeared in 12.9%
measuring at least 1.5 cm of the patients in the acute phase, which typically resolved within 1
or 2 weeks. Massive pericardial effusion or cardiac tamponade was
Patients may be diagnosed with KD with fewer criteria accompanied
by dilated coronary arteries (Z score ≥ 2.5 SD units) documented by
rare. Clinically mild myocarditis was observed in about 28.3% of
echocardiography. the patients in the acute phase, especially in the first and second
weeks of illness, regardless of the presence of coronary aneurysms.
B. Other associated clinical signs
However, echocardiographic evidence of diastolic dysfunction sug-
1. Cardiovascular: gests that impaired relaxation due to myocardial inflammation may
Auscultation abnormalities: gallop rhythm be more common (Sato et al. 2013; Selamet Tierney et al. 2011). The
2. GI tract: presence of a cardiac gallop, distant heart sounds, ST-T segment
Diarrhoea changes, and decreased voltage of R waves on electrocardiogram
Vomiting (ECG) may suggest the presence of myocarditis. In many instances,
Abdominal pain cardiac enzyme levels did not change significantly. Acute myocardi-
Hydrops of gall bladder tis may cause cardiomegaly or decreased ejection fraction of the left
Paralytic ileus ventricle, but usually resolves following treatment with IVIg. A KD
Mild jaundice shock syndrome has been recognized but appears related more to
3. Skin: inappropriate peripheral vascular resistance rather than decreased
Erythema and crusting at the site of BCG inoculation left ventricular contractility (Dominguez et  al. 2008; Kanegaye
Micropustular rash et al. 2009).
Transverse furrows of the nails (Beau’s lines) in convalescent phase
Cardiac evaluation
6. Respiratory:
Cough The evaluation of the coronary artery lesions in KD in the acute
Rhinorrhoea stage of illness is essential, and is usually done by two-dimensional
echocardiography (2DE). The internal diameter of the right and
7. Axial arthropathy and small joint arthritis of PIP joints
left anterior descending coronary arteries should be expressed as
8. Neurological: standard deviation units from the mean based on body surface
Facial nerve palsy area in order to appropriately classify abnormalities (de Zorzi et al.
Sensorineural hearing loss 1998; McCrindle et  al. 2007; Manlhiot et  al. 2010; Dallaire et  al.
2012). Low-radiation computed tomographic angiography (CTA)
and magnetic resonance angiography (MRA) have replaced inva-
tended to have one-vessel obstruction, particularly in the right cor- sive coronary angiography at most centres for more detailed evalu-
onary artery (Figure 28.6). ation of the coronary arteries in children with aneurysms (Duan
et al. 2012; Tacke et al. 2013).
Systemic artery involvement In patients with severe coronary lesions, evaluation for other
In a Japanese series, aneurysms in non-coronary arteries were systemic vascular involvement (such as aneurysms in the axil-
observed in 1.0% of the patients (Kato et al. 1996). In an angi- lary, iliac, renal, or intrathoracic arteries) should be considered
ographic study of 22 patients with systemic artery aneurysms, based on clinical findings. Because regression of coronary artery
axillary arteries were affected in 18 cases; common iliac arteries aneurysms or progression to stenotic lesions occur over time,
in 16; internal iliac arteries in 12; renal arteries in six; mesenteric serial evaluations of patients with coronary artery abnormalities
arteries in two; and internal thoracic arteries in two. One patient are essential (Kato et  al. 1996). Intravascular ultrasound imag-
had a large common iliac artery aneurysm associated with aortic ing of the coronary arteries in patients with KD may show mild
dilatation. Although the prognosis of systemic artery aneurysms or marked intimal thickening at the site of aneurysms that have
is generally favourable, renovascular hypertension may develop resolved (Mitani et al. 2009).
378 SECTION 5 vasculitic diseases and syndromes and related disorders

(a) (b1) (b2)


LAD
RCA

CX

(c1) (c2)

Fig. 28.6 Regression of coronary aneurysms. This patient was diagnosed with Kawasaki’s disease as an 11 month old. (a) Aneurysms in both the right and the left
coronary arteries. (b-1, b-2) One year and 2 months later the follow-up coronary angiography demonstrated disappearance of the coronary aneurysms. There were no
stenoses or irregularities of the arterial lumen. (c-1,c-2) The follow-up coronary angiography 12 years later demonstrated no aneurysms, stenosis, or irregularities of the
arterial lumen in either the left or right coronary arteries.

Management Japanese population, their positive and negative predictive value


are low for genetically mixed populations (Tremoulet et  al.
Treatment of acute kawasaki’s disease 2008; Kobayashi et  al. 2009; Sleeper et  al. 2011). Treatment of
The major therapeutic approach to acute KD is a combination IVIg-resistant KD patients remains controversial. In Japan, addi-
of aspirin (30–100 mg/kg body weight per day until afebrile for tion of oral methylprednisolone to standard IVIg therapy for 2
48 hours, then 3–5 mg/kg/day until the platelet count and ESR to 3 weeks in selected, high-risk subjects reduced IVIg resistance
have returned to normal, usually 4–6 weeks), and high-dose IVIg and the number of subjects with coronary artery abnormalities
(Newburger et al. 1991; Newburger et al. 2004). Clinical improve- (Kobayashi et  al. 2012). Such treatment approaches, however,
ment is usually noted by the end of the IVIg infusion. The mecha- may be problematic in mixed ethnic populations for whom the
nisms of action of IVIg may be various and include: Fc-mediated scoring systems fail to identify patients at greatest risk. A trial of
expansion of natural regulatory T-cell population; modulation of a single pulse of methylprednisolone (30 mg/kg intravenously)
T/B-cell functions through binding to Fc receptors; and inhibition added to standard IVIg plus aspirin therapy in unselected North
of activated complement binding to targets such as endothelium American KD patients failed to show a clinical benefit (Newburger
(Franco et al. 2010). Potential Fab-mediated mechanisms include et al. 2007).
neutralization of microbial toxins, cytokines, and anti-idiotypic Other treatments for IVIg-resistant KD patients include the
reactions against autoantibodies. anti-TNF-α antibody infliximab (Burns et al. 2008), the neutrophil
IVIg resistance is defined as persistent or recrudescent fever at elastase inhibitor ulinastatin (Kanai et al. 2011), plasma exchange
least 36 hours but less than 7 days after completion of the IVIg (Hokosaki et al. 2012), and cytotoxic agents such as cyclophospha-
infusion (Burns et al. 1998). Rates of IVIg resistance vary among mide and ciclosporin (Wallace et al. 2000; Tremoulet et al. 2012).
series but generally range from 15 to 25% (Tremoulet et al. 2008). All these drugs may have some efficacy for treatment of KD; how-
Scoring systems to predict IVIg resistance have been developed in ever, there are no results from randomized, controlled trials in
Japan and while they work well in the genetically homogeneous IVIg-resistant patients to guide therapeutic decisions.
CHAPTER 28 kawasaki’s disease 379

Long-term management Table 28.3 (Continued)


Long-term management of patients with KD depends on the degree Patients with regressed coronary aneurysms
of coronary arterial involvement. If coronary artery abnormalities discontinue aspirin
are detected, low-dose aspirin is commonly continued until these follow with stress echocardiography every several years
have resolved. Since progression to ischaemic heart disease in chil- no restriction of physical activity
dren with coronary artery aneurysms noted during the acute phase Patients with giant coronary aneurysms (Z ≥10)
may occur, particularly in those with aneurysms with a Z score aspirin with warfarin
of 10 or greater, these patients should be managed by paediatric follow-up every 3–6 months
cardiologists. Strong historical evidence supports the treatment of computed tomographic angiography and stress echocardiography at
patients with aneurysms with a Z score of 10 or greater with war- intervals
farin (target INR 2.0–2.5) and aspirin (Suda et al. 2011). Table 28.3 Patients with coronary stenosis with or without aneurysms
follow-up every 3–6 months
summarizes recommendations for long-term management.
physical activity restricted depending on stress test
assess the need for percutaneous intervention or bypass surgery
Table 28.3 Therapeutic recommendations for KD
Indications for percutaneous interventions:
Acute phase: Ischaemic symptoms or findings
Aspirin
Localized stenosis
30 to 100 mg/kg (4 divided daily doses) until afebrile for 48 hours, then
PTCA is preferable in younger patients without severe calcification
3–5 mg/kg for 6 to 8 weeks
Stenting is indicated in older children
IVIg
Rotational ablation is indicated for patients with severe calcification
2 g/kg as a single infusion over 12 hours
For cases refractory to initial IVIg Indications for coronary bypass surgery:
additional 2 g/kg as a single infusion over 12 hours Ischaemic symptoms or findings
or Severe multivessel obstruction or stenosis
infliximab 5 mg/kg as a single infusion over 2 hours Stenosis in the left main coronary artery
or
Indication for heart transplantation:
methylprednisolone 30 mg/kg/dose once daily i.v. for 3 days or 2 mg/kg
orally for 2–3 weeks depending on C-reactive protein and clinical response Severe ischaemic cardiomyopathy
Optional treatments for highly refractory cases:
infliximab, ciclosporin, or cyclophosphamide
For cases with coronary artery aneurysms: Long-term complications may include coronary artery stenosis,
Consider adding clopidogrel 1 mg/kg/day as a single dose for patients myocardial ischaemia, congestive heart failure, myocardial infarc-
with moderate coronary aneurysms Z≥5.0 but <10
tion, and arrhythmia (Gordon et al. 2009; Tsuda et al. 2011a; Tsuda
For patients with giant coronary aneurysms (internal dimension ≥8 mm
or Z score ≥10)
et al. 2011b). Interventions for myocardial ischaemia may include
Enoxaparin 0.75 mg/kg/day subcutaneously every 12 hours in combination angioplasty with or without stenting, rotational ablation, internal
with aspirin thoracic artery bypass grafting, and cardiac transplantation (Ino
or et al. 1996; Checchia et al. 1997; Tsuda et al. 2006a; Tsuda et al.
warfarin (0.05–0.12 mg/kg/day to keep INR 2.0–2.5) in combination
2006b; Kitamura et al. 2009; Muta and Ishii 2010). Percutaneous
with aspirin interventions may be complicated by the presence of extensive cal-
cification in the arterial lesions (Kahn et al. 2012) (Figure 28.7).
Convalescent phase:
Aspirin 3–5 mg/kg/day as a single dose for patients with small coronary
aneurysms (Z ≥ 2.5 but <5.0) Prognosis and long-term issues
Consider adding clopidogrel 1 mg/kg/day as a single dose for patients with
Long-term consequences of coronary
moderate coronary aneurysms Z ≥5.0 but <10)
aneurysms
For patients with giant coronary aneurysms (internal dimension ≥8 mm or Z
score ≥10): The natural history or fate of coronary aneurysms was studied by
Enoxaparin 0.75 mg/kg/day subcutaneously every 12 hours Kato and colleagues in 594 consecutive patients with acute KD
Warfarin (0.05–0.12 mg/kg/day to keep INR 2.0–2.5) in combination with aspirin
between 1973 and 1983, with follow-up ranging from 10 to 21 years
(Kato et al. 1996). In all patients, coronary artery lesions were evalu-
Long-term management:
ated by coronary angiography in the subacute stage. One hundred
Patients with no coronary lesions
stop aspirin, follow at 1 year and intervals thereafter and forty-six patients (24.6%) were diagnosed as having coronary
no restriction of physical activity aneurysms. A second angiogram obtained 1 to 2 years later in all
Patients with small or moderate-sized aneurysms 146 cases who previously had coronary aneurysms, demonstrated
continue aspirin (3–5 mg/kg/day) that 72 (49.3%) of these 146 had regression in coronary aneurysms.
follow with echocardiography and computed tomographic angiography to None of the patients with regression of coronary aneurysms had car-
check for regression of coronary aneurysms or progression to stenosis diac symptoms in the long-term follow-up periods; results of EKGs,
no restriction of physical activity exercise stress tests, thallium myocardial scintigraphy, and left ven-
(continued) tricular function assessments were all within normal limits. Of the
380 SECTION 5 vasculitic diseases and syndromes and related disorders

a-1 a-2 a-3


stenosis stent

LAD

LCX

b-1 b-2 b-3

Fig. 28.7 Percutaneous interventions for stenotic lesions. Left coronary artery angiography and intravascular ultrasound imaging in a 13-year-old boy with severe stenosis
of the left anterior descending artery (LAD). (a-1, b-1) 95% localized stenosis of the LAD (arrow) before stent implantation. (a-2, b-2) Coronary stenosis improved after
stent placement. (b-2, b-3) Intravascular ultrasound imaging image at the site of the coronary artery stenosis after stent placement showing the stent and improved
coronary artery lumen diameter. (a-3, b-3) Follow-up coronary angiography and intravascular ultrasound imaging images 1 year later showed maintenance of the
successfully dilated coronary artery lumen.

594 patients, stenosis in the coronary aneurysms developed in 28 cause of acquired heart disease in children. The vasculitis affects
cases. Myocardial infarction occurred in 11 patients, five of whom predominantly medium-sized, extraparenchymal arteries, particu-
died. From this study, it is estimated that about 4% of patients with larly the coronary arteries. Timely treatment with IVIg and aspirin
KD may develop ischaemic heart disease. The timing and the inci- reduces the incidence of coronary artery aneurysms from 25% to
dence of regression or progression to obstructive lesions from the 5%. Long-term follow-up studies are needed to clarify the progno-
onset of KD was assessed using the Kaplan–Meyer plots. Regression sis for these children in adulthood.
of coronary aneurysms mostly occurred within 2  years from the
onset of illness, whereas progression to obstructive lesions gradu- References
ally increased after 2 years. In the 26 patients with giant coronary
Akagi, T., Kato, H., Inoue, O., et al. (1990). Valvular heart disease in Kawasaki
aneurysms (internal diameter >8mm), stenotic lesions developed in
syndrome: incidence and natural history. American Heart Journal, 120,
12, and no regression occurred during the follow-up study. Thus, the 366–72.
presence of giant coronary aneurysms is a critical problem because Amano, S., Hazama, F., Kubagawa, H., et al. (1980). General pathology of
it has a high likelihood to progress to ischaemic heart disease. Kawasaki disease. On the morphological alterations corresponding to
It has now been more than 45 years since the first description of the clinical manifestations. Acta Pathologica Japonica, 30, 681–94.
KD as a new clinical entity, and a number of the early KD children Bratincsak, A., Reddy, V.D., Purohit, P.J., et al. (2012). Coronary artery dila-
are now adults. A recent study of young adults less 40 years of age tion in acute Kawasaki disease and acute illnesses associated with Fever.
who underwent cardiac catheterization for evaluation of possible Pediatric Infectious Disease Journal, 31, 924–6.
myocardial ischaemia showed that 5% had coronary artery lesions Breunis, W.B., Biezeveld, M.H., Geissler, J., et  al. (2007). Polymorphisms
in chemokine receptor genes and susceptibility to Kawasaki disease.
consistent with antecedent KD (Daniels et al. 2012).
Clinical and Experimental Immunology,150, 83–90.
Breunis, W.B., Davila, S., Shimizu, C., et al. (2012). Disruption of vascular
Summary homeostasis in patients with Kawasaki disease: involvement of vascular
endothelial growth factor and angiopoietins. Arthritis and Rheumatism,
KD is a common, acute, self-limited illness characterized by fever 64, 306–15.
in association with mucocutaneous involvement that affects pri- Brown, T.J., Crawford, S.E., Cornwall, M.L., et al. (2001). CD8 T lympho-
marily infants and young children. In Japan, North America, and cytes and macrophages infiltrate coronary artery aneurysms in acute
Western Europe, KD has replaced rheumatic fever as the leading Kawasaki disease. Journal of Infectious Diseases, 184, 940–3.
CHAPTER 28 kawasaki’s disease 381

Burns, J.C., Best, B.M., Mejias, A., et  al. (2008). Infliximab treatment of Hsueh, K.C., Lin, Y.J., Chang, J.S., et  al. (2008). Association of vascular
intravenous immunoglobulin-resistant Kawasaki disease. Journal of endothelial growth factor C-634 g polymorphism in taiwanese children
Pediatrics, 153, 833–8. with Kawasaki disease. Pediatric Cardiology, 29, 292–6.
Burns, J.C., Capparelli, E.V., Brown, J.A., et  al. (1998). Intravenous Ino, T., Akimoto, K., Ohkubo, M., et al. (1996). Application of percutane-
gamma-globulin treatment and retreatment in Kawasaki disease. US/ ous transluminal coronary angioplasty to coronary arterial stenosis in
Canadian Kawasaki Syndrome Study Group. Pediatric Infectious Disease Kawasaki disease. Circulation, 93, 1709–15.
Journal, 17, 1144–8. Jaggi, P., Kajon, A.E., Mejias, A., et  al. (2013). Human adenovirus infec-
Burns, J.C., Cayan, D.R., Tong, G., et al. (2005a). Seasonality and temporal tion in Kawasaki disease: A confounding bystander? Clinical Infectious
clustering of Kawasaki syndrome. Epidemiology, 16, 220–5. Diseases, 56, 58–64.
Burns, J.C., Kushner, H.I., Bastian, J.F., et al. (2000). Kawasaki disease: A brief Jennette, J.C., Falk, R.J., Andrassy, K., et  al. (1994). Nomenclature of sys-
history. Pediatrics, 106, E27. temic vasculitides. Proposal of an international consensus conference.
Burns, J.C., Shimizu, C., Gonzalez, E., et al. (2005b). Genetic variations in Arthritis and Rheumatism, 37, 187–92.
the receptor-ligand pair CCR5 and CCL3L1 are important determi- Jhang, W.K., Kang, M.J., Jin, H.S., et al. (2009). The CCR5 (-2135C/T)
nants of susceptibility to Kawasaki disease. Journal of Infectious Diseases, Polymorphism may be associated with the development of Kawasaki
192, 344–9. disease in Korean children. Journal of Clinical Immunology,
Burns, J.C., Shimizu, C., Shike, H., et al. (2005c). Family-based association 29, 22–8.
analysis implicates IL-4 in susceptibility to Kawasaki disease. Genes and Kahn, A.M., Budoff, M.J., Daniels, L.B., et  al. (2012). Calcium scoring in
Immunity, 6, 438–44. patients with a history of Kawasaki disease. JACC Cardiovascular
Burns, J.C., Wiggins, Jr, J.W., Toews, W.H., et al. (1986). Clinical spectrum Imaging, 5, 264–72.
of Kawasaki disease in infants younger than 6 months of age. Journal of Kanai, T., Ishiwata, T., Kobayashi, T., et  al. (2011). Ulinastatin, a urinary
Pediatrics, 109, 759–63. trypsin inhibitor, for the initial treatment of patients with Kawasaki dis-
Chang, R.K. (2002). Hospitalizations for Kawasaki disease among children in ease: a retrospective study. Circulation, 124, 2822–8.
the United States, 1988-1997. Pediatrics, 109, e87. Kanegaye, J.T., Wilder, M.S., Molkara, D., et  al. (2009). Recognition of a
Checchia, P.A., Pahl, E., Shaddy, R.E., et al. (1997). Cardiac transplantation Kawasaki disease shock syndrome. Pediatrics, 123, e783–9.
for Kawasaki disease. Pediatrics, 100, 695–9. Kariyazono, H., Ohno, T., Khajoee, V., et al. (2004). Association of vascu-
Dallaire, F., Fournier, A., Breton, J., et  al. (2012). Marked variations in lar endothelial growth factor (VEGF) and VEGF receptor gene poly-
serial coronary artery diameter measures in Kawasaki disease:  a new morphisms with coronary artery lesions of Kawasaki disease. Pediatric
indicator of coronary involvement. Journal of the American Society of Research, 56, 953–9.
Echocardiography, 25, 859–65. Kato, H., Ichinose, E., and Kawasaki, T. (1986). Myocardial infarc-
Daniels, L.B., Tjajadi, M.S., Walford, H.H., et  al. (2012). Prevalence of tion in Kawasaki disease:  clinical analyses in 195 cases. Journal of
Kawasaki disease in young adults with suspected myocardial ischemia. Pediatrics,108, 923–7.
Circulation, 125, 2447–53. Kato, H., Koike, S., Yamamoto, M., et  al. (1975). Coronary aneurysms in
de Zorzi, A., Colan, S.D., Gauvreau, K., et al. (1998). Coronary artery dimen- infants and young children with acute febrile mucocutaneous lymph
sions may be misclassified as normal in Kawasaki disease. Journal of node syndrome. Journal of Pediatrics,86, 892–8.
Pediatrics,133, 254–8. Kato, H., Sugimura, T., Akagi, T., et  al. (1996). Long-term consequences
Dominguez, S.R., Friedman, K., Seewald, R., et al. (2008). Kawasaki disease of Kawasaki disease. A 10- to 21-year follow-up study of 594 patients.
in a pediatric intensive care unit: a case-control study. Pediatrics, 122, Circulation, 94, 1379–85.
e786–90. Kawasaki, T., Kosaki, F., Okawa, S., et al. (1974). A new infantile acute febrile
Duan, Y., Wang, X., Cheng, Z., et  al. (2012). Application of prospective mucocutaneous lymph node syndrome (MLNS) prevailing in Japan.
ECG-triggered dual-source CT coronary angiography for infants and Pediatrics, 54, 271–6.
children with coronary artery aneurysms due to Kawasaki disease. Khor, C.C., Davila, S., Breunis, W.B., et al. (2011a). Genome-wide association
British Journal of Radiology, 85, e1190–7. study identifies FCGR2A as a susceptibility locus for Kawasaki disease.
Fujiwara, H., Fujiwara, T., Kao, T.C., Ohshio, G., Hamashima, Y. (1986). Nature Genetics, 43, 1241–6.
Pathology of Kawasaki disease in the healed stage. Relationships Khor, C.C., Davila, S., Shimizu, C., et  al. (2011b). Genome-wide linkage
between typical and atypical cases of Kawasaki disease. Acta Pathol Jpn. and association mapping identify susceptibility alleles in ABCC4 for
36, 857–67. Kawasaki disease. Journal of Medical Genetics, 48, 467–72.
Franco, A., Shimizu, C., Tremoulet, A.H., et al. (2010). Memory T-cells and Kitamura, S., Tsuda, E., Kobayashi, J., et  al. (2009). Twenty-five-year out-
characterization of peripheral T-cell clones in acute Kawasaki disease. come of pediatric coronary artery bypass surgery for Kawasaki disease.
Autoimmunity, 43, 317–24. Circulation, 120, 60–8.
Gidding, S.S., Shulman, S.T., Ilbawi, M., et al. (1986). Mucocutaneous lymph Kobayashi, T., Inoue, Y., Otani, T., et al. (2009). Risk stratification in the deci-
node syndrome (Kawasaki disease): delayed aortic and mitral insuffi- sion to include prednisolone with intravenous immunoglobulin in pri-
ciency secondary to active valvulitis. Journal of the American College of mary therapy of Kawasaki disease. Pediatric Infectious Disease Journal,
Cardiology, 7, 894–7. 28, 498–502.
Gordon, J.B., Kahn, A.M., Burns, J.C., et al. (2009). When children with Kobayashi, T., Saji, T., Otani, T., et al. (2012). Efficacy of immunoglobulin
Kawasaki disease grow up:  Myocardial and vascular complications plus prednisolone for prevention of coronary artery abnormalities in
in adulthood. Journal of the American College of Cardiology, 54, severe Kawasaki disease (RAISE study):  a randomised, open-label,
1911–20. blinded-endpoints trial. Lancet, 379, 1613–20.
Hokosaki, T., Mori, M., Nishizawa, T., et al. (2012). Long-term efficacy of Komatsu, H. and Tateno, A. (2007). Failure to distinguish systemic-onset
plasma exchange treatment for refractory Kawasaki disease. Pediatrics juvenile idiopathic arthritis from incomplete Kawasaki disease in an
International, 54, 99–103. infant. Journal of Paediatrics and Child Health, 43, 707–9.
Holman, R.C., Belay, E.D., Christensen, K.Y., et al. (2010). Hospitalizations Kumar, S., Vaidyanathan, B., Gayathri, S., et  al. (2013). Systemic onset
for Kawasaki syndrome among children in the United States, 1997-2007. juvenile idiopathic arthritis with macrophage activation syndrome
Pediatric Infectious Disease Journal, 29, 483–8. misdiagnosed as Kawasaki disease:  case report and literature review.
Hong, Y.M., Jin, H.S., Park, I.S., et  al. (2008). Association of the matrix Rheumatology International, 33, 1047–52.
metalloproteinase-3 (-439C/G) promoter polymorphism with Kawasaki Kuo, H.C., Chao, M.C., Hsu, Y.W., et al. (2012). CD40 Gene polymorphisms
disease in Korean children. Heart and Vessels, 23, 341–7. associated with susceptibility and coronary artery lesions of Kawasaki
382 SECTION 5 vasculitic diseases and syndromes and related disorders

disease in the Taiwanese population. Scientific World Journal, 2012, Onouchi, Y., Gunji, T., Burns, J.C., et al. (2008). ITPKC functional polymor-
520865. phism associated with Kawasaki disease susceptibility and formation of
Kuo, H.C., Onouchi, Y., Hsu, Y.W., et al. (2011a). Polymorphisms of trans- coronary artery aneurysms. Nature Genetics, 40, 35–42.
forming growth factor-beta signaling pathway and Kawasaki disease in Onouchi, Y., Ozaki, K., Burns, J.C., et al. (2010). Common variants in CASP3
the Taiwanese population. Journal of Human Genetics, 56, 840–5. confer susceptibility to Kawasaki disease. Human Molecular Genetics,
Kuo, H.C., Yang, K.D., Juo, S.H., et al. (2011b). ITPKC single nucleotide poly- 19, 2898–906.
morphism associated with the Kawasaki disease in a Taiwanese popula- Onouchi, Y., Ozaki, K., Burns, J.C., et al. (2012). A genome-wide associa-
tion. PLoS ONE, 6, e17370. tion study identifies three new risk loci for Kawasaki disease. Nature
Kushner, H.I., Bastian, J.F., Turner, C.H., et al. (2003). Rethinking the bound- Genetics, 44, 517–21.
aries of Kawasaki disease: toward a revised case definition. Perspectives Orenstein, J.M., Shulman, S.T., Fox, L.M., et al. (2012). Three linked vasculo-
in Biology and Medicine, 46, 216–33. pathic processes characterize Kawasaki disease: a light and transmission
Kushner, H.I., Turner, C.L., Bastian, J.F., et  al. (2004). The narratives of electron microscopic study. PLoS One, 7, e38998.
Kawasaki disease. Bulletin of the History of Medicine, 78, 410–39. Pannaraj, P.S., Turner, C.L., Bastian, J.F., et  al. (2004). Failure to diagnose
Landing, B.H. and Larson, E.J. (1977). Are infantile periarteritis nodosa Kawasaki disease at the extremes of the pediatric age range. Pediatric
with coronary artery involvement and fatal mucocutaneous lymph Infectious Disease Journal, 23, 789–91.
node syndrome the same? Comparison of 20 patients from North Peng, Q., Chen, C., Zhang, Y., et al. (2012). Single-nucleotide polymorphism
America with patients from Hawaii and Japan. Pediatrics, 59, rs2290692 in the 3'UTR of ITPKC associated with susceptibility to
651–62. Kawasaki disease in a Han Chinese population. Pediatric Cardiology, 33,
Latino, G.A., Manlhiot, C., Yeung, R.S., et al. (2010). Macrophage activation 1046–53.
syndrome in the acute phase of Kawasaki disease. Journal of Pediatric Rodo, X., Ballester, J., Cayan, D., et al. (2011). Association of Kawasaki dis-
Hematology/ Oncology, 32, 527–31. ease with tropospheric wind patterns. Scientific Reports, 1, 152.
Lee, Y.C., Kuo, H.C., Chang, J.S., et al. (2012). Two new susceptibility loci for Rowley, A.H., Baker, S.C., Shulman, S.T., et al. (2011). Ultrastructural, immu-
Kawasaki disease identified through genome-wide association analysis. nofluorescence, and RNA evidence support the hypothesis of a “new”
Nature Genetics, 44, 522–5. virus associated with Kawasaki disease. Journal of Infectious Diseases,
Manlhiot, C., Millar, K., Golding, F., et al. (2010). Improved classification of 203, 1021–30.
coronary artery abnormalities based only on coronary artery z-scores Sato, Y.Z., Molkara, D.P., Daniels, L.B., et  al. (2013). Cardiovascular bio-
after Kawasaki disease. Pediatric Cardiology, 31, 242–9. markers in acute Kawasaki disease. International Journal of Cardiology,
Matsubara, T., Furukawa, S., and Yabuta, K. (1990). Serum levels of tumor 164, 58–63.
necrosis factor, interleukin 2 receptor, and interferon-gamma in Selamet Tierney, E.S., Newburger, J.W., Graham, D., et al. (2011). Diastolic
Kawasaki disease involved coronary-artery lesions. Clinical Immunology function in children with Kawasaki disease. International Journal of
and Immunopathology, 56, 29–36. Cardiology, 148, 309–12.
McCrindle, B.W., Li, J.S., Minich, L.L., et al. (2007). Coronary artery involve- Sengupta, D., Kahn, A.M., Burns, J.C., et al. (2012). Image-based modeling of
ment in children with Kawasaki disease: risk factors from analysis of hemodynamics in coronary artery aneurysms caused by Kawasaki dis-
serial normalized measurements. Circulation, 116, 174–9. ease. Biomechanics and Modeling in Mechanobiology, 11, 915–32.
Mitani, Y., Ohashi, H., Sawada, H., et al. (2009). In vivo plaque composition Shike, H., Burns, J.C., et  al. (2002). English Translation of Dr.  Tomisaku
and morphology in coronary artery lesions in adolescents and young Kawasakis original report of fifty patients in 1967 (Japanese). Pediatric
adults long after Kawasaki disease:  a virtual histology-intravascular Infectious Disease Journal, 21, 993.
ultrasound study. Circulation, 119, 2829–36. Shim, Y. H., Kim, H.S., Sohn, S., Hong, Y.M. (2006). Insertion/deletion poly-
Muniz, J.C., Dummer, K., Gauvreau, K., et  al. (2013). Coronary artery morphism of angiotensin converting enzyme gene in Kawasaki disease.
dimensions in febrile children without Kawasaki disease. Circulation J Korean Med Sci. 21, 208–11.
Cardiovascular Imaging, 6, 239–44. Shimizu, C., Jain, S., Davila, S., et al. (2011). Transforming growth factor-beta
Muta, H. and Ishii, M. (2010). Percutaneous Coronary Intervention versus signaling pathway in patients with Kawasaki disease. Circulation
Coronary Artery Bypass Grafting for Stenotic Lesions after Kawasaki Cardiovascular Genetics, 4, 16–25.
Disease. Journal of Pediatrics, 157, 120–6. Shimizu, C., Matsubara, T., Onouchi, Y., et al. (2010). Matrix metallopro-
Nakamura, Y., Yashiro, M., Uehara, R., et al. (2012). Epidemiologic features of teinase haplotypes associated with coronary artery aneurysm forma-
Kawasaki disease in Japan: Results of the 2009-2010 nationwide survey. tion in patients with Kawasaki disease. Journal of Human Genetics, 55,
Journal of Epidemiology/ Japan Epidemiological Association, 22, 216–21. 779–84.
Naoe, S., Takahashi, K., Masuda, H., et al. (1991). Kawasaki disease. With par- Shimizu, C., Oharaseki, T., Takahashi, K., et al. (2012). The role of TGF-beta
ticular emphasis on arterial lesions. Acta Pathologica Japonica, 41, 785–97. and myofibroblasts in the arteritis of Kawasaki disease. Human
Newburger, J.W., Sleeper, L.A., McCrindle, B.W., et al. (2007). Randomized Pathology, 44, 189–98.
trial of pulsed corticosteroid therapy for primary treatment of Kawasaki Sleeper, L.A., Minich, L.L., McCrindle, B.M., et  al. (2011). Evaluation of
disease. New England Journal of Medicine, 356, 663–75. Kawasaki disease risk-scoring systems for intravenous immunoglobulin
Newburger, J.W., Takahashi, M., Beiser, A.S., et al. (1991). A single intrave- resistance. Journal of Pediatrics, 158, 831–35, e833.
nous infusion of gamma globulin as compared with four infusions in Straface, E., Marchesi, A., Gambardella, L., et al. (2012). Does oxidative stress
the treatment of acute Kawasaki syndrome. New England Journal of play a critical role in cardiovascular complications of Kawasaki disease?
Medicine,324, 1633–9. Antioxidants and Redox Signaling, 17, 1441–6.
Newburger, J.W., Takahashi, M., Gerber, M.A., et  al. (2004). Diagnosis, Suda, K., Iemura, M., Nishiono, H., et  al. (2011). Long-term prognosis of
treatment, and long-term management of Kawasaki disease:  a state- patients with Kawasaki disease complicated by giant coronary aneu-
ment for health professionals from the Committee on Rheumatic Fever, rysms: a single-institution experience. Circulation, 123, 1836–42.
Endocarditis and Kawasaki Disease, Council on Cardiovascular Disease Tacke, C.E., Romeih, S., Kuipers, I.M., et  al. (2013). Evaluation of cardiac
in the Young, American Heart Association. Circulation, 110, 2747–71. function by magnetic resonance imaging during the follow-up of
Onouchi, Y. (2012). Genetics of Kawasaki disease: what we know and dont patients with Kawasaki disease. Circulation Cardiovascular Imaging,
know. Circulation Journal, 76, 1581–6. 6, 67–73.
CHAPTER 28 kawasaki’s disease 383

Takahashi, K., Oharaseki, T., Naoe, S., et al. (2005). Neutrophilic involvement Tsuda, E., Hirata, T., Matsuo, O., et  al. (2011b). The 30-year outcome for
in the damage to coronary arteries in acute stage of Kawasaki disease. patients after myocardial infarction due to coronary artery lesions
Pediatrics International, 47, 305–10. caused by Kawasaki disease. Pediatric Cardiology, 32, 176–82.
Takeuchi, K., Yamamoto, K., Kataoka, S., Kakihara, T., Tanaka, A., Sato, Tsuda, E., Miyazaki, S., Takamuro, M., et al. (2006a). Strategy for localized stenosis
S., Uchiyama, M. (1997). High incidence of angiotensin I converting caused by Kawasaki disease: midterm results of percutaneous transluminal
enzyme genotype II in Kawasaki disease patients with coronary aneu- coronary balloon angioplasty in two infants. Pediatric Cardiology, 27, 272–5.
rysm. Eur J Pediatr. 156, 266–8. Tsuda, E., Miyazaki, S., Yamada, O., et al. (2006b). Percutaneous translumi-
Tanaka, N. (1975). Kawasaki disease (acute febrile infantile muco-cutaneous nal coronary rotational atherectomy for localized stenosis caused by
lymph node syndrome) in Japan; relationship with infantile periarteritis Kawasaki disease. Pediatric Cardiology, 27, 447–53.
nodosa. Pathologia et Microbiologia (Basel), 43, 204–18. Wallace, C.A., French, J.W., Kahn, S.J., et  al. (2000). Initial intravenous
Tomita, S., Chung, K., Mas, M., et al. (1992). Peripheral gangrene associated gammaglobulin treatment failure in Kawasaki disease. Pediatrics,
with Kawasaki disease. Clinical Infectious Diseases, 14, 121–6. 105, E78.
Tremoulet, A.H., Best, B.M., Song, S., et  al. (2008). Resistance to intrave- Wu, S.F., Chang, J.S., Peng, C.T., Shi, Y.R., Tsai, F.J. (2004) Polymorphism
nous immunoglobulin in children with Kawasaki disease. Journal of of angiotensin-1 converting enzyme gene and Kawasaki disease.
Pediatrics, 153, 117–21. Pediatr Cardiol. 25, 529–33.
Tremoulet, A.H., Jain, S., Chandrasekar, D., et al. (2011). Evolution of labo- Yellen, E.S., Gauvreau, K., Takahashi, M., et  al. (2010). Performance of
ratory values in patients with Kawasaki disease. Pediatric Infectious 2004 American Heart Association recommendations for treatment of
Disease Journal, 30, 1022–6. Kawasaki disease. Pediatrics, 125, e234–41.
Tremoulet, A.H., Pancoast, P., Franco, A., et al. (2012). Calcineurin inhibitor Yonesaka, S., Nakada, T., Sunagawa, Y., et  al. (1989). Endomyocardial
treatment of intravenous immunoglobulin-resistant Kawasaki disease. biopsy in children with Kawasaki disease. Acta Paediatrica Japonica,
Journal of Pediatrics, 161, 506–12, e501. 31, 706–11.
Tsuda, E., Abe, T., and Tamaki, W. (2011a). Acute coronary syndrome in adult Yutani, C., Okano, K., Kamiya, T., et al. (1980). Histopathological study on
patients with coronary artery lesions caused by Kawasaki disease: review right endomyocardial biopsy of Kawasaki disease. British Heart Journal,
of case reports. Cardiology in the Young, 21, 74–82. 43, 589–92.
CHAPTER 29

Granulomatosis with
polyangiitis (Wegener’s
granulomatosis): pathogenesis
Peter Lamprecht, Konstanze
Holl-Ulrich, and Wolfgang L. Gross

Granulomatosis with polyangiitis (GPA) is a rare, chronic, inflamma- Genetic risk factors
tory disorder of unknown aetiology. GPA is characterized by necrotiz-
Familial cases of GPA are rare (Knudsen et  al. 1988). Similar to
ing granulomatous inflammation involving the upper and/or lower
other autoimmune disorders, disease-associated alleles individually
respiratory tract and necrotizing vasculitis affecting predominantly
carry very modest degrees of risk in GPA. Thus, multiple genetic
small to medium-sized vessels. Necrotizing glomerulonephritis is
factors of relatively small effect combine to create a state of sus-
common. GPA is strongly associated with antineutrophil cytoplas-
ceptibility to autoimmune activation. Genetic associations suggest
mic autoantibodies specific for proteinase 3 (PR3-ANCA) (Jennette
that some immune pathomechanisms are shared among chronic
et al. 2013). While data from in vitro and in vivo studies suggest that
inflammatory and autoimmune diseases, while others are not. Most
PR3-ANCA play an important role in the pathogenesis of systemic vas-
associations relate to the activation of cells of innate and adaptive
culitis, it has remained largely unclear to date how necrotizing granu-
immunity and intracellular signalling (Cho and Gregersen 2011).
lomatous inflammation and autoimmunity are related and induced in
GPA shares its association with the 620 tryptophan risk allele of the
GPA (Sarraf and Sneller 2005; Lamprecht et al. 2006; Lamprecht et al.
protein tyrosine phosphatase non-receptor 22 (PTPN22) with other
2009a; Kallenberg 2011; Gadola and Gross 2012). Data from animal
autoimmune diseases such as rheumatoid arthritis, type 1 diabetes
models suggest that the pathogenesis of granulomatous inflammation
mellitus, and systemic lupus erythematosus (Table  29.1) (Jagiello
is separate from acute vasculitis (Salama and Little 2012).
et al. 2005). The PTPN22 R620W single-nucleotide polymorphism
(SNP) is associated with faster progress to end-stage kidney disease
Epidemiology and environmental and relapse in AAV, in particular in GPA. The gain-of-function
and genetic risk factors PTPN22 R620W polymorphism results in an up-regulated basal
PTPN22 phosphatase activity, altered gene expression profile with
Epidemiology decreased extracellular signal-regulated kinase (ERK) activity, and
In Central Europe the incidence of GPA is 9 new cases per million increased p38 mitogen-activated protein kinase (MAPK) activity
inhabitants per year and the prevalence 98 per million inhabitants. and reduced anti-inflammatory IL-10 transcription in GPA (Cao
Doubling of the prevalence during the last decade has been attrib- et al. 2012). The geographic distribution of the disease-associated
uted to better treatment with improved outcome (Herlyn et al. 2010). PTPN22 allele mirrors the north–south gradient of the GPA inci-
Similar incidence and prevalence rates are reported from the USA. dence in Europe. Moreover, the allele is nearly absent in African and
The incidence rate is somewhat higher in the United Kingdom and Asian populations (Vang et al. 2007; Cao et al. 2012).
lower in Southern Europe, suggestive of a European north–south gra- GPA is strongly associated with the human leukocyte antigen
dient. In contrast, GPA is much less common in Japan (Ntatsaki et al. allele HLA-DPB1*0401. GPA shares its HLA-DP association with
2010). A cyclical pattern of occurrence with a periodicity of 7–8 years another granulomatous disorder, chronic beryllium disease, but
between the peak and the lowest incidence is observed in GPA, but not not with MPA and eosinophilic granulomatosis with polyangii-
in microscopic polyangiitis (MPA) in the United Kingdom. This study tis (EGPA, Churg–Strauss syndrome) (Arning et al. 2011). Genes
lends support to differences in the aetiology of ANCA-associated within the major histocompatibility complex (MHC) generally exert
vasculitides (AAV) and a potential role of infectious triggers in GPA by far the strongest single genetic effect in autoimmune disorders
(Watts et al. 2012). Identification of environmental components that (Cho and Gregersen 2011). The difference in the HLA background
interact with host genetic factors will be critical in developing a deeper suggests that AAV constitute separate diseases with shared clinical
understanding of GPA and new approaches to its treatment and pre- and pathological features, rather than one disease entity with differ-
vention in the future (Cho and Gregersen 2011). ent phenotypes (Arning et al. 2011). Moreover, genetic risk factors
386 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 29.1 Genetic risk factors in GPA; most associations are descriptive, their functional impact is unknown

Candidate gene Encoded protein and function Reference


HLA-DPB1*0401 Human leukocyte antigen: antigen presentation. Lyons et al. 2012, Arning et al. 2011
PRTN3 Proteinase 3: Neutrophil serine protease. Autoantigen in GPA. Lyons et al. 2012
SERPINA1 Alpha1 antitrypsin: serine protease inhibition. Lyons et al. 2012
CTLA-4 Cytotoxic T-lymphocyte-associated protein 4: inhibition of T-cell function. Chung et al. 2012
LEPR Leptin receptor: activation of effector T cells, suppression of regulatory T cells. Wieczorek et al. 2010a
IRF5 Interferon regulatory factor 5: diverse roles including virus-mediated activation of interferon Wieczorek et al. 2010b
production, cell growth, and apoptosis.
CD226 DNAX accessory molecule-1 (DNAM-1): Th1-type polarization. Wieczorek et al. 2009
FCGR3B (low copy Fc fragment of immunoglobulin G, low affinity IIIb, receptor: neutrophil activation. Kelley et al. 2011, Fanciulli et al. 2007
number)
PTPN22 Protein tyrosine phosphatase non-receptor 22 (PTPN22): T- and B-cell signalling, altered gene Cao et al. 2012, Jagiello et al. 2005
expression, decreased IL-10 transcription.

appear to determine the clinical course of GPA. Generalized GPA, Environmental factors
that is GPA with renal or other organ-threatening disease, is seen
To date, the most convincing evidence for an association of a bac-
in the vast majority of patients seeking clinical care. Localized GPA,
terial infection with GPA stems from studies showing that nasal
that is GPA solely restricted to the respiratory tract, accounts for 5%
carriage of Staphylococcus aureus (S.  aureus) is associated with
of all cases with GPA (Holle et al. 2010). Localized GPA differs from
endonasal activity and an increased risk for a relapse (Stegeman
generalized GPA with respect to its association with interferon
et al. 1994; Laudien et al. 2010). Transient induction of PR3- and/
regulatory factor (IRF)5 haplotypes controlling interferon response
or MPO-ANCA and vasculitic manifestations is also found in suba-
genes (Wieczorek et al. 2010b).
cute bacterial endocarditis with S. aureus and other Staphylococcus
The first genome-wide association study in AAV confirmed dif-
species, providing circumstantial clinical evidence for the induc-
ferences in the genetic association between GPA and MPA driven
tion and pathogenicity of ANCA (Csernok et al. 2010). Moreover,
by the underlying autoantibody specificity. PR3-ANCA seropositiv-
bacterial DNA motifs (unmethylated CpG oligonucleotides) stimu-
ity typical of GPA is strongly associated with HLA-DP. In contrast,
late ANCA production of circulating autoreactive B cells in patients
myeloperoxidase-specific (MPO)-ANCA seropositivity typical of
with AAV (Hurtado et al. 2008). In contrast, there is little evidence
MPA is associated with HLA-DQ. Further, the Z allele of SERPINA1,
for a causal role of viral infections in the pathogenesis of GPA as
which encodes the serine protease inhibitor alpha1 antitrypsin, and
yet (Table  29.2) (Csernok et  al. 2010). Cytomegalovirus (CMV)
a single nucleotide polymorphism in the PRTN3 gene encoding pro-
infection drives memory T-cell inflation in healthy individuals
teinase 3 are associated with PR3-ANCA (Lyons et al. 2012). Of note,
and patients with GPA (Morgan et al. 2011; O’Hara et al. 2012).
epigenetic control of PR3 gene silencing is also perturbed, result-
However, differences in the phenotype of CMV-specific T cells
ing in increased PR3 expression in neutrophils and monocytes of
and mode of distribution of memory T-cell frequencies between
patients with GPA (Yang et al. 2004; Ciavatta et al. 2010). Genetically
healthy controls and patients with GPA suggest that factors other
determined high constitutive membrane expression of PR3 is asso-
than CMV may have a role in driving T-cell memory inflation
ciated with an increased risk for relapse (Rarok et al. 2002).
in GPA (Lamprecht et al. 2003a; Fagin et al. 2012a). Finally, high

Table 29.2 Environmental factors associated with GPA or association excluded

Environmental factor Specific findings Reference


Staphylococcus aureus Nasal carriage is associated with higher endonasal activity and an increased rate of relapse. Laudien et al. 2010, Stegeman et al. 1994
Gram-negative bacteria LAMP-2 ANCA cross-react with bacterial adhesin FimH suggestive of molecular mimicry. Kain et al. 2012
Cytomegalovirus (CMV) CMV seropositivity is associated with the expansion of CD28− effector memory T cells, Morgan et al. 2011
increased mortality, and risk of infection.
Parvovirus B19 Association excluded in case–control studies. Eden et al. 2003, Nikkari et al. 1995
V9 erythrovirus Association excluded in case–control study. Eden et al. 2003
Silica Inhalation of silicon-containing compounds such as silica and grain dust increases the risk Lee et al. 2012, Hogan et al. 2007, Nuyts
of GPA, silica activate T cells. et al. 1995
CHAPTER 29 granulomatosis with polyangiitis (wegener’s granulomatosis) 387

lifetime exposure to silica (inorganic mineral form of silicone) and Table 29.3 Major pathological manifestations (diagnostic criteria)
silicone (synthetic polymers with a SiO backbone) is also associated of GPA of the lung
with an increased risk of AAV (Nuyts et al. 1995; Hogan et al. 2007;
Lee et al. 2012). Manifestation Features
Drug-induced ANCA-positive vasculitis constitutes an impor-
Vasculitis Arteritis, capillaritis, venulitis
tant differential diagnosis and provides additional circumstantial
Six types: acute, chronic, necrotizing
clinical evidence for the induction and pathogenicity of ANCA. Of
granulomatous, non-necrotizing granulomatous,
note, MPO-ANCA are more frequently detected in drug-induced fibrinoid necrosis, cicatricial changes
ANCA-positive vasculitis than PR3-ANCA. ANCA forma-
tion may evolve as a consequence of propylthiouracil-induced Parenchymal necrosis Neutrophilic microabscess
MPO-neoantigen formation and sulfasalazine-induced Geographic necrosis
up-regulation of PR3 and MPO expression on apoptotic neutro- Granulomatous Microabscess surrounded by granulomatous
phils (Bonaci-Nikolic et al. 2005; Csernok et al. 2010). Necrotizing inflammation / mixed inflammation
inflammation of the nasal mucosa and detection of ANCA with inflammatory infiltrate Palisading histiocytes
specificity for human neutrophil elastase (HNE-ANCA) has been Scattered giant cells
reported in cocaine-induced midline destructive lesions (CIMDL). Poorly formed granulomas
PR3-ANCA are less often found in this disorder. Clinically, CIMDL Sarcoid-like granulomas (rare)
may mimic localized GPA, but it lacks features of systemic AAV
(Trimarchi et al. 2001). (Adapted from Travis et al. 2002.)

Malignancy
Patients with GPA have an increased risk of a preceding or con- Some patients show a bronchially centred disease with necrotiz-
current malignancy with bladder cancer, non-melanoma skin can- ing and/or granulomatous bronchitis, bronchiolitis, and subglot-
cer, leukaemia, and malignant lymphoma (Heijl et al. 2011). The tic stenosis (Figure  29.4). In contrast, well-defined sarcoid-like
cause for this association has remained unclear so far. GPA con- granulomas are rare (Mark et  al. 1988; Kradin and Mark 2002;
veys an increased risk for renal cancer. PR3 mRNA is expressed in Jennette 2011). Individual cases with a marked eosinophilic tissue
renal epithelial and glomerular cell lines and ANCA induce sig- infiltration have been reported (‘eosinophilic variant’) (Yousem and
nalling and growth of renal epithelial cells (Tatsis et al. 1999; Relle Lombard 1988).
et al. 2003). Treatment with cyclophosphamide is associated with In GPA, the inflammatory infiltrate includes monohistiocytic
a 9- to 45-fold increased risk for bladder cancer. Co-medication epithelioid cells and giant cells as well as a large and heterogene-
with 2-mercaptoethane sulfonate sodium (mesna) reduces the ous population of other cells, including clusters of T and B lym-
risk of bladder cancer in cyclophosphamide-treated patients. phocytes, partially with follicular organization, activated and
Cyclophosphamide-induced haemorrhagic cystitis is a risk factor follicular dendritic cells, plasma cells, PR3-expressing neutrophils,
for the development bladder cancer (Hellmich et al. 2004). scattered eosinophils, mast cells, debris-laden macrophages, and
high endothelial venules suggestive of ectopic lymphoid-like tis-
sue neoformation (Figure 29.5) (Mackiewicz et al. 2005; Voswinkel
Pathology
The key defining pathological features of GPA are necrotizing gran-
ulomatous inflammation, with a predilection for the upper and/or
lower respiratory tract, and systemic necrotizing vasculitis, prefer-
entially affecting small to medium-sized pulmonary and renal ves-
sels. Owing to its systemic nature the vasculitis may, however, affect
any organ (Lamprecht et  al. 2009b; Holl-Ulrich 2010; Jennette
2011; Jennette et al. 2013).

Necrotizing granulomatous inflammation


Granulomatous and non-granulomatous extravascular inflam-
mation is common and displays various morphologies in GPA
(Table  29.3) (Holl-Ulrich et  al. 2002; Travis et  al. 2002; Jennette
2011; Jennette et  al. 2013). During the early phase, neutrophilic
microabscesses with scattered multinucleated giant cells prevail
(Figures 29.1 and 29.2). In later phases, degenerating neutrophils
and necrotic debris form a central necrotic irregularly outlined
zone (geographic necrosis), which is surrounded by poorly defined
granulomatous inflammation with palisades of elongated mac-
rophages and scattered multinucleated giant cells (Figure  29.3). Fig. 29.1 GPA—granulomatous and vasculitic lesions in nasal biopsy: diffuse
Large zones of geographic necrosis with irregular serpiginous mar- granulomatous tissue, several dispersed multinucleated giant cells, and
gins bounded by palisading histiocytes and giant cells may be the neutrophilic microabscesses. Vasculitis of small venule (upper right) with
only pattern of inflammation seen with treated chronic disease. onion-skin appearance of vessel wall and intramural inflammatory infiltrates.
388 SECTION 5 vasculitic diseases and syndromes and related disorders

Fig. 29.4 GPA—ulcerating, necrotizing, and granulomatous bronchiolitis (Giemsa


Fig. 29.2 GPA—granulomatous lesions in nasal biopsy: typical ill-defined stain): residual bronchiolar epithelium (left), ulceration and necrosis (centre),
epithelioid cell granuloma (left) with several dispersed multinucleated giant cells bordered by ill-defined granuloma (top), and giant cell (right).
(right), dense mixed inflammatory infiltrate with neutrophilic microabscesses,
lymphocytes, and plasma cells.

et al. 2006; Capraru et al. 2008; Mueller et al. 2008). IL-17, lym- All characteristic hallmarks of GPA can be found in nasal biopsies
phoid chemokines (B-cell attracting chemokine-1, BCA-1), and of active disease (Table 29.4) (Matsubara et al. 1996; Schnabel et al.
B-cell survival factors (B-cell-activating factor of the tumour 1997; Garske et al. 2012).
necrosis factor/TNF family, BAFF, a proliferation-inducing ligand,
APRIL) are expressed within areas of clustering B cells in renal and Vasculitis
nasal inflammatory lesions (Steinmetz et al. 2008; Zhao et al. 2012; In GPA, necrotizing vasculitis predominantly affects small and
Mueller et al. 2008; Mueller personal communication). Similar to medium-sized vessels, that is arteries, arterioles, capillaries, and
findings in the kidney, the majority of IL-17-expressing cells are venules (Figure  29.7). The vasculitis is termed ‘pauci-immune’
neutrophils (Velden et al. 2012; Mueller personal communication). because few or no immunoglobulin and complement deposits are
Moreover, the activating NK-receptor NKG2D, its ligand MHC detected in glomerular lesions and at other inflammatory sites
class I chain-related protein A (MICA), as well as IL-15 are found (Holl-Ulrich 2010; Jennette 2011; Jennette et al. 2013). Classical
in granulomatous lesions (Capraru et al. 2007; De Menthon et al. patterns of vasculitis in GPA comprise necrotizing crescentic
2011). Fibroblasts within the inflammatory infiltrate invade and glomerulonephritis and acute pulmonary capillaritis with alveo-
destroy contiguous nasal cartilage and bone (Figure 29.6) (Kesel lar haemorrhage. Apart from acute capillaritis, other patterns of
et  al. 2012). Endonasal activity ranges from areas of unaltered vasculitis may affect the lung. Necrotizing granulomatous vascu-
mucosa or mild lesions to severe ulcerative and destructive lesions. litis involving medium-sized vessels may be found close to large,
necrotizing, granulomatous pulmonary lesions (Figure  29.8).
Granulomatous vasculitis, that is granulomas within the vessel wall,
is extremely rare outside the lung in GPA. Non-granulomatous
fibrinoid necrosis of vessel walls is rare in the lung, but may affect
bronchial arteries. Cutaneous lesions usually display leukocyto-
clastic vasculitis. As a systemic disease, GPA may evoke vasculitic
lesions in virtually any organ. Vasculitis morphologically identi-
cal to that seen in classical polyarteritis and microscopic poly-
angiitis, both in size of the vessels involved and the presence of
fibrinoid necrosis, may also be seen in GPA (Holl-Ulrich 2010;
Jennette 2011).

Necrotizing crescentic glomerulonephritis


In the kidney, endothelial cells are the target of earliest changes,
giving rise to swelling, necrosis, and de-adherence of the vascular
endothelium (Figure  29.9). Lysed neutrophils are found within
the affected vessels. Degranulation of the neutrophils induces rup-
ture of the basement membrane and glomerular capillary throm-
bosis followed by segmental necrosis of the tuft. Necrotic debris,
Fig. 29.3 GPA—granulomatous lesions in lung biopsy: large, necrotizing blood, and fibrin spill into the Bowman’s space. As a consequence,
granuloma with palisading histiocytes and large, central, geographical necrosis. monocytes and parietal (Bowman’s) epithelial cells accumulate and
CHAPTER 29 granulomatosis with polyangiitis (wegener’s granulomatosis) 389

(a) (b)

Fig. 29.5 GPA—ectopic lymphoid tissue in active nasal GPA: secondary lymphoid follicle in vicinity of granulomatous inflammation; (a) immunohistochemistry showing
B-cell antigen CD20; (b) same area, CD23-positive follicular dendritic cells.

proliferate to form crescents (Figure  29.10) (Harper and Savage glomerulonephritis, analysis of kidney biopsies from patients with
2000). Neutrophils displaying NETosis, that is a death pathway char- AAV showed that the majority of IL-17-expressing cells are neu-
acterized by the formation of neutrophil extracellular traps (NETs), trophils. Th17 cells and mast cells were found less frequently (Gan
are found in inflammatory lesions in the kidney (Kessenbrock et al. et al. 2010; Turner et al. 2010; Velden et al. 2012).
2009). Proinflammatory cytokines (TNF-α, IL-1β, IL-8, IL-18) and Based on light microscopy findings, the histopathological clas-
the IL-2 receptor are expressed in the interstitium, periglomerular, sification of AAV-associated glomerulonephritis considers pre-
and perivascular space (Noronha et al. 1993; Cockwell et al. 1999; dominant focal, crescentic, mixed, and sclerotic lesions (Table 29.5)
Hewins et al. 2006). Clusters of dendritic cells and T and B cells (Berden et al. 2010). Renal outcome correlates with the number of
are also detected in glomerular lesions of patients with AVV, sug- normal glomeruli, tubulointerstitial infiltration with T cells, and
gestive of neoformation of ectopic lymphoid tissue in chronically tubular atrophy at the time of renal biopsy and during follow-up.
inflamed renal tissue (Steinmetz et  al. 2008; Wilde et  al. 2009a). Sclerotic glomerulonephritis is associated with the worst renal
While IL-17-producing CD4+ T-helper cells (Th17) promote survival (de Lind van Wijngaarden et al. 2006; Berden et al. 2010;
inflammation and recruitment of neutrophils in experimental Berden et al. 2012).

(a) (b)

Fig. 29.6 GPA—granulomatous lesions in nasal biopsy: Destruction of nasal cartilage (a) and bone (b) by inflammatory cells and fibroblasts.
390 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 29.4 GPA score: authors’ proposal for a histopathological classification of diagnostic certainty in nasal mucosa biopsies

GPA score of nasal biopsya Histopathological features


I. Non-specific Slight, moderate or dense lymphoplasmocytic infiltrates and/or scattered neutrophils.
II. Suggestive for GPA, but not diagnostic Single or few microabscess(es) or scattered giant cells or deep inflammation down to cartilage and bone
or extensive fibrosis without prior surgery.
III. GPA likely One or two of the following criteriab:
Geographic necrosis
and/or ill-defined granuloma
and/or scattered giant cells
and/or vasculitis
IV. GPA characteristic At least three of the following criteriab:
Geographic necrosis
and/or ill-defined granuloma
and/or scattered giant cells
and/or vasculitis
a Other causes of rhinosinusitis, e.g. mycotic infection, tuberculosis, sarcoidosis, have to be excluded.
b Neutrophilic microabscess and/or deep fibrosis may also be present, but do not count as separate criterion for scores III and IV.
The proposal is based on light microscopy findings in a cohort of GPA patients from the German Vasculitis Pathology Reference Center in Lübeck.

Pathophysiology Barrier dysfunction


As mentioned in the first paragraph of this chapter, data from in One of the striking features of GPA is its peculiar propensity for
vitro and in vivo studies suggest that PR3-ANCA play an important necrotizing granulomatous inflammation of the upper and lower
role in the pathogenesis of systemic vasculitis. However, it is still respiratory tract (Lamprecht et  al. 2009a). Accordingly, respira-
largely unclear how granulomatous inflammation and autoimmun- tory tract manifestations are the most frequent presenting features.
ity are induced in GPA (Sarraf and Sneller 2005; Lamprecht et al. They affect virtually all patients with GPA during follow-up in large
2006; Lamprecht et al. 2009a; Kallenberg 2011; Gadola and Gross cohorts. Moreover, so-called ‘grumbling disease’ related to persis-
2012). Barrier dysfunction and formation of ectopic lymphoid-like tent ENT disease activity is observed in most patients with GPA who
structures potentially sustaining autoimmunity and/or chronic are defined as being otherwise in clinical remission (Hoffman et al.
inflammation are important pathophysiological mechanisms in 1992; Holle et al. 2011). Barrier dysfunction predisposing to abnor-
various disorders such as Crohn’s disease and rheumatoid arthritis. mal microbial composition and mucosa susceptibility to invasion
Evidence is accumulating that these mechanisms may also have a
role in GPA (Lamprecht et al. 2006; Lamprecht et al. 2009a; Wilde
et al. 2011).

Fig. 29.8 GPA—medium-size-vessel granulomatous vasculitis of lung, late


changes (van Gieson stain): narrowing of venous lumen, intimal fibrosis, disruption
of elastic fibres, transmural inflammatory infiltrate of vessel wall, several giant cells
Fig. 29.7 GPA—small -necrotizing vasculitis in nasal biopsy, early (left). Note that granulomatous vasculitis in GPA is exceedingly rare as compared
changes: endothelial swelling of capillaries and venules, adhesion and to necrotizing vasculitis, and mostly confined to the lungs (medium-sized and
transmigration of neutrophils, subendothelial fibrinoid necrosis. large vessels).
CHAPTER 29 granulomatosis with polyangiitis (wegener’s granulomatosis) 391

Table 29.5 Histopathological classification of AAV-associated


glomerulonephritis based on light microscopy findings

Class Inclusion criteria


Focal ≥50% normal glomeruli
Crescentic ≥50% glomeruli with cellular crescents
Sclerotic ≥50% globally sclerotic glomeruli
Mixed <50% normal, <50% crescentic, <50% globally
sclerotic glomeruli

Any biopsy that does not display a predominant focal, crescentic, or sclerotic phenotype is
assigned to the mixed category
(Berden et al. 2010.)

2011). Baseline and inducible IL-8 secretion of nasal epithelial cells


Fig. 29.9 GPA—focal necrotizing glomerulonephritis, acute lesion (PAS
is also reduced, whereas granulocyte colony-stimulating factor
stain): acute focal necrosis of glomerular tufts with early crescent formation (right)
(Courtesy of Dr T. Wiech, Hamburg, Germany). (G-CSF) secretion is increased (Wohlers et al. 2012). Moreover, the
respiratory ciliary function is severely impaired (Ullrich et al. 2009).
These data suggest a potential role of primary or secondary barrier
favouring chronic inflammation is a fundamental aspect of various dysfunction in the pathogenesis of GPA; however, further studies
disorders, for example Crohn’s disease (McGuckin et al. 2009). The are needed to elucidate the role of barrier dysfunction in triggering
nasal mucosa displays a unique transcriptional profile of antimicro- disease activity and infection-associated relapse in GPA.
bial peptide and cytokine expression in GPA, which differs from
that of other inflammatory disorders (Laudien et al. 2011). As men- Neutrophilic microabscesses and
tioned in Section Environmental Factors, nasal carriage of S. aureus geographic necrosis
is associated with endonasal activity and relapses (Stegeman et al. Neutrophilic microabscesses and/or large geographic necrosis are
1994; Laudien et  al. 2010). Bacterial DNA motifs induce ANCA pathological hallmarks of GPA lesions in the upper and/or lower
production by autoreactive B cells in AAV (Hurtado et al. 2008). respiratory tract. Extravascular neutrophil-rich necrosis is not typi-
Physiological elimination of S. aureus and other bacteria depends cal for inflammatory lesions of the two other AAV, that is MPA and
on the expression of antimicrobial peptides in barrier tissues. For EGPA (Churg–Strauss syndrome) (Mark et al. 1988; Kradin and
instance, S. aureus colonization is facilitated by decreased human Mark 2002; Travis et al. 2002; Holl-Ulrich et al. 2010; Jennette 2011).
beta-defensin-3 (hBD-3) production of keratinocytes in atopic Serum levels of high mobility group box protein 1 (HMGB1), an
dermatitis (Kisich et  al. 2008). Intriguingly, the inducible hBD-3 alarmin passively released from necrotic cells and actively secreted
response of nasal epithelial cells is significantly reduced in GPA, sug- by macrophages, correlate with disease activity and the size of
gesting barrier dysfunction favouring S. aureus carriage (Hui et al. inflammatory pulmonary lesions determined by computed tomog-
raphy volumetry in GPA, but not in MPA (Wibisono et al. 2010;
Henes et al. 2011).
How are neutrophilic microabscesses and geographic necrosis
induced in respiratory tract lesions? Previous studies by Fienberg
suggested that the earliest lesions in the lung are foci of swollen
collagen fibres representing tissue injury and ensuing necrosis in
GPA. Neutrophils are attracted by these lesions (Fienberg 1981).
As early lesions (neutrophilic microabscesses) age, they develop a
central zone of degenerated neutrophil and apoptotic debris and
the periphery of the lesions accrues palisades of elongated mac-
rophages and scattered giant cells. The recruitment of macrophages
and fusion of macrophages to giant cells could represent a second-
ary reactive response to the acute necrotic lesions (Mark et al. 1988;
Kradin and Mark 2002; Jennette 2011). Apoptotic debris-laden
macrophages and giant cells surrounding necrotic areas have been
shown in necrotizing granulomatous lesions of the respiratory
tract in GPA (Mackiewicz et al. 2005; Jennette 2011). It is unclear
to what extent necrotic lesions are solely ANCA-induced or result
Fig. 29.10 GPA—focal necrotizing glomerulonephritis, chronic lesion (PAS
from exposure to another initiating injurious agent. Animal mod-
stain): destruction and sclerosis of glomerular tufts, prominent fibrocellular els showing PR3-ANCA-induced acute pulmonary and renal
crescent formation, periglomerular inflammatory infiltrate (top) (Courtesy of Dr vascular injury with focal and segmental necrotizing crescentic
T. Wiech, Hamburg, Germany). glomerulonephritis lack necrotizing granulomatous inflammation
392 SECTION 5 vasculitic diseases and syndromes and related disorders

of the upper and/or lower respiratory tract typical of the patho- PR3-ANCA
logical lesions in GPA. Refinement of the modelling of GPA may
While PR3-ANCA are detected in about 50% of the patients with
further elucidate the chain of events resulting in the formation of
localized GPA, PR3-ANCA are present in more than 90% of the
necrotizing granulomatous lesions in the upper and lower respira-
patients with generalized GPA (Csernok et al. 2006a; Holle et al.
tory tract and systemic vasculitis (Lamprecht et al. 2009a; Salama
2011). ANCA levels may be associated with disease activity and
and Little 2012).
relapse. Persistence of ANCA after induction of remission is
NETosis represents a unique death pathway, differing from
associated with relapse (Sanders et al. 2006). The strength of the
apoptosis and necrosis. It depends on the generation of reactive
association between ANCA and disease activity is a matter of con-
oxygen species and is characterized by the formation of neutro-
tinuing debate as not all studies to date have consistently demon-
phil extracellular traps (NETs), which are chromatin-containing
strated a close correlation (Finkielman et al. 2007; Tomasson et al.
filamentous structures displaying various proteins, including PR3
2012). Apart from their diagnostic value, data from in vitro and
and MPO (Brinkmann and Zychlinsky 2012). NETs are released by
in vivo studies suggest that PR3-ANCA play an important role
ANCA-stimulated neutrophils. Inflammatory lesions in the kidney
in the pathogenesis of systemic vasculitis in GPA. It is inferred
contain NETs, suggesting a role of PR3-ANCA induced NETosis
that ANCA-mediated activation of cytokine-primed neutrophils
in the pathogenesis of GPA (Kessenbrock et al. 2009). Uploading
induces premature intravascular degranulation of neutrophils and
of myeloid dendritic cells with NETs induces ANCA in susceptible
endothelial cell damage (Harper and Savage 2000; Csernok et al.
mice (Sangaletti et al. 2012). Moreover, priming of T cells by NETs
2006b; Falk and Jennette 2008; Lamprecht et al. 2009b; Kallenberg
increases T-cell responses to various antigens (Tillack et al. 2012). As
2011; Wilde et al. 2011).
mentioned in Section Necrotizing Crescentic Glomerulonephritis,
the majority of IL-17-expressing cells are neutrophils in inflamma- PR3-ANCA-induced neutrophil activation and degranulation
tory renal and nasal lesions in AAV (Velden et al. 2012; Mueller PR3-ANCA-induced activation of neutrophils primed with com-
personal communication). Interestingly, neutrophils and mast cells plement factor C5a and cytokines (TNF-α, IL-1) results in respira-
produce IL-17 upon extracellular trap formation in psoriasis sug- tory burst and degranulation of neutrophils with release of oxygen
gestive of a link between NETosis and IL-17 production by innate radicals, superoxide, endothelial cell-activating microparticles, and
immune cells (Lin et al. 2011). serine proteases in vitro (Csernok et al. 1994; Kettritz et al. 1997;
Franssen et al. 1999; Hao et al. 2012; Hong et al. 2012). Interaction
Ectopic lymphoid tissue of ANCA with both its target antigen on the membrane and
Neoformation of ectopic lymphoid-like structures in chronically Fcγ-receptors IIa or IIIb is required for full activation of neutrophils
inflamed tissue is observed in many infectious and autoimmune (Csernok et al. 1994; Porges et al. 1994; Reumaux et al. 1995; Kettritz
diseases (Aloisi and Pujol-Borrell 2006). Nobel laureate Rolf et al. 1997; David et al. 2005). ANCA-induced neutrophil activation
Zinkernagel argues that while physiologically antigen is trans- requires adherence of cells (Reumaux et al. 1995). Membrane-bound
ferred to a draining lymph node to mount an immune response, PR3 is constitutively expressed by neutrophils (Rarok et al. 2002).
in autoimmune disease lymphoid tissue is ‘exported’ into periph- Its expression is further enhanced by the translocation of PR3 from
eral tissue. Ectopic lymphoid-like tissue may help to maintain an intracellular granules to the surface membrane upon stimulation
immunopathological response against an autoantigen, which is with proinflammatory cytokines such as TNF-α and IL-1 (Csernok
otherwise tolerated. In this scenario the autoimmune response et  al. 1994; Witko-Sarsat et  al. 1999; Brachemi et  al. 2007). The
does not subside unless the antigen expressed in the target cells adhesion molecule Mac-1 (CD11b/CD18) and glycoprotein NB1
is eliminated (Zinkernagel 2002). As mentioned in Section (CD177) form a receptor complex binding PR3 on the neutrophil
Necrotizing Granulomatous Inflammation, chronic inflammatory cell surface (David et al. 2003; von Vietinghoff et al. 2007; Jerke et al.
infiltrates in GPA also display features of ectopic lymphoid-like 2011). NB1-negative individuals also display membrane expression
tissue neoformation. PR3-expressing neutrophils, dendritic cells, of PR3 and are susceptible to PR3-ANCA-mediated respiratory
and cells of the adaptive immune response (T and B cells, plasma burst, suggestive of membrane anchors for PR3 other than CD177
cells) are clustered in the infiltrate (Voswinkel et al. 2006; Capraru (Hu et al. 2009). Binding of ANCA with membrane-bound PR3 and
et al. 2007; Steinmetz et al. 2008; Mueller et al. 2008). Local fol- Fcγ-receptors IIa or IIIb could account for differences in the acti-
licle formation and B-cell survival appears to be sustained by vation of neutrophils compared to immunoglobulin-induced acti-
IL-17, lymphoid chemokines (BCA-1), and B-cell survival factors vation solely via Fcγ-receptors. ANCA activate the (p101/p110g)
(e.g. APRIL) (Steinmetz et al. 2008; Zhao et al. 2012; Mueller per- isoform of phosphatidylinositol-3-kinase (PI3K) in vitro. In con-
sonal communication). Immunoglobulin variable heavy (IgVH) trast, sole Fcγ-receptor ligation activates the (p85/p110) PI3K iso-
chain gene analysis suggests antigen-driven affinity maturation form (Ben-Smith et al. 2001). PI3K, p38 mitogen-activated protein
and clonal B-cell expansion in these areas (Voswinkel et al. 2006). kinase (MAPK), extracellular signal-regulated kinase (ERK), and
Lesional B cells display reactivity for various target antigens, for diacylglycerol kinase have a role in mediating C5a-induced activa-
example lysosomal transmembrane protein 9b and tetraspanin tion, translocation of PR3 to the outer membrane of neutrophils,
7 (CD231) (Thurner et al. 2011). Local memory T-cell differen- and ANCA-induced respiratory burst (Kettritz et al. 2002; Holden
tiation and expression of the activating NK-receptor NKG2D and et al. 2011a; van der Veen et al. 2011; Hao et al. 2012). Various genes,
its ligand MICA appears to be induced by IL-15 in granuloma- including DIF-2, COX-2, and IL-8, are up-regulated following stim-
tous lesions (Capraru et al. 2007; De Menthon et al. 2011). Future ulation with ANCA (Yang et al. 2002).
studies will clarify whether and how neoformation of ectopic PR3-ANCA recognize a limited number of conformational
lymphoid-like tissue plays an integral and necessary role in the epitopes (Silva et al. 2010). They may interfere with the enzymatic
autoimmune response to PR3. activity of PR3 and complexation of PR3 with alpha 1-antitrypsin
CHAPTER 29 granulomatosis with polyangiitis (wegener’s granulomatosis) 393

(van der Geld et  al. 2002). Epitope-specific PR3-ANCA subsets 2001). In line with these studies, it was shown that macrophages
with variable impact on biological functions of PR3 have been and giant cells contain phagocytosed neutrophil and apoptotic
postulated. Epitope specificity apparently changes during the dis- debris in necrotizing granulomatous lesions of the respiratory tract
ease course as a result of epitope spreading or other factors (Rarok in GPA (Mackiewicz et al. 2005; Jennette 2011). The presence of
et  al. 2003; Silva et  al. 2010). Moreover, the IgG subclass as well debris-laden macrophages was also reported in inflammatory
as glycosylation and sialylation levels are associated with disease infiltrates in a PR3-ANCA treated chimeric mice model (Little
activity and differences in neutrophil activation, for example IL-8 et al. 2012). It has been speculated that local antigen presentation
release and oxidative burst (Colman et al. 2007; Espy et al. 2011). is maintained by such macrophages (Mackiewicz et al. 2005). As
Deficiency or dysfunction of serine-protease inhibitors, such as mentioned in Section Neutrophilic microabscesses and geographic
alpha 1-antitypsin, may facilitate enzymatic activity of PR3 and necrosis, NETosis represents a unique death pathway differing from
PR3-ANCA-mediated neutrophil activation (Esnault et  al. 1993; apoptosis and necrosis. NETs are released by ANCA-stimulated
Callea et al. 1997; Lyons et al. 2012). neutrophils, potentially sustaining autoimmunity and vasculitis and
PR3 is also a constituent of monocytic granules. Similar to neu- they are present in inflammatory renal lesions (Kessenbrock et al.
trophil activation by ANCA, cross-linking of the target antigen and 2009). As mentioned in Section Neutrophilic Microabscesses and
Fcγ-receptor is important in ANCA-induced monocyte activation. Geographic Necrosis, NETs facilitate T-cell responses to antigen.
PR3-ANCA induces monocyte cytokine (TNF-α, IL-1β, IL-6, IL-8) NETs-stimulated myeloid dendritic cells induces ANCA produc-
and prostanoid (thromboxane A2) release (Ralston et  al. 1997; tion in susceptible mice (Sangaletti et al. 2012; Tillack et al. 2012).
Hattar et al. 2002). PR3 expression is lost with monocytic differ-
Ex Vivo and in vivo models of PR3-ANCA-induced vasculitis
entiation to macrophages (Charles et  al. 1992). Finally, antibod-
ies with dual reactivity to plasminogen and complementary PR3 While the neutrophil rolling-flux fraction remains relatively con-
possibly facilitating venous thrombosis have been reported in GPA stant, adhesion is stabilized and transmigration of cytokine-primed
(Pendergraft et al. 2004; Bautz et al. 2008). ANCA-stimulated neutrophils increased on an endothelial cell layer
in vitro (Radford et al. 2001). Moreover, PR3-ANCA, and to a lesser
PR3-ANCA: apoptosis and NETosis extent MPO-ANCA, interfere with endothelial repair (Le Roux
ANCA accelerate apoptosis and secondary necrosis in TNF-primed et al. 2012). MPO-ANCA-induced neutrophil adhesion and trans-
neutrophils (Harper et al. 2000). PR3 is translocated to the surface migration was observed by intravital microscopy in a rat model of
of apoptotic neutrophils, where it associates with calreticulin and ANCA-associated vasculitis (Little et al. 2005). As GPA and MPA
scramblase-1. The PR3/scramblase-1 and PR3/calreticulin com- display diverse clinical and pathological characteristics, one has to
plexes interfere with the phagocytosis of apoptotic neutrophils by be careful to generalize findings from MPO-ANCA vasculitis mod-
macrophages and elicit an inflammatory response in vitro (Kantari els to both AAV. Owing to structural differences between human
et al. 2007; Gabillet et al. 2012). Opsonization of apoptotic neutro- PR3 and its rodent homologue and human and rodent neutrophil
phils with PR3-ANCA enhances their clearance by macrophages PR3 expression, modelling of PR3-ANCA-induced vasculitis is
and induces TNF-α production (Moosig et al. 2000; Harper et al. challenging (Table  29.6) (Salama and Little 2012). Pathogenicity

Table 29.6 Animal models of PR3-ANCA-induced pathology

Model Specific findings Reference


Co-implantation of inflamed nasal mucosa from Fibroblast-mediated destruction of contiguous nasal cartilage and bone. Kesel et al. 2012
GPA-patients and allogeneic cartilage into pfp/
rag(−/−) immunodeficient mice.
Humanized chimeric mouse: transplantation of Induction of acute vasculitic lung injury and pauci-immune crescentic glomerulonephritis Little et al. 2012
human haematopoietic stem cells into irradiated by passive transfer of human PR3-ANCA. Majority of recruited leukocytes were of murine
NOD/scid/IL-2Rγ(−/−) mice. origin. No granuloma formation.
PR3-humanized transgenic mice. No induction of renal or other pathology by ex vivo generated (hybridoma) monoclonal Relle et al. 2013
mouse anti-human PR3-ANCA.
Isolated lung perfusion model in rats. Respiratory burst and protease-dependent acute lung injury induced by human Hattar et al. 2010
PR3-ANCA-activated TNF-α-primed neutrophils. No granuloma formation.
Transfer of splenocytes from recombinant murine Induction of crescentic glomerulonephritis in NOD-SCID recipient mice. Presence or Primo et al. 2010
PR3 immunized autoimmunity prone NOD mice absence of immunoglobulin and/or complement deposits is not mentioned. No granuloma
into NOD-SCID recipients. formation. By contrast, no pathology developed in C57BL/6-Rag-1(−/−) mice suggestive of a
role of the genetic background and degree of immunodeficiency on disease pathology.
Immunization of mice and rats with different Induction of PR3-ANCA in mice and rats. No pulmonary or renal pathology. Van der Geld
combinations of recombinant murine and et al. 2007
human PR3
Immunization of PR3 and elastase-deficient No pulmonary or renal pathology. Aggravation of TNF-α-induced dermal panniculits by Pfister et al. 2004
mice with murine PR3 and transfer of resultant transfer of murine PR3-ANCA.
autoantibodies into LPS-primed mice.
394 SECTION 5 vasculitic diseases and syndromes and related disorders

of PR3-ANCA resulting in acute vascular pulmonary and renal TEM contain distinct subsets including so-called CD45RA ‘rever-
damage has been demonstrated in three different animal mod- tants’ (TEMRA) and Th1-type CD4+ T cells lacking co-stimulatory
els. Vasculitis developed in the absence or presence of only few T CD28 expression, that is CD28− T cells (Moosig et  al. 1998;
and B cells (Hattar et al. 2010; Primo et al. 2010; Little et al. 2012). Komocsi et al. 2002; Lamprecht et al. 2003b; Fagin et al. 2012a).
Granuloma formation was not observed in these models, lending Anomalous expression of various receptors has been reported on
support for the hypothesis that the pathogenesis of necrotizing CD4+ TEM, for example the activating NKG2D receptor, the inhibi-
granulomatous inflammation predominantly found in the respira- tory receptor PD-1, the co-stimulatory receptors CD134 and GITR
tory tract may be separate from acute systemic vasculitis and in (glucocorticoid-induced TNF-receptor-related protein), and vari-
particular T-cell-dependent in GPA (Salama and Little 2012; Little ous CC-type chemokine receptors indicating activation and migra-
et al. 2012). In a different model, fibroblast-mediated destruction of tory potential (Lamprecht et al. 2003b; Capraru et al. 2008; Wilde
contiguous nasal cartilage and bone was demonstrated, suggestive et al. 2009b; de Menthon et al. 2011; Fagin et al. 2012b; Wilde et al.
of a role of fibroblasts in the formation of saddle-nose deformity in 2012). CD28− TEM are also enriched in bronchoalveolar fluid of
GPA (Kesel et al. 2012). patients with active pulmonary involvement (Lamprecht et al. 2001).
Furthermore, the majority of CD4+ T cells lack CD28 expression
LAMP-2 ANCA in granulomatous lesions (Komocsi et  al. 2002). As discussed in
To what extent autoantibodies with specificity for lysosomal Section Environmental Factors, CMV drives memory T-cell infla-
membrane protein-2 (LAMP-2 ANCA) play a role as a biomarker tion in healthy individuals and patients with GPA (Morgan et al.
and in the pathogenesis of AAV is subject to continuing research 2011; O’Hara et al. 2012). However, differences in the phenotype of
and debate (Kain et  al. 2012; Roth et  al. 2012). LAMP-2 ANCA CMV-specific T cells and the mode of distribution of memory cell
display cross-reactivity to bacterial adhesin FimH and induce frequencies between healthy controls and patients with GPA sug-
pauci-immune crescentic glomerulonephritis in susceptible rats, gest a role of factors other than CMV in driving memory cell infla-
suggestive of molecular mimicry as potential immune mechanism tion in GPA (Lamprecht et al. 2003a; Fagin et al. 2012a). Of note,
for the break of tolerance and induction of autoimmunity. However, an altered dendritic cell response is seen along with the alterations
FimH-immunized rats do not develop PR3- or MPO-ANCA (Kain in the T-cell compartment. Circulating plasmacytoid dendritic cells
et al. 2008). display an altered phenotype. Similar to TEM, they decrease with
disease flares in AAV (Rimbert et al. 2011).
T lymphocytes IL-15 promotes memory T-cell differentiation and induces
T cells play a crucial role in regulating immune responses. Failure NKG2D expression (de Menthon et  al. 2011). It is expressed in
of T-cell control predisposes to chronic inflammation and autoim- granulomatous lesions where it could sustain local T-cell differ-
munity, that is two of the key pathological and immunological fea- entiation and survival (Capraru et  al. 2007). Increased IL-7 and
tures of GPA (Lamprecht et al. 2009a). T cells are abundant and IL-15 receptor alpha chain (CD127, CD215) co-expression could
to some extent clustered in granulomatous and vasculitic lesions favour the response of CD4+ T cells to IL-7 and IL-15 (Klapa et al.
in GPA (Cunningham et al. 1999; Mueller et al. 2000; Wilde et al. 2012). Intriguingly, CD4+NKG2D+ T cells display NK-cell-like
2009b). A  CD8+ T-cell transcription signature, including genes cytotoxicity towards microvascular endothelial cells in vitro (de
of the interleukin-7 receptor (IL-7R) pathway and T-cell receptor Menthon et  al. 2011). Subject to confirmation by other studies,
(TCR) signalling, is associated with poor prognosis (McKinney T-cell-mediated vascular damage might constitute a novel mecha-
et al. 2010). Studies in a delayed-type hypersensitivity (DTH) model nism in the induction of vasculitis adding to PR3-ANCA-induced
suggest that few antigen-specific T cells are sufficient to initiate cel- damage.
lular migration to an antigen-presenting site in peripheral tissue.
The immune response is sustained by concomitant recruitment of Skewed cytokine response
non-specific bystander cells and local proliferation of memory T Early studies showed a skewed Th1-type cytokine response of the
cells (Ghani et al. 2009; Doebis et al. 2011). total population of circulating CD4+ T cells in GPA (Lúdvíksson
et  al. 1998; Csernok et  al. 1999). Still unproven, granulomatous
Altered phenotype and potential vascular cytotoxicity inflammation was hypothesized to be initiated by an aberrant
Persistent expansion of circulating effector memory T cells (TEM) cell-mediated immune response and driven by activated Th1-type
suggests a fundamental alteration of the T-cell response in GPA cells (Sarraf and Sneller 2005). IL-17-producing subsets have been
(Popa et  al. 1999; Marinaki et  al. 2006; Abdulahad et  al. 2006). identified in the total circulating T-cell population. CD45RClow
Oligoclonality and shortened telomers result from clonal expan- Th17 cells are increased in peripheral blood (Ordonez et al. 2009).
sion and replicative senescence of TEM (Grunewald et  al. 1998; Circulating CCR6+ T cells produce IL-17 and IL-22 in GPA (Fagin
Vogt et al. 2003). Percentages of circulating TEM decrease during et al. 2012b). T cells proliferate in response to PR3, but clear-cut
active disease and increase with remission, possibly reflecting recir- evidence of an immunodominant linear PR3-epitope is missing in
culation from blood to inflammatory sites and back (Abdulahad GPA (Popa et  al. 2002). The frequency of PR3-specific T cells is
et al. 2006). In line with this notion, TEM are found in urine dur- low in peripheral blood (Winek et al. 2004). PR3-stimulated den-
ing renal activity (Abdulahad et al. 2009). Interestingly, memory dritic cells induce a stronger Th1-type response in patients with
cell ‘prestages’, so-called very early memory T cells (T VEM), are also GPA compared to healthy controls in vitro (Csernok et al. 2006b).
increased in the circulation (Fagin et  al. 2012b). Conversely, the Nevertheless, the cytokine response of circulating PR3-specific T
fraction of circulating naïve T cells (TN) is decreased. TN display cells is found to be skewed towards an increase of Th2-type, Th17,
an activated phenotype in individual patients (Marinaki et al. 2005; Th17/Th1, Th17/Th2, and Th22 cell fractions (Popa et  al. 2002;
Abdulahad et al. 2006; Fagin et al. 2012b). Abdulahad et al. 2008; Nogueira et al. 2010; Fagin et al. 2011). The
CHAPTER 29 granulomatosis with polyangiitis (wegener’s granulomatosis) 395

cause of the skewed T-cell response to PR3 is unclear. In EGPA Abdulahad, W.H., Stegeman, C.A., van der Geld, Y.M., Doornbos-van der
(Churg–Strauss) PR3-specific cells display a Th2-type dominated Meer, B., Limburg, P.C., and Kallenberg, C.G. (2007). Functional defect
response suggesting that the cytokine response of PR3-specific cells of circulating regulatory CD4+ T cells in patients with Wegener’s granu-
lomatosis in remission. Arthritis and Rheumatism, 56, 2080–91.
is influenced by the underlying disorder (Fagin et al. 2011).
Abdulahad, W.H., van der Geld, Y.M., Stegeman, C.A., and Kallenberg,
Both Th1-type and Th17 cells are considered important in chronic C.G. (2006). Persistent expansion of CD4+ effector memory T cells in
inflammation and autoimmunity (Steinman 2008). In animal mod- Wegener’s granulomatosis. Kidney International, 70, 938–47.
els of encephalitis and glomerulonephritis, infiltration with Th17 Aloisi, F. and Pujol-Borrell, R. (2006). Lymphoid neogenesis in chronic
cells during the early phase is followed by Th1-type cell infiltration inflammatory diseases. Nature Reviews Immunology, 6, 205–17.
at later stages (Murphy et al. 2010; Paust et al. 2012). Transition Arning, L., Holle, J.U., Harper, L., et al. (2011). Are there specific genetic risk
from Th17 to Th1-type cellular infiltration gives rise to a succession factors for the different forms of ANCA-associated vasculitis? Annals of
from neutrophil- to macrophage-dominated lesions (Kroenke et al. the Rheumatic Diseases, 70, 707–8.
2008). Tissue-related chemokine- and cytokine-expression as well Bautz, D.J., Preston, G.A., Lionaki, S., et al. (2008). Antibodies with dual reac-
as Th17 cell plasticity are important in Th17 to Th1-type transition tivity to plasminogen and complementary PR3 in PR3-ANCA vasculitis.
Journal of the American Society of Nephrology, 19, 2421–9.
(Lee et al. 2009; Murphy et al. 2010; Paust et al. 2012). In line with
Ben-Smith, A., Dove, S.K., Martin, A., Wakelam, M.J., and Savage, C.O.
the data from animal models, Th1-type CD4+CD28− T cells have
(2001). Antineutrophil cytoplasm autoantibodies from patients with
been reported in granulomatous lesions (Komocsi et al. 2002), but systemic vasculitis activate neutrophils through distinct signaling cas-
the majority of IL-17-expressing cells in nasal and kidney biopsies cades: comparison with conventional Fcgamma receptor ligation. Blood,
from patients are neutrophils and not Th17 cells, suggesting IL-17 98, 1448–55.
cross-presentation and/or genuine neutrophil-derived production Berden, A.E., Ferrario, F., Hagen, E.C., et al. (2010). Histopathologic classifi-
in GPA (Velden et al. 2012; Mueller personal communication). cation of ANCA-associated glomerulonephritis. Journal of the American
Society of Nephrology, 21, 1628–36.
Regulatory T cells Berden, A.E., Jones, R.B., Erasmus, D.D., et al. (2012). Tubular lesions predict
Regulatory T cells (Treg) limit immune responses (Lamprecht et al. renal outcome in antineutrophil cytoplasmic antibody-associated glo-
2009a; Wilde et  al. 2011). Regulatory T cells display an altered merulonephritis after rituximab therapy. Journal of the American Society
phenotype and impaired suppressor activity towards PR3-induced of Nephrology, 23, 313–21.
T-cell proliferation in GPA, potentially favouring inflammation Bonaci-Nikolic, B., Nikolic, M.M., Andrejevic, S., Zoric, S., and Bukilica,
and break of tolerance (Abdulahad et al. 2007; Morgan et al. 2010; M. (2005). Antineutrophil cytoplasmic antibody (ANCA)-associated
autoimmune diseases induced by antithyroid drugs: comparison with
Klapa et al. 2010; Rimbert et al. 2011). Treg are interspersed in the
idiopathic ANCA vasculitides. Arthritis Research and Therapy, 7,
inflammatory infiltrate in GPA (Klapa et al. 2010). Future studies R1072–81.
will show whether plasticity of CD4+ T cells is altered, resulting Brachemi, S., Mambole, A., Fakhouri, F., et al. (2007). Increased membrane
in transition from PR3-specific Treg to Th17 cells (Lee et al. 2009; expression of proteinase 3 during neutrophil adhesion in the pres-
Kallenberg 2011). ence of anti proteinase 3 antibodies. Journal of the American Society of
Nephrology, 18, 2330–9.
B Lymphocytes Brinkmann, V. and Zychlinsky, A. (2012). Neutrophil extracellular traps: Is
Circulating B cells from patients with high ANCA titres spontaneously immunity the second function of chromatin? Journal of Cellular Biology,
198, 773–83.
secrete ANCA (Clayton and Savage 2003; Tadema et al. 2011). As men-
Callea, F., Gregorini, G., Sinico, A., et  al. (1997). Alpha 1-Antitrypsin
tioned earlier, bacterial DNA motifs (unmethylated CpG oligonucleo- (AAT) deficiency and ANCA-positive systemic vasculitis: genetic and
tides) stimulate ANCA production by circulating autoreactive B cells in clinical implications. European Journal of Clinical Investigation, 27,
patients with AAV, thereby possibly playing a role in infection-triggered 696–702.
relapses (Hurtado et al. 2008; Tadema et al. 2011). PR3-ANCA induce Cao, Y., Yang, J., Colby, K., et al. (2012). High basal activity of the PTPN22
B lymphocyte stimulator (BLys) production from primed neutrophils, gain-of-function variant blunts leukocyte responsiveness negatively
potentially perpetuating the autoimmune response. Serum BLys levels affecting IL-10 production in ANCA vasculitis. PLOS one, 7, e42783.
are increased during active disease (Holden et al. 2011b). As discussed Capraru, D., Müller, A., Csernok, E., et al. (2008). Expansion of circulating
in Section Necrotizing Granulomatous Inflammation, inflammatory NKG2D+ effector memory T-cells and expression of NKG2D-ligand
MIC in granulomatous lesions in Wegener’s granulomatosis. Clinical
infiltrates in GPA display features of ectopic lymphoid-like tissue neo-
Immunology, 127, 144–50.
formation with clusters of B cells and plasma cells (Voswinkel et al.
Charles, L.A., Falk, R.J., and Jennette, J.C. (1992). Reactivity of antineutro-
2006; Steinmetz et al. 2008; Mueller et al. 2008). However, how loss phil cytoplasmic autoantibodies with mononuclear phagocytes. Journal
of tolerance and PR3-ANCA production by autoreactive plasma cells of Leukocyte Biology, 51, 65–8.
are induced at the onset of disease and subsequently sustained still Cho, J.H. and Gregersen, P.K. (2011). Genomics and the multifactorial nature
remains an enigma. of human autoimmune disease. New England Journal of Medicine, 365,
1612–23.
References Chung, S.A., Xie, G., Roshandel, D., et al. (2012). Meta-analysis in granulo-
Abdulahad, W.H., Kallenberg, C.G., Limburg, P.C., and Stegeman, C.A. matosis with polyangiitis reveals shared susceptibility loci with rheuma-
(2009). Urinary CD4+ effector memory T cells reflect renal disease toid arthritis. Arthritis and Rheumatism, 64, 3463–71.
activity in antineutrophil cytoplasmic antibody-associated vasculitis. Ciavatta, D.J., Yang, J., Preston, G., et al. (2010). Epigenetic basis for aberrant
Arthritis and Rheumatism, 60, 2830–8. upregulation of autoantigen genes in humans with ANCA vasculitis.
Abdulahad, W.H., Stegeman, C.A., Limburg, P.C., and Kallenberg, C.G. Journal of Clinical Investigation, 120, 3209–19.
(2008). Skewed distribution of Th17 lymphocytes in patients with Clayton, A.R. and Savage, C.O. (2003). Production of antineutrophil cyto-
Wegener’s granulomatosis in remission. Arthritis and Rheumatism, 58, plasm antibodies derived from circulating B cells in patients with sys-
2196–205. temic vasculitis. Clinical and Experimental Immunology, 132, 174–9.
396 SECTION 5 vasculitic diseases and syndromes and related disorders

Cockwell, P., Brooks, C.J., Adu, D., and Savage, C.O. (1999). Fagin, U., Pitann, S, Gross, W.L., and Lamprecht, P. (2012b). Increased
Interleukin-8:  A  pathogenetic role in antineutrophil cytoplasmic frequency of CCR4+ and CCR6+ memory T-cells including
autoantibody-associated glomerulonephritis. Kidney International, 55, CCR7+CD45RAmed very early memory cells in granulomatosis with
852–63. polyangiitis (Wegener’s). Arthritis Research and Therapy, 14, R73.
Colman, R., Hussain, A., Goodall, M., et al. (2007). Chimeric antibodies to Falk, R.J. and Jennette, J.C. (2008). New insight into the pathogensis of vascu-
proteinase 3 of IgG1 and IgG3 subclasses induce different magnitudes of litis associated with antineutrophil cytoplasmic autoantibodies. Current
functional responses in neutrophils. Annals of the Rheumatic Diseases, Opinion in Rheumatology, 20, 55–60.
66, 676–82. Fanciulli, M., Norsworthy, P.J., Petretto, E., et  al. (2007). FCGR3B copy
Csernok, E., Ai, M., Gross, W.L., et al. (2006b). Wegener autoantigen induces number variation is associated with susceptibility to systemic, but not
maturation of dendritic cells and licenses them for Th1 priming via the organ-specific, autoimmunity. Nature Genetics, 39, 721–3.
protease-activated receptor-2 pathway. Blood, 107, 4440–8. Fienberg, R. (1981). The protracted superficial phenomenon in pathergic
Csernok, E., Ernst, M., Schmitt, W.H., Bainton, D.F., and Gross, W.L. (1994). (Wegener’s) granulomatosis. Human Pathology, 12, 458–67.
Activated neutrophils express proteinase 3 on their plasma membrane Finkielman, J.D., Merkel, P.A., Schroeder, D., et al. (2007). Antiproteinase 3
in vitro and in vivo. Clinical and Experimental Immunology, 95, 244–50. antineutrophil cytoplasmic antibodies and disease activity in Wegener
Csernok, E., Lamprecht P., and Gross, W.L. (2006a). Diagnostic significance granulomatosis. Annals of Internal Medicine, 147, 611–9.
of ANCA in vasculitis. Nature Clinical Practise Rheumatology 2, 174–5. Franssen, C.F., Huitema, M.G., and Muller Kobold, A.C. (1999). In vitro
Csernok, E., Lamprecht, P., and Gross, W.L. (2010). Clinical and neutrophil activation by antibodies to proteinase 3 and myeloperoxi-
immunological features of drug-induced and infection-induced dase from patients with crescentic glomerulonephritis. Journal of the
proteinase 3-antineutrophil cytoplasmic antibodies and American Society of Nephrology, 10, 1506–15.
myeloperoxidase-antineutrophil cytoplasmic antibodies and vasculitis. Gabillet, J., Millet, A., Pederzoli-Ribeil, M., et al. (2012). Proteinase 3, the
Current Opinion in Rheumatology, 22, 43–8. autoantigen in granulomatosis with polyangiitis, associates with calreti-
Csernok, E., Trabandt, A., Müller, A., et  al. (1999). Cytokine profiles in culin on apoptotic neutrophils, impairs macrophage phagocytosis, and
Wegener’s granulomatosis: predominance of type 1 (Th1) in the granu- promotes inflammation. Journal of Immunology, 189, 2574–83.
lomatous inflammation. Arthritis and Rheumatism, 42, 742–50. Gadola, S.D. and Gross, W.L. (2012). Vasculitis in 2011: the renaissance of
Cunningham, M.A., Huang, X.R., Dowling, J.P., Tipping, P.G., and granulomatous inflammation in AAV. Nature Reviews Rheumatology,
Holdsworth, S.R. (1999). Prominence of cell-mediated immunity effec- 8, 74–6.
tors in ‘pauci-immune’ glomerulonephritis. Journal of the American Gan, P.Y., Steinmetz, O.M., Tan, D.S., et  al. (2010). Th17 cells promote
Society of Nephrology, 10, 499–506. autoimmune anti-myeloperoxidase glomerulonephritis. Journal of the
David, A., Fridlich, R., and Aviram, I. (2005). The presence of membrane American Society of Nephrology, 21, 925–31.
Proteinase 3 in neutrophil lipid rafts and its colocalization with Garske, U., Haack, A., Beltran, O., et al. (2012). Intra- and inter-rater reliabil-
FcgammaRIIIb and cytochrome b558. Experimental Cell Research, 308, ity of endonasal activity estimation in granulomatosis with polyangii-
156–65. tis (Wegener’s). Clinical and Experimental Rheumatology, 30 (1 Suppl.
David, A., Kacher, Y., Specks, U., and Aviram, I. (2003). Interaction of pro- 70), S22–8.
teinase 3 with CD11b/CD18 (beta2 integrin) on the cell membrane of Ghani, S., Feuerer, M., Doebis, C., et al. (2009). T cells as pioneers: antigen-specific
human neutrophils Journal of Leukocyte Biology, 74, 551–7. T cells condition inflamed sites for high-rate antigen-non-specific effector
de Lind van Wijngaarden, R.A., Hauer, H.A., Wolterbeek, R., et  al. cell recruitment. Immunology, 128 (Suppl. 1), e870–80.
(2006). Clinical and histologic determinants of renal outcome in Grunewald, J., Halapi, E., Wahlström, J., et  al. (1998). T-cell expansions
ANCA-associated vasculitis: A prospective analysis of 100 patients with with conserved T-cell receptor β chain motifs in the peripheral blood
severe renal involvement. Journal of the American Society of Nephrology, of HLA-DRB1*0401 positive patients with necrotizing vasculitis. Blood,
17, 2264–74. 92, 3737–44.
De Menthon, M., Lambert, M., Guiard, E., et  al. (2011). Excessive Hao, J., Meng, L.Q., Xu, P.C., Chen, M., and Zhao, M.H. (2012). p38MAPK,
interleukin-15 transpresentation endows NKG2D+CD4+ T cells with ERK and PI3K signaling pathways are involved in C5a-primed neutro-
innate-like capacity to lyse vascular endothelium in granulomatosis with phils for ANCA-mediated activation. PLOS One, 7, e38317.
polyangiitis (Wegener’s). Arthritis and Rheumatism, 63, 2116–26. Harper, L., Cockwell, P., Adu, D., and Savage, C.O. (2001). Neutrophil priming
Doebis, C., Menning, A., Neumann, K., et al. (2011). Accumulation and local and apoptosis in anti-neutrophil cytoplasmic autoantibody-associated
proliferation of antigen-specific CD4+ T cells in antigen-bearing tissue. vasculitis. Kidney International, 59, 1729–38.
Immunology and Cell Biology, 89, 566–72. Harper, L., Ren, Y., Savill, J., Adu, D., and Savage, C.O. (2000). Antineutrophil
Eden, A., Mahr, A., Servant, A., et al. (2003). Lack of association between cytoplasmic antibodies induce reactive oxygen-dependent dysregula-
B19 or V9 erythrovirus infection and ANCA-positive vasculitides:  a tion of primed neutrophil apoptosis and clearance by macrophages.
case-control study. Rheumatology, 42, 660–4. American Journal of Pathology, 157, 211–20.
Esnault, V.L., Testa, A., Audrain, M., et  al. (1993). Alpha 1-antitrypsin Harper, L. and Savage, C.O.S. (2000). Pathogenesis of ANCA-associated sys-
genetic polymorphism in ANCA-positive systemic vasculitis. Kidney temic vasculitis. Journal of Patholology, 190, 349–359.
International, 43, 1329–32. Hattar, K., Bickenbach, A., Csernok, E., et al. (2002). Wegener’s granuloma-
Espy, C., Morelle, W., Kavian, N., et  al. (2011). Sialylation levels of tosis: antiproteinase 3 antibodies induce monocyte cytokine and pros-
anti-proteinase 3 antibodies are associated with the activity of granu- tanoid release-role of autocrine cell activation. Journal of Leukocyte
lomatosis with polyangiitis (Wegener’s). Arthritis and Rheumatism, 63, Biology, 71, 996–1004.
2105–15. Hattar, K., Oppermann, S., Ankele, C., et  al. (2010). C-ANCA-induced
Fagin, U., Csernok, E., Müller, A., et al. (2011). Distinct proteinase 3-induced neutrophil-mediated lung injury: a model of acute Wegener’s granulo-
cytokine patterns in Wegener’s granulomatosis, Churg-Strauss syn- matosis. European Respiratory Journal, 36, 187–95.
drome, and healthy controls. Clinical and Experimental Rheumatology, Heijl, C., Harper, L., Flossmann, O., et al. (2011). Incidence of malignancy in
29 (Suppl. 64), S57–62. patients treated for antineutrophil cytoplasm antibody-associated vas-
Fagin, U., Pitann, S, Gross, W.L., and Lamprecht, P. (2012a). Flow cytomet- culitis: follow-up data from European Vasculitis Study Group clinical
ric characterization of ‘early’ and ‘late differentiated' T-cells including trials. Annals of the Rheumatic Diseases, 70, 1415–21.
PR3-specific cells in granulomatosis with polyangiitis (Wegener’s). Hellmich, B., Kausch, I., Doehn, C., Jocham, D., Holl-Ulrich, K., and Gross,
Cytometry Part B (Clinical Cytometry), 82, 173–5. W.L. (2004). Urinary bladder cancer in Wegener’s granulomatosis: Is it
CHAPTER 29 granulomatosis with polyangiitis (wegener’s granulomatosis) 397

more than cyclophosphamide? Annals of the Rheumatic Diseases, 63, Jerke, U., Rolle, S., Dittmar, G., et al. (2011). Complement receptor Mac-1 is
1183–85. an adaptor for NB1 (CD177)-mediated PR3-ANCA neutrophil activa-
Henes, F.O., Chen, Y., Bley, T.A., et al. (2011). Correlation of serum level of tion. Journal of Biological Chemistry, 286, 7070–81.
high mobility group box 1 with the burden of granulomatous inflam- Kain, R., Exner, M., Brandes, R., et  al. (2008a). Molecular mimicry in
mation in granulomatosis with polyangiitis (Wegener’s). Annals of the pauci-immune focal necrotizing glomerulonephritis. Nature Medicine,
Rheumatic Diseases, 70, 1926–9. 14, 1088–96.
Herlyn, K., Buckert, F., Gross, W.L., and Reinhold-Keller, E. (2010). Verdopplung Kain, R., Tadema, H., McKinney, E.F., et al. (2012). High prevalence of autoan-
der Periodenprävalenz von ANCA-assoziierten Vaskulitiden und tibodies to hLAMP-2 in anti-neutrophil cytoplasmic antibody-associated
Riesenzellarteriitis von 1994 bis 2006 in Schleswig-Holstein [Doubling vasculitis. Journal of the American Society of Nephrology, 23, 556–66.
oft the periodic prevalence of ANCA-associated vasculitides and giant Kallenberg, C.G. (2011). Pathogenesis of ANCA-associated vasculitides.
cell arteritis from 1994 until 2006 in Schleswig-Holstein]. Medizinische Annals of the Rheumatic Diseases, 70, i59–i63.
Klinik, 105 (Suppl. 1), 37. Kantari, C., Pederzoli-Ribeil, M., Amir-Moazami, O., et  al. (2007).
Hewins, P., Morgan, M.D., Holden, N., et al. (2006). IL-18 is upregulated in Proteinase 3, the Wegener autoantigen, is externalized during neutro-
the kidney and primes neutrophil responsiveness in ANCA-associated phil apoptosis: evidence for a functional association with phospholipid
vasculitis. Kidney International, 69, 605–15. scramblase 1 and interference with macrophage phagocytosis. Blood,
Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., et al. (1992). Wegener’s granulomato- 110, 4086–95.
sis: an analysis of 158 patients. Annals of Internal Medicine, 116, 488–98. Kelley, J.M., Monach, P.A., Ji, C., et al. (2011). IgA and IgG antineutrophil
Hogan, S.L., Cooper, G.S., Savitz, D.A., et  al. (2007). Association of silica cytoplasmic antibody engagement of Fc receptor genetic variants influ-
exposure with anti-neutrophil cytoplsamic autoantibody small-vessel ences granulomatosis with polyangiitis. Proceedings of the National
vasculitis: a population-based, case-control study. Clinical Journal of the Academy of Science USA, 108, 20736–41.
American Society of Nephrology, 2, 290–9. Kesel, N., Köhler, D., Herich, L., et al. (2012). Cartilage destruction in granu-
Holden, N.J., Savage, C.O., Young, S.P., Wakelam, M.J., Harper, L., and lomatosis with polyangiitis (Wegener’s granulomatosis) is mediated by
Williams, J.M. (2011a). A dual role for diacylglycerol kinase gener- human fibroblasts after transplantation into immunodeficient mice.
ated phosphatidic acid in autoantibody-induced neutrophil exocytosis. American Journal of Pathology, 180, 2144–55.
Molceular Medicine, 17, 1242–52. Kessenbrock, K., Krumbholz, M., Schönermarck, U., et al. (2009). Netting
Holden, N.J., Williams, J.M., Morgan, M.D., et  al. (2011b). neutrophils in autoimmune small-vessel vasculitis. Nature Medicine,
ANCA-stimulated neutrophils release BLyS and promote B cell sur- 15, 623–5.
vival:  a clinically relevant cellular process. Annals of the Rheumatic Kettritz, R., Choi, M., Butt, W., Rane, M., Rolle, S., Luft, F.C., and Klein, J.B.
Diseases, 70, 2229–33. (2002). Phosphatidylinositol 3-kinase controls antineutrophil cytoplas-
Holle, J.U., Gross, W.L., Holl-Ulrich, K., et al. (2010). Prospective long-term mic antibodies-induced respiratory burst in human neutrophils. Journal
follow-up of patients with localised Wegener’s granulomatosis: does it of the American Society of Nephrology, 13, 1740–9.
occur as persistent disease stage? Annals of the Rheumatic Diseases, 69, Kettritz, R., Jennette, J.C., and Falk, R.J. (1997). Crosslinking of
1934–9. ANCA-antigens stimulates superoxide release by human neutrophils.
Holle, J.U., Gross, W.L., Latza, U., et al. (2011). Improved outcome in 445 Journal of the American Society of Nephrology, 8, 386–94.
patients with Wegener’s granulomatosis in a German Vasculitis Center Kisich, K.O., Carspecken, C.W., Fiéve, S., Boguniewicz, M., and Leung, D.Y.
over four decades. Arthritis and Rheumatism, 63, 257–66. (2008). Defective killing of Staphylococcus aureus in atopic dermatitis is
Holl-Ulrich, K. (2010). Histopathology of systemic vasculitis. Pathologie, associated with reduced mobilization of human beta-defensin-3. Journal
31, 67–76. of Allergy and Clinical Immunology, 122, 62–8.
Holl-Ulrich, K., Reinhold-Keller, E., Müller, A., and Feller, A.C. (2002). Klapa, S., Mueller, A, Csernok, E., et al. (2010). Lower numbers of FoxP3
Pathology of vasculitis:  Differential diagnosis and selected disorders. and CCR4 co-expressing cells in an elevated subpopulation of
Verhandlungen der Deutsche Gesellschaft für Pathologie, 86, 83–90. CD4+CD25high regulatory T cells from Wegener’s granulomatosis.
Hong, Y., Eleftheriou, D., Hussain, A.A., et  al. (2012). Anti-neutrophil Clinical and Experimental Rheumatology, 28 (1 Suppl. 57), 72–80.
cytoplasmic antibodies stimulate release of neutrophil microparticles. Klapa, S., Schüler, S., Pitann, S., Klenerman, P., Gross, W.L., and Lamprecht,
Journal of the American Society of Nephrology, 23, 49–62. P. (2012). Increased frequency of IL-7 and IL-15 receptor alpha chain
Hu, N., Westra, J., Huitema, M.G., et al. (2009). Coexpression of CD177 and (CD127, CD215) co-expressing CD4(+) T cells in granulomatosis with
membrane proteinase 3 on neutrophils in antineutrophil cytoplasmic polyangiitis (Wegener’s). Clinical and Experimental Rheumatology, 30 (1
autoantibody-associated systemic vasculitis: anti-proteinase 3-mediated Suppl. 70), S171.
neutrophil activation is independent of the role of CD177-expressing Knudsen, B.B., Joergensen, T., and Munch-Jensen, B. (1988). Wegener’s
neutrophils. Arthritis and Rheumatism, 60, 1548–57. granulomatosis in a family. Scandinavian Journal of Rheumatology,
Hui, Y., Wohlers, J., Podschun, R., et al. (2011). Antimicrobial peptides in 17, 225–7.
nasal secretion and mucosa with respect to S.  aureus colonisation in Komocsi, A., Lamprecht, P., Csernok, E., et  al. (2002). Peripheral blood
Wegener’s granulomatosis. Clinical and Experimental Rheumatology, 29 and granuloma CD4(+)CD28(-) T cells are a major source of
(Suppl. 64), S49–56. interferon-gamma and tumor necrosis factor-alpha in Wegener’s granu-
Hurtado, P.R., Jeffs, L., Nitschke, J., et al. (2008). CpG oligodeoxynucleotide lomatosis. American Journal of Pathology, 160, 1717–24.
stimulates production of anti-neutrophil cytoplasmic antibodies in Kradin, R.L. and Mark, E.J. (2002). Case records of the Massachusetts
ANCA associated vasculitis. Biomed Central Immunology, 9, 34. General Hospital:  Weekly clnicopathological exercises. Case 18-2002,
Jagiello, P., Aries, P., Arning, L., et al. (2005). The PTPN22 620W polymor- A  48-year-old man with a cough and bloody sputum. New England
phism is associated with the occurrence of autoantibodies to proteinase Journal of Medicine, 346, 1892–9.
3 in Wegener’s granulomatosis. Arthritis and Rheumatism, 52, 4039–43. Kroenke, M.A., Carlson, T.J., Andjelkovic, A.V., and Segal, B.M. (2008).
Jennette, J.C. (2011). Nomenclature and classification of vasculitis: lessons IL-12- and IL-23-modulated T cells induce distinct types of EAE based
learned from granulomatosis with polyangiitis (Wegener’s granuloma- on histology, CNS chemokine profile, and response to cytokine inhibi-
tosis). Clinical and Experimental Immunology, 164 (Suppl. 1), 7–10. tion. Journal of Experimental Medicine, 205, 1535–41.
Jennette, J.C., Falk, R., Bacon, P.A., et al. (2013). 2012 revised international Lamprecht, P., Csernok, E., and Gross, W.L. (2006). Effector memory T cells
Chapel Hill consensus conference nomenclature of vasculitides. Arthritis as driving force of granuloma formation and autoimmunity in Wegener’s
and Rheumatism, 65, 1–11. granulomatosis. Journal of Internal Medicine, 260, 187–91.
398 SECTION 5 vasculitic diseases and syndromes and related disorders

Lamprecht, P., Erdmann, A., Mueller, A., et  al. (2003b). Heterogeneity of McGuckin, M.A., Eri, R., Simms, L.A., Florin, T.H., and Radford-Smith, G.
CD4 and CD8+ memory T cells in localized and generalized Wegener’s (2009). Intestinal barrier dysfunction in inflammatory bowel diseases.
granulomatosis. Arthritis Research and Therapy, 5, R25–31. Inflammatory Bowel Diseases, 15, 100–13.
Lamprecht, P., Holle, J., and Gross, W.L. (2009b). Update on clinical, patho- McKinney, E.F., Lyons, P.A., Carr, E.J., et  al. (2010). A CD8+ T cell tran-
physiological and therapeutic aspects in ANCA-associated vasculitides. scription signature predicts prognosis in autoimmune disease. Nature
Current Drug Discovery Technologies, 6, 241–51. Medicine, 16, 586–91.
Lamprecht, P., Moosig, F., Csernok, E., et al. (2001). CD28 negative T cells are Moosig, F., Csernok, E., Kumanovics, G., and Gross, W.L. (2000).
enriched in granulomatous lesions of the respiratory tract in Wegener’s Opsonization of apoptotic neutrophils by anti-neutrophil cytoplasmic
granulomatosis. Thorax, 56, 751–7. antibodies (ANCA) leads to enhanced uptake by macrophages and
Lamprecht, P., Vargas Cuero, A.L., Muller, A., et al. (2003a). Alterations in increased release of tumour necrosis factor-alpha (TNF-alpha). Clinical
the phenotype of CMV-specific and total CD8+ T-cell populations in and Experimental Immunology, 122, 499–503.
Wegener’s granulomatosis. Cellular Immunology, 224, 1–7. Moosig, F., Csernok, E., Wang, G., and Gross, W.L. (1998). Costimulatory
Lamprecht, P., Wieczorek, S., Epplen, J.T., Ambrosch, P., and Kallenberg, C.G. molecules in Wegener’s granulomatosis (WG):  lack of expression of
(2009a). Granuloma formation in ANCA-associated vasculitides. Acta CD28 and preferential up-regulation of its ligands B7-1 (CD80) and B7-2
Pathologica Microbiologica et Immunologica, 127 (Suppl.), 32–6. (CD86) on T cells. Clinical and Experimental Immunology, 114, 113–8.
Laudien, M., Gadola, S.D., Podschun, R., et  al. (2010). Nasal carriage of Morgan, M.D., Day, C.J., Piper, K.P., et al. (2010). Patients with Wegener’s
Staphylococcus aureus and endonasal activity in Wegener s granuloma- granulomatosis demonstrate a relative deficiency and functional impair-
tosis as compared to rheumatoid arthritis and chronic Rhinosinusitis ment of T-regulatory cells. Immunology, 130, 64–73.
with nasal polyps. Clinical and Experimental Rheumatology, 28 (Suppl. Morgan, M.D., Pachnio, A., Begum, J., et  al. (2011). CD4+CD28- T cell
57), 51–5. expansion in granulomatosis with polyangiitis (Wegener’s) is driven by
Laudien, M., Häsler, R., Wohlers, J., et  al. (2011). Molecular signatures of latent cytomegalovirus infection and is associated with an increased risk
a disturbed nasal barrier function in the primary tissue of Wegener’s of infection and mortality. Arthritis and Rheumatism, 63, 2127–37.
granulomatosis. Mucosal Immunology, 4, 564–73. Mueller, A., Holl-Ulrich, K., Lamprecht, P., and Gross, W.L. (2008). Germinal
Lee, Y.K., Mukasa, R., Hatton, R.D., and Weaver, C.T. (2009). Developmental centre-like structures in Wegener’s granuloma: the morphological basis
plasticity of Th17 and Treg cells. Current Opinion in Immunology, 21, for autoimmunity? Rheumatology, 47, 1111–3.
274–80. Mueller, A., Trabandt, A., Gloeckner-Hofmann, et  al. (2000). Localized
Lee, S., Hayashi, H., Maeda M., et al. (2012). Environmental factors produc- Wegener’s granulomatosis:  predominance of CD26 and IFN-gamma
ing autoimmune dysregulation—chronic activation of T cells caused by expression. Journal of Pathology, 192, 113–20.
silica exposure. Immunobiology, 217, 743–8. Murphy, A.C., Lalor, S.J., Lynch, M.A., and Mills, K.H. (2010). Infiltration of
Le Roux, S., Pepper, R.J., Dufay, A., et al. (2012). Elevated soluble Flt1 inhib- Th1 and Th17 cells and activation of microglia in the CNS during the
its endothelial repair in PR3-ANCA-associated vasculitis. Journal of the course of experimental autoimmune encephalomyelitis. Brain Behavior
American Society of Nephrology, 23, 155–64. and Immunity, 24, 641–51.
Lin, A.M., Rubin, C.J., Khandpur, R., et al. (2011). Mast cells and neutrophils Nikkari, S., Vainionpää, R., Toivanen, P., et al. (1995). Wegener’s granulo-
release IL-17 through extracellular trap formation in psoriasis. Journal matosis and parvovirus B19 infection. Arthritis and Rheumatism, 38,
of Immunology, 187, 490–500. 1175–6.
Little, M.A., Smyth, C.L., Yadav, R., et  al. (2005). Antineutrophil cyto- Nogueira, E., Hamour, S., Sawant, D., et al. (2010). Serum IL-17 and IL-23
plasm antibodies directed against myeloperoxidase augment levels and autoantigen-specific Th17 cells are elevated in patients with
leukocyte-microvascular interactions in vivo. Blood, 106, 2050–8. ANCA-associated vasculitis. Nephrology Dialysis and Transplantation,
Little, M.A., Al-Ani, B., Ren, S., et al. (2012). Anti-proteinase 3 anti-neutrophil 25, 2209–17.
cytoplasm autoantibodies recapitulate systemic vasculitis in mice with a Noronha, I.L., Krüger, C., Andrassy, K., Ritz, E., and Waldherr, R. (1993). In
humanized immune system. PLOS One, 7, e28626. situ production of TNF-alpha, IL-1 beta and IL-2R in ANCA-positive
Lúdvíksson, B.R., Sneller, M.C., Chua, K.S., et al. (1998). Active Wegener’s glomerulonephritis. Kidney International, 43, 682–92.
granulomatosis is associated with HLA-DR+ CD4+ T cells exhibiting Ntatsaki, E., Watts, R.A., and Scott, D.G.I. (2010). Epidemiology of
an unbalanced Th1-type T cell cytokine pattern:  reversal with IL-10. ANCA-associated vasculitis. Rheumatic Disease Clinics of North
Journal of Immunology, 160, 3602–9. America, 36, 447–61.
Lyons, P.A., Rayner, T.F., Trivedi, S., et al. (2012). Genetically distinct subsets Nuyts, G.D., van Vlem, E., de Vos, A., et al. (1995). Wegener’s granulomato-
within ANCA-associated vasculitis. New England Journal of Medicine, sis is associated to exposure to silicon compounds: a case-control study.
367, 214–23. Nephrology Dialysis and Transplantation, 10, 1162–5.
Mackiewicz, Z., Rimkevicius, A., Petersen, J., et  al. (2005). Macrophages O’Hara, G.A., Welten, S.P.M., Klenerman, P., and Arens, R. (2012). Memory
overloaded with tissue debris in Wegener’s granulomatosis. Annals of T-cell inflation: understanding cause and effect. Trends in Immunology,
the Rheumatic Diseases, 64, 1239–29. 33, 84–90.
Marinaki, S., Kälsch, A.I., Grimminger, P., et al. (2006). Persistent T-cell Ordonez, L., Bernard, I., l’Faqihi-Olive, F.E., Tervaert, J.W., Damoiseaux, J.,
activation and clinical correlations in patients with ANCA-associated and Saoudi, A. (2009). CD45RC isoform expression identifies function-
systemic vasculitis. Nephrology Dialysis and Transplantation, 21, ally distinct T cell subsets differentially distributed between healthy
1825–32. individuals and AAV patients. PLOS One, 4, e5287.
Marinaki, S., Neumann, I., Kälsch, A.I., et al. (2005). Abnormalities of CD4 Paust, H.J., Turner, J.E., Riedel, J.H., et al. (2012). Chemokines play a criti-
T cell subpopulations in ANCA-associated vasculitis. Clinical and cal role in the cross-regulation of Th1 and Th17 immune responses
Experimental Immunology, 140, 181–91. in murine crescentic glomerulonephritis. Kidney International,
Mark, E.J., Matsubara, O., Tan-Liu, N.S., and Fienberg, R. (1988). The pulmo- 82, 72–83.
nary biopsy in the early diagnosis of Wegener’s (pathergic) granuloma- Pendergraft, W.F. 3rd, Preston, G.A., Shah, R.R., et al. (2004). Autoimmunity
tosis. Human Pathology, 19, 1065–71. is triggered by cPR-3(105-201), a protein complementary to human
Matsubara, O., Yoshimura, N., Doi, Y, Tamura, A., and Mark, E.J. (1996). autoantigen proteinase-3. Nature Medicine, 10, 72–9.
Nasal biopsy in the early diagnosis of Wegener’s (pathergic) granuloma- Pfister, H., Ollert, M., Fröhlich, L.F., et al. (2004). Antineutrophil cytoplasmic
tosis. Significance of palisading granuloma and leukocytoklastic vascu- autoantibodies against the murine homolog of proteinase 3 (Wegener
litis. Virchows Archiv, 428, 13–9. autoantigen) are pathogenic in vivo. Blood, 104, 1411–18.
CHAPTER 29 granulomatosis with polyangiitis (wegener’s granulomatosis) 399

Popa, E., Stegeman, C.A., Bos, N.A., Kallenberg, C.G., and Tervaert, J.W. Silva, F., Hummel, A.M., Jenne, D.E., and Specks, U. (2010). Discrimination
(1999). Differential B- and T-cell activation in Wegener’s granulomato- and variable impact of ANCA binding to different surface epitopes on
sis. Journal of Allergy and Clinical Immunology, 103, 885–94. proteinase 3, the Wegener’s autoantigen. Journal of Autoimmunity, 35,
Popa, E.R., Franssen, C.F., Limburg, P.C., Huitema, M.G., Kallenberg, 299–308.
C.G., and Tervaert, J.W. (2002). In vitro cytokine production and Stegeman, C.A., Tervaert, J.W., Sluiter, W.J., Manson, W.L., de Jong, P.E., and
proliferation of T cells from patients with anti-proteinase 3- and Kallenberg, C.G.M. (1994). Association of chronic nasal carriage of
antimyeloperoxidase-associated vasculitis, in response to proteinase 3 Staphylococcus aureus and higher relapse rates in Wegener’s granulo-
and myeloperoxidase. Arthritis and Rheumatism, 46, 1894–1904. matosis. Annals of Internal Medicine, 120, 12–7.
Porges, A.J., Redecha, P.B., Kimberly, W.T., Csernok, E., Gross, W.L., Steinman, L. (2008). A rush to judgment on Th17. Journal of Experimental
and Kimberly, R.P. (1994). Antineutrophil cytoplasmic antibodies Medicine, 205, 1517–22.
engage and activate human neutrophils via Fc gamma RIIa. Journal of Steinmetz, O.M., Velden, J., Kneissler, U., et al. (2008). Analysis and classifica-
Immunology, 153, 1271–80. tion of B-cell infiltrates in lupus and ANCA-associated nephritis. Kidney
Primo, V.C., Marusic, S., Franklin, C.C., et  al. (2010). Anti-PR3 immune International, 74, 448–67.
responses induce segmental and necrotizing glomerulonephritis. Tadema, H., Abdulahad, W.H., Lepse, N., Stegeman, C.A., Kallenberg, C.G.,
Clinical and Experimental Immunology, 159, 327–37. and Heeringa, P. (2011). Bacterial DNA motifs trigger ANCA produc-
Radford, D.J., Luu, N.T., Hewins, P., Nash, G.B., and Savage, C.O. (2001). tion in ANCA-associated vasculitis in remission. Rheumatology, 50,
Antineutrophil cytoplasmic antibodies stabilize adhesion and promote 689–96.
migration of flowing neutrophils on endothelial cells. Arthritis and Tatsis, E., Reinhold-Keller, E., Steindorf, K., Feller, A.C., and Gross, W.L.
Rheumatism, 44, 2851–61. (1999). Wegener’s granulomatosis associated with renal cell carcinoma.
Ralston, D.R., Marsh, C.B., Lowe, M.P., and Wewers, M.D. (1997). Arthritis and Rheumatism, 42, 751–6.
Antineutrophil cytoplasmic antibodies induce monocyte IL-8 release. Thurner, L., Müller, A., Cerutti, M., et al. (2011). Wegener’s granuloma harbors
Role of surface proteinase-3, alpha1-antitrypsin, and Fcgamma recep- B lymphocytes with specificities against proinflammatory transmem-
tors. Journal of Clinical Investigation, 100, 1416–24. brane protein and a tetraspanin. Journal of Autoimmunity, 36, 87–90.
Rarok, A.A, Stegeman, C.A., Limburg, P.C., and Kallenberg, C.G. (2002). Tillack, K., Breiden, P., Martin, R., and Sospedra, M. (2012). T lymphocyte
Neutrophil membrane expression of proteinase 3 (PR3) is related to priming by neutrophil extracellular traps links innate and adaptive
relapse in PR3-ANCA-associated vasculitis. Journal of the American immune responses. Journal of Immunology, 188, 3150–9.
Society of Nephrology, 13, 2232–8. Tomasson, G., Grayson, P.C., Mahr, A., LaValley, M. and Merkel, P. (2012).
Rarok, A.A., van der Geld, Y.M., Stegeman, C.A., Limburg, P.C., and Value of ANCA measurements during remission to predict a relapse of
Kallenberg, C.G. (2003). Diversity of PR3-ANCA epitope specificity ANCA-associated vasculitis—a meta-analysis. Rheumatology, 51, 100–9.
in Wegener’s granulomatosis. Analysis using the biosensor technology. Travis, W.D., Colby, T.V., and Koss, M.N. (2002). Non-neoplastic disorders of
Journal of Clinical Immunology, 23, 460–8. the lower respiratory tract. Atlas of nontumor pathology, 1st Series, Fasc.2.
Relle, M., Cash, H., Schommers, N., Reifenberg, K., Galle, P.R., and American Registry of Pathology/Armed Forces Institute of Pathology,
Schwarting, A. (2013). PR3 antibodies do not induce renal pathology Washington, DC.
in a novel PR3-humanized mouse model for Wegener’s granulomatosis. Trimarchi, M., Gregorini, G., Facchetti, F., et  al. (2001). Cocaine-induced
Rheumatology International, 33, 613–22. midline destructive lesions: clinical, radiographic, histopathologic, and
Relle, M., Mayet, W.J., Strand, D., Brenner, W., Galle, P.R., and Schwarting, A. serologic features and their differentiation from Wegener granulomato-
(2003). Proteinase 3/myeloblastin as a growth factor in human kidney sis. Medicine, 80, 391–404.
cells. Journal of Nephrology, 16, 831–40. Turner, J.E., Paust, H.J., Steinmetz, O.M., et al. (2010). CCR6 recruits regula-
Reumaux, D., Vossebeld, P.J., Roos, D., and Verhoeven, A.J. (1995). Effect of tory T cells and Th17 cells to the kidney in glomerulonephritis. Journal
tumor necrosis factor-induced integrin activation on Fc gamma receptor of the American Society of Nephrology, 21, 974–85.
II-mediated signal transduction: relevance for activation of neutrophils by Ullrich, S., Gustke, H., Lamprecht, P., et al. (2009). Severely impaired res-
anti-proteinase 3 or anti-myeloperoxidase antibodies. Blood, 86, 3189–95. piratory ciliary function in Wegener’s granulomatosis. Annals of the
Rimbert, M., Hamidou, M., Braudeau, C., et al. (2011). Decreased numbers Rheumatic Diseases, 68, 1067–71.
of blood dendritic cells and defective function of regulatory T cells in Van der Geld, Y.M., Tool, A.T., Videler, J., et  al. (2002). Interference of
antineutrophil cytoplasmic antibody-associated vasculitis. PLOS One, 6, PR3-ANCA with the enzymatic activity of PR3: differences in patients
e18734. during active disease or remission of Wegener’s granulomatosis. Clinical
Roth, A.J., Brown, M.C., Smith, R.N., et al. (2012). Anti-LAMP-2 antibodies are and Experimental Immunology, 129, 562–70.
not prevalent in patients with antineutrophil cytoplasmic autoantibody glo- Van der Geld, Y.M., Hellmark, T., Selga, D., et  al. (2007). Rats and mice
merulonephritis. Journal of the American Society of Nephrology, 23, 545–55. immunised with chimeric human/mouse proteinase 3 produce autoan-
Salama, A.D. and Little, M. (2012). Animal models of antineutrophil cyto- tibodies to mouse PR3 and rat granulocytes. Annals of the Rheumatic
plasm antibody-associated vasculitis. Current Opinion in Rheumatology, Diseases, 66, 1679–82.
24, 1–7. Van der Veen, B.S., Chen, M., Müller, R., et  al. (2011). Effects of p38
Sanders, J.S., Huitma, M.G., Kallenberg, C.G., and Stegeman, C.A. (2006). mitogen-activated protein kinase inhibition on anti-neutrophil cyto-
Prediction of relapses in PR3-ANCA-associated vasculitis by assess- plasmic autoantibody pathogenicity in vitro and in vivo. Annals of the
ing responses of ANCA titres to treatment. Rheumatology (Oxford), Rheumatic Diseases, 70, 356–65.
45, 724–9. Vang, T., Miletic, A.V., Bottini, N., and Mustelin, T. (2007). Protein tyros-
Sangaletti, S., Tripodo, C., Chiodoni, C., et al. (2012). Neutrophil extracellu- ine phosphatase PTPN22 in human autoimmunity. Autoimmunity 10,
lar traps mediate transfer of cytoplasmic neutrophil antigens to myeloid 453–61.
dendritic cells towards ANCA induction and associated autoimmunity. Velden, J., Paust, H.J., Hoxha, E., et  al. (2012). Renal IL-17 expression in
Blood, 120, 3007–18. human ANCA-associated glomerulonephritis. American Journal of
Sarraf, P. and Sneller, M. (2005). Pathogenesis of Wegener’s granulomato- Physiology—Renal Physiology, 302, F1663–73.
sis: current concepts. Expert Reviews in Molecular Medicine, 7, 1–19. Vogt, S., Iking-Konert, C., Hug, F., Andrassy, K., and Hänsch, G.M. (2003).
Schnabel, A., Holl-Ulrich, K., Dalhoff, K., Reuter, M., and Gross, W.L. (1997). Shortening of telomeres:  Evidence for replicative senescence of T
Efficacy of transbronchial biopsy in pulmonary vaculitides. European cells derived from patients with Wegener’s granulomatosis. Kidney
Respiratory Journal, 10, 2738–43. International, 63, 2144–51.
400 SECTION 5 vasculitic diseases and syndromes and related disorders

Von Vietinghoff, S., Tunnemann, G., Eulenberg, C., et al. (2007). NB1 medi- Wilde, B., van Paassen, P., Damoiseaux, J., et al. (2009a). Dendritic cells in
ates surface expression of the ANCA antigen proteinase 3 on human renal biopsies of patients with ANCA-associated vasculitis. Nephrology
neutrophils. Blood, 109, 4487–93. Dialysis and Transplantation, 24, 2151–6.
Voswinkel, J., Mueller, A., Kraemer, J.A., et al. (2006). B lymphocyte matura- Wilde, B., van Paassen, P., Witzke, O., and Cohen Tervaert, J.C. (2011). New
tion in Wegener’s granulomatosis: a comparative analysis of VH genes pathophysiological insights and treatment of ANCA-associated vasculi-
from endonasal lesions. Annals of the Rheumatic Diseases, 65, 859–64. tis. Kidney International, 79, 599–612.
Watts, R.A., Mooney, J., Skinner, J., Scott, D.G.I., and MacGregor, A.J. Winek, J., Mueller, A., Csernok, E., Gross, W.L., and Lamprecht, P. (2004).
(2012). The contrasting epidemiology of granulomatosis with poly- Frequency of proteinase 3 (PR3)-specific autoreactive T cells deter-
angiitis (Wegener’s) and microscopic polyangiitis. Rheumatology, 51, mined by cytokine flow cytometry in Wegener’s granulomatosis. Journal
926–31. of Autoimmunity, 22, 79–85.
Wibisono, D., Csernok, E., Lamprecht, P., Holle, J.U., Gross, W.L., and Moosig, Witko-Sarsat, V., Cramer, E.M., Hieblot, C., et al. (1999). Presence of pro-
F. (2010). Serum HMGB1 levels are increased in active Wegener’s granu- teinase 3 in secretory vesicles:  evidence of a novel, highly mobiliz-
lomatosis and differentiate between active forms of ANCA-associated able intracellular pool distinct from azurophil granules. Blood, 94,
vasculitis. Annals of the Rheumatic Diseases, 69, 1888–9. 2487–96.
Wieczorek, S., Hoffjan, S., Chan, A., et al. (2009). Novel association of the Wohlers, J., Breucker, K., Podschun, R., et al. (2012). Aberrant cytokine pat-
CD226 (DNAM-1) Gly307Ser polymorphism in Wegener’s granuloma- tern of the nasal mucosa in granulomatosis with polyangiitis. Arthritis
tosis and confirmation for multiple sclerosis in German patients. Genes Research and Therapy, 14, R203
and Immunity, 10, 591–5. Yang, J.J., Pendergraft, W.F., Alcorta, D.A., et al. (2004). Circumvention of
Wieczorek, S., Holle, J.U., Bremer, J.P., et al. (2010a). Contrasting associa- normal constraints on granule protein gene expression in peripheral
tion of a non-synonymous leptin receptor gene polymorphism with blood neutrophils and monocytes of patients with antineutrophil cyto-
Wegener’s granulomatosis and Churg-Strauss syndrome. Rheumatology, plasmic autoantibody-associated glomerulonephritis. Journal of the
49, 907–14. American Society of Nephrology, 15, 2103–14.
Wieczorek, S., Holle, J.U., Müller, S., Fricke, H., Gross, W.L., and Epplen, J.T. Yang, J.J., Preston, G.A., Alcorta, D.A., et al. (2002). Expression profile of leu-
(2010b). A functionally relevant IRF5 haplotype is associated with reduced kocyte genes activated by anti-neutrophil cytoplasmic autoantibodies
risk to Wegener’s granulomatosis. Journal of Molecular Medicine, 88, 413–21. (ANCA). Kidney International, 62, 1638–49.
Wilde, B., Dolff, S., Cai, X., et  al. (2009b). CD4+CD25+ T-cell popula- Yousem, S.A. and Lombard, C.M. (1988). The eosinophilic variant of
tions expressing CD134 and GITR are associated with disease activity Wegener’s granulomatosis. Human Pathology, 19, 682–8.
in patients with Wegener’s granulomatosis. Nephrology Dialysis and Zhao, Y., Odell, E., Choong, L.M., et al. (2012). Granulomatosis with poly-
Transplantation, 24, 161–71. angiitis involves sustained mucosal inflammation that is rich in B-cell
Wilde, B., Hua, F., Dolff, S., et al. (2012). Aberrant expression of the nega- survival factors and autoantigen. Rheumatology, 51, 1580–6.
tive costimulator PD-1 on T cells in granulomatosis with polyangiitis. Zinkernagel, R.M. (2002). Antiinfection immunity and autoimmunity.
Rheumatology (Oxford), 51, 1188–97. Annals of the New York Academy of Science, 958, 3–6.
CHAPTER 30

Granulomatosis with
polyangiitis (Wegener’s
granulomatosis): clinical and
immunodiagnostic aspects
Susanne Schinke, Wolfgang L. Gross, and
Elena Csernok

Introduction Yet the pathological hallmark of GPA is the coexistence of granu-


lomatous inflammation and vasculitis.
Granulomatosis with polyangiitis (GPA; Wegener’s granulomatosis)
is a systemic inflammatory disease with a broad clinical spectrum.
The disease was first described in 1931 by a medical student at the Nomenclature, definitions,
Charité in Berlin, Heinz Klinger, who mistakenly believed it to be and classification
an atypical form of polyarteritis nodosa (Klinger 1932). At the 29th The nomenclature for all vasculitides have been revised by the
Meeting of the German Society of Pathology in Breslau, Friedrich Chapel Hill Consensus Conference (CHCC) 2012 (see Chapter 1)
Wegener, a good friend of Klinger’s, reported on the post-mortem (Jennette et al. 2013) replacing historical eponyms with a patho-
findings in three of his patients. He was able to convince famous logical description within the name of the disease. The revised
German pathologists, including Ludwig Aschoff (Freiburg), that CHCC nomenclature 2012 defines GPA as necrotizing granu-
the findings represented a new disease entity (reviewed in Socias lomatous inflammation of the upper and lower respiratory tract
et al. 1987 and Wegener 1990). At that time, Wegener like his friend and necrotizing vasculitis affecting small to medium-sized vessels
Klinger, was fascinated by the vascular lesions that dominate the final (e.g. capillaries, venules, arterioles, and arteries), with necrotiz-
disease stage, and entitled his report About generalized septic vessel ing glomerulonephritis, granulomatous, and non-granulomatous
diseases (Wegener 1935). However, in its complete form, the disease extravascular inflammation being common, and ocular involve-
is characterized by a fulminant, life-threatening, necrotizing granu- ment and pulmonary capillaritis being frequent.
lomatous inflammation of the upper and lower respiratory tracts, The 1990 ACR criteria classify a vasculitis as GPA if two out of
vasculitis, and glomerulonephritis (Wegener’s triad). A second report the following apply (Leavitt et al. 1990):
by Wegener in 1939 further detailed the clinical course of the three
patients, with special emphasis on the granulomatous lesions About a 1. nasal or oral inflammation with painful or painless oral ulcers or
peculiar rhinogenic granulomatosis with particular involvement of the bloody or purulent nasal discharge;
arteries and kidneys (Wegener 1939). He noted that symptoms began 2. abnormal chest radiograph with the presence of nodules, fixed
with severe upper respiratory infections followed by a fulminant infiltrates, or cavities;
vasculitic course. This sequence of events (spreading of granuloma- 3. abnormal urinary sediment (microhaematuria with more than
tous inflammation and, secondarily, the development of systemic five red blood cells/high power field or red cell casts);
vasculitis) has been confirmed both by clinical means and histo-
logical studies (reviewed in Gross et al. 2000). The term pathergic 4. granulomatous inflammation in biopsy, that shows inflamma-
granulomatosis was introduced in 1955 in order to include a local- tion within the wall of an artery or arteriole or in the perivascular
ized form of GPA in the lung, a new concept at the time (Fienberg or extravascular area.
1955), later confirmed by Carrington and Liebow (Carrington and The presence of any two or more criteria yields a sensitivity of
Liebow 1966). Subsequently, this concept was revived with the grow- 88.2% and a specificity of 92.0%.
ing recognition of GPA clinical subgroups, including ‘limited disease’ In order to overcome the limitations of the ACR and CHCC crite-
and localized GPA (EUVAS) (Hellmich et al. 2007b; Mukhtyar et al. ria (ANCA testing and vasculitis/ granuloma surrogate markers not
2009). The analysis of a larger GPA cohort revealed that 5% of GPA included) the EMEA study group published a classification algorithm for
patients remain with localized disease for years (Holle et al. 2010). primary systemic vasculitides in 2006 (Watts et al. 2007) (Figure 30.1).
402 SECTION 5 vasculitic diseases and syndromes and related disorders

Cases with a clinical diagnosis of ANCA-


associated vasculitis

Fulfils ACR or Lanham criteria for CSS? 1

Yes No

Fulfils ACR criteria for WG 2a


CSS
No
Yes
Histology compatible with CHCC WG 2b
Yes
No
WG
Yes 2c
Histology compatible with CHCC MPA
+WG surrogate markers

Yes No

No Histology, WG surrogate markers 2d


present AND +ve serology for PR3/
MPO

No

Histology compatible with small-vessel 3a


vasculitis. No WG surrogate markers.

Yes No

No histology. No WG surrogate markers.


MPA 3b
Surrogate markers for renal vasculitis AND +ve
Yes
serology for PR3/MPO
*includes renal limited vasculitis
No

cPAN Yes Histology compatible with CHCC cPAN or 4


Angiographic Features typical of cPAN
No

Unclassified

Fig. 30.1 EMEA classification algorithm for ANCA-associated vasculitides. Reproduced from Ann Rheum Dis, Watts, R., et al., 66(2): 222–7, 2007, with permission from
BMJ Publishing Group Ltd.

More recently, the EUVAS categorized AAV disease stages and (Wegener’s)’ replacing the old eponym of ‘Wegener’s granulomato-
activity (Hellmich et al. 2007b; Mukhtyar et al. 2009) for the assess- sis’. As the classifications and definitions (ACR 1990, EMEA 2007,
ment of treatment response in clinical studies (see Chapter  31, CHCC 2012) of vasculitides were intended for study purposes and
and Table 30.1 for EUVAS definitions for GPA disease stages and differentiating vasculitis entities from each other, diagnostic crite-
Table 30.2 for EUVAS definitions for disease activity states used for ria do not exist yet but are currently being developed (DCVAS).
assessment of outcome and therapy response).
Furthermore, for the first time diagnostic criteria for vasculitides
are being developed by the ACR/EULAR diagnosis and classification Epidemiology
of vasculitis study group (DCVAS) for two reasons (Luqmani et al. The estimated disease prevalence of GPA varies between 29 (UK
2011b): first, diagnostic criteria have never been developed. The 1990 1990)  (Watts et  al. 2009)  and 160 (Sweden 2002)  (Mohammad
American College of Rheumatology (ACR) classification criteria and et al. 2007) cases per million population. Also incidence rates vary
the CHCC definitions only serve to differentiate the diverse vasculitis between countries (0.5 per million population and year in Peru
entities after the diagnosis has already been established. Second, new and 11.9 in Sweden). However, results of a population-based vas-
diagnostic criteria have evolved (ANCA testing, imaging techniques) culitis registry over 7 years for the incidence of primary systemic
which need to be integrated in the diagnostic pathway. vasculitides among 2.8  million habitants in northern Germany
and in another population-based study from the UK from 1990
Summary to 2005 reflect a very stable incidence for GPA (Table 30.2) (Watts
The revised nomenclature has been published by the Chapel Hill et  al. 2009; Herlyn et  al. 2008). There is a lack of data from the
Consensus Conference in 2012: ‘granulomatosis with polyangiitis non-developed parts of the world.
CHAPTER 30 granulomatosis with polyangiitis (wegener’s granulomatosis) 403

Table 30.1 EMEA classification: surrogate markers for GPA and renal diagnostics and screening, also reflected by the establishment of
vasculitis centres specialized in vasculitis;
3. the relatively large difference between Peru and Japan compared
I. Surrogate markers for GPA (granulomatous disease)
to the western developed countries probably reflects a differ-
Only one surrogate marker is necessary to support the diagnosis
ent genetic background (rate twice as high among Europeans
of GPA:
as Asians or Maori) (Fujimoto et al. 2011; Lyons et al. 2012;
(a) Lower airways:
X-ray evidence of fixed pulmonary infiltrates, nodules, or cavitations O’Donnell et al. 2007).
present for >1 month. The rising prevalence comparing older to more recent study
Bronchial stenosis populations (e.g. the prevalence of GPA rose in northern Germany
(b) Upper airways: from 58 (1994) to 98 (2006) cases per million) might be explained
Bloody nasal discharge and crusting for >1 month, or nasal ulceration by better survival, besides better diagnostic means, reporting of
Chronic sinusitis, otitis media, or mastoiditis for >3 months data, and a true rise in disease occurrence (Herlyn et al. 2014).
Retro-orbital mass or inflammation (pseudotumour) Studies regarding seasonal variations (Sweden: no (Mohammad
Subglottic stenosis et  al. 2009); New Zealand:  yes (O’Donnell et  al. 2007)), envi-
Saddle nose deformity/ destructive sinonasal disease
ronmental (urban versus country) and geographical, that is
II. Surrogate markers for renal vasculitis (glomerulonephritis) are latitude-dependent factors (lower prevalence towards the equator in
either: New Zealand, Europe, Spain, Peru, and Japan sharing a similar low
Haematuria associated with red cell casts or >10% dysmorphic incidence and latitude, but opposing low incidence in Lithuania, see
erythrocytes Table 30.2), as well as occupational factors remain inconsistent.
or 2+ haematuria AND 2+ proteinuria on urinalysis
GPA may present at any age, the mean age at presentation being
(Reproduced from Ann Rheum Dis, Watts, R., et al., 66(2): 222–7, 2007, with permission 53–68  years; the highest GPA incidence occurs in the elderly
from BMJ Publishing Group Ltd.) (65–79 years) (O’Donnell et al. 2007; Mohammad et al. 2009; Takala
et al. 2008) and it seems to rise with age. However, the incidence of
GPA in a Canadian paediatric population is almost the same as in
There are several explanations why incidence rates vary in differ- adults (2.8–6.4 cases per million and year) (Grisaru et al. 2010).
ent countries:
Summary
1. methodological approach of data acquisition: either population-
The incidence of GPA in central Europe is estimated to be approxi-
based or hospital-based;
mately 8 per million, whereas differences in disease frequencies from
2. study period: before or after introduction of ANCA testing in other regions might reflect genetic, environmental or technical issues
the mid-1980s; a rising awareness of vasculitides with improved of data acquisition. While the incidence seems to remain stable over

Table 30.2 Incidence rates for GPA in different regions and periods: new cases per year and per one million

Country Study region Annual incidence/ Study period Study design Study Mean age Reference
million population
Sweden Sweden 3.3 1975–1985 Population 8 000 000 Knight et al. 2006
11.9 1991–2000
Norway North 8.0 1984–1998 Population 464 000 Haugeberg et al. 1998
Finland 1.9 1981–1985 Population 4 000 000 53 Takala et al. 2008
9.3 1986–2000
Lithuania Vilnius 2.1 1990–1999 Hospital 470 000 Dadoniene et al. 2005
USA Olmsted 8.3 1990–1999 Population Ng et al. 2003
Spain Lugo 2.9 1988–2001 Hospital 208 271 Gonzalez–Gay et al. 2003
Australia South Australia 5.1 1985–2004 Hospital 1 500 000 57 Hissaria et al. 2008
Peru 0.5 1990–2004 Population 930 306 Sanchez et al. 2006
UK UK 8.4 1990–2005 Population 3 600 000 59 Watts et al. 2009
Australia Capital/ New South Wales 8.6 1995–2005 Hospital 440 000 55 Ormerod et al. 2008
Germany Schleswig Holstein 8.5 1998–2005 Population 2 830 000 60 Herlyn et al. 2008
Sweden Skåne 9.8 1997–2006 Hospital 641 000 67.6 Mohammad et al. 2009
Japan Miyazaki 2.1 2005–2009 Population 759 000 Fujimoto et al. 2011
404 SECTION 5 vasculitic diseases and syndromes and related disorders

the last two decades, the prevalence rises, reflecting a better sur- Generalization of GPA is defined by the occurrence of systemic
vival, possibly due to earlier diagnosis and start of therapy, as well disease such as constitutional symptoms (fatigue, night sweats, (sub)
as improved therapeutic options with less therapy-related mortality. febrile temperature, weight loss), non-destructive arthralgia/ arthri-
tis, or vasculitis. The hallmarks of severe generalized disease are
alveolar haemorrhage and glomerulonephritis. While neutrophil
Clinical presentation of GPA or lymphocytic alveolitis are considered a surrogate of pulmonary
The most common presenting symptoms in GPA are related to the capillaritis, lung granuloma are considered to be localized disease.
upper respiratory tract. Typically, GPA starts with a flu-like pres- Whereas granuloma predominate in the respiratory tract, any
entation such as nasal obstruction, serosanguineous discharge, organ system can be affected by either granulomatous mass forma-
epistaxis, sinusitis, external ear chondritis, otitis media, mastoiditis, tion (the pituitary gland with hormonal insufficiency and diabetes
middle or inner ear hearing impairment, or oral inflammation such insipidus, skin, muscles, urinary tract and ovaries, prostate, mam-
as ulcerative stomatitis and hyperplastic gingivitis. This can easily be mary glands) or by or vasculitis manifestations: skin with purpura,
mistaken for a respiratory tract infection due to these non-specific digital gangrene or ulcerations; eye with (epi)scleritis, retinitis, or
symptoms. Therefore the diagnosis of GPA is often delayed until sus- optical nerve ischaemia; serositis; muscle; peripheral nerves with
picion of GPA is raised by either generalization of the disease with mononeuritis multiplex or polyneuropathy and central nerv-
vasculitic features, or by persistent ENT complaints not improving ous system including sterile pachymeningitis; and less frequently
on antibiotic or surgical treatment. More specific GPA symptoms the gastrointestinal tract and the heart with (peri)myocarditis or
are spontaneous septal perforation, saddle nose deformity, destruc- ischaemia due to small-vessel vasculitis (Table  30.3). Even the
tion of conchae or sinus walls, granulomatous mass formation in venous system can be affected as the incidence of thromboembolic
paranasal sinuses, lacrimal glands or orbits leading to proptosis, or events is increased in GPA (Allenbach et al. 2009) and frequently
subglottic stenosis with inspirational stridor. venulitis can be found in histology samples.

Table 30.3 GPA disease manifestations in different study cohorts

Mahr et al. 2012 Holle et al. 2011 Stone et al. 2003 WGET Hoffman et al. 1992 NIH
trial cohort cohort
2012 2011 2003 1992
Total no. of patients 396 445 180 158
Male/female (%) 54/46 50/50 60/40 50/50
Mean age at diagnosis (range) 55 51 (12–85) 47 41
Study period 1995–2003 1966–2002 2000–2002 1968–1992
Mean follow-up (years) 4.75 6 not applicable 8
cANCA/PR3 positive (%) n/a/79 81/80 88/73 88
Involved organ systems at
diagnosis/
throughout course of disease
(%)
ENT 84/n/a 87/98 90/n/a 92/n/a
Arthralgia/ arthritis, myalgia n/a 55/73 81/n/a 67/n/a
Lung 67/n/a 51/60 75/n/a 85/n/a
Kidney 77/n/a 49/60 68/n/a 77/n/a
Dialysis at end of FU n/a 5 n/a
Eye 39/n/a 35/40 31/n/a 52/n/a
Peripheral nervous system 28/n/a 23/40 14/n/a 15/n/a
Skin 30/n/a 19/26 39/n/a 46/n/a
Heart 10/n/a 8/11 4/n/a 8/n/a
Central nervous system n/a 3/10 5/n/a 8/n/a
Gastrointestinal tract 7/n/a 1/3 7/n/a 0/n/a
Infections (%) n/a 35 n/a
Relapses (%) during FU 47 50 n/a

n/a, not available; FU, follow-up.


CHAPTER 30 granulomatosis with polyangiitis (wegener’s granulomatosis) 405

Early recognition of disease progression (rapidly progressive three histological features have been considered to confirm GPA
glomerulonephritis; alveolar haemorrhage syndrome) is important (Devaney et al. 1990). Further typical but non-specific histologi-
because, if untreated, it can lead to life-threatening situations inher- cal features are the presence of multinucleated giant cells and
ent to this disease. neutrophilic microabscesses. Granulomatous inflammation may
Patients often report the disease progression in two stages (ENT be related to bone or cartilage erosions. The granuloma may be
complaints followed by vasculitic manifestations) but GPA can discrete or confluent, forming an irregular geographic pattern of
manifest with a fulminant pulmonary–renal syndrome as a pre- lung parenchymal necrosis (for more detailed discussion and fig-
senting manifestation in 30% of cases. A subgroup of patients (5%) ures see Chapter 29). Eosinophils may be present in the inflam-
may persist in the localized disease stage without progressing ever matory infiltrate but seldom in excessive numbers (Yousem and
into vasculitis. Hence, it is postulated that localized GPA can appear Lombard 1988). In the presence of tissue eosinophilia, the clinical
as a singular disease entity (Holle et al. 2010). presentation and course of disease differentiate it from eosino-
philic granulomatosis with polyangiitis (EGPA). Extravascular
Summary granuloma with or without vasculitis or necrosis can be found
GPA should be suspected in patients who present with chronic or most often in the respiratory tract, but also in any other organ
destructive ENT complaints refractory to antibiotic treatment or system, while the typical histological presentation in the kidney
surgery; unexplained symptoms of systemic inflammatory disease is a focal, segmental, necrotizing glomerulonephritis with cres-
(malaise, fever, night sweats, weight loss, arthralgias); in patients cent formation. Characteristically, no immune deposits are seen
with granulomatous disease or with visible signs of vasculitis such by immunohistochemistry (pauci-immune glomerulonephritis) in
as purpura or episcleritis or surrogate markers of vasculitis such contrast to the immune complex diseases, such as IgA vasculitis
as nephritic sediment. In localized GPA there are no systemic or (Henoch–Schönlein purpura), SLE, cryoglobulinaemic vasculitis,
vasculitic symptoms. or anti-GBM-disease (Goodpasture’s syndrome).
The nasal mucosa is frequently involved in GPA and is eas-
Diagnosis of GPA ily accessible for biopsy but only 20–50% reveal a typical histol-
ogy (Devaney et  al. 1990; Del Buono and Flint 1991; Lombard
As discussed in Section Nomenclature, Definitions, and et  al. 1990). In the case of a typical clinical setting with definite
Classification, classification criteria (ACR, CHCC, EMEA) were GPA or vasculitis surrogate markers and the presence of cANCA
never intended for diagnosis of individual patients. Rao et  al. (PR3) diagnosis can be made without histological confirmation. In
(1998) showed that the ACR classification criteria work poorly less-clear cases, especially when ANCA cannot be found and dif-
in the diagnosis of specific vasculitides as the positive predictive ferential diagnosis may apply, biopsy should be obtained. In the
values ranged only between 29% and 75% for confirmed vascu- presence of renal abnormalities, most often the kidney biopsy will
litis. Therefore GPA should be diagnosed if a typical history and lead to diagnosis. While tracheal or bronchial biopsy can gener-
clinical presentation (see Section Clinical Presentation of GPA), ate diagnostic samples in up to 25% of cases, the surgical biopsy
or characteristic morphological changes in imaging (for example of pulmonary nodules contributes to diagnosis in up to 60%. Also,
granulomatous masses), or clinical correlates of vasculitis (glomer- a neuromuscular biopsy (e.g. sural nerve) in clinically suspected
ulonephritis, alveolar haemorrhage, purpura, uveitis) occur and peripheral nerve involvement has an equally high diagnostic yield
diagnosis should be confirmed by histology showing epitheloid but may lead to a permanent sensory deficit.
granuloma, geographical necrosis, or small-vessel vasculitis and/or
presence of ANCA, most often cANCA with PR3 specificity, less Laboratory tests
common pANCA with MPO specificity. Furthermore, mimickers There is no single laboratory finding that is exclusively diagnos-
should be excluded (see Section Differential Diagnosis). tic for GPA but the presence of cANCA with PR3 subspecificity
is highly indicative (see Section Biomarkers of Disease Activity).
Clinical examination Besides the clinical presentation, elevations in acute phase reac-
In every GPA patient, a full body examination, including all organ tants such as ESR, CRP, and ferritin and elevated complement
systems and a neurological assessment, should be done with special factors reflect inflammatory disease activity. Leukocytosis, which
attention to nasal saddle deformity, oral/nasal ulcerations, propto- can be associated with moderate eosinophilia, thrombocytosis,
sis or exophthalmos, double vision due to impaired eye movements and normochromic normocytic anaemia, are also common. These
(due to orbital mass), pain on percussion of paranasal sinuses and parameters can be normal in the localized disease stage of GPA
mastoid, stridor on forced inspiration (subglottic stenosis), wheez- and, by contrast, can be maximally elevated in the fulminant gen-
ing (bronchial stenosis), haemoptysis due to alveolar haemorrhage, eralized stage, which sometimes is associated with leukaemoid
arthritis, peripheral oedema, skin vasculitis, splinter haemorrhage, reactions. While these tests do not allow a distinction between vas-
as well as sensorimotor deficits, muscular atrophy, and alopecia of culitic disease activity and a concomitant infection or other source
limbs of as signs of neuropathy. of inflammation, procalcitonin can be normal or moderately
elevated, but does not reach levels seen in septicaemia. Therefore
Histopathological features PCT levels >0.5 ng/ml are strongly suggestive of bacterial (super-)
GPA is a necrotizing granulomatous disease as well as a pri- infection (Schinke et al. 2012). Indispensable tests for diagnosis
mary vasculitic disorder (Lie et al. 1990). The typical histological and monitoring of renal involvement are: creatinine, urinary sedi-
lesion in GPA shows the classic triad of (extra)vascular epith- ment (acanthocytes, red cell casts, granular casts), and quantifi-
eloid cell granuloma, geographical necrosis, and vasculitis in cation and differentiation (glomerular versus tubular damage) of
capillaries, arterioles, and, less often, venules. Two out of these proteinuria.
406 SECTION 5 vasculitic diseases and syndromes and related disorders

In contrast to the group of connective tissue diseases (e.g. sys- Although the diagnostic value of ANCA tests is well established,
temic lupus erythematosus), generalized GPA is associated with the usefulness of measuring ANCA titres in assessing disease activ-
no or only mild hypergammaglobulinaemia, no complement con- ity and guiding therapy is somewhat controversial (Langford 2005).
sumption, and no cryoglobulins. Rheumatoid factor may be pre- Generally, because rises in ANCA occur in some patients prior to a
sent in up to 50% of patients. relapse, serial measurements of ANCA titres in patients with AAV
during remission can help predict relapses. However, ANCA levels
ANCA alone should not be used to guide treatment. A significant increase
Antineutrophil cytoplasmic antibodies (ANCA) represent a in ANCA titres, or reappearance of ANCA, should alert the clini-
class of autoantibodies that are specifically directed to multiple cians and lead to stricter disease control.
intracellular antigens in neutrophils and monocytes. ANCA are Although the methods of ANCA detection have been stand-
routinely detected by indirect immunofluorescence (IIF) with ardized internationally and an international consensus statement
ethanol-fixed neutrophils (Hagen et  al. 1993). At least three on testing and reporting on ANCA have been published (Savige
different patterns of fluorescence can be distinguished:  the et al. 1999), uncertainties over testing techniques and strategies for
cytoplasmic granular fluorescence with central interlobular ANCA screening persist. The international guidelines for ANCA
accentuation (cANCA), the perinuclear with nuclear extension testing recommended screening for ANCA by IIF and demand
pattern (pANCA), and the combination of cytoplasmic and per- that in cases of positive IIF testing for ANCA by an ELISA test
inuclear staining (atypical aANCA). In vasculitis, cANCA is a is obligatory as a minimum requirement. However, many hos-
seromarker for GPA and pANCA is a marker for microscopic pital/ referral laboratories do not perform the tests according to
polyangiitis (MPA); the cytoplasmic pattern associated with GPA established guidelines and use only commercially available ELISA
is characteristically induced by proteinase 3 (PR3)-ANCA, the kits to detect ANCA. Over the years, many studies have investi-
perinuclear pattern seen in MPA by myeloperoxidase (MPO)- gated the performances of commercial direct ANCA ELISA and
ANCA. These associations are not absolute and PR3-ANCA is have shown significant differences in sensitivity, specificity, and
sometimes seen in MPA and MPO-ANCA in GPA (reviewed in predictive value among available commercial ELISA kits (Holle
Hoffman and Specks 1998). et al. 2012). Studies have suggested that the sensitivity of capture
The early literature on the clinical utility of ANCA as a diag- ELISA as well anchor ELISA is superior to conventional ELISA and
nostic marker for GPA has been evaluated in a meta-analysis by even to IIF (Hellmich et al. 2007a). The new generation of ANCA
Rao and colleagues (Rao et al. 1995). The sensitivities of cANCA ELISA tests has only recently become available and is currently
testing for GPA ranged from 34 to 92%, and the specificities undergoing evaluation.
range from 88 to 100%. The pooled sensitivity was 66%, and the Although it is currently unclear why among all the described
pooled specificity was 98%. Over the past few years, several new ANCA target antigens only ANCA against these two antigens (PR3
studies have been conducted, most of them being retrospec- and MPO) are closely associated with small-vessel vasculitis, several
tive, and the reported sensitivities and specificities of ANCA studies have compared the clinical and histopathological associations
tests range widely, depending not only on the test characteris- between patients with PR3-ANCA and MPO-ANCA. A direct com-
tics of the assays used but also on the population under study. parison of clinical features of patients categorized by their ANCA
Between 60 and 95% of all ANCA found in GPA are cANCA status revealed that extrarenal manifestations, granuloma formation,
and PR3-ANCA. Hagen et  al. (1998) showed that the value of and relapse were more frequent in patients with PR3-ANCA than in
the immunofluorescence test for ANCA detection can be greatly those with MPO-ANCA (Franssen et al. 1998; Schonermarck et al.
increased by the addition of a well-standardized antigen-specific 2001). Analysis of renal biopsy specimens from 173 patients obtained
ELISA. When the results of IIF were combined with those of the at the time of diagnosis suggested that active and chronic renal
ELISA (cANCA/PR3-ANCA and pANCA/MPO-ANCA), the lesions are more common in MPO-ANCA-positive patients than in
diagnostic specificity increased to 99%. The sensitivity of the PR3-ANCA-positive patients (Hauer et al. 2002). The recent find-
combination of cANCA/PR3-ANCA and pANCA/MPO-ANCA ings from the EUVAS-based GWAS study in GPA and MPA confirm
for GPA, MPA, or primary pauci-immune crescentic glomeru- a genetic component to AAV (GPA and MPA), demonstrate genetic
lonephritis was 73%, 67%, and 82%, respectively (Hagen et  al. distinctions between GPA and MPA that are associated with ANCA
1998). The sensitivity of cANCA/PR3-ANCA for GPA is related specificity, and suggest that the response against autoantigen PR3
to the extent, severity, and activity of disease at the time of (encoded by PRTN3) is a central feature of PR3-ANCA-associated
sampling. The association of MPA with pANCA/MPO-ANCA vasculitis (Lyons et al. 2012). Thus, despite substantial overlap, there
is reported to be in the range of 40–80% (Hagen et  al. 1998). seems to be clinical and pathological differences between patients
Few patients with GPA are pANCA/MPO-ANCA positive and with PR3-ANCA and patients with MPO-ANCA, which may reflect
few patients with MPA cANCA/PR3-ANCA positive. In general, different pathogenic interactions between ANCA, their target anti-
ANCA levels are usually high at presentation and ANCA test- gens, and organ involvement at the molecular level. About 10% of
ing can been used to monitor disease activity and to differentiate patients with GPA do not have ANCA that can be demonstrated by
disease flares from intercurrent illness. PR3-ANCA has also been IFT or in antigen-specific assays. Patients with ANCA-negative GPA
observed in other disorders such as cryoglobulinaemic vasculitis, are likely to have localized disease. ANCA-negative patients with
ulcerative colitis, infectious disorders (i.e. tuberculosis, leprosy, generalized GPA are younger, more likely to be female, and have less
subacute bacterial endocarditis), and drug-induced syndrome lung and kidney involvement, a lower rate of relapse, and a better
(cocaine-induced midline destructive lesions). However, the outcome than ANCA-positive patients.
clinical value of PR3-ANCA testing in these conditions is very Within the last 30  years, ANCA were the subject of intensive
limited. studies and a growing body of evidence indicates a specific role of
CHAPTER 30 granulomatosis with polyangiitis (wegener’s granulomatosis) 407

ANCA in the pathogenesis of AAV. Recently, a direct causal link haemorrhage, ulcerations, crusts, destructive disease with fistulas,
between ANCA and the development of glomerulonephritis and nasal septum perforation, or loss of conchae, and revealing a mono-
vasculitis has been demonstrated in animal models (for review see cavum nasi in the worst case. Diagnostic procedures will include
van Timmeren and Heeringa 2012). ultrasound of paranasal sinuses, otoscopy, and laryngeal and sub-
glottic endoscopy to assess subglottic stenosis (Figure 30.2). ENT
Biomarkers of disease activity
interventions include biopsies for histological diagnosis and local
Although the diagnostic sensitivity and specificity of PR3-ANCA therapies such as dilatation and submucosal infiltration of steroids
and MPO-ANCA for AAV in the appropriate clinical context are or mitomycin for subglottic stenosis or paracentesis to relieve mid-
very high, its use as a biomarker of disease activity is insufficient dle ear effusion.
(Tomasson et al. 2012). In general, with respect to diagnosis and Cranial MRI proved to be a more helpful technique for the visu-
follow-up, sensitive biomarkers reflecting disease activity or smoul- alization of nasal, sinusoidal, orbital (retrobulbar), mastoidal, and
dering disease are still required. even subglottic inflammatory lesions in GPA than conventional
Cytokines, cytokine receptor molecules, and adhesion molecules radiography (Figures  30.3a–c and 30.4). Not only can it help in
in serum or plasma of GPA patients were shown to be associated locating biopsy sites but it also differentiates characteristic patterns
with clinical disease activity. Soluble IL-2 receptor (sIL2-R) levels such as mucosal disease, granuloma formation, and cicatricial
were found to be higher in sera of patients with generalized GPA changes (Muhle et al. 1997). While MRI is vastly superior to CT
than in sera from patients with localized GPA. Persistent elevation in detecting soft tissue changes, including vascular cerebral lesions
of sIL2-R during remission may indicate a higher risk of relapse in and meningeal changes, CT is more sensitive if osseous destruction
these patients (Schmitt et al. 1992). Soluble CD30 has been shown is suspected.
to correlate with disease activity in the generalized vasculitic phase
of GPA (Wang et al. 1997). Elevated procalcitonin levels are a useful Assessing pulmonary involvement
variable in the differential diagnosis of flares and bacterial infection For assessment of pulmonary involvement, the chest radiograph
in GPA patients (Schinke et al. 2012; Eberhard et al. 1997; Moosig (CXR) is required and may show characteristic lesions such as
et al. 1998). nodules, cavities, or infiltrates, which might resemble pneumonia
Markers of endothelial activation and damage have been elevated but often are atypical: that is, bilateral, fluctuating, or interstitial.
in patients with active AAV, including endothelial microparticles However, as subtle changes can easily be missed by conventional
(Erdbruegger et  al. 2008), von Willebrand factor, and thrombo- radiography, a CT scan should be performed if there is clinical
modulin (Boehme et  al. 1996). Antiendothelial cell antibodies suspicion of pulmonary involvement or if pulmonary CO diffu-
(AECA) have been detected in GPA but do not seem to play a major sion is reduced in body plethysmography as a surrogate of inter-
role as a diagnostic tool. stitial disease. Furthermore, the lung function testing can detect
a fixed extrathoracic major airway stricture in tracheobronchial
Imaging and further investigations stenosis.
Assessing ENT involvement The most common abnormalities among the latter are small nod-
The ENT surgeon’s opinion should be sought for diagnosis and ules, septal and non-septal linear opacities, and low-attenuation
grading of visible disease activity (ENTAS, ENT activity score), (ground-glass) opacities. A large study comparing high-resolution
see Section Measurement of Disease Activity and Extent (Garske CT (HRCT) with conventional radiography found the former to
et  al. 2012)  and to follow up on efficacy of therapy. ENT diag- be three times more sensitive in detecting nodules and masses
nostics include rhinoscopy, which can show mucosal oedema, and 1.7 times more sensitive in detecting linear opacities than the

(a) (b)

Fig. 30.2 (a) Palatinal ulcerations and (b) perforation in GPA.


408 SECTION 5 vasculitic diseases and syndromes and related disorders

(a) (b) (c)

Fig. 30.3 Cranial MRI in GPA with orbital granulomatous mass surrounding the right ocular bulbus. (a) T2-weighted image showing bulbar compression. (b) Orbital
granulomatous tissue enhanced in T1-weighted image. (c) Coronary plane; T1-weighted image showing orbital granuloma and increased soft tissues in the maxillary
sinuses, with high signal intensity, indicating active sinusitis.

Fig. 30.4 MRI imaging (STIR T2) of subglottic inflammatory stenosis in GPA. Nearly circumferential thickening of subglottic tracheal wall with oedema and contrast
enhancement reflecting inflammatory activity.

latter (Reuter et  al. 1998). Focal low-attenuation infiltrates, which Assessing renal involvement
evade detection in the conventional radiograph, were found in 10% Elevated creatinine, peripheral oedema, and new-onset or worsen-
of patients. Longitudinal observation suggests that nodules and ing hypertension raise suspicion of renal involvement, but subclini-
low-attenuation opacities are ordinarily a reflection of active disease, cal early renal disease can only be detected by urinalysis looking for
whereas linear opacities can be either active or cicatricial lesions
(Figure 30.5).
In order to differentiate vasculitic alveolitis and alveolar haem-
orrhage from infection, CT will identify the best accessible lung
segment for obtaining bronchoalveolar lavage (Figure 30.6a,b).
Bronchoscopy also discloses tracheobronchial lesions, which are
found in 26–59% of GPA patients (Cordier et al. 1990; Daum et al.
1995; Schnabel et  al. 1997). The abnormalities comprise ulcera-
tions, inflammatory pseudotumours, inflammatory or cicatricial
bronchial or tracheal stenosis, diffuse inflammation, and focal
bleeding.
The bronchoalveolar lavage (BAL) cell profile in GPA depends
on the underlying lesion (Schnabel et  al. 1999). In highly active
vasculitic disease, the BAL cell profile is dominated by neutrophils.
In lung disease of low or moderate disease activity characterized
by nodular or linear opacities, lymphocytes may prevail. For the
most part these are CD4+ T cells with a predominant Th1 cytokine
profile (Schnabel et al. 1999; Csernok et al. 1999). It is of foremost
importance to examine all BAL materials microbiologically to rule
out infection, especially in the immunosuppressed patient as the
risk of atypical infection such as Pneumocystis jiroveci is signifi-
cantly increased. Fig. 30.5 Multiple cavitating pulmonary granuloma in GPA.
CHAPTER 30 granulomatosis with polyangiitis (wegener’s granulomatosis) 409

(a) (b)

Fig. 30.6 High-resolution CT of the lungs in GPA showing a left granuloma (3.2 cm) with consecutive filiform bronchial stenosis and atelectasis of the upper lobe
(a) before and (b) after stenting.

dysmorphic erythrocytes, that is acanthocytes in particular, red cell lesions, especially in the absence of cardiovascular risk, strongly
casts, or granular casts as a sign of proteinuria. The 24-hour urine supports the rare event of cerebral vasculitis. Furthermore, granu-
collection allows quantification and differentiation of glomerular lomatous inflammation may affect the pituitary gland leading to
from tubular proteinuria and is easily obtained for disease moni- hormonal insufficiency.
toring. In rapidly progressing glomerulonephritis, a renal biopsy Only histology is able to confirm vasculitis; a peripheral nerve
is compulsory for establishing diagnosis and differentiating acute biopsy is easily achievable but will lead to permanent nerve dam-
(treatable) from chronic renal changes in relapse or slowly worsen- age, while the cerebral biopsy will require a careful analysis of ben-
ing renal function, respectively. efits and risks. Less frequent is a muscular small-vessel vasculitis
(clinical hints: myalgia, weakness, CK elevation) that can change
Other organ involvement electromyography and presents as muscular oedema or necrosis in
As eye involvement is frequent in GPA, an ophthalmological MRI scan (for review see Holle and Gross 2011).
opinion is often required for diagnosis and monitoring of granu- Cardiac involvement is an infrequent event (<10%, compare
lomatous orbital involvement and ocular vasculitis (episcleritis, Table 30.3) and might present as ischaemic heart disease, myocar-
scleritis, uveitis, retinitis, optical nerve neuritis). The most com- ditis, pericarditis, arrhythmias, or valvular disease. Therefore the
mon finding of granulomatous disease may lead to proptosis, exclusion of arteriosclerotic coronary disease (coronary angiog-
diplopia by ocular muscle or nerve compression (Figure  30.7), raphy) and infection is mandatory (endocarditis, viral serology).
and compression of the optical nerve threatens the eyesight. Only myocardial biopsy will allow definite diagnosis of cardiac
Enophthalmus may result from fibrous orbital socket contracture. small-vessel vasculitis. In cardiac involvement, MRI might show
Also, lacrimal glands and lacrimal ducts can be affected by granu- myocardial oedema and late enhancement (Goodfield et al. 1995).
lomatous disease (for review see Schmidt et al. 2011). Moreover, Furthermore, in new-onset cardiomyopathy, in particular under
therapy-related side-effects such as steroid-induced glaucoma or cyclophosphamide, a drug-induced side-effect should be consid-
cataract need to be monitored. ered, although this is very uncommon in cyclophosphamide dos-
The neurologist’s opinion should be sought for either the fre- ages used for immunosuppression.
quent vasculitic affliction of peripheral nerves (mononeuritis Endoscopy is required in abdominal complaints such as abdom-
multiplex, sensorimotor polyneuropathy) or central nervous inal claudication, GI bleeding, or weight loss in order to diagnose
involvement, such as cerebral or spinal vasculitis, cranial nerve vasculitic involvement histologically or to exclude differential
palsy, or pachymeningitis. Meningitis can also result from ENT diagnosis such as therapy-related side-effects (steroid-induced
granuloma invading the cerebral structures. In cerebral involve- gastritis) or neoplasia. In severe cases, gastrointestinal GPA
ment, a sterile inflammatory cerebrospinal fluid in association may present as ischaemic disease with intestinal necrosis or
with new-onset microangiopathic vascular or steroid-sensitive perforation.
Angiography (including non-invasive techniques) is a valuable
technique for detecting abnormalities in large and intermediate
vessels. Since vasculitic involvement of intermediate vessels, which
needs to be differentiated from panarteritis nodosa, is not uncom-
mon in GPA, angiography can be helpful for the discrimination of
vasculitic from arteriosclerotic disease. Intermediate-vessel vascu-
Fig. 30.7 Bilateral orbital granuloma in GPA with proptosis, eye deviation, scleritis, litis typically presents with angina pectoris, intestinal ischaemia
and marginal keratitis. (abdominal claudication), or hypoperfusion of an extremity such as
410 SECTION 5 vasculitic diseases and syndromes and related disorders

Raynaud’s syndrome, digital gangrene, or acral ulcerations. However, Small-vessel vasculitides frequently occur secondary to under-
angiography cannot document small-vessel disease in GPA. lying conditions, mainly due to infection, haematological malig-
nancies, rheumatic, and other chronic inflammatory diseases
Summary (inflammatory bowel disease, sarcoidosis) or in association with
Until the new millennium, GPA diagnosis was based on the obser- immunodeficiency syndromes. If the disease course appears atypi-
vation of a clinicopathological entity with its typical histological cal, lacking specific GPA features and does not respond to therapy
triad and clinical signs. At present, new and more sophisticated as expected, secondary vasculitides (especially neoplastic, or infec-
imaging techniques and the diagnostic role of ANCA, most spe- tious disease, or non-vasculitic mimics such as immunodeficiency
cifically PR3-ANCA, have immensely increased the sensitivity and syndromes) should be thoroughly investigated.
specificity of the diagnostic process. Many drugs can induce ‘allergic’ leukocytoclastic vasculitis, which
is self-limiting if the causative drug is stopped. Some drugs can
induce ANCA with or without clinical evidence of ANCA-associated
Differential diagnosis vasculitis. Typically, drug-induced ANCA are directed against mul-
In the absence of GPA-specific characteristics (macroscopic gran- tiple antigens other than MPO, such as cathepsin G, lactoferrin, or
uloma mass formation, histological epitheloid granulomatous bactericidal permeability increasing protein (BPI). MPO-ANCA
disease, cANCA directed against proteinase 3, typical ENT involve- frequently occurs with antithyroid drugs but vasculitis can also be
ment such as granulomatous masses in sinuses or orbita) other pri- secondary to the underlying endocrine disorder (Grave’s disease).
mary small-vessel vasculitides and secondary vasculitides should be Cocaine can mimic GPA with nasal septum perforation, secondary
considered. vasculitis, and with a cANCA directed against PR3 but concurrently
As for primary vasculitides, MPA should be suspected if pANCA/ against human leukocyte elastase (HLE) distinguishing it from GPA.
MPO-ANCA is present without evidence of extravascular granuloma Also, endocarditis can present with a cANCA/PR3-ANCA and vas-
formation or marked ENT disease, while EGPA should be diagnosed culitic aspects such as splinter haemorrhages or nephritis.
if eosinophilia and asthma predominate. In hypocomplementaemic Table 30.4 reviews common and important differential diagnoses
small-vessel vasculitis, cryoglobulinaemic, rheumatoid, or secondary conditions of typical GPA features.
vasculitis due to connective tissue disease, infection, or drugs should
be taken into account. If oral or genital lesions associated with ocular Summary
or skin involvement are cardinal symptoms, especially in people with In the presence of histologically proven small-vessel vasculitis,
a genetic background from the Mediterranean or Silk Road areas, GPA needs to be differentiated from other primary ANCA and
Behçet’s disease should be considered. non-ANCA-associated vasculitides as well as from secondary

Table 30.4 Differential diagnosis of GPA: other vasculitides and non-vasculitic diseases according to cardinal symptoms

Cardinal symptoms Rheumatic disorders, (Para)infectious Malignancy Drugs Other


autoimmune disorders
(Destructive) ENT Granulomatous nasal Klebsiella Lethal midline Cocaine (cANCA/ Trauma cholesterol granuloma
inflammation (Fuchs Crohn’s nasal sarcoid rhinoscleromatis, granuloma elastase +ve) immunodeficiencies:
and Tanner 2009) aktinomycosis, (NK-T-cell-lymphoma) intranasal narcotic MHC I -(TAP)-deficiency
rhinosporidiosis, abuse (BPI-ANCA +ve) (Schultz
Aspergillus flavus, et al. 2003),
mycobacteria, lues, hypomorphic Rag mutation
leprosy, leishmania (De Ravin et al. 2010)
Oral ulcerations CTD, Behçet’s Viral infections
Macroscopic nodules Rheumatic nodules Mycobacteria, fungus, Neoplasia, lymphoma Orbital mass/ proptosis:
(any organ) (also pulmonary) Echinoccocus, Klebsiella Tolosa-Hunt-syndrome.,
sarcoid (also pituitary gland) neoplasia
IgG4 disease (Stone et al. 2012)
Rapid progressive AAV, Goodpasture’s, Hantavirus, HUS Paraneoplastic Interstitial nephritis,
glomerulonephritis, CTD, HSP, CV poststreptococcal drug-induced
acute renal failure renal crisis in scleroderma glomerulonephritis vasculitis
urosepsis
Diffuse alveolar AAV, Goodpasture’s, CTD, Idiopathic pulmonary siderosis
haemorrhage HSP, CV, PAN, Behçet’s
Tissue hypoperfusion Small-vessel vasculitis Vasculitis/ embolism Paraneoplastic Thromboangiitis Diabetic small-vessel disease
(acral, skin, or PAN, scleroderma, in endocarditis (can be vasculitis or Raynaud obliterans (smokers) mesenterial VOD
organ-related) secondary Raynaud’s, cANCA/PR3 +ve) ergotism (migraine)
necrosis thrombotic:
HUS, TTP, APS,
cholesterol embolism
(continued)
CHAPTER 30 granulomatosis with polyangiitis (wegener’s granulomatosis) 411

Table 30.4 (continued)
Cardinal symptoms Rheumatic disorders, (Para)infectious Malignancy Drugs Other
autoimmune disorders
Nervous system SVV, HES, CTD, Lyme disease, HIV, Paraneoplastic, Toxic (alcohol, drugs, Diabetes, deficiency in vitamin
polyneuropathy, GCA-associated leprosy, paraproteinaemia e.g. leflunomide) B1, B6, or B12, folate (under
mononeuritis CSS, MPA, PAN, CV, progressive MTX),
multiplex, CTD, RV, sarcoidosis leukoencephalopathy hereditary
cerebral lesions EGPA, MPA, RV, SVV, (JC-virus) amyloidosis,
CTD, PACNS, diabetes
multiple sclerosis arteriosclerotic
(Tissue) EGPA Parasitosis (helminths) Eosinophilic leukaemia Drug reaction Organ-related
Eosinophilia HES (no ANCA, (FIP1L1-PDGFR-fusion, eosinophilia: eosinophilic
vasculitis, or granuloma) T-cell clonality, or oesophagitis, pneumonia,
other) or fasciitis
Microscopic Crohn’s, Mycobacteria, bartonellosis,
epitheloid granuloma sarcoidosis brucellosis, toxoplasmosis,
tularaemia, EBV
Isolated organ PACNS, skin, GI,
vasculitis mesenterial VOD, testicles,
(granulomatous or uterus, ovary, breast,
small-vessel type) peripheral nerves, muscles
(Holl-Ulrich et al. 2009)
Secondary RV, CTD, relapsing Endocarditis, Often haematological, Cocaine, Immunodeficiencies
vasculitides polychondritis toxoplasmosis, solid malignancy leukocytoclastic skin Wiskott-Aldrich-syndrome.
inflammatory bowel CMV, VZV, fungus vasculitis, Hyper-IgE-syndrome. (aortitis,
disease aortitis luica pANCA/MPO ± coronaritis, aneurysms)
clinical vasculitis after
PAN/CV (hepatitis B /
PTU, thiamazole,
C, HIV)
hydralazine, SSZ,
TNF-blocker,
D-penicillamine;
gemcitabine,
transretinoic acid

AAV, ANCA associated vasculitis; APS, antiphospholipid syndrome; BPI, bactericidal permeability increasing protein; CTD, connective tissue disease; CV, cryoglobulinaemic vasculitis;
EGPA, eosinophilic granulomatosis and polyangiitis; GI, gastrointestinal; HES, hypereosinophilia syndrome; HSP, Henoch–Schönlein purpura; HUS, haemolytic uraemic syndrome; MPA,
microscopic polyangiitis; MTX, methotrexate; PACNS, primary angiitis of the central nervous system; PAN, panarteritis nodosa; PTU, propylthiouracil; Rag, recombination activating
gene; RV, rheumatoid vasculitis; SSZ, sulfasalazine; SVV, small-vessel vasculitis; TAP, transporter associated with antigen processing; TTP, thrombotic thrombocytopenic purpura; VOD,
veno-occlusive disease.

vasculitides and conditions mimicking vasculitis. If clinical suspi- new or worsening symptoms from nine organ systems within the
cion of GPA is raised, dependent on the cardinal symptoms such previous 4 weeks. The BVAS was modified in 1997 (BVAS v.2),
as ENT disease, mass formation, histological granuloma, impaired in 2001 (BVAS/WG), and most recently in 2009 (BVAS v.3) The
perfusion/ ischaemia, and haemorrhage, a large variety of different BVAS/WG and BVAS 2003 incorporate items specific for GPA,
diagnoses have to be considered including infections, malignancy, such as orbital mass or subglottic stenosis (Stone et al. 2001) (see
non-vasculitic granulomatous diseases, and vasculitis mimics that Chapter 22).
impair small-vessel perfusion. From the extended ELK classification (De Remee 1976) a disease
extent index (DEI) was derived (de Groot et al. 2001), a more fea-
Measurement of disease activity sible score in daily routine as it sums up disease activity within ten
organ systems and constitutional symptoms without grading activ-
and extent ity (Table 30.5). The DEI has proven to be easy and quick to apply,
Recognizing the diversity of disease courses in GPA, treatment can be used prospectively or retrospectively, and shows a very low
should be individualized according to disease severity, determined inter- and intraobserver variability.
by extent and activity, and should take the response to current or The ENT activity score (ENTAS) has been published for stand-
previous treatment into account. ardizing the ENT assessment, including activity and damage to the
As a standardized patient assessment serves as the basis for inner and external nose, paranasal sinus, ear, larynx, and subglottis
therapeutic decisions, a range of tools to assess GPA disease activ- (Garske et al. 2012).
ity has been validated. Most frequently used is the Birmingham Most importantly, disease activity needs to be differentiated from
vasculitis activity score (BVAS), as recommended by the EULAR chronic damage due to vasculitis (scarring) or its therapy to prevent
for conducting clinical trials (Luqmani et al. 1994). It measures unnecessary immunosuppression. The vasculitis damage index
412 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 30.5 The extended ELK classification leading to the Disease (VDI) measures 64 symptoms in 11 organ systems that persist for
Extent Index (DEI) longer than 3 months and thus are considered not to respond to
immunosuppression any more. However, this needs to be definitely
Involved organ Score Standard diagnostic procedure differentiated from refractory active disease insufficiently treated
and requiring treatment escalation. In more than 80% of patients,
E ENT/upper 2 Clinical assessment by ENT specialist,
respiratory tract cranial MRI, sinoscopy chronic damage is an inevitable consequence of persisting GPA.
VDI can help to predict outcome as patients with more than five
L Lung 2 Chest radiograph, HRCT, bronchoscopy,
items of damage have an increased risk of mortality (Bhamra and
bronchoalveolar
Luqmani 2012) (Table 30.6).
lavage Recently, another outcome score, the five factor score (FFS), has
EY Eye 2 Ophthalmological referral, cranial MRI proven its predictive value also for GPA as the 5-year mortality was
K Kidney 2 Urinalysis, serum creatinine, abdominal increased in GPA patients with chronic renal impairment (HR 3.6)
ultrasound or cardiac insufficiency (HR 2.0) (Guillevin et al. 2011).
To assess disease activity and irreversible damage, and hence
H Heart 2 ECG, chest radiograph, echocardiogram,
therapy need, the patients should be evaluated on a regular basis
myocardial scintigram, coronary angiogram
by an interdisciplinary team of physicians, ENT surgeons, oph-
GI Gastrointestinal 2 Abdominal ultrasound, endoscopy, thalmologists, neurologist, dermatologist, urologist, etc., as well
tract (mesenteric angiogram, laparotomy) as technical investigations (such as imaging, lung function, urine
S Skin 2 Clinical assessment, biopsy collection). Even if no disease activity is evident to the patient or
P Peripheral 2 Neurological referral, ENG, EMG the physician, a closer look can reveal subclinical disease activity or
nervous system (nerve biopsy) damage such as granuloma formation, proteinuria, neuropathy, etc.
C Central nervous 2 Neurological referral, lumbar puncture,
Furthermore, it is important to assess therapy-related side-effects,
system cranial MRI such as cataract, osteoporosis, urocystitis or malignancy, especially
bladder and skin cancer. Evidently, the frequency of disease assess-
A Rheumatic 2 Radiographs, ultrasound, diagnostic
ment depends on the severity of disease and toxicity of current
complaints arthrocentesis, bone scan,
treatment. Thus, recommended routine follow-ups of 3–6 months
serum-CK, EMG, MRI, muscle biopsy intervals should be adjusted to individual needs.
B Constitutional 1
symptoms Summary
Maximum DEI score 21 In order to monitor disease burden in GPA, there are a variety of
tools available that assess in a standardized manner both active
ENG, electroneurography; EMG, electromyography inflammation requiring immunosuppression (BVAS, DEI, ENTAS)
(de Groot, K., et al., Development and validation of a disease extent index for Wegener's
granulomatosis. Clin Nephrol, 2001, 55(1): p. 31–8). and chronic damage from GPA and its treatment, as well as associ-
ated co-morbidity (VDI). Evaluation of GPA activity requires an
Table 30.6 Items documented in the vasculitis damage index

Ear, Nose, Throat Pulmonary Cardiovascular Gastrointestinal


Hearing loss, nasal blockage/chronic Pulmonary hypertension Pulmonary Angina/angioplasty Gut infarction /resection Mesenteric
discharge/crusting fibrosis Pulmonary infarction Myocardial infarction Cardiomyopathy insufficiency / pancreatitis
nasal bridge collapse/ septal Pleural fibrosis Valvular disease Chronic peritonitis Oesophageal
perforation, chronic sinusitis/ Chronic asthma stricture/ surgery
Pericarditis ≥3 months or
radiological damage, subglottic Chronic breathlessness Impaired lung Neuropsychiatric
pericardectomy
stenosis ± surgery function Cognitive impairment
Diastolic BP ≥95 mmHg or
Skin/mucous membranes Renal Major psychosis
antihypertensives
Alopecia, cutaneous or mouth ulcers Estimated/measured GFR ≤50 Seizures
Peripheral vascular disease
Ocular Proteinuria ≥0.5 g/24 h Cerebrovascular accident
Absent pulses in one limb
Cataract, retinal change, optic atrophy, End-stage renal disease Cranial nerve lesion Peripheral
Major vessel stenosis Claudication
visual impairment, diplopia, blindness, Musculoskeletal neuropathy Transverse myelitis
>3 months
orbital wall destruction
Muscle atrophy / weakness Minor tissue loss Other
Deforming/erosive arthritis Major tissue loss Gonadal failure
Osteoporosis/vertebral collapse Complicated venous thrombosis Marrow failure
Avascular necrosis Diabetes
Osteomyelitis Chemical cystitis
Malignancy
Other

(Exley, A.R., et al., Development and initial validation of the Vasculitis Damage Index for the standardized clinical assessment of damage in the systemic vasculitides. Arthritis Rheum, 1997,
40(2): p. 371–80.)
CHAPTER 30 granulomatosis with polyangiitis (wegener’s granulomatosis) 413

interdisciplinary approach on a regular basis, usually 3–6 monthly infection (pneumonia, CMV, sepsis, Pneumocystis) (Flossmann
intervals in order to offer the best and individualized therapy. et  al. 2011; Little et  al. 2010)  and due to severe vasculitis activ-
ity requiring intensive care (mortality rate 29%) (Burkhardt et al.
2007) especially in young men (standardized mortality ratio 8.9)
Disease course and prognosis (Holle and Gross 2011). Other poor prognostic factors are older age
GPA progresses without therapy, in most cases, but the rate of >50 years, renal impairment (Mohammad et al. 2009; Takala et al.
progression varies greatly among individual patients. Up to the 2010; Walsh et al. 2012), and cardiac and gastrointestinal involve-
mid-1980s, before the institution of effective therapy, the prognosis ment (the Five Factor Score from the French vasculitis study group)
of GPA was extremely grave. In untreated GPA, 82% of patients (Mahr et al. 2012; Guillevin et al. 2011).
died within 1 year; more than 90% within 2 years (Fauci et al. 1983). Main long-term morbidities beside high relapse rates (35–64%),
Walton (1958) reported in a retrospective study of 56 GPA patients which are decreasing in more recent cohorts (Holle et al. 2011), and
that ‘though a few cases have lived for up to 4 years, in the majority refractory disease (10–15%) (Reinhold-Keller and Moosig 2011;
death rapidly ensues from renal or respiratory failure, the average Reinhold-Keller and Zink 2011), are arteriosclerotic cardiovascular
course being about 5 months’. He further reported that the natural disease (Hissaria et al. 2008) and malignancies. Cardiovascular dis-
course of GPA involves two phases and that ‘the initial lesion in ease might occur due to endothelial dysfunction in vasculitis, due
GPA is not vasculitic, but the ulceration in the respiratory tract’. to renal impairment as an independent cardiovascular risk factor
This observation of two distinct disease GPA stages, that is local- but it also might be increased, as it is in other chronic inflammatory
ized and systemic vasculitic disease, could also be seen in a NIH diseases such as rheumatoid arthritis or SLE.
cohort (n=158; indolent course in patients without renal manifesta- Malignancies are associated with the use of cytotoxic agents,
tion for up to 16 years before the definitive diagnosis of GPA was in particular cyclophosphamide, in a dose-dependent asso-
established; mean period from the onset of symptoms to a diag- ciation. Previous observations revealed an overall malignancy
nosis 15 months) (Hoffman et al. 1992) and in a British study of incidence of 1.6–2.4, especially in bladder cancer, leukae-
265 GPA patients (Anderson et al.1985). Only recently could it be mia, lymphoma, and non-melanotic skin cancer (Table  30.8)
shown in a large GPA cohort of 1024 patients that 5% of patients (Hoffman et al. 1992; Knight et al. 2002; Faurschou et al. 2008;
remained in the localized disease stage more than 1 year, thus rep- Westman et al. 1998).
resenting a discrete entity that usually does not generalize. Merely In more recent cohorts the risk of malignancies seems to adjust
half of those patients were ANCA-positive. Although long-term to the general populations risk with less-intensive use of cytotoxic
survival in localized GPA is excellent, there are particular charac- drugs (Holle et al. 2011; Heijl et al. 2011).
teristics in localized disease, such as tumour-like mass formation With improving survival and long-term outcome patients have
with space obturation and invasiveness, that can lead to organ- or to cope with chronic morbidity and therapy-related side-effects. In
life-threatening complications, such as visual loss in orbital granu- a self-assessment of disease burden (patient-reported outcome) the
loma, meningitis due to intracranial perforation of sinus granu- majority of patients suffer from general complaints such as fatigue,
loma, or respiratory insufficiency due to subglottic stenosis. The musculoskeletal pain, and weight gain besides ENT symptoms that
number of relapses (46%) and refractory disease (16%) was similar reduce their quality of life most (Herlyn et al. 2010).
to those in generalized disease (Holle et al. 2010).
Whereas older studies found an increased mortality rate com- Summary
pared to the reference population, newer data show an improved Despite a high burden of disease in GPA the outcome has signifi-
survival with a decreasing excess mortality rate (Table 30.7). This cantly improved over the last decades, mainly due to improved
is due to improved diagnostic and therapeutic options by prevent- diagnostic means, greater disease awareness, and a more focused
ing relapses more efficiently (relapse rate declined from 64 to 50% use of cytotoxic agents. However, mortality still is increased, in
between 1966 and 1998) despite lower cumulative cyclophospha- particular in young men, intensive care patients, and in those
mide doses (67 vs. 36 g) with fewer therapy-related deaths (Holle with renal and visceral organ involvement. Patients remain at risk
2011). Five-year survival rates between 74 and 81% have been of dying, especially within the first year after diagnosis. Probably,
reported (Mohammad et  al. 2009; Takala et  al. 2010). However, therapy-related toxicity contributes significantly to early mortality.
there is still a significant early excess mortality within the first Long-term morbidity is determined by relapsing disease, leading
year of diagnosis (HR 9.0) (Luqmani et al. 2011a) mainly due to to chronic damage and by therapy side-effects as well as resulting
co-morbidities.
Table 30.7 Standardized mortality ratios (SMR) in GPA

Country Study period Study design n SMR Reference Table 30.8 Standardized incidence of cancer (SIR) in GPA

Canada, 1978–87 Multicentre 77 4.7 Matteson et al. 1996 Country Study period Study design n SIR Reference
Mexico, USA
USA 1967–1990 Monocentre 158 2.4 Hoffman et al. 1992
Sweden 1969–1994 Multicentre 1065 4.0 Knight et al. 2002
Sweden 1969–94 Population 1065 2.0 Knight et al. 2002
Norway 1988–1998 Multicentre 108 3.8 Aasarod et al. 2000
Denmark 1973–99 Population 293 2.1 Faurschou et al. 2008
Germany 1966–1993 Monocentre 155 2.1 Holle et al. 2011
1994–1998 123 1.4 Europe 1995–2007 Multicentre 281 1.9 Heijl et al. 2011
1999–2002 167 1.0 Germany 1966–2002 Monocentre 445 0.8 Holle et al. 2011
414 SECTION 5 vasculitic diseases and syndromes and related disorders

References Faurschou, M., Sorensen, I.J., Mellemkjaer, L., et al. (2008). Malignancies in
Wegener’s granulomatosis: incidence and relation to cyclophosphamide
Aasarod, K., Iversen, B.M., Hammerstrøm, J., et al. (2000). Wegener’s gran- therapy in a cohort of 293 patients. Journal of Rheumatology, 35, 100–5.
ulomatosis:  clinical course in 108 patients with renal involvement.
Fienberg, R. (1955). Pathergic granulomatosis. American Journal of Medicine,
Nephrology Dialysis Transplantation, 15, 611–8.
19, 829–31.
Allenbach, Y., Seror, R., Pagnoux, C., et al. (2009). High frequency of venous
Flossmann, O., Berden, A., de Groot, K., et al. (2011). Long-term patient sur-
thromboembolic events in Churg–Strauss syndrome, Wegener’s granu-
vival in ANCA-associated vasculitis. Annals of the Rheumatic Diseases,
lomatosis and microscopic polyangiitis but not polyarteritis nodosa: a
70, 488–94.
systematic retrospective study on 1130 patients. Annals of the Rheumatic
Franssen, C., Gans, R., Kallenberg, C., et al. (1998). Disease spectrum of patients
Diseases, 68, 564–7.
with antineutrophil cytoplasmic autoantibodies of defined specificity: distinct
Anderson, G., Crane, M., Douglas, A.C., et al. (1992). Wegener’s granuloma.
differences between patients with anti-proteinase 3 and anti-myeloperoxidase
A series of 265 British cases seen between 1975 and 1985. A report by
autoantibodies. Journal of Internal Medicine, 244, 209–16.
a sub-committee of the British Thoracic Society Research Committee.
Fuchs, H.A. and Tanner, S.B. (2009). Granulomatous disorders of the nose
Quarterly Journal of Medicine, 83, 427–38.
and paranasal sinuses. Current Opinion in Otolaryngology and Head and
Bhamra, K. and Luqmani, R. (2012). Damage assessment in ANCA-associated
Neck Surgery, 17, 23–7.
vasculitis. Current Rheumatology Reports, 14, 494–500.
Fujimoto, S., Watts, R.A., Kobayashi, S., et  al. (2011). Comparison of the
Boehme, M.W., Schmitt, W.H., Youinou, P., et al. (1996). Clinical relevance of
epidemiology of anti-neutrophil cytoplasmic antibody-associated vas-
elevated serum thrombomodulin and soluble E-selectin in patients with
culitis between Japan and the U.K. Rheumatology (Oxford), 50, 1916–20.
Wegener’s granulomatosis and other systemic vasculitides. American
Garske, U., Haack, A., Beltrán, O., et al. (2012). Intra- and inter-rater reliabil-
Journal of Medicine, 101, 387–94.
ity of endonasal activity estimation in granulomatosis with polyangii-
Burkhardt, O., Köhnlein, T., Wrenger, E., et al. (2007). Predicting outcome
tis (Wegener s). Clinical and Experimental Rheumatology, 30 (1 Suppl.
and survival in patients with Wegener’s granulomatosis treated on the
70), S22–8.
intensive care unit. Scandinavian Journal of Rheumatology, 36, 119–24.
Gonzalez-Gay, M.A., Garcia-Porrua, C., Guerrero, J., et al. (2003). The epide-
Carrington, C.B. and Liebow, A. (1966). Limited forms of angiitis and granu- miology of the primary systemic vasculitides in northwest Spain: impli-
lomatosis of Wegener’s type. American Journal of Medicine, 41, 497–527. cations of the Chapel Hill Consensus Conference definitions. Arthritis
Cordier, J.F., Valeyre, D., Guillevin, L., et al. (1990). Pulmonary Wegener’s gran- and Rheumatism, 49, 388–93.
ulomatosis. A clinical and imaging study of 77 cases. Chest, 97, 906–12. Goodfield, N.E., Bhandari, S., Plant, W.D., et al. (1995). Cardiac involvement
Csernok, E., Trabandt, A., Müller, A., et  al. (1999). Cytokine profiles in in Wegener’s granulomatosis. British Heart Journal, 73, 110–5.
Wegener’s granulomatosis: predominance of type 1 (Th1) in the granu- Grisaru, S., Yuen, G.W., Miettunen, P.M., et al. (2010). Incidence of Wegener’s
lomatous inflammation. Arthritis and Rheumatism, 42, 742–50. granulomatosis in children. Journal of Rheumatology, 37, 440–2.
Dadoniene, J., Kirdaite, G., Mackiewicz, Z., et al. (2005). Incidence of pri- Gross, W.L., Schnabel, A., and Trabandt, A. (2000). New perspectives in pul-
mary systemic vasculitides in Vilnius: a university hospital population monary angiitis. From pulmonary angiitis and granulomatosis to ANCA
based study. Annals of the Rheumatic Diseases, 64, 335–6. associated vasculitis. Sarcoidosis Vasculitis, and Diffuse Lung Diseases,
Daum, T.E., Specks, U., Colby, T.V., et al. (1995). Tracheobronchial involve- 17, 33–52.
ment in Wegener’s granulomatosis. American Journal of Respiratory and Guillevin, L., Pagnoux, C., Seror, R., et  al. (2011). The Five-Factor Score
Critical Care Medicine, 151, 522–6. revisited: assessment of prognoses of systemic necrotizing vasculitides
de Groot, K., Gross, W.L., Herlyn, K., et al. (2001). Development and vali- based on the French Vasculitis Study Group (FVSG) cohort. Medicine
dation of a disease extent index for Wegener’s granulomatosis. Clinical (Baltimore), 90, 19–27.
Nephrology, 55, 31–8. Hagen, E.C., Daha, M.R., Hermans, J., et al. (1998). Diagnostic value of stand-
Del Buono, E.A. and Flint, A. (1991). Diagnostic usefulness of nasal biopsy in ardized assays for anti-neutrophil cytoplasmic antibodies in idiopathic
Wegener’s granulomatosis. Human Pathology, 22, 107–10. systemic vasculitis. EC/BCR Project for ANCA Assay Standardization.
De Ravin, S.S., Cowen, E.W., Zarember, K.A., et al. (2010). Hypomorphic Rag Kidney International, 53, 743–53.
mutations can cause destructive midline granulomatous disease. Blood, Hagen, E.C., Andrassy, K., Csernok, E., et al. (1993). The value of indirect
116, 1263–71. immunofluorescence and solid phase techniques for ANCA detection.
De Remee, R., McDonald, T.J., Harrison, E.G.J.R., et al. (1976). Wegener’s A report on the first phase of an international cooperative study on the
granulomatosis. Anatomic correlates, a proposed classification. Mayo standardization of ANCA assays. EEC/BCR Group for ANCA Assay
Clinic Proceedings, 51, 777–81. Standardization. Journal of Immunological Methods, 159, 1–16.
Devaney, K.O., Travis, W.D., Hoffman, G., et  al. (1990). Interpretation of Hauer, H.A., Bajema, I.M., van Houwelingen, H.C., et al. (2002). Renal histol-
head and neck biopsies in Wegener’s granulomatosis. A pathologic study ogy in ANCA-associated vasculitis: differences between diagnostic and
of 126 biopsies in 70 patients. American Journal of Surgical Pathology, serologic subgroups. Kidney International, 61, 80–9.
14, 555–64. Haugeberg, G., Bie, R., Bendvold, A., et  al. (1998). Primary vasculitis in
Eberhard, O.K., Haubitz, M., Brunkhorst, F.M., et al. (1997). Usefulness of a Norwegian community hospital:  a retrospective study. Clinical
procalcitonin for differentiation between activity of systemic autoim- Rheumatology, 17, 364–8.
mune disease (systemic lupus erythematosus/systemic antineutrophil Heijl, C., Harper, L., Flossmann, O., et al. (2011). Incidence of malignancy in
cytoplasmic antibody-associated vasculitis) and invasive bacterial infec- patients treated for antineutrophil cytoplasm antibody-associated vas-
tion. Arthritis and Rheumatism, 40, 1250–6. culitis: follow-up data from European Vasculitis Study Group clinical
Erdbruegger, U., Grossheim, M., Hertel, B., et  al. (2008). Diagnostic role of trials. Annals of the Rheumatic Diseases, 70, 1415–21.
endothelial microparticles in vasculitis. Rheumatology (Oxford), 47, 1820–5. Hellmich, B., Csernok, E., Fredenhagen, G., et  al. (2007a). A novel high
Exley, A.R., Bacon, P.A., Luqmani, R.A., et  al. (1997). Development and sensitivity ELISA for detection of antineutrophil cytoplasm antibod-
initial validation of the Vasculitis Damage Index for the standardized ies against proteinase-3. Clinical and Experimental Rheumatology, 25
clinical assessment of damage in the systemic vasculitides. Arthritis and (Suppl. 44), S1–5.
Rheumatism, 40, 371–80. Hellmich, B., Flossmann, O., Gross, W.L., et al. (2007b). EULAR recommen-
Fauci, A.S., Haynes, B.F., Katz, P., et  al. (1983). Wegener’s granulomato- dations for conducting clinical studies and/or clinical trials in systemic
sis: prospective clinical and therapeutic experience with 85 patients for vasculitis: focus on anti-neutrophil cytoplasm antibody-associated vas-
21 years. Annals of Internal Medicine, 98, 76–85. culitis. Annals of the Rheumatic Diseases, 66, 605–17.
CHAPTER 30 granulomatosis with polyangiitis (wegener’s granulomatosis) 415

Herlyn, K., Buckert F., Gross W.L., et al. Doubled prevalence rates of ANCA- Luqmani, R.A., Suppiah, R., Grayson, P.C., et al. (2011b). Nomenclature and
associated vasculitides and giant cell arteritis between 1994 and 2006 in classification of vasculitis—update on the ACR/EULAR diagnosis and
North Germany. Rheumatology, in press 2014 classification of vasculitis study (DCVAS). Clinical and Experimental
Herlyn, K., et al. Doubled prevalence rates of ANCA-associated vasculitides Immunology, 164 (Suppl. 1), 11–3.
and giant cell arteritis between 1994 and 2006 in North Germany. Lyons, P.A., Rayner, T.F., Trivedi, S., et al. (2012). Genetically distinct subsets
Arthritis Care and Research (Hoboken) in press 2014. within ANCA-associated vasculitis. New England Journal of Medicine,
Herlyn, K., Hellmich, B., Seo, P., et  al. (2010). Patient-reported outcome 367, 214–23.
assessment in vasculitis may provide important data and a unique per- Mahr, A., Katsahian, S., Varet, H., et al. (2013). Revisiting the classification of
spective. Arthritis Care and Research (Hoboken), 62, 1639–45. clinical phenotypes of anti-neutrophil cytoplasmic antibody-associated
Herlyn, K., Hellmich, B., Gross, W.L., et al. (2008). Stable incidence of sys- vasculitis:  a cluster analysis. Annals of the Rheumatic Diseases, 72,
temic vasculitides in schleswig-holstein, Germany. Deutsches Ärzteblatt 1003–10.
International, 105, 355–61. Matteson, E.L., Gold, K. N., Bloch, D. A., et al. (1996). Long-term survival
Hissaria, P., Cai, F.Z., Ahern, M., et al. (2008). Wegener’s granulomatosis: epi- of patients with Wegener’s granulomatosis from the American College
demiological and clinical features in a South Australian study. Internal of Rheumatology Wegener’s Granulomatosis Classification Criteria
Medicine Journal, 38, 776–80. Cohort. American Journal of Medicine, 101, 129–34.
Hoffman, G.S. and Specks, U. (1998). Antineutrophil cytoplasmic antibodies. Mohammad, A.J., Jacobsson, L.T., Westman, K.W., et al. (2009). Incidence
Arthritis and Rheumatism, 41, 1521–37. and survival rates in Wegener’s granulomatosis, microscopic polyangii-
Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., et al. (1992). Wegener granulomato- tis, Churg-Strauss syndrome and polyarteritis nodosa. Rheumatology
sis: an analysis of 158 patients. Annals of Internal Medicine, 116, 488–98. (Oxford), 48, 1560–5.
Holle, J.U. and Gross, W.L. (2011). Neurological involvement in Wegener’s Mohammad, A.J., Jacobsson, L.T., Mahr, A.D., et al. (2007). Prevalence of
granulomatosis. Current Opinion in Rheumatology, 23, 7–11. Wegener’s granulomatosis, microscopic polyangiitis, polyarteritis
Holle, J.U., Herrmann, K., Gross, W.L., et al. (2012). Comparative analysis of nodosa and Churg-Strauss syndrome within a defined population in
different commercial ELISA systems for the detection of anti-neutrophil southern Sweden. Rheumatology (Oxford), 46, 1329–37.
cytoplasm antibodies in ANCA-associated vasculitides. Clinical and Moosig, F., Csernok, E., Reinhold-Keller, E., et al. (1998). Elevated procalci-
Experimental Rheumatology, 30(Suppl. 70), S66–9. tonin levels in active Wegener’s granulomatosis. Journal of Rheumatology,
Holle, J.U., Gross, W.L., Latza, U., et al. (2011). Improved outcome in 445 25, 1531–3.
patients with Wegener’s granulomatosis in a German vasculitis center Muhle, C., Reinhold-Keller, E., Richter, C., et al. (1997). MRI of the nasal
over four decades. Arthritis and Rheumatism, 63, 257–66. cavity, the paranasal sinuses and orbits in Wegener’s granulomatosis.
Holle, J.U., Gross, W.L., Holl-Ulrich, K., et  al. (2010). Prospective long-term European Radiology, 7, 566–70.
follow-up of patients with localised Wegener’s granulomatosis: does it occur Mukhtyar, C., Guillevin, L., Cid, M.C., et al. (2009). EULAR recommenda-
as persistent disease stage? Annals of the Rheumatic Diseases, 69, 1934–9. tions for the management of primary small and medium vessel vasculi-
Holl-Ulrich, K., Noack, F., and Feller, A.C. (2009). [Vasculitis: histopathol- tis. Annals of the Rheumatic Diseases, 68, 310–7.
ogy and differential diagnosis]. Zeitschrift für Rheumatologie, 68, 320–8. Ng, B., Specks, U., Offord, K.P., et al. (2003). Epidemiology of Wegener
Jennette, J., Falk, R.J., Bacon, P.A., et al. (2013). 2012 Revised International granulomatosis since the introduction of ANCA testing in Olmsted
Chapel Hill Consensus Conference Nomenclature of Vasculitides. County, MN, 1990-1999. Journal of Clinical Rheumatology,
Arthritis and Rheumatism, 65, 1–11. 9, 387–8.
Klinger, H. (1932). Grenzformen der Periarteritis nodosa. Frankfurter O’Donnell, J.L., Stevanovic, V.R., Frampton, C., et al. (2007). Wegener’s gran-
Zeitschrift für Pathologie, 42, 455–80. ulomatosis in New Zealand: evidence for a latitude-dependent incidence
Knight, A., Ekbom, A., Brandt, L., et  al. (2006). Increasing incidence gradient. Internal Medicine Journal, 37, 242–6.
of Wegener’s granulomatosis in Sweden, 1975-2001. Journal of Ormerod, A.S. and Cook, M.C. (2008). Epidemiology of primary systemic
Rheumatology, 33, 2060–3. vasculitis in the Australian Capital Territory and south-eastern New
Knight, A., Askling, J., and Ekbom, A. (2002). Cancer incidence in a South Wales. Internal Medicine Journal, 38, 816–23.
population-based cohort of patients with Wegener’s granulomatosis. Rao, J.K., Weinberger, M., Oddone, E.Z., et al. (1995). The role of antineu-
International Journal of Cancer, 100, 82–5. trophil cytoplasmic antibody (c-ANCA) testing in the diagnosis of
Langford, C.A. (2005). How can relapses be detected and prevented in pri- Wegener granulomatosis. A literature review and meta-analysis. Annals
mary systemic small-vessel vasculitides? Best Practice and Research of Internal Medicine, 123, 925–32.
Clinical Rheumatology, 19, 307–20. Rao, J.K., Allen, N.B., and Pincus, T. (1998). Limitations of the 1990 American
Leavitt, R.Y., Fauci, A.S., Bloch, D.A., et al. (1990). The American College of College of Rheumatology classification criteria in the diagnosis of vascu-
Rheumatology 1990 criteria for the classification of Wegener’s granulo- litis. Annals of Internal Medicine, 129, 345–52.
matosis. Arthritis and Rheumatism, 33, 1101–7. Reinhold-Keller, E. and Zink, A. (2011 ). [Morbidity and mortality in sys-
Lie, J.T. (1990). Illustrated histopathologic classification criteria for selected temic rheumatoid diseases:  how did they change?]. Zeitschrift für
vasculitis syndromes. American College of Rheumatology Subcommittee Rheumatologie, 70, 462–3.
on Classification of Vasculitis. Arthritis and Rheumatism, 33, 1074–87. Reinhold-Keller, E. and Moosig, F. (2011 ). [Development of morbidity and
Little, M.A., Nightingale, P., Verburgh, C.A., et al. (2010). Early mortality in mortality in ANCA-associated vasculitis]. Zeitschrift für Rheumatologie,
systemic vasculitis:  relative contribution of adverse events and active 70, 486–92.
vasculitis. Annals of the Rheumatic Diseases, 69, 1036–43. Reuter, M., Schnabel, A., Wesner, F., et  al. (1998). Pulmonary Wegener’s
Lombard, C.M., Duncan, S.R., Rizk, N.W., et  al. (1990). The diagnosis of granulomatosis: correlation between high-resolution CT findings and
Wegener’s granulomatosis from transbronchial biopsy specimens. clinical scoring of disease activity. Chest, 114, 500–6.
Human Pathology, 21, 838–42. Sanchez, A., Acevedo, E., and Sanchez, C. (2006). Epidemiology of the
Luqmani, R., Suppiah, R., Edwards, C.J., et al. (2011a). Mortality in Wegener’s primary vasculitides in a Latin-American population. Arthritis and
granulomatosis:  a bimodal pattern. Rheumatology (Oxford), 50, Rheumatism, 54 (Suppl.), S757.
697–702. Savige, J., Gillis, D., Benson, E., et  al. (1999). International Consensus
Luqmani, R.A., Bacon, P.A., Moots, R.J., et al. (1994). Birmingham Vasculitis Statement on Testing and Reporting of Antineutrophil Cytoplasmic
Activity Score (BVAS) in systemic necrotizing vasculitis. Quarterly Antibodies (ANCA). American Journal of Clinical Pathology, 111,
Journal of Medicine, 87, 671–8. 507–13.
416 SECTION 5 vasculitic diseases and syndromes and related disorders

Schinke, S., Herrmann, K., Gross, W., et al. (2012). PS 216 Differenzialdiagnostischer Takala, J.H., Kautiainen, H., and Leirisalo-Repo, M. (2010). Survival of
Nutzen von Procalcitonin (PCT) bei Granulomatose mit Polyangiitis (GPA, patients with Wegener’s granulomatosis diagnosed in Finland in
Wegener) und CRP-Anstieg. Der Internist, 53 (Suppl. 1), 98. 1981-2000. Scandinavian Journal of Rheumatology, 39, 71–6.
Schmidt, J., Pulido, J.S., and Matteson, E.L. (2011). Ocular manifestations of Tomasson, G., Grayson, P.C., Mahr, A.D., et al. (2012). Value of ANCA meas-
systemic disease: antineutrophil cytoplasmic antibody-associated vascu- urements during remission to predict a relapse of ANCA-associated
litis. Current Opinion in Ophthalmology, 22, 489–95. vasculitis—a meta-analysis. Rheumatology (Oxford), 51, 100–9.
Schmitt, W.H., Heesen, C., Csernok, E., et al. (1992). Elevated serum levels van Timmeren, M.M. and Heeringa, P. (2012). Pathogenesis of
of soluble interleukin-2 receptor in patients with Wegener’s granuloma- ANCA-associated vasculitis:  recent insights from animal models.
tosis. Association with disease activity. Arthritis and Rheumatism, 35, Current Opinion in Rheumatology, 24, 8–14.
1088–96. Walsh, M., Flossmann, O., Berden, A., et al. (2012). Risk factors for relapse of
Schnabel, A., Reuter, M., Gloeckner, K. et al. (1999). Bronchoalveolar lav- antineutrophil cytoplasmic antibody-associated vasculitis. Arthritis and
age cell profiles in Wegener’s granulomatosis. Respiratory Medicine, 93, Rheumatism, 64, 542–8.
498–506. Walton, E.W. (1958). Giant-cell granuloma of the respiratory tract (Wegener’s
Schnabel, A., Holl-Ullrich, K., Dalhoff, K., et al. (1997). Efficacy of transbron- granulomatosis). British Medical Journal, 2 (5091), 265–70.
chial biopsy in pulmonary vaculitides. European Respiratory Journal, 10, Wang, G., Hansen, H., Tatsis, E., et al. (1997). High plasma levels of the solu-
2738–43. ble form of CD30 activation molecule reflect disease activity in patients
Schonermarck, U., Lamprecht, P., Csernok, E.. et  al. (2001). Prevalence with Wegener’s granulomatosis. American Journal of Medicine, 102,
and spectrum of rheumatic diseases associated with protein- 517–23.
ase 3-antineutrophil cytoplasmic antibodies (ANCA) and Watts, R., Lane, S., Hanslik, T., et al. (2007). Development and validation of
myeloperoxidase-ANCA. Rheumatology (Oxford), 40, 178–84. a consensus methodology for the classification of the ANCA-associated
Schultz, H., Schinke, S., Weiss, J., et  al. (2003). BPI-ANCA in transporter vasculitides and polyarteritis nodosa for epidemiological studies. Annals
associated with antigen presentation (TAP) deficiency:  possible role of the Rheumatic Diseases, 66, 222–7.
in susceptibility to Gram-negative bacterial infections. Clinical and Watts, R.A., Al-Taiar, A., Scott, D.G., et al. (2009). Prevalence and incidence
Experimental Immunology, 133, 252–9. of Wegener’s granulomatosis in the UK general practice research data-
Socias, R., James, D.G., and Pozniak, A. (1987). Wegener and Wegener’s gran- base. Arthritis and Rheumatism, 61, 1412–6.
ulomatosis. Thorax, 42, 920–1. Wegener, F. (1935). Über generalisierte, septische Gefässerkrankungen.
Stone, J.H. (2003). Limited versus severe Wegener’s granulomatosis:  base- Verhandlungen der Deutschen Pathologischen Gesellschaft, 29, 202–10.
line data on patients in the Wegener’s granulomatosis etanercept trial. Wegener, F. (1939). Über eine eigenartige rhinogene Granulomatose mit
Arthritis and Rheumatism, 48, 2299–309. besonderer Beteiligung der des Arteriensystems und der Nieren. Beiträge
Stone, J.H. (2012). IgG4-related disease: nomenclature, clinical features, and zur pathologischen Anatomie und zur allgemeinen Pathologie, 102, 36–8.
treatment. Seminars in Diagnostic Pathology, 29, 177–90. Wegener, F. (1990). [50 years of Wegener’s granulomatosis]. Immunität und
Stone, J.H., Hoffman, C.S., Merkel, P.A., et al. (2001). A disease-specific activ- Infektion, 18, 11–9.
ity index for Wegener’s granulomatosis: modification of the Birmingham Westman, K.W., Bygren, P.G., Olsson, H., et al. (1998). Relapse rate, renal
Vasculitis Activity Score. International Network for the Study of the survival, and cancer morbidity in patients with Wegener’s granuloma-
Systemic Vasculitides (INSSYS). Arthritis and Rheumatism, 44, 912–20. tosis or microscopic polyangiitis with renal involvement. Journal of the
Takala, J.H., Kautiainen, H., Malmberg, H., et  al. (2008). Incidence American Society of Nephrology, 9, 842–52.
of Wegener’s granulomatosis in Finland 1981-2000. Clinical and Yousem, S.A. and Lombard, C.M. (1988). The eosinophilic variant of
Experimental Rheumatology, 26 (Suppl. 49), S81–5. Wegener’s granulomatosis. Human Pathology, 19, 682–8.
CHAPTER 31

Granulomatosis with
polyangiitis (Wegener’s
granulomatosis): treatment
Julia U. Holle, Bernhard Hellmich,
and Wolfgang L. Gross

Introduction Key points
Granulomatosis with polyangiitis (GPA) is characterized by granu- 1. GPA treatment follows evidence-based strategies and is adapted
lomatous inflammation primarily affecting the upper and lower according to disease stage and activity.
respiratory tract, a necrotizing vasculitis and a rapidly progres-
2. Chronicle of treatment:
sive glomerulonephritis. GPA is a disease that can affect multiple
organ systems at one time and has a variable clinical presentation 1970s CYC and CS (NIH studies): improvement of outcome
(see Chapter  30), which can range from isolated granulomatous 1990s controlled trials with cytotoxic agents (European Vasculitis
inflammation (localized disease) to generalized vasculitis. A treat- Study Group: EUVAS)
ment plan for an individual patient should primarily take into
Principle of remission induction (e.g. CYC and CS) and mainte-
account the stage of disease (Table 31.1) and the current activity
nance (e.g. AZA and low-dose CS)
state (Table 31.2). Therefore, a thorough evaluation of the extent
2000s controlled trials with biological agents. Controlled trials
of organ involvement and disease activity at the time of presenta-
with RTX in GPA: RTX emerges as an alternative to CYC for
tion and a basic understanding of the underlying immunopatho-
remission induction of generalized GPA/MPA and is approved
genetic processes (see Chapter 29) are important requirements for
by the FDA in 2011.
the development of a treatment plan that is tailored to the needs of
the individual patient. 3. Research agenda:
Friedrich Wegener’s first description of the disease was based on
an autopsy case (Wegener 1939), reflecting the fact that GPA gener- a. reducing damage by targeted and less toxic therapies
ally had a fatal outcome (prior to the advent of immunosuppres- b. individualized treatment in genetically characterized patients
sive therapy) (Walton 1958). Although corticosteroids (CS) are an c. patient education.
integral part of any treatment regimen for GPA, particularly when
given in high doses during phases of active disease, they are alone
not sufficient for disease control (Hollander and Manning 1967; Design and interpretation of therapeutic
Walton 1958). trials in GPA: points to consider
Only the combination of CS with oral cyclophosphamide
(CYC), as first introduced by Fauci and Wolff in the early 1970s,
Classification and definitions
has clearly prolonged survival, with median survival times of more of disease stages
than 20 years (Hoffman et al. 1992; Reinhold-Keller et al. 2000). As Although in most studies both the vasculitis classification criteria
long-term toxicity of oral CYC limits its use, alternative treatment of the American College of Rheumatology (ACR) (Leavitt et  al.
protocols (e.g. intravenous CYC) as well as alternative immuno- 1990) and the definition of the Chapel Hill consensus conference
suppressive medications (e.g. methotrexate (MTX), azathioprine (CHCC) (Jennette et al. 1994; Jennette et al. 2013) for vasculitides
(AZA), leflunomide (LEF), or mycophenolate mofetil (MMF)) for were applied for identifying patients with GPA, investigators from
less severe disease and maintenance of remission have been utilized. both sides of the Atlantic have often used differing definitions of
Furthermore, for the first time in over 30 years, a new treatment disease stages for subclassification of patients with GPA into clini-
option, rituximab (RTX), has emerged as an alternative to CYC cal subgroups (Table 31.1). Furthermore, some studies of patients
for remission induction in generalized disease (Stone et al. 2010; with GPA have also included those with MPA or even polyarteri-
Jones et al. 2010). Long-term data are needed to evaluate safety and tis nodosa (PAN). This can be problematic because outcome differs
long-term outcomes of RTX-treated patients. between patients with GPA and other ANCA-associated vasculitides
418 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 31.1 Definitions for disease stages used for subclassification of patients with granulomatosis with polyangiitis in clinical trials

Study group Clinical Systemic vasculitis outside Threatened vital Other definitions Serum creatinine Clinical trials
subgroup ENT tract and lung organ function (µmol/l) using definition
EUVAS Localized no no No constitutional symptoms, < 120
ANCA typically negative
Early systemic yes no Constitutional symptoms present, < 120 NORAM
ANCA positive or negative
Generalized yes yes ANCA positive < 500 CYCAZAREM
Severe yes Organ failure ANCA positive > 500 MEPEX
Refractory yes yes Refractory to standard therapy any SOLUTION
WGET Limited allowed no Not severe ≤ 124, if haematuria, WEGET
Research but no red blood cell
Group/VCRC casts present
Severe yes yes Organ- or life-threatening disease any WEGET

EUVAS, European Vasculitis Study Group; WGET, Wegener’s granulomatosis etanercept trial; VCRC, Vasculitis Clinical Research Consortium; NORAM, Non-renal Wegener’s Granulomatosis
treated Alternatively with Methotrexate; CYCAZAREM, Cyclophosphamide or Azathioprine as Remission Therapy for Vasculitis; MEPEX, Methylprednisolone Versus Plasma Exchange;
SOLUTION, Antithymocyte Globulin in Refractory Wegener’s Granulomatosis.
(Hellmich B. 2007)

(AAV) despite the same treatment, as demonstrated in the different in trials and in every-day practice (de Groot et al. 2001b). A uni-
relapse rates in the cyclophosphamide or azathioprine as remission form and internationally accepted disease assessment tool in GPA
therapy for vasculitis (CYCAZAREM) trial (Jayne et al. 2003). or ANCA-associated vasculitis is highly desirable and initiatives
towards such an consensus instruments, involving the OMERACT
Outcome Parameters and End Points process, are in progress (Merkel et al. 2011).
Definitions of outcome parameters have not been uniform across Differences in the evaluation of patients in the course of a clinical
clinical trials. Outcome is usually defined in terms of disease states trial represent another important factor influencing interpretation
such as remission or relapse (Table 31.2) and is quantified by the
use of disease assessment instruments. While in the past com-
plete remission was defined as the absence of any disease activ- Table 31.2 Definitions for disease states used for assessment of
ity related to active GPA, American investigators designing the outcome in granulomatosis with polyangiitis
WGET and RAVE trials (Wegener's Granulomatosis Etanercept
Trial (WGET) Research Group  2005; Stone et  al. 2010)  (see Study group Disease state Definitions
Section Corticosteroids) felt that, in addition, remission should EUVAS Complete Absence of any disease activitya, BVAS = 0
be maintained in the absence of any corticosteroid (CS) treatment remission
for a specified period before patients were judged to be in remis-
Partial remission Significant decrease of disease activity with
sion (Table  31.2), while other investigators (European Vasculitis persisting low-grade activity believed to
Society: EUVAS) allow low-dose CS. Although a complete with- regress by continued treatment
drawal of CS is desirable, it seems questionable whether it is a real-
Minor relapse Recurrent or new disease activity not
istic goal for all patients and whether it should be an end point for
threatening vital organs which is controllable
clinical studies in GPA given the comparably high relapse rates dur- by an increase of CS dosage only
ing maintenance therapy with currently available drugs without CS.
With respect to maintenance studies, time to relapse is a common Major relapse Recurrent or new disease activity threatening
vital organs or leading to functional
end point across controlled trials.
impairment which usually requires
Disease assessment tools reinstitution of CYC

Disease activity of GPA is quantified by disease assessment tools, WEGET Sustained BVAS/WG of 0 for 6 months
which record disease activity in different organ systems in speci- Research remission
Group/VCRC
fied item lists (see Chapter  30). Most frequently used are the
original Birmingham vasculitis activity score (BVAS) (Luqmani Low level of BVAS/WG <3 for 6 months
et  al. 1994)  or modified versions:  the BVAS/WG, which incor- disease activity
porates several items specific for GPA (Stone et al. 2001), or the Flare Increase of at least one point in BVAS/WG
BVAS 2003, which is a further modification of the initial BVAS
a Clinical features, serology, and imaging.
(Mukhtyar et al. 2009b). In contrast to the BVAS and its derivatives,
EUVAS, European Vasculitis Study Group; WGET, Wegener’s Granulomatosis Etanercept
the disease extent index (DEI) incorporates no subjective weight- Trial; VCRC, Vasculitis Clinical Research Consortium.
ing according to the seriousness of the items, but is easier to use (Mukhtyar et al. 2009, Hellmich et al. 2007)
CHAPTER 31 granulomatosis with polyangiitis (wegener’s granulomatosis) 419

of outcomes. For example, close surveillance with frequent evalua- active GPA and MPA both in the USA and in Europe (see Section
tion of subclinical disease using sensitive imaging procedures, such Rituximab) (Stone et al. 2010). In case of organ failure due to vas-
as magnetic resonance imaging or high-resolution CT, is likely to culitic activity—especially renal failure—plasma exchange may be
detect more disease activity in a specified period. Furthermore, the used to induce remission. Methotrexate (MTX) may be used for
assessment of certain symptoms is likely to vary among investiga- remission induction in non-organ-threatening (early systemic) dis-
tors. For example, the evaluation of granulomatous manifestations, ease. Given the high cumulative toxicity of CYC, treatment should
such as nasal crusting or orbital masses (‘granuloma’), is par- be switched to less toxic agents such as MTX, AZA, LEF, or MMF
ticularly challenging as it is often difficult to distinguish between as soon as remission is achieved. The duration of maintenance
damage (‘scar tissue’) and persistent low-grade activity (‘grum- treatment is unclear. Currently, the EULAR/EUVAS recommends
bling disease’). In addition, the length of follow-up is crucial for maintenance treatment for at least 18 months. However, in spite of
the interpretation of relapse rates. Finally, concomitant treatments maintenance strategies, relapse rates are still high (35 to 51% in 3
and CS doses often contribute significantly to outcome, especially to 5 years) (Holle et al. 2011). In the future, biomarkers may help
in uncontrolled observational studies. In summary, variations in to identify patients at risk for relapse and to adapt immunosup-
trial design, and particularly disease assessment, are likely to have pression in accordance with personal risk and prognosis. Recently,
a major impact on reported outcomes in therapeutic trials in GPA. a CD8+ T-cell transcription signature has been identified as a risk
factor for relapse in AAV (McKinney et al. 2010) but requires fur-
ther validation.
Approaches to treatment of GPA In approximately 10% of cases, administration of CYC and CS is
The current approach to treating GPA is strongly influenced by insufficient to control disease activity completely. In these cases of
EUVAS, VCRC, and NIH trials. The initial goal of treatment is refractory disease, biological agents, or alternative treatments such
induction of remission; the next is maintenance of remission. One as deoxyspergualin may be needed (Table 31.5).
of today`s major challenges is the treatment of refractory disease
states. Treatment recommendations have been published in 2009 Standard therapeutic regimens in GPA
by the EULAR/EUVAS group (Hellmich et al. 2007; Mukhtyar et al.
2009a) and will be discussed in detail in this chapter; moreover, Corticosteroids
data from controlled trials have been published since then and It is generally accepted that CS alone are insufficient to control
will be incorporated into the treatment schemes despite the fact active generalized GPA. In historic cohorts, CS monotherapy
that they are not yet formally incorporated into the recommenda- prolonged median survival by only 7.5  months (Hollander and
tions. Treatment for induction of remission during phases of active Manning 1967). However, when given as an adjunct to other
disease (Table 31.3) is usually followed by less aggressive therapy immunosuppressive agents (CYC, MTX, etc.) CS represent a cor-
for maintenance of remission (Table 31.4). As detailed in Section nerstone in the treatment of GPA for remission induction and
Standard Therapeutic Regimens in GPA, cyclophosphamide (CYC) maintenance. For induction of remission, 1 mg/kg prednisolone
is one option for patients with active generalized and severe GPA. (PRD) is given as a starting dose in conjunction with CYC or MTX
RTX has emerged as an alternative treatment to CYC, especially (Mukhtyar et al. 2009a). In case of life- or organ-threatening dis-
in young patients or in patients with high cumulative CYC doses ease (e.g. alveolar haemorrhage, rapid progressive glomerulone-
or contraindications to CYC and has been licensed to treat severe, phritis) high-dose intravenous methylprednisolone (250–1000 mg

Table 31.3 Recommendations for induction of remission in granulomatosis with polyangiitis

Protocol Disease stage Dose Category of Grade of References


evidenced recommendationsd
Cyclophosphamide Generalizedc 2 mg/kg/day p.o. I-b A Fauci et al. 1983; de Groot et al. 2009; Guillevin
(daily oral)a et al. 1997; Harper et al. 2010; Hoffman et al. 1992;
Jayne et al. 2003; Reinhold-Keller et al. 2000
Cyclophosphamide Generalizedc 15–20 mg/kg i.v. every I-a A Adu et al. 1997; de Groot et al. 2001a; de
(Pulse)a 3rd week Groot et al. 2009; Guillevin et al. 1997; Harper L et al. 2010
Rituximab Generalized/ 375 mg/m2 4 weekly I-b A Jones et al. 2010; Stone et al. 2010, Specks
severe infusions et al. 2013
Methotrexatea Early systemicc 0.3 mg/kg/week. I-b A Faurschou et al. 2012; de Groot et al. 1998;
s.c. or p.o de Groot et al. 2005; Sneller et al. 1995
Trimethoprim/ Localizedc 2 × 960 mg/day p.o. II-a B Reinhold-Keller et al. 1996
sulfamethoxazole
Plasma exchangeb Severec 40–60 ml/kg (4–7×) I-b A Jayne et al. 2007; Walsh et al. 2013
a Plus prednisone (starting dose 1mg/kg).
b Plus cyclophosphamide (daily oral) and prednisone (starting dose 1 mg/kg).
c For definitions of disease stages see Table 31.1.
d Based on the study(ies) with the highest level of evidence or grade of recommendations according to (Shekelle et al. 1999), see Table 31.6 for definitions.
420 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 31.4 Recommendations for maintenance of remission in granulomatosis with polyangiitis

Protocol Dose Category of evidenceb Grade of recommendationsb References


Azathioprinea 2 mg/kg/day p.o. I-b A Jayne et al. 2003; Pagnoux et al. 2008
Methotrexatea 0.3 mg/kg/week I-b A Langford et al. 1999; Metzler et al. 2005;
p.o. Pagnoux et al. 2008; Reinhold-Keller et al. 2002

Leflunomidea 30–40 mg/day I-b A Metzler et al. 2004; Metzler et al. 2005
Trimethoprim/ 2 × 960 mg/day p.o. I-bc Ac Reinhold-Keller et al. 1996; Stegeman et al. 1996
sulfamethoxazolea
Mycophenolate mofetila 2 g/day I-b A Haubitz et al. 2002; Hiemstra et al. 2010;
Langford et al. 2004; Nowack et al. 1999
Deoxyspergualin 0.5 mg/kg/day III C Schmitt et al. 2005
a Prednisone should be tapered to ≤7.5 mg/day.
b Based on the study(ies) with the highest level of evidence or grade of recommendations according (Shekelle et al. 1999), see Table 31.6 for definitions.
c Significantly reduced incidence of relapses for upper airway disease only.

daily on 3 consecutive days) should be used. The dosage is then trial used CYC 2 mg/kg/day orally together with prednisone (PRD)
subsequently tapered in steps of 5 to 10 mg/day according to dis- at an initial dose of 1 mg/kg/day to induce and maintain remission.
ease activity. In view of the predictable long-term damage related In a cohort of 133 patients treated with this regimen at the NIH,
to the use of high-dose CS (e.g. osteoporosis, diabetes), CS should 91% responded with 75% patients achieving complete remission
be tapered as low as possible. EULAR recommends tapering PRD (Hoffman et al. 1992). Mortality related to GPA or its therapy was
to 10 mg/day or less. In general, a PRD dose below 7.5 mg per day 13%. Based on these data, CYC and CS were used in many cases
should be the target to minimize CS-induced side-effects. However, for several years not only to induce but also to maintain remis-
due to persistent or ‘grumbling’ disease activity upon complete CS sion, leading to high cumulative doses of both CYC and CS and
withdrawal, it is often necessary to continue a low dose of CS (5 high treatment-related morbidity and mortality. The extended use
mg PRD or below) even during the maintenance phase. In fact, in of CYC is limited by often serious morbidity, including infections,
studies where CS were rapidly withdrawn (such as in the WGET leukopenia related to bone marrow failure, infertility, and haemor-
or RAVE study), remission induction rates were relatively low and rhagic cystitis. Besides the risk of developing haemorrhagic cysti-
relapse rates high (Wegener's Granulomatosis Etanercept Trial tis, a large body of evidence suggests that administration of CYC
(WGET) Research Group 2005; Stone et al. 2010), which argues in is linked to a nine- to 45-fold increased risk for bladder cancer in
favour of the continuation of low-dose CS. GPA patients (Hellmich et al. 2004; Hoffman et al. 1992; Knight
et al. 2002; Reinhold-Keller et al. 2000; Talar-Williams et al. 1996).
Cyclophosphamide In a large cohort of 1065 patients with GPA from Sweden, Knight
Oral cyclophosphamide and co-workers reported a 31-fold relative risk and a cumulative
Investigators at the National Institutes of Health (NIH) were the risk of 3% at 10  years and 10% at 15  years after treatment with
first to formally study the effects of CYC in combination with CS cyclophosphamide (Knight et al. 2002). Interestingly, eight patients
in patients with GPA (Fauci et al. 1983; Fauci and Wolff 1973). The within this cohort had a diagnosis of bladder cancer before GPA

Table 31.5 Recommendations for treatment of refractory disease in granulomatosis with polyangiitis

Protocol Dose Category of evidenceb Grade of recommendationsb References


Intravenous immunoglobulin 5 × 400 mg/kg i.v. I-b A Jayne et al. 2000
Rituximaba 375 mg/m2/week II-c B Numerous studies; see Rutgers and
for 4 weeks Kallenberg 2012 for review
Infliximaba 5 mg/kg twice monthly II-b B Booth et al. 2004; Lamprecht et al. 2002
Deoxyspergualin 0.5 mg/kg/day II-b B Birck et al. 2003; Flossmann et al. 2009;
Schmitt et al. 2005
Mycophenolate mofetil 2 g/day III C Koukoulaki et al. 2006
Antithymocyte Globulin (ATG) 5 mg/kg i.v. for 10 days III C Schmitt et al. 2004
a Plus standard therapy (cyclophosphamide and prednisone).
b Based on the study(ies) with the highest level of evidence or grade of recommendations according to (Shekelle et al. 1999), see Table 31.6 for definitions.

(Modified from Mukhytar et al. 2009.)


CHAPTER 31 granulomatosis with polyangiitis (wegener’s granulomatosis) 421

Table 31.6 Categorization of evidence and recommendations randomized trials mentioned in Section Oral Cyclophosphamide
according to Shekelle et al. 1999 indicated that compared to daily oral CYC, pulse CYC appeared to
be equally effective in inducing remission with less adverse events
Category of evidence Strength of recommendations (de Groot et al. 2001a); however, relapse rates tended to be higher
Ia Evidence from meta-analysis of A directly based on category I evidence with intermittent pulse therapy.
randomized controlled trials The methodological limitations of the studies mentioned in the
previous paragraph stimulated the CYCLOPS trial (Randomized
Ib Evidence from at least one
randomized controlled trial Trial of Daily Oral versus Pulse Cyclophosphamide as Therapy for
ANCA-associated Vasculitis) (de Groot et al. 2009). In this trial,
IIa Evidence from at least one B directly based on category II evidence 149 AAV patients with renal involvement (creatinine <500 µmol/l)
controlled study without or extrapolated recommendation from
were randomized to intravenous pulse CYC (15 mg/kg bw pulse
randomization category I evidence
every 2 to 3 weeks until remission plus 3 months) or to oral CYC
IIb Evidence from at least one other (initially 2 mg/kg bw/day until remission, then 1.5 mg/kg bw/day
type of quasiexperimental study for 3  months) and then switched to AZA (2 mg/kg bw/day plus
III Evidence from non-experimental C directly based on category III evidence low-dose CS). There was no difference in the remission rate at
descriptive studies, and case– or extrapolated recommendation from 9 months (pulse CYC 78.7% vs. oral CYC 88.1%) and in median
control studies category I or II evidence time to remission (in both groups 3 months) as well as the devel-
IV Evidence form expert committee D directly based on category IV evidence opment of end-stage renal disease, serious adverse events, and
reports or opinions or clinical or extrapolated recommendation from adverse events. There was a significant difference in the cumula-
experience of respected category I, II, or III evidence tive CYC dose (8.2 vs. 15.9 g, P <0.001) and episodes of leucope-
authorities, or both nia. Interestingly, there were 13 relapses in the pulse CYC group
compared to six in the oral CYC group. This trend was confirmed
in the long-term follow-up (median 4.3 years), which showed no
was diagnosed, resulting in a relative risk of 2.1 (95% CI 0.6–3.6) differences in survival between the two groups but confirmed a sig-
compared to the expected prevalence of bladder cancer in Sweden nificantly lower relapse rate in the oral CYC compared to the pulse
(Knight et al. 2004), further substantiating earlier observations sug- CYC group (20.8% versus 39.5% with at least one relapse, P=0.029)
gesting that GPA may be associated with cancer (i.e. renal cell can- (Harper et al. 2012).
cer) (Tatsis et al. 1999). Although epidemiological studies have not
yet shown a clear cut-off, the risk of cyclophosphamide-associated Rituximab
malignancies appears substantially increased at cumulative cyclo-
Rituximab (RTX) has recently emerged as an alternative to CYC
phosphamide doses above 100 g.
for remission induction in generalized and severe GPA and MPA.
In view of the fact that long-term toxicity of CYC is correlated
In the RAVE trial, oral CYC (2 mg/kg bw/day for 3–6 months) ver-
with the cumulative dose, approaches to limit the total exposure
sus four weekly infusions of 375 mg/m2 RTX were compared as
to CYC have been studied. Cumulative CYC doses can be lim-
remission induction protocol in GPA/MPA (creatinine <4 mg/dl).
ited by either shortening the period of oral CYC therapy, by using
In both arms, patients received three pulses of 1 g methylpredni-
intravenous pulse therapy (CYCLOPS trial, see Section Pulse
solone initially, and were tapered from CS after 6 months. The oral
Cyclophosphamide), by substituting another, less toxic immuno-
CYC group additionally received AZA for remission maintenance
suppressant such as AZA once remission is achieved (Jayne et al.
over a period of 18 months. Of 197 patients, 84 on RTX and 81 on
2003) (CYCAZAREM trial, see Section Pulse Cyclophosphamide),
CYC completed 6 months, after which there was no difference in
or by completely avoiding CYC and using other immunosuppres-
the rate of patients achieving remission (defined as BVAS/WG = 0
sant medications for remission induction such as rituximab (Stone
under no CS). Thus, 64% of patients in the RTX group and 55% in
et al. 2010) (RAVE trial, see Section Rituximab). Cohort studies,
the CYC group reached this primary end point. Interestingly, a sub-
including our own, suggest that limiting use of CYC and applying a
group analysis including the patients treated for relapse only dem-
strict maintenance regimen had a significant impact on the reduc-
onstrated significantly higher remission rates in patients treated
tion of therapy-related morbidity and mortality and also led to a
with RTX compared to CYC (Stone et  al. 2010). Follow-up data
decline in relapse rates (Holle et al. 2011).
confirmed these findings (Specks et al. 2013). To conclude, RTX
Pulse cyclophosphamide was non-inferior for inducing remission compared to oral CYC in
The intermittent use of intravenous (i.v.). pulses is an alternative patients with generalized GPA and MPA and was superior in the
strategy to reduce the cumulative exposure of CYC. To date, five subgroup of patients treated for relapse. In general, remission rates
prospective trials on the efficacy of pulse CYC therapy in GPA have were low, which is probably a result of the strict definition of remis-
been published, of which three had a randomized design (Adu et al. sion (BVAS=0 and no CS at month 6) and argues in favour of keep-
1997; Guillevin et al. 1997; Haubitz et al. 1998). However, none of ing a low-dose CS treatment beyond the remission induction phase
these trials was sufficiently sized to permit confident conclusions (see Section Corticosteroids).
regarding the efficacy of pulse versus oral CYC. Furthermore, the Similar results were noted in the RITUXVAS trial (Jones et al. 2010),
inclusion of patients with MPA and PAN in two of the three ran- which assessed the efficacy of RTX (375 mg/m2 four times weekly)
domized trials represents an important confounding factor for the plus two CYC pulses compared to CYC pulses for 3–6 months only
efficacy analysis as it has been shown in the CYCAZAREM trial that (44 patients, 3:1 randomization). Patients of both arms received con-
patients with GPA relapsed more frequently (18%) than patients comitant CS, which were discontinued after 12 months. Patients who
with MPA (8%) (Jayne et al. 2003). A meta-analysis of the three received CYC pulses only were switched to remission maintenance
422 SECTION 5 vasculitic diseases and syndromes and related disorders

with AZA after 3–6 months. In this trial there were no differences treatment in GPA (de Groot et al. 1998; De Groot et al. 2005; Sneller
in the primary end point (remission at 12 months) (76% vs. 82%) et al. 1995). In the two prospective open-label studies MTX was
(Jones et al. 2010). From this trial it was concluded that the com- given at doses of around 0.3 mg/kg/week combined with CS to GPA
bined CYC-RTX regimen is not inferior to CYC pulse and that RTX patients who did not have evidence for life-threatening disease and
was effective in sparing CYC pulses. The data of these two studies who had a normal or near to normal renal function. Successful
provide evidence for an alternative, non-inferior treatment to CYC induction of remission was seen in 10 of 17 (59%) and 33 of 42
in the remission induction of generalized/severe AAV. Side-effects patients (79%), respectively.
were not significantly different in both treatment arms and need to In the prospective, unblinded, randomized, controlled trial
be carefully assessed in the long run. Based on these data, RTX was (NORAM), there were 100 patients with AAV; 89 (94%) had
approved as treatment for severe GPA/MPA in the USA in 2011 and GPA without life- or organ-threatening disease (a creatinine
Europe in 2013. Current British guidelines suggest that RTX should <150 µmol/l) and were randomized to treatment with either MTX
especially be considered for remission induction in patients in whom at a target dose of 20–25 mg/week or daily oral CYC (2 mg/kg).
avoidance of CYC is desirable (i.e. in patients with high cumulative After 12  months from study entry, therapy was stopped in both
CYC doses), in patients who relapse, and in young patients to pre- arms without subsequent maintenance treatment. Results of the
serve fertility (Guerry et al. 2012). NORAM trial show that methotrexate (MTX) can be as effective as
Data from uncontrolled trials and case series and preliminary data CYC, with remission rates of 89.8% for MTX and 93.5% for CYC
from one controlled trial (MAINRITSAN trial) suggest that RTX and similar times to remission (De Groot et al. 2005). Relapse rates
may also be an effective maintenance treatment when administered at 6  months after discontinuation of therapy were high for both
intermittently (Terrier et al. 2013). Rhee et al. showed that repetitive MTX (69.5%) and CYC (46.5%). Furthermore, the long-term out-
administration of RTX at month 0 (two times 1 g), month 6 (1 g), come data of the NORAM trial showed the relapse-free survival
and month 12 (1 g) in 39 patients with AAV on conventional immu- tended to be higher in the CYC group compared to the MTX group
nosuppression led to a significant decrease of the BVAS/WG score (P=0.65) with no differences in major adverse events (Faurschou
and to the reduction of the concomitant immunnosuppression (Rhee et al. 2012). The administration of long-term CYC is definitely not a
et al. 2010). Likewise Cartin-Ceba et al. (in a series of 53 patients with viable treatment option despite its effectiveness due to its associated
AAV) and Smith et al. (in a series of 92 patients with AAV) showed morbidity. A major lesson of the NORAM trial is that some form
that RTX successfully induced remission in relapsing patients and of immunosuppressive treatment should be continued beyond
maintains remission effectively when administered pre-emptively 12 months for maintenance of remission.
(Cartin-Ceba et al. 2012; Smith et al. 2012). Preliminary data from Two standardized, randomized, open-label trials have investi-
the MAINRITSAN trial suggest that intermittent RTX at a low dose gated the efficacy of MTX for maintenance of remission in GPA
of 500 mg every 6 months may be superior to AZA in preventing after successful induction of remission with oral CYC (Langford
major relapses after remission induction with CYC. A full publica- et  al. 1999; Langford et  al. 2003; Reinhold-Keller et  al. 2002).
tion of these data is still pending. A second controlled trial to assess Furthermore, a large randomized, open-label, controlled trial, the
the efficacy of RTX as maintenance treatment in AAV (compared to WEGENT study, assessed toxicity (adverse events requiring dis-
AZA) is underway (RITAZAREM study). Therefore, RTX may have continuation of the study drug or death) in 126 patients with GPA
an important role in maintenance treatment in the future. or MPA receiving either oral MTX or AZA for maintenance after
remission induction with intravenous CYC (Pagnoux et al. 2008).
Practice points: Cyclophosphamide and Rituximab There was no significant difference in terms of adverse events
CYC has been the mainstay of treatment for many years. Toxicity requiring stopping of the drug between the medications, although
seems to be related especially to long-term use and high cumulative there was a trend favouring AZA with respect to the primary end
doses. Even if oral CYC is more effective in preventing relapses in the point (drug discontinuation/death: seven in the AZA group vs. 12
long-term compared to pulse CYC, short periods of CYC adminis- in the MTX group, P=0.21). Moreover relapse free survival did not
tration (3–4 months) and pulse CYC should be considered in order differ significantly (23 relapses under AZA vs. 21 relapses under
to minimize side-effects and a sufficient maintenance treatment MTX; P=0.71).
should be routinely used. RTX is non-inferior in inducing remis-
sion in generalized GPA/MPA and superior in patients who relapse. Practice points: Methotrexate
However, additional data are required to assess long-term efficacy In summary, in daily practice MTX and PRD can be used in early
and potential side-effects. RTX may especially be chosen in order systemic GPA to induce remission (according to NORAM) when
to avoid CYC (high cumulative CYC doses), in patients who relapse there is no significant kidney involvement and no renal insuffi-
and in young patients. In severe disease (i.e. renal failure) plasma ciency. Additionally, treatment with MTX or AZA may be used for
exchange is recommended in addition to immunosuppressive treat- remission maintenance in GPA/MPA, as shown by the WEGENT
ment (with CYC or RTX, see Section Plasma Exchange). RTX may trial. Importantly, the use of MTX is limited to patients with pre-
also have a role in maintenance treatment in the future as data from served kidney function.
a preliminary trial suggest superiority in preventing major relapses
compared to AZA. The efficacy of RTX as maintenance treatment is Azathioprine
currently being assessed in a second large, controlled trial. As mentioned in Section Pulse Cyclophosphamide, the
CYCAZAREM trial conducted by the EUVAS (Jayne et al. 2003) was
Methotrexate the first controlled trial to demonstrate that CYC can be substituted
Two open-label studies and one randomized trial evaluated the by a less potent and less toxic maintenance drug after successful
efficacy of MTX as a potentially less toxic alternative for induction remission induction: GPA/MPA patients (n=155), who went into
CHAPTER 31 granulomatosis with polyangiitis (wegener’s granulomatosis) 423

remission after treatment with CYC 2 mg/kg and PRD for 3 to microscopic polyangiitis in a small, open-label study (17 patients)
6 months were randomized to either continued CYC therapy for a (Silva et  al. 2010), it should currently not be used as a routine
total of 12 months or AZA as a substitute for CYC. After 12 months, remission-inducing agent in generalized GPA due to the lack of
the CYC group was also switched to AZA and followed for an addi- controlled trials. A randomized controlled trial to assess the effi-
tional 6 months. Relapse rates during the 18-month period were cacy of CYC versus MMF in generalized GPA/MPA is currently
similar for the long-term (13.7%) compared to the short-term CYC underway. A full publication of the trial (MYCYC) is pending.
group (15.5%). Preliminary data assessing the long-term outcome The safety and efficacy of MMF for maintenance of remission has
of the CYCAZAREM study demonstrated slightly higher relapse been evaluated in three small, open-label clinical trials including
rates in the AZA arm compared to the continued CYC arm with no five to 14 patients (Haubitz and de Groot 2002; Langford et al. 2004;
differences regarding renal outcome in both arms. A full publica- Nowack et al. 1999) and one randomized controlled trial (Hiemstra
tion of the longterm follow-up data is pending. Given the increased et al. 2010) (IMRPOVE). In the study by Nowack and co-workers
risk of toxicity with prolonged CYC administration, results of the only one relapse in 11 patients was seen during a follow-up time of
CYCAZAREM trial suggest that CYC exposure should be limited 15 months. In contrast, in the study by Langford and co-workers
to phases of active disease and should be replaced by a less toxic six of 14 patients relapsed during a median 18 months of follow-up.
therapy as soon as remission is accomplished. Dosage and duration of CYC therapy for induction of remission
Furthermore, data of the WEGENT trial demonstrated equal as well as MMF dosage (2 g/day) were similar in both studies.
toxicity (leading to discontinuation of treatment) and equal relapse Differences in CS therapy may account for the divergent outcome,
rates of AZA compared to MTX (WEGENT trial, see Section as in the study by Langford and colleagues CS was completely dis-
Methotrexate). Therefore, AZA is a first-line option in the mainte- continued after a median of 8 months while in the study by Nowack
nance of remission of GPA/MPA. et  al. a median dose of 5 mg PRD was maintained. Tolerability
of MMF was reported to be good in both studies, but in a small
Practice points: Azathioprine
series of five patients with AAV and end-stage renal disease MMF
AZA is one of the first-line options to maintain remission in doses of >1 g were not well tolerated with anaemia, leukopenia,
GPA/MPA, as demonstrated by randomized controlled trials and gastrointestinal symptoms being the most frequently reported
(CYCAZAREM, WEGENT). In terms of toxicity, there was a trend adverse events (Haubitz and de Groot 2002). In a large, randomized
of fewer discontinuations due to adverse events under AZA as com- controlled trial, MMF (2 g/day) was compared to AZA (2 mg/kg
pared to CYC in the WEGENT trial. AZA should be preferred over bw/day) for the maintenance of remission in GPA/MPA patients
MTX especially in patients with impaired kidney function. A role (n=156) after remission induction with CYC and CS (Hiemstra
of AZA for induction of remission has not yet been established. et al. 2010). After a median follow-up of 39 months, relapses were
Leflunomide more common in the MMF group (42/76 patients) compared to the
AZA group (30/80 patients) with an unadjusted hazard ratio of 1.68
Leflunomide (LEF) inhibits de novo pyrimidine synthesis and
for MMF. Severe adverse events did not differ significantly. On the
inhibits responses of activated T and B cells. In a phase II open-label
basis of this trial, MMF should currently not be used as a first-line
clinical trial LEF (30 mg/day) was given to 20 patients with GPA
treatment option for the maintenance of remission in GPA/MPA.
for maintenance of remission following CYC induction therapy
(Metzler et al. 2004). During a median follow-up of 21 months only Practice points: Mycophenolate
one major relapse requiring reinstitution of CYC was recorded. The efficacy of MMF for remission induction in GPA/MPA is yet to
Eight minor flares were successfully treated by increasing the LEF be evaluated. MMF may be regarded as an agent of second choice
dose to 40 mg/day. In a multicentre randomized controlled clini- in the maintenance of remission. AZA (and probably MTX due to
cal trial (LEM) the potency of LEF (30 mg) to prevent relapses in data from the WEGENT trial) should be preferred in this situation.
patients with WG after successful induction of remission with CYC
was compared to MTX (20 mg weekly). There was a significantly Trimethoprim/sulfamethoxazole
higher rate of severe relapses in the MTX treatment group (n=7)
The use of trimethoprim/ sulfamethoxazole (T/S) in GPA was first
compared to the LEF treatment group (n=1) (Metzler et al. 2007).
introduced by DeRemee in 1985 (DeRemee et  al. 1985)  and has
The study was therefore prematurely terminated but adverse events
since then been studied for its potential to induce and maintain
(hypertension, neuropathy, leukopenia) necessitated a withdrawal
remission. In a prospective, open-label clinical trial, T/S was given to
of LEF in four of 26 patients (Metzler et al. 2007). A slow dose esca-
72 patients with GPA and different disease stages (Reinhold-Keller
lation scheme for MTX and the use of the oral formulation may par-
et al. 1996). Treatment with T/S induced partial or complete remis-
tially account for the higher relapse rate in the MTX treated group.
sion in 11 of 19 patients (58%) with localized GPA, which lasted
Practice points: Leflunomide for a median of 43  months (Reinhold-Keller et  al. 1996). Of the
LEF is a promising drug for maintenance therapy of GPA, which remaining eight patients, five showed local disease progression and
warrants further investigation. In clinical practice, LEF can be used three progressed to generalized disease. In contrast, in patients
as an alternative treatment for maintenance of remission if AZA with generalized GPA neither TS alone or in combination with CS
or MTX are contraindicated or have previously failed to maintain induced sustained remissions.
remission. The efficacy of T/S for maintenance of remission following
standard therapy has been evaluated in a prospective, randomized,
Mycophenolate mofetil placebo-controlled clinical trial (Stegeman et  al. 1996). After
Although mycophenolate mofetil (MMF) was effective for the 24 months, relapse rates in the T/S limb were lower (18%) com-
induction of remission and maintenance in non-severe renal pared to the placebo group (40%); however, subgroup analyses
424 SECTION 5 vasculitic diseases and syndromes and related disorders

revealed that only relapses in the ENT region were significantly was continued until 12 months if patients were in remission (de
reduced by T/S while relapses in all major organ systems were not Menthon et  al. 2011). In the IFX group, three (of nine patients)
prevented. had a response or remission at the end of follow-up, whereas five
Uncontrolled data from a cohort study in localized GPA in of eight RTX patients had responses or remissions at the end of
which treatment was adapted according to disease activity and follow-up. To summarize, both medications were partially effective
severity in a standardized step-up regimen demonstrated that with a slightly better outcome in the RTX group, although a defi-
T/S was not sufficient to prevent disease progression in around nite conclusion cannot be drawn from the study as patient numbers
two-thirds of patients receiving T/S for initially mild disease were too small.
(Holle et al. 2010). Treatment with ATG induced remission in 13 of 15 patients
in an open study of refractory AAV patients. Notwithstanding,
Practice points: Trimethoprim/Sulfamethoxazole
relapse rates and side-effects were high (relapse in seven patients
Summarizing the available data, T/S can therefore only be recom- after 21.8 months, infections in five patients, serum sickness in two
mended in patients with localized disease restricted to the upper patients) (Schmitt et al. 2004).
respiratory tract who do not show signs of rapid disease progres- Deoxyspergualin is a synthetic analogue of spergualin, a natural
sion or destructive disease. product of the bacterium Bacillus lactosporus. Although its exact
mechanism of action remains obscure, DSG has immunosuppres-
Treatment of refractory GPA sive properties and is licensed in Japan for treatment of recurrent
In 10 to 25% of patients, standard CYC therapy fails to induce remis- kidney transplant rejection. Deoxyspegualin was investigated
sion in patients with GPA (Holle et al. 2011). Treatment options in three open trials for remission induction (Birck et  al. 2003;
in refractory disease comprise RTX, TNF- inhibitors, intrave- Flossmann et al. 2009) or maintenance in refractory disease in GPA
nous immunoglobulins (IVIg), deoxyspergualin and antithymo- (Schmitt et al. 2005). Remission induction studies showed remis-
cyte globulin (ATG), and MMF; however, most of the treatments sion rates of 70–95%. In the largest study (Flossmann et al. 2009), a
lack evidence from controlled trials (Mukhtyar et  al. 2009a). In high rate of side-effects and relapses was documented (43% relapse
refractory patients on pulse CYC, a switch to the daily oral dosing after median 170 days of remission in spite of maintenance with
(2 mg/kg) is sometimes successful, possibly related to the higher AZA, 53% SAEs mostly due to leucopenia and 27% lower respira-
cumulative doses applied (Seror et  al. 2010). Currently, RTX is tory tract infections).
probably the most widely used drug in refractory disease with a Intravenous immunoglobulins (IVIg) represent another
large number of open-label studies published so far (Rutgers and treatment option. Several uncontrolled studies demonstrated
Kallenberg 2012). One of the largest studies (Jones et al. 2009) com- a beneficial effect of IVIg in refractory and/or relapsing AAV,
prised 65 patients (46 with GPA) and demonstrated high response in particular when other treatments were not sufficient to con-
rates with RTX: 75% of patients achieved complete remission, 23% trol disease activity (Jayne et al. 1991; Levy et al. 1999; Martinez
partial remission, and no efficacy was documented in only 2%. et al. 2008; Richter et al. 1995; Tuso et al. 1992). Furthermore,
Furthermore, RTX enabled CS reduction from median 12.5 mg/ a small randomized, placebo-controlled trial (34 AAV patients)
day to 9 mg/day (at 6 months) and the withdrawal of immunosup- (Jayne et  al. 2000)  showed a significant higher rate of remis-
pression in 62%. Nevertheless, relapse was frequent, occurring in sion in the IVIg arm (IVIg 400 mg/kg/day for 4 consecutive
half of the patients who achieved complete remission. Cartin-Ceba days) compared to placebo (15 vs. six remissions, P=0.015);
et al. (Cartin-Ceba et al. 2012) demonstrated that Rituximab was however, this effect was not maintained beyond 3  months.
effective in inducing and maintaining remission in 53 refractory Arthritis/ arthralgias, ENT, and pulmonary symptoms, as well
GPA patients. However, it is unclear how to proceed with patients as peripheral neuropathy, were the manifestations that primarily
who received RTX but are refractory. In these cases the following responded to IVIg therapy. As patients with glomerulonephritis
options may be used. and severe lung disease were not included (Jayne et  al. 2000),
In spite of disappointing results of the WGET trial demonstrat- the efficacy of IVIg in patients with severe renal or pulmonary
ing that etanercept, as additional induction and maintenance disease remains uncertain. Further trials are needed to assess
treatment in conjunction with standard treatment, did not yield efficacy of IVIg in this situation. IVIg may in particular be used
higher sustained remission rates compared to standard treatment in refractory situations with concomitant infection in order to
plus placebo (Wegener's Granulomatosis Etanercept Trial (WGET) treat both conditions.
Research Group  2005), TNF- inhibitors may be considered for MMF was efficient in the induction of remission in recurrent,
refractory disease. CYC-resistant AAV or in patients with contraindications for CYC,
Remission rates of 70–88% have been reported from five open as shown by small open studies (Joy et al. 2005; Koukoulaki and
studies using infliximab (IFX) in refractory AAV (Booth et al. 2004; Jayne 2006; Stassen et al. 2007) but should not be considered as a
Booth et al. 2002; Josselin et al. 2008; Lamprecht et al. 2002), but first-line option to treat refractory disease.
patient numbers were relatively small (six to 32 AAV patients, six
to 19 with GPA) and treatment duration as well as follow-up was Specific non-medical therapies
short. In addition, the occurrence of two deaths and a high rate for organ failure
of infections (21%) in the largest study of 32 patients (Booth et al.
2004) mandate close monitoring and cautious use of such inten- Plasma exchange
sive immunosuppression. In a controlled trial comparing IFX to Severe (generalized) disease is defined as organ failure, in par-
RTX in refractory GPA, 17 patients were randomized to receive ticular occurring as respiratory or renal failure (creatinine
either IFX or RTX. After a first assessment at 2 months, treatment >500 µmol/l). Plasma exchange (PLEX) in addition to standard
CHAPTER 31 granulomatosis with polyangiitis (wegener’s granulomatosis) 425

remission induction has been shown to improve renal survival tracheostomy. Intralesional injection of methylprednisolone into
compared to methylprednisolone (MP) pulses in a randomized the stenotic segment, followed by microsurgical lysis of the stenotic
controlled trial in severe renal disease (Jayne et al. 2007). In 137 ring and serial dilatation, was reported to prevent tracheostomy in
newly diagnosed GPA and MPA patients with a creatinine level a series of 21 patients with GPA (Hernandez-Rodriguez et al. 2010;
>500 µmol/l, dialysis independence at 3 months was significantly Hoffman et al. 2003). This therapeutic approach is therefore rec-
higher in the PLEX group compared to the MP group (69% vs. ommended before surgical tracheostomy is performed. In the rare
49%) and the difference was sustained after 1  year; however, event of severe bronchial narrowing with consecutive atelectasis,
patient survival did not differ in the two groups. Furthermore, endobronchial placement of stents can prevent respiratory failure
long-term follow-up data from the MEPEX trial showed that (Hernandez-Rodriguez et al. 2010).
the benefit of a preserved kidney function in the PLEX groups is
not preserved after 4 years, suggesting that PLEX performed in Prophylaxis and treatment
patients with severe renal disease leads to delay of end-stage renal
disease but does not prevent it (Walsh et al. 2013). This is in line
of complications
with a recent meta-analysis (Walsh et al. 2011), demonstrating a The high rate and severity of adverse effects of drugs commonly
moderate benefit of PLEX with respect to prevention of end-stage used in the treatment of GPA warrants close monitoring and, for
renal failure with no effect on survival. certain medications, specific prophylaxis (Table  31.7). Although
No controlled trials are available with respect to alveolar haem- prophylactic therapies help to reduce the incidence of complica-
orrhage and PLEX; however, a retrospective analysis of 20 patients tions, education is the fundamental cornerstone in the prevention
with alveolar haemorrhage who received PLEX reported resolution of adverse events in patients with GPA. In addition to repeated
of alveolar haemorrhage in all of them (Klemmer et al. 2003). patient education the following measures are recommended.
PLEX is currently recommended by the EUVAS in severe renal
disease (creatinine >500 mmol/l) in addition to standard remission Pneumocystis jirovecii prophylaxis
induction. The British Society for Rheumatology suggests perform- Patients with GPA are at a particularly high risk to develop
ing PLEX in alveolar haemorrhage as well (Lapraik et  al. 2007). Pneumocystis jirovecii pneumonia (PJP) and mortality rates of over
A randomized, controlled trial initiated by the EUVAS and VCRC 35% have been reported (Ognibene et al. 1995; Sneller 1998). The
to investigate the efficacy of PLEX in non-severe renal and/or lung largest retrospective review of cases in patients with autoimmune
involvement (GFR <50 ml/min and/or alveolar haemorrhage with- disease revealed that 8.9 of 1000 hospitalizations of patients with
out requirement for mechanical ventilation) is currently underway GPA are caused by PJP (Ward and Donald 1999). Lymphopenia
(PEXIVAS trial). (<1000 cells/mm3), CYC therapy, and high CS doses have been
identified as the three major risk factors for PJ infection (Sowden
Renal transplantation and Carmichael 2004). Patients with a CD4+ T-cell count of <200
Retrospective studies from several centres indicate that the sur- cells/mm3 and a corticosteroid dose of >15 mg prednisolone
vival and graft function of patients with GPA after renal trans- equivalent, or a duration of treatment of >3 months are at a par-
plantation is comparable to other non-diabetic patients following ticularly high risk (Sowden and Carmichael 2004). Although there
transplantation (Deegens et al. 2003; Elmedhem et al. 2003; Geetha are no prospective studies on the use of PJP prophylaxis in patients
et al. 2011; Gera et al. 2007; Schmitt et al. 1993; Shen et al. 2011). with GPA, the administration of co-trimoxazole during induc-
Immunosuppressive therapy after transplantation was not uni- tion therapy with CYC is recommended (Mukhtyar et al. 2009a).
form in the different cohorts and consisted of ciclosporin, AZA, Furthermore, maintaining PJP prophylaxis independent of current
MMF, tacrolimus, PRD or combinations of the respective drugs. treatment in patients with CD4+ counts of <200 cells/mm3 has been
Extrarenal flares can occur despite immunosuppression, yet recur- advocated (Sowden and Carmichael 2004). Although full doses of
rence of GPA in the grafts is rare and seems not to cause graft loss. co-trimoxazole increase the risk of adverse events in MTX-treated
Patient and graft survival rates were 100% at 1 year, 93.4% at 5 years, patients, the recommended three times weekly dose can be admin-
and 67.4% at 10 years in a large retrospective cohort on 85 patients istered safely (Sneller et al. 1995). In sulfa allergic patients, inhaled
with AAV receiving renal transplants (42 with GPA) (Geetha et al. pentamidine 300 mg every 3 to 4 weeks may be used (Table 31.7).
2011). Therefore, renal transplantation should be offered to GPA
patients who progress to end-stage renal disease. Osteoporosis prophylaxis
In view of the often extended need for CS, a risk assessment for the
Subglottic stenosis development of CS-induced osteoporosis is advisable, which ideally
Subglottic stenosis occurs in around 10 to 20% of GPA patients. should include bone density measurements. Prophylaxis with vita-
In a series of 43 patients with GPA and subglottic stenosis fol- min D and calcium should be instituted according to local guide-
lowed by the NIH, the diagnosis of subglottic stenosis was made lines for the prophylaxis and treatment of CS-induced osteoporosis.
when no disease activity of WG was seen at other sites (Langford
et  al. 1996). In ten of 18 patients who were receiving immuno- Prophylaxis of transitional bladder cancer
suppressive therapy, tracheostomy was later required (Langford Comparison of the two largest cohorts of GPA patients suggests
et  al. 1996). Therefore subglottic stenosis may occur in spite of that the use of mesna during CYC therapy limits the risk of devel-
on-going immunosuppression, suggesting that immunosuppres- oping bladder cancer (Hellmich et al. 2004; Hoffman et al. 1992;
sion may not always be effective and scarring contributes to the Holle et  al. 2011; Reinhold-Keller et  al. 2000). Patients in the
progressive narrowing. Intralesional local endoscopic therapy in large cohort of GPA patients observed at the NIH did not receive
patients unresponsive to systemic immunosuppression can prevent mesna along with cyclophosphamide treatment (Hoffman et  al.
426 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 31.7 Frequently used drugs for treatment of granulomatosis with polyangiitis: adverse effects, monitoring, prophylaxis

Medication Common adverse effects Monitoring Prophylaxis


(expected frequency)
Corticosteroids Osteoporosisa Dual-energy X-ray absorbtiometry Calcium 1000–1500 mg daily, vitamin D 400–800 IU daily,
(CS) (in case of other risk factors and/or consider bisphosphonate if T-score is <1.5mg and/or
high cumulative CS exposure) fractures are present
Diabetes mellitusa Blood glucose 3-monthly Patient education: diets
Infectionsa Clinical Patient education: avoid contact with infected people,
recognize early symptoms
Cushing’s syndromea Clinical Early tapering of CS
Skin atrophya Clinical Early tapering of CS
Cyclophosphamide Haemorrhagic cystitis Urinalysis, cystoscopy in case of Give CYC only in the morning
(CYC) (1–50%), transitional cell bladder non-renal haematuria Advise patients to drink sufficient fluids (2–3 l/day)
cancer (1–5%)
Give mesna (dosage in mg = CYC dosage, oral
CYC: divide on two doses in morning and afternoon,
pulse CYC divide on three doses at 0, 4, and 8 h)
Limit cumulative CYC exposure
Haematological: leukopenia Blood cell counts, 2–3 times weekly Frequent monitoring
(50–100%), anaemia, during daily oral therapy, at days 8, 10,
thrombocytopenia and 12 during pulse therapy
Severe infections (up to 30%) Clinical Patient education: avoid contact with infected people,
recognize early symptoms
Pneumocystis jirovecii pneumonia Clinical Thrice weekly doses of co-trimoxazole
Lymphocyte counts in sulfa-allergic patients inhaled pentamidine 300 mg
every 3 to 4 weeks
Nausea, vomiting (variable, up to >50%) Clinical Ondansetron 4–8 mg
Myelodysplasia (1–8%) Blood cell counts, MCV Limit cumulative CYC exposure
Rituximabb(RTX) Allergic infusion reaction (leading to Clinical : monitoring of blood pressure Pretreatment with 100 mg prednisolone i.v. 30 min
termination of the infusion : 1%) and pulse during infusion prior to infusion, pretreatment with analgesics and
antihistamines
Severe infections (7%) Clinical Patient education; vaccination before RTX initiation if
possible
PML (progressive multifocal clinical
leukoencephalopathy)
Depletion of immunoglobulins Assess immunoglobulins levels before Consider retreatment carefully in the event of
(very frequent) and after treatment infections associated with low immunoglobulins
Reduced response towards Assess vaccination status before Administer vaccinations
vaccination treatment
Haematological : leukopenia (3%), Regular blood cell counts No infusion during severe cytopenia
neutropenia, thrombocytopenia (3%)
Methotrexate Stomatitis (11%), alopecia Clinical Folate 15–20 mg 24 h after MTX
(MTX)C
Haematological (2–22%): leukopenia, Blood cell counts, weekly at first Stop if renal function declines
anaemia, thrombocytopenia 4 weeks, then every 2 to 4weeks Folate 15–20 mg 24 h after MTX
Serum creatinine
Elevation of liver enzymes (6%) ALT, AST, weekly at first 4 weeks, Folate 15–20 mg 24 h after MTX
then every 2 to 4 weeks
Infections (24%) Clinical Patient education: avoid contact to infected people,
recognize early symptoms
Pneumocystis jirovecii pneumoniaa ClinicalLymphocyte counts Thrice weekly doses of co-trimoxazole
in sulfa-allergic patients inhaled pentamidine
300 mg every 3 to 4 weeks
(continued)
CHAPTER 31 granulomatosis with polyangiitis (wegener’s granulomatosis) 427

Table 31.7 (Continued)


Medication Common adverse effects Monitoring Prophylaxis
(expected frequency)
Pneumonitis Clinical, consider bronchoalveolar Patient education: symptoms (cough, fever)
lavage in case of unexplained
pneumonitis
Nausea, vomiting Clinical Give MTX parenterally
In severe cases, theophylline on day of MTX
Azathioprine Haematological (3–16%): leukopenia, Blood cell counts, weekly at first 4 Avoid co-medication with allopurinol
(AZA)c anaemia, thrombopenia weeks, then every 2 to 4weeks Patient education: regular lab checks
Serum creatinine Check thiopurine methyltransferase (TPMT) in
case of frequent leukopenia
Elevation of liver enzymes (6%) ALT, AST, weekly at first 4 weeks, Patient education: regular lab checks
then every 2 to 4 weeks
Infections (around 20%) Clinical Patient education: avoid contact with infected
people, recognize early symptoms
Drug fevera Clinical Patient education: consult physician in case of
unexplained fever
Nausea, vomiting, abdominal paina Clinical Check TPMT
Leflunomide Haematological: leukopenia, anaemia, Blood cell counts, weekly at first 4 Patient education: regular lab checks
thrombopeniaa weeks, then every 2 to 4 weeks
Serum creatinine
Elevation of liver enzymesa ALT, AST, weekly at first 4 weeks, Patient education: regular lab checks
then every 2 to 4 weeks
Infectionsa Clinical Patient education: avoid contact to infected
people, recognize early symptoms
Diarrhoeaa Clinical Patient education: report symptoms
Hypertensiona Blood pressure twice-weekly Avoid in patients with pre-existing severe
hypertension
Mycophenolate Haematologicala: leukopenia, Blood cell counts, weekly at first Patient education: regular lab checks
mofetild anaemia, thrombopenia 4 weeks, then every 2 to 4 weeks
Serum creatinine
Elevation of liver enzymesa (0/76 ALT, AST, weekly at first 4 weeks, Patient education: regular lab checks
patients in the IMPROVE trial, then every 2 to 4 weeks
Hiemstra T et al., 2010)
Infections (general)a (12/76 patients, Clinical Patient education: avoid contact with infected
IMPROVE trial, Hiemstra T et al., 2010) people, recognize early symptoms
Diarrhoeaa Clinical Patient education: report symptoms
Nausea, vomiting, abdominal paina Clinical Check TPMT
Hypertensiona Blood pressure twice-weekly Avoid in patients with pre-existing severe
hypertension
a Insufficient data on frequency in GPA.
b Frequencies are given from the RAVE trial (Stone et al. 2010).
c data derived from the WEGENT trial (Pagnoux C et al., 2008).
d data derived from the IMPROVE trial (Hiemstra T et al., 2010).

The frequencies of adverse events were obtained form different randomized trials and cohort studies involving patients with GPA with different times of observation and therefore only
represent rough estimates.
There are no sufficient data on long-term side-effects of RTX.

1992; Talar-Williams et al. 1996). In that cohort, 68 of 158 patients diagnosed from 1999 to 2002 had cyclophosphamide-induced cys-
(43%) and, after extended follow-up, 42 of 145 patients (29%) titis, respectively (Holle et al. 2011; Reinhold-Keller et al. 2000).
developed haemorrhagic cystitis due to cyclophosphamide treat- The decline of cyclophosphamide-induced cystitis over time
ment (Hoffman et al. 1992; Talar-Williams et al. 1996). In contrast, within the German cohort may be explained by the reduction of
in patients receiving mesna together with cyclophosphamide in the cumulative CYC dose per patient. Furthermore, the incidence
the German GPA cohort reported by Reinhold-Keller and Holle, of bladder cancer was significantly lower in the mesna-treated
12% of patients diagnosed from 1966 to 1993 and 6% of patients cohort of Reinhold-Keller (one of 155 patients) compared to the
428 SECTION 5 vasculitic diseases and syndromes and related disorders

not-mesna-treated NIH cohort (seven of 158 patients; P=0.034) maintenance treatment. Additionally, the patient`s individual his-
(Reinhold-Keller et al. 2000). We therefore recommend the use of tory needs to be considered part of the toxicity profile of the respec-
mesna in any patients receiving CYC. Mesna can be given orally tive compounds. Previous intolerability or failure (relapse) of certain
split in three daily doses of 50 mg during oral CYC therapy or medications as well as impaired organ function (kidney, liver, bone
in three doses during the day of intravenous CYC pulses. Urine marrow, etc.) are important points to be considered in the choice
cytology should be checked every 6 months. Allergies, which may of a maintenance treatment for an individual patient with GPA. In
sometimes be difficult to distinguish from cutaneous vasculitic refractory disease, RTX is currently the most widely used drug.
rashes, and an elevation of liver enzymes are the most frequently TNF- inhibitors, deoxyspergualin, and ATG may represent other
adverse events of mesna. In addition, CYC should be given as a alternatives although experience from controlled trials is lacking.
single dose in the morning and patients should be advised to drink IVIg may represent another option for refractory/ relapsing GPA,
frequently. Patients who already experienced haemorrhagic cysti- in particular in the setting of concomitant infection. Preliminary
tis should have cystoscopy every year. data suggest that RTX may have a role in maintenance treatment;
furthermore, trials assessing the duration of maintenance treat-
Treatment of drug-induced ment are underway. In the future, patients may be treated accord-
Myelosuppression ing to their personal risk for relapse and outcome, as first studies
Myelosuppression is frequently observed in patients with GPA who demonstrated that the transcription signature of CD8+ T-cells may
have been treated with CYC (Bradley et al. 1989). In most cases neu- help to identify patients at risk for relapse (McKinney et al. 2010).
tropenia is modest and resolves after a couple of days. Sometimes
neutropenia can be severe and may last for a week or even longer, References
and involved patients are at particularly high risk for developing Adu, D., Pall, A., Luqmani, R.A., Richards, N.T., Howie, A.J., Emery, P.,
opportunistic infections (Bradley et al. 1989). In patients with GPA Michael, J., Savage, C.O., and Bacon, P.A. (1997). Controlled trial of
and agranulocytosis due to CYC therapy lasting more than 3 days, pulse versus continuous prednisolone and cyclophosphamide in the
the administration of recombinant human granulocyte-colony treatment of systemic vasculitis. QJM, 90, 401–9.
stimulating factor (rhG-CSF) was shown to induce a significantly Birck, R., Warnatz, K., Lorenz, H.M., Choi, M., Haubitz, M., Grunke, M.,
Peter, H.H., Kalden, J.R., Gobel, U., Drexler, J.M., Hotta, O., Nowack, R.,
faster rise in neutrophil counts compared to historic controls with a
and Van Der Woude, F.J. (2003). 15-Deoxyspergualin in patients with
similar degree of neutropenia (Hellmich et al. 1999a). In addition, refractory ANCA-associated systemic vasculitis: a six-month open-label
the application of rhG-CSF was associated with a lower incidence trial to evaluate safety and efficacy. Journal of the American Society of
of infections (Hellmich et al. 1999a). Nephrology, 14, 440–7.
A particular concern of administering recombinant cytokines Booth, A., Harper, L., Hammad, T., Bacon, P., Griffith, M., Levy, J., Savage, C.,
to patients with autoimmune diseases is the potential of these Pusey, C., and Jayne, D. (2004). Prospective study of TNFalpha blockade
cytokines to trigger the activity of the underlying inflammatory with infliximab in anti-neutrophil cytoplasmic antibody-associated sys-
disease. Clinical observations suggest that the administration of temic vasculitis. Journal of the American Society of Nephrology, 15, 717–21.
rhG-CSF to patients with CYC-induced agranulocytosis does not Booth, A.D., Jefferson, H.J., Ayliffe, W., Andrews, P.A., and Jayne, D.R. (2002).
Safety and efficacy of TNFalpha blockade in relapsing vasculitis. Annals
cause flares of GPA unless rhG-CSF is not stopped when neutrophil
of the Rheumatic Diseases, 61, 559.
counts are at 1000/µl or above (Hellmich et al. 1999a). Nevertheless, Bradley, J.D., Brandt, K.D., and Katz, B.P. (1989). Infectious complications of
flares of other autoimmune disease such as Felty’s syndrome were cyclophosphamide treatment for vasculitis. Arthritis and Rheumatism,
observed when overshooting neutrophilia developed as a result of 32, 45–53.
growth factor treatment (Hellmich et al. 1999b; Starkebaum 1997). Cartin-Ceba, R., Golbin, J.M., Keogh, K.A., et  al. (2012). Rituximab for
Thus, a routine administration of rhG-CSF is not recommended remission induction and maintenance in refractory granulomatosis with
but may be used in particular situations with agranulocytosis and poylangiitis (Wegener`s): ten-year experience at a single center. Arthritis
severe infection. In contrast to rhG-CSF, granulocyte-macrophage and Rheumatism, 64, 3770–8.
colony-stimulating factor (GM-CSF) has the potential to increase Deegens, J.K., Artz, M.A., Hoitsma, A.J., and Wetzels, J.F. (2003). Outcome of
renal transplantation in patients with pauci-immune small vessel vascu-
the surface expression of the ANCA target antigen PR3 on neutro-
litis or anti-GBM disease. Clinical Nephrology, 59, 1–9.
phils in vitro, and therefore theoretically bears an additional risk to de Groot, K., Adu, D., and Savage, C.O. (2001a). The value of pulse cyclo-
trigger GPA (Hellmich et al. 2000). phosphamide in ANCA-associated vasculitis: meta-analysis and critical
review. Nephrology Dialysis Transplantation, 16, 2018–27.
Summary de Groot, K., Gross, W.L., Herlyn, K., and Reinhold-Keller, E. (2001b).
Development and validation of a disease extent index for Wegener’s
Data from randomized controlled clinical trials suggest that CYC granulomatosis. Clinical Nephrology, 55, 31–8.
or RTX in combination with CS should be used for remission de Groot, K., Harper, L., Jayne, D.R., Flores Suarez, L.F., Gregorini, G., Gross,
induction in generalized disease. RTX may be preferred in relaps- W.L., Luqmani, R., Pusey, C.D., Rasmussen, N., Sinico, R.A., Tesar, V.,
ing patients, in young patients to preserve fertility, and in patients Vanhille, P., Westman, K., and Savage, C.O. (2009). Pulse versus daily
with contraindications to CYC or high cumulative CYC doses. In oral cyclophosphamide for induction of remission in antineutrophil
cytoplasmic antibody-associated vasculitis: a randomized trial. Annals
early systemic GPA without renal involvement, MTX can be used
of Internal Medicine, 150, 670–80.
instead of CYC, but maintenance of remission treatment and close
de Groot, K., Muhler, M., Reinhold-Keller, E., Paulsen, J., and Gross, W.L.
monitoring are mandatory. Efficacy for maintenance treatment in (1998). Induction of remission in Wegener’s granulomatosis with low
GPA is documented in randomized controlled trials for azathio- dose methotrexate. Journal of Rheumatology, 25, 492–5.
prine, methotrexate, leflunomide, and mycophenolate (Table 31.4). de Groot, K., Rasmussen, N., Bacon, P.A., Tervaert, J.W., Feighery, C.,
AZA and MTX should be considered as first-choice options for Gregorini, G., Gross, W.L., Luqmani, R., and Jayne, D.R. (2005).
CHAPTER 31 granulomatosis with polyangiitis (wegener’s granulomatosis) 429

Randomized trial of cyclophosphamide versus methotrexate for granulocyte colony-stimulating factor (G-CSF) induces plasma mem-
induction of remission in early systemic antineutrophil cytoplasmic brane expression of proteinase 3 (PR3). on neutrophils in vitro. Clinical
antibody-associated vasculitis. Arthritis and Rheumatism, 52, 2461–9. and Experimental Immunology, 120, 392–8.
de Menthon, M., Cohen, P., Pagnoux, C., Buchler, M., Sibilia, J., Detree, F., Hellmich, B., Flossmann, O., Gross, W.L., Bacon, P., Cohen-Tervaert, J.W.,
Gayraud, M., Khellaf, M., Penalba, C., Legallicier, B., Mouthon, L., and Guillevin, L., Jayne, D., Mahr, A., Merkel, P.A., Raspe, H., Scott, D.G.,
Guillevin, L. (2011). Infliximab or rituximab for refractory Wegener’s Witter, J., Yazici, H., and Luqmani, R.A. (2007). EULAR recommenda-
granulomatosis: long-term follow up. A prospective randomised mul- tions for conducting clinical studies and/or clinical trials in systemic
ticentre study on 17 patients. Clinical and Experimental Rheumatology, vasculitis: focus on anti-neutrophil cytoplasm antibody-associated vas-
29, S63–71. culitis. Annals of the Rheumatic Diseases, 66, 605–17.
DeRemee, R.A., McDonald, T.J., and Weiland, L.H. (1985). Wegener’s granu- Hellmich, B., Kausch, I., Doehn, C., Jocham, D., Holl-Ulrich, K., and Gross,
lomatosis: observations on treatment with antimicrobial agents. Mayo W.L. (2004). Urinary bladder cancer in Wegener’s granulomatosis: is it
Clinic Proceedings, 60, 27–32. more than cyclophosphamide? Annals of the Rheumatic Diseases, 63,
Elmedhem, A., Adu, D., and Savage, C.O. (2003). Relapse rate and out- 1183–5.
come of ANCA-associated small vessel vasculitis after transplantation. Hellmich, B., Schnabel, A., and Gross, W.L. (1999a). Granulocyte
Nephrology Dialysis Transplantation, 18, 1001–4. colony-stimulating factor treatment for cyclophosphamide-induced
Fauci, A.S., Haynes, B.F., Katz, P., and Wolff, S.M. (1983). Wegener’s granulo- severe neutropenia in Wegener’s granulomatosis. Arthritis and
matosis: prospective clinical and therapeutic experience with 85 patients Rheumatism, 42, 1752–6.
for 21 years. Annals of Internal Medicine, 98, 76–85. Hellmich, B., Schnabel, A., and Gross, W.L. (1999b). Treatment of severe
Fauci, A.S. and Wolff, S.M. (1973). Wegener’s granulomatosis:  studies in neutropenia due to Felty’s syndrome or systemic lupus erythematosus
eighteen patients and a review of the literature. Medicine, 52, 535–61. with granulocyte colony-stimulating factor. Seminars in Arthritis and
Faurschou, M., Westman, K., Rasmussen, N., de Groot, K., Flossmann, O., Rheumatism, 29, 82–99.
Hoglund, P., and Jayne, D.R. (2012). Brief report: long-term outcome Hernandez-Rodriguez, J., Hoffman, G.S., and Koening, C.L. (2010). Surgical
of a randomized clinical trial comparing methotrexate to cyclophos- interventions and local therapy for Wegener’s granulomatosis. Current
phamide for remission induction in early systemic antineutrophil cyto- Opinion in Rheumatology, 22, 29–36.
plasmic antibody-associated vasculitis. Arthritis and Rheumatism, 64, Hiemstra, T.F., Walsh, M., Mahr, A., Savage, C.O., de Groot, K., Harper,
3472–7. L., Hauser, T., Neumann, I., Tesar, V., Wissing, K.M., Pagnoux, C.,
Flossmann, O., Baslund, B., Bruchfeld, A., Tervaert, J.W., Hall, C., Heinzel, Schmitt, W., and Jayne, D.R. (2010). Mycophenolate mofetil vs aza-
P., Hellmich, B., Luqmani, R.A., Nemoto, K., Tesar, V., and Jayne, D.R. thioprine for remission maintenance in antineutrophil cytoplasmic
(2009). Deoxyspergualin in relapsing and refractory Wegener’s granulo- antibody-associated vasculitis:  a randomized controlled trial. JAMA,
matosis. Annals of the Rheumatic Diseases, 68, 1125–30. 304, 2381–8.
Geetha, D., Eirin, A., True, K., Valentina Irazabal, M., Specks, U., Seo, P., Hoffman, G.S., Kerr, G.S., Leavitt, R.Y., Hallahan, C.W., Lebovics, R.S., Travis,
Nachman, P., and Fervenza, F.C. (2011). Renal transplantation in anti- W.D., Rottem, M., and Fauci, A.S. (1992). Wegener granulomatosis: an
neutrophil cytoplasmic antibody-associated vasculitis:  a multicenter analysis of 158 patients. Annals of Internal Medicine, 116, 488–98.
experience. Transplantation, 91, 1370–5. Hoffman, G.S., Thomas-Golbanov, C.K., Chan, J., Akst, L.M., and Eliachar,
Gera, M., Griffin, M.D., Specks, U., Leung, N., Stegall, M.D., and Fervenza, I. (2003). Treatment of subglottic stenosis, due to Wegener’s granu-
F.C. (2007). Recurrence of ANCA-associated vasculitis following renal lomatosis, with intralesional corticosteroids and dilation. Journal of
transplantation in the modern era of immunosupression. Kidney Rheumatology, 30, 1017–21.
International, 71, 1296–1301. Hollander, D. and Manning, R.T. (1967). The use of alkylating agents in the treat-
Guerry, M.J., Brogan, P., Bruce, I.N., D’Cruz, D.P., Harper, L., Luqmani, ment of Wegener’s granulomatosis. Annals of Internal Medicine, 67, 393–8.
R., Pusey, C.D., Salama, A.D., Scott, D.G., Savage, C.O., Watts, R.A., Holle, J.U., Dubrau, C., Herlyn, K., Heller, M., Ambrosch, P., Noelle, B.,
and Jayne, D.R. (2012). Recommendations for the use of rituximab in Reinhold-Keller, E., and Gross, W.L. (2012). Rituximab for refractory
anti-neutrophil cytoplasm antibody-associated vasculitis. Rheumatology granulomatosis with polyangiitis (Wegener’s granulomatosis): compari-
(Oxford), 51, 634–43. son of efficacy in granulomatous versus vasculitic manifestations. Annals
Guillevin, L., Cordier, J.F., Lhote, F., Cohen, P., Jarrousse, B., Royer, I., Lesavre, of the Rheumatic Diseases, 71, 327–33.
P., Jacquot, C., Bindi, P., Bielefeld, P., Desson, J.F., Detree, F., Dubois, A., Holle, J.U., Gross, W.L., Holl-Ulrich, K., Ambrosch, P., Noelle, B., Both, M.,
Hachulla, E., Hoen, B., Jacomy, D., Seigneuric, C., Lauque, D., Stern, M., Csernok, E., Moosig, F., Schinke, S., and Reinhold-Keller, E. (2010).
and Longy-Boursier, M. (1997). A prospective, multicenter, randomized Prospective long-term follow-up of patients with localised Wegener’s
trial comparing steroids and pulse cyclophosphamide versus steroids granulomatosis: does it occur as persistent disease stage? Annals of the
and oral cyclophosphamide in the treatment of generalized Wegener’s Rheumatic Diseases, 69, 1934–9.
granulomatosis. Arthritis and Rheumatism, 40, 2187–98. Holle, J.U., Gross, W.L., Latza, U., Nolle, B., Ambrosch, P., Heller, M.,
Harper, L., Morgan, M.D., Walsh, M., Hoglund, P., Westman, K., Flossmann, Fertmann, R., and Reinhold-Keller, E. (2011). Improved outcome in 445
O., Tesar, V., Vanhille, P., Groot, K.D., Luqmani, R., Flores-Suarez, patients with Wegener’s granulomatosis in a German vasculitis center
L.F., Watts, R., Pusey, C., Bruchfeld, A., Rasmussen, N., Blockmans, over four decades. Arthritis and Rheumatism, 63, 257–66.
D., Savage, C.O., and Jayne, D. (2012). Pulse versus daily oral cyclo- Jayne, D., Rasmussen, N., Andrassy, K., Bacon, P., Tervaert, J.W., Dadoniene,
phosphamide for induction of remission in ANCA-associated vasculi- J., Ekstrand, A., Gaskin, G., Gregorini, G., de Groot, K., Gross, W.,
tis: long-term follow-up. Annals of the Rheumatic Diseases, 71, 955–60. Hagen, E.C., Mirapeix, E., Pettersson, E., Siegert, C., Sinico, A., Tesar,
Haubitz, M. and de Groot, K. (2002). Tolerance of mycophenolate mofetil V., Westman, K., and Pusey, C. (2003). A randomized trial of mainte-
in end-stage renal disease patients with ANCA-associated vasculitis. nance therapy for vasculitis associated with antineutrophil cytoplasmic
Clinical Nephrology, 57, 421–4. autoantibodies. New England Journal of Medicine, 349, 36–44.
Haubitz, M., Schellong, S., Gobel, U., Schurek, H.J., Schaumann, D., Koch, Jayne, D.R., Chapel, H., Adu, D., Misbah, S., O’Donoghue, D., Scott,
K.M., and Brunkhorst, R. (1998). Intravenous pulse administration of D., and Lockwood, C.M. (2000). Intravenous immunoglobulin for
cyclophosphamide versus daily oral treatment in patients with antineutro- ANCA-associated systemic vasculitis with persistent disease activity.
phil cytoplasmic antibody-associated vasculitis and renal involvement: a QJM, 93, 433–9.
prospective, randomized study. Arthritis and Rheumatism, 41, 1835–44. Jayne, D.R., Davies, M.J., Fox, C.J., Black, C.M., and Lockwood, C.M. (1991).
Hellmich, B., Csernok, E., Trabandt, A., Gross, W.L., and Ernst, M. (2000). Treatment of systemic vasculitis with pooled intravenous immunoglob-
Granulocyte-macrophage colony-stimulating factor (GM-CSF) but not ulin. Lancet, 337, 1137–9.
430 SECTION 5 vasculitic diseases and syndromes and related disorders

Jayne, D.R., Gaskin, G., Rasmussen, N., Abramowicz, D., Ferrario, F., regimen for the treatment of Wegener’s granulomatosis:  extended
Guillevin, L., Mirapeix, E., Savage, C.O., Sinico, R.A., Stegeman, C.A., follow-up and rate of relapse. American Journal of Medicine, 114, 463–9.
Westman, K.W., van der Woude, F.J., de Lind van Wijngaarden, R.A., Langford, C.A., Talar-Williams, C., and Sneller, M.C. (2004). Mycophenolate
and Pusey, C.D. (2007). Randomized trial of plasma exchange or mofetil for remission maintenance in the treatment of Wegener’s granu-
high-dosage methylprednisolone as adjunctive therapy for severe renal lomatosis. Arthritis and Rheumatism, 51, 278–83.
vasculitis. Journal of the American Society of Nephrology, 18, 2180–8. Lapraik, C., Watts, R., Bacon, P., Carruthers, D., Chakravarty, K., D’Cruz, D.,
Jennette, J.C., Falk, R.J., Andrassy, K., Bacon, P.A., Churg, J., Gross, W.L., Guillevin, L., Harper, L., Jayne, D., Luqmani, R., Mooney, J., and Scott,
Hagen, E.C., Hoffman, G.S., Hunder, G.G., Kallenberg, C.G., et  al. D. (2007). BSR and BHPR guidelines for the management of adults with
(1994). Nomenclature of systemic vasculitides. Proposal of an interna- ANCA associated vasculitis. Rheumatology (Oxford), 46, 1615–6.
tional consensus conference. Arthritis and Rheumatism, 37, 187–192. Leavitt, R.Y., Fauci, A.S., Bloch, D.A., Michel, B.A., Hunder, G.G., Arend,
Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F., W.P., Calabrese, L.H., Fries, J.F., Lie, J.T., Lightfoot, R.W. Jr, et al. (1990).
Flores-Suarez, L.F., Gross, W.L., Guillevin, L., Hagen, E.C., Hoffman, The American College of Rheumatology 1990 criteria for the classifi-
G.S., Jayne, D.R., Kallenberg, C.G.M., Lamprecht, P., Langford, C.A., cation of Wegener’s granulomatosis. Arthritis and Rheumatism, 33,
Luqmani, R.A., Mahr, A.D., Matteson, E.L., Merkel, P.A., Ozen, S., 1101–7.
Pusey, C.D., Rasmussen, N., Rees, A.J., Scott, D.G.I., Specks, U., Stone, Levy, Y., Sherer, Y., George, J., Langevitz, P., Ahmed, A., Bar-Dayan, Y.,
J.H., Takahashi, K., and Watts, R.A. (2013). 2012 Revised International Fabbrizzi, F., Terryberry, J., Peter, J., and Shoenfeld, Y. (1999). Serologic
Chapel Hill Consensus Conference Nomenclature of Vasculitides. and clinical response to treatment of systemic vasculitis and associated
Arthritis and Rheumatism, 65, 1–11. autoimmune disease with intravenous immunoglobulin. International
Jones, R.B., Ferraro, A.J., Chaudhry, A.N., Brogan, P., Salama, A.D., Archives of Allergy and Immunology, 119, 231–8.
Smith, K.G., Savage, C.O., and Jayne, D.R. (2009). A multicenter sur- Luqmani, R.A., Bacon, P.A., Moots, R.J., Janssen, B.A., Pall, A., Emery, P.,
vey of rituximab therapy for refractory antineutrophil cytoplasmic Savage, C., and Adu, D. (1994). Birmingham Vasculitis Activity Score
antibody-associated vasculitis. Arthritis and Rheumatism, 60, 2156–68. (BVAS) in systemic necrotizing vasculitis. QJM, 87, 671–8.
Jones, R.B., Tervaert, J.W., Hauser, T., Luqmani, R., Morgan, M.D., Peh, C.A., Martinez, V., Cohen, P., Pagnoux, C., Vinzio, S., Mahr, A., Mouthon,
Savage C.O., Segelmark, M., Tesar, V., van Paassen, P., Walsh, D., Walsh, L., Sailler, L., Delaunay, C., Sadoun, A., and Guillevin, L. (2008).
M., Westman, K., and Jayne, D.R. (2010). Rituximab versus cyclophos- Intravenous immunoglobulins for relapses of systemic vasculitides asso-
phamide in ANCA-associated renal vasculitis. New England Journal of ciated with antineutrophil cytoplasmic autoantibodies: results of a mul-
Medicine, 363, 211–20. ticenter, prospective, open-label study of twenty-two patients. Arthritis
Josselin, L., Mahr, A., Cohen, P., Pagnoux, C., Guaydier-Souquieres, G., and Rheumatism, 58, 308–17.
Hayem, G., Job-Deslandre, C., Liferman, F., Pourrat, J., and Guillevin, McKinney, E.F., Lyons, P.A., Carr, E.J., Hollis, J.L., Jayne, D.R., Willcocks,
L. (2008). Infliximab efficacy and safety against refractory systemic L.C., Koukoulaki, M., Brazma, A., Jovanovic, V., Kemeny, D.M., Pollard,
necrotising vasculitides: long-term follow-up of 15 patients. Annals of A.J., Macary, P.A., Chaudhry, A.N., and Smith, K.G. (2010). A CD8+ T
the Rheumatic Diseases, 67, 1343–6. cell transcription signature predicts prognosis in autoimmune disease.
Joy, M.S., Hogan, S.L., Jennette, J.C., Falk, R.J., and Nachman, P.H. (2005). Nature Medicine, 16, 586–91, 1 p. following 591.
A pilot study using mycophenolate mofetil in relapsing or resistant Merkel, P.A., Aydin, S.Z., Boers, M., Direskeneli, H., Herlyn, K., Seo, P.,
ANCA small vessel vasculitis. Nephrology Dialysis Transplantation, 20, Suppiah, R., Tomasson, G., and Luqmani, R.A. (2011). The OMERACT
2725–32. core set of outcome measures for use in clinical trials of ANCA-associated
Klemmer, P.J., Chalermskulrat, W., Reif, M.S., Hogan, S.L., Henke, D.C., vasculitis. Journal of Rheumatology, 38, 1480–6.
and Falk, R.J. (2003). Plasmapheresis therapy for diffuse alveolar hem- Metzler, C., Fink, C., Lamprecht, P., Gross, W.L., and Reinhold-Keller, E.
orrhage in patients with small-vessel vasculitis. American Journal of (2004). Maintenance of remission with leflunomide in Wegener’s granu-
Kidney Diseases, 42, 1149–53. lomatosis. Rheumatology (Oxford), 43, 315–20.
Knight, A., Askling, J., and Ekbom, A. (2002). Cancer incidence in a Metzler, C., Miehle, N., Manger, K., Iking-Konert, C., de Groot, K., Hellmich,
population-based cohort of patients with Wegener’s granulomatosis. B., Gross, W.L., Reinhold-Keller, E.; German Network of Rheumatic
International Journal of Cancer, 100, 82–5. Diseases (2007). Elevated relapse rate under oral methotrexate versus
Knight, A., Askling, J., Granath, F., Sparen, P., and Ekbom, A. (2004). Urinary leflunomide for maintenance of remission in Wegener’s granulomatosis.
bladder cancer in Wegener’s granulomatosis: risks and relation to cyclo- Rheumatology (Oxford), 46, 1087–91.
phosphamide. Annals of the Rheumatic Diseases, 63, 1307–11. Mukhtyar, C., Guillevin, L., Cid, M.C., Dasgupta, B., de Groot, K., Gross,
Koukoulaki, M. and Jayne, D.R. (2006). Mycophenolate mofetil in W., Hauser, T., Hellmich, B., Jayne, D., Kallenberg, C.G., Merkel, P.A.,
anti-neutrophil cytoplasm antibodies-associated systemic vasculitis. Raspe, H., Salvarani, C., Scott, D.G., Stegeman, C., Watts, R., Westman,
Nephron Clinical Practice, 102, c100–107. K., Witter, J., Yazici, H., and Luqmani, R. (2009a). EULAR recommenda-
Lamprecht, P., Voswinkel, J., Lilienthal, T., Nolle, B., Heller, M., Gross, W.L., tions for the management of primary small and medium vessel vasculi-
and Gause, A. (2002). Effectiveness of TNF-alpha blockade with inflixi- tis. Annals of the Rheumatic Diseases, 68, 310–7.
mab in refractory Wegener’s granulomatosis. Rheumatology (Oxford), Mukhtyar, C., Lee, R., Brown, D., Carruthers, D., Dasgupta, B., Dubey, S.,
41, 1303–7. Flossmann, O., Hall, C., Hollywood, J., Jayne, D., Jones, R., Lanyon, P.,
Langford, C.A., Sneller, M.C., Hallahan, C.W., Hoffman, G.S., Kammerer, Muir, A., Scott, D., Young, L., and Luqmani, R.A. (2009b). Modification
W.A., Talar-Williams, C., Fauci, A.S., and Lebovics, R.S. (1996). Clinical and validation of the Birmingham Vasculitis Activity Score (version 3).
features and therapeutic management of subglottic stenosis in patients Annals of the Rheumatic Diseases, 68, 1827–32.
with Wegener’s granulomatosis. Arthritis and Rheumatism, 39, 1754–60. Nowack, R., Gobel, U., Klooker, P., Hergesell, O., Andrassy, K., and van der
Langford, C.A., Talar-Williams, C., Barron, K.S., and Sneller, M.C. (1999). A Woude, F.J. (1999). Mycophenolate mofetil for maintenance therapy of
staged approach to the treatment of Wegener’s granulomatosis: induc- Wegener’s granulomatosis and microscopic polyangiitis: a pilot study in
tion of remission with glucocorticoids and daily cyclophosphamide 11 patients with renal involvement. Journal of the American Society of
switching to methotrexate for remission maintenance. Arthritis and Nephrology, 10, 1965–71.
Rheumatism, 42, 2666–73. Ognibene, F.P., Shelhamer, J.H., Hoffman, G.S., Kerr, G.S., Reda, D., Fauci,
Langford, C.A., Talar-Williams, C., Barron, K.S., and Sneller, M.C. (2003). A.S., and Leavitt, R.Y. (1995). Pneumocystis carinii pneumonia: a major
Use of a cyclophosphamide-induction methotrexate-maintenance complication of immunosuppressive therapy in patients with Wegener’s
CHAPTER 31 granulomatosis with polyangiitis (wegener’s granulomatosis) 431

granulomatosis. American Journal of Respiratory and Critical Care granulomatosis patients treated with methotrexate and prednisone.
Medicine, 151, 795–9. Arthritis and Rheumatism, 38, 608–13.
Pagnoux, C., Mahr, A., Hamidou, M.A., Boffa, J.J., Ruivard, M., Ducroix, J.P., Sowden, E. and Carmichael, A.J. (2004). Autoimmune inflammatory disor-
Kyndt, X., Lifermann, F., Papo, T., Lambert, M., Le Noach, J., Khellaf, ders, systemic corticosteroids and pneumocystis pneumonia: a strategy
M., Merrien, D., Puechal, X., Vinzio, S., Cohen, P., Mouthon, L., Cordier, for prevention. BMC Infectious Diseases, 4, 42.
J.F., and Guillevin, L. (2008). Azathioprine or methotrexate maintenance Specks, U., Merkel, P.A., Seo, P., et al. (2013). Efficacy of remission-induction
for ANCA-associated vasculitis. New England Journal of Medicine, 359, regimens for ANCA-associated vasculitis. New England Journal of
2790–803. Medicine, 369, 417–27.
Reinhold-Keller, E., Beuge, N., Latza, U., de Groot, K., Rudert, H., Nolle, B., Starkebaum, G. (1997). Use of colony-stimulating factors in the treatment of
Heller, M., and Gross, W.L. (2000). An interdisciplinary approach to the neutropenia associated with collagen vascular disease. Current Opinion
care of patients with Wegener’s granulomatosis: long-term outcome in in Hematology, 4, 196–9.
155 patients. Arthritis and Rheumatism, 43, 1021–32. Stassen, P.M., Tervaert, J.W., and Stegeman, C.A. (2007). Induction of remis-
Reinhold-Keller, E., De Groot, K., Rudert, H., Nolle, B., Heller, M., and Gross, sion in active anti-neutrophil cytoplasmic antibody-associated vasculi-
W.L. (1996). Response to trimethoprim/sulfamethoxazole in Wegener’s tis with mycophenolate mofetil in patients who cannot be treated with
granulomatosis depends on the phase of disease. QJM, 89, 15–23. cyclophosphamide. Annals of the Rheumatic Diseases, 66, 798–802.
Reinhold-Keller, E., Fink, C.O., Herlyn, K., Gross, W.L., and De Groot, K. Stegeman, C.A., Tervaert, J.W., de Jong, P.E., and Kallenberg, C.G. (1996).
(2002). High rate of renal relapse in 71 patients with Wegener’s granu- Trimethoprim-sulfamethoxazole (co-trimoxazole) for the prevention of
lomatosis under maintenance of remission with low-dose methotrexate. relapses of Wegener’s granulomatosis. Dutch Co-Trimoxazole Wegener
Arthritis and Rheumatism, 47, 326–32. Study Group. New England Journal of Medicine, 335, 16–20.
Rhee, E.P., Laliberte, K.A., and Niles, J.L. (2010). Rituximab as maintenance Stone, J.H., Hoffman, G.S., Merkel, P.A., Min, Y.I., Uhlfelder, M.L., Hellmann,
therapy for anti-neutrophil cytoplasmic antibody-associated vasculitis. D.B., Specks, U., Allen, N.B., Davis, J.C., Spiera, R.F., Calabrese, L.H.,
Clinical Journal of the American Society of Nephrology, 5, 1394–400. Wigley, F.M., Maiden, N., Valente, R.M., Niles, J.L., Fye, K.H., McCune,
Richter, C., Schnabel, A., Csernok, E., De Groot, K., Reinhold-Keller, E., and J.W., St Clair, E.W., and Luqmani, R.A. (2001). A disease-specific activity
Gross, W.L. (1995). Treatment of anti-neutrophil cytoplasmic antibody index for Wegener’s granulomatosis: modification of the Birmingham
(ANCA)-associated systemic vasculitis with high-dose intravenous Vasculitis Activity Score. International Network for the Study of the
immunoglobulin. Clinical and Experimental Immunology, 101, 2–7. Systemic Vasculitides (INSSYS). Arthritis and Rheumatism, 44, 912–20.
Rutgers, A. and Kallenberg, C.G. (2012). Refractory disease in antineutro- Stone, J.H., Merkel, P.A., Spiera, R., Seo, P., Langford, C.A., Hoffman, G.S.,
phil cytoplasmic antibodies associated vasculitis. Current Opinion in Kallenberg, C.G., St Clair, E.W., Turkiewicz, A., Tchao, N.K., Webber,
Rheumatology, 24, 245–51. L., Ding, L., Sejismundo, L.P., Mieras, K., Weitzenkamp, D., Ikle, D.,
Schmitt, W.H., Birck, R., Heinzel, P.A., Gobel, U., Choi, M., Warnatz, K., Peter, Seyfert-Margolis, V., Mueller, M., Brunetta, P., Allen, N.B., Fervenza,
H.H., and van der Woude, F.J. (2005). Prolonged treatment of refractory F.C., Geetha, D., Keogh, K.A., Kissin, E.Y., Monach, P.A., Peikert, T.,
Wegener’s granulomatosis with 15-deoxyspergualin: an open study in Stegeman, C., Ytterberg, S.R., and Specks, U. (2010). Rituximab ver-
seven patients. Nephrology Dialysis Transplantation, 20, 1083–92. sus cyclophosphamide for ANCA-associated vasculitis. New England
Schmitt, W.H., Hagen, E.C., Neumann, I., Nowack, R., Flores-Suarez, L.F., Journal of Medicine, 363, 221–32.
and van der Woude, F.J. (2004). Treatment of refractory Wegener’s Talar-Williams, C., Hijazi, Y.M., Walther, M.M., Linehan, W.M., Hallahan,
granulomatosis with antithymocyte globulin (ATG): an open study in C.W., Lubensky, I., Kerr, G.S., Hoffman, G.S., Fauci, A.S., and Sneller,
15 patients. Kidney International, 65, 1440–8. M.C. (1996). Cyclophosphamide-induced cystitis and bladder cancer in
Schmitt, W.H., Haubitz, M., Mistry, N., Brunkhorst, R., Erbsloh-Moller, B., patients with Wegener granulomatosis. Annals of Internal Medicine, 124,
and Gross, W.L. (1993). Renal transplantation in Wegener’s granuloma- 477–84.
tosis. Lancet, 342, 860. Tatsis, E., Reinhold-Keller, E., Steindorf, K., Feller, A.C., and Gross, W.L.
Seror, R., Pagnoux, C., Ruivard, M., Landru, I., Wahl, D., Riviere, S., Aussant, (1999). Wegener’s granulomatosis associated with renal cell carcinoma.
S., Mahr, A., Cohen, P., Mouthon, L., and Guillevin, L. (2010). Treatment Arthritis and Rheumatism, 42, 751–6.
strategies and outcome of induction-refractory Wegener’s granuloma- Terrier B, Pagnoux C, Karras A, Khoutra C, Aumaitre O, Cohen P, Maurier
tosis or microscopic polyangiitis: analysis of 32 patients with first-line F, Decaux O, Demurs-Clavel H, Gobert P, Quemeneur T, Godmer
induction-refractory disease in the WEGENT trial. Annals of the P, Puechal X, Mouthon L, Guillevin L. (2013). Rituximab versus
Rheumatic Diseases, 69, 2125–30. Azathioprine for maintenance in Anineutrophil cytoplasmic antibod-
Shekelle PG, Woolf SH, Eccles M, Grimshaw J (1999). Clinical guidelines: ies (ANCA)-associated vasculitis (MAINRITSAN): Follow up at 34
developing guidelines. BMJ 318, 593–6. months. Ann Rheum Dis, 72 (Suppl 3): 124.
Shen, J., Gill, J., Shangguan, M., Sampaio, M.S., and Bunnapradist, S. (2011). Tuso, P., Moudgil, A., Hay, J., Goodman, D., Kamil, E., Koyyana, R.,
Outcomes of renal transplantation in recipients with Wegener’s granulo- and Jordan, S.C. (1992). Treatment of antineutrophil cytoplasmic
matosis. Clinical Transplantation, 25, 380–7. autoantibody-positive systemic vasculitis and glomerulonephritis
Silva, F., Specks, U., Kalra, S., Hogan, M.C., Leung, N., Sethi, S., and Fervenza, with pooled intravenous gammaglobulin. American Journal of Kidney
F.C. (2010). Mycophenolate mofetil for induction and maintenance of Diseases, 20, 504–8.
remission in microscopic polyangiitis with mild to moderate renal Walsh, M., Catapano, F., Szpirt, W., Thorlund, K., Bruchfeld, A., Guillevin,
involvement—a prospective, open-label pilot trial. Clinical Journal of the L., Haubitz, M., Merkel, P.A., Peh, C.A., Pusey, C., and Jayne, D. (2011).
American Society of Nephrology, 5, 445–53. Plasma exchange for renal vasculitis and idiopathic rapidly progres-
Smith, R., Jones, R.B., Guerry, M.J., et al. (2012). Rituximab for remission sive glomerulonephritis:  a meta-analysis. American Journal of Kidney
maintenance in relapsing antineutrophil cytoplasmic antibody associ- Diseases, 57, 566–74.
ated vasculitis. Arthritis and Rheumatism, 64, 3760–9. Walsh, M., Casian, A., Flossmann, O., et  al. (2013). Long-term follow-up
Sneller, M.C. (1998). Evaluation, treatment, and prophylaxis of infections of patients with severe ANCA-associated vasculitis comparing plasma
complicating systemic vasculitis. Current Opinion in Rheumatology, exchange to intravenous methylprednisolone treatment is unclear.
10, 38–44. Kidney International, 84, 397–402.
Sneller, M.C., Hoffman, G.S., Talar-Williams, C., Kerr, G.S., Hallahan, Walton, E.W. (1958). Giant-cell granuloma of the respiratory tract (Wegener’s
C.W., and Fauci, A.S. (1995). An analysis of forty-two Wegener’s granulomatosis). British Medical Journal, 2, 265–70.
432 SECTION 5 vasculitic diseases and syndromes and related disorders

Ward, M.M. and Donald, F. (1999). Pneumocystis carinii pneumonia in Beitrage zur Pathologischen Anatomie und zur Allgemeinen Pathologie,
patients with connective tissue diseases: the role of hospital experience 102, 36–68.
in diagnosis and mortality. Arthritis and Rheumatism, 42, 780–9. Wegener's Granulomatosis Etanercept Trial (WGET) Research Group
Wegener, F. (1939). Über eine eigenartige rhinogene Granulomatose (2005). Etanercept plus standard therapy for Wegener’s granulomatosis.
mit besonderer Beteiligung des Arteriensystems und der Nieren. New England Journal Of Medicine, 352, 351–61.
CHAPTER 32

Eosinophilic granulomatosis
with polyangiitis (Churg–
Strauss syndrome)
Jeremy M. Clain and Ulrich Specks

Original description, disease clinical and histopathological features:  a patient fulfils the defi-
nition if asthma, peripheral eosinophilia, eosinophil-rich and
definitions, and classification necrotizing granulomatous inflammation involving the respiratory
Eosinophilic granulomatosis with polyangiitis (EGPA, for- tract, and necrotizing vasculitis affecting small to medium-sized
merly Churg–Strauss syndrome) was first described as ‘allergic vessels are found; radiographic or clinical surrogates for vasculitis
granulomatosis and angiitis’ in patients with asthma who had are accepted (Jennette et al. 2013). Finally, the American College of
three pathological features:  necrotizing vasculitis, eosinophilic Rheumatology criteria were designed to differentiate patients with
inflammation, and extravascular granulomas (Churg and Strauss EGPA from those with other forms of vasculitis; a patient with vas-
1951). It is now recognized as a small-vessel vasculitis, which culitis is said to have EGPA if four of the following six features are
preferentially affects the capillaries, arterioles, and venules, even present:  asthma, peripheral eosinophilia, peripheral neuropathy
though the disease process occasionally extends to larger ves- attributed to vasculitis, transient pulmonary infiltrates, paranasal
sels (Jennette et al. 2013). Because of the frequent occurrence of sinus disease, and a biopsy containing a blood vessel with extravas-
antineutrophil cytoplasmic antibodies (ANCA) and clinical and cular eosinophils (Masi et  al. 1990). Even though these three
histopathological similarities, EGPA has been classified as one schemes are not meant to serve as stand-alone diagnostic criteria,
of the three ANCA-associated vasculitides, along with granulo- they are frequently used in that capacity. None of the three schemes
matosis with polyangiitis (GPA, formerly Wegener’s granuloma- captures all patients (Keogh and Specks 2003).
tosis) and microscopic polyangiitis (MPA) (Tervaert et al. 1991; Furthermore, diagnosing EGPA is complicated because the
Tervaert and Kallenberg 1993; Guillevin et  al. 1993; Reid et  al. various clinical manifestations of the syndrome rarely coin-
1998; Guillevin et al. 1999; Solans et al. 2001; Keogh and Specks cide. Lanham and colleagues distinguished three clinical phases
2003; Jennette et al. 2013). While previously referred to as Churg– (Lanham et al. 1984). The first, prodromal phase consists of asthma,
Strauss syndrome (CSS), the 2012 Revised International Chapel
Hill Consensus Conference on the Nomenclature of Vasculitides
changed the name to reflect the prominence of eosinophilia as
an essential feature of the syndrome and to achieve nomenclature
continuity across the spectrum of the ANCA-associated vascu-
litides (Jennette et al. 2013).
The original description of the disease was based on autopsy
observations. All three pathological features of the syndrome
defined by Churg and Strauss (necrotizing vasculitis, eosinophilic
inflammation, and extravascular granulomas (Figures  32.1, 32.2,
and 32.3)) (Churg and Strauss 1951) are rarely present in patients
at the time of original presentation.
Three schemes of clinical criteria and definitions have subse-
quently been proposed for the disease. Lanham’s criteria emphasize
the clinical presentation: a patient is said to have EGPA if asthma,
peripheral blood eosinophilia, and systemic vasculitis in two or
more organs are detected (Lanham et al. 1984). The Chapel Hill Fig. 32.1 Extravascular granuloma in the lung, also referred to as the Churg–
Consensus Conference definition is based on the combination of Strauss or allergic granuloma (courtesy of Dr. Jeffrey L. Myers).
434 SECTION 5 vasculitic diseases and syndromes and related disorders

seems to be common to all patients, and allergens have been pro-


posed as triggering factors. Direct tissue injury is attributed to toxic
eosinophil and neutrophil degranulation products. Activation of
T lymphocytes seems to be a significant driving force maintain-
ing the eosinophilic inflammation. Reports of successful treatment
of EGPA with B-lymphocyte depletion suggest that B lympho-
cytes may also play a yet unrecognized but important role in the
pathogenesis of the disease. Finally, there is evidence in support of
a pathogenic role of ANCA, which seem to modulate the disease
phenotype.
Genetic susceptibilities to EGPA have been suggested by
gene association studies investigating both HLA genes and
single-nucleotide polymorphisms (SNPs). In an Italian cohort, an
investigation of selected HLA foci found that the HLA-DRB1*07
allele and the related HLA-DRB4 gene confer increased suscep-
Fig. 32.2 Necrotizing vasculitis of a small pulmonary artery with prominent tibility to EGPA, while the HLA-DRB1*13 allele and the related
eosinophilic infiltration (courtesy of Dr. Jeffrey L. Myers).
HLA-DRB3 gene are protective (Vaglio et al. 2007). An independent
study of a German cohort subsequently found similar HLA associa-
tions (Wieczorek et al. 2008b). These findings, which associate HLA
class II genes with EGPA susceptibility, suggest the possibility that
antigens may be key mediators of the disease and that CD4+ T cells
may be involved in its pathogenesis (Vaglio et al. 2012). A separate
gene association study investigated the impact of polymorphisms
in the gene that encodes the precursor of interleukin-10 (IL-10)
(Wieczorek et  al. 2008a). IL-10 is a potent pleiotropic cytokine
produced by numerous immune cells, and its level of production
is largely genetically determined. By analysing the SNPs that tag
the promoter haplotypes of the IL10 gene, this study found that the
IL10.2 haplotype—which is correlated with increased IL-10 pro-
duction—was strongly associated with ANCA-negative EGPA. This
association raises the possibility of a pathogenic role for IL-10, and
further supports the hypothesis that genetic determinants contrib-
Fig. 32.3 Necrotizing vasculitis of a small extrapulmonary artery (courtesy
ute to the development of EGPA.
of Dr. Jeffrey L. Myers). Peripheral blood and tissue eosinophilia are hallmarks of EGPA.
These eosinophils are activated, and markers of eosinophil activa-
with or without allergic rhinitis and nasal polyposis. It may pre- tion parallel disease activity and predict relapses (Cottin et al. 1995;
cede other manifestations of the disease, particularly vasculitis, by Schmitt et al. 1998; Hurst et al. 2000; Kurosawa et al. 2000). Both
a number of years (Lanham et al. 1984). The second phase is char- eosinophil and neutrophil degranulation products are found in
acterized by peripheral blood eosinophilia and eosinophilic tissue areas of prominent inflammation and tissue necrosis in affected
infiltration. Eosinophilic manifestations may also remit and reoc- organs (Schnabel et al. 1999; Peen et al. 2000; Ramakrishna et al.
cur over a number of years. The third and most severe phase of 2000; Drage et  al. 2002). It has been suggested that the eosino-
Churg–Strauss syndrome consists of systemic vasculitis, and may philic inflammation and possibly the allergic background in EGPA
be life-threatening. These three phases do not always occur sequen- are the result of abnormally activated T lymphocytes. Markers of
tially, and in a minority of cases the onset of asthma may follow T-lymphocyte activation (soluble interleukin-2 receptor) are ele-
the onset of vasculitis (Chumbley et al. 1977; Keogh and Specks vated in patients with active EGPA and correlate with markers of
2003; Comarmond et al. 2013). Occasionally, eosinophilic vascu- endothelial cell damage (soluble thrombomodulin) (Schmitt et al.
litis and/or eosinophilic granulomatous inflammation is limited to 1998). T lymphocytes of patients with EGPA release predominantly
one organ in asthmatic patients who lack other signs of systemic Th-2-type cytokines, such as interleukin-4 (IL-4), interleukin-5
disease. Moreover, overt manifestations of vasculitis may be sup- (IL-5), and interleukin-13 (IL-13), and are thought to be responsi-
pressed by oral glucocorticoid therapy of asthma, only to emerge ble for the recruitment, activation, and delayed apoptosis of eosin-
when the glucocorticoids are withdrawn. These have been referred ophils (Schonermarck et al. 2000; Kiene et al. 2001). In addition,
to as ‘limited forms’ or as ‘formes frustes’ of EGPA (Lie 1993; Churg serum levels of the eosinophil-attracting chemokine eotaxin-3,
et al. 1995; Churg 2001). which is secreted by endothelial cells and eosinophils in EGPA
lesions, has been found to be markedly increased in patients with
active EGPA, and may promote disease-associated inflammation
Pathogenesis (Zwerina et al. 2011).
The pathogenesis of EGPA remains poorly understood. Genetic Abnormalities in the CD95/CD95 ligand system have also been
association studies have suggested certain genotypic suscepti- observed in EGPA (Muschen et  al. 1999). This mechanism may
bilities for development of the syndrome. An allergic background contribute to clonal expansion of autoaggressive T cells and delayed
CHAPTER 32 eosinophilic granulomatosis with polyangiitis (churg–strauss syndrome) 435

lymphocyte and eosinophil apoptosis (Muschen et  al. 1999), et al. 2000), possibly compounded by the original case reports of
though other evidence suggests that eosinophils in EGPA may LRA-associated EGPA syndrome themselves. The estimated annual
not truly have an intrinsic apoptotic defect, but may instead owe incidence of EGPA cases per million asthma patients (0–67) over-
their prolonged survival to the stimulating effects of IL-5 (Jakiela laps with the estimated incidence of about 60 cases per million
et al. 2009). It has been documented that clonally expanded T cells LRA users (Wechsler et  al. 2000; Loughlin et  al. 2002; Harrold
from EGPA patients exhibit preferential V-gene usage for a gene et  al. 2005). The clinical disease manifestations, including time
from the Vβ21 family, and that they display similar T-cell recep- from onset of asthma to onset of vasculitis symptoms, do not dif-
tor specificities. As with the finding of a restricted HLA repertoire, fer between EGPA patients exposed to these drugs and those never
this restriction in T-cell receptor variability suggests recognition of exposed (Keogh and Specks 2003). One-hundred and sixty-five
a limited number of shared antigens among the patients studied patients who developed EGPA while receiving LRA were analysed
(Muschen et al. 1999). These findings also support clinical observa- at a workshop sponsored by the National Institutes of Health and
tions, which have implicated inhaled antigen(s) as triggers of EGPA the US Food and Drug Administration, and in 88% of these cases
(Guillevin et al. 1991; Muschen et al. 1999; Lane et al. 2003). the onset of vasculitis was associated with a dose reduction of oral
Two types of ANCA are associated with small-vessel vasculi- glucocorticoids (Weller et al. 2001). Finally, there is little chemical
tis. Those reacting with proteinase 3 (PR3) predominate in GPA, similarity between the different agents in this class or other drugs
whereas ANCA reacting with myeloperoxidase (MPO) are more reported in association with EGPA, including acetaminophen,
commonly found in MPA and EGPA. The reported frequency of macrolides, carbamazepine, cocaine, and other asthma treatments
ANCA in active EGPA varies between 31 and 75% (Tervaert and used to reduce the use of oral glucocorticoids, such as inhaled glu-
Kallenberg 1993; Schmitt et al. 1998; Solans et al. 2001; Keogh and cocorticoids, cromolyn sodium, beta-agonists, and the anti-IgE
Specks 2003; Sinico et al. 2005; Comarmond et al. 2013), and the monoclonal antibody omalizumab (Sheffer et al. 1975; Churg et al.
frequency as well as the levels of detected ANCA seem to correlate 1995; Orriols et al. 1996; Hubner et al. 1997; Bili et al. 1999; Le Gall
with disease activity and clinical manifestations (Keogh and Specks et al. 2000; Lilly et al. 2002; Masuzawa et al. 2005; Wechsler et al.
2003; Sinico et al. 2005; Comarmond et al. 2013). 2009). All of these arguments suggest that LRA unmask a smoul-
In vitro and animal model studies have documented a variety dering vasculitis by allowing a dose reduction of systemic glucocor-
of proinflammatory effects of ANCA, suggesting that ANCA are ticoids used for asthma management, rather than causing EGPA.
important for the leukocyte activation and endothelial cell injury
observed in ANCA-associated vasculitis (Savage et al. 2002; Xiao Epidemiology
et  al. 2002; Pfister et  al. 2004; Specks 2004). For MPO-ANCA,
which are the predominant type of ANCA encountered in EGPA, Given the described diagnostic uncertainties and the rare nature
the evidence from rodent models is particularly strong (Xiao et al. of the disease, the interpretation of reported incidence and preva-
2002). The clinical observations made in two retrospective stud- lence numbers for EGPA is difficult. However, it is the rarest of the
ies further support a potentially pathogenic and disease-modifying three ANCA-associated vasculitides. The estimated annual inci-
role of ANCA in EGPA (Sinico et  al. 2005; Comarmond et  al. dence is 1–3 cases per million (Watts et al. 1995; Watts et al. 2000;
2013). In an Italian cohort of EGPA patients, manifestations of Gonzalez-Gay et  al. 2003; Reinhold-Keller et  al. 2005). Among
small-vessel vasculitis, including purpura, alveolar haemorrhage, asthma patients, the incidence of EGPA is as high as 67 per mil-
mononeuritis multiplex, and glomerulonephritis, were found to lion (Harrold et al. 2005; Loughlin et al. 2002). EGPA affects men
be significantly more common in ANCA-positive patients than slightly more often than women. The disease is typically diag-
in those who lacked these autoantibodies (Sinico et  al. 2005). nosed in middle-age patients, but it has been reported in chil-
Similarly, in a French EGPA cohort, histological features of vascu- dren (Chumbley et al. 1977; Lanham et al. 1984; Reid et al. 1998;
litis were more frequent among ANCA-positive patients, as were Guillevin et al. 1999; Solans et al. 2001; Keogh and Specks 2003;
ENT manifestations, peripheral nerve involvement, and renal dis- Hernandez-Bautista et al. 2006).
ease (Comarmond et al. 2013).
Specific organ manifestations
Leukotriene receptor antagonists The reported frequencies of organ manifestations and clinical fea-
and other drugs tures of EGPA are summarized in Table 32.1.
There has been considerable discussion about a possible pathogenic
link between leukotriene receptor antagonists (LRA) and the onset Respiratory tract
of EGPA. The introduction of LRA for the treatment of asthma Asthma is the disease-defining feature of EGPA, which is central
was followed by anecdotal reports of EGPA in patients exposed to to all three classification schemes. It occurs in almost all patients
agents of this class (Knoell et al. 1998; Wechsler et al. 1998; Tuggey (>90%) (Keogh and Specks 2003; Sinico et al. 2005; Comarmond
and Hosker 2000; Wechsler et  al. 2000; Hashimoto et  al. 2001; et  al. 2013). In 83% of patients, it precedes the onset of vasculi-
Guilpain et al. 2002; Solans et al. 2002; Hauser et al. 2008). The most tis, with a median time between onset of asthma and a diagnosis
significant argument put forth in favour of a pathogenic role was an of EGPA of 4 years (interquartile range 2–11.5 years) (Keogh and
observed two- to threefold increase in incidence of EGPA coincid- Specks 2003). In 14% of patients the diagnosis of asthma coincided
ing with the wide-spread use of LRA (Watts et al. 1995; Watts et al. with that of vasculitis, and in rare cases vasculitis precedes the
2000) but many arguments speak against a pathogenic role of LRA. asthma by over a decade (Keogh and Specks 2003).
The observed increased incidence of EGPA can easily be attrib- The paranasal sinuses are affected in approximately 75% of
uted to improved physician awareness (Watts et  al. 1995; Watts patients (Keogh and Specks 2003; Sinico et  al. 2005). It usually
436 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 32.1 Frequency of clinical manifestations in eosinophilic stage of the disease (Figure 32.4) (Worthy et al. 1998). They are
granulomatosis with polyangiitis radiographically and histopathologically indistinguishable from
chronic eosinophilic pneumonitis (Silva et  al. 2005). Poorly
Organ involvement defined small centrilobular nodules are occasionally identified
Upper respiratory tract 50–60% on high-resolution computed tomography, and larger nodules
Lungs with a diameter of 1–3 cm or granulomatous mass lesions are rare
Asthma 90–100% (Worthy et al. 1998; Choi et al. 2000; Keogh and Specks 2003; Silva
et al. 2005).
Lung nodules < 10%
Pleural effusions, occurring in up to 10% of patients, can be
Eosinophilic infiltrates 30–50% caused by congestive heart failure resulting from cardiac involve-
Alveolar haemorrhage/ capillaritis 0–20% ment or by eosinophilic pleuritis. The latter is typically associated
Skin 51–70% with eosinophils found in the pleural fluid. Mediastinal adenopa-
Nervous system thy caused by either reactive hyperplasia or eosinophilic infiltra-
Central nervous system 6–39% tion is also rare (Choi et al. 2000; Lesens et al. 2002; Comarmond
Peripheral neuropathy, multiple mononeuropathies 66–76%
et al. 2013).
Alveolar haemorrhage has been reported in EGPA patients
Gastrointestinal tract 13–59%
(Clutterbuck and Pusey 1987; Lai et  al. 1998). Three large case
Kidneys 25–58% series have reported a frequency ranging from 0 to 25% (Keogh
Heart and Specks 2003; Sinico et al. 2005; Comarmond et al. 2013). One
Endomyocardial 13–47% study has found that alveolar haemorrhage is more frequent in
Pericardial 8–32% ANCA-positive patients (Sinico et al. 2005).
Eyes < 5% The differential diagnosis of eosinophilic lung disease includes
acute and chronic eosinophilic pneumonia, allergic bronchopul-
Laboratory abnormalities monary aspergillosis, parasitic infections, drug reactions, and
Peripheral blood eosinophilia 90–100% idiopathic hypereosinophilic syndrome. Of these, only the lat-
ANCA 40–75% ter myeloproliferative disorder affects extrapulmonary organs, as
does EGPA.
(Data from Chumbley et al. 1977; Lanham et al. 1984; Guillevin et al. 1999; Solans et al. 2001;
Keogh and Specks 2003; Sinico et al. 2005; Comarmond et al. 2013.)
Skin
About half of the patients with EGPA have skin involvement,
takes the form of nasal polyposis and/or allergic or granulomatous resembling that of the other systemic small-vessel vasculitic disor-
inflammation. Severe necrotizing inflammation is much less com- ders (Davis et al. 1997; Keogh and Specks 2003; Sinico et al. 2005;
mon in EGPA than in GPA. Comarmond et al. 2013). Nodules and papules on extensor surfaces
Roentographic imaging studies reveal pulmonary parenchy- of joints are called ‘Churg–Strauss granuloma’ and represent cuta-
mal abnormalities in up to two-thirds of patients. Mostly, these neous extravascular granulomas. The term refers to the histological
are caused by eosinophilic or granulomatous inflammation with pattern of palisading granulomas with central necrosis. Despite its
or without vasculitis (Guillevin et  al. 1999; Keogh and Specks name, this is a non-specific finding, which may occur in isolation
2003). Fleeting patchy pulmonary infiltrates with a peripheral or in the setting of other systemic diseases including GPA, sys-
predominance occur in half of the patients with EGPA at some temic lupus erythematosus, and rheumatoid arthritis (Finan and

(a) (b)

Fig. 32.4 Computed tomography of the chest showing peripheral infiltrates in the right middle lobe (a) and left lower lobe (b) caused by eosinophilic inflammation of
eosinophilic granulomatosis with polyangiitis.
CHAPTER 32 eosinophilic granulomatosis with polyangiitis (churg–strauss syndrome) 437

Winkelmann 1983). Vasculitic cutaneous involvement of EGPA (Marmursztejn et  al. 2009; Dennert et  al. 2010; Szczeklik et  al.
may also present as a petechial rash, palpable purpura, or erythe- 2011). However, the clinical significance of isolated CMRI abnor-
matous maculopapules, typically developing on the lower extremi- malities has not been determined.
ties and expanding proximally. The histopathological correlate is Endomyocardial fibrosis, restrictive cardiomyopathy, and
leukocytoclastic vasculitis with or without prominent eosinophilic superimposed intracavitary thrombus can cause life-threatening
infiltration (Davis et al. 1997). complications (Ramakrishna et  al. 2000). Contiguous valvular
involvement, most commonly of the atrioventricular valves, may
Nervous system lead to valvular destruction (Ramakrishna et  al. 2000). Acute or
Neurological manifestations are common in patients with EGPA. chronic constrictive pericarditis, and pericardial effusion with or
They are similar to those found in MPA and GPA and are a conse- without cardiac tamponade, can result from eosinophilic peri-
quence of vasculitis affecting epineural vessels or the central nerv- cardial disease (Ramakrishna et al. 2000; Stollberger et al. 2005).
ous system (Vital et al. 2006). Up to 76% of patients have peripheral Whereas the original autopsy studies found small-vessel coronary
nerve involvement, typically in the form of multiple mononeuropa- vasculitis in the majority of patients (Churg and Strauss 1951), and
thies (Sehgal et al. 1995; Keogh and Specks 2003; Sinico et al. 2005; ischaemia is thought to contribute to myocardial dysfunction, angi-
Comarmond et al. 2013). Also seen are distal symmetrical poly- ographically detectable involvement of the major coronary vessels
neuropathy (24%) and less commonly asymmetric polyneuropathy is rare (Hasley et al. 1990; Kozak et al. 1995; Hellemans et al. 1997).
(3%) and lumbar radiculopathy (3%) (Sehgal et al. 1995). Central
nervous system involvement presents as confusion, seizures, or Kidneys
coma, with or without cranial nerve palsies or evidence of cere- Renal involvement occurs in one-quarter of patients with EGPA
bral infarction. It is less common than peripheral nerve involve- (Keogh and Specks 2003; Sinico et  al. 2006; Comarmond et  al.
ment, but causes significant morbidity and mortality in patients 2013), far less commonly than in GPA or MPA, where renal
with EGPA. Neurological manifestations may be more frequent in involvement affects up to 80% of patients. Furthermore, renal dis-
patients with ANCA-positive EGPA. One study found an associa- ease is usually mild and rarely causes renal failure. Only investiga-
tion between ANCA positivity and central nervous system involve- tors from the United Kingdom have reported a higher frequencies
ment (Keogh and Specks 2003), while two other studies reported of renal involvement in EGPA (50–80%), and in the report from
increased frequencies of mononeuritis multiplex among those who the Hammersmith Hospital nephrology group, renal outcome
are ANCA positive (Sinico et al. 2005; Comarmond et al. 2013). seemed similar to that of MPA (Clutterbuck et al. 1990; Reid et al.
1998). ANCA positivity has been associated with the development
Gastrointestinal manifestations of renal involvement, particularly rapidly progressive glomerulo-
Gastrointestinal tract involvement occurs in 21–31% of patients nephritis, in EGPA (Sinico et al. 2005; Comarmond et al. 2013).
(Keogh and Specks 2003; Sinico et  al. 2005; Comarmond et  al. The most common histopathological abnormality on renal biopsy
2013). Symptoms may range from abdominal pain, nausea, vomit- is that of a pauci-immune focal segmental necrotizing glomerulo-
ing, diarrhoea and gastrointestinal bleeding to an acute abdomen nephritis with crescent formation, indistinguishable from the other
(Pagnoux et al. 2005; Comarmond et al. 2013). Eosinophilic gas- ANCA-associated vasculitides (Clutterbuck et al. 1990; Sinico et al.
troenteritis or colitis and small-vessel vasculitis affecting the small 2006). Eosinophilic or granulomatous inflammation of the kidney
or large bowel can cause bowel ischaemia and may lead to mucosal are rare findings in EGPA (Clutterbuck et al. 1990; Kikuchi et al.
ulcerations and possibly perforation (Peen et  al. 2000; Memain 2001; Sinico et al. 2006).
et al. 2002).
Other clinical manifestations
Heart In addition to these most frequent clinical manifestations, any
Reports of cardiac involvement in large case series of EGPA vary organ system may be affected in EGPA. Eye, ear, and salivary gland
widely (10–49%) (Chumbley et al. 1977; Lanham et al. 1984; Keogh involvement have been reported (Robin et  al. 1985; Vitali et  al.
and Specks 2003; Sable-Fourtassou et al. 2005; Sinico et al. 2005; 1996; Granata et al. 2001; Ishiyama and Canalis 2001; Takanashi
Comarmond et al. 2013). Clinical manifestations range from car- et al. 2001; Hoffman et al. 2005; Boin et al. 2006; Cackett and Singh
diac arrest, congestive heart failure, and myocardial infarction to 2006; Comarmond et al. 2013). Myositis and even breast involve-
valvular and pericardial disease, and the predominant finding is ment may occur (De Vlam et al. 1995). Lastly, patients with EGPA
eosinophilic myocarditis (Morgan et al. 1989; Ramakrishna et al. seem to have a propensity for thromboembolic events, as in patients
2000; Val-Bernal et  al. 2003; Stollberger et  al. 2005). Two retro- with GPA (Ames et al. 1996; Garcia et al. 2005; Merkel et al. 2005;
spective studies have found that ANCA-negative patients develop Comarmond et al. 2013).
cardiomyopathy more frequently (Sinico et al. 2005; Comarmond
et al. 2013). Like other forms of eosinophilic infiltration of the myo-
cardium, it typically affects the ventricles and the diagnosis was tra- Differential diagnosis
ditionally made by echocardiography (Morgan et al. 1989). Cardiac The diagnostic approach to patients suspected of EGPA is governed
magnetic resonance imaging (CMRI) has been shown to be an even by the predominant symptom. If symptoms are centred on the res-
more sensitive test of cardiac involvement in EGPA (Marmursztejn piratory tract, the differential diagnosis needs to exclude other pul-
et al. 2009; Dennert et al. 2010). In small studies that have investi- monary eosinophilic syndromes. Once parasitic infections and drug
gated the utility of CMRI in EGPA, cardiac abnormalities have fre- reactions are excluded, EGPA needs to be differentiated from acute
quently been demonstrated in asymptomatic patients, suggesting and chronic eosinophilic pneumonia, from allergic bronchopulmo-
that clinical series may underreport the rate of cardiac involvement nary aspergillosis, and from idiopathic hypereosinophilic syndrome.
438 SECTION 5 vasculitic diseases and syndromes and related disorders

Of these, only the latter myeloproliferative disorder affects extrapul- that would provide guidance. Systemic glucocorticoids are the
monary organs, as in EGPA. If the clinical presentation features mainstay of therapy, often used in combination with immunosup-
prominent peripheral eosinophilia and multiorgan involvement, but pressants. The choice of initial therapy often depends on the extent
vasculitic features and asthma are not obvious, idiopathic hypere- of disease manifestations. Based on the observation that certain
osinophilic syndrome is the more likely diagnosis, and additional clinical findings at the time of diagnosis portend a worse outcome,
evaluations should focus on identification of specific subsets, such as Guillevin et al. created the Five Factor Score as a prognostic tool,
clonal abnormalities in the eosinophil lineage, atypical myeloprolif- which has been used to guide initial management (Guillevin et al.
erative variants, and T-lymphocytic variants. If multisystem disease 1996; Pagnoux et al. 2007). The original five factors were elevation
and vasculitis are apparent, EGPA also needs to be differentiated of serum creatinine (>1.58 mg/dl), proteinuria due to vasculitis
from GPA and MPA, particularly if the patient has ANCA. or glomerulonephritis (>1 g/ 24 h), gastrointestinal involvement,
cardiomyopathy, and central nervous system involvement. The
presence of one or more of these manifestations at diagnosis was
Diagnostic procedures and associated with increased mortality, and thus patients with at least
laboratory testing one of the factors have typically been treated with cytotoxic agents
Laboratory tests and diagnostic procedures should be performed to in addition to glucocorticoids, whereas those without any of the
firmly establish the diagnosis and differentiate it from other eosino- five factors (or any other organ- or life-threatening manifestations)
philic and vasculitic disorders, to establish the extent and severity have typically been treated with glucocorticoids alone (Guillevin
of organ involvement, and to provide baseline markers of disease et al. 1996; Vaglio et al. 2012). The elements of the Five Factor Score
activity for future assessments of treatment response. have now been revised (see Section: Prognosis), but the practice of
Peripheral blood eosinophilia detected on complete blood count initially reserving cytotoxic therapy for more severe cases of EGPA
with differential is an important component of the diagnosis and a has continued (Guillevin et al. 2011; Comarmond et al. 2013).
marker of disease activity. Immunoglobulin E levels, erythrocyte The optimal choice of immunosuppressant agent remains an area
sedimentation rate, and C-reactive protein levels are other markers of ongoing investigation (Vaglio et al. 2012). The standard treat-
of inflammatory activity that should be followed. Serum chemis- ment for EPGA associated with poor prognostic factors has been
tries and urine microscopy are critical to detect any evidence of cyclophosphamide in combination with glucocorticoids (Pagnoux
renal involvement and follow its severity. A variety of other sur- et al. 2007). Following induction of remission, conversion to a less
rogate markers of disease activity have been proposed, including toxic immunosuppressant such as azathioprine may be appropri-
serum markers of endothelial damage, lymphocyte and eosinophil ate (Pagnoux et al. 2007). Patients without poor prognostic factors
activation such as soluble thrombomodulin, vascular endothelial who are initially responding adequately to glucocorticoids alone
growth factor, soluble interleukin-2 receptor, eosinophil cationic may subsequently require glucocorticoid-sparing agents, such as
protein, and eotaxin-3. Routine measurements of these parameters methotrexate, azathioprine, mycophenolate mofetil, or hydroxyu-
in clinical practice are currently not practical (Schmitt et al. 1998; rea, if glucocorticoids cannot be tapered to acceptably low doses
Mitsuyama et al. 2006; Zwerina et al. 2011). (Della Rossa et al. 2002; Assaf et al. 2004; Metzler et al. 2004).
Chest roentgenogram or computed tomography of the chest For patients who are resistant to standard immunosuppressive
are indicated to detect pulmonary parenchymal involvement agents, particularly if eosinophilic inflammation dominates the
(Figure 32.4). Flexible bronchoscopy with bronchoalveolar lavage clinical presentation, interferon-α has been proposed as an alter-
can help to establish the presence of eosinophilic inflammation in native (Tatsis et al. 1998; Termeer et al. 2001; Lesens et al. 2002).
patients with roentgenographically detected infiltrates. This may Interferon-α therapy may have to be continued long term, in order
obviate the need for more invasive lung biopsy procedures. The dif- to prevent relapses in such patients, and there is evidence for poten-
ferential cell count of the bronchoalveolar lavage fluid usually con- tially serious treatment toxicity, which may limit its use in EGPA
firms the presence of eosinophils in patients with lung infiltrates (Gordon et al. 1998; Metzler et al. 2005; Pagnoux et al. 2005).
caused by EGPA (Wallaert et al. 1993; Schnabel et al. 1999). More recently, certain targeted biological response modifiers
Pulmonary function testing and peak flow measure- have been considered for use in EGPA. Rituximab, a chimeric
ments are integral components of the management of asthma. monoclonal antibody that targets the B-lymphocyte-specific
Pulmonary function testing should go beyond simple spirometry. cell-surface protein CD20, induces profound B-cell depletion
Abnormalities of the shape of the flow–volume curve may suggest and has been shown to be effective in the treatment of severe,
large airway involvement, and a reduction in diffusion capacity ANCA-positive GPA and MPA (Stone et  al. 2010). Several case
can be a clue to the presence of venous thromboembolic complica- reports have described rituximab as beneficial in refractory EGPA,
tions (Matsushima et al. 2006). If there is any concern for cardiac and one small open-label pilot study showed promise for its use
involvement, patients with suspected or confirmed EGPA should in ANCA-positive EGPA with active renal disease (Kaushik et al.
also undergo echocardiography, and consideration may be given 2006; Koukoulaki et al. 2006; Pepper et al. 2008; Saech et al. 2010;
to performing cardiac magnetic resonance imaging. A histopatho- Donvik and Omdal 2011; Cartin-Ceba et al. 2011). Mepolizumab,
logical diagnosis should be sought whenever possible, with biopsy a humanized monoclonal antibody against the eosinophil survival
of the most accessible affected tissue. factor IL-5, has also been considered in EGPA (Kahn et al. 2010; Kim
et al. 2010; Moosig et al. 2011). Two recent uncontrolled studies in
relapsing, refractory, or glucocorticoid-dependent EGPA demon-
Therapy strated that mepolizumab provides a steroid-sparing effect and can
The management of EGPA remains challenging, and there are no induce disease remission (Kim et al. 2010; Moosig et al. 2011) but
conclusive clinical trial results in well-defined patient populations relapse rates were high after discontinuation of mepolizumab in
CHAPTER 32 eosinophilic granulomatosis with polyangiitis (churg–strauss syndrome) 439

both studies. In fact, in patients with relapsing or refractory EGPA Assaf, C., Mewis, G., Orfanos, C.E., and Geilen, C.C. (2004). Churg-Strauss
who achieved remission with mepolizumab, the majority suffered syndrome:  successful treatment with mycophenolate mofetil. British
relapses after discontinuing the medication, despite converting to Journal of Dermatology, 150, 598–600.
Bili, A., Condemi, J.J., Bottone, S.M., and Ryan, C.K. (1999). Seven cases of
methotrexate maintenance therapy (Herrmann et al. 2012). Further
complete and incomplete forms of Churg-Strauss syndrome not related
investigations are required to better define the utility of both rituxi- to leukotriene receptor antagonists. Journal of Allergy and Clinical
mab and mepolizumab in EGPA. Immunology, 104, 1060–5.
Boin, F., Sciubba, J.J., and Stone, J.H. (2006). Churg-Strauss syndrome
Prognosis presenting with salivary gland enlargement and respiratory distress.
Arthritis and Rheumatism, 55, 167–70.
The long-term outcome and prognosis of EGPA is not entirely clear. Cackett, P. and Singh, J. (2006). Churg-Strauss syndrome in association with
Before therapy with glucocorticoids, the disease appeared univer- proliferative retinopathy. Eye, 20, 394–6.
sally fatal (Churg and Strauss 1951) but this perception may have Cartin-Ceba, R., Keogh, K.A., Specks, U., Sethi, S., and Fervenza, F.C. (2011).
been confounded by selection bias towards the most severe cases. Rituximab for the treatment of Churg-Strauss syndrome with renal
In the 1970s, EGPA was estimated to carry a mortality of about involvement. Nephrology, Dialysis, Transplantation, 26, 2865–71.
40% (Chumbley et  al. 1977). A  subsequent analysis of patients Choi, Y.H., Im, J.G., Han, B.K., Kim, J.H., Lee, K.Y., and Myoung, N.H.
evaluated at the same centre during the 1990s did not detect a sig- (2000). Thoracic manifestation of Churg-Strauss syndrome: radiologic
and clinical findings. Chest, 117, 117–24.
nificantly increased mortality for EGPA (n=91) (Keogh and Specks
Chumbley, L.C., Harrison, Jr E.G., and DeRemee, R.A. (1977). Allergic gran-
2003). This improvement in prognosis may be the result of ear- ulomatosis and angiitis (Churg-Strauss syndrome). Report and analysis
lier diagnosis and optimized use of immunosuppressive therapy of 30 cases. Mayo Clinic Proceedings, 52, 477–84.
but it may also reflect a referral bias, favouring patients with less Churg, A. (2001). Recent advances in the diagnosis of Churg-Strauss syn-
life-threatening disease manifestations. A population-based study drome. Modern Pathology, 14, 1284–93.
conducted in a cohort of 18 EGPA patients treated at the Norwich Churg, A., Brallas, M., Cronin, S.R., and Churg, J. (1995). Formes frustes of
University Hospital, UK, between 1988 and 2000, revealed 1- and Churg-Strauss syndrome. Chest, 108, 320–3.
5-year survival rates of 83.2 % and 68.1%, respectively (Lane et al. Churg, J. and Strauss, L. (1951). Allergic granulomatosis, allergic angiitis, and
2005). These patients were significantly older, and a significant periarteritis nodosa. American Journal of Pathology, 27, 277–301.
increase in mortality with age was observed (Lane et al. 2005). The Clutterbuck, E.J., Evans, D.J., and Pusey, C.D. (1990). Renal involvement
in Churg-Strauss syndrome. Nephrology, Dialysis, Transplantation,
French Vasculitis Study Group described a cohort of 383 patients
5, 161–7.
with EGPA diagnosed between 1957 and 2009, for whom 5- and Clutterbuck, E.J. and Pusey, C.D. (1987). Severe alveolar haemorrhage in
10-year survival rates were 88.9% and 78.6% (Comarmond et al. Churg-Strauss syndrome. European Journal of Respiratory Diseases, 71,
2013). As with the UK cohort, older age was associated with 158–63.
increased mortality. Among the 348 patients who underwent Comarmond, C., Pagnoux, C., Khellaf, M., Cordier, J.F., Hamidou, M.,
ANCA testing upon diagnosis, ANCA-positive patients had a Viallard, J.F., Maurier, F., Jouneau, S., Bienvenu, B., Puechal, X.,
lower mortality rate, but slightly increased risk of disease relapse Aumaitre, O., Guenno, G. L., Quellec, A.L., Cevallos, R., Fain, O.,
(Comarmond et al. 2013). Godeau, B., Seror, R., Dunogue, B., Mahr, A., Guilpain, P., Cohen,
P., Aouba, A., Mouthon, L., and Guillevin, L. (2013). Eosinophilic
The Five Factor Score, a simple prognostic tool for systemic
granulomatosis with polyangiitis (Churg-Strauss):  Clinical charac-
necrotizing vasculitides, recently underwent revision (Guillevin teristics and long-term followup of the 383 patients enrolled in the
et al. 2011). The revised factors are age greater than 65 years, eleva- French Vasculitis Study Group cohort. Arthritis and Rheumatism, 65,
tion of serum creatinine (>1.70 mg/dl), symptomatic cardiac insuf- 270–281.
ficiency, severe gastrointestinal manifestations, and the absence of Cottin, V., Tardy, F., Gindre, D., Vernet, G., Deviller, P., and Cordier, J.F.
ENT involvement. Discovery of one or more of the factors at diag- (1995). Urinary eosinophil-derived neurotoxin in Churg-Strauss syn-
nosis is associated with increased 5-year mortality. Among patients drome. Journal of Allergy and Clinical Immunology, 96, 261–4.
with EGPA, advanced age, cardiac insufficiency, and the absence Davis, M.D., Daoud, M.S., McEvoy, M.T., and Su, W.P. (1997). Cutaneous
of ENT involvement have been found to be particularly important manifestations of Churg-Strauss syndrome: a clinicopathologic corre-
lation. Journal of the American Academy of Dermatology, 37, 199–203.
prognostic features (Guillevin et al. 2011).
Della Rossa, A., Baldini, C., Tavoni, A., Tognetti, A., Neglia, D., Sambuceti,
Beyond mortality, the long-term morbidity associated with G., Puccini, R., Colangelo, C., and Bombardieri, S. (2002). Churg-Strauss
the disease and its treatment are frequently the major concern of syndrome: clinical and serological features of 19 patients from a single
patients with EGPA. The vasculitis manifestations of the disease Italian centre. Rheumatology, 41, 1286–94.
usually respond promptly to appropriate therapy. However, asthma Dennert, R.M., van Paassen, P., Schalla, S., Kuznetsova, T., Alzand, B.S.,
usually persists and is often difficult to manage. Most patients Staessen, J.A., Velthuis, S., Crijns, H.J., Tervaert, J.W., and Heymans, S.
require long-term oral glucocorticoids to control their asthma, (2010). Cardiac involvement in Churg-Strauss syndrome. Arthritis and
and many patients therefore develop steroid-associated toxicities Rheumatism, 62, 627–34.
De Vlam, K., De Keyser, F., Goemaere, S., Praet, M., and Veys, E.M. (1995).
(Comarmond et  al. 2013). Neuropathic sequelae of the disease
Churg-Strauss syndrome presenting as polymyositis. Clinical and
are another long-term management challenge, which persists long Experimental Rheumatology, 13, 505–7.
after the acute inflammation has subsided. Donvik, K.K. and Omdal, R. (2011). Churg-Strauss syndrome successfully
treated with rituximab. Rheumatology International, 31, 89–91.
References Drage, L.A., Davis, M.D., De Castro, F., Van Keulen, V., Weiss, E.A., Gleich,
Ames, P.R., Roes, L., Lupoli, S., Pickering, M., Brancaccio, V., Khamashta, G.J., and Leiferman, K.M. (2002). Evidence for pathogenic involve-
M.A., and Hughes, G.R. (1996). Thrombosis in Churg-Strauss syn- mentof eosinophils and neutrophilsin Churg-Strauss syndrome. Journal
drome. Beyond vasculitis? British Journal of Rheumatology, 35, 1181–3. of the American Academy of Dermatology, 47, 209–16.
440 SECTION 5 vasculitic diseases and syndromes and related disorders

Finan, M.C. and Winkelmann, R.K. (1983). The cutaneous extravascular Hubner, C., Dietz, A., Stremmel, W., Stiehl, A., and Andrassy, H. (1997).
necrotizing granuloma (Churg-Strauss granuloma) and systemic dis- Macrolide-induced Churg-Strauss syndrome in a patient with atopy.
ease: a review of 27 cases. Medicine, 62, 142–58. Lancet, 350, 563.
Garcia, G., Achouh, L., Cobarzan, D., Fichet, D., and Humbert, M. (2005). Hurst, S., Chizzolini, C., Dayer, J.M., Olivieri, J., and Roux-Lombard, P.
Severe venous thromboembolic disease in Churg-Strauss syndrome. (2000). Usefulness of serum eosinophil cationic protein (ECP) in pre-
Allergy, 60, 409–10. dicting relapse of Churg and Strauss vasculitis. Clinical and Experimental
Gonzalez-Gay, M.A., Garcia-Porrua, C., Guerrero, J., Rodriguez-Ledo, P., Rheumatology, 18, 784–5.
and Llorca, J. (2003). The epidemiology of the primary systemic vas- Ishiyama, A. and Canalis, R.F. (2001). Otological manifestations of
culitides in northwest Spain: implications of the Chapel Hill Consensus Churg-Strauss syndrome. Laryngoscope, 111, 1619–24.
Conference definitions. Arthritis and Rheumatism, 49, 388–93. Jakiela, B., Szczeklik, W., Sokolowska, B., Mastalerz, L., Sanak, M., Plutecka,
Gordon, A.C., Edgar, J.D., and Finch, R.G. (1998). Acute exacerbation of vas- H., and Szczeklik, A. (2009). Intrinsic pathway of apoptosis in periph-
culitis during interferon-alpha therapy for hepatitis C-associated cryo- eral blood eosinophils of Churg-Strauss syndrome. Rheumatology, 48,
globulinaemia. Journal of Infection, 36, 229–30. 1202–7.
Granata, M., Ammendolea, C., Trudu, R., Spimpolo, N., and Martelletti, P. Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F.,
(2001). Thrombosis of the retinal artery in a patient with Churg-Strauss Flores-Suarez, L.F., Gross, W.L., Guillevin, L., Hagen, E.C., Hoffman,
syndrome. Clinical and Experimental Rheumatology, 19, 234. G.S., Jayne, D.R., Kallenberg, C.G., Lamprecht, P., Langford, C.A.,
Guillevin, L., Amouroux, J., Arbeille, B., and Boura, R. (1991). Churg-Strauss Luqmani, R.A., Mahr, A.D., Matteson, E.L., Merkel, P.A., Ozen, S.,
angiitis. Arguments favoring the responsibility of inhaled antigens. Pusey, C.D., Rasmussen, N., Rees, A.J., Scott, D.G., Specks, U., Stone,
Chest, 100, 1472–3. J.H., Takahashi, K., and Watts, R.A. (2013). 2012 revised international
Guillevin, L., Cohen, P., Gayraud, M., Lhote, F., Jarrousse, B., and Casassus, chapel hill consensus conference nomenclature of vasculitides. Arthritis
P. (1999). Churg-Strauss syndrome. Clinical study and long-term and Rheumatism, 65, 1–11.
follow-up of 96 patients. Medicine, 78, 26–37. Kahn, J. E., C. Grandpeix-Guyodo, I. Marroun, E. Catherinot, F. Mellot, F.
Guillevin, L., Lhote, F., Gayraud, M., Cohen, P., Jarrousse, B., Lortholary, Roufosse, and O. Bletry. (2010). Sustained response to mepolizumab
O., Thibult, N., and Casassus, P. (1996). Prognostic factors in polyarte- in refractory Churg-Strauss syndrome. Journal of Allergy and Clinical
ritis nodosa and Churg-Strauss syndrome. A prospective study in 342 Immunology, 125, 267–70.
patients. Medicine, 75, 17–28. Kaushik, V.V., Reddy, H.V., and Bucknall, R.C. (2006). Successful use of ritux-
Guillevin, L., Pagnoux, C., Seror, R., Mahr, A., Mouthon, L., and Le Toumelin, imab in a patient with recalcitrant Churg-Strauss syndrome. Annals of
P. (2011). The Five-Factor Score revisited: assessment of prognoses of the Rheumatic Diseases, 65, 1116–7.
systemic necrotizing vasculitides based on the French Vasculitis Study Keogh, K. A. and Specks, U. (2003). Churg-Strauss syndrome: clinical pres-
Group (FVSG) cohort. Medicine, 90, 19–27. entation, antineutrophil cytoplasmic antibodies, and leukotriene recep-
Guillevin, L., Visser, H., Noel, L.H., Pourrat, J., Vernier, I., Gayraud, M., tor antagonists. American Journal of Medicine, 115, 284–90.
Oksman, F., and Lesavre, P. (1993). Antineutrophil cytoplasm anti- Kiene, M., Csernok, E., Muller, A., Metzler, C., Trabandt, A., and Gross,
bodies in systemic polyarteritis nodosa with and without hepatitis B W.L. (2001). Elevated interleukin-4 and interleukin-13 production by
virus infection and Churg-Strauss syndrome—62 patients. Journal of T cell lines from patients with Churg-Strauss syndrome. Arthritis and
Rheumatology, 20, 1345–9. Rheumatism, 44, 469–73.
Guilpain, P., Viallard, J.F., Lagarde, P., Cohen, P., Kambouchner, M., Pellegrin, Kikuchi, Y., Ikehata, N., Tajima, O., Yoshizawa, N., and Miura, S. (2001).
J.L., and Guillevin, L. (2002). Churg-Strauss syndrome in two patients Glomerular lesions in patients with Churg-Strauss syndrome and the
receiving montelukast. Rheumatology, 41, 535–9. anti-myeloperoxidase antibody. Clinical Nephrology, 55, 429–35.
Harrold, L.R., Andrade, S.E., Go, A.S., Buist, A.S., Eisner, M., Vollmer, Kim, S., Marigowda, G., Oren, E., Israel, E., and Wechsler, M.E. (2010).
W.M., Chan, K.A., Frazier, E.A., Weller, P.F., Wechsler, M.E., Yood, Mepolizumab as a steroid-sparing treatment option in patients with
R.A., Davis, K.J., and Platt, R. (2005). Incidence of Churg-Strauss syn- Churg-Strauss syndrome. Journal of Allergy and Clinical Immunology,
drome in asthma drug users: a population-based perspective. Journal of 125, 1336–43.
Rheumatology, 32, 1076–80. Knoell, D.L., Lucas, J., and Allen, J.N. (1998). Churg-Strauss syndrome asso-
Hashimoto, M., Fujishima, T., Tanaka, H., Kon, H., Saikai, T., Suzuki, A., ciated with zafirlukast. Chest, 114, 332–4.
Nakatsugawa, M., and Abe, S. (2001). Churg-Strauss syndrome after Koukoulaki, M., Smith, K.G., and Jayne, D.R. (2006). Rituximab in
reduction of inhaled corticosteroid in a patient treated with pranlukast Churg-Strauss syndrome. Annals of the Rheumatic Diseases, 65, 557–9.
for asthma. Internal Medicine, 40, 432–4. Kozak, M., Gill, E.A., and Green, L.S. (1995). The Churg-Strauss syndrome.
Hasley, P.B., Follansbee, W.P., and Coulehan, J.L. (1990). Cardiac manifes- A case report with angiographically documented coronary involvement
tations of Churg-Strauss syndrome: report of a case and review of the and a review of the literature. Chest, 107, 578–80.
literature. American Heart Journal, 120, 996–9. Kurosawa, M., Nakagami, R., Morioka, J., Inamura, H., Mizushima, Y.,
Hauser, T., Mahr, A., Metzler, C., Coste, J., Sommerstein, R., Gross, W.L., Sugawara, N., Yamashita, T., Yokoseki, T., Kitamura, S., Omura, Y.,
Guillevin, L., and Hellmich, B. (2008). The leucotriene receptor Shibata, M., and Chihara, J. (2000). Interleukins in Churg-Strauss syn-
antagonist montelukast and the risk of Churg-Strauss syndrome:  a drome. Allergy, 55, 785–7.
case-crossover study. Thorax, 63, 677–82. Lai, R.S., Lin, S.L., Lai, N.S., and Lee, P.C. (1998). Churg-Strauss syndrome
Hellemans, S., Dens, J., and Knockaert, D. (1997). Coronary involvement in presenting with pulmonary capillaritis and diffuse alveolar hemorrhage.
the Churg-Strauss syndrome. Heart, 77, 576–8. Scandinavian Journal of Rheumatology, 27, 230–2.
Hernandez-Bautista, V.M., Espinosa-Padilla, S.E., Yamazaki-Nakashimada, Lane, S.E., Watts, R.A., Bentham, G., Innes, N.J., and Scott, D.G. (2003).
M.A., Lopez-Lara, D., Gonzalez-Serrano, E., Staines-Boone, T., and Are environmental factors important in primary systemic vasculitis?
Espinosa-Rosales, F. (2006). Pediatric Churg-Strauss syndrome in A case-control study. Arthritis and Rheumatism, 48, 814–23.
Mexico. Pediatric Pulmonology, 41, 379–82. Lane, S.E., Watts, R.A., Shepstone, L., and Scott, D.G. (2005). Primary sys-
Herrmann, K., Gross, W.L., and Moosig, F. (2012). Extended follow-up after temic vasculitis: clinical features and mortality. QJM, 98, 97–111.
stopping mepolizumab in relapsing/refractory Churg-Strauss syn- Lanham, J.G., Elkon, K.B., Pusey, C.D., and Hughes, G.R. (1984). Systemic
drome. Clinical and Experimental Rheumatology, 30 (Suppl. 70), S62–5. vasculitis with asthma and eosinophilia:  a clinical approach to the
Hoffman, P.M., Godfrey, T., and Stawell, R.J. (2005). A case of Churg-Strauss Churg-Strauss syndrome. Medicine, 63, 65–81.
syndrome with visual loss following central retinal artery occlusion. Le Gall, C., Pham, S., Vignes, S., Garcia, G., Nunes, H., Fichet, D., Simonneau,
Lupus, 14, 174–5. G., Duroux, P., and Humbert, M. (2000). Inhaled corticosteroids and
CHAPTER 32 eosinophilic granulomatosis with polyangiitis (churg–strauss syndrome) 441

Churg-Strauss syndrome:  a report of five cases. European Respiratory Pagnoux, C., Guilpain, P., and Guillevin, L. (2007). Churg-Strauss syndrome.
Journal, 15, 978–81. Current Opinion in Rheumatology, 19, 25–32.
Lesens, O., Hansmann, Y., Nerson, J., Pasquali, J., Gasser, B., Wihlm, J., and Pagnoux, C., Mahr, A., Cohen, P., and Guillevin, L. (2005). Presentation and
Christmann, D. (2002). Severe Churg-Strauss syndrome with mediasti- outcome of gastrointestinal involvement in systemic necrotizing vas-
nal lymphadenopathy treated with interferon therapy. European Journal culitides: analysis of 62 patients with polyarteritis nodosa, microscopic
of Internal Medicine, 13, 458. polyangiitis, Wegener granulomatosis, Churg-Strauss syndrome, or
Lie, J.T. (1993). Limited forms of Churg-Strauss syndrome. Pathology Annual, rheumatoid arthritis-associated vasculitis. Medicine, 84, 115–28.
28, 199–220. Peen, E., Hahn, P., Lauwers, G., Williams, R.C. Jr, Gleich, G., and Kephart,
Lilly, C.M., Churg, A., Lazarovich, M., Pauwels, R., Hendeles, L., Rosenwasser, G.M. (2000). Churg-Strauss syndrome:  localization of eosinophil
L.J., Ledford, D., and Wechsler, M.E. (2002). Asthma therapies and major basic protein in damaged tissues. Arthritis and Rheumatism, 43,
Churg-Strauss syndrome. Journal of Allergy and Clinical Immunology, 1897–900.
109, S1–19. Pepper, R.J., Fabre, M.A., Pavesio, C., Gaskin, G., Jones, R.B., Jayne, D., Pusey,
Loughlin, J.E., Cole, J.A., Rothman, K.J., and Johnson, E.S. (2002). Prevalence C.D., and Salama, A.D. (2008). Rituximab is effective in the treatment of
of serious eosinophilia and incidence of Churg-Strauss syndrome in a refractory Churg-Strauss syndrome and is associated with diminished
cohort of asthma patients. Annals of Allergy, Asthma and Immunology, T-cell interleukin-5 production. Rheumatology, 47, 1104–5.
88, 319–25. Pfister, H., Ollert, M., Frohlich, L.F., Quintanilla-Martinez, L., Colby, T.V.,
Marmursztejn, J., Vignaux, O., Cohen, P., Guilpain, P., Pagnoux, C., Gouya, Specks, U., and Jenne, D.E. (2004). Antineutrophil cytoplasmic autoan-
H., Mouthon, L., Legmann, P., Duboc, D., and Guillevin, L. (2009). tibodies against the murine homolog of proteinase 3 (Wegener autoanti-
Impact of cardiac magnetic resonance imaging for assessment of gen) are pathogenic in vivo. Blood, 104, 1411–8.
Churg-Strauss syndrome: a cross-sectional study in 20 patients. Clinical Ramakrishna, G., Connolly, H.M., Tazelaar, H.D., Mullany, C.J., and
and Experimental Rheumatology, 27 (1 Suppl. 52), S70–6. Midthun, D.E. (2000). Churg-Strauss syndrome complicated by eosino-
Masi, A.T., Hunder, G.G., Lie, J.T., Michel, B.A., Bloch, D.A., Arend, W.P., philic endomyocarditis. Mayo Clinic Proceedings, 75, 631–5.
Calabrese, L.H., Edworthy, S.M., Fauci, A. S., Leavitt, R.Y., et al. (1990). Reid, A.J., Harrison, B.D., Watts, R.A., Watkin, S.W., McCann, B.G., and
The American College of Rheumatology 1990 criteria for the classifica- Scott, D.G. (1998). Churg-Strauss syndrome in a district hospital. QJM,
tion of Churg-Strauss syndrome (allergic granulomatosis and angiitis). 91, 219–29.
Arthritis and Rheumatism, 33, 1094–100. Reinhold-Keller, E., Herlyn, K., Wagner-Bastmeyer, R., and Gross, W.L.
Masuzawa, A., Moriguchi, M., Tsuda, T., Sugawara, H., Otsuka, M., Yamada, (2005). Stable incidence of primary systemic vasculitides over five
S., Tabei, K., and Kawakami, M. (2005). Churg-Strauss syndrome years:  results from the German vasculitis register. Arthritis and
associated with hypersensitivity to acetaminophen. Internal Medicine, Rheumatism, 53, 93–9.
44, 496–8. Robin, J.B., Schanzlin, D.J., Meisler, D.M., deLuise, V.P., and Clough, J.D.
Matsushima, H., Takayanagi, N., Kurashima, K., Tokunaga, D., Ubukata, M., (1985). Ocular involvement in the respiratory vasculitides. Survey of
Kawabata, Y., and Sugita, Y. (2006). Multiple tracheobronchial mucosal Ophthalmology, 30, 127–40.
lesions in two cases of Churg-Strauss syndrome. Respirology, 11, 109–12. Sable-Fourtassou, R., Cohen, P., Mahr, A., Pagnoux, C., Mouthon, L., Jayne,
Memain, N., De, B.M., Guillevin, L., Wechsler, B., and Meyer, O. (2002). D., Blockmans, D., Cordier, J.F., Delaval, P., Puechal, X., Lauque, D.,
Delayed relapse of Churg-Strauss syndrome manifesting as colon ulcers Viallard, J.F., Zoulim, A., and Guillevin, L. (2005). Antineutrophil cyto-
with mucosal granulomas: 3 cases. Journal of Rheumatology, 29, 388–91. plasmic antibodies and the Churg-Strauss syndrome. Annals of Internal
Merkel, P.A., Lo, G.H., Holbrook, J.T., Tibbs, A. K., Allen, N.B., Davis Jr, J.C., Medicine, 143, 632–8.
Hoffman, G.S., McCune, W.J., St Clair, E.W., Specks, U., Spiera, R., Petri, Saech, J., Owczarczyk, K., Rosgen, S., Petereit, H., Hallek, M., and
M., and Stone, J.H. (2005). Brief communication:  high incidence of Rubbert-Roth, A. (2010). Successful use of rituximab in a patient with
venous thrombotic events among patients with Wegener granulomato- Churg-Strauss syndrome and refractory central nervous system involve-
sis: the Wegeners Clinical Occurrence of Thrombosis (WeCLOT) Study. ment. Annals of the Rheumatic Diseases, 69, 1254–5.
Annals of Internal Medicine, 142, 620–6. Savage, C.O., Harper, L., and Holland, M. (2002). New findings in pathogen-
Metzler, C., Hellmich, B., Gause, A., Gross, W.L., and de Groot, K. (2004). esis of antineutrophil cytoplasm antibody-associated vasculitis. Current
Churg Strauss syndrome—successful induction of remission with meth- Opinion in Rheumatology, 14, 15–22.
otrexate and unexpected high cardiac and pulmonary relapse ratio dur- Schmitt, W.H., Csernok, E., Kobayashi, S., Klinkenborg, A., Reinhold-Keller,
ing maintenance treatment. Clinical and Experimental Rheumatology, 22 E., and Gross, W.L. (1998). Churg-Strauss syndrome:  serum mark-
(6 Suppl. 36), S52–61. ers of lymphocyte activation and endothelial damage. Arthritis and
Metzler, C., Lamprecht, P., Hellmich, B., Reuter, M., Arlt, A.C., and Gross, Rheumatism, 41, 445–52.
W.L. (2005). Leucoencephalopathy after treatment of Churg-Strauss syn- Schnabel, A., Csernok, E., Braun, J., and Gross, W.L. (1999). Inflammatory
drome with interferonα Annals of the Rheumatic Diseases, 64, 1242–3. cells and cellular activation in the lower respiratory tract in Churg-Strauss
Mitsuyama, H., Matsuyama, W., Iwakawa, J., Higashimoto, I., Watanabe, M., syndrome. Thorax, 54, 771–8.
Osame, M., and Arimura, K. (2006). Increased serum vascular endothe- Schonermarck, U., Csernok, E., Trabandt, A., Hansen, H., and Gross, W.L.
lial growth factor level in Churg-Strauss syndrome. Chest, 129, 407–11. (2000). Circulating cytokines and soluble CD23, CD26 and CD30 in
Moosig, F., Gross, W.L., Herrmann, K., Bremer, J.P., and Hellmich, B. (2011). ANCA-associated vasculitides. Clinical and Experimental Rheumatology,
Targeting interleukin-5 in refractory and relapsing Churg-Strauss syn- 18, 457–63.
drome. Annals of Internal Medicine, 155, 341–3. Sehgal, M., Swanson, J.W., DeRemee, R.A., and Colby, T.V. (1995). Neurologic
Morgan, J.M., Raposo, L., and Gibson, D.G. (1989). Cardiac involvement manifestations of Churg-Strauss syndrome. Mayo Clinic Proceedings, 70,
in Churg-Strauss syndrome shown by echocardiography. British Heart 337–41.
Journal, 62, 462–6. Sheffer, A.L., Rocklin, R.E., and Goetzl, E.J. (1975). Immunologic compo-
Muschen, M., Warskulat, U., Perniok, A., Even, J., Moers, C., Kismet, B., nents of hypersensitivity reactions to cromolyn sodium. New England
Temizkan, N., Simon, D., Schneider, M., and Haussinger, D. (1999). Journal of Medicine, 293, 1220–4.
Involvement of soluble CD95 in Churg-Strauss syndrome. American Silva, C.I., Muller, N.L., Fujimoto, K., Johkoh, T., Ajzen, S.A., and Churg, A.
Journal of Pathology, 155, 915–25. (2005). Churg-Strauss syndrome:  high resolution CT and pathologic
Orriols, R., Munoz, X., Ferrer, J., Huget, P., and Morell, F. (1996). findings. Journal of Thoracic Imaging, 20, 74–80.
Cocaine-induced Churg-Strauss vasculitis. European Respiratory Sinico, R.A., Di Toma, L., Maggiore, U., Bottero, P., Radice, A., Tosoni, C.,
Journal, 9, 175–7. Grasselli, C., Pavone, L., Gregorini, G., Monti, S., Frassi, M., Vecchio, F.,
442 SECTION 5 vasculitic diseases and syndromes and related disorders

Corace, C., Venegoni, E., and Buzio, C. (2005). Prevalence and clinical Val-Bernal, J.F., Mayorga, M., Garcia-Alberdi, E., and Pozueta, J.A. (2003).
significance of antineutrophil cytoplasmic antibodies in Churg-Strauss Churg-Strauss syndrome and sudden cardiac death. Cardiovascular
syndrome. Arthritis and Rheumatism, 52, 2926–35. Pathology, 12, 94–7.
Sinico, R.A., Di Toma, L., Maggiore, U., Tosoni, C., Bottero, P., Sabadini, Vital, A., Vital, C., Viallard, J.F., Ragnaud, J.M., Canron, M.H., and Lagueny,
E., Giammarresi, G., Tumiati, B., Gregorini, G., Pesci, A., Monti, S., A. (2006). Neuro-muscular biopsy in Churg-Strauss syndrome: 24 cases.
Balestrieri, G., Garini, G., Vecchio, F., and Buzio, C. (2006). Renal Journal of Neuropathology and Experimental Neurology, 65, 187–92.
involvement in Churg-Strauss syndrome. American Journal of Kidney Vitali, C., Genovesi-Ebert, F., Romani, A., Jeracitano, G., and Nardi, M.
Diseases, 47, 770–9. (1996). Ophthalmological and neuro-ophthalmological involvement in
Solans, R., Bosch, J.A., Perez-Bocanegra, C., Selva, A., Huguet, P., Alijotas, J., Churg-Strauss syndrome: a case report. Graefe’s Archive for Clinical and
Orriols, R., Armadans, L., and Vilardell, M. (2001). Churg-Strauss syn- Experimental Ophthalmology, 234, 404–8.
drome: outcome and long-term follow-up of 32 patients. Rheumatology, Wallaert, B., Gosset, P., Prin, L., Bart, F., Marquette, C.H., and Tonnel, A.B.
40, 763–71. (1993). Bronchoalveolar lavage in allergic granulomatosis and angiitis.
Solans, R., Bosch, J.A., Selva, A., Orriols, R., and Vilardell, M. (2002). European Respiratory Journal, 6, 413–7.
Montelukast and Churg-Strauss syndrome. Thorax, 57, 183–5. Watts, R.A., Carruthers, D.M., and Scott, D.G. (1995). Epidemiology of sys-
Specks, U. (2004). Antineutrophil cytoplasmic antibodies: are they patho- temic vasculitis: changing incidence or definition? Seminars in Arthritis
genic? Clinical and Experimental Rheumatology, 22 (6 Suppl. 36), S7–12. and Rheumatism, 25, 28–34.
Stollberger, C., Finsterer, J., and Winkler, W.B. (2005). Eosinophilic pericardial Watts, R.A., Lane, S.E., Bentham, G., and Scott, D.G. (2000). Epidemiology
effusion in Churg-Strauss syndrome. Respiratory Medicine, 99, 377–9. of systemic vasculitis: a ten-year study in the United Kingdom. Arthritis
Stone, J.H., Merkel, P.A., Spiera, R., Seo, P., Langford, C.A., Hoffman, G.S., and Rheumatism, 43, 414–9.
Kallenberg, C.G., St Clair, E.W., Turkiewicz, A., Tchao, N.K., Webber, Wechsler, M.E., Finn, D., Gunawardena, D., Westlake, R., Barker, A.,
L., Ding, L., Sejismundo, L.P., Mieras, K., Weitzenkamp, D., Ikle, D., Haranath, S.P., Pauwels, R.A., Kips, J.C., and Drazen, J.M. (2000).
Seyfert-Margolis, V., Mueller, M., Brunetta, P., Allen, N.B., Fervenza, Churg-Strauss syndrome in patients receiving montelukast as treatment
F.C., Geetha, D., Keogh, K. A., Kissin, E.Y., Monach, P.A., Peikert, T., for asthma. Chest, 117, 708–13.
Stegeman, C., Ytterberg, S.R., and Specks, U. (2010). Rituximab ver- Wechsler, M.E., Garpestad, E., Flier, S.R., Kocher, O., Weiland, D.A., Polito,
sus cyclophosphamide for ANCA-associated vasculitis. New England A.J., Klinek, M.M., Bigby, T.D., Wong, G.A., Helmers, R.A., and Drazen,
Journal of Medicine, 363, 221–32. J.M. (1998). Pulmonary infiltrates, eosinophilia, and cardiomyopathy
Szczeklik, W., Miszalski-Jamka, T., Mastalerz, L., Sokolowska, B., Dropinski, following corticosteroid withdrawal in patients with asthma receiving
J., Banys, R., Hor, K.N., Mazur, W., and Musial, J. (2011). Multimodality zafirlukast. JAMA, 279, 455–7.
assessment of cardiac involvement in Churg-Strauss syndrome patients Wechsler, M.E., Wong, D.A., Miller, M.K., and Lawrence-Miyasaki, L. (2009).
in clinical remission. Circulation Journal, 75, 649–55. Churg-strauss syndrome in patients treated with omalizumab. Chest,
Takanashi, T., Uchida, S., Arita, M., Okada, M., and Kashii, S. (2001). 136, 507–18.
Orbital inflammatory pseudotumor and ischemic vasculitis in Weller, P.F., Plaut, M., Taggart, V., and Trontell, A. (2001). The relationship of
Churg-Strauss syndrome: report of two cases and review of the litera- asthma therapy and Churg-Strauss syndrome: NIH workshop summary
ture. Ophthalmology, 108, 1129–33. report. Journal of Allergy and Clinical Immunology, 108, 175–83.
Tatsis, E., Schnabel, A., and Gross, W.L. (1998). Interferon-alpha treatment Wieczorek, S., Hellmich, B., Arning, L., Moosig, F., Lamprecht, P., Gross,
of four patients with the Churg-Strauss syndrome. Annals of Internal W.L., and Epplen, J.T. (2008a). Functionally relevant variations of
Medicine, 129, 370–4. the interleukin-10 gene associated with antineutrophil cytoplasmic
Termeer, C.C., Simon, J.C., and Schopf, E. (2001). Low-dose interferon antibody-negative Churg-Strauss syndrome, but not with Wegeners
alfa-2b for the treatment of Churg-Strauss syndrome with prominent granulomatosis. Arthritis and Rheumatism, 58, 1839–48.
skin involvement. Archives of Dermatology, 137, 136–8. Wieczorek, S., Hellmich, B., Gross, W.L., and Epplen, J.T. (2008b). Associations
Tervaert, J.W. and Kallenberg, C.G. (1993). Anti-myeloperoxidase antibodies of Churg-Strauss syndrome with the HLA-DRB1 locus, and relationship
in Churg-Strauss syndrome. Journal of Neurology, 240, 449–50. to the genetics of antineutrophil cytoplasmic antibody-associated vascu-
Tervaert, J.W., Limburg, P.C., Elema, J.D., Huitema, M.G., Horst, G., The, litides: comment on the article by Vaglio et al. Arthritis and Rheumatism,
T.H., and Kallenberg, C.G. (1991). Detection of autoantibodies against 58, 329–30.
myeloid lysosomal enzymes: a useful adjunct to classification of patients Worthy, S.A., Muller, N.L., Hansell, D.M., and Flower, C.D. (1998).
with biopsy-proven necrotizing arteritis. American Journal of Medicine, Churg-Strauss syndrome: the spectrum of pulmonary CT findings in 17
91, 59–66. patients. American Journal of Roentgenology, 170, 297–300.
Tuggey, J.M. and Hosker, H.S. (2000). Churg-Strauss syndrome associated Xiao, H., Heeringa, P., Hu, P., Liu, Z., Zhao, M., Aratani, Y., Maeda, N., Falk,
with montelukast therapy. Thorax, 55, 805–6. R.J., and Jennette, J.C. (2002). Antineutrophil cytoplasmic autoantibod-
Vaglio, A., Martorana, D., Maggiore, U., Grasselli, C., Zanetti, A., Pesci, A., ies specific for myeloperoxidase cause glomerulonephritis and vasculitis
Garini, G., Manganelli, P., Bottero, P., Tumiati, B., Sinico, R.A., Savi, M., in mice. Journal of Clinical Investigation, 110, 955–63.
Buzio, C., and Neri, T.M. (2007). HLA-DRB4 as a genetic risk factor for Zwerina, J., Bach, C., Martorana, D., Jatzwauk, M., Hegasy, G., Moosig,
Churg-Strauss syndrome. Arthritis and Rheumatism, 56, 3159–66. F., Bremer, J., Wieczorek, S., Moschen, A., Tilg, H., Neumann, T.,
Vaglio, A., Moosig, F., and Zwerina, J. (2012). Churg-Strauss syn- Spriewald, B.M., Schett, G., and Vaglio, A. (2011). Eotaxin-3 in Churg-
drome: update on pathophysiology and treatment. Current Opinion in Strauss syndrome: a clinical and immunogenetic study. Rheumatology,
Rheumatology, 24, 24–30. 50, 1823–7.
CHAPTER 33

Vasculitis in primary
connective tissue diseases
Laura B. Hughes

Introduction as well as other extra-articular manifestations of RA appear to be


declining (Nyhall-Wahlin et al. 2012).
In 1957, Bywaters suggested that vasculitis was an inherent com- Examples of clinical and angiographic manifestations of RV are
ponent of all connective tissue diseases (Bywaters 1957). While shown in Figure 33.1.
vasculitis arising in the context of a pre-existing connective tis- RV appears to affect patients with more aggressive RA. It occurs
sue disease (CTD) is well documented, its presence varies widely on average of 10–14 years after the onset of RA; however, it can
among such diseases. Characterized as multisystem inflammatory occur before or at the time of RA diagnosis (Chen et al. 2002; Gray
processes, CTDs such as rheumatoid arthritis (RA), Sjögren’s syn- and Poppo 1983; Vollertsen et  al. 1986). There are many patient
drome (SS), systemic sclerosis (SSc), and systemic lupus erythe- factors that have been associated with an increased risk for the
matosus (SLE) may have vascular involvement as a major clinical development of RV. Some of these factors include: a high titre rheu-
manifestation. Vasculitis in these various CTDs often shares char- matoid factor (RF), ANA positivity, male gender, smoking, longer
acteristics:  (1)  it may affect vessels of any calibre, concurrently disease duration, presence of joint erosions or subcutaneous nod-
or sequentially; (2) it can vary from mild protean symptoms to ules or other extra-articular manifestations, increased number of
severe, life-threatening disease; and (3)  it rarely precedes other disease-modifying antirheumatic drugs (DMARDs), and longer
manifestations of the particular CTD (Danning et  al. 1998). In disease duration when compared with age-matched RA patients
general, vasculitis in CTD is often associated with more severe without RV (Struthers et al. 1981; Voskuyl et al. 1996). Similarly,
disease activity and with longer disease duration. The clinical RV is reported in as many as 30% of persons with Felty’s syndrome
manifestations of vasculitis associated with CTDs, along with (Campion et al. 1990). The association with smoking may be due to
therapeutic considerations, are described in this chapter. Key the immunomodulatory effects or to endothelial damage that pre-
clinical features of vasculitis in these conditions are summarized disposes to vascular inflammation (Struthers et al. 1981; Turesson
in Table 33.1. et al. 2004).
Significant morbidity and mortality can occur in patients with
Rheumatoid vasculitis RV. The mortality has been reported to be as high as 43%, most
notably within the first 6  months after onset of vasculitis (Scott
Epidemiology et al. 1981c). Predictors of decreased survival include younger age
The occurrence of vasculitis in RA underscores the systemic inflam- at diagnosis of RV, delayed diagnosis, abnormal urinary sediment,
matory nature of the disease. RA is primarily manifested by sym- high titre RF, and hypergammaglobulinaemia (Geirsson et al. 1987;
metric inflammation in the synovial lining of diarthrodial joints. Vollertsen et al. 1986). Cardiac involvement, gangrene, mononeuri-
Extra-articular disease, particularly involving the skin (rheumatoid tis multiplex, and bowel infarctions suggest a fatal prognosis (Chen
nodules), lungs (e.g. pleuritis, interstitial lung disease (ILD)), and et al. 2002; Geirsson et al. 1987).
heart (pericarditis, myocarditis), is also well recognized. Vasculitis
in RA can lead to a variety of extra-articular conditions, ranging Pathogenesis
from mild to severe. The overall annual incidence of rheumatoid In 1959, the dominant scheme for RV pathogenesis was attributed
vasculitis (RV) is <1%, with a lifetime risk of 1 in 9 for males and to RF action in the vessel wall. Several clinical and experimental
1 in 38 for females with RA (Panush et al. 1983; Watts et al. 1994). studies offer support to this theory, describing circulating immune
Post-mortem studies place the incidence of RV closer to 25% of complexes containing IgG and IgA rheumatoid factor, deposition
RA patients, perhaps consistent with the view that RV is associ- of complement, and the presence of autoantibodies such as antien-
ated with more severe RA (Bacon and Carruthers 1995). The severe dothelial cell antibodies, which form deposits in vessel walls trig-
form of RV (threatening end-organ damage) is rare; it is estimated gering an inflammatory reaction (Breedveld et al. 1988; Jans et al.
to occur in less than 1 per 100  000 of the general population 1983; Scott et al. 1981a). Indeed, patients with RV typically have
(Goronzy and Weyand 1994). In the present era of tight disease polyclonal hypergammaglobulinaemia with high titre rheuma-
control and availability of biological agents, the incidence of RV toid factor, detectable serum immune complexes (IC), and often
444 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 33.1 Characteristic of vasculitis associated with primary connective tissue diseases

Disease Vessel size Comments


Rheumatoid arthritis Small vessel, typically leukocytoclastic vasculitis. Vasculitis is a complication of RA in 10–15% of patients, usually those
Necrotizing vasculitis of larger arteries occurs. with more severe disease.

SLE Usually small vessel/leukocytoclastic. Diseases resulting in pseudovasculitis (atherosclerosis;


Can involve the great vessels. thromboembolism with or without APLs) are common in SLE.

Sjögren’s syndrome Usually small vessel/leukocytoclastic. Mixed cryoglobulinaemia may occur and concurrent infection with
hepatitis C has been reported.
Latent HIV infection should also be considered. Hyperviscosity can
also produce ‘vasculitis’.
Systemic sclerosis Cases reported involve small vessels. Vasculitis is decidedly rare in primary, diffuse SSc.
Most instances of vasculitis occur with limited variant (with
anticentromere antibodies) or in overlap with Sjögren’s syndrome
SNSA Vasculitis reported in anecdotal cases Clinical suspicion should be raised for underlying HIV infection when
vasculitis complicates the SNSAs.
Relapsing polychondritis Valvular (aortic) disease is most common but necrotizing RP may also occur in other rheumatic conditions where vasculitis is
vasculitis of medium to large arteries has been reported. common as well as in certain malignancies.
Inflammatory myopathies Rare and involves small vessels/ capillaries when present. Vasculitis is more common in paediatric cases.
Aortitis, periaortitis, and IgG-4 related syndromes with <10% of cases have true Treatment requires long-term immunosuppression.
retroperitoneal fibrosis vasculitis of the aorta.

RA, rheumatoid arthritis; SLE, systemic lupus erythematosus; APLs, antiphospholipid antibodies; HIV, human immunodeficiency virus; SSc, systemic sclerosis; SNSA, seronegative
spondyloarthropathies; RP, relapsing polychondritis.

diminished levels of serum complement (Bacons and Kitas 1994). TNF-α expression in endothelial and perivascular infiltrative cells in
Rapoport et al. reported an association between cutaneous immune active RV lesions (Flipo et al. 1994). There is also evidence to suggest
complex deposition, detected via direct immunofluorescence, and dysregulated B cell–T cell interactions in RV (Turesson et al. 2005).
cutaneous RV (Rapoport et al. 1980). Increased levels of circulating fibronectin have been detected in
More recently, T-cell mediated processes have been postulated. patients with RV (versus other CTDs), perhaps indicating activa-
Because ICs cannot always be detected, vasculitis mediated by direct tion of vascular endothelium (Voskuyl et al. 1998a). Autoantibodies
cell–cell contacts has been postulated by Goronzy et al. (Goronzy reportedly associated with RV include antiendothelial cell antibod-
and Weyand 1994). T-cell abnormalities observed in patients with ies (AECA), which were detected in 87% of patients with RV in
RV (and extra-articular RA) include expansion of CD8+ large granu- one series (van der Zee et al. 1991). It is not clear whether these
lar lymphocytes, and immunosenescent CD4+CD28− cells (Goronzy represent pathogenically active molecules or are simply indicators
and Weyand 2003; Martens et al. 1997). Flipo et al. proposed a TH-1 of the ongoing inflammatory process.
mediated process leading to an increase in ICAM-1, E-selectin, and There is increasing evidence to support the role of genetics in
RV. An association of RV with the MHC class II HLA-DR4 genes
containing the RA-associated shared epitope (SE) alleles has been
described in several studies (Gonzalez et al. 1997; Hillarby et al.
1991; Jaraquemada et  al. 1986; Toussirot et  al. 1999; Turesson
et al. 2005; Voskuyl et al. 1997; Weyand et al. 1992). These asso-
ciations tend to be stronger in studies of northern European
and Caucasian American RA populations. The presence of two
HLA-DRB1*04 SE alleles was associated with RV (OR 2.44, 95%
CI 1.22–4.89) in a multicentre study of patients from Minnesota
and Sweden (Turesson et al. 2005). Other studies have found an
association between mild cutaneous RV and the *0401 and *0404
alleles (Voskuyl et al. 1997). In a meta-analysis, RV was found to
be associated with three specific genotypes containing a dou-
ble dose of the SE, HLE-DRB1*0401/*0401, *0401/*0404, and
*0401/*0101(Gorman et al. 2004). Other genes, such as the MHC
class I alleles HLA-C*03 and HLA-C*05 and the KIR2DS2 gene,
encoding a stimulatory killer-immunoglobulin-like receptor (KIR)
Fig. 33.1 Vasculitis in a young woman with systemic lupus erythematosus. on natural killer (NK) cells, have also been associated with vasculi-
(Courtesy of Dr Gene Ball.) tis in RA (Yen et al. 2001).
CHAPTER 33 vasculitis in primary connective tissue diseases 445

RV can affect vessels of varying size with varying degrees of of the cerebral parenchyma and meninges (Chin and Latov 2005;
inflammation, which may occur concurrently in the same patient. Spath et al. 2012). CNS RV can lead to significant morbidity and
Milder forms of RV typically involve smaller vessels (i.e. digital mortality unless it is recognized early and properly treated (Kim
arteries and postcapillary venules) of the skin characterized by et al. 1982). Treatment with intravenous pulse glucocorticoids and
intimal proliferation, immune complex deposition, and intra- cyclophosphamide have shown the best reported outcomes (Chin
vascular thrombosis (Chen et al. 2002; Turesson and Jacobsson and Latov 2005; Spath et al. 2012).
2004). Medium and large artery involvement, manifest as
necrotizing granulomatous vasculitis and abdominal microaneu- Pulmonary
rysms, represent more severe forms of RV (Chen et al. 2002). Mild Pulmonary involvement in RA is common and can be due to the
cutaneous vasculitis may coexist with, or be followed by, more disease itself as well as to the therapies used to treat it. Lung disease
severe systemic necrotizing vasculitis (Chen et al. 2002; Lindsay has been reported to effect between 1% and 40% of patients with
et al. 1973). RA (Horton 2004). RA-associated interstitial lung disease (ILD) is
often subtle in onset, slowly progressive, and of unclear aetiology
Cutaneous (Horton 2004). The two most common patterns of ILD in patients
Cutaneous RV is the most common vasculitis reported in RA, seen with RA are usual interstitial pneumonia (UIP) and non-specific
in 75–89% of RV patients (Chen et al. 2002; Geirsson et al. 1987; interstitial pneumonia (NSIP)(Vij and Strek 2013). The therapies
Scott et al. 1981c; Voskuyl et al. 1996; Watts et al. 1994). The cuta- commonly used to treat RA, such as methotrexate and less com-
neous findings reported in RV include ulcers, petechiae or palpable monly leflunomide, can cause drug-induced pneumonitis and
purpura, digital infarcts including innocuous nail-fold infarcts, and are an important consideration in the differential diagnosis of
gangrene (Sayah and English 2005). Ulcers of the lower extremi- RA-associated ILD (Vij and Strek 2013). Diffuse alveolar haemor-
ties have been well described in up to 38% of subjects with RV and rhage (DAH) due to pulmonary capillaritis has been documented
are typically acute in onset, painful, punched-out lesions occurring in a patient with RA and mixed connective tissue disease (MCTD),
along the lateral malleolus or pretibial region. Other less common and in two RA patients where the onset of DAH was thought to be
cutaneous manifestations include non-specific maculopapular related to treatment with etanercept and leflunomide (Carloni et al.
or nodular erythema, haemorrhagic blisters, livedo reticularis, 2008; Khaja et al. 2012; Schwarz et al. 1998). The first line of treat-
erythema elevatum diutinum, and atrophie blanche (Sayah and ment for RA-associated lung disease is prednisone and removal of
English 2005). Among a study of 11 subjects with RV, all but one any possible offending agent (Vij and Strek 2013).
had coexistence of different forms of cutaneous vasculitis (Chen
et al. 2002).
Cardiac and peripheral vascular
Cardiac vasculitis affecting medium and small intramyocardial
Neurological arteries has been described in up to 20% of autopsy cases (Bonfiglio
The most common extracutaneous manifestation of RV is a periph- and Atwater 1969). This may lead to patchy myocardial necrosis
eral, non-compressive neuropathy. Mononeuritis multiplex and due to microinfarction or ischaemia (Bonfiglio and Atwater 1969;
distal symmetric sensory or sensorimotor neuropathies occur Cathcart and Spodick 1962; Sokoloff 1953). A non-occlusive vascu-
in up to 50% of subjects with RV (Bacon and Carruthers 1995; litis of the epicardial vessels has also been described (Morris et al.
Puechal et al. 1995; Schneider et al. 1985). In a study of 32 subjects 1986; Voyles et al. 1980).
with biopsy-confirmed RV of the peripheral nerves, morphologi- Aortitis of the thoracic and abdominal aorta or both has been
cal analysis revealed epi- or perineural necrotizing vasculitis, with described in RV patients (Gravallese et al. 1989). More recently,
axonal degeneration in the majority of the nerve fibres (Puechal aortic inflammation diagnosed by 18F-fluorodeoxyglucose posi-
et al. 1995). In this study, the coexistence of cutaneous vasculitis, a tron emission tomography (PET) imaging, thought to represent
multifocal neuropathy, and/or a depressed C4 level were associated subclinical vasculitis, was found in all 17 patients studied with
with increased mortality (Puechal et al. 1995). Vasculitis in the CNS severe RA but who were without clinical cardiovascular dis-
is uncommon in RA and in published cases it is typically described ease (Maki-Petaja et  al. 2012). Treatment of these patients with
in patients with seropositive, long-standing disease often with evi- anti-TNF-α therapy for 8 weeks resulted in reduced inflammation
dence of other extra-articular involvement (Guadalupe Loya-de la along the entire aorta, suggesting that treatment of RA results in
Cerda et al. 2013; Kim et al. 1982; Spath et al. 2012). The published improvement of subclinical aortic vasculitis (Maki-Petaja et  al.
cases of CNS RV have reported either the presence of: (1) rheuma- 2012). Mesenteric vasculitis is rare in RA but has been reported
toid nodules in the CNS, (2) meningeal vasculitis, or (3) cerebral and is associated with significant mortality (Chen et  al. 2002;
vasculitis (Bathon et al. 1989; Guadalupe Loya-de la Cerda et al. Pagnoux et al. 2005). This should be suspected in RV patients with
2013; Kim et al. 1982; Spath et al. 2012). CNS vasculitis is char- skin or peripheral nerve involvement who develop acute abdomi-
acterized microscopically by lymphocyte infiltration and fibrinoid nal pain, colonic ulcerations, or appendicitis (Chen et  al. 2002;
deposition and necrosis of the vessel wall layers causing the typi- Nagahama et al. 2000; Pagnoux et al. 2005; Scott and Bacon 1983;
cal angiographic findings of beading and stenosis of the involved van Laar et al. 1998). Necrotizing vasculitis of renal vessels is rare
arteries (Kurne et al. 2009; Spath et al. 2012). Clinical manifesta- (Bacons and Kitas 1994).
tions are varied and depend on the vessels involved and range from
headache, mental status changes, stroke, cranial nerve palsies, and Ocular
seizures (Chin and Latov 2005; Kurne et al. 2009; Spath et al. 2012). Scleritis/ keratitis has been reported to occur in up to 20% of
Methods used to diagnose CNS RV include MRI imaging (FLAIR patients with other manifestations of RV and has been described as
weighted), cerebral angiography, CSF fluid analysis, and biopsy a herald of diffuse vasculitis (Jifi-Bahlool et al. 1995).
446 SECTION 5 vasculitic diseases and syndromes and related disorders

Diagnosis and treatment Schneider et al. 1985; Scott et al. 1981b). The efficacy of anti-TNF-α
Ideally, the diagnosis of RV is made by histological examination of agents, as well as other biological agents for RV, is encouraging
a biopsy specimen of an affected organ (skin or nerve) or by angio- (Armstrong et al. 2005; den Broeder et al. 2001; Garcia-Porrua et al.
2006; Richter et al. 2000; van der Bijl et al. 2005).
gram. Biopsy of clinically unaffected muscle, rectum, and labial sal-
ivary glands have also shown promise in diagnosing RV (Flipo et al.
1994; Tribe et al. 1981; Voskuyl et al. 1998b; Voskuyl et al. 2003). Systemic lupus erythematosus
There is also an argument whether the diagnosis of RV can be made
according to classic clinical signs alone. This is demonstrated by
and vasculitis
the 1984 Scott and Bacon criteria for the diagnosis of RV, which Systemic lupus erythematosus (SLE) is a chronic, immune-mediated,
includes the presence of one or more of the following in a patient inflammatory, multisystemic disorder (Mills 1994). Involvement of
with RA:  (1)  mononeuritis multiplex or peripheral neuropathy; blood vessels in SLE is common, reported in 10–40% of patients
(2) peripheral gangrene; (3) biopsy evidence of acute necrotizing (d’Cruz 1998). Vascular lesions in SLE occur by a variety mecha-
arteritis plus systemic illness (e.g. fever, weight loss); (4) deep cuta- nisms including: (1) non-complicated vascular deposits of immune
neous ulcers or extra-articular disease (e.g. pleurisy, pericarditis, complex, (2) non-inflammatory necrotic vasculopathy, (3) throm-
scleritis) if associated with typical digital infarcts or biopsy evi- botic microangiopathy, and (4) true lupus vasculitis (Appel et al.
dence of vasculitis (Scott and Bacon 1984). More recently, Voskuyl 1994; Sung et al. 2002). The differentiation of the specific type of
and others sought to determine the optimal diagnostic strategy for vascular involvement in SLE can sometimes be difficult. A thor-
RA patients suspected of having RV (Voskuyl et al. 2003). In their ough investigation to determine the specific type of vascular lesion
cohort of 81 subjects with suspected RV, they found peripheral in SLE patients requires clinical expertise and often additional lab-
neuropathy or purpura/ petechiae were the most important clini- oratory studies, histopathological analysis, and/or vascular imag-
cal features to discriminate patients with RA with and without his- ing. The correct diagnosis of the specific vascular lesion will help
tological proven RV (Voskuyl et al. 2003). The presence of a high initiate appropriate treatment, based on the operative mechanisms
number of recent onset (≤6 months) extra-articular RA manifesta- involved, as well as predict prognosis (Cieslik et al. 2008; Greisman
tions, an elevated IgA RF level, and decreased C3 also were associ- et al. 1991; Somer 1993).
ated with increased probability of RV (Voskuyl et al. 2003).
There are no randomized controlled trials of the treatment of RV. Epidemiology
Similar to treatment of RA, the approach to treatment of those with Vasculitis as a manifestation of SLE is not rare. In a largely Caucasian
RV varies according to clinical severity. Several diseases may mimic population, the reported incidence of SLE vasculitis was 3.6/million
RV and should be considered prior to initiation of RV treatment. persons, nearly the same as one estimate of vasculitis due to infec-
These include cholesterol embolization syndrome (cutaneous tion (Watts et al. 1995). When patients with the diagnosis of SLE are
lesions and infarcts), peripheral vascular disease or venous insuf- examined as a group, most studies report the prevalence of vasculitis
ficiency (leg ulcers), diabetes mellitus (mononeuritis multiplex), ranging between 11% and 56% (Cardinali et al. 2000; Cieslik et al.
other forms of primary vasculitis such as polyarteritis nodosa, and 2008; Drenkard et al. 1997; Gonzalez-Gay and Garcia-Porrua 1999;
cryoglobulinaemia. Ramos-Casals et al. 2006; Vitali et al. 1992; Vlachoyiannopoulos
In its mildest form, simple nail-fold infarcts may be treated either et al. 1993). Cutaneous vasculitis involving small arteries and ven-
by adjustment of DMARD or biological therapy as one would for ules of the skin is most common, accounting for more than 60% of
routine synovitis, followed by close observation (Vollertsen and cases (Appel et al. 1994; Carlson and Chen 2007). Medium-vessel
Conn 1990). Leg ulcerations and leukocytoclastic vasculitis are involvement is less frequent, accounting for less than 10% of cases,
typically managed with aggressive topical therapy, moderate-dose manifested as mononeuritis multiplex in the majority of cases fol-
corticosteroids, and DMARD or biological therapy (McRorie et al. lowed by abdominal vasculitis (Drenkard et al. 1997; Ramos-Casals
1998). Other therapies with limited evidence in the treatment of et al. 2006). Lupus-associated large-vessel vasculitis is rare with a
vasculitic leg ulcers include topical nerve growth factor (not avail- few reported cases of lower extremity large-artery vasculitis and
able in the United States), and pinch skin grafting (Oien et al. 2001; coronary vasculitis (Drenkard et  al. 1997). Visceral involvement
Tuveri et al. 2000). portends increased morbidity and mortality (Drenkard et al. 1997;
More severe forms of RV, such as mononeuritis multiplex, digital Medina et al. 1997). Between 10% and 40% of lupus vasculitis cases
infarction, mesenteric vasculitis, or cutaneous vasculitis, that fail display both cutaneous and visceral involvement (Drenkard et al.
to respond to more conservative measures require more aggressive 1997; Ramos-Casals et al. 2006).
therapy. Standard treatment includes high-dose pulse or continu- In a large cohort of SLE subjects (n=667) described by Drenkard
ous cyclophosphamide along with corticosteroids (Scott and Bacon et al., patients with vasculitis were more commonly male, had onset
1984). If remission is maintained for 9–12 months then cyclophos- of disease at an earlier age, and had a longer duration of disease
phamide can be slowly tapered (Sundy et al. 2004). Methotrexate than those without vasculitis (Drenkard et al. 1997). Clinical fea-
(MTX) by intrathecal administration has been reported to be effec- tures associated with the presence of vasculitis included Raynaud’s
tive in CNS vasculitis complicating RA (Bathon et al. 1989). Success phenomenon, APL syndrome, neuropsychiatric lupus, myocardi-
using MTX by the oral route has been reported in one case (Ohno tis, serositis, and lymphopenia (Drenkard et al. 1997). In a large
et al. 1994). Other therapies used for RV, either alone or in com- Spanish cohort of SLE subjects, patients with vasculitis had a sig-
bination with corticosteroids, include ciclosporin, azathioprine, nificantly higher prevalence of livedo reticularis, higher SLE dis-
leflunomide, anti-TNF-α therapy, rituximab, intravenous immuno- ease activity, anaemia, a higher ESR, and presence of anti-La/SS-B
globulin (IVIg), and plasma exchange (Axson 1979; Heurkens et al. antibodies, compared to those without vasculitis (Ramos-Casals
1991; Jaffe and Smith 1968; Knab et al. 2005; Nicholls et al. 1973; et al. 2006).
CHAPTER 33 vasculitis in primary connective tissue diseases 447

Pathogenesis
Vasculitis in SLE is thought to be mediated by IC deposition in
vessel walls, leading to an inflammatory cascade through comple-
ment activation (Ansari et al. 1986; Fauci et al. 1978). Most biopsy
specimens of vasculitic lesions from SLE patients demonstrate
deposits of IgG, components of complement, and fibrin deposi-
tion. Complement components, including the C5b-9 complex,
C1q, and C5a, have been reported to play a role in SLE vasculitis
(d’Cruz 1998; Meroni et al. 2003). Cytokine ‘priming’ and autoanti-
body stimulation of the endothelium may also play roles in vascular
activation (Alexander et al. 1985; Belmont et al. 1996). In addition
to IC and cytokine-driven endothelium activation, antiendothe-
lial cell antibodies (AECAs) have been proposed as ‘endothelial
activators’ in this context and several investigators have noted
an increased occurrence of AECAs in SLE patients with vasculi-
tis (Alarcon-Segovia et  al. 1992; d’Cruz 1998; d’Cruz et  al. 1997;
Li et al. 1996). Two groups have noted that antiphospholipid anti-
bodies (APLs) tend to occur along with AECAs in SLE patients
with vasculitis and therefore a causal role for AECAs in vasculitis
remains speculative (Drenkard et al. 1997; Navarro et al. 1997).
There is a well-established correlation between cutaneous vas-
culitis (as well as subacute cutaneous lupus) and the presence of
anti-SSA/Ro antibodies (Lee and Norris 1989; Simmons-O’Brien
et al. 1995). Others have reported associations between cutaneous
vasculitis and anti-Smith, anti-SSB/La, and anti-dsDNA antibodies
(Beaufils et al. 1983; Ramos-Casals et al. 2006; Tikly et al. 1996).
Anti-dsDNA antibodies possess antiendothelial cell activity dem-
Fig. 33.2 Extensive, rapidly developing, necrosis due to rheumatoid vasculitis in
onstrated by increased synthesis of IL-1 and IL-6 by the endothelial a 60-year-old woman, who subsequently died with infl ammation in the coronary
cells (Praprotnik et al. 2001). Increased soluble forms of adhesion arteries as well. (Courtesy of Dr Gene Ball.)
molecules such as E-selectin, and ICAM-1, as well as IL-6, are
increased in the plasma of SLE patients with vasculitis, providing
indirect evidence of endothelial cell activation (Egerer et al. 2000). purpura is also common (~25%) followed by ischaemic lesions or
A considerable literature exists regarding the existence and clinical ulcers (~14%), urticarial lesions (~5–10%), nodular lesions (~5%),
significance of antineutrophil cytoplasmic antibody (ANCA) in SLE and a combination of lesions are found in approximately 30% of
(Sen and Isenberg 2003). Approximately 15–20% of SLE patients patients (Drenkard et al. 1997; Ramos-Casals et al. 2006). The most
have ANCA antibodies, usually perinuclear ANCA (pANCA) (Sen frequent location of the lesions were the lower extremities (42%),
and Isenberg 2003). There appears to be a link between pANCA lev- hands (40%), upper extremities (20%), trunk (5%), and face (3%)
els and disease activity in some patients; however, there is no consen- (Ramos-Casals et al. 2006).
sus regarding a correlation between ANCA levels and the presence Leukocytoclastic vasculitis is the most common pattern reported
of vasculitis (Chin et al. 2000; Lee and Lawton 1992; Nishiya et al. on biopsy with classic findings of fibrinoid changes in the vessel
1997; Schnabel et al. 1995; Sen and Isenberg 2003). walls composed of immunoglobulin, especially IgM, and comple-
ment (Calamia and Balabanova 2004). Diffuse neutrophilic and
Cutaneous mononuclear/ lymphocytic perivascular infiltrates have also been
The most common organ affected by vasculitis in SLE is the skin described, possibly associated with the age of the lesion (Calamia
Figure 33.2. Cutaneous vasculitis has diverse manifestations, pri- and Balabanova 2004). Lesions of small arterioles or venules high in
marily due to the variation in skin depth and intensity of vessel the dermal layer produce a petechial morphology but necrosis can
involvement. Most commonly affected are small vessels, mainly occur if the vasculitis is particularly robust (Lee and Norris 1989).
postcapillary venules, as seen in leukocytoclastic vasculitis and Not all petechial lesions represent vasculitis, as thrombocytopenia
urticarial vasculitis; less frequently, larger vessels in the deeper can also produce such lesions. Palpable purpura or urticaria result
dermis are involved producing cutaneous lesions similar to PAN from vascular involvement deeper in the dermis. Extensive damage
(Rothfield et  al. 2006). Although morphological features usu- at this level may result in bullous-type lesions. Such lesions almost
ally suggest vasculitis, punch biopsy or deeper tissue biopsy is the always are due to leukocytoclastic vasculitis (Laman and Provost
standard diagnostic method used to confirm the diagnosis. This is 1994; Tsuchida et al. 1994). Subcutaneous nodules akin to polyarte-
particularly important in SLE where causes of pseudovasculitis due ritis nodosa are the consequence of vasculitis at the dermal–epider-
to atherosclerotic disease or thrombotic disease are prevalent and mal junction. Ulcerations from these lesions may result if collateral
the treatments are sharply different from that of vasculitis. circulation is impaired (e.g. from coexistent atherosclerotic or
Erythematous punctate lesions on the fingertips and palms are thrombotic disease) (Martens et al. 1999; Sanchez et al. 1981).
the most common cutaneous lesion occurring in up to two-thirds of Urticarial lesions that persist for more than 24 hours may be a
patients (Drenkard et al. 1997; Ramos-Casals et al. 2006). Palpable vasculitis manifestation of SLE (Calamia and Balabanova 2004).
448 SECTION 5 vasculitic diseases and syndromes and related disorders

Urticarial lesions in SLE have been shown to be commonly associated and splenic infarcts have also been reported in an SLE patient with
with hypocomplementaemia (d’Cruz 1998). Hypocomplementaemic medium-vessel vasculitis (Ashouri et al. 2012).
urticarial vasculitis (HUV) is associated with anti-C1q antibodies Abdominal pain is common in SLE patients (~20%) and differ-
(Trendelenburg et al. 1999). Anti-C1q antibodies are also present in entiating benign from life-threatening, causes can be difficult (Lee
SLE; therefore, the distinction between these two disease entities can et al. 2002; Medina et al. 1997). It is also critical to consider and rule
sometimes be blurred (Trendelenburg et al. 1999). out non-SLE-related causes of abdominal pain, including pancrea-
Treatment of cutaneous vasculitis in SLE is tailored to the under- titis, cholecystitis, appendicitis, ovarian cysts, diverticular disease,
lying disease process. Antimalarials such as hydroxychloroquine and others. Antiphospholipid antibody syndrome may also cause
may be effective in mild cases. Dapsone (100–200 mg/day) has vessel obstruction from thrombosis and it also can be difficult to
also been utilized (Holtman et al. 1990). Reported side-effects of distinguish from vasculitis (Asherson et al. 1985; Asherson et al.
dapsone (haemolytic anaemia, agranulocytosis, and neuropathy) 1986; Provenzale and Ortel 1995).
are not trivial and all patients should have a documented normal In SLE patients with active disease, gastrointestinal vasculitis as
complete blood count and G6PD level prior to starting dapsone. the underlying cause of acute abdominal pain is high, approaching
Thalidomide has been considered for use in carefully selected 50% (Medina et al. 1997). Conversely, in patients with inactive SLE,
patients (Godfrey et  al. 1998). Glucocorticoids are often used as the overall incidence of mesenteric vasculitis is less than 1% (Sultan
‘bridge’ therapy to control moderate to severe disease while waiting et al. 1999).
for a more potent cytotoxic agent such as azathioprine to produce Although arteriography is considered the gold standard test for
clinical benefit (Callen et al. 1991). Cyclophosphamide is effective, diagnosing mesenteric or abdominal vasculitis, abdominal CT is
but should be reserved for severe, refractory cases of cutaneous vas- the most useful tool due to its advantages in terms of wide avail-
culitis (Nishijima et al. 1999). Mycophenolate mofetil (MMF) has ability and promptness of result (Ko et al. 1997; Zizic et al. 1982).
been used for several dermatological conditions and for other man- Characteristic CT findings include target signs, comb signs, and
ifestations of SLE, but its role in SLE cutaneous vasculitis remains to other associated findings (Ashouri et  al. 2012; Ju et  al. 2009).
be defined (Goyal and Nousari 2001; Liu and Mackool 2003; Pisoni High-dose intravenous methylprednisolone and cyclophospha-
et al. 2005). Similarly, rituximab, or tocilizumab have shown util- mide are the usual treatments (Grimbacher et al. 1998; Laing 1988).
ity in case reports of severe or treatment-resistant cutaneous lupus
vasculitis (Makol et al. 2012; Van den et al. 2005; Weide et al. 2003). Pulmonary
Pulmonary involvement occurs in approximately 60% of patients
Visceral with SLE during the course of the disease (Paran et  al. 2004).
Extracutaneous vasculitis is observed in 12 to 20% of subjects According to an autopsy study, pulmonary vasculitis is rare, occur-
with lupus (Drenkard et  al. 1997; Ramos-Casals et  al. 2006). ring in less than 2% of patients (Haupt et  al. 1981). Pulmonary
Mononeuritis multiplex accounts for over half of the cases of vasculitis can occur with or without diffuse alveolar haemor-
extracutaneous SLE vasculitis (see Section Neurological) followed rhage (DAH) (Zamora et al. 1997). Pulmonary haemorrhage has
by vasculitis of the abdominal vasculature (Drenkard et al. 1997; been the initial clinical event in SLE in 10–20% of the few cases
Ramos-Casals et al. 2006). Mesenteric vasculitis typically involving where DAH was secondary to SLE (Schwab et  al. 1993; Zamora
the ileum or the colon is most commonly described with a reported et al. 1997). Most patients with DAH have active SLE manifested
prevalence of 0.2 to 9.7% among patients with SLE and in 30–65% by lupus-related nephritis or neuropsychiatric involvement, and
of those who present with an acute abdomen (Ju et al. 2009; Lee a diagnosis of SLE of less than 5 years (Calamia and Balabanova
et  al. 2002). The clinical presentation is usually an abrupt onset 2004; Kwok et al. 2011; Liu et al. 1998; Virdi et al. 2012). DAH may
of severe, postprandial, diffuse abdominal pain, frequently with have an occult presentation with patients initially reporting abrupt
vomiting and fever, which may be associated with haematochezia onset of dyspnoea (Blanche et al. 1996). Haemoptysis is a present-
or guaiac-positive stool. Mesenteric vasculitis involves small ves- ing complaint less than 50% of the time. Fever, hypoxaemia, patchy
sels of the bowel submucosa (Ashouri et al. 2012; Ju et al. 2009). infiltrates on chest radiographs, and decreasing haematocrit should
The pathological process consists of immune-complex deposition raise clinical suspicion (Zamora et al. 1997). The latter feature dis-
and complement activation with subsequent submucosal oedema, tinguishes DAH from the otherwise similar presentation of acute
leukocytoclastic vasculitis, and thrombus formation (Ju et al. 2009). lupus pneumonitis. An increased single breath carbon monoxide
An accurate and prompt diagnosis is critical to prevent the potential diffusion determination further suggests DAH (Greening and
catastrophic sequelae of bowel ischaemia, necrosis, and perforation Hughes 1981). Bloody fluid on bronchoalveolar lavage in these
(Ashouri et al. 2012; Ju et al. 2009). In one report of 13 SLE subjects circumstances strongly supports DAH (Leatherman et  al. 1984).
with mesenteric vasculitis, the mortality approached 50%, particu- Radiographic features of DAH are indistinguishable from pneu-
larly if the patient presented with an acute abdomen or if surgery monia and pulmonary oedema; therefore, aggressive procure-
was delayed (Medina et al. 1997). Other less frequent sites of vis- ment of material for culture as well as broad antibiotic/ antifungal/
ceral, small-vessel vasculitic involvement include the pancreas, antiviral coverage should be considered (Zamora et al. 1997). The
gallbladder, urinary bladder, liver, and uterus (Alarcon-Segovia histological lesions seen in DAH are those of capillaritis. An associ-
et al. 1984; Bando et al. 2003; Feriozzi et al. 1997; Karrar et al. 2001; ated lesion is ‘microangiitis,’ where capillaries as well as arterioles
Matsumoto et al. 1992; Medina et al. 1997; Newbold et al. 1987). and small muscular arteries are involved (Myers and Katzenstein
Abdominal medium- and large-vessel vasculitis is rare in SLE and 1986; Santos-Ocampo et al. 2000; Zamora et al. 1997). Pulmonary
limited to case reports. Necrotizing medium-vessel vasculitis simi- manifestations of antiphospholipid antibody syndrome (APS)
lar to PAN has been reported in gallbladders and hepatic vessels of can be strikingly similar to SLE-related pulmonary vasculitis and
SLE patients (Matsumoto et al. 2000; Swanepoel et al. 1983). Renal DAH (Nguyen et al. 2005). Apart from pulmonary embolism and
CHAPTER 33 vasculitis in primary connective tissue diseases 449

pulmonary hypertension, catastrophic APS can cause diffuse pul- dysfunction in SLE may be secondary to other causes such as coro-
monary infiltrates, capillaritis, and diffuse alveolar haemorrhage nary artery disease due to premature atherosclerosis, hypertension,
(Nguyen et al. 2005). Fortunately, pulmonary APS and SLE vascu- valvular disease, or toxicity to medications (e.g. cyclophosphamide,
litis are reported to exhibit a similar clinical course and response to chloroquine) (Doria et al. 2005; Kao and Manzi 2002).
treatment (Nguyen et al. 2005). Coronary artery disease occurs in 6 to 10% of SLE patients and
The mortality in SLE patients with DAH remains high, greater is most often due to atherosclerosis (Petri et al. 1992; Toloza et al.
than 50% in some studies (Kwok et  al. 2011; Shen et  al. 2010; 2004). The risk of developing coronary artery disease is four to
Virdi et al. 2012; Zamora et al. 1997). Early diagnosis and aggres- eight times higher than controls and the risk of myocardial infarc-
sive therapy has been shown to improve survival considerably tion is increased 50-fold in young women with SLE (Manzi et al.
(Santos-Ocampo et al. 2000; Schwab et al. 1993). Multiple inter- 1997). Current knowledge indicates that atherosclerosis is an
ventions have been utilized with pulse-dose corticosteroids being active inflammatory and immune-mediated process. Therefore,
the mainstay of therapy, which is typically combined with cyclo- the processes characteristic of SLE can undoubtedly contribute to
phosphamide with or without plasmapheresis, depending on the the accelerated vascular disease seen in these patients (Kao et al.
severity of the disease (Kwok et al. 2011; Shen et al. 2010; Virdi et al. 2003). Coronary arteritis is rare and may be exacerbated by coex-
2012). SLE patients with DAH who survive are at increased risk istent atherosclerosis (Moder et al. 1999). Differentiating coronary
of future episodes of DAH as well as other severe manifestations arteritis from lesions due to atherosclerosis may be difficult but
of SLE; therefore, they have diminished life expectancy compared important for therapeutic decisions. The presence of aneurysmal
with SLE patients without DAH (Zamora et al. 1997). dilatation and rapid progression of lesions on arteriography sup-
Vasculitis in SLE may also involve the upper airway, albeit rarely. port vasculitis (Heibel et  al. 1976; Nobrega et  al. 1996). Active
Ulcerations of the larynx as well as overt necrotizing vasculitis SLE and a short duration of symptoms should also heighten clini-
of the larynx, tracheal structures, and large bronchi have been cal suspicion for coronary arteritis (Badui et al. 1985; Doria et al.
reported (Boumpas et al. 1995; Teitel et al. 1992). 2005). In cases of coronary vasculitis, corticosteroids at high doses
are recommended. When atherosclerotic coronary artery disease is
Cardiac and peripheral vascular suspected, diagnosis and treatment should follow current recom-
Pericarditis is the most common cardiac manifestation of SLE, mended guidelines. Additionally, a concerted effort to modify both
found in up to 60% of cases examined post-mortem (Doherty et al. traditional risk factors and immune and inflammatory processes
1988). Echocardiographic studies show pericardial abnormalities in SLE patients is critical in preventing premature cardiovascular
in 11 to 54% of patients with SLE (Doria et al. 2005). Pericardial disease (Kao et al. 2003).
disease appears more frequently at disease onset or during relapses Large-vessel vasculitis indistinguishable from that of Takayasu’s
(Doria et al. 2005). Immune complex deposition in small vessels arteritis has been reported in SLE. Due to the lack of common patho-
of the pericardium is the underlying cause (Bidani et  al. 1980). logical and serological features between SLE vasculitis and Takayasu’s
Pericardial effusions vary in size, but complications such as tam- arteritis, it has been suggested that these ‘overlap’ cases represent
ponade, constrictive pericarditis, or purulent pericarditis are rare random occurrences of two different autoimmune diseases in one
(Doria et  al. 2005). Non-steroidal anti-inflammatory drugs or individual (Lorber et al. 1994). Most reported cases of large-vessel
low-dose corticosteroids are effective in mild, symptomatic peri- vasculitis have responded to moderate doses of oral prednisone (1
carditis; a moderate dose of oral glucocorticoids (prednisone 1mg/ mg/kg/day), while others have required the addition of cyclophos-
kg/day) is usually effective for more severe cases. In patients with phamide (Saxe and Altman 1992). Azathioprine and MMF are other
refractory or recurring pericarditis, chronic immunosuppression therapeutic options (Daina et al. 1999; Kao et al. 2003).
with methotrexate, azathioprine, MMF, or IVIg may be beneficial
(Doria et al. 2005). Invasive procedures such as pericardiocentesis Renal
or pericardial window are rarely required (Doria et al. 2005). Vasculitis involving renal vessels, including the specialized vas-
Valvular abnormalities in SLE are common. Libman–Sachs (ver- cular bed of the glomeruli, is at particular risk for immune com-
rucous) endocarditis is the most characteristic lesion; however, plex binding or deposition (Calamia and Balabanova 2004; Hricik
valvular thickening and regurgitation are more common than ver- et al. 1998). Histologically, the pattern of glomerular injury may
rucous endocarditis (Doria et al. 2005). Anatomic valvular lesions vary to include capillary proliferation, fibrinoid necrosis, or hya-
are observed in 18–74% of all patients with SLE and tend to occur line thrombi (Calamia and Balabanova 2004; Hricik et al. 1998).
in the mitral and aortic valves (Doria et  al. 2005; Moder 1999). The most common form of renal vasculitis is capillaritis and is
Actual valvulitis with vasculitis and resultant fibrinoid necrosis may the underlying vascular lesion in the proliferative forms of lupus
be seen on histological examination in the rare instances of severe nephritis (WHO classes III and IV). Less commonly, lesions remi-
valvular disease. Severe valvulitis is managed by valve replacement niscent of necrotizing vasculitis may involve larger renal arteries,
when clinically indicated. which is associated with severe renal disease (Bacon and Carruthers
Clinical myocarditis occurs in 7 to 10% of SLE patients (Bidani 1995). Glucocorticoids with MMF or cyclophosphamide have been
et al. 1980; Bulkley and Roberts 1975; Doria et al. 2005; Roberts and the cornerstones of therapy for renal vasculitis (Boumpas et  al.
High 1999). When severe, the condition presents as congestive heart 1995; Ginzler et al. 2005).
failure and the diagnosis is confirmed by the clinical context and
endomyocardial biopsy (Fairfax et al. 1988). Biopsy material shows Neurological
intravascular IC deposition with vasculitis and adjacent myocarditis. Neuropsychiatric SLE (NPSLE) affects 17 to 72% of patients and is
Treatment with moderate to high doses of glucocorticoids has been manifested by a wide variety of symptoms (Eber et al. 2005; West
effective in some cases (Fairfax et al. 1988). Importantly, myocardial 1994). In 1999, the ACR produced a standard nomenclature and
450 SECTION 5 vasculitic diseases and syndromes and related disorders

diagnostic criteria for 19 neuropsychiatric syndromes known to et al. 2001). The diagnosis should be established by nerve conduc-
occur in SLE (American College of Rheumatology (ACR) 1999). tion velocity assessment followed by nerve biopsy (usually the sural
The most frequent NPSLE symptoms include cognitive dysfunc- nerve) for histological confirmation. Non-specific axonal degenera-
tion, headache, mood disorder, cerebrovascular disease, seizures, tion with demyelination is the most common finding, but necrotiz-
polyneuropathy, anxiety, and psychosis (Hanly and Harrison 2005). ing vasculitis of perineural vessels has been reported (Griffin 2001).
Three primary immunopathogenic mechanisms are most often The distinction may be important in terms of therapeutic approach
implicated in NPSLE: vasculopathy of primarily small intracranial as IVIg may be the preferred treatment for the former, while com-
vessels, autoantibody production (e.g. antineuronal, antiribosomal, bination prednisone/ cytotoxic agent therapy is indicated for the
antiphospholipid), and the generation of inflammatory mediators latter (Enevoldson and Wiles 1991; Lesprit et al. 1996).
(e.g. IL-6, IL-10, IL-2, IL-8, MMP-9)(Hanly 2005). Vasculitis is an Management of NPSLE needs to be tailored according to the
uncommon cause of NPSLE, accounting for less than 10% of cases individual patient and the manifestation of the disease. A EULAR
(Drenkard et al. 1997). Smaller-vessel involvement and occlusions taskforce has recently published recommendations for treatment of
are responsible for the majority of signs and symptoms of NPSLE NPSLE (Bertsias et al. 2010). Initial attention should be focused on
(Boumpas et al. 1995). Large-vessel occlusive disease is rare and treating any non-SLE-related factors, including infection, hyperten-
typically presents as a stroke or intracerebral haemorrhage and car- sion, or metabolic abnormalities. Symptomatic therapy with anticon-
ries a relatively poor prognosis (Mitsias and Levine 1994; Weiner vulsants, antidepressants, or antipsychotic medications should be
and Allen 1991). Arterial thrombus, dissection, fibromuscular considered (Hanly and Harrison 2005). Immunosuppressive therapy
dysplasia, and vasculitis should be considered with large-vessel with high-dose or pulse glucocorticoids is the main stay of treatment
involvement (Mitsias and Levine 1994). in the acute stage of active NPSLE. Concomitant use of intravenous
In diagnosing NPSLE, cerebrospinal fluid studies are impor- cyclophosphamide has been shown to have a higher response rate
tant to exclude infection; although cell count, total protein, and and a more prolonged treatment response in severe cases of NPSLE
oligoclonal band determination are not reliable indicators of (Barile-Fabris et al. 2005; Boumpas et al. 1995; Bertsias et al. 2010).
NPSLE (West 1994). Studies have demonstrated an association Azathioprine, MMF, rituximab, IVIg, and plasma exchange are
of antiribosomal-P antibodies and psychiatric manifestations of other therapeutic options (Bertsias et al. 2010; Boumpas et al. 1995;
NPSLE (Eber et  al. 2005). Imaging studies are more likely to be Damato et al. 2011; Hanly and Harrison 2005; Papadaki et al. 2006).
revealing in patients with focal neurological deficits rather than
diffuse encephalopathy (Boumpas et al. 1995). MRI is more likely Ophthalmological
to show small-vessel vasculopathy, but asymptomatic periventricu- Systemic lupus erythematosus can affect every structure in the eye
lar white matter changes are found in a large number of patients and ocular inflammation can antedate the diagnosis of the disease.
with SLE (Kent et al. 1994; Rovaris et al. 2000). MR arteriography About one-third of patients with SLE have ocular manifestations
(MRA), computed tomography (CT), and radiocontrast arteriogra- during the course of their disease (Palejwala et al. 2012). The most
phy are useful only in detecting the rare instances of larger-vessel common ocular manifestation is keratoconjunctivitis sicca (Peponis
vasculitis (Boumpas et al. 1995; Weiner and Allen 1991). Positron et al. 2006). Recurrent episcleritis has been reported in up to 28% of
emission spectroscopy has reportedly proven useful in demonstrat- SLE patients and has a clinically benign course (Frith et al. 1990).
ing diminished CNS blood flow in SLE vasculitis, but this finding Less frequently, scleritis occurs in SLE and cause severe ocular
awaits confirmation in larger series (Hanly 1998). morbidity (Peponis et al. 2006). SLE retinopathy has been reported
Transverse myelitis consists of segmental spinal cord compromise in up to 25% and is one of the most common vision-threatening
due to vascular occlusion and occurs in SLE due to either throm- complications of the disease (Palejwala et al. 2012). In most cases
bosis or vasculitis. A strong association between transverse myelitis it is due to an immune complex-mediated vasculopathy (Peponis
and APLs exists in patients with SLE (Lavalle et al. 1990). Patients et al. 2006). Abnormalities in the retinal vasculature include vas-
present with hemiparesis, often complicated by urinary sphincter cular sheathing, arterial narrowing, capillary and venous dila-
dysfunction. The diagnosis is confirmed by MRI, which shows tion, tortuosity, and microaneurysms (Peponis et al. 2006). Severe
increased cord diameter as well as increased T1 and T2-weighted vaso-occlusive retinopathy due to vasculitis or associated with
signal in the affected cord segments (Provenzale et  al. 1994). antiphospholipid antibodies is rare; however, unlike the immune
When recognized early, myelitis has been successfully treated with complex-mediated retinopathy, it carries a high risk of ensuing
high-dose methylprednisolone and cyclophosphamide (Barile and visual loss (Au and O’Day 2004; Klinkhoff et al. 1986). A strong
Lavalle 1992; Harisdangkul et al. 1995). Treatment should not be correlation exists between presence of vaso-occlusive retinopathy
abandoned early, as there are reports of response after months of and involvement of the CNS vasculature (Jabs et al. 1986; Klinkhoff
treatment. et al. 1986). Orbital vasculitis is a rare manifestation of SLE, which
Peripheral neuropathy has been reported in up to 28% of SLE can lead to non-perfusion of the globe and extraocular muscles,
patients (Ainiala et  al. 2001; Brey et  al. 2002; Hanly et  al. 2004; and can cause irreversible vision loss (Palejwala et al. 2012; Stavrou
Hanly and Harrison 2005). It is usually a sensorimotor neuropa- et al. 2002). Treatment of retinal vasculitis is with systemic gluco-
thy which is mild, persistent, and frequently occurs independently corticoids, but ischaemic retinal vasculopathy may require anti-
of other disease characteristics (Hanly and Harrison 2005; Omdal coagulation. Other treatments that have been reported in severe
et  al. 2001). Other less frequent forms of peripheral neuropathy disease include plasmapheresis, plasma exchange, rituximab, and
include mononeuritis multiplex, autonomic neuropathies, cranial MMF (Damato et al. 2011; Papadaki et al. 2006). Several reports
neuropathies, plexopathy, and Guillain–Barré syndrome (Bloch indicate SLE retinopathy both parallels systemic disease activ-
et al. 1979; Hanly and Harrison 2005; Liote and Osterland 1994; ity and is a marker of poor prognosis for survival (Klinkhoff et al.
Martinez-Taboada et  al. 1996; Straub et  al. 1996; van Laarhoven 1986; Stafford-Brady et al. 1988). SLE patients should, therefore,
CHAPTER 33 vasculitis in primary connective tissue diseases 451

have regular ophthalmological examinations, even if they are not SS patients without cutaneous vasculitis (Alexander et  al. 1983;
on antimalarial therapy. Garcia-Carrasco et al. 2002; Ramos-Casals et al. 2002; Ramos-Casals
In summary, vasculitis has widespread and diverse manifesta- et al. 2004; Ramos-Casals et al. 2008; Tsokos et al. 1987). The lower
tions in SLE. It is most commonly recognized in the skin but may extremities are the most common site of SS-related cutaneous vas-
be present in a number of organs and its presence is associated with culitis and in about 50% of the patients it will be a single episode
increased morbidity and mortality. Causes of pseudovasculitis, (Ramos-Casals et al. 2004). Treatment of cutaneous vasculitis typi-
including APS, are also common in SLE and should be considered cally involves corticosteroids in low to moderate doses. Other drugs,
in the evaluation of ischaemic events. The degree of disease sever- such as hydroxychloroquine, dapsone, IVIg, methotrexate, lefluno-
ity dictates therapy, ranging from careful observation, to high-dose mide, azathioprine, and cyclophosphamide, can be used alone or
glucocorticoids and cytotoxic agents. The increased availability in combination with corticosteroids depending on disease sever-
and research utilizing targeted immunotherapy holds promise for ity (Fox 2005; Price et al. 1998; Skopouli et al. 1996; van Woerkom
improved treatment and understanding of disease pathogenesis et al. 2007).
(Calamia and Balabanova 2004).
Neurological
Sjögren’s syndrome and vasculitis The nervous system can be globally involved in SS, observed
in approximately 20–25% of cases (Delalande et  al. 2004).
Epidemiology Peripheral nervous system involvement is most common, affect-
Sjögren’s syndrome (SS) is an autoimmune inflammatory disease ing 10–20% of cases (Garcia-Carrasco et  al. 2002; Gono et  al.
that primarily involves the exocrine glands. The disorder is charac- 2011; Govoni et al. 1999; Ramos-Casals et al. 2002). Peripheral
terized by lymphocytic infiltration of lacrimal and salivary glands sensory neuropathies are the most common manifestation (Chai
resulting in their functional impairment, leading to characteristic and Logigian 2010; Delalande et al. 2004; Gono et al. 2011; Grant
symptoms of xerostomia and keratoconjunctivitis sicca. The pres- et al. 1997). Small-fibre neuropathy is being increasingly recog-
ence of autoantibodies, particularly anti-Ro (SSA) and anti-La nized in patients with SS characterized by neuropathic pain, with
(SSB), are also part of the classification criteria for Sjögren’s syn- loss of pinprick or temperature sensation, but normal tendon
drome (Vitali et al. 2002). Sjögren’s syndrome is the second most reflexes and nerve conduction studies (Gono et  al. 2011). It is
common autoimmune rheumatic disease and can present either usually mild and is diagnosed by skin biopsy, which demonstrates
alone (primary Sjögren’s syndrome (pSS)) or in the context of either reduced or abnormal intraepidermal nerve fibres (Gono
another connective tissue disease (secondary Sjögren’s syndrome et al. 2011). Sensory ataxic neuropathy is another pure sensory
(sSS)), most commonly RA or SLE (Peri et al. 2012). Systemic or neuropathy described in patients with pSS, which is manifested
extraglandular manifestations occur in 30 to 70% of patients with as sensory ataxia of the limbs due to a T-cell mediated dorsal
pSS (Garcia-Carrasco et al. 2002; Seror et al. 2012; Skopouli et al. root ganglionopathy, often with a peripheral nerve vasculitis
2000; ter Borg et  al. 2011). Vasculitis is a common extraglandu- (Mori et  al. 2005). Cranial neuropathies, including trigeminal
lar manifestation of pSS with biopsy-proven vasculitis reported in sensory neuropathy, are well described in pSS (Delalande et al.
approximately 13% of patients (Scofield 2011; Tsokos et al. 1987). 2004; Gono et al. 2011; Grant et al. 1997). Multiple mononeu-
Vasculitis in SS can affect arteries of different sizes, veins, and capil- ropathy, although far less common, has been well recognized in
laries in various organs, with cutaneous small-vessel vasculitis and pSS. It frequently occurs in patients with more severe systemic
vasculitis involving the peripheral nerves being the most common disease and presents with an acute or subacute onset of par-
(Ramos-Casals et al. 2006). Several studies have described an asso- aesthesia, pain, and weakness in the distal extremities. It is fre-
ciation of vasculitis with the presence of anti-Ro (SSA) and anti-La quently diagnosed by a sural nerve biopsy (Delalande et al. 2004;
(SSB) antibodies (Alexander et al. 1983; Ramos-Casals et al. 2004). Fox 2005; Mori et  al. 2005). Autonomic neuropathy, including
abnormal pupillary responses (e.g. Adie’s pupils) and orthostatic
Cutaneous hypotension, have been reported in patients with pSS-associated
Cutaneous vasculitis has been reported in 9 to 32% of patients neuropathy (Griffin et al. 1990; Mori et al. 2005). Treatment for
with SS, with a predominance of small-vessel vasculitis (SVV) peripheral neuropathies in pSS consists mainly of corticoster-
(Garcia-Carrasco et al. 2002; Ramos-Casals et al. 2002; Ramos-Casals oids, which are typically effective for the multiple mononeu-
et al. 2004; Ramos-Casals et al. 2008). Cutaneous SVV can be further ropathies and the cranial neuropathies (Mori et  al. 2005). The
categorized, in order of frequency, as: (1) non-cryoglobulinaemic, small-fibre and sensory ataxic neuropathies have been reported
non-urticarial vasculitis presenting as palpable purpura in the lower to be more responsive to IVIg therapy rather than corticosteroids
extremities; (2) cryoglobulinaemic vasculitis manifesting as palpa- (Mori et al. 2005; Morozumi et al. 2009). Rituximab has shown
ble purpura primarily affecting the lower extremities; and (3) urti- clinical and biological efficacy in patients with pSS with vasculi-
carial vasculitis with erythematous, urticariform macules on the tis (and cryoglobulin) related peripheral neuropathy with a high
lower or upper extremities (Ramos-Casals et al. 2006). Cutaneous response rate (Mekinian et al. 2012a). A recent study found low
medium-vessel, necrotizing vasculitis is rare and typically pre- levels of vitamin D associated with increased risk for peripheral
sents as ischaemic/ ulcerative lesions (Ramos-Casals et  al. 2006). neuropathy (and lymphoma) in subjects with pSS (Agmon-Levin
Venous and arterial thrombotic lesions can also occur. SS patients et al. 2012). Therefore, checking vitamin D levels and vitamin D
with cutaneous vasculitis typically have more severe disease with repletion should be considered in patients with pSS and periph-
more extraglandular involvement, including a higher prevalence of eral neuropathy (Agmon-Levin et al. 2012).
hypocomplementaemia, anti-Ro and anti-La antibodies, anaemia, Central nervous system (CNS) involvement can be present in pSS
elevated ESR, and hypergammaglobulinaemia compared to those but the reported prevalence and type of involvement varies widely
452 SECTION 5 vasculitic diseases and syndromes and related disorders

from 10 to 60% (Delalande et al. 2004; Gono et al. 2011; Ramos-Casals subclinical (Alexander et al. 1983; Lindvall et al. 2002). Studies of
et al. 2008). Vasculitis affecting the spinal column is well reported in pSS patients who had normal muscle strength, normal serum cre-
the literature and typically presents as an acute or chronic myelopa- atinine kinase, and mild muscle pain often had abnormal muscle
thy (Delalande et al. 2004; Rogers et al. 2004; Williams et al. 2001). biopsies, which demonstrated perivascular inflammation with per-
Acute transverse myelitis in pSS carries a high mortality and requires imysial or endomysial infiltrates (Alexander et al. 1983; Lindvall
prompt, aggressive immunosuppressive treatment (Delalande et al. et al. 2002). Myopathy has also been shown to coexist with neu-
2004; Rogers et al. 2004). Chronic or progressive myelitis, often pre- ropathy in pSS (Vrethem et al. 1990).
senting as a progressive paraparesis, can mimic primary progressive
multiple sclerosis in patients with pSS (Delalande et al. 2004; Rogers Cardiac and pulmonary
et al. 2004). Less common spinal cord manifestations include lower Cardiopulmonary involvement can occur in pSS. The most common
motor neuron disease, neurogenic bladder, and Brown–Sequard syn- pulmonary symptom is cough, observed in up to 41% of patients,
drome (Delalande et al. 2004; Rogers et al. 2004). which is often a symptom of tracheobronchial sicca (Papiris et al.
Brain involvement in pSS can present either (1) focally, mimick- 1999). Pulmonary manifestations are among the most prevalent
ing a stroke, (2) diffusely, presenting as seizures, cognitive dysfunc- extraglandular complications in pSS, with reported prevalence vary-
tion, or encephalopathy, or (3) neuromyelitis optica (Delalande et al. ing from 9 to 75%, depending on the methods of detection and
2004). There have been several reports examining the relationship patient selection (Constantopoulos et al. 1985; Papiris et al. 1999;
between neuromyelitis optica, characterized by optic neuritis, trans- Wright et al. 2003). In a recent study of 216 patients with pSS, clini-
verse myelitis and autoantibodies to aquaporin 4, with pSS and other cal lung involvement was found in 27% of patients and abnormal
CTDs. Interestingly, aquaporin-4 is expressed on salivary glands as HRCTs were found in 23% of patients (Palm et al. 2013). The most
well as on perivascular astrocytes (Gresz et al. 2001; Lennon et al. common HRCT findings included reticular pattern (44%), cysts
2004). Anti-aquaporin 4 appears to be as highly specific for neuro- (42%), air trapping (22%), and ground-glass attenuation (20%)
myelitis optica in patients with pSS and other CTDs as it is in those (Palm et al. 2013). Histological findings in patients with pSS from
without an underlying CTD. It should, therefore, be suspected in transbronchial biopsy typically demonstrate peribronchial and/or
pSS patients who present with recurrent optic neuritis and longi- peribronchiolar mononuclear inflammation, interstitial inflamma-
tudinally extensive transverse myelitis and treated as such (Jarius tion, or lymphocytic aveolitis rather than acute vasculitis (Dalavanga
et al. 2011; Lennon et al. 2004; Wandinger et al. 2010). Diagnosis of et al. 1991; Papiris et al. 1999). Several studies have now shown an
suspected CNS SS begins with a high index of suspicion and an MRI increased mortality risk in pSS patients with pulmonary involvement
of the brain and/or spinal cord. Spinal cord MRI typically reveals (Dalavanga et al. 2006; Palm et al. 2013; Papiris et al. 1999). Other
T2-weighted hyperintensities, which can involve large, longitudinal SS-related pulmonary disorders include pulmonary hypertension,
segments of the spinal cord in cases of acute myelopathy (Delalande hypersensitivity pneumonitis, toxic effects of drugs (i.e. methotrex-
et al. 2004). In the brain, multiple areas of increased signal inten- ate), and opportunistic infections in patients on immunosuppressive
sity in the periventricular and subcortical white matter are seen on drugs (Fox 2005; Kim et al. 2002). Pericarditis, pulmonary hyperten-
FLAIR and T2-weighted MRI images (Morgen et al. 2004; Tzarouchi sion, and cardiovascular tests consistent with an autonomic neuropa-
et al. 2011). The white matter lesions in pSS can appear to be similar thy can also occur in SS (Fox 2005; Gyongyosi et al. 1996).
to lesions found in multiple sclerosis (MS). Some differences that
can help distinguish CNS SS from MS include: (1) the localization of Renal
MRI lesions (rare corpus callosum lesions, involvement of the basal Renal involvement in SS occurs in approximately 30% of subjects;
ganglia), (2) associated peripheral nerve involvement (common in however, most often it is unrecognized (Bossini et  al. 2001). The
SS), (3) lower prevalence of oligoclonal bands in the spinal fluid, and majority of patients have subclinical renal tubular dysfunction, due
(4) the presence of sicca symptoms or extraglandular features of SS to tubulointerstitial nephritis. A frank distal tubular acidosis can be
(Delalande et al. 2004). Cutaneous vasculitis is frequently found in found in approximately 5% of subjects who present with hypokalae-
patients with neurological SS, as are laboratory abnormalities such mia, hyperchloraemic acidosis, nephrolithiasis, and rarely hypokalae-
as lymphopenia, hypergammaglobulinaemia, ANA, anti-SSA/Ro mic paralysis (Bossini et al. 2001). Histological examination typically
antibodies, and cryoglobulins (Delalande et al. 2004). reveals a focal or diffuse lymphocytic infiltrate in the interstitial tissue
Severe disability can result rapidly from CNS SS, therefore, inten- (Bossini et al. 2001). Interstitial fibrosis and tubular atrophy are asso-
sive and early treatment is recommended. In patients with mye- ciated with chronic disease (Bossini et al. 2001). Immune-mediated
lopathy, i.v. corticosteroids plus i.v. cyclophosphamide is effective glomerulonephritis is rare in SS, occurring in less than 5% of patients.
(Alexander 1992; Williams et al. 2001). Unlike the positive results The most frequent types of glomerular involvement include mem-
of rituximab in SS-associated peripheral neuropathies, rituximab branoproliferative, membranous, or mesangioproliferative glomerlu-
was not shown to be an effective treatment for CNS manifestations lonephropathies (Bossini et al. 2001; Goules et al. 2000; Moutsopoulos
(Mekinian et al. 2012a; Mekinian et al. 2012b). Other immunosup- et al. 1978). Subclinical tubular abnormalities require no treatment.
pressive treatments have been tried with variable efficacy including Mild to moderate renal tubular acidosis requires potassium replace-
azathioprine, chlorambucil, and plasmapheresis (Alexander 1992; ment and alkalinization with potassium citrate. For cases resistant to
Hermisson et al. 2002; Konttinen et al. 1987; Williams et al. 2001; replacement therapy or those with renal insufficiency, corticosteroids
Wright et al. 1999). are the treatment of choice (Carsons 2001).

Muscle Gastrointestinal
Muscle pain is common in patients with pSS; however, sympto- Vasculitis of the gastrointestinal tract is rare in SS and is associated
matic myopathy has been reported but it is uncommon and often with a high mortality. Acute necrotizing arteritis of medium-sized
CHAPTER 33 vasculitis in primary connective tissue diseases 453

arteries resembling polyarteritis nodosa has been described involv- cartilage of the tracheobronchial tree. Inflammation can involve
ing the pancreatic and mesenteric arteries as well as the spleen and other non-cartilagenous structures, including the eyes, heart, blood
gallbladder (Ramos-Casals et al. 2004; Tsokos et al. 1987). vessels, inner ear, and kidneys (Letko et al. 2002). Approximately
In summary, vasculitis can be one of the many extraglandular one-third of patients with RP have an associated haematological
manifestation of SS. The skin and nervous system are the most disorder, connective tissue disease, systemic inflammatory arthri-
common organ systems affected. Treatment depends on the sever- tis, or other autoimmune disease (Letko et al. 2002).
ity of vasculitis and the site of involvement. CNS vasculitis may be Vasculitis is an accepted feature of RP, occurring in as many as
more common than previously reported and carries a high risk of 56% of patients with the disease (McAdam et al. 1976). The spec-
morbidity if not recognized and treated aggressively. trum varies from leukocytoclastic vasculitis of small cutaneous ves-
sels to necrotizing vasculitis of the great vessels (Letko et al. 2002;
Systemic sclerosis and vasculitis Michet 1990). Dermatological manifestations of RP are found in
up to 35% of patients, with the majority of lesions associated with
Systemic sclerosis (SSc) is a disease characterized by immunological
leukocytoclastic vasculitis (Frances et al. 2001). These skin lesions
abnormalities, microangiopathy, and excessive deposition of colla-
include purpura, livedo reticularis, urticarial papules, erythema
gen in unregulated fashion. Organ systems affected include skin,
nodosum-like lesions, erythema elevatum diutinum, and distal
GI tract, lungs, heart, and kidneys. The pathology of SSc has three
ulceration of the limbs (Frances et al. 2001). Other non-vasculitic
major components:  (1)  small-vessel vasculopathy characterized
RP-related skin lesions include oral aphthosis (common), neu-
by endothelial cell injury and activation, (2) inflammation caused
trophilic dermatosis, superficial phlebitis, and septal panniculitis
by a perivascular monocytic/ macrophage infiltrate, and (3) tissue
(Frances et al. 2001).
fibrosis (Denton and Black 2004). Despite the systemic vasculopa-
Ocular manifestations occur in up to 60% of RP patients (Isaak
thy associated with SSc, true vasculitis is rare occurring in less than
et al. 1986). Episcleritis and scleritis is most common, followed by
1% of patients (Oddis et al. 1987; Quemeneur et al. 2013).
conjunctivitis sicca, iritis, retinopathy, keratitis, optic neuritis, and
Vasculitis in SSc, when it is found, usually occurs in the set-
corneal melt (Isaak et al. 1986; Kent et al. 2004). Orbital pseudo-
ting of the limited cutaneous form, which is seropositive for anti-
tumour, extraocular muscle palsy, and lid oedema can also occur
centromere antibody (Oddis et al. 1987; Quemeneur et al. 2013).
(Isaak et al. 1986; Kent et al. 2004).
Histologically, the vasculitis is typically cutaneous and leukocyto-
Renal lesions occur in approximately 10–20% of RP patients
clastic. Rarely, necrotizing vasculitis of larger arteries may occur in
and are usually associated with extrarenal vasculitis and a worse
the limited form and has been reported as digital gangrene and CNS
prognosis (Chang-Miller et  al. 1987; Kent et  al. 2004). Kidney
disease (Herrick et al. 1994a; Herrick et al. 1994b). Vasculitis has
involvement is caused by RP itself, or with an associated under-
also been described in SSc patients who have concomitant Sjögren’s
lying connective tissue disease-associated vasculitis, such as SLE
syndrome, ANCA associated vasculitis, or mixed cryoglobulinae-
(Isaak et al. 1986). Mild mesangial proliferation is the most com-
mia (Oddis et al. 1987; Quemeneur et al. 2013). ANCA-associated
mon histological finding. Other findings include IgA nephropathy,
vasculitis occurs in patients with limited SSc and typically presents
tubulointerstitial nephritis, and segmental necrotizing crescentic
with crescentic glomerulonephritis and/or a pulmonary–renal syn-
glomerulonephritis (Chang-Miller et al. 1987; Letko et al. 2002).
drome (Anders et al. 1999; Carvajal et al. 1997; Mizutani et al. 2000;
Peripheral neuropathies (mononeuritis multiplex and mixed
Omote et al. 1997; Quemeneur et al. 2013; Yamashita et al. 2000).
motor-sensory types) can occur in RP due to vasculitis of small,
SSc patients with mixed cryoglobulinaemia-associated vasculitis
epineurial vessels (Michet 1990). Cerebral vasculitis affecting
have no evidence of hepatitis C infection and the cryoglobulinaemia
both small and medium vessels has also been reported (Stewart
was thought to be due to secondary Sjögren’s syndrome (Buskila
et al. 1988).
et al. 1990; Husson et al. 1976; Quemeneur et al. 2013). Clinically,
Cardiovascular disease manifestations in RP are well docu-
these patients typically present with cutaneous and neurological
mented (Michet 1990). The spectrum of involvement includes
manifestations with evidence of circulating cryoglobulins and low
aortitis, thoracic, and abdominal aortic aneurysms, Takayasu-like
C4 values (Buskila et al. 1990; Husson et al. 1976; Quemeneur et al.
aortic arch syndrome, conduction system abnormalities, pericar-
et al. 2013). Although rare in SSc, prompt recognition of vasculitis,
ditis, coronary vasculitis, and valvulitis. The aortic valve is most
especially in those with limited disease, should lead to further eval-
commonly involved followed by the mitral valve, which can cause
uation including, but not limited to, testing for ANCAs, Sjögren’s
catastrophic heart failure, even if other features of RP are clinically
related autoantibodies, serum cryoglobulins, and tissue biopsy of
quiet (Pappas and Johnson 1972). Baseline echocardiography has
affected organs (Quemeneur et al. 2013). Treatment should be dic-
been advocated to screen for aortic valve disease, but no evidence
tated by the underlying cause of the vasculitis and the severity of
supports its routine use (Buckley and Ades 1992).
the disease. Careful dosing of corticosteroids is required in patients
Glucocorticoids are used in therapy of vasculitis associated
with SSc in order to avoid precipitating renal crisis. Early and
with RP. Azathioprine, methotrexate, cyclophosphamide, and
appropriate use of immunosuppressive agents, such as cyclophos-
ciclosporin have been used for glucocorticoid-resistant cases or
phamide, MMF, rituximab, or plasmapheresis, is also indicated and
as steroid-sparing agents (Kent et  al. 2004; Michet 1990). There
directed by the specific type of vasculitis (Quemeneur et al. 2013).
are no controlled studies documenting the effectiveness of any of
these agents. The use of several biologicals has been reported in
Polychondritis and vasculitis RP, including anti-TNF-α blockers, rituximab, anakinra, tocili-
Relapsing polychondritis (RP) is a rare systemic disorder char- zumab, and abatacept, which show efficacy in some but not all cases
acterized by episodic inflammation of cartilaginous struc- (Abdwani et  al. 2012; Kaly and Rosner 2012; Kemta et  al. 2012;
tures, including the elastic cartilage of the ear and nose, and Narshi and Allard 2012). Autologous stem cell transplantation for
454 SECTION 5 vasculitic diseases and syndromes and related disorders

refractory connective tissue diseases including RP may also be a gross haematuria and proteinuria (Satko et  al. 2000). Other glo-
viable option (Rosen et al. 2000; Tomomatsu et al. 2012). Finally, merulopathies reported include mesangial proliferative glomeru-
valve replacement or aneurysm grafting should be performed lonephritis, amyloid nephropathy, and membranous nephropathy
when indicated, although the inflammatory process may continue (Botey et al. 1981; Malaviya et al. 1981; Satko et al. 2000; Shu et al.
in proximity to installed devices, resulting in therapeutic failures 1986). An important note of caution is that patients with SNSA
(Lang-Lazdunski et al. 1995; Michet 1990). may develop renal insufficiency unrelated to their primary disease,
but rather as a consequence of treatment with NSAIDs (Satko et al.
2000; Shu et al. 1986).
Seronegative spondyloarthropathies Vasculitic skin lesions are rarely described in the SNSAs. There
The seronegative spondyloarthropathies (SNSAs) consist of a clus- are cases of leukocytoclastic vasculitis that have occurred in asso-
ter of interrelated, overlapping inflammatory rheumatic diseases ciation with reactive arthritis, psoriatic arthritis, and in those with
that include ankylosing spondylitis (AS), reactive arthritis, psori- IgA nephropathy (Jennette et al. 1982; Magro et al. 1995; Wong and
atic arthritis, arthritis associated with Crohn’s disease and ulcera- Marks 1994). When vasculitis is seen in the context of SNSA, latent
tive colitis, and undifferentiated spondyloarthropathies (Khan HIV infection should be excluded as a cause (Kaye 1989).
2002). These diseases share several characteristics including: (1) an Nervous system involvement in SNSA is limited to case reports
asymmetric erosive inflammatory arthropathy, (2)  inflamma- describing transverse myelitis, and cauda equina syndrome (Bilgen
tion at the site of ligament-to-bone insertion (enthesopathy), and et  al. 1999; Gabay et  al. 1978; Haddad et  al. 1990; Koenigsberg
(3) inflammation in the axial skeleton and sacroiliac joints (Kerr et al. 1995; Kushwaha and Steinberg 1992; Oh et al. 2001; Sant and
and Sturrock 1999). Extra-articular features of the spondyloar- O’Connell 1995; Schroder et al. 1994; Travis and Byrne 1987).
thropathies include anterior uveitis, colitis, aortitis, and apical lung
fibrosis (especially in the case of AS) (Kerr and Sturrock 1999; Idiopathic inflammatory
Momeni et al. 2011).
Acute anterior uveitis occurs in 20–40% of patients with AS, myopathies and vasculitis
but is less common in the other spondyloarthropathies (Gran and Polymyositis (PM) and dermatomyositis (DM) are members of
Skomsvoll 1997). The uveitis is characterized by recurrent, alternat- a group of inflammatory myopathies that have in common the
ing, unilateral attacks of a painful, inflamed eye which is typically presence of muscle weakness and inflammation in the muscle
confined to those who are HLA-B27 positive (Monnet et al. 2004; (Dalakas and Hohlfeld 2003). Dermatomyositis is distinguished
Rothova et al. 1992). Musculoskeletal symptoms precede the first by characteristic skin findings (heliotrope rash, erythema on the
attack of anterior uveitis in more than 80% of cases (Monnet et al. face, neck and anterior chest, and Gottron’s papules) associated
2004). Corticosteroids are effective initial treatment in topical, peri- with the muscle disease. Pathologically, DM is a humorally medi-
ocular, or systemic formulations. Sulfasalazine and methotrexate are ated microangiopathy with activation and deposition of comple-
also recommended for those who fail to respond to systemic cor- ment in the endomysial capillaries, resulting in muscle ischaemia
ticosteroids or who have frequent recurrences (Munoz-Fernandez in a perivascular and perifasicular distribution (Arahata and Engel
et  al. 2003; Munoz-Fernandez and Martin-Mola 2006). The use 1988; Dalakas and Hohlfeld 2003; Engel and Arahata 1984). In pol-
of anti-TNF agents, specifically adalimumab and infliximab, have ymyositis there is no rash and the inflammation is due to invasion
been very effective treatment options for SNSA-associated uveitis of CD8-positive cytotoxic T cells, which invade muscle fibres that
(Baraliakos et al. 2012; Braun et al. 2005; Gouveia et al. 2012). express MHC class I antigens (Arahata and Engel 1988; Engel and
Cardiovascular manifestations of AS include aortic root thicken- Arahata 1984; Engel et al. 1990). Vasculitis is not a common com-
ing and dilation, aortic valve thickening and insufficiency, mitral ponent of either DM or PM.
valve insufficiency, and conduction system abnormalities. Vasculitis Small-vessel cutaneous vasculitis characterized by periungual
and/or a vasculopathy affecting the small arteries of the cardiac and infarcts and digital ulcerations has been reported, which is more
aortic tissues result in intimal proliferation (obliterative endarteritis) frequent in child-onset DM as compared to adult-onset DM
and fibrosis (Bergfeldt 1997). Overall, aortic regurgitation has been (Feldman et al. 1983; Kadoya et al. 1994). In some, but not all stud-
described in 2 to 40% of patients with AS and in 2.8% of patients ies, leukocytoclastic vasculitis in adult DM has been a marker of an
with reactive arthritis (Bachmann et  al. 1976; Bergfeldt 1997; underlying malignancy (Feldman et al. 1983; Hunger et al. 2001).
Kinsella et al. 1974; Roldan et al. 1998). Atrioventricular conduc- Interstitial lung disease is well described in PM/DM, frequently
tion blocks have been described in up to one-third of patients with associated with the antibodies to antihistidyl transfer RNA syn-
AS (Bergfeldt et al. 1982; O’Neill and Bresnihan 1992). Myocarditis thetase (anti-Jo-l) or mucin-like glycoprotein (KL-6)(Hirakata and
and pericarditis are rare features of SNSA (Bergfeldt 1997). Medium Nagai 2000). In a recent study of 70 subjects with PM or DM and
and large-vessel vasculitis has been described in patients with SNSA diffuse interstitial lung disease, the majority (~81%) of lung biopsies
including polyarteritis nodosa, Takayasu’s arteritis, and aneurysms revealed non-specific interstitial pneumonia (NSIP), while there
with arteritis of the ascending, thoracic, and abdominal aorta were isolated cases of diffuse alveolar damage (DAD), bronchioli-
(Duvernoy and Schatz 1966; Kawasuji et al. 1982; Paloheimo et al. tis obliterans organizing pneumonia (BOOP), and usual interstitial
1966; Somer and Siltanen 1970; Stamp et al. 2000; Takagi et al. 2003). pneumonia (UIP)(Douglas et al. 2001). In a separate report, pul-
Interstitial lung disease is a recognized pulmonary manifestation of monary capillaritis with DAH is described in two patients with PM
SNSAs (Momeni et al. 2011). Parenchymal change can occur early (Schwarz et al. 1995).
or late in the course of the SNSA (Momeni et al. 2011). Renal involvement is uncommon in DM/PM and glomerulone-
IgA nephropathy has been reported to occur in association with phritis (GN) is rare. In a review of the literature, Takizawa et al.
all types of SNSAs. Patients typically present with microscopic or found 15 cases of biopsy-proven cases of glomerulonephritis in
CHAPTER 33 vasculitis in primary connective tissue diseases 455

patients with PM and six cases in DM patients (Takizawa et  al. et al. 1998; Harreby et al. 1994; Khosroshahi et al. 2012; Marzano
2007). Mesangioproliferative GN is the primary lesion found in et al. 2001). Case reports have supported the use of tamoxifen in
PM, while membranous GN was more common in DM (Takizawa retroperitoneal fibrosis due to the ability of the drug to inhibit
et al. 2007). The GN responds like the muscle disease to corticos- transforming growth factor-beta (Al Salman and Makhdomi 2002;
teroids and/or immunosuppressive agents (Takizawa et al. 2007). Bourouma et  al. 1997; Clark et  al. 1991; Dedeoglu et  al. 2001;
Large-vessel vasculitis is rare, and is reportedly more common in Loffeld and van Weel 1993; Vaglio et al. 2006a). The treatment of
children than adults. One survey of 43 adult cases found evidence IgG-4-RSD may need to be prolonged in order to prevent disease
of large-vessel vasculitis in five patients (Kalovidouris et al. 1988; relapse (Harreby et al. 1994; Kamisawa and Okamoto 2008; Kardar
Pachman 1995). Mesenteric vasculitis has rarely been described et al. 2002; Khosroshahi et al. 2012).
in adult-onset DM, but is a known complication of paediatric
DM (Eshraghi et  al. 1998; Pachman et  al. 1998; See et  al. 1997). Summary
A peripheral polyneuropathy due to microangiopathic changes is
Vasculitis occurring in the context of connective tissue diseases
reported in two cases of child-onset DM (Vogelgesang et al. 1995).
presents difficult diagnostic challenges due to the variety of ves-
sels and organ systems that can become involved. Fortunately, most
Aortitis, periaortitis, and idiopathic are responsive to currently available therapies, especially when the
retroperitoneal fibrosis diagnosis is made and appropriate therapy instituted in a timely
fashion. The most important factors in successfully making these
Non-infectious aortitis, periaortitis, and retroperitoneal fibrosis
diagnoses are familiarity with common patterns of vasculitis within
are mostly seen in middle-aged to older men. It has recently been
a given disease process and appropriate consideration of vasculitis
recognized that a significant portion of these cases are caused by
in the differential diagnosis.
IgG4-related systemic disease (IgG4-RSD)(Stone 2011). IgG4-RSD
can affect multiple organ systems including the aorta, pancreas, References
biliary tree, lung, thyroid gland, kidneys, salivary glands, and Abdwani, R., Kolethekkat, A.A., and Al Abri, R. (2012). Refractory relaps-
other organs (Khosroshahi et  al. 2013; Khosroshahi et  al. 2011; ing polychondritis in a child treated with antiCD20 monoclonal anti-
Khosroshahi and Stone 2011). These diseases are characterized by body (rituximab):  first case report. International Journal of Pediatric
elevated serum IgG4 levels, infiltration of IgG4 plasma cells in the Otorhinolaryngology, 76, 1061–4.
affected organ, and accompanying fibrosis (Kamisawa and Okamoto Agmon-Levin, N., Kivity, S., Tzioufas, A.G., Lopez, H.M., Rozman, B., Efes, I.,
2008; Stone 2011). The characteristic histological findings include Shapira, Y., Shamis, A., Amital, H., Youinou, P., and Shoenfeld, Y. (2012).
Low levels of vitamin-D are associated with neuropathy and lymphoma
a lymphoplasmacytic infiltrate with a high percentage of plasma
among patients with Sjogren’s syndrome. Journal of Autoimmunity,
cells staining for IgG4, obliterative phlebitis, storiform fibrosis, and 39, 234–9.
modest tissue eosinophilia (Khosroshahi and Stone 2011). Ainiala, H., Hietaharju, A., Loukkola, J., Peltola, J., Korpela, M., Metsanoja,
IgG4-related aortitis is most commonly found in the ascending R., and Auvinen, A. (2001). Validity of the new American College
portion and arch of the thoracic aorta and the characteristic inflam- of Rheumatology criteria for neuropsychiatric lupus syndromes:  a
matory findings are located within the aortic wall (Kamisawa and population-based evaluation. Arthritis and Rheumatism, 45, 419–23.
Okamoto 2008; Stone 2011). Inflammatory abdominal aortic aneu- Alarcon-Segovia, D., Abud-Mendoza, C., Reyes-Gutierrez, E.,
rysms and abdominal periaortitis are also described in IgG4-RSD Iglesias-Gamarra, A., and Diaz-Jouanen, E. (1984). Involvement of the
urinary bladder in systemic lupus erythematosus. A pathologic study.
(Stone 2011). Idiopathic retroperitoneal fibrosis, which can occur
Journal of Rheumatology, 11, 208–10.
in the setting of abdominal aortitis or periaortitis or alone, is also
Alarcon-Segovia, D., Perez-Vazquez, M.E., Villa, A.R., Drenkard, C., and
commonly due to an IgG4-related disease (Kamisawa and Okamoto Cabiedes, J. (1992). Preliminary classification criteria for the antiphos-
2008; Stone 2011). pholipid syndrome within systemic lupus erythematosus. Seminars in
Due to the anatomic location of the inflammation, patients Arthritis and Rheumatism, 21, 275–86.
often complain early of vague lower back or abdominal pain. Alexander, E. (1992). Central nervous system disease in Sjogren’s syndrome.
Hydronephrosis due to ureteral compression can occur in retro- New insights into immunopathogenesis. Rheumatic Diseases Clinics of
peritoneal fibrosis (Rhee et  al. 1994). Constitutional symptoms, North America, 18, 637–72.
including fatigue, low-grade fever, nausea, anorexia, weight loss, Alexander, E.L., Arnett, F.C., Provost, T.T., and Stevens, M.B. (1983).
Sjogren’s syndrome: association of anti-Ro(SS-A) antibodies with vascu-
and myalgias, are also common (Vaglio et al. 2006b). CT or MRI
litis, hematologic abnormalities, and serologic hyperreactivity. Annals of
scans can be used to detect and follow the course of these IgG4-RSD. Internal Medicine, 98, 155–9.
A tissue biopsy should be obtained for a definitive diagnosis and to Alexander, E.L., Moyer, C., Travlos, G.S., Roths, J.B., and Murphy, E.D.
rule out other possible aetiologies, including infection, malignancy, (1985). Two histopathologic types of inflammatory vascular disease in
and atherosclerotic disease. MRL/Mp autoimmune mice. Model for human vasculitis in connective
The goals of treatment of these IgG4-RSD are to stop the progres- tissue disease. Arthritis and Rheumatism, 28, 1146–55.
sion of the disease, relieve or prevent ureteral obstruction in ret- Al Salman, J. and Makhdomi, A.R. (2002). Treatment of retroperitoneal
roperitoneal fibrosis, normalize acute phase reactants, and prevent fibrosis with tamoxifen. Southern Medical Journal, 95, 947.
disease relapse (Vaglio et al. 2006b). Corticosteroids are most often American College of Rheumatology (ACR) (1999). The American College of
Rheumatology nomenclature and case definitions for neuropsychiatric
used with initial doses of prednisone of 40 to 60 mg per day (Kardar
lupus syndromes. Arthritis and Rheumatism, 42, 599–608.
et  al. 2002; Marcolongo et  al. 2004). Other immunosuppressive
Anders, H.J., Wiebecke, B., Haedecke, C., Sanden, S., Combe, C., and
agents (cyclophosphamide, azathioprine, methotrexate, MMF, Schlondorff, D. (1999). MPO-ANCA-Positive crescentic glomerulone-
ciclosporin, rituximab) have been used both as steroid-sparing phritis: a distinct entity of scleroderma renal disease? American Journal
agents as well as rescue therapy for steroid-resistant cases (Grotz of Kidney Diseases, 33, e3.
456 SECTION 5 vasculitic diseases and syndromes and related disorders

Ansari, A., Larson, P.H., and Bates, H.D. (1986). Vascular manifestations of of inflammatory cells and activated endothelium. Arthritis and
systemic lupus erythematosus. Angiology, 37, 423–32. Rheumatism, 39, 9–22.
Appel, G.B., Pirani, C.L., and D’Agati, V. (1994). Renal vascular complica- Bergfeldt, L. (1997). HLA-B27-associated cardiac disease. Annals of Internal
tions of systemic lupus erythematosus. Journal of the American Society Medicine, 127, 621–9.
of Nephrology, 4, 1499–515. Bergfeldt, L., Edhag, O., and Vallin, H. (1982). Cardiac conduction distur-
Arahata, K. and Engel, A.G. (1988). Monoclonal antibody analysis of mono- bances, an underestimated manifestation in ankylosing spondylitis.
nuclear cells in myopathies. V: Identification and quantitation of T8+ A  25-year follow-up study of 68 patients. Acta Medica Scandinavica,
cytotoxic and T8+ suppressor cells. Annals of Neurology, 23, 493–99. 212, 217–23.
Armstrong, D.J., McCarron, M.T., and Wright, G.D. (2005). Successful treat- Bertsias, G.K., Ioannidis, J.P., Aringer, M., Bollen, E., Bombardieri, S.,
ment of rheumatoid vasculitis-associated foot-drop with infliximab. Bruce, I.N., Cervera, R., Dalakas, M., Doria, A., Hanly, J.G., Huizinga,
Journal of Rheumatology, 32, 759–60. T.W., Isenberg, D., Kallenberg, C., Piette, J.C., Schneider, M., Scolding,
Asherson, R.A., Mackworth-Young, C.G., Harris, E.N., Gharavi, A.E., and N., Smolen, J., Stara, A., Tassiulas, I., Tektonidou, M., Tincani, A.,
Hughes, G.R. (1985). Multiple venous and arterial thromboses associ- van Buchem, M.A., van Vollenhoven, R., Ward, M., Gordon, C., and
ated with the lupus anticoagulant and antibodies to cardiolipin in the Boumpas, D.T. (2010). EULAR recommendations for the management
absence of SLE. Rheumatology International, 5, 91–3. of systemic lupus erythematosus with neuropsychiatric manifesta-
Asherson, R.A., Morgan, S.H., Harris, E.N., Gharavi, A.E., Krausz, T., and tions: report of a task force of the EULAR standing committee for clini-
Hughes, G.R. (1986). Arterial occlusion causing large bowel infarction— cal affairs. Annals of the Rheumatic Diseases, 69, 2074–82.
a reflection of clotting diathesis in SLE. Clinical Rheumatology, 5, 102–6. Bidani, A.K., Roberts, J.L., Schwartz, M.M., and Lewis, E.J. (1980).
Ashouri, J.F., Davis, J.L., Farkas, A., Durack, J.C., Ramachandran, R., and Immunopathology of cardiac lesions in fatal systemic lupus erythema-
Dall’Era, M. (2012). A young woman with systemic lupus erythematosus tosus. American Journal of Medicine, 69, 849–58.
and extensive mesenteric vasculitis involving small and medium vessels. Bilgen, I.G., Yunten, N., Ustun, E.E., Oksel, F., and Gumusdis, G. (1999).
Arthritis Care and Research (Hoboken), 64, 1928–33. Adhesive arachnoiditis causing cauda equina syndrome in ankylosing
Au, A. and O’Day, J. (2004). Review of severe vaso-occlusive retinopathy in spondylitis: CT and MRI demonstration of dural calcification and a dor-
systemic lupus erythematosus and the antiphospholipid syndrome: asso- sal dural diverticulum. Neuroradiology, 41, 508–11.
ciations, visual outcomes, complications and treatment. Clinical and Blanche, P., Krebs, S., Renaud, B., Dusser, D., and Sicard, D. (1996). Systemic
Experimental Ophthalmology, 32, 87–100. lupus erythematosus presenting as iron deficiency anemia due to pul-
Axson, F.A. (1979). D-penicillamine in the treatment of rheumatoid vasculi- monary alveolar hemorrhage. Clinical and Experimental Rheumatology,
tis. Journal of the South Carolina Medical Association, 75, 211–5. 14, 28.
Bachmann, F., Hartl, W., Veress, M., and Frind, W. (1976). [Cardiovascular Bloch, S.L., Jarrett, M.P., Swerdlow, M., and Grayzel, A.I. (1979). Brachial
complications of ankylosing spondylitis (Bechterew’s disease)]. plexus neuropathy as the initial presentation of systemic lupus erythe-
Medizinische Welt, 27, 2149–50. matosus. Neurology, 29, 1633–4.
Bacon, P.A. and Carruthers, D.M. (1995). Vasculitis associated with connective Bonfiglio, T. and Atwater, E.C. (1969). Heart disease in patients with sero-
tissue disorders. Rheumatic Diseases Clinics of North America, 21, 1077–96. positive rheumatoid arthritis; a controlled autopsy study and review.
Bacons, P.A. and Kitas, G.D. (1994). The significance of vascular inflam- Archives of Internal Medicine, 124, 714–9.
mation in rheumatoid arthritis. Annals of the Rheumatic Diseases, Bossini, N., Savoldi, S., Franceschini, F., Mombelloni, S., Baronio, M.,
53, 621–3. Cavazzana, I., Viola, B.F., Valzorio, B., Mazzucchelli, C., Cattaneo, R.,
Badui, E., Garcia-Rubi, D., Robles, E., Jimenez, J., Juan, L., Deleze, M., Diaz, A., Scolari, F., and Maiorca, R. (2001). Clinical and morphological fea-
and Mintz, G. (1985). Cardiovascular manifestations in systemic lupus tures of kidney involvement in primary Sjogren’s syndrome. Nephrology
erythematosus. Prospective study of 100 patients. Angiology, 36, 431–41. Dialysis Transplantation, 16, 2328–36.
Bando, H., Kobayashi, S., Matsumoto, T., Tamura, N., Yamanaka, K., Yamaji, Botey, A., Torras, A., and Revert, L. (1981). Membranous nephropathy in
C., Takasaki, C., Takasaki, Y., and Hashimoto, H. (2003). Acute acalculous ankylosing spondylitis. Nephron, 29, 203.
cholecystitis induced by mesenteric inflammatory veno-occlusive dis- Boumpas, D.T., Austin, H.A., III, Fessler, B.J., Balow, J.E., Klippel, J.H., and
ease (MIVOD) in systemic lupus erythematosus. Clinical Rheumatology, Lockshin, M.D. (1995). Systemic lupus erythematosus: emerging con-
22, 447–9. cepts. Part 1: Renal, neuropsychiatric, cardiovascular, pulmonary, and
Baraliakos, X., van den Berg, R., Braun, J., and Van Der Heijde, D. (2012). hematologic disease. Annals of Internal Medicine, 122, 940–50.
Update of the literature review on treatment with biologics as a basis for Bourouma, R., Chevet, D., Michel, F., Cercueil, J.P., Arnould, L., and Rifle, G.
the first update of the ASAS/EULAR management recommendations of (1997). Treatment of idiopathic retroperitoneal fibrosis with tamoxifen.
ankylosing spondylitis. Rheumatology (Oxford), 51, 1378–87. Nephrology Dialysis Transplantation, 12, 2407–10.
Barile, L. and Lavalle, C. (1992). Transverse myelitis in systemic lupus erythe- Braun, J., Baraliakos, X., Listing, J., and Sieper, J. (2005). Decreased inci-
matosus—the effect of IV pulse methylprednisolone and cyclophospha- dence of anterior uveitis in patients with ankylosing spondylitis treated
mide. Journal of Rheumatology, 19, 370–2. with the anti-tumor necrosis factor agents infliximab and etanercept.
Barile-Fabris, L., Ariza-Andraca, R., Olguin-Oretega, L., Jara, L.J., Arthritis and Rheumatism, 52, 2447–51.
Fraga-Mouret, A., Miranda-Limon, J.M., Fuentes delaMata, J., Clark, Breedveld, F.C., Heurkens, A.H., Lafeber, G.J., van Hinsbergh, V.W., and
P., Vargas, F., and Alocer-Varela, J. (2005). Controlled clinical trial of Cats, A. (1988). Immune complexes in sera from patients with rheu-
IV cyclophosphamide versus IV methylprednisolone in severe neuro- matoid vasculitis induce polymorphonuclear cell-mediated injury
logical manifestations in systemic lupus erythematosus. Annals of the to endothelial cells. Clinical Immunology and Immunopathology, 48,
Rheumatic Diseases, 64, 620–5. 202–13.
Bathon, J.M., Moreland, L.W., and DiBartolomeo, A.G. (1989). Inflammatory Brey, R.L., Holliday, S.L., Saklad, A.R., Navarrete, M.G., Hermosillo-Romo,
central nervous system involvement in rheumatoid arthritis. Seminars in D., Stallworth, C.L., Valdez, C.R., Escalante, A., del Rincón, I, Gronseth,
Arthritis and Rheumatism, 18, 258–66. G., Rhine, C.B., Padilla, P., and McGlasson, D. (2002). Neuropsychiatric
Beaufils, M., Kouki, F., Mignon, F., Camus, J.P., Morel-Maroger, L., and syndromes in lupus:  prevalence using standardized definitions.
Richet, G. (1983). Clinical significance of anti-Sm antibodies in systemic Neurology, 58, 1214–20.
lupus erythematosus. American Journal of Medicine, 74, 201–5. Buckley, L.M. and Ades, P.A. (1992). Progressive aortic valve inflamma-
Belmont, H.M., Abramson, S.B., and Lie, J.T. (1996). Pathology and patho- tion occurring despite apparent remission of relapsing polychondritis.
genesis of vascular injury in systemic lupus erythematosus. Interactions Arthritis and Rheumatism, 35, 812–4.
CHAPTER 33 vasculitis in primary connective tissue diseases 457

Bulkley, B.H. and Roberts, W.C. (1975). The heart in systemic lupus erythe- Dalavanga, Y.A., Constantopoulos, S.H., Galanopoulou, V., Zerva, L., and
matosus and the changes induced in it by corticosteroid therapy. A study Moutsopoulos, H.M. (1991). Alveolitis correlates with clinical pulmo-
of 36 necropsy patients. American Journal of Medicine, 58, 243–64. nary involvement in primary Sjogren’s syndrome. Chest, 99, 1394–7.
Buskila, D., Langevitz, P., and Lee, P. (1990). The frequency of cryoglobulinae- Dalavanga, Y.A., Voulgari, P.V., Georgiadis, A.N., Leontaridi, C., Katsenos,
mia in systemic sclerosis (scleroderma). British Journal of Rheumatology, S., Vassiliou, M., Drosos, A.A., and Constantopoulos, S.H. (2006).
29, 234. Lymphocytic alveolitis: A surprising index of poor prognosis in patients
Bywaters, E.G.L. (1957). Peripheral vascular obstruction in rheumatoid with primary Sjogren’s syndrome. Rheumatology International, 26,
arthritis and its relationship to other vascular lesions. Annals of the 799–804.
Rheumatic Diseases, 16, 84–103. Damato, E., Chilov, M., Lee, R., Singh, A., Harper, S., and Dick, A. (2011).
Calamia, K.T. and Balabanova, M. (2004). Vasculitis in systemic lupus ery- Plasma exchange and rituximab in the management of acute occlusive
thematosis. Clinical Dermatology, 22, 148–56. retinal vasculopathy secondary to systemic lupus erythematosus. Ocular
Callen, J.P., Spencer, L.V., Burruss, J.B., and Holtman, J. (1991). Azathioprine. Immunology and Inflammation, 19, 379–81.
An effective, corticosteroid-sparing therapy for patients with recalci- Danning, C.L., Illei, G.G., and Boumpas, D.T. (1998). Vasculitis associated
trant cutaneous lupus erythematosus or with recalcitrant cutaneous leu- with primary rheumatologic diseases. Current Opinion in Rheumatology,
kocytoclastic vasculitis. Archives of Dermatology, 127, 515–22. 10, 58–65.
Campion, G., Maddison, P.J., Goulding, N., James, I., Ahern, M.J., Watt, I., d’Cruz, D. (1998). Vasculitis in systemic lupus erythematosus. Lupus, 7, 270–4.
and Sansom, D. (1990). The Felty syndrome: a case-matched study of d’Cruz, D., Khamashta, M., and Hughes, G. (1997). Antiendothelial cell anti-
clinical manifestations and outcome, serologic features, and immunoge- bodies (AECA) in systemic lupus erythematosus (SLE). Clinical Reviews
netic associations. Medicine (Baltimore), 69, 69–80. in Allergy and Immunology, 15, 53–63.
Cardinali, C., Caproni, M., Bernacchi, E., Amato, L., and Fabbri, P. (2000). Dedeoglu, F., Rose, C.D., Athreya, B.H., Conard, K., Grissom, L., and
The spectrum of cutaneous manifestations in lupus erythematosus—the Magnusson, M. (2001). Successful treatment of retroperitoneal fibrosis
Italian experience. Lupus, 9, 417–23. with tamoxifen in a child. Journal of Rheumatology, 28, 1693–5.
Carloni, A., Piciucchi, S., Giannakakis, K., Nori, G., Zobel, B.B., and Poletti, Delalande, S., de Seze, J., Fauchais, A.L., Hachulla, E., Stojkovic, T., Ferriby,
V. (2008). Diffuse alveolar hemorrhage after leflunomide therapy in a D., Dubucquoi, S., Pruvo, J.P., Vermersch, P., and Hatron, P.Y. (2004).
patient with rheumatoid arthritis. Journal of Thoracic Imaging, 23, 57–9. Neurologic manifestations in primary Sjogren syndrome: a study of 82
Carlson, J.A. and Chen, K.R. (2007). Cutaneous vasculitis update: neutro- patients. Medicine (Baltimore), 83, 280–91.
philic muscular vessel and eosinophilic, granulomatous, and lympho- den Broeder, A.A., van den Hoogen, F.H., and van de Putte, L.B. (2001).
cytic vasculitis syndromes. American Journal of Dermatopathology, Isolated digital vasculitis in a patient with rheumatoid arthritis: good
29, 32–43. response to tumour necrosis factor alpha blocking treatment. Annals of
Carsons, S. (2001). A review and update of Sjogren’s syndrome: manifesta- the Rheumatic Diseases, 60, 538–9.
tions, diagnosis, and treatment. American Journal of Managed Care, 7 Denton, C.P. and Black, C.M. (2004). Scleroderma—clinical and pathological
(Suppl.), S433–43. advances. Best Practice and Research Clinical Rheumatology, 18, 271–90.
Carvajal, I., Bernis, C., Sanz, P., Garcia, A., Garcia-Vadillo, A., and Traver, Doherty, N.E., III, Feldman, G., Maurer, G., and Siegel, R.J. (1988).
J.A. (1997). Antineutrophil cytoplasmic autoantibodies (ANCA) and Echocardiographic findings in systemic lupus erythematosus. American
systemic sclerosis. Nephrology Dialysis Transplantation, 12, 576–7. Journal of Cardiology, 61, 1144.
Cathcart, E.S. and Spodick, D.H. (1962). Rheumatoid heart disease. A study Doria, A., Iaccarino, L., Sarzi-Puttini, P., Atzeni, F., Turriel, M., and Petri, M.
of the incidence and nature of cardiac lesions in rheumatoid arthritis. (2005). Cardiac involvement in systemic lupus erythematosus. Lupus,
New England Journal of Medicine, 266, 959–64. 14, 683–6.
Chai, J. and Logigian, E.L. (2010). Neurological manifestations of primary Douglas, W.W., Tazelaar, H.D., Hartman, T.E., Hartman, R.P., Decker,
Sjogren’s syndrome. Current Opinion in Neurology, 23, 509–13. P.A., Schroeder, D.R., and Ryu, J.H. (2001). Polymyositis-
Chang-Miller, A., Okamura, M., Torres, V.E., Michet, C.J., Wagoner, R.D., dermatomyositis-associated interstitial lung disease. American Journal
Donadio, J.V. Jr, Offord, K.P., and Holley, K.E. (1987). Renal involvement of Respiratory and Critical Care Medicine, 164, 1182–5.
in relapsing polychondritis. Medicine (Baltimore), 66, 202–17. Drenkard, C., Villa, A.R., Reyes, E., Abello, M., and Alarcon-Segovia, D.
Chen, K.R., Toyohara, A., Suzuki, A., and Miyakawa, S. (2002). Clinical (1997). Vasculitis in systemic lupus erythematosus. Lupus, 6, 235–42.
and histopathological spectrum of cutaneous vasculitis in rheumatoid Duvernoy, W.F. and Schatz, I.J. (1966). Rheumatoid spondylitis associated
arthritis. British Journal of Dermatology, 147, 905–13. with aneurysmal dilatation of the entire thoracic aorta. Henry Ford
Chin, H.J., Ahn, C., Lim, C.S., Chung, H.K., Lee, J.G., Song, Y.W., Lee, H.S., Hospital Medical Journal, 14, 309–12.
Han, J.S., Kim, S., and Lee, J.S. (2000). Clinical implications of antineu- Eber, T., Chapman, J., and Shoenfeld, Y. (2005). Anti-ribosomal P-protein
trophil cytoplasmic antibody test in lupus nephritis. American Journal of and its role in psychiatric manifestations of systemic lupus erythemato-
Nephrology, 20, 57–63. sus: myth or reality? Lupus, 14, 571–5.
Chin, R.L. and Latov, N. (2005). Central nervous system manifestations of Egerer, K., Feist, E., Rohr, U., Pruss, A., Burmester, G.R., and Dorner, T.
rheumatologic diseases. Current Opinion in Rheumatology, 17, 91–9. (2000). Increased serum soluble CD14, ICAM-1 and E-selectin corre-
Cieslik, P., Hrycek, A., and Klucinski, P. (2008). Vasculopathy and vascu- late with disease activity and prognosis in systemic lupus erythematosus.
litis in systemic lupus erythematosus. Polskie Archiwum Medycyny Lupus, 9, 614–21.
Wewnętrznej, 118, 57–63. Enevoldson, T.P. and Wiles, C.M. (1991). Severe vasculitic neuropathy in sys-
Clark, C.P., Vanderpool, D., and Preskitt, J.T. (1991). The response of retrop- temic lupus erythematosus and response to cyclophosphamide. Journal
eritoneal fibrosis to tamoxifen. Surgery, 109, 502–6. of Neurology Neurosurgery and Psychiatry, 54, 468–9.
Constantopoulos, S.H., Papadimitriou, C.S., and Moutsopoulos, H.M. Engel, A.G. and Arahata, K. (1984). Monoclonal antibody analysis of mono-
(1985). Respiratory manifestations in primary Sjogren’s syndrome. nuclear cells in myopathies. II: Phenotypes of autoinvasive cells in poly-
A clinical, functional, and histologic study. Chest, 88, 226–9. myositis and inclusion body myositis. Annals of Neurology, 16, 209–15.
Daina, E., Schieppati, A., and Remuzzi, G. (1999). Mycophenolate mofetil Engel, A.G., Arahata, K., and Emslie-Smith, A. (1990). Immune effec-
for the treatment of Takayasu arteritis: report of three cases. Annals of tor mechanisms in inflammatory myopathies. Research Publications
Internal Medicine, 130, 422–6. Association for Research in Nervous and Mental Disease, 68, 141–57.
Dalakas, M.C. and Hohlfeld, R. (2003). Polymyositis and dermatomyositis. Eshraghi, N., Farahmand, M., Maerz, L.L., Campbell, S.M., Deveney, C.W.,
Lancet, 362, 971–82. and Sheppard, B.C. (1998). Adult-onset dermatomyositis with severe
458 SECTION 5 vasculitic diseases and syndromes and related disorders

gastrointestinal manifestations: case report and review of the literature. Goules, A., Masouridi, S., Tzioufas, A.G., Ioannidis, J.P., Skopouli,
Surgery, 123, 356–8. F.N., and Moutsopoulos, H.M. (2000). Clinically significant and
Fairfax, M.J., Osborn, T.G., Williams, G.A., Tsai, C.C., and Moore, T.L. biopsy-documented renal involvement in primary Sjogren syndrome.
(1988). Endomyocardial biopsy in patients with systemic lupus erythe- Medicine (Baltimore), 79, 241–9.
matosus. Journal of Rheumatology, 15, 593–6. Gouveia, E.B., Elmann, D., and Morales, M.S. (2012). Ankylosing spondylitis
Fauci, A.S., Steinberg, A.D., Haynes, B.F., and Whalen, G. (1978). and uveitis: overview. Revista Brasileira de Reumatologia, 52, 742–56.
Immunoregulatory aberrations in systemic lupus erythematosus. Govoni, M., Bajocchi, G., Rizzo, N., Tola, M.R., Caniatti, L., Tugnoli, V.,
Journal of Immunology, 121, 1473–9. Colamussi, P., and Trotta, F. (1999). Neurological involvement in pri-
Feldman, D., Hochberg, M.C., Zizic, T.M., and Stevens, M.B. (1983). mary Sjogren’s syndrome:  clinical and instrumental evaluation in a
Cutaneous vasculitis in adult polymyositis/dermatomyositis. Journal of cohort of Italian patients. Clinical Rheumatology, 18, 299–303.
Rheumatology, 10, 85–9. Goyal, S. and Nousari, H.C. (2001). Treatment of resistant discoid lupus ery-
Feriozzi, S., Muda, A.O., Amini, M., Faraggiana, T., and Ancarani, E. (1997). thematosus of the palms and soles with mycophenolate mofetil. Journal
Systemic lupus erythematosus with membranous glomerulonephritis of the American Academy of Dermatology, 45, 142–4.
and uterine vasculitis. American Journal of Kidney Diseases, 29, 277–9. Gran, J.T. and Skomsvoll, J.F. (1997). The outcome of ankylosing spondyli-
Flipo, R.M., Janin, A., Hachulla, E., Houvenagel, E., Foulet, A., Cardon, T., tis: a study of 100 patients. British Journal of Rheumatology, 36, 766–71.
Desbonnet, A., Grardel, B., Duquesnoy, B., and Delcambre, B. (1994). Grant, I.A., Hunder, G.G., Homburger, H.A., and Dyck, P.J. (1997). Peripheral
Labial salivary gland biopsy assessment in rheumatoid vasculitis. Annals neuropathy associated with sicca complex. Neurology, 48, 855–62.
of the Rheumatic Diseases, 53, 648–52. Gravallese, E.M., Corson, J.M., Coblyn, J.S., Pinkus, G.S., and Weinblatt, M.E.
Fox, R.I. (2005). Sjogren’s syndrome. Lancet, 366, 321–31. (1989). Rheumatoid aortitis: a rarely recognized but clinically significant
Frances, C., el Rassi, R., Laporte, J.L., Rybojad, M., Papo, T., and Piette, entity. Medicine (Baltimore), 68, 95–106.
J.C. (2001). Dermatologic manifestations of relapsing polychondritis. Gray, R.G. and Poppo, M.J. (1983). Necrotizing vasculitis as the initial mani-
A study of 200 cases at a single center. Medicine (Baltimore), 80, 173–9. festation of rheumatoid arthritis. Journal of Rheumatology, 10, 326–8.
Frith, P., Burge, S.M., Millard, P.R., and Wojnarowska, F. (1990). External Greening, A.P. and Hughes, J.M. (1981). Serial estimations of carbon monox-
ocular findings in lupus erythematosus: a clinical and immunopatho- ide diffusing capacity in intrapulmonary haemorrhage. Clinical Science
logical study. British Journal of Ophthalmology, 74, 163–7. (London), 60, 507–12.
Gabay, R., Guignard, D., and Chantraine, A. (1978). A rare extra-articular Greisman, S.G., Thayaparan, R.S., Godwin, T.A., and Lockshin, M.D. (1991).
manifestation of ankylosing spondylitis:  cauda equina syndrome. Occlusive vasculopathy in systemic lupus erythematosus. Association
Journal of Rheumatology, 5, 234–5. with anticardiolipin antibody. Archives of Internal Medicine, 151, 389–92.
Garcia-Carrasco, M., Ramos-Casals, M., Rosas, J., Pallares, L., Calvo-Alen, Gresz, V., Kwon, T.H., Hurley, P.T., Varga, G., Zelles, T., Nielsen, S., Case,
J., Cervera, R., Font, J., and Ingelmo, M. (2002). Primary Sjogren syn- R.M., and Steward, M.C. (2001). Identification and localization of aqua-
drome:  clinical and immunologic disease patterns in a cohort of 400 porin water channels in human salivary glands. American Journal of
patients. Medicine (Baltimore), 81, 270–80. Physiology Gastrointestinal and Liver Physiology, 281, G247–54.
Garcia-Porrua, C., Gonzalez-Gay, M.A., and Quevedo, V. (2006). Should Griffin, J.W. (2001). Vasculitic neuropathies. Rheumatic Diseases Clinics of
anti-tumor necrosis factor-alpha be the first therapy for rheumatoid vas- North America, 27, 751–60, vi.
culitis? Journal of Rheumatology, 33, 433–4. Griffin, J.W., Cornblath, D.R., Alexander, E., Campbell, J., Low, P.A., Bird, S.,
Geirsson, A.J., Sturfelt, G., and Truedsson, L. (1987). Clinical and serological and Feldman, E.L. (1990). Ataxic sensory neuropathy and dorsal root
features of severe vasculitis in rheumatoid arthritis: prognostic implica- ganglionitis associated with Sjogren’s syndrome. Annals of Neurology,
tions. Annals of the Rheumatic Diseases, 46, 727–33. 27, 304–15.
Ginzler, E.M., Dooley, M.A., Aranow, C., Kim, M.Y., Buyon, J., Merrill, J.T., Grimbacher, B., Huber, M., von Kempis, J., Kalden, P., Uhl, M., Kohler, G.,
Petri, M., Gilkeson, G.S., Wallace, D.J., Weisman, M.H., and Appel, G.B. Blum, H.E., and Peter, H.H. (1998). Successful treatment of gastrointes-
(2005). Mycophenolate mofetil or intravenous cyclophosphamide for tinal vasculitis due to systemic lupus erythematosus with intravenous
lupus nephritis. New England Journal of Medicine, 353, 2219–28. pulse cyclophosphamide: a clinical case report and review of the litera-
Godfrey, T., Khamashta, M.A., and Hughes, G.R. (1998). Therapeutic advances ture. British Journal of Rheumatology, 37, 1023–8.
in systemic lupus erythematosus. Current Opinion in Rheumatology, 10, Grotz, W., von Zedtwitz, I, Andre, M., and Schollmeyer, P. (1998). Treatment
435–41. of retroperitoneal fibrosis by mycophenolate mofetil and corticosteroids.
Gono, T., Kawaguchi, Y., Katsumata, Y., Takagi, K., Tochimoto, A., Baba, S., Lancet, 352, 1195.
Okamoto, Y., Ota, Y., and Yamanaka, H. (2011). Clinical manifestations Guadalupe Loya-de la Cerda, D., Aviles-Solis, J.C., Delgado-Montemayor,
of neurological involvement in primary Sjogren’s syndrome. Clinical M.J., Camara-Lemarroy, C.R., and Galarza-Delgado, D.A. (2013).
Rheumatology, 30, 485–90. Isolated rheumatoid arthritis-associated cerebral vasculitis: a diagnostic
Gonzalez, A., Nicovani, S., Massardo, L., Aguirre, V., Cervilla, V., Lanchbury, challenge. Joint Bone Spine, 80, 88–90.
J.S., and Jacobelli, S. (1997). Influence of the HLA-DR beta shared Gyongyosi, M., Pokorny, G., Jambrik, Z., Kovacs, L., Kovacs, A., Makula, E.,
epitope on susceptibility to and clinical expression of rheumatoid arthri- and Csanady, M. (1996). Cardiac manifestations in primary Sjogren’s
tis in Chilean patients. Annals of the Rheumatic Diseases, 56, 191–3. syndrome. Annals of the Rheumatic Diseases, 55, 450–4.
Gonzalez-Gay, M.A. and Garcia-Porrua, C. (1999). Systemic vasculitis in Haddad, F.S., Sachdev, J.S., and Bellapravalu, M. (1990). Neuropathic blad-
adults in northwestern Spain, 1988-1997. Clinical and epidemiologic der in ankylosing spondylitis with spinal diverticula. Urology, 35, 313–6.
aspects. Medicine (Baltimore), 78, 292–308. Hanly, J.G. (1998). Evaluation of patients with CNS involvement in SLE.
Gorman, J.D., David-Vaudey, E., Pai, M., Lum, R.F., and Criswell, L.A. (2004). Baillieres Clinical Rheumatology, 12, 415–31.
Particular HLA-DRB1 shared epitope genotypes are strongly associated Hanly, J.G. (2005). Neuropsychiatric lupus. Rheumatic Diseases Clinics of
with rheumatoid vasculitis. Arthritis and Rheumatism, 50, 3476–84. North America, 31, 273–98, vi.
Goronzy, J.J. and Weyand, C.M. (1994). Vasculitis in rheumatoid arthritis. Hanly, J.G. and Harrison, M.J. (2005). Management of neuropsychiatric
Current Opinion in Rheumatology, 6, 290–4. lupus. Best Practice and Research Clinical Rheumatology, 19, 799–821.
Goronzy, J.J. and Weyand, C.M. (2003). Aging, autoimmunity and arthri- Hanly, J.G., McCurdy, G., Fougere, L., Douglas, J.A., and Thompson,
tis:  T-cell senescence and contraction of T-cell repertoire diversity— K. (2004). Neuropsychiatric events in systemic lupus erythemato-
catalysts of autoimmunity and chronic inflammation. Arthritis Research sus: attribution and clinical significance. Journal of Rheumatology, 31,
and Therapy, 5, 225–34. 2156–62.
CHAPTER 33 vasculitis in primary connective tissue diseases 459

Harisdangkul, V., Doorenbos, D., and Subramony, S.H. (1995). Lupus trans- T., Kristoferitsch, W., Melms, A., Wandinger, K.P., and Wildemann, B.
verse myelopathy:  better outcome with early recognition and aggres- (2011). Frequency and syndrome specificity of antibodies to aquaporin-4
sive high-dose intravenous corticosteroid pulse treatment. Journal of in neurological patients with rheumatic disorders. Multiple Sclerosis, 17,
Neurology, 242, 326–31. 1067–73.
Harreby, M., Bilde, T., Helin, P., Meyhoff, H.H., Vinterberg, H., and Nielsen, Jennette, J.C., Ferguson, A.L., Moore, M.A., and Freeman, D.G. (1982).
V.A. (1994). Retroperitoneal fibrosis treated with methylprednisolon IgA nephropathy associated with seronegative spondylarthropathies.
pulse and disease-modifying antirheumatic drugs. Scandinavian Journal Arthritis and Rheumatism, 25, 144–9.
of Urology and Nephrology, 28, 237–42. Jifi-Bahlool, H., Saadeh, C., and O’Connor, J. (1995). Peripheral ulcera-
Haupt, H.M., Moore, G.W., and Hutchins, G.M. (1981). The lung in systemic tive keratitis in the setting of rheumatoid arthritis:  treatment with
lupus erythematosus. Analysis of the pathologic changes in 120 patients. immunosuppressive therapy. Seminars in Arthritis and Rheumatism,
American Journal of Medicine, 71, 791–98. 25, 67–73.
Heibel, R.H., O’Toole, J.D., Curtiss, E.I., Medsger, T.A., Jr, Reddy, S.P., and Ju, J.H., Min, J.K., Jung, C.K., Oh, S.N., Kwok, S.K., Kang, K.Y., Park, K.S.,
Shaver, J.A. (1976). Coronary arteritis in systemic lupus erythematosus. Ko, H.J., Yoon, C.H., Park, S.H., Cho, C.S., and Kim, H.Y. (2009). Lupus
Chest, 69, 700–3. mesenteric vasculitis can cause acute abdominal pain in patients with
Hermisson, M., Klein, R., Schmidt, F., Weller, M., and Kuker, W. (2002). SLE. Nature Reviews Rheumatology, 5, 273–81.
Myelopathy in primary Sjogren’s syndrome: diagnostic and therapeutic Kadoya, A., Akahoshi, T., Sekiyama, N., Hosaka, S., and Kondo, H. (1994).
aspects. Acta Neurologica Scandinavica, 105, 450–3. Cutaneous vasculitis in a patient with dermatomyositis without muscle
Herrick, A.L., Oogarah, P., Brammah, T.B., Freemont, A.J., and Jayson, M.I. involvement. Internal Medicine, 33, 809–12.
(1994a). Nervous system involvement in association with vasculitis and Kalovidouris, A.E., Stoesz, E., Muller, J., Kimes, T., and Brandt, K.D. (1988).
anticardiolipin antibodies in a patient with systemic sclerosis. Annals of Relationships between clinical features and distribution of mononuclear
the Rheumatic Diseases, 53, 349–50. cells in muscle of patients with polymyositis. Journal of Rheumatology,
Herrick, A.L., Oogarah, P.K., Freemont, A.J., Marcuson, R., Haeney, M., and 15, 1401–6.
Jayson, M.I. (1994b). Vasculitis in patients with systemic sclerosis and Kaly, L. and Rosner, I. (2012). Tocilizumab—a novel therapy for
severe digital ischaemia requiring amputation. Annals of the Rheumatic non-organ-specific autoimmune diseases. Best Practice and Research
Diseases, 53, 323–6. Clinical Rheumatology, 26, 157–65.
Heurkens, A.H., Westedt, M.L., and Breedveld, F.C. (1991). Prednisone plus Kamisawa, T. and Okamoto, A. (2008). IgG4-related sclerosing disease.
azathioprine treatment in patients with rheumatoid arthritis compli- World Journal of Gastroenterology, 14, 3948–55.
cated by vasculitis. Archives of Internal Medicine, 151, 2249–54. Kao, A.H. and Manzi, S. (2002). How to manage patients with cardiopul-
Hillarby, M.C., Clarkson, R., Grennan, D.M., Bate, A.S., Ollier, W., Sanders, monary disease?. Best Practice and Research Clinical Rheumatology, 16,
P.A., Chattophadhyay, C., Davis, M., O’Sullivan, M.M., and Williams, 211–27.
B. (1991). Immunogenetic heterogeneity in rheumatoid disease as illus- Kao, A.H., Sabatine, J.M., and Manzi, S. (2003). Update on vascular disease
trated by different MHC associations (DQ, Dw and C4) in articular and in systemic lupus erythematosus. Current Opinion in Rheumatology, 15,
extra-articular subsets. British Journal of Rheumatology, 30, 5–9. 519–27.
Hirakata, M. and Nagai, S. (2000). Interstitial lung disease in polymyositis Kardar, A.H., Kattan, S., Lindstedt, E., and Hanash, K. (2002). Steroid ther-
and dermatomyositis. Current Opinion in Rheumatology, 12, 501–8. apy for idiopathic retroperitoneal fibrosis: dose and duration. Journal of
Holtman, J.H., Neustadt, D.H., Klein, J., and Callen, J.P. (1990). Dapsone is an Urology, 168, 550–55.
effective therapy for the skin lesions of subacute cutaneous lupus erythe- Karrar, A., Sequeira, W., and Block, J.A. (2001). Coronary artery disease in
matosus and urticarial vasculitis in a patient with C2 deficiency. Journal systemic lupus erythematosus: A review of the literature. Seminars in
of Rheumatology, 17, 1222–5. Arthritis and Rheumatism, 30, 436–43.
Horton, M.R. (2004). Rheumatoid arthritis associated interstitial lung dis- Kawasuji, M., Hetzer, R., Oelert, H., Stauch, G., and Borst, H.G. (1982).
ease. Critical Reviews in Computed Tomography, 45, 429–40. Aortic valve replacement and ascending aorta replacement in ankylos-
Hricik, D.E., Chung-Park, M., and Sedor, J.R. (1998). Glomerulonephritis. ing spondylitis: report of three surgical cases and review of the literature.
New England Journal of Medicine, 339, 888–99. Thoracic and Cardiovascular Surgeon, 30, 310–4.
Hunger, R.E., Durr, C., and Brand, C.U. (2001). Cutaneous leukocytoclas- Kaye, B.R. (1989). Rheumatologic manifestations of infection with human
tic vasculitis in dermatomyositis suggests malignancy. Dermatology, immunodeficiency virus (HIV). Annals of Internal Medicine, 111,
202, 123–6. 158–67.
Husson, J.M., Druet, P., Contet, A., Fiessinger, J.N., and Camilleri, J.P. (1976). Kemta, L.F., Kraus, V.B., and Chevalier, X. (2012). Biologics in relapsing poly-
Systemic sclerosis and cryoglobulinemia. Clinical Immunology and chondritis: a literature review. Seminars in Arthritis and Rheumatism,
Immunopathology, 6, 77–82. 41, 712–9.
Isaak, B.L., Liesegang, T.J., and Michet, C.J., Jr (1986). Ocular and systemic Kent, D.L., Haynor, D.R., Longstreth, W.T. Jr, and Larson, E.B. (1994). The
findings in relapsing polychondritis. Ophthalmology, 93, 681–9. clinical efficacy of magnetic resonance imaging in neuroimaging. Annals
Jabs, D.A., Fine, S.L., Hochberg, M.C., Newman, S.A., Heiner, G.G., and of Internal Medicine, 120, 856–71.
Stevens, M.B. (1986). Severe retinal vaso-occlusive disease in systemic Kent, P.D., Michet, C.J. Jr, and Luthra, H.S. (2004). Relapsing polychondritis.
lupus erythematous. Archives of Ophthalmology, 104, 558–63. Current Opinion in Rheumatology, 16, 56–61.
Jaffe, I.A. and Smith, R.W. (1968). Rheumatoid vasculitis. Report of a second Kerr, H.E. and Sturrock, R.D. (1999). Clinical aspects, outcome assessment,
case treated with penicillamine. Arthritis and Rheumatism, 11, 585–92. disease course, and extra-articular features of spondyloarthropathies.
Jans, H., Halberg, P., and Lorenzen, I. (1983). Circulating immune com- Current Opinion in Rheumatology, 11, 235–7.
plexes in rheumatoid arthritis with extra-articular manifestations. Khaja, M., Menon, L., Niazi, M., and Fuentes, G.D. (2012). Diffuse alveolar
Scandinavian Journal of Rheumatology, 12, 215–8. hemorrhage and acute respiratory distress syndrome during treatment
Jaraquemada, D., Ollier, W., Awad, J., Young, A., Silman, A., Roitt, I.M., of rheumatoid arthritis with etanercept. Journal of Bronchology and
Corbett, M., Hay, F., Cosh, J.A., and Maini, R.N. (1986). HLA and rheu- Interventional Pulmonology, 19, 228–31.
matoid arthritis: a combined analysis of 440 British patients. Annals of Khan, M.A. (2002). Update on spondyloarthropathies. Annals of Internal
the Rheumatic Diseases, 45, 627–36. Medicine, 136, 896–907.
Jarius, S., Jacobi, C., de Seze, J., Zephir, H., Paul, F., Franciotta, D., Rommer, Khosroshahi, A., Carruthers, M.N., Deshpande, V., Unizony, S., Bloch, D.B.,
P., Mader, S., Kleiter, I., Reindl, M., Akman-Demir, G., Seifert-Held, and Stone, J.H. (2012). Rituximab for the treatment of IgG4-related
460 SECTION 5 vasculitic diseases and syndromes and related disorders

disease:  lessons from 10 consecutive patients. Medicine (Baltimore), Lee, C.K., Ahn, M.S., Lee, E.Y., Shin, J.H., Cho, Y.S., Ha, H.K., Yoo, B., and
91, 57–66. Moon, H.B. (2002). Acute abdominal pain in systemic lupus erythema-
Khosroshahi, A., Carruthers, M.N., Stone, J.H., Shinagare, S., Sainani, N., tosus: focus on lupus enteritis (gastrointestinal vasculitis). Annals of the
Hasserjian, R.P., and Deshpande, V. (2013). Rethinking Ormond’s dis- Rheumatic Diseases, 61, 547–50.
ease: ‘idiopathic’ retroperitoneal fibrosis in the era of IgG4-related dis- Lee, L.A. and Norris, D.A. (1989). Mechanisms of cutaneous tissue damage in
ease. Medicine (Baltimore), 92, 82–91. lupus erythematosus. Immunology Series, 46, 359–86.
Khosroshahi, A., Deshpande, V., and Stone, J.H. (2011). The clinical and Lee, S.S. and Lawton, J.M. (1992). Antimyeloperoxidase antibody in systemic
pathological features of IgG(4)-related disease. Current Rheumatology lupus erythematosus. Journal of Internal Medicine, 232, 283–4.
Reports, 13, 473–81. Lennon, V.A., Wingerchuk, D.M., Kryzer, T.J., Pittock, S.J., Lucchinetti, C.F.,
Khosroshahi, A. and Stone, J.H. (2011). A clinical overview of IgG4-related Fujihara, K., Nakashima, I., and Weinshenker, B.G. (2004). A serum
systemic disease. Current Opinion in Rheumatology, 23, 57–66. autoantibody marker of neuromyelitis optica: distinction from multiple
Kim, E.A., Lee, K.S., Johkoh, T., Kim, T.S., Suh, G.Y., Kwon, O.J., and Han, J. sclerosis. Lancet, 364, 2106–12.
(2002). Interstitial lung diseases associated with collagen vascular dis- Lesprit, P., Mouloud, F., Bierling, P., Schaeffer, A., Cesaro, P., Brun-Buisson,
eases: radiologic and histopathologic findings. Radiographics, 22, S151–65. C., and Godeau, B. (1996). Prolonged remission of SLE-associated poly-
Kim, R.C., Collins, G.H., and Parisi, J.E. (1982). Rheumatoid nodule for- radiculoneuropathy after a single course of intravenous immunoglobu-
mation within the choroid plexus. Report of a second case. Archives of lin. Scandinavian Journal of Rheumatology, 25, 177–9.
Pathology and Laboratory Medicine, 106, 83–4. Letko, E., Zafirakis, P., Baltatzis, S., Voudouri, A., Livir-Rallatos, C., and
Kinsella, T.D., Johnson, L.G., and Ian, R. (1974). Cardiovascular manifesta- Foster, C.S. (2002). Relapsing polychondritis: a clinical review. Seminars
tions of ankylosing spondylitis. Canadian Medical Association Journal, in Arthritis and Rheumatism, 31, 384–95.
111, 1309–11. Li, J.S., Liu, M.F., and Lei, H.Y. (1996). Characterization of anti-endothelial cell
Klinkhoff, A.V., Beattie, C.W., and Chalmers, A. (1986). Retinopathy in sys- antibodies in the patients with systemic lupus erythematosus: a potential
temic lupus erythematosus: relationship to disease activity. Arthritis and marker for disease activity. Clinical Immunology and Immunopathology,
Rheumatism, 29, 1152–6. 79, 211–6.
Knab, J., Goos, M., and Dissemond, J. (2005). Successful treatment of a Lindsay, M.K., Tavadia, H.B., Whyte, A.S., Lee, P., and Webb, J. (1973). Acute
leg ulcer occurring in a rheumatoid arthritis patient under lefluno- abdomen in rheumatoid arthritis due to necrotizing arteritis. British
mide therapy. Journal of the European Academy of Dermatology and Medical Journal, 2 (5866), 592–3.
Venereology, 19, 243–6. Lindvall, B., Bengtsson, A., Ernerudh, J., and Eriksson, P. (2002). Subclinical
Ko, S.F., Lee, T.Y., Cheng, T.T., Ng, S.H., Lai, H.M., Cheng, Y.F., and Tsai, C.C. myositis is common in primary Sjogren’s syndrome and is not related to
(1997). CT findings at lupus mesenteric vasculitis. Acta Radiologica, 38, muscle pain. Journal of Rheumatology, 29, 717–25.
115–20. Liote, F. and Osterland, C.K. (1994). Autonomic neuropathy in systemic
Koenigsberg, R.A., Klahr, J., Zito, J.L., Patel, M., and Carsons, S. (1995). lupus erythematosus:  cardiovascular autonomic function assessment.
Magnetic resonance imaging of cauda equina syndrome in ankylosing Annals of the Rheumatic Diseases, 53, 671–4.
spondylitis: a case report. Journal of Neuroimaging, 5, 46–8. Liu, M.F., Lee, J.H., Weng, T.H., and Lee, Y.Y. (1998). Clinical experience of
Konttinen, Y.T., Kinnunen, E., von Bonsdorff, M., Lillqvist, P., Immonen, I., 13 cases with severe pulmonary hemorrhage in systemic lupus erythe-
Bergroth, V., Segerberg-Konttinen, M., and Friman, C. (1987). Acute matosus with active nephritis. Scandinavian Journal of Rheumatology,
transverse myelopathy successfully treated with plasmapheresis and 27, 291–5.
prednisone in a patient with primary Sjogren’s syndrome. Arthritis and Liu, V. and Mackool, B.T. (2003). Mycophenolate in dermatology. Journal of
Rheumatism, 30, 339–44. Dermatological Treatment, 14, 203–11.
Kurne, A., Karabudak, R., Karadag, O., Yalcin-Cakmakli, G., Karli-Oguz, K., Loffeld, R.J. and van Weel, T.F. (1993). Tamoxifen for retroperitoneal fibrosis.
Yavuz, K., Calguneri, M., and Topcuoglu, M.A. (2009). An unusual cen- Lancet, 341, 382.
tral nervous system involvement in rheumatoid arthritis: combination Lorber, M., Gershwin, M.E., and Shoenfeld, Y. (1994). The coexistence of sys-
of pachymeningitis and cerebral vasculitis. Rheumatology International, temic lupus erythematosus with other autoimmune diseases: the kalei-
29, 1349–53. doscope of autoimmunity. Seminars in Arthritis and Rheumatism, 24,
Kushwaha, S.S. and Steinberg, V.L. (1992). Cauda equina syndrome asso- 105–13.
ciated with ankylosing spondylitis in a female. Postgraduate Medical Magro, C.M., Crowson, A.N., and Peeling, R. (1995). Vasculitis as the basis of
Journal, 68, 485–6. cutaneous lesions in Reiter’s disease. Human Pathology, 26, 633–8.
Kwok, S.K., Moon, S.J., Ju, J.H., Park, K.S., Kim, W.U., Cho, C.S., Kim, H.Y., Maki-Petaja, K.M., Elkhawad, M., Cheriyan, J., Joshi, F.R., Ostor, A.J.,
and Park, S.H. (2011). Diffuse alveolar hemorrhage in systemic lupus Hall, F.C., Rudd, J.H., and Wilkinson, I.B. (2012). Anti-tumor necro-
erythematosus: risk factors and clinical outcome: results from affiliated sis factor-alpha therapy reduces aortic inflammation and stiffness in
hospitals of Catholic University of Korea. Lupus, 20, 102–7. patients with rheumatoid arthritis. Circulation, 126, 2473–80.
Laing, T.J. (1988). Gastrointestinal vasculitis and pneumatosis intestinalis due Makol, A., Gibson, L.E., and Michet, C.J. (2012). Successful use of interleukin
to systemic lupus erythematosus: successful treatment with pulse intra- 6 antagonist tocilizumab in a patient with refractory cutaneous lupus
venous cyclophosphamide. American Journal of Medicine, 85, 555–8. and urticarial vasculitis. Journal of Clinical Rheumatology, 18, 92–5.
Laman, S.D. and Provost, T.T. (1994). Cutaneous manifestations of lupus ery- Malaviya, A.N., Raina, V., Mittal, V., Narayanan, K., Malhotra, K.K., Bhuyan,
thematosus. Rheumatic Diseases Clinics of North America, 20, 195–212. U.N., and Mehra, N.K. (1981). Glomerulonephritis in seronegative
Lang-Lazdunski, L., Hvass, U., Paillole, C., Pansard, Y., and Langlois, J. spondylarthritis syndrome. Arthritis and Rheumatism, 24, 751–2.
(1995). Cardiac valve replacement in relapsing polychondritis. A review. Manzi, S., Meilahn, E.N., Rairie, J.E., Conte, C.G., Medsger, T.A., Jr,
Journal of Heart Valve Disease, 4, 227–35. Jansen-McWilliams, L., D’Agostino, R.B., and Kuller, L.H. (1997).
Lavalle, C., Pizarro, S., Drenkard, C., Sanchez-Guerrero, J., and Age-specific incidence rates of myocardial infarction and angina in
Alarcon-Segovia, D. (1990). Transverse myelitis: a manifestation of sys- women with systemic lupus erythematosus:  comparison with the
temic lupus erythematosus strongly associated with antiphospholipid Framingham Study. American Journal of Epidemiology, 145, 408–15.
antibodies. Journal of Rheumatology, 17, 34–7. Marcolongo, R., Tavolini, I.M., Laveder, F., Busa, M., Noventa, F., Bassi, P.,
Leatherman, J.W., Davies, S.F., and Hoidal, J.R. (1984). Alveolar hemorrhage and Semenzato, G. (2004). Immunosuppressive therapy for idiopathic
syndromes: diffuse microvascular lung hemorrhage in immune and idi- retroperitoneal fibrosis: a retrospective analysis of 26 cases. American
opathic disorders. Medicine (Baltimore), 63, 343–61. Journal of Medicine, 116, 194–7.
CHAPTER 33 vasculitis in primary connective tissue diseases 461

Martens, P.B., Goronzy, J.J., Schaid, D., and Weyand, C.M. (1997). Expansion patients with HLA-B27 uveitis:  a study of 175 cases. Ophthalmology,
of unusual CD4+ T cells in severe rheumatoid arthritis. Arthritis and 111, 802–9.
Rheumatism, 40, 1106–14. Morgen, K., McFarland, H.F., and Pillemer, S.R. (2004). Central nervous sys-
Martens, P.B., Moder, K.G., and Ahmed, I. (1999). Lupus panniculitis: clini- tem disease in primary Sjogrens syndrome: the role of magnetic reso-
cal perspectives from a case series. Journal of Rheumatology, 26, 68–72. nance imaging. Seminars in Arthritis and Rheumatism, 34, 623–30.
Martinez-Taboada, V.M., Alonso, R.B., Armona, J., Fernandez-Sueiro, J.L., Mori, K., Iijima, M., Koike, H., Hattori, N., Tanaka, F., Watanabe, H., Katsuno,
Gonzalez, V.C., and Rodriguez-Valverde, V. (1996). Mononeuritis mul- M., Fujita, A., Aiba, I., Ogata, A., Saito, T., Asakura, K., Yoshida, M.,
tiplex in systemic lupus erythematosus: response to pulse intravenous Hirayama, M., and Sobue, G. (2005). The wide spectrum of clinical
cyclophosphamide. Lupus, 5, 74–6. manifestations in Sjogren’s syndrome-associated neuropathy. Brain, 128,
Marzano, A., Trapani, A., Leone, N., Actis, G.C., and Rizzetto, M. (2001). 2518–34.
Treatment of idiopathic retroperitoneal fibrosis using cyclosporin. Morozumi, S., Kawagashira, Y., Iijima, M., Koike, H., Hattori, N., Katsuno,
Annals of the Rheumatic Diseases, 60, 427–8. M., Tanaka, F., and Sobue, G. (2009). Intravenous immunoglobulin
Matsumoto, T., Kobayashi, S., Shimizu, H., Nakajima, M., Watanabe, S., treatment for painful sensory neuropathy associated with Sjogren’s syn-
Kitami, N., Sato, N., Abe, H., Aoki, Y., Hoshi, T., and Hashimoto, H. drome. Journal of the Neurological Sciences, 279, 57–61.
(2000). The liver in collagen diseases:  pathologic study of 160 cases Morris, P.B., Imber, M.J., Heinsimer, J.A., Hlatky, M.A., and Reimer, K.A.
with particular reference to hepatic arteritis, primary biliary cirrhosis, (1986). Rheumatoid arthritis and coronary arteritis. American Journal
autoimmune hepatitis and nodular regenerative hyperplasia of the liver. of Cardiology, 57, 689–90.
Liver, 20, 366–73. Moutsopoulos, H.M., Balow, J.E., Lawley, T.J., Stahl, N.I., Antonovych, T.T.,
Matsumoto, T., Yoshimine, T., Shimouchi, K., Shiotu, H., Kuwabara, N., and Chused, T.M. (1978). Immune complex glomerulonephritis in sicca
Fukuda, Y., and Hoshi, T. (1992). The liver in systemic lupus erythema- syndrome. American Journal of Medicine, 64, 955–60.
tosus: pathologic analysis of 52 cases and review of Japanese Autopsy Munoz-Fernandez, S., Hidalgo, V., Fernandez-Melon, J., Schlincker,
Registry Data. Human Pathology, 23, 1151–8. A., Bonilla, G., Ruiz-Sancho, D., Fonseca, A., Gijon-Banos, J., and
McAdam, L.P., O’Hanlan, M.A., Bluestone, R., and Pearson, C.M. (1976). Martin-Mola, E. (2003). Sulfasalazine reduces the number of flares of
Relapsing polychondritis: prospective study of 23 patients and a review acute anterior uveitis over a one-year period. Journal of Rheumatology,
of the literature. Medicine (Baltimore), 55, 193–215. 30, 1277–9.
McRorie, E.R., Ruckley, C.V., and Nuki, G. (1998). The relevance of Munoz-Fernandez, S. and Martin-Mola, E. (2006). Uveitis. Best Practice and
large-vessel vascular disease and restricted ankle movement to the Research Clinical Rheumatology, 20, 487–505.
aetiology of leg ulceration in rheumatoid arthritis. British Journal of Myers, J.L. and Katzenstein, A.A. (1986). Microangiitis in lupus-induced pul-
Rheumatology, 37, 1295–8. monary hemorrhage. American Journal of Clinical Pathology, 85, 552–6.
Medina, F., Ayala, A., Jara, L.J., Becerra, M., Miranda, J.M., and Fraga, A. Nagahama, T., Matsui, T., Matsumura, M., Matake, H., Tsuda, S., Sakurai, T.,
(1997). Acute abdomen in systemic lupus erythematosus:  the impor- Yao, T., and Schlemper, R. (2000). Rheumatoid arthritis accompanied by
tance of early laparotomy. American Journal of Medicine, 103, 100–5. colonic lesions. Internal Medicine, 39, 235–8.
Mekinian, A., Ravaud, P., Hatron, P.Y., Larroche, C., Leone, J., Gombert, B., Narshi, C.B. and Allard, S.A. (2012). Sustained response to tocilizumab,
Hamidou, M., Cantagrel, A., Marcelli, C., Rist, S., Breban, M., Launay, anti-IL-6 antibody, following anti-TNF-alpha failure in a patient
D., Fain, O., Gottenberg, J.E., and Mariette, X. (2012a). Efficacy of rituxi- with relapsing polychondritis complicated by aortitis. Rheumatology
mab in primary Sjogren’s syndrome with peripheral nervous system (Oxford), 51, 952–3.
involvement:  results from the AIR registry. Annals of the Rheumatic Navarro, M., Cervera, R., Font, J., Reverter, J.C., Monteagudo, J., Escolar, G.,
Diseases, 71, 84–7. Lopez-Soto, A., Ordinas, A., and Ingelmo, M. (1997). Anti-endothelial
Mekinian, A., Ravaud, P., Larroche, C., Hachulla, E., Gombert, B., cell antibodies in systemic autoimmune diseases: prevalence and clinical
Blanchard-Delaunay, C., Cantagrel, A., Fain, O., Sibilia, J., Gottenberg, significance. Lupus, 6, 521–6.
J.E., and Mariette, X. (2012b). Rituximab in central nervous system man- Newbold, K.M., Allum, W.H., Downing, R., Symmons, D.P., and Oates, G.D.
ifestations of patients with primary Sjogren’s syndrome: results from the (1987). Vasculitis of the gall bladder in rheumatoid arthritis and sys-
AIR registry. Clinical and Experimental Rheumatology, 30, 208–12. temic lupus erythematosus. Clinical Rheumatology, 6, 287–9.
Meroni, P.L., Tincani, A., Sepp, N., Raschi, E., Testoni, C., Corsini, E., Nguyen, V.A., Gotwald, T., Prior, C., Oberrnoser, G., and Sepp, N. (2005).
Cavazzana, I., Pellegrini, S., and Salmaggi, A. (2003). Endothelium and Acute pulmonary edema, capillaritis and alveolar hemorrhage: pulmo-
the brain in CNS lupus. Lupus, 12, 919–28. nary manifestations coexistent in antiphospholipid syndrome and sys-
Michet, C.J. (1990). Vasculitis and relapsing polychondritis. Rheumatic temic lupus erythematosus? Lupus, 14, 557–60.
Diseases Clinics of North America, 16, 441–4. Nicholls, A., Snaith, M.L., Maini, R.N., and Scott, J.T. (1973).
Mills, J.A. (1994). Systemic lupus erythematosus. New England Journal of Proceedings: Controlled trial of azathioprine in rheumatoid vasculitis.
Medicine, 330, 1871–9. Annals of the Rheumatic Diseases, 32, 589–91.
Mitsias, P. and Levine, S.R. (1994). Large cerebral vessel occlusive disease in Nishijima, C., Hatta, N., Inaoki, M., Sakai, H., and Takehara, K. (1999).
systemic lupus erythematosus. Neurology, 44, 385–93. Urticarial vasculitis in systemic lupus erythematosus: fair response to
Mizutani, A., Tanaka, I., Katayama, M., Oshima, H., Komatsu, Y., Asano, S., prednisolone/dapsone and persistent hypocomplementemia. European
Kato, K., Matsumura, H., Ishii, K., Miyama, H., Nagai, T., Kato, K., Fukaya, Journal of Dermatology, 9, 54–6.
S., Yoshida, S., Hasegawa, M., Kawashima, S., and Torikai, K. (2000). [A case Nishiya, K., Chikazawa, H., Nishimura, S., Hisakawa, N., and Hashimoto,
of myeloperoxidase-antineutrophil cytoplasmic antibody (MPO-ANCA) K. (1997). Anti-neutrophil cytoplasmic antibody in patients with sys-
related glomerulonephritis associated with systemic sclerosis treated by temic lupus erythematosus is unrelated to clinical features. Clinical
steroid pulse therapy: a case report]. Ryumachi, 40, 828–32. Rheumatology, 16, 70–5.
Moder, K.G., Miller, T.D., and Tazelaar, H.D. (1999). Cardiac involvement Nobrega, T.P., Klodas, E., Breen, J.F., Liggett, S.P., Higano, S.T., and Reeder,
in systemic lupus erythematosus. Mayo Clinic Proceedings, 74, 275–84. G.S. (1996). Giant coronary artery aneurysms and myocardial infarc-
Momeni, M., Taylor, N., and Tehrani, M. (2011). Cardiopulmonary manifes- tion in a patient with systemic lupus erythematosus. Catheterization and
tations of ankylosing spondylitis. International Journal of Rheumatology, Cardiovascular Diagnosis, 39, 75–9.
2011, 728471. Nyhall-Wahlin, B.M., Petersson, I.F., Jacobsson, C., Geborek, P., Nilsson, J.A.,
Monnet, D., Breban, M., Hudry, C., Dougados, M., and Brezin, A.P. (2004). Nilsson, K., Jacobsson, L.T., and Turesson, C. (2012). Extra-articular
Ophthalmic findings and frequency of extraocular manifestations in manifestations in a community-based sample of patients with
462 SECTION 5 vasculitic diseases and syndromes and related disorders

rheumatoid arthritis: incidence and relationship to treatment with TNF Peponis, V., Kyttaris, V.C., Tyradellis, C., Vergados, I., and Sitaras, N.M.
inhibitors. Scandinavian Journal of Rheumatology, 41, 434–7. (2006). Ocular manifestations of systemic lupus erythematosus: a clini-
Oddis, C.V., Eisenbeis, C.H. Jr, Reidbord, H.E., Steen, V.D., and Medsger, cal review. Lupus, 15, 3–12.
T.A. Jr (1987). Vasculitis in systemic sclerosis: association with Sjogren’s Peri, Y., Agmon-Levin, N., Theodor, E., and Shoenfeld, Y. (2012). Sjogren’s
syndrome and the CREST syndrome variant. Journal of Rheumatology, syndrome, the old and the new. Best Practice and Research Clinical
14, 942–8. Rheumatology, 26, 105–17.
Oh, D.H., Jun, J.B., Kim, H.T., Lee, S.W., Jung, S.S., Lee, I.H., and Kim, S.Y. Petri, M., Perez-Gutthann, S., Spence, D., and Hochberg, M.C. (1992). Risk
(2001). Transverse myelitis in a patient with long-standing ankylosing factors for coronary artery disease in patients with systemic lupus ery-
spondylitis. Clinical and Experimental Rheumatology, 19, 195–6. thematosus. American Journal of Medicine, 93, 513–9.
Ohno, T., Matsuda, I., Furukawa, H., and Kanoh, T. (1994). Recovery from Pisoni, C.N., Obermoser, G., Cuadrado, M.J., Sanchez, F.J., Karim, Y., Sepp,
rheumatoid cerebral vasculitis by low-dose methotrexate. Internal N.T., Khamashta, M.A., and Hughes, G.R. (2005). Skin manifestations of
Medicine, 33, 615–20. systemic lupus erythematosus refractory to multiple treatment modali-
Oien, R.F., Hakansson, A., and Hansen, B.U. (2001). Leg ulcers in patients ties: poor results with mycophenolate mofetil. Clinical and Experimental
with rheumatoid arthritis—a prospective study of aetiology, wound Rheumatology, 23, 393–6.
healing and pain reduction after pinch grafting. Rheumatology (Oxford), Praprotnik, S., Blank, M., Meroni, P.L., Rozman, B., Eldor, A., and Shoenfeld,
40, 816–20. Y. (2001). Classification of anti-endothelial cell antibodies into anti-
Omdal, R., Loseth, S., Torbergsen, T., Koldingsnes, W., Husby, G., and bodies against microvascular and macrovascular endothelial cells: the
Mellgren, S.I. (2001). Peripheral neuropathy in systemic lupus erythe- pathogenic and diagnostic implications. Arthritis and Rheumatism, 44,
matosus—a longitudinal study. Acta Neurologica Scandinavica, 103, 1484–94.
386–91. Price, E.J., Rigby, S.P., Clancy, U., and Venables, P.J. (1998). A double blind
Omote, A., Muramatsu, M., Sugimoto, Y., Hosono, S., Murakami, R., Tanaka, placebo controlled trial of azathioprine in the treatment of primary
H., Watanabe, Y., Sano, H., and Kato, K. (1997). Myeloperoxidase-specific Sjogren’s syndrome. Journal of Rheumatology, 25, 896–9.
anti-neutrophil cytoplasmic autoantibodies—related scleroderma renal Provenzale, J.M., Barboriak, D.P., Gaensler, E.H., Robertson, R.L., and
crisis treated with double-filtration plasmapheresis. Internal Medicine, Mercer, B. (1994). Lupus-related myelitis: serial MR findings. American
36, 508–13. Journal of Neuroradiology, 15, 1911–7.
O’Neill, T.W. and Bresnihan, B. (1992). The heart in ankylosing spondylitis. Provenzale, J.M. and Ortel, T.L. (1995). Anatomic distribution of venous
Annals of the Rheumatic Diseases, 51, 705–6. thrombosis in patients with antiphospholipid antibody: imaging find-
Pachman, L.M. (1995). An update on juvenile dermatomyositis. Current ings. American Journal of Roentgenology, 165, 365–8.
Opinion in Rheumatology, 7, 437–41. Puechal, X., Said, G., Hilliquin, P., Coste, J., Job-Deslandre, C., Lacroix, C.,
Pachman, L.M., Hayford, J.R., Chung, A., Daugherty, C.A., Pallansch, and Menkes, C.J. (1995). Peripheral neuropathy with necrotizing vascu-
M.A., Fink, C.W., Gewanter, H.L., Jerath, R., Lang, B.A., Sinacore, J., litis in rheumatoid arthritis. A clinicopathologic and prognostic study of
Szer, I.S., Dyer, A.R., and Hochberg, M.C. (1998). Juvenile dermato- thirty-two patients. Arthritis and Rheumatism, 38, 1618–29.
myositis at diagnosis: clinical characteristics of 79 children. Journal of Quemeneur, T., Mouthon, L., Cacoub, P., Meyer, O., Michon-Pasturel, U.,
Rheumatology, 25, 1198–204. Vanhille, P., Hatron, P.Y., Guillevin, L., and Hachulla, E. (2013). Systemic
Pagnoux, C., Mahr, A., Cohen, P., and Guillevin, L. (2005). Presentation vasculitis during the course of systemic sclerosis: report of 12 cases and
and outcome of gastrointestinal involvement in systemic necrotizing review of the literature. Medicine (Baltimore), 92, 1–9.
vasculitides:  analysis of 62 patients with polyarteritis nodosa, micro- Ramos-Casals, M., Anaya, J.M., Garcia-Carrasco, M., Rosas, J., Bove, A.,
scopic polyangiitis, Wegener granulomatosis, Churg-Strauss syndrome, Claver, G., Diaz, L.A., Herrero, C., and Font, J. (2004). Cutaneous vascu-
or rheumatoid arthritis-associated vasculitis. Medicine (Baltimore), 84, litis in primary Sjogren syndrome: classification and clinical significance
115–28. of 52 patients. Medicine (Baltimore), 83, 96–106.
Palejwala, N.V., Walia, H.S., and Yeh, S. (2012). Ocular manifestations of Ramos-Casals, M., Font, J., Garcia-Carrasco, M., Brito, M.P., Rosas, J.,
systemic lupus erythematosus: a review of the literature. Autoimmune Calvo-Alen, J., Pallares, L., Cervera, R., and Ingelmo, M. (2002). Primary
Diseases, 2012, 290898. Sjogren syndrome: hematologic patterns of disease expression. Medicine
Palm, O., Garen, T., Berge, E.T., Jensen, J.L., Lund, M.B., Aalokken, T.M., and (Baltimore), 81, 281–92.
Gran, J.T. (2013). Clinical pulmonary involvement in primary Sjogren’s Ramos-Casals, M., Nardi, N., Lagrutta, M., Brito-Zeron, P., Bove, A.,
syndrome:  prevalence, quality of life and mortality—a retrospective Delgado, G., Cervera, R., Ingelmo, M., and Font, J. (2006). Vasculitis in
study based on registry data. Rheumatology (Oxford), 52, 173–9. systemic lupus erythematosus: prevalence and clinical characteristics in
Paloheimo, J.A., Julkunen, H., Siltanen, P., and Kajander, A. (1966). Takayasu’s 670 patients. Medicine (Baltimore), 85, 95–104.
arteritis and ankylosing spondylitis. Report of four cases. Acta Medica Ramos-Casals, M., Solans, R., Rosas, J., Camps, M.T., Gil, A., Pino-Montes,
Scandinavica, 179, 77–85. J., Calvo-Alen, J., Jimenez-Alonso, J., Mico, M.L., Beltran, J., Belenguer,
Panush, R.S., Katz, P., Longley, S., Carter R, Love, J., and Stanley, H. (1983). R., and Pallares, L. (2008). Primary Sjogren syndrome in Spain: clini-
Rheumatoid vasculitis:  diagnostic and therapeutic decisions. Clinical cal and immunologic expression in 1010 patients. Medicine (Baltimore),
Rheumatology, 2, 321–30. 87, 210–9.
Papadaki, T.G., Zacharopoulos, I.P., Papaliodis, G., Iaccheri, B., Fiore, T., and Rapoport, R.J., Kozin, F., Mackel, S.E., and Jordon, R.E. (1980). Cutaneous
Foster, C.S. (2006). Plasmapheresis for lupus retinal vasculitis. Archives vascular immunofluorescence in rheumatoid arthritis. Correlation
of Ophthalmology, 124, 1654–6. with circulating immune complexes and vasculitis. American Journal of
Papiris, S.A., Maniati, M., Constantopoulos, S.H., Roussos, C., Moutsopoulos, Medicine, 68, 325–31.
H.M., and Skopouli, F.N. (1999). Lung involvement in primary Sjogren’s Rhee, R.Y., Gloviczki, P., Luthra, H.S., Stanson, A.W., Bower, T.C., and
syndrome is mainly related to the small airway disease. Annals of the Cherry, K.J., Jr (1994). Iliocaval complications of retroperitoneal fibro-
Rheumatic Diseases, 58, 61–4. sis. American Journal of Surgery, 168, 179–83.
Pappas, G. and Johnson, M. (1972). Mitral and aortic valvular insufficiency in Richter, C., Wanke, L., Steinmetz, J., Reinhold-Keller, E., and Gross, W.L.
chronic relapsing polychondritis. Archives of Surgery, 104, 712–4. (2000). Mononeuritis secondary to rheumatoid arthritis responds to
Paran, D., Fireman, E., and Elkayam, O. (2004). Pulmonary disease in etanercept. Rheumatology (Oxford), 39, 1436–7.
systemic lupus erythematosus and the antiphospholpid syndrome. Roberts, W.C. and High, S.T. (1999). The heart in systemic lupus erythemato-
Autoimmunity Reviews, 3, 70–5. sus. Current Problems in Cardiology, 24, 1–56.
CHAPTER 33 vasculitis in primary connective tissue diseases 463

Rogers, S.J., Williams, C.S., and Roman, G.C. (2004). Myelopathy in Scott, D.G. and Bacon, P.A. (1984). Intravenous cyclophosphamide plus
Sjogren’s syndrome: role of nonsteroidal immunosuppressants. Drugs, methylprednisolone in treatment of systemic rheumatoid vasculitis.
64, 123–32. American Journal of Medicine, 76, 377–84.
Roldan, C.A., Chavez, J., Wiest, P.W., Qualls, C.R., and Crawford, M.H. Scott, D.G., Bacon, P.A., Allen, C., Elson, C.J., and Wallington, T. (1981a). IgG
(1998). Aortic root disease and valve disease associated with anky- rheumatoid factor, complement and immune complexes in rheumatoid
losing spondylitis. Journal of the American College of Cardiology, 32, synovitis and vasculitis: comparative and serial studies during cytotoxic
1397–404. therapy. Clinical and Experimental Immunology, 43, 54–63.
Rosen, O., Thiel, A., Massenkeil, G., Hiepe, F., Haupl, T., Radtke, H., Scott, D.G., Bacon, P.A., Bothamley, J.E., Allen, C., Elson, C.J., and
Burmester, G.R., Gromnica-Ihle, E., Radbruch, A., and Arnold, R. Wallington, T.B. (1981b). Plasma exchange in rheumatoid vasculitis.
(2000). Autologous stem-cell transplantation in refractory autoimmune Journal of Rheumatology, 8, 433–9.
diseases after in vivo immunoablation and ex vivo depletion of mononu- Scott, D.G., Bacon, P.A., and Tribe, C.R. (1981c). Systemic rheumatoid vas-
clear cells. Arthritis and Rheumatism, 2, 327–36. culitis: a clinical and laboratory study of 50 cases. Medicine (Baltimore),
Rothfield, N., Sontheimer, R.D., and Bernstein, M. (2006). Lupus erythema- 60, 288–97.
tosus: systemic and cutaneous manifestations. Clinical Dermatology, 24, See, Y., Martin, K., Rooney, M., and Woo, P. (1997). Severe juvenile dermato-
348–62. myositis complicated by pancreatitis. British Journal of Rheumatology,
Rothova, A., Buitenhuis, H.J., Meenken, C., Brinkman, C.J., Linssen, A., 36, 912–6.
Alberts, C., Luyendijk, L., and Kijlstra, A. (1992). Uveitis and systemic Sen, D. and Isenberg, D.A. (2003). Antineutrophil cytoplasmic autoantibod-
disease. British Journal of Ophthalmology, 76, 137–41. ies in systemic lupus erythematosus. Lupus, 12, 651–8.
Rovaris, M., Viti, B., Ciboddo, G., Gerevini, S., Capra, R., Iannucci, G., Comi, Seror, R., Bootsma, H., Bowman, S.J., Dorner, T., Gottenberg, J.E., Mariette,
G., and Filippi, M. (2000). Brain involvement in systemic immune medi- X., Ramos-Casals, M., Ravaud, P., Theander, E., Tzioufas, A., and Vitali,
ated diseases: magnetic resonance and magnetisation transfer imaging C. (2012). Outcome measures for primary Sjogren’s syndrome. Journal
study. Journal of Neurology Neurosurgery and Psychiatry, 68, 170–77. of Autoimmunity, 39, 97–102.
Sanchez, N.P., Peters, M.S., and Winkelmann, R.K. (1981). The histopa- Shen, M., Zeng, X., Tian, X., Zhang, F., Zeng, X., Zhang, X., and Xu, W.
thology of lupus erythematosus panniculitis. Journal of the American (2010). Diffuse alveolar hemorrhage in systemic lupus erythematosus: a
Academy of Dermatology, 5, 673–80. retrospective study in China. Lupus, 19, 1326–30.
Sant, S.M. and O’Connell, D. (1995). Cauda equina syndrome in ankylos- Shu, K.H., Lian, J.D., Yang, Y.F., Lu, Y.S., Wang, J.Y., Lan, J.L., and Chou,
ing spondylitis:  a case report and review of the literature. Clinical G. (1986). Glomerulonephritis in ankylosing spondylitis. Clinical
Rheumatology, 14, 224–6. Nephrology, 25, 169–74.
Santos-Ocampo, A.S., Mandell, B.F., and Fessler, B.J. (2000). Alveolar hemor- Simmons-O’Brien, E., Chen, S., Watson, R., Antoni, C., Petri, M., Hochberg,
rhage in systemic lupus erythematosus: presentation and management. M., Stevens, M.B., and Provost, T.T. (1995). One hundred anti-Ro (SS-A)
Chest, 118, 1083–90. antibody positive patients: a 10-year follow-up. Medicine (Baltimore),
Satko, S.G., Iskandar, S.S., and Appel, R.G. (2000). IgA nephropathy and 74, 109–30.
Reiter’s syndrome. Report of two cases and review of the literature. Skopouli, F.N., Dafni, U., Ioannidis, J.P., and Moutsopoulos, H.M. (2000).
Nephron, 84, 177–82. Clinical evolution, and morbidity and mortality of primary Sjogren’s
Saxe, P.A. and Altman, R.D. (1992). Takayasu’s arteritis syndrome associ- syndrome. Seminars in Arthritis and Rheumatism, 29, 296–304.
ated with systemic lupus erythematosus. Seminars in Arthritis and Skopouli, F.N., Jagiello, P., Tsifetaki, N., and Moutsopoulos, H.M. (1996).
Rheumatism, 21, 295–305. Methotrexate in primary Sjogren’s syndrome. Clinical and Experimental
Sayah, A. and English, J.C. III (2005). Rheumatoid arthritis:  a review of Rheumatology, 14, 555–8.
the cutaneous manifestations. Journal of the American Academy of Sokoloff, L. (1953). The heart in rheumatoid arthritis. American Heart
Dermatology, 53, 191–209. Journal, 45, 635–43.
Schnabel, A., Csernok, E., Isenberg, D.A., Mrowka, C., and Gross, W.L.
Somer, T. (1993). Thrombo-embolic and vascular complications in vasculitis
(1995). Antineutrophil cytoplasmic antibodies in systemic lupus erythe-
syndromes. European Heart Journal, 14 (Suppl. K), 24–9.
matosus. Prevalence, specificities, and clinical significance. Arthritis and
Somer, T. and Siltanen, P. (1970). Aneurysm of the descending thoracic aorta,
Rheumatism, 38, 633–7.
amyloidosis and renal carcinoma in a patient with ankylosing spondyli-
Schneider, H.A., Yonker, R.A., Katz, P., Longley, S., and Panush, R.S. (1985).
tis. American Journal of Medicine, 49, 408–15.
Rheumatoid vasculitis: experience with 13 patients and review of the
Spath, N.B., Amft, N., and Farquhar, D. (2012). Cerebral vasculitis in rheu-
literature. Seminars in Arthritis and Rheumatism, 14, 280–6.
matoid arthritis. QJM, in press.
Schroder, R., Urbach, H., and Zierz, S. (1994). Cauda equina syndrome with
multiple lumbar diverticula complicating long-standing ankylosing Stafford-Brady, F.J., Urowitz, M.B., Gladman, D.D., and Easterbrook, M.
spondylitis. Clinical Investigator, 72, 1056–9. (1988). Lupus retinopathy. Patterns, associations, and prognosis.
Arthritis and Rheumatism, 31, 1105–10.
Schwab, E.P., Schumacher, H.R. Jr, Freundlich, B., and Callegari, P.E. (1993).
Pulmonary alveolar hemorrhage in systemic lupus erythematosus. Stamp, L., Lambie, N., and O’Donnell, J. (2000). HLA-B27 associated spon-
Seminars in Arthritis and Rheumatism, 23, 8–15. dyloarthropathy and severe ascending aortitis. Journal of Rheumatology,
27, 2038–40.
Schwarz, M.I., Sutarik, J.M., Nick, J.A., Leff, J.A., Emlen, J.W., and Tuder, R.M.
(1995). Pulmonary capillaritis and diffuse alveolar hemorrhage. A pri- Stavrou, P., Murray, P.I., Batta, K., and Gordon, C. (2002). Acute ocular
mary manifestation of polymyositis. American Journal of Respiratory ischaemia and orbital inflammation associated with systemic lupus ery-
and Critical Care Medicine, 151, 2037–40. thematosus. British Journal of Ophthalmology, 86, 474–5.
Schwarz, M.I., Zamora, M.R., Hodges, T.N., Chan, E.D., Bowler, R.P., and Stewart, S.S., Ashizawa, T., Dudley, A.W., Jr, Goldberg, J.W., and Lidsky,
Tuder, R.M. (1998). Isolated pulmonary capillaritis and diffuse alveolar M.D. (1988). Cerebral vasculitis in relapsing polychondritis. Neurology,
hemorrhage in rheumatoid arthritis and mixed connective tissue dis- 38, 150–2.
ease. Chest, 113, 1609–15. Stone, J.R. (2011). Aortitis, periaortitis, and retroperitoneal fibrosis, as
Scofield, R.H. (2011). Vasculitis in Sjogren’s syndrome. Current Rheumatology manifestations of IgG4-related systemic disease. Current Opinion in
Reports, 13, 482–8. Rheumatology, 23, 88–94.
Scott, D.G. and Bacon, P.A. (1983). Rheumatoid vasculitis. Clinical Straub, R.H., Zeuner, M., Lock, G., Rath, H., Hein, R., Scholmerich, J., and
Rheumatology, 2, 311–4. Lang, B. (1996). Autonomic and sensorimotor neuropathy in patients
464 SECTION 5 vasculitic diseases and syndromes and related disorders

with systemic lupus erythematosus and systemic sclerosis. Journal of leukocytoclastic vasculitis. Journal of the American Academy of
Rheumatology, 23, 87–92. Dermatology, 31, 387–90.
Struthers, G.R., Scott, D.L., Delamere, J.P., Sheppeard, H., and Kitt, M. (1981). Turesson, C. and Jacobsson, L.T. (2004). Epidemiology of extra-articular
Smoking and rheumatoid vasculitis. Rheumatology International, manifestations in rheumatoid arthritis. Scandinavian Journal of
1, 145–6. Rheumatology, 33, 65–72.
Sultan, S.M., Ioannou, Y., and Isenberg, D.A. (1999). A review of gastroin- Turesson, C., Schaid, D.J., Weyand, C.M., Jacobsson, L.T., Goronzy, J.J.,
testinal manifestations of systemic lupus erythematosus. Rheumatology Petersson, I.F., Sturfelt, G., Nyhall-Wahlin, B.M., Truedsson, L., Dechant,
(Oxford), 38, 917–32. S.A., and Matteson, E.L. (2005). The impact of HLA-DRB1 genes on
Sundy, J.S., Jaffe, G.J., and McCallum, R.M. (2004). Extraarticular disease. In extra-articular disease manifestations in rheumatoid arthritis. Arthritis
Rheumatoid arthritis (ed. E.W. St Clair, D.S. Pisetsky, and B.F. Haynes), Research and Therapy, 7, R1386–93.
pp. 483–95. Lippincott Williams and Wilkins, Philadelphia, PA. Turesson, C., Weyand, C.M., and Matteson, E.L. (2004). Genetics of rheuma-
Sung, J.M., Hsu, S.C., Chen, F.F., and Huang, J.J. (2002). Systemic toid arthritis: Is there a pattern predicting extraarticular manifestations?
lupus erythematosus presented as non-inflammatory necrotiz- Arthritis and Rheumatism, 51, 853–63.
ing vasculopathy-induced ischemic glomerulopathy and small Tuveri, M., Generini, S., Matucci-Cerinic, M., and Aloe, L. (2000). NGF, a
vessels-related ischemic cardiomyopathy. Lupus, 11, 458–62. useful tool in the treatment of chronic vasculitic ulcers in rheumatoid
Swanepoel, C.R., Floyd, A., Allison, H., Learmonth, G.M., Cassidy, M.J., arthritis. Lancet, 356, 1739–40.
and Pascoe, M.D. (1983). Acute acalculous cholecystitis complicating Tzarouchi, L.C., Tsifetaki, N., Konitsiotis, S., Zikou, A., Astrakas, L., Drosos,
systemic lupus erythematosus: case report and review. British Medical A., and Argyropoulou, M.I. (2011). CNS involvement in primary Sjogren
Journal (Clinical Research Ed.), 286, 251–2. Syndrome: assessment of gray and white matter changes with MRI and
Takagi, H., Mori, Y., Umeda, Y., Fukumoto, Y., Kato, Y., Shimokawa, K., and voxel-based morphometry. American Journal of Roentgenology, 197,
Hirose, H. (2003). Abdominal aortic aneurysm with arteritis in ankylos- 1207–12.
ing spondylitis. Journal of Vascular Surgery, 38, 613–6. Vaglio, A., Greco, P., and Buzio, C. (2006a). Tamoxifen therapy for retroperi-
Takizawa, Y., Kanda, H., Sato, K., Kawahata, K., Yamaguchi, A., Uozaki, toneal fibrosis. Annals of Internal Medicine, 144, 619–20.
H., Shimizu, J., Tsuji, S., Misaki, Y., and Yamamoto, K. (2007). Vaglio, A., Salvarani, C., and Buzio, C. (2006b). Retroperitoneal fibrosis.
Polymyositis associated with focal mesangial proliferative glo- Lancet, 367, 241–51.
merulonephritis with depositions of immune complexes. Clinical Van den Bergh, B., Selleslag, D., Boelaert, J.R., Matthys, E.G., Schurgers,
Rheumatology, 26, 792–6. M., Vandecasteele, S., and De Vriese, A. (2005). Management of
Teitel, A.D., MacKenzie, C.R., Stern, R., and Paget, S.A. (1992). Laryngeal therapy-resistant systemic lupus erythematosus with rituximab: report
involvement in systemic lupus erythematosus. Seminars in Arthritis and of a case and review of the literature. Acta Clinica Belgica, 60, 102–5.
Rheumatism, 22, 203–14. van der Bijl, A.E., Allaart, C.F., Van Vugt, J., Van Duinen, S., and Breedveld,
ter Borg, E.J., Risselada, A.P., and Kelder, J.C. (2011). Relation of systemic F.C. (2005). Rheumatoid vasculitis treated with infliximab. Journal of
autoantibodies to the number of extraglandular manifestations in pri- Rheumatology, 32, 1607–9.
mary Sjogren’s Syndrome: a retrospective analysis of 65 patients in the van der Zee, J.M., Heurkens, A.H., van der Voort, E.A., Daha, M.R., and
Netherlands. Seminars in Arthritis and Rheumatism, 40, 547–51. Breedveld, F.C. (1991). Characterization of anti-endothelial antibodies
Tikly, M., Burgin, S., Mohanlal, P., Bellingan, A., and George, J. (1996). in patients with rheumatoid arthritis complicated by vasculitis. Clinical
Autoantibodies in black South Africans with systemic lupus erythe- and Experimental Rheumatology, 9, 589–94.
matosus:  spectrum and clinical associations. Clinical Rheumatology, van Laar, J.M., Smit, V.T., de Beus, W.M., Collee, G., Jansen, E.H., and
15, 143–7. Breedveld, F.C. (1998). Rheumatoid vasculitis presenting as appendici-
Toloza, S.M., Uribe, A.G., McGwin, G. Jr, Alarcon, G.S., Fessler, B.J., Bastian, tis. Clinical and Experimental Rheumatology, 16, 736–8.
H.M., Vila, L.M., Wu, R., Shoenfeld, Y., Roseman, J.M., and Reveille, van Laarhoven, H.W., Rooyer, F.A., van Engelen, B.G., van Dalen, R., and
J.D. (2004). Systemic lupus erythematosus in a multiethnic US cohort Berden, J.H. (2001). Guillain-Barre syndrome as presenting feature in a
(LUMINA). XXIII. Baseline predictors of vascular events. Arthritis and patient with lupus nephritis, with complete resolution after cyclophos-
Rheumatism, 50, 3947–57. phamide treatment. Nephrology Dialysis Transplantation, 16, 840–2.
Tomomatsu, J., Hamano, Y., Ando, J., Komatsu, N., and Sugimoto, K. (2012). van Woerkom, J.M., Kruize, A.A., Geenen, R., van Roon, E.N., Goldschmeding,
Non-myeloablative allogenic BMT for myelodysplastic syndrome suc- R., Verstappen, S.M., van Roon, J.A., and Bijlsma, J.W. (2007). Safety and
cessfully controlled accompanying relapsing polychondritis. Bone efficacy of leflunomide in primary Sjogren’s syndrome: a phase II pilot
Marrow Transplantation, 47, 742–3. study. Annals of the Rheumatic Diseases, 66, 1026–32.
Toussirot, E., Auge, B., Tiberghien, P., Chabod, J., Cedoz, J.P., and Wendling, Vij, R. and Strek, M.E. (2013). Diagnosis and treatment of connective tissue
D. (1999). HLA-DRB1 alleles and shared amino acid sequences in dis- disease-associated interstitial lung disease. Chest, 143, 814–24.
ease susceptibility and severity in patients from eastern France with Virdi, R.P., Bashir, A., Shahzad, G., Iqbal, J., and Mejia, J.O. (2012). Diffuse
rheumatoid arthritis. Journal of Rheumatology, 26, 1446–51. alveolar hemorrhage: a rare life-threatening condition in systemic lupus
Travis, R.C. and Byrne, P. (1987). The cauda equina syndrome:  a rare erythematosus. Case Reports in Pulmonology, 2012, 836017.
extra-articular manifestation of ankylosing spondylitis—a case report. Vitali, C., Bencivelli, W., Isenberg, D.A., Smolen, J.S., Snaith, M.L., Sciuto,
Australasian Radiology, 31, 395–6. M., d’Ascanio, A., and Bombardieri, S. (1992). Disease activity in sys-
Trendelenburg, M., Courvoisier, S., Spath, P.J., Moll, S., Mihatsch, M., Itin, temic lupus erythematosus: report of the Consensus Study Group of the
P., and Schifferli, J.A. (1999). Hypocomplementemic urticarial vasculitis European Workshop for Rheumatology Research. I. A descriptive analy-
or systemic lupus erythematosus? American Journal of Kidney Diseases, sis of 704 European lupus patients. European Consensus Study Group
34, 745–51. for Disease Activity in SLE. Clinical and Experimental Rheumatology,
Tribe, C.R., Scott, D.G., and Bacon, P.A. (1981). Rectal biopsy in the diagno- 10, 527–39.
sis of systemic vasculitis. Journal of Clinical Pathology, 34, 843–50. Vitali, C., Bombardieri, S., Jonsson, R., Moutsopoulos, H.M., Alexander, E.L.,
Tsokos, M., Lazarou, S.A., and Moutsopoulos, H.M. (1987). Vasculitis in pri- Carsons, S.E., Daniels, T.E., Fox, P.C., Fox, R.I., Kassan, S.S., Pillemer,
mary Sjogren’s syndrome. Histologic classification and clinical presenta- S.R., Talal, N., and Weisman, M.H. (2002). Classification criteria for
tion. American Journal of Clinical Pathology, 88, 26–31. Sjogren’s syndrome: a revised version of the European criteria proposed
Tsuchida, T., Furue, M., Kashiwado, T., and Ishibashi, Y. (1994). Bullous by the American-European Consensus Group. Annals of the Rheumatic
systemic lupus erythematosus with cutaneous mucinosis and Diseases, 61, 554–8.
CHAPTER 33 vasculitis in primary connective tissue diseases 465

Vlachoyiannopoulos, P.G., Karassa, F.B., Karakostas, K.X., Drosos, A.A., and Watts, R.A., Carruthers, D.M., and Scott, D.G. (1995). Epidemiology of sys-
Moutsopoulos, H.M. (1993). Systemic lupus erythematosus in Greece. temic vasculitis: changing incidence or definition? Seminars in Arthritis
Clinical features, evolution and outcome: a descriptive analysis of 292 and Rheumatism, 25, 28–34.
patients. Lupus, 2, 303–12. Watts, R.A., Carruthers, D.M., Symmons, D.P., and Scott, D.G. (1994). The
Vogelgesang, S.A., Gutierrez, J., Klipple, G.L., and Katona, I.M. (1995). incidence of rheumatoid vasculitis in the Norwich Health Authority.
Polyneuropathy in juvenile dermatomyositis. Journal of Rheumatology, British Journal of Rheumatology, 33, 832–3.
22, 1369–72. Weide, R., Heymanns, J., Pandorf, A., and Koppler, H. (2003). Successful
Vollertsen, R.S. and Conn, D.L. (1990). Vasculitis associated with rheuma- long-term treatment of systemic lupus erythematosus with rituximab
toid arthritis. Rheumatic Diseases Clinics of North America, 16, 445–61. maintenance therapy. Lupus, 12, 779–82.
Vollertsen, R.S., Conn, D.L., Ballard, D.J., Ilstrup, D.M., Kazmar, R.E., and Weiner, D.K. and Allen, N.B. (1991). Large vessel vasculitis of the central
Silverfield, J.C. (1986). Rheumatoid vasculitis: survival and associated nervous system in systemic lupus erythematosus: report and review of
risk factors. Medicine (Baltimore), 65, 365–75. the literature. Journal of Rheumatology, 18, 748–51.
Voskuyl, A.E., Emeis, J.J., Hazes, J.M., van Hogezand, R.A., Biemond, I., West, S.G. (1994). Neuropsychiatric lupus. Rheumatic Diseases Clinics of
and Breedveld, F.C. (1998a). Levels of circulating cellular fibronec- North America, 20, 129–58.
tin are increased in patients with rheumatoid vasculitis. Clinical and Weyand, C.M., Hicok, K.C., Conn, D.L., and Goronzy, J.J. (1992). The influ-
Experimental Rheumatology, 16, 429–34. ence of HLA-DRB1 genes on disease severity in rheumatoid arthritis.
Voskuyl, A.E., Hazes, J.M., Schreuder, G.M., Schipper, R.F., de Vries, R.R., Annals of Internal Medicine, 117, 801–6.
and Breedveld, F.C. (1997). HLA-DRB1, DQA1, and DQB1 genotypes Williams, C.S., Butler, E., and Roman, G.C. (2001). Treatment of myelopathy
and risk of vasculitis in patients with rheumatoid arthritis. Journal of in Sjogren syndrome with a combination of prednisone and cyclophos-
Rheumatology, 24, 852–5. phamide. Archives of Neurology, 58, 815–9.
Voskuyl, A.E., Hazes, J.M., Zwinderman, A.H., Paleolog, E.M., van der Meer, Wong, S.S. and Marks, R. (1994). Cutaneous vasculitis in psoriasis. Acta
F.J., Daha, M.R., and Breedveld, F.C. (2003). Diagnostic strategy for the Dermato-Venereologica, 74, 57–60.
assessment of rheumatoid vasculitis. Annals of the Rheumatic Diseases, Wright, R.A., O’Duffy, J.D., and Rodriguez, M. (1999). Improvement of
62, 407–13. myelopathy in Sjogren’s syndrome with chlorambucil and prednisone
Voskuyl, A.E., van Duinen, S.G., Zwinderman, A.H., Breedveld, F.C., and therapy. Neurology, 52, 386–8.
Hazes, J.M. (1998b). The diagnostic value of perivascular infiltrates in Wright, S.A., Convery, R.P., and Liggett, N. (2003). Pulmonary involvement
muscle biopsy specimens for the assessment of rheumatoid vasculitis. in Sjogren’s syndrome. Rheumatology (Oxford), 42, 697–8.
Annals of the Rheumatic Diseases, 57, 114–7. Yamashita, K., Yorioka, N., Kyuden, Y., Naito, T., Tanji, C., Ueda, C., Usui, K.,
Voskuyl, A.E., Zwinderman, A.H., Westedt, M.L., Vandenbroucke, J.P., Shigemoto, K., Harada, S., and Yamakido, M. (2000). A case of CREST
Breedveld, F.C., and Hazes, J.M. (1996). Factors associated with the devel- syndrome and myeloperoxidase-specific anti-neutrophil cytoplasmic
opment of vasculitis in rheumatoid arthritis: results of a case-control autoantibody-associated glomerulonephritis. Clinical Nephrology, 53,
study. Annals of the Rheumatic Diseases, 55, 190–2. 296–300.
Voyles, W.F., Searles, R.P., and Bankhurst, A.D. (1980). Myocardial infarction Yen, J.H., Moore, B.E., Nakajima, T., Scholl, D., Schaid, D.J., Weyand,
caused by rheumatoid vasculitis. Arthritis and Rheumatism, 23, 860–3. C.M., and Goronzy, J.J. (2001). Major histocompatibility complex
Vrethem, M., Lindvall, B., Holmgren, H., Henriksson, K.G., Lindstrom, F., class I-recognizing receptors are disease risk genes in rheumatoid arthri-
and Ernerudh, J. (1990). Neuropathy and myopathy in primary Sjogren’s tis. Journal of Experimental Medicine, 193, 1159–67.
syndrome:  neurophysiological, immunological and muscle biopsy Zamora, M.R., Warner, M.L., Tuder, R., and Schwarz, M.I. (1997). Diffuse
results. Acta Neurologica Scandinavica, 82, 126–31. alveolar hemorrhage and systemic lupus erythematosus. Clinical pres-
Wandinger, K.P., Stangel, M., Witte, T., Venables, P., Charles, P., Jarius, S., entation, histology, survival, and outcome. Medicine (Baltimore), 76,
Wildemann, B., Probst, C., Iking-Konert, C., and Schneider, M. (2010). 192–202.
Autoantibodies against aquaporin-4 in patients with neuropsychiatric Zizic, T.M., Classen, J.N., and Stevens, M.B. (1982). Acute abdominal com-
systemic lupus erythematosus and primary Sjogren’s syndrome. Arthritis plications of systemic lupus erythematosus and polyarteritis nodosa.
and Rheumatism, 62, 1198–1200. American Journal of Medicine, 73, 525–31.
CHAPTER 34

Behçet’s syndrome:
pathogenesis, clinical
manifestations,
and treatment
Emire Seyahi, Gulen Hatemi, Izzet Fresko,
Melike Melikoglu, and Hasan Yazici

Introduction Epidemiology
There is little doubt that elements of Behçet’s syndrome (BS) were BS has a distinct geographical distribution (see Chapter  2). As
discussed in the medical literature before Behçet first proposed it as shown in Figure 34.1, most of the reported cases are from the coun-
a distinct entity (Behçet 1937). Hippocrates described something tries around the Mediterranean basin and Japan. There is a paucity
akin to BS but the entity had not been mentioned in the medical of patients, for unknown reasons, in Northern Europe, Northern
literature until the early part of the last century. Asia, most of continental Africa, Australia, and the Americas.
In his original paper Hulusi Behçet, a professor of dermatove- Travel along the Silk Route has been repeatedly implicated as the
nereology at the University of Istanbul, described three patients, means through which a putative aetiological agent was spread.
two men and a woman, with aphthous and genital ulceration along Perhaps the reason Pakistan and India are mostly spared from BS is
with hypopyon uveitis (Saylan 1997). It is interesting to note that that the Silk Route ran north of these regions.
these three patients were studied over a course of 17 years. Starting The prevalence of Behçet’s syndrome in Turkey is between 20 and
with skin disease, it soon became apparent that many other organ 420 per 100 000 adult population—the highest prevalence around
systems were involved in addition to the initial ‘trisymptom disease’ the globe—based on five field surveys (Demirhindi et  al. 1981;
as originally proposed. Yurdakul et al. 1988; Azizlerli et al. 2003; Idil et al. 2002; Cakir et al.
Recent years have seen a global interest in BS reflected in a large 2004). The prevalence rate is higher in the Asian part of Turkey
number of scientific publications each year. In 2012, the 15th (370 and 421 per 100 000) compared with the European region (20
International Conference on Behçet’s Disease was held in Japan and 80 per 100 000).
with world-wide participants from many disciplines. BS is the lead- The reported prevalences of BS in various geographic regions
ing cause of acquired blindness in Japan and the Japanese govern- are summarized in Table 34.1 (Al Dalaan et al. 1996; Al-Rawi et al.
ment has been supporting a National Behçet’s Disease Research 2003; Davatchi et al. 2008; Gonzalez-Gay et al. 2000; Kaklamani
Committee since 1972. Also, there are dedicated, university-based et al. 1998; Kurosawa et al. 2004; Mahr et al. 2008; O’Duffy 1994;
units in Turkey, Iran, and recently the USA. In 2010, a compre- Salvarani et al. 2007; Shimizu et al. 1979; Zouboulis et al. 1996).
hensive book, describing basic and clinical aspects of Behçet’s syn- It has to be also noted that the frequency of Behçet’s syndrome
drome, written by multinational experts, was published (Yazici and (7: 100 000) (Mahr et al. 2008) was as great as that of all other vas-
Yazici 2010). The wide interest in BS is not simply due to the obvious culitides (9 : 100 000), including polyarteritis nodosa, microscopic
concerns of clinical medicine and health services. There are many polyangiitis, Wegener’s granulomatosis, and Churg–Strauss syn-
physicians and scientists interested in BS from the standpoints of drome, as found in a French survey (Mahr et al. 2004).
endothelial injury, genetics, and relations between inflammatory An ethnic predilection to BS has been shown in a number of
and immunological insult and tissue localization and inflamma- surveys. A higher prevalence of Behçet’s syndrome among immi-
tion, to name but a few. grants of North African and Asian ancestry compared to the
There is debate whether the entity described by Behçet is better European population in Paris (Mahr et  al. 2008)  and a lower
designated as a disease or a syndrome. We prefer the latter as we do frequency in the ethnic Armenian population compared to that
not know the pathogenesis of the illness, nor is there a definitive found in the Turkish population (421 :  100  000) are examples
diagnostic test. (Seyahi et al. 2010).
468 SECTION 5 vasculitic diseases and syndromes and related disorders

Fig. 34.1 Geographical distribution of Behçet’s syndrome. From: Yazici, H. (1994). Behçet’s syndrome. In Rheumatology (eds J. H. Klippel and P. A. Dieppe). Mosby, pp. 20,
1–6 and H. Yazici, I. Fresko, R. Tunç, and M. Melikoglu (2002). Behçet’s syndrome: pathogenesis, clinical manifestations and treatment. In: Vasculitis, 1st edn. G. V. Ball and
S. L. Bridges, Jr, eds. Oxford University Press.

BS has been reported in patients from every race; however, there BS show regional differences as well. Gastrointestinal (GI) manifes-
is a definite under-representation of black patients from Africa and tations are common among the Far Eastern patients (Shimizu et al.
elsewhere (Poon et al. 2003). Even though it can affect every age 1979; Chang and Kim 2002); however, these are rather infrequent
group, onset before puberty or after the sixth decade is relatively in Turkey (Yurdakul et al. 1996) and the Middle East. The same is
rare. There has been a recent awareness of paediatric BS (Kone-Paut true for the prevalence of pathergy, the non-specific hypersensitiv-
et al. 1998; Uziel et al. 1998; Seyahi and Ozdogan et al. 2010); how- ity of the skin to a needle prick, which is less common in North
ever, a survey of 47 000 children in Turkey (Ozen et al. 1998) did European and United States patients (Yazici et al. 1984b). Finally,
not identify a single patient with BS. the HLA B51 association is most pronounced among patients from
The usual onset is in the third decade. No socioeconomic class the Middle and the Far East.
seems to be spared and no environmental agent has been identified
as causative. Clinical findings
Apart from the unexplained differences in disease prevalence
BS is a systemic vasculitis that affects many organs. The clini-
between different geographical areas, some of the manifestations of
cal manifestations include: a variety of skin and mucosal lesions;
uveitis that may result in blindness; a wide range of central nerv-
Table 34.1 Prevalence of BS in various countries ous system (CNS) abnormalities; major-vessel disease; and mus-
culoskeletal and GI problems. The diagnosis is based on clinical
Region Prevalence per 100 000 findings. Initial manifestations typically include one or more of the
following:  oral and genital ulcers; ocular lesions; papulopustular
Turkey 20–420 (aged 10 or over)
skin lesions; erythema nodosum; and arthritis. Table 34.2 shows
Japan 12–100 the frequency of clinical manifestations of BS. The course is char-
Iran 16–100 acterized by remissions and exacerbations that generally abate in
Iraq 17 intensity with the passage of time. A set of diagnostic (classifica-
tion) criteria was published in 1990 by International Study Group
Israel 26–120
(International Study Group for Behçet’s Disease 1990) (Table 34.3).
Saudi Arabia 20 These criteria define oral ulceration as the sine qua non and addi-
France 7 tionally require involvement of two other organ for the diagnosis.
Italy 4 Mucocutaneous findings
Germany (West Berlin) 1.6 Oral ulcers
Sweden 1.5 Oral ulcers occur in virtually all patients and are frequently the first
Scotland, UK 0.3–0.6 disease manifestation (Alpsoy et al. 2007) (Figure 34.2); however,
1–3% of patients can have several of the other features of the disease
USA (Olmstead County, Minn.) 5–30
without ever having oral ulcers. They may precede the diagnosis
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 469

Table 34.2 Frequency of clinical manifestations of BS

Manifestation Frequency
Oral ulcers 97–99%
Genital ulcers ~85%
Scar ~50% (probably more prevalent in males)
Skin lesions
Papulopustular lesions ~85%
Erythema nodosum ~50%
Pathergy reaction ~60% (Mediterranean countries and Japan)
Uveitis ~50%
Arthritis 30–50%
Subcutaneous thrombophlebitis 25%
Deep vein thrombosis ~5%
Fig. 34.2 Oral ulcers. From: Yazici, H. (1994). Behçet’s syndrome. In Rheumatology (eds
Arterial occlusion/ aneurysm ~4%
J.H. Klippel and P.A. Dieppe). Mosby, pp. 20, 1–6 and H. Yazici, I. Fresko, R. Tunç, and
CNS involvement ~5% M. Melikoglu (2002). Behçet’s Syndrome: Pathogenesis, Clinical Manifestations and
Treatment. In: Vasculitis. 1st edn. G.V. Ball and S.L. Bridges, Jr, eds. Oxford University Press.
Epididymitis ~5%
Gastrointestinal lesions 1–30% (more prevalent in Japan) and pharynx are less commonly involved; in contrast to herpetic
lesions, the skin-covered portions of the lips are spared. The ulcers
of BS usually have regular borders, unlike the lesions of Stevens–
of BS by a mean of 9 years (Ideguchi et al. 2011b). It is sometimes Johnson’s or reactive arthritis. Minor ulcers constitute around 90%
difficult or impossible to tell when the disease actually started. The of all oral ulcers in BS and usually heal in about 15 days without
majority of the oral ulcers are indistinguishable from ordinary can- scarring. Like ordinary canker sores, they cause varying amounts
ker sores; however, they are more frequent and multiple. They fre- of discomfort and occasionally make eating difficult.
quently appear as erythematous, circular, and slightly raised areas Major aphthous ulcers are seen less often. These lesions range
evolving into oval or round ulcers within 48 hours (Bang et al. 1995). from 1–3 cm in diameter, have a grey-white ulcer base and may
The oral ulcerations in BS are classified as minor, major, or involve any region of the oral mucosa. They are usually more pain-
herpetiform (Grattan and Scully 1986). Minor aphthous ulcers ful than minor ulcers and have a tendency to heal with a scar. They
are the most common clinical form and are round or oval, shal- may cause dysphagia and lead to pharyngeal stenosis, resulting in
low ulcers, usually multiple, less than 10 mm in diameter, with a malnutrition or even death (Kobayashi et al. 1982). Herpetiform
grey-white pseudomembrane surrounded by a thin erythematous aphthous ulcers are rather uncommon. They consist of crops of
inflammatory halo. The involved sites are usually the mucous mem- ulcers, 2–3 mm in diameter, distributed throughout the oral cavity.
branes of the lips, gingiva, cheeks, and tongue. The palate, tonsils, They heal in a short time without scarring.
The frequency and type of oral ulcers in BS patients were evalu-
ated at a single time point in a hospital-based population in Istanbul.
Table 34.3 International Study Group Criteria for the Diagnosis of Minor aphthous ulcers were the most common type of aphthae in
Behçet’s Syndrome BS (85%); the frequency of major oral ulcerations, more common
in females, was 14% (Coskun et al. 2004). Furthermore, it has been
Criterion Definition reported that frequent occurrence of oral ulcerations during the
Recurrent oral Aphthous or herpetiform lesions; observed by the initial years of the disease may predict the development of major
ulceration physician or patient; recurring at least 3 times a year. organ involvement in men with BS (Hamuryudan et al. 2012).
In the general population, the prevalence of recurrent aphthous
Recurrent genital Aphthous ulceration or scarring observed by the
stomatitis (RAS) is roughly 10 to 20% (Crivelli et al. 1988; Yurdakul
ulceration physician or reliably described by the patient
et al. 1988; Azizlerli et al. 2003). The prevalence of BS, on the other
Eye lesions Anterior or posterior uveitis or cells in the vitreus body hand was 2.8% among patients with RAS, suggesting that BS and
on slit –lamp examination or retinal vasculitis detected RAS represent two ends of a clinical spectrum (Lehner and Batchelor
by an ophthalmologist
1979). This view has been challenged from the clinical standpoint,
Skin lesions Erythema nodosum, pseudofolliculitis, papulopustular in that few patients present with aphthous ulcers plus one other
lesions or acneiform nodules, not related to symptom, and, from the genetic point of view, in that there is not
glucocorticoids treatment or adolescence an increased frequency of the HLA B51 gene among patients with
Positive Test interpreted as positive by the physician at 24–48 h RAS (Ozbakir et al. 1987). A study from Israel, however, did report
pathergy test an association of HLA B51 with RAS (Shohat-Zabarski et al. 1992).
There is preliminary evidence for a negative correlation between
A set of diagnostic (classification) criteria was published in 1990 by International Study
Group for Behçet’s Disease (1990). These criteria define oral ulceration as the sine qua non oral ulcers and tobacco use; cessation of smoking exacerbated oral
and additionally require two other organ involvements for the diagnosis. ulcers (Soy et al. 2000; Kaklamani et al. 2003).
470 SECTION 5 vasculitic diseases and syndromes and related disorders

Histopathological examination of oral ulcers in BS shows lym-


phocytic and monocytic inflammatory infiltrates in the basal layer
and dermis with an eroded epidermis. In the early stages, plasma
cells are usually absent; as the ulcer ages, neutrophils become the
dominant cell (Muller and Lehner 1982) and there is vascular pro-
liferation along with endothelial swelling. Leukocytoclastic vascu-
litis is also occasionally observed (Chun et al. 1990). There is no
difference between the findings on macroscopic examination of the
aphthae in patients with BS and those with RAS.
Genital ulcers
Genital ulcers of BS usually begin as papules or pustules that ulcer-
ate in a short time period. They are usually round or oval with a
punched-out appearance (Figure  34.3). The base is covered with
grey-white fibrin, and at times with surrounding oedema with
induration. They tend to be more readily infected than oral ulcers.
In males they are usually localized on the scrotum and commonly
leave scars. They are less frequent on the shaft and the glans penis, Fig. 34.4 Erythema nodosum like lesion.
inguinal area, pubis, and perineum. Compared with oral ulcers,
perianal ulcers are larger, deeper, less recurrent, and more resistant disease-associated erythema nodosum (Figure 34.4), but they tend
to healing. to be deeper and less well demarcated. Erythema nodosum lesions
In females, ulcers are commonly found on both major and minor related to BS have more histopathological features of vasculitis than
labiae; vaginal and cervical lesions are less frequent and may go erythema nodosum due to other causes (Demirkesen et al. 2001).
unnoticed unless complicated by infection and associated dis- Superficial thrombophlebitis lesions morphologically look very
charge. Therefore, gynaecological examination should be carried much like erythema nodosum, which might explain the negative
out in all female patients with suspected BS. Genital scarring is association described between the two (Krause et  al. 1999). On
usually good evidence of BS in a patient suspected of having this biopsy, superficial thrombophlebitis lesions have a central throm-
syndrome. bosed vein. Ultrasonography can usually differentiate between ery-
The genital ulcers of BS usually heal in 10–30 days if they are not thema nodosum-like lesions and superficial thrombophlebitis.
secondarily infected. Not surprisingly, large ulcers have a higher The acne-like lesions of BS (papulopustular lesions) were once
rate of scarring compared to small ulcers both in males and females considered to represent dermal vasculitis and thus to be different
(Mat et al. 2006). The histopathological features of genital ulcers are from ordinary acne; however, the two can hardly be differentiated.
similar to that of the oral ulcers. Acne-like lesions of BS are frequently seen at the usual sites of acne
Other skin lesions vulgaris, such as the face, upper chest, and upper back, and it is not
There are three broad categories of other skin lesions in BS: (1) ery- uncommon for them to appear on the legs and arms (Figure 34.5).
thema nodosum-like lesions and superficial thrombophlebi- Histologically, the acne-like lesions of BS are indistinguishable
tis; (2)  papulopustular lesions; and (3)  other lesions, including from those of acne vulgaris (Ergun et al. 1998). Sebum production
ulcers, Sweet’s syndrome, and pyoderma gangrenosum. Erythema is increased in BS, as it is in acne vulgaris (Yazici et al. 1987). Acne
nodosum-like lesions are observed in 50% of patients. They lesions of BS are mainly of cosmetic concern.
may be indistinguishable clinically from idiopathic or other

Fig. 34.3 Genital ulcers. Fig. 34.5 Osteofollicular (acne) like lesions.


CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 471

It is curious why BS patients, usually in their late 20s or mid-30s, have not been consistent. It is worth noting that wound healing
suddenly begin having acne lesions, which are more characteristic appears to be normal in BS patients (Mat et al. 1998).
of adolescence. It is well known that acne is an androgen-dependent It is thought by some groups that pathergy occurs during the
lesion and in view of the more severe disease in BS among males, active phases of the disease and disappears when the disease remits,
one could speculate that acne lesions represent androgen hyper- but others find no such correlation (Krause et  al. 2000). Male
sensitivity but data do not consistently support this hypothesis. patients have stronger pathergy reactions (Yazici et al. 1985).
Androgen levels are normal in BS (Yazici et  al. 1986)  with no A positive pathergy test is an important parameter in the diag-
evidence for increased androgen receptors on the skin (Mat et al. nosis of BS in Middle Eastern countries. The specificity of the
1998); however, increased androgen receptor activity has been pathergy test is high (Tuzun et al. 1980) but sensitivity varies in
demonstrated in specimens of skin pathergy reactions from men different studies. The frequency of a positive reaction decreases
(Alpsoy et al. 2005). Although papulopustular lesions in BS were sharply in countries outside the Middle East, Far East, and the
thought to be sterile, it has recently been shown that these lesions Mediterranean regions (Yazici et al. 1984b). Furthermore, analy-
are colonized with bacteria, most commonly Staphylococcus aureus sis of case series suggests that the rate of pathergy has decreased
and less frequently by Prevotella species (Hatemi et al. 2004). One steadily since 1968 (Saylan et al. 1999). It has been hypothesized
interesting observation is the association of acne lesions with the that the use of less traumatic disposable needles may account for
presence of arthritis in BS (Diri et al. 2001; Tunc et al. 2002). this decrease (Ozarmagan et al. 1991). The pathergy test has lim-
Skin ulcers may also be observed rarely in the axillary and inter- ited reproducibility due to intra- and interobserver variation in
digital areas (Azizlerli et al. 1992) and an occasional patient might its assessment, and the reaction may vary in intensity in the same
have dermal punched-out lesions similar to those of polyarteritis patient over time (Altaç et al. 1982).
nodosa (PAN). The association of BS with neutrophilic dermatitis
(Sweet’s syndrome) can be confusing. Typical lesions of Sweet’s syn- Ocular involvement
drome, slightly elevated and somewhat shiny, can be seen in 2.4 to Eye involvement is the most serious manifestation of BS when one
4% of patients with BS (Shi and Huang 1993). On the other hand, takes into consideration both its frequency and morbidity. It is a
some clinical features of BS, such as oral ulceration and arthritis, common cause of blindness in the Mediterranean basin, Middle
can be seen during the course of primary Sweet’s syndrome (Oguz East, and Far East. In a multicentre survey, BS was the most fre-
et al. 1992). Thus the differential diagnosis between the two may quent diagnosis (32%) among uveitis patients who presented to
occasionally be a semantic exercise. ophthalmology clinics in different parts of Turkey (Kazokoglu et al.
2008). Similar surveys report that this frequency was 6.2% in Japan
Pathergy (Goto et al. 2007) and 2.5% in the USA (Rodriguez et al. 1996). Eye
The pathergy phenomenon is a non-specific hyper-reactivity in disease in BS is a non-granulomatous panuveitis and retinal vascu-
response to minor trauma. It is rarely observed in normal individu- litis with exacerbations and remissions of the inflammatory process
als or those with other diseases except for pyoderma gangreno- (Pazarli et al. 1986; Shimizu et al. 1979) (see Chapter 12). While the
sum and chronic myeloid leukaemia treated with interferon-alpha overall prevalence is about 50%, it occurs in close to 70% of males
(IFN-α) (Tuzun et al. 1980; Budak Alpdogan et al. 1998). It is one aged less than 25 years (Yazici et al. 1984a). Eye disease is frequently
of the characteristic manifestations of BS and is usually elicited for present at the onset of the disease or within the first 2 years and it
diagnostic purposes by inserting a 20 gauge needle into the dermis is quite rare for this to develop after the initial 5 years of disease
of the forearm. A positive response is defined by a papule or pustule (Kural-Seyahi et al. 2003).
that forms 48 hours later (Figure 34.6). It has been claimed that sur- Eye involvement is bilateral in 70–80% of patients (Tugal-Tutkun
gical procedures might also lead to pathergic-type reactions, which et al. 2004; Kural-Seyahi et al. 2003). It affects the posterior uvea
may influence the outcome of the procedures, but such findings more often and more severely than the anterior uvea. Patients usu-
ally complain of ocular discomfort and visual blurring. Anterior
uveitis appears as a solitary finding in 10% of those with ocular
involvement. It is not associated with a poor visual outcome.
Hypopyon formation, which represents a profuse accumulation
of inflammatory cells forming a visible layering of pus anterior
to the iris (Figure  34.7), occurs in about 20% of patients with
eye involvement. As a rule, patients with hypopyon suffer from
associated severe retinal vasculitis and thereby have a grave visual
prognosis. It has been shown that optic atrophy ensues within a
few months in about half of those with hypopyon uveitis (Pazarli
et al. 1986).
Gross pathological changes include vitreous deposits, choroidi-
tis, and retinitis. Retinitis involves capillaries at the disc, macula,
and the periphery. Early retinal vasculitis can be seen as patches of
fluorescein leaks along vessels in otherwise asymptomatic patients
with a normal ophthalmoscopic examination. In well-established
disease, fundoscopic examination reveals: vitreal deposits; choroi-
dal or retinal exudates with sheathing of the blood vessels; haem-
Fig. 34.6 Positive pathergy reaction. orrhages; cytoid bodies or white patches; venous thrombosis;
472 SECTION 5 vasculitic diseases and syndromes and related disorders

Fig. 34.7 Hypopyon uveitis.


Fig. 34.8 Radiographic appearance of pulmonary arterial aneurysms.
papilloedema; and macular disease. It is sometimes difficult to dif-
ferentiate papilloedema due to primary eye disease from that due to
increased intracranial pressure. The majority of the vascular events occur within 5 years of dis-
Recurrent inflammatory activity in the anterior chamber may ease onset (Kural-Seyahi et al. 2003; Hamuryudan and Melikoglu
lead to anterior and posterior synechiae, secondary glaucoma, 2010). However, in up to 30% of the patients vascular disease
optic atrophy, and macular degeneration. Cataracts may form with may occur as an initial feature even before the full disease onset
or without the additional insult of glucocorticoid therapy. Ocular (Hamuryudan and Melikoglu 2010). Peripheral venous involve-
inflammation can persist among patients with complete loss of ment is earlier in the course (median 2 years), while pulmonary
vision and total global atrophy. Enucleation might be the only (median 5  years) and non-pulmonary arterial (median 7  years),
way to control the pain of ongoing inflammation in these patients. and central venous involvement (median 5  years) appear later
Histological examination of the enucleated blind eye reveals atro- (Melikoglu et al. 2008). Dural sinus thrombosis also seems to occur
phy and marked lymphocyte infiltration (predominantly T cells) early as it is the most frequent manifestation among juvenile BS
of all intraocular tissues (Shimizu et al. 1979; George et al. 1997). patients (Uluduz et al. 2011).
Episcleritis, conjunctivitis, corneal ulcerations, and lid lesions Recurrences are common; in a retrospective study by our group,
are not characteristic features of eye disease in BS, but are noted the recurrence rate was reported to be 23% and 38% over 2 and
occasionally. 5  years, respectively (Melikoglu et  al. 2008). The most common
More than two decades ago, BenEzra and Cohen (1986) noted the (65%) recurrent vascular event was lower extremity deep vein
eventual loss of vision in three-quarters of their patients. We are cer- thrombosis. The onset is unilateral in most of the patients; however,
tainly doing better today—loss of useful vision ensues in one-fifth progression to bilateral involvement is common.
of our BS patients. Severely impaired vision does not always mean Vascular disease may be considered as a distinct cluster among
an eventual loss of useful vision, and those patients with late onset the many phenotypes of BS (Hamuryudan and Melikoglu 2010).
of eye disease have a better visual prognosis (Kural-Seyahi et al. Chronic relapsing deep vein thromboses in the legs tend to precede
2003; Tugal-Tutkun et al. 2004). other sites of major vessel involvement. Furthermore, superficial
thrombophlebitis tends to be associated with lower extremity deep
Major vessel involvement vein thrombosis and inferior vena cava. Dural sinus thrombosis is
The spectrum of vascular disease is broad and unique in BS. It is also known to be associated with major-vessel disease (Tunc et al.
one of the few vasculitides that can involve large vessels, both arte- 2004). We recently became aware of the association of pulmonary
rial and venous. It is virtually the only disorder that leads to the artery aneurysms (PAA) with vena cava and dural sinus thrombosis
formation of pulmonary artery aneurysms (Figure  34.8). Close (Melikoglu et al. 2008). On the other hand, non-pulmonary arterial
scrutiny suggests that the Hughes–Stovin syndrome (pulmonary disease seems to represent rather a different phenotype of vascular
arterial aneurysms with systemic thrombosis) is indeed BS (Erkan involvement, based on older age at manifestation and lesser asso-
et al. 2004). Vascular disease develops in up to 40% of the patients ciation with venous thrombosis (Melikoglu et al. 2008).
in Turkey and has a definite male preponderance (Kural-Seyahi The pattern of vascular inflammation in BS is diffuse rather than
et al. 2003). However, its prevalence may be as low as 6% in Japan patchy, and large segments of the vessel wall are involved, with
(Ideguchi et al. 2011a). superimposed inflammatory thrombi. Sometimes the entire length
Deep veins of the lower extremity are the most common sites of the inferior vena cava is affected. Clinical signs of venous lesions
of venous thrombosis, which constitute 60–80% of the vascular vary according to their site. Chronic recurrent thrombosis of the
lesions. In the remaining, the veins affected in descending order of lower extremities leads to erythema, dermatitis, hyperpigmenta-
frequency are: inferior and superior vena cavae, dural sinuses; axil- tion, and subsequently to crural ulcers (Figure 34.9). Chronic oblit-
lary; brachial; hepatic; and portal veins (Seyahi and Yurdakul 2011). eration of the caval systems leads to the appearance of prominent
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 473

remission can be seen in about 60% of the cases. About 40% of the
patients may still complain of dyspnoea or small bouts of haemop-
tysis, although PAA or PAT disappeared completely, after a mean
follow-up of 7 years, Arterial lesions may leave occlusion as a seque-
lae. Additionally, PAI patients, especially those with poor progno-
sis, may have increased systolic pulmonary artery pressure (Seyahi
et al. 2011). Early detection and use of immunosuppressive drugs
have decreased the mortality to as low as 26% (Seyahi et al. 2012),
which was 50% about a decade ago (Hamuryudan et  al. 1994).
Histopathologically, the inflammatory process is located primarily
in the vasa vasorum. Necrosis of the vessel wall occurs with the
formation of true or false aneurysms (Hamuryudan et al. 2004b).
They in turn erode into the bronchial tree and cause haemopty-
sis, the most frequent symptom. No association between pulmo-
nary embolism and deep vein thrombosis is found in BS, perhaps
because thrombi are tightly adherent to inflamed veins. It follows
that anticoagulant therapy is not only unhelpful but is dangerous in
Fig. 34.9 Chronic thrombosis of the leg veins with hyperpigmentation and a
skin ulcer. this situation (Efthimiou et al. 1986).
Aortic and peripheral arterial aneurysms are also major causes
of death because of the risk of rupture. Arterial occlusions are rare.
venous collaterals on the thoracic and abdominal walls, forming Non-pulmonary arterial disease is mostly unilateral and in a single
caput medusae. Obstruction of superior vena cava may also cause vessel, but multiple, mainly false, aneurysms are possible. Mostly
oesophageal varices, chylothorax and chylous pericardial effusions, abdominal aorta, femoral, iliac, popliteal, and carotid arteries were
but pericardial involvement is rare. involved. The commonest localization of the aneurysms was the aor-
Budd–Chiari syndrome is caused by hepatic vein occlusions tic bifurcation (Tuzun et al. 1997; Tuzun et al. 2012; Saadoun et al.
and usually presents with ascites or eventually liver failure. In 2012). Abdominal aortic aneurysms sometimes produce abdominal
series from Turkey, BS is reported as the most common cause of pain. Bruits can be heard over the affected large arteries and a pul-
Budd–Chiari syndrome (Saatci et al. 1993; Bayraktar et al. 1997). sating mass may be found. Most patients with arterial lesions have
It has a mortality rate of 60% (Kural-Seyahi et al. 2003; Bayraktar constitutional signs and symptoms, such as fever and weight loss.
et al. 1997). Peripheral arterial involvement causes reduced or absent pulses with
Arterial involvement is less common then venous disease with a intermittent claudication, cold extremities, or even gangrene.
prevalence of 1.5–7.5%. It is one of the most serious complications A histopathological study of six resected peripheral arterial
of BS with high morbidity and mortality (Kural-Seyahi et al. 2003). aneurysms reported a predominantly neutrophilic vascular injury
There is a marked male predominance. Arterial lesions can cause of the vasa vasorum, with a significantly increased number of vasa
aneurysms, which can be complicated by in situ thrombosis, lead- vasorum. The internal elastic membrane was intact, and there was
ing to variable degrees of occlusion. Any part of the arterial tree can increased expression of HLA-DR-positive cells in the endothelium
be diseased, but the pulmonary arteries are the most frequent sites (Kobayashi et al. 2000). Chronic aneurysmal lesions are character-
followed by abdominal aorta, femoral, popliteal, iliac and carotid ized by periadventitial fibrosis with the entire vessel being encased
arteries. Renal, cerebral, and coronary arteritis have been reported in fibrous tissue. Large segments of thrombi are observed, and as a
rarely. Abdominal visceral ischaemia is uncommon. rule there are no free-floating thrombus tails. Reactive lymph nodes
Pulmonary arterial involvement (PAI) mainly manifests as PAA have also been reported in the immediate vicinity of peripheral
or less often solely ‘in situ’ pulmonary artery thrombosis (PAT). arterial aneurysms (Tuzun et al. 1997).
Thrombosis develops usually as a complication to underlying
extensive vasculitis (Hamuryudan et al. 1994; Hamuryudan et al. Cardiac disease
2004a; Seyahi et al. 2012). Clinical features and outcome of isolated Cardiac involvement due to BS is rare (Mogulkoc et  al. 2000;
PAT and PAA is similar except that haemoptysis is less frequent Saadoun et al. 2012) and has mainly been mentioned in sporadic
and less abundant in patients with isolated PAT. Thorax CT scans case reports. It appears in the form of valvular heart disease, intra-
are more helpful than chest radiographs in the diagnosis of PAI. cardiac thrombosis, aortitis, coronary artery vasculitis, ventricular
There is a risk of pseudoaneurysm formation while doing pulmo- aneurysms, and pericarditis. Valvular heart disease mostly involves
nary angiography (Kingston et al. 1979). Aneurysms are partially the aortic valve, with or without aortitis, and is relatively frequent
or totally thrombosed in about one-third of the cases. The involve- in the Far East. Intracardiac thrombosis is associated with vascu-
ment is usually bilateral and affects mostly inferior lobes. Thorax lar involvement elsewhere in the body, mostly with PAI (Mogulkoc
CT scans may show parenchymal lesions such as nodules, consoli- et al. 2000). It is found more frequently among young males and
dations, cavities, and ground-glass lesions in addition to the main located usually in the right side of the heart, right ventricle being
arterial lesions. Mediastinal lymphadenopathy, mild pleural and the most common place. The thrombosis was reported to be tightly
pericardial effusions, and intracardiac filling defects may be also adhered to underlying endocardium or myocardium. Coronary
present. Ventilation–perfusion lung scans may show persistent artery vasculitis in the form of aneurysms or occlusion may lead
perfusion defects, a finding that is not expected in true pulmo- to myocardial infarction or rupture of the aneurysm. In a recent
nary emboli. Relapses may occur in 20% of the patients; however, French series, pericarditis was listed as the most common cardiac
474 SECTION 5 vasculitic diseases and syndromes and related disorders

lesion (Geri et  al. 2012). Histopathological examinations of val- positivity and sacroiliitis was not frequent among these patients
vular, aortic, and endomyocardial lesions show mixed acute and (Hatemi et al. 2008b; Hatemi et al. 2010). We have also reported that
chronic inflammation at various stages and fibrosis. Atherosclerotic this papulopustular lesion–arthritis association clustered among
changes are absent (Mogulkoc et al. 2000). Occasionally, endomyo- the families (Karaca et al. 2012), suggesting a separate genetic pre-
cardial fibrosis may be seen (Houng et al. 1997). disposition as compared to other clinical manifestations.

Atherosclerosis Central nervous system disease


There is no evidence that clinical cardiovascular disease or its asso- Neurological disease, of acute and chronic onset, is one of the most
ciated mortality is increased in BS. An increase in the mortality serious features of BS. Although the prevalence of neurological
rate late in the course, indicating mainly atherosclerotic cardio- involvement has been reported to be between 5% and 59% (Farah
vascular disease mortality, as reported for SLE, was not observed et al. 1998), a prevalence of 5% was found in a prospective survey
(Kural-Seyahi et  al. 2003; Saadoun et  al. 2010a). The prevalence of Turkish patients (Serdaroglu et al. 1989). Japanese autopsy reg-
of angina and history of ischaemic heart disease was found to be istry data (Lakhanpal et al. 1985) suggest that cerebrospinal tract
similar between patients and controls even after adjustment for demyelination is the most common pathological finding, followed
risk factors (Ugurlu et al. 2008). Evidence for increased frequency by encephalomalacia, perivascular cellular infiltration, and cerebral
of carotid artery plaques is weak; however, there are studies in atrophy. The role, if any, of vasculitis per se in the pathogenesis of
which carotid artery intima media thickness is increased among CNS involvement is not certain. Histopathological studies show
BS patients compared to age and gender matched healthy controls that active neurological involvement was associated with perivas-
(Seyahi and Yazici 2007). cular infiltration of T lymphocytes, and monocytes with few B lym-
phocytes, and most neurons undergoing apoptosis (Arai et al. 2006;
Musculoskeletal manifestations Hirohata 2008). Absence of fibronoid necrosis is noted. Clinically
Arthritis or arthralgias have been noted at some point in the disease inactive lesions were characterized by atrophy and isomorphic glio-
in as many as 50% of patients (Yurdakul et al. 1983). It is usually sis with formation of cystic or moth-eaten lesions (Hirohata et al.
monoarticular or oligoarticular and may occasionally precede the 2003; Hirohata 2008).
diagnosis of BS by months or years. Arthritis most often involves There are mainly two types of CNS involvement in BS
large joints: knees, ankles, elbows, and wrists, in more or less that (Akman-Demir et  al. 1999; Siva and Saip 2009). About 75% of
order. The arthritis is usually non-destructive and typically resolves patients with CNS disease have parenchymal lesions (Figure 34.10)
in several days or weeks, but synovitis, lasting months to years with and the remainder have dural sinus thrombi (Figure  34.11).
deformity and erosions, has been encountered. It is accompanied Dural sinus thrombosis in the CNS typically causes headaches
by a moderately elevated erythrocyte sedimentation rate. The issue with or without bilateral papilloedema and increased cerebrospi-
of sacroiliitis as a component of BS and the inclusion of BS among nal fluid pressure (Aguiar de Souza et al. 2011). Venous infarcts
the seronegative spondyloarthritides is controversial, but the con- may be observed (Yesilot et  al. 2009). A  recent study showed
sensus is that BS should not be included in this group (Chamberlain that dural sinus thrombosis is the dominant type of neurological
and Robertson 1993; Olivieri et al. 1997; Yazici et al. 1981a).
Synovial fluid in BS-associated arthritis is inflammatory, with a
predominance of polymorphonuclear leukocytes. The histological
changes of synovium in BS are not disease specific. Ulceration and
replacement of the superficial layer by heavily inflamed granulation
tissue, and paucity of plasma cells, are the salient features. Articular
symptoms may also arise from aseptic osteonecrosis, where it is
taken as evidence of vasculitis and not necessarily the result of glu-
cocorticoid use (Sugioka et al. 1966).
Inflammatory myositis is usually local and confined to the leg,
but generalized forms have been described (Arkin et al. 1980; Yazici
et al. 1981b). Serum levels of muscle enzymes are not elevated in
localized forms. The histological picture of these lesions is similar
to that observed in polymyositis. The prevalence of ‘fibromyalgia’ is
around 10%, and is mainly in female patients with mild to moder-
ate disease activity (Yavuz et al. 1998).
Patients with arthritis tend to have a high risk for papulopus-
tular lesions compared to BS patients with no arthritis (Diri et al.
2001). Moreover the association of papulopustular lesions and joint
involvement was confirmed in a factor analysis in BS (Tunc et al.
2002). It was hypothesized that this papulopustular lesion–arthritis
cluster in BS may have a distinct pathogenesis, similar to that of
acne-associated reactive arthritis, involving an infectious aetiology.
Interestingly, it was later shown that enthesopathy is also associated
with this papulopustular lesion–arthritis association (Hatemi et al. Fig. 34.10 CT scan of parenchymal central nervous system involvement. Note
2004). Similar to the other acne associated arthritides, HLA B27 the hyperintense lesion in the brain stem.
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 475

Table 34.4 Features of neurological involvement in BS

Symptoms and signs %


Pyramidal signs 82
Headache 65
Cerebellar signs 24
Sensory symptoms and/or signs 24
Pseudobulbar signs 12
Intracranial hypertension 12
Meningeal irritation signs 12
Hemianopsia 6
Dementia 6
Personality changes 6
Tremor 6
Myoclonic jerks 6

favourable prognosis. Autonomic nervous system involvement was


Fig. 34.11 CT scan of dural sinus thrombosis.
also reported to occur in rare cases (Karatas et al. 2002).
Cerebrospinal fluid analysis usually reveals non-specific find-
ings such as mild pleocytosis and slightly elevated protein con-
centration. Glucose levels are usually normal but occasionally
involvement in juvenile patients with BS (Uluduz et al. 2011). It they can be low. Despite the lack of specificity of these findings,
is rare to have both lesions in the same patient (Siva et al. 2004). patients with abnormal CSF had the worst prognosis during the
Table 34.4 summarizes the clinical symptoms and findings of CNS follow-up (Akman-Demir et al. 1996; Akman-Demir et al. 1999).
involvement in BS. Additionally, increased levels of interleukin (IL)-6 in the cerebro-
The brain stem is the most commonly affected site, followed by spinal fluid can be found and seem to be a marker of disease activity
the spinal cord, cerebrum, and cerebellum (Lakhanpal et al. 1985). and long-term outcome (Akman-Demir et al. 2008b).
Pyramidal motor signs are the most common clinical features, fol- If there is localized neurological disease, CT scans and magnetic
lowed by cerebellar and sensory signs and symptoms (Siva and Saip resonance imaging can correlate anatomical findings with clinical
2009). Isolated cerebellar involvement, however, is most unusual features. Lesions include hypodense areas on CT scans and hyper-
(Serdaroglu 1998). Signs and symptoms of meningeal irritation and dense areas on MRI, similar to those seen in vasculitis. The brain
increased intracranial pressure also occur. stem and basal ganglia are the sites most frequently abnormal on
Headache is the most common neurological symptom seen in MRI of patients with acute BS. Seventy-five per cent of these lesions
patients with BS (Saip et al. 2005; Aykutlu et al. 2006; Kidd 2006). were large and confluent, mainly extending from the brain stem to
Headaches are ascribed to CNS lesions only if other associated neu- the diencephalon and basal ganglia. The brain stem or basal ganglia
rological signs and symptoms are present (Serdaroglu et al. 1989), were involved in one-third of patients with chronic BS. Brain stem
but one should keep in mind that a non-structural migraine-type atrophy was seen in 21% of the patients with a specificity of 100%
headache unrelated to neurological involvement is also common in the absence of cortical atrophy (Coban et al. 1999). MRI findings
among patients with BS (Saip et al. 2005). It can also be seen in in BS indicate the presence of a small-vessel vasculitis with mainly
association with ocular inflammation. Hearing loss and vertigo venular involvement (Kocer et al. 1999). There are reports that sin-
may also occur (Kulahli et al. 2005). Lesions of the nervous system gle photon emission computed tomography (SPECT) may be more
may progress to produce fatal bulbar paralysis. As with almost all sensitive in detecting early CNS lesions than MRI but controlled
other manifestations, neurological involvement runs a more severe studies are lacking (Avci et al. 1998; Nobili et al. 2002).
course among males.
Problems related to higher cortical function, such as character
disorders, impairment of memory and dementia, and other psychi- Gastrointestinal involvement
atric symptoms, are sometimes noted (Akman-Demir et al. 1999; Lesions of BS occur throughout the GI tract (Yurdakul et  al.
Monastero et al. 2004). Some of these disturbances are related to 1996)  (see Chapter  14). Mucosal ulcers dominate and are his-
the situational response to a chronic disease or possibly to gluco- tologically indistinguishable from those found in inflammatory
corticoid use. Peripheral nervous system involvement is distinctly bowel disease. These are multiple, deep, round, and not specific,
unusual in BS. sometimes granulomatous, ulcers associated with chronic inflam-
Silent neurological involvement in the form of abnormali- mation. Normal intervening mucosa is also characteristic (Fukuda
ties in magnetic resonance imaging or neuropsychological test- and Watanabe 1979). Three-quarters of the mucosal ulcers in BS
ing is also reported (Yesilot et  al. 2006). These patients have a are found in the terminal ileum and caecum and less often in the
476 SECTION 5 vasculitic diseases and syndromes and related disorders

stomach and duodenum (Mori et al. 1983). In contrast to ulcerative questionnaire, the prevalence of epididymitis was found to be as
colitis, rectal involvement in BS is rare. Oesophageal lesions include high as 19% (Cetinel et al. 1998). In the same survey, the frequency
oesophagitis and ulcers in various stages of healing and stenosis of lower urinary tract symptoms was also significantly higher in BS
(Griffin et al. 1982). than in controls. Clinicians have noted voiding disturbances due to
GI complaints include dysphagia, vomiting, colicky abdominal neurological involvement, as well as primary bladder dysfunction
pain, and diarrhoea, which can be bloody. They are characterized due to mucosal ulcers or what resembles interstitial cystitis, with-
by exacerbations and remissions. During exacerbations, a mass is out demonstrable neurological defects (Cetinel et al. 1999). Erectile
often palpable in the abdomen. Ileocaecal lesions are quite prone to dysfunction has been reported to have a frequency of 63% among
perforation (Kasahara et al. 1981; Shimizu et al. 1979). GI involve- patients with neurological involvement (Erdogru et al. 1999).
ment in BS is sometimes difficult to distinguish from Crohn’s dis-
ease in which oral ulcers, erythema nodosum, and intermittent
arthritis are also features. Although these features are common
Pathogenesis
to both BS and inflammatory bowel diseases, International Study The pathogenesis of BS remains unclear. The scope and the speci-
Group (ISG) criteria may be helpful in differentiating the two ficity of immunological alterations suggest that immunologically
conditions. When inflammatory bowel disease patients were for- mediated mechanisms are the main trigger of the vasculitis of
mally assessed, it was observed that none of the 93 Crohn’s patients BS. The arguments against a primary autoimmune process are as
and two of the 130 ulcerative colitis patients fulfilled ISG criteria follows:
(Hatemi et al. 2008a). However, it is not uncommon for BS patients 1. Clinical and laboratory findings usually found in autoimmune
to present with gastrointestinal involvement before fulfilling ISG diseases (Raynaud’s phenomenon, serosal involvement, photo-
criteria. Thus, a study attempted to identify gastrointestinal features sensitivity, thrombocytopenia, haemolytic anaemia) are not usu-
to distinguish BS from Crohn’s disease. When specificity and sen- ally seen in BS. Most importantly, there is no association with
sitivity of several clinical and endoscopic features were analysed in Sjögren’s syndrome.
BS and Crohn’s disease patients, it was observed that having a focal
2. There is no increased prevalence of autoimmune diseases among
single ulcer, rectal bleeding, and perforation were the feature that
BS patients or their families.
had the highest specificity, and thus a positive likelihood ratio for
BS (Celik et al. 2010). A study of enteroclysis findings has shown 3. The HLA gene associated with BS, HLA B51, is not found in
that GI involvement in BS is milder than that of Crohn’s disease. autoimmune diseases and the HLA alleles associated with other
Scalloping, ulceronodular patterns, and abscess and fistula forma- autoimmune diseases are not frequent in BS.
tion are not part of BS (Iida et al. 1993; Korman et al. 2003). 4. Although there is evidence of polyclonal B-cell activation, there
As noted in Section Major Vessel Involvement, Budd–Chiari syn- is no specific association with autoantibodies (Gunaydin et al.
drome occurs in BS patients, but primary involvement of the liver 1994; Yazici 1997).
without occlusion of the hepatic vein is uncommon. Splenomegaly
is noted in 20% of patients, even without portal hypertension sec- The absence of the classical features of autoimmunity along with
ondary to venous thrombosis (Soysal et al. 1990). the paucity of specific autoantibodies has led to the suggestion that
BS is an autoinflammatory disorder (Gul 2005). On the other hand,
Amyloidosis and renal involvement there is no association of BS with mutations in genes implicated
As in other chronic inflammatory states, there may be secondary in autoinflammatory disorders such as familial Mediterranean
amyloidosis of the AA type. Widely different frequencies, ranging fever or TNF-receptor-associated periodic syndrome (MEVF and
from 400/100  000 to 2000/100  000, have been reported (Dilsen TNFR2, respectively). Furthermore, BS is more frequent than the
et al. 1988; Yurdakul et al. 1990). In BS, amyloidosis predominantly classical autoinflammatory conditions and tends to abate with time,
occurs in males, and nephrotic syndrome is the typical clinical pres- which also argues against its inclusion as an autoinflammatory dis-
entation. The onset of amyloidosis is about a decade after the dis- ease (Yazici and Fresko 2005).
ease begins; however, it can be seen as early as the third year. There
seems to be a close association with large-vessel disease and arthri- Lymphocytes and cytokines
tis. The nephrotic syndrome due to amyloidosis has a mortality rate Increasing evidence suggests that Th1-type T cells are the main
of over 50% within 3.5 years of its onset (Melikoglu et al. 2001). cells responsible for tissue destruction (Frassanito et  al. 1999).
Glomerulonephritis attributable to BS has been increasingly Additionally, there is growing evidence that Th17 cells, which pro-
reported (Akpolat et al. 2003; Altiparmak et al. 2002), with lesions liferate from naive T cells through IL-6 and TGF-β stimulation and
varying from IgA nephropathy to rapidly progressive glomerulone- are characterized by mainly IL-17 production, also play a role in the
phritis. Immune complexes are not a constant feature. We have had development of BS, similar to other inflammatory and autoimmune
a successful kidney transplant,with 4-year follow-up, in a patient conditions (Direskeneli et al. 2011; Hamzaoui et al. 2011). This Th17
with renal failure (Apaydin et al. 1999). dominance was observed in BS patients with neurological (Geri
et al. 2011) and eye involvement (Kim et al. 2010), but not those
Urological problems with gastrointestinal involvement (Ferrante et al. 2010). The possi-
Urethritis and urogenital infections are rare in BS. An occasional bility of specific or non-specific T-cell activation is still a matter of
patient may present with a meatal ulcer. Apart from the genital debate. The former is supported by: increased susceptibility to infec-
ulcerations there are three primary problems related to the genitou- tious triggers (Kaneko et al. 2003); the up-regulation of heat-shock
rinary system in BS: epididymitis, cystitis, and erectile dysfunction. protein (HSP60) expression in mucosal lesions of BS (Ergun et al.
In a recent prospective survey based on a standard urological history 2001; Lehner et al. 1997); and a strong oligoclonal T-cell response
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 477

to human HSP60 and to other strains of streptoccocci or various decreased prostaglandin I2 (PGI2) synthesis; decreased nitrous
microbial agents (Direskeneli et al. 1999; Esin et al. 1997). In this oxide (NO) production; and enhanced E-selectin expression in
context, one attractive hypothesis is the involvement of heat-shock endothelium (see Chapter 4). Von Willebrand factor and endothe-
proteins in a Th1-type immune response without T cell receptor lin are synthesized and stored in the endothelium and both are
engagement, linking the innate immune system to T-cell immunity elevated in BS (Direskeneli et al. 1995; Ural et al. 1994). Elevated
(Direskeneli and Saruhan-Direskeneli 2003). On the other hand, the levels of these proteins in the peripheral blood might be due to
overproduction of proinflammatory cytokines by innate immune increased synthesis, perhaps secondary to an inflammatory insult.
cells of BS (Mege et al. 1993; Zierhut et al. 2003) may result in a There is also decreased production of PGI2 (Kansu et al. 1986) and
non-specific T-cell response. An interesting, preliminary observa- nitric oxide synthetase (NOS). Both molecules are potent vasodila-
tion is the production of oral tolerization in BS patients with uvei- tors and are strong inhibitors of platelet aggregation and thrombo-
tis, by the uveitogenic BS specific peptide of the human HSP60 sis. There are two lines of evidence for decreased NOS production
(Stanford et al. 2004). A possible role of HSPs was also observed from the endothelium in BS. NOS levels in the peripheral blood
in the local immune response, in peripheral blood and cerebrospi- are decreased (Orem et  al. 1999), and flow-mediated vasodila-
nal fluids of BS patients with neurological involvement (Tasci et al. tion, a well established function of the endothelium, is impaired
1998), in oral ulcer biopsy samples (Deniz 2010), and intestinal (Chambers et al. 2001; Oflaz et al. 2005). The VNTR polymorphism
lesions of BS patients with gastrointestinal involvement (Imamura in the endothelial NOS gene has been associated with BS, suggest-
et al. 2005; Nara et al. 2008). However, whether the HSPs observed ing the role of genetic factors in endothelial dysfunction (Karasneh
in BS lesions have a role in the pathogenesis, or their presence is et al. 2005). The presence of antiendothelial antibodies in BS is con-
caused by the ongoing inflammation, and whether they have a pro- troversial. One study suggested that their quantity is related to dis-
tective role against inflammation needs to be further studied. ease activity (Direskeneli et al. 1995), while another study did not
Another potential connection between the innate and adaptive find increased levels (Dinc et al. 2003).
immune systems involves the interaction of MHC class I molecules
with the killer cell immunoglobulin-like receptors (KIRs), which are Genetics
expressed by natural killer cells and T cells. A particular association Most cases of BS are sporadic. Nevertheless, the presence of famil-
between HLA B51 and KIR3DL1 or KIR3DS1 receptors on inflamma- ial occurrence in about 10% of the patients, the increased prev-
tory cells has been reported in BS (Saruhan-Direskeneli et al. 2004). alence of the syndrome along the so-called Silk Road (30–45°
latitude north; from Japan to the Mediterranean Basin), and the
Neutrophils association with HLA-B51 in many ethnic groups favour the role
For many years it was thought that polymorphonuclear neutro- of genetic factors (Gul and Ohno 2010). Differences in the fre-
phil (PMN) hyper-reactivity in BS was central to its pathogenesis quencies of BS between various ethnic groups and locals living
(Takeuchi et al. 1981), which explained the usefulness of colchi- in the same environment (e.g. France and Turkey) also suggest a
cine. Closer scrutiny indicates that the data are, in fact, somewhat hereditary basis (Mahr et al. 2008; Seyahi et al. 2010). Although
contradictory. In a well-controlled study, there was no evidence multicase family studies have suggested a multifactorial mode
of increased PMN activity in BS (Tuzun et al. 1999). Carletto and of inheritance, an autosomal recessive pattern of inheritance has
colleagues found increased activity of PMNs only during active been implicated in childhood BS (Molinari et al. 2003). Familial
BS (Carletto et al. 1997); under basal conditions, no evidence for cases were also more common in the paediatric group compared
increased superoxide production or adhesion was noted. Another to adults, suggesting a higher genetic load in children (Kone-Paut
study found an increased oxidative burst response in neutrophils et al. 1999). A recent twin study in BS showed that the pairwise
and monocytes in patients with BS although the reaction was not concordance rate for BS was 2/6 for monozygotic twins and 1/8
specific to this condition (Gogus et al. 2005). for dizygotic twins, again suggesting a genetic predisposition;
however, discordance persisted in the remaining monozygotic
Immune complexes and hypercoagulability twins after 8 years, confirming the complex genetic nature of the
It is controversial whether immune complexes or B cells play a role syndrome (Masatlioglu et al. 2010). HLA B51, a class I major his-
in the pathogenesis of BS. Antineutrophil cytoplasmic antibodies tocompatibility complex (MHC) has been the most consistently
(ANCA) (Ben Hmida et al. 1997) and anticardiolipin antibodies reported HLA association in BS. A  meta-analysis demonstrated
(Tokay et  al. 2001)  are absent. Antibodies to Saccharomyces cer- that the pooled odds ratio of HLA-B51/B5 carriers to develop BS
evisiae (Fresko et  al. 2005; Krause et  al. 2002), to α-tropomysin compared to non-carriers was 5.78 (95% CI 5.00–6.67). The risk
(Mahesh et al. 2005), and to α-enolase (Lee et al. 2003) have been was consistent across different populations with varying frequen-
described, but their role in the pathogenesis of BS is not clear. cies of BS (De Menthon et  al. 2009). Potential linkage disequi-
A detailed analysis has revealed that no procoagulant factors are librium in the Class I region has also been studied. Many genes
definitely associated with the tendency to thrombosis observed in have been implicated, including the MICA 009 allele and its trans-
BS (Espinosa et al. 2002; Leiba et al. 2004). It has been suggested, membrane microsatellite polymorphism A6 (Mizuki et al. 2007),
however, that there is a defect in fibrinolysis related to a deficiency and fine mapping of the region has shown that HLA B-51 has the
of tissue plasminogen activator (tPA) production in acute throm- strongest association. The only exception to this is the HLA-A26
bosis of BS (Yurdakul et al. 2005). allele and the HLA-A*26-F*010101-G*010102 haplotype, which
associates with BS independently from HLA-B51 (Meguro et al.
Endothelial cells 2012). The mechanism whereby HLA B51 confers susceptibility to
Several findings suggest endothelial dysfunction in BS: increased BS is not known. The main function of the HLA Class I system is to
levels of von Willebrand factor; increased endothelin I production; present peptides to CD 8+ T cells. The configuration of the peptide
478 SECTION 5 vasculitic diseases and syndromes and related disorders

binding pocket of HLA-B51 has been delineated (Sakaguchi et al. further bring to our understanding of BS. The divergent nature of
1997) but a specific peptide associated with BS has not been deter- the genetic associations we summarize in this section might sup-
mined. Cross-reactivity with another antigen, such as retinal port the contention that BS may indeed be more than one nosologi-
S (Kurhan-Yavuz et  al. 2000), interactions between KIR (killer cal entity (Yazici et al. 2012).
immunoglobulin-like) receptors of natural killer (NK), CD 8+, and
gamma delta T cells (Duymaz-Tozkir et al. 2008) and neutrophil Pathogenesis of pathergy
hyperactivity (Takeno et al. 1995) are other possibilities. HLA B51 The pathergy reaction is related to the increased inflammatory
may account for no more than 20% of the sibling relative risk (Gul state that characterizes BS, but the exact pathogenesis of the phe-
et al. 2000), which supports the view that susceptibility is depend- nomenon is unknown. Pathogenetically, it shows features in com-
ent on other genetic loci in addition to environmental triggers. mon with T-cell-mediated immune response (Gul et  al. 1995).
Case–control studies with a small number of patients and controls One study suggested that injury to the skin of BS patients elicits
have shown a multitude of associations with various candidate a T helper 1 (TH1)-cell response due to IL-12-mediated produc-
genes. However, these studies lack statistical power and the data tion of interferon-gamma (IFN-γ) by CD4+ T cells (Melikoglu
they provide are not robust. Moreover, only a few of them have et al. 2002). The intradermal inoculation of monosodium urate
been replicated in different ethnic groups (e.g. polymorphisms in (MSU) crystals also produces a longer lasting erythema in BS
the TNF, MEVF, ICAM1, and eNOS genes) (Gul and Ohno 2010). patients than in healthy or diseased controls (Cakir et al. 1991).
Genome-wide association studies (GWAS) have provided new An interesting observation is the induction of the pathergy reac-
insights. The first GWAS was conducted in a small number of tion with IFN-α (Budak-Alpdogan et al. 1997). In these patients
Turkish patients (156 with BS and 167 controls). The single nucleo- the symptoms of BS subsided on withdrawal of IFN-α therapy.
tide polymorphisms (SNPs) KIAA1529, CPVL, LOC100129342, The same group (Budak-Alpdogan et al. 1998) reported a positive
UBASH3B, and UBAC2 were associated with BS. The functions pathergy test in 25% of chronic myelogenous leukaemia (CML)
of two of these genes were unknown. UBASH3B and UBAC2 are patients using IFN-α. A  number of patients with BS and coex-
involved in the ubiquitination pathway whereas CPVL encodes istent myelodysplastic syndrome have been reported (Ohno et al.
a carboxypeptidase (Fei et  al. 2009). The same group repeated 1997). The majority of these patients had trisomy 8. The associa-
the study with 376 patients and 369 controls and the relationship tion of BS with the myelodysplastic syndrome and the induction
between UBAC2 and BS was confirmed (Sawalha et  al. 2011). It of the pathergy reaction among patients with CML brings us
was also demonstrated in two independent Chinese sets of patients back to the argument of whether the neutrophil is central to the
with BS (Hou et  al. 2012). The second and third GWAS studies pathogenesis of BS.
were carried out with 1215 Turkish BS patients and 1278 controls In summary, several points are worth emphasizing regarding
(Remmer et al. 2010) and with 612 Japanese individuals with BS the pathogenesis of BS. Most immunological data have been based
and 740 controls (Mizuki et al. 2010). Both studies revealed similar either on healthy controls alone or patients with other autoimmune
results. The most significant association was on the HLA polymor- diseases. Data from patients with other vasculitides, infectious
phisms on chromosome 6 as expected. A second association was diseases such as tuberculosis, and those with other inflammatory
seen around the HLA-A region, supporting the previous finding of diseases such as gout and familial Mediterranean fever (FMF) are
Meguro et al. (2012) concerning HLA-A26. The third concerned clearly needed. Second, some of the observations related to the
the IL-10 gene resulting in decreased production of IL-10, an aetiopathogenesis might be more relevant to the severity of disease
important finding because IL-10 is an inhibitory cytokine that reg- expression than to susceptibility. The data related to HLA-B51, as
ulates both innate and adaptive immune responses. On the other discussed in Section Genetics, are a case in point. Finally, perhaps
hand, it is perhaps counterintuitive to suggest that IL-10 plays a not all the clinical findings of BS are explained by the same patho-
pivotal role in pathogenesis because IL-10 secretions do decrease genetic mechanism. The previously mentioned clinical association
with age (de Craen et al. 2005) and BS is most severe among the between acne-like lesions and arthritis certainly brings to mind
young. The fourth interesting association observed in the two stud- reactive arthritis. On the other hand, eye and genital lesions in BS do
ies concerned the IL-23R gene, a proinflammatory cytokine that not resemble the findings seen in reactive arthritis such as Reiter’s
stimulates T helper cells and increases the production of IL-1, IL-6, syndrome.
IL-17, and tumour necrosis factor-alpha (TNF-α). Polymorphisms
of this gene are associated with ankylosing spondylitis, inflam- Laboratory findings and
matory bowel disease, and psoriasis and may suggest that at least
a subgroup of BS patients may have features reminiscent of the differential diagnosis
spondyloarthritides (Yazici et al. 2012). Data presented in the 15th The laboratory features of BS are non-specific. Mild anaemia of
International Conference of Behçet’s disease also reported associa- chronic disease and neutrophilic leucocytosis are observed in 15%
tions with CCR1, STAT4 and KLRC4 (encoding a chemokine recep- of the patients, but they do not correlate with clinical activity of
tor, a transcription factor implicated in IL-12 and IL-23 signalling, the disease. The erythrocyte sedimentation (ESR) rate and the
and a natural killer cell receptor) and with ERAP1, which encodes C-reactive protein (CRP) may be moderately elevated; the latter
an endoplasmic reticulum-expressed aminopeptidase that trims may correlate with erythema nodosum and acute thrombophlebitis
peptides and loads them to MHC Class I. An epistatic interaction (Muftuoglu et al. 1986). Elevated serum alkaline phosphatase levels
between HLA B51 and ERAP was demonstrated and MEVF, NOD2, have been noted in approximately 10% of patients; the levels were
and TLR4, loci pertinent to innate immunity, were also implicated correlated with ESR and CRP (Takeuchi et al. 1989).
(Fresko et al. 2012). It remains to be seen what the new ENCODE Serum immunoglobulins (especially IgA) may be increased, but
revelation (Stamatoyannopoulos 2012) concerning epigenetics will rheumatoid factor and antinuclear antibody are absent. Tests for
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 479

antineutrophil cytoplasmic antibodies (ANCA) are usually nega- to an increased awareness and diagnosis of a second disease, the
tive. Patients with BS who have gastrointestinal involvement seem so-called Berkson’s bias (Ben-Chetrit and Yazici 2002). Important
to have high levels of anti-Saccharomyces cerevisiae antibodies considerations in the differential diagnosis are summarized in
(ASCA), but larger numbers of patients are needed to confirm this Table 34.5 (Barnes and Yazici 1999).
finding (Fresko et al. 2005).
A group of physicians involved in the care of large numbers of Table 34.5 Highlights of the clinical manifestations of BS
patients with BS formed an International Study Group (ISG) and
in 1990 published the ISG Criteria of Diagnosis of Behçet’s Disease Area affected Clinical features
(International Study Group for Behçet’s Disease 1990). After deter-
Mouth ulcers Majority similar to common aphthous ulcers regarding
mining the sensitivity and specificity of the various findings, a set
appearance, localization, and discomfort/ pain
of criteria that seemed to perform better than all the previously
More frequent and frequently multiple
proposed criteria emerged (International Study Group for Behçet’s
Disease 1992). Oral ulceration was the sine qua non, as its presence May scar
was almost universal. Genital ulcers Most commonly scrotal or vulval, painful, recurrent and
In the original control groups, the representation of Reiter’s syn- usually with scarring
drome and inflammatory bowel disease was deficient. The perfor- Urethral discharge and penile lesions very rare
mance of the criteria remained robust when a group of patients Skin Acneiform lesions as common acne in appearance
with inflammatory bowel disease was included in the control group and histology but also at uncommon sites such as the
(Tunc et  al. 2001). In the differential diagnosis of the individual extremities
patient it is important to remember that BS very rarely occurs with- Erythema nodosum-like lesions leaving pigmentation
out recurrent oral ulceration, which is also rather common in oth- Not psoriasis
erwise healthy people. On the other hand, oral ulcers of BS tend to Eyes Panuveitis and retinal vasculitis, usually bilateral occurring
occur more frequently. within about 2 years of the onset of the disease
The so-called MAGIC syndrome (oral and genital ulcers with Conjunctivitis and sicca syndrome most unusual
cartilage inflammation), which is relevant to the differential diag-
Joints Monarthritis in 50%, otherwise oligoarticular or
nosis of BS (Firestein et al. 1985), is probably nothing other than BS polyarticular involving relatively few joints
with secondary relapsing polychondritis. RAS in AIDS patients can
May be symmetrical; knees most frequently
also be confusing. The absence of dysuria or urethritis and the usual
Intermittent resolving in 2–4 weeks or chronic and
sparing of glans penis are helpful in diagnosis. It must be stressed
continuous
that in females secondary infection is very common.
Not involving sacroiliac joints or spine
Finally, there remains a group of patients in whom it is not pos-
sible to make a firm diagnosis of BS. Particularly bothersome are Deformity and erosions rare
those patients with no recurrent oral ulceration but with other Synovial fluid usually inflammatory with good mucin clot
manifestations, patients with isolated CNS or major vascular dis- Peripheral Subclinical peripheral large vein disease uncommon
ease, and those patients with inflammatory bowel disease. Among arterial and Usually involves large segments with skip areas without
these patients the presence of scar-forming genital disease is most venous disease embolization
helpful in diagnosis. Arteritis with occlusion and/or pseudoaneurysms
As discussed in Section Pathogenesis, there has been a debate Microaneurysms of the polyarteritic type very uncommon
concerning the inclusion of BS among the so called autoinflam-
Neurological Peripheral neuropathy and isolated cerebellar involvement
matory diseases (Gul 2005). These diseases, TRAPS (tumour involvement very unusual
necrosis factor receptor-associated periodic syndrome), MWS
Headaches with dural sinus thrombosis
(Muckle–Wells syndrome), HIDS (hyperimmunoglobulinaemia
Vascular CNS lesions including transverse myelitis-type
D with periodic fever syndrome), FMF, and to a lesser extent
manifestation
Crohn’s disease, are characterized by the absence of specific
Multiple sclerosis with aphthous ulcers a problem but no
high-titre antibodies, and specific mutations that are related to
plaques on MRI
the syndromes (CARD, NOD, and various pyrin mutations).
Aphthae, genital ulcers, acneiform skin lesions, pathergy like skin Pulmonary Haemoptysis associated with pulmonary arterial aneurysm
involvement Pulmonary artery occlusion
reactions, uveitis, arthritis, and orchioepididymitis are occasion-
ally seen during their course. However, apart from FMF, all of Pleural involvement uncommon
these are very rare. Most of these entities begin in infancy, but Interstitial involvement very rare
paediatric BS is infrequent. Paroxysmal attacks that last from a Gastrointestinal Severe abdominal pain
few hours to a couple of weeks with frequent serosal involvement involvement Ulcerative lesions at any level but mainly in the
and fever characterize the former whereas they are not typical ileocaecal region
of BS. Arthritis and skin involvement are non-specific findings Mild GI symptoms should not be associated with BS
that are also observed in SLE, a prototype autoimmune disease.
Cardiac disease Pericarditis, valve lesions, and coronary artery involvement
Extensive vasculitis, hypercoagulability, increased severity among
uncommon
males, and less severe disease after age 40 are findings that are
Rarely intracardiac thrombi
peculiar to BS (Yazici and Fresko 2005). It should also be kept
in mind that the presence of one of these conditions may lead (Barnes and Yazici 1999)
480 SECTION 5 vasculitic diseases and syndromes and related disorders

Prognosis manifestations of the disease can be summarized as in the Section


Management.
In a survey of the 20-year outcome of a cohort of patients with BS,
mortality was specifically increased among young males, among Mucocutaneous manifestations
whom morbidity was also the highest (Kural-Seyahi et al. 2003). Mucocutaneous lesions of BS follow a course with exacerbations
The main cause of mortality in this age group was major-vessel and remissions. Although they are not life or organ threatening as
disease (especially pulmonary arterial aneurysms) and neurologi- some of the other manifestations, they can severely affect the qual-
cal involvement. The mortality rate decreased with the passage of ity of life in some patients. In the presence of other major organ
time, as did all mucocutaneous and articular manifestations. Both lesions, the immunosuppressives that are prescribed for their con-
the onset of eye disease and its greatest damage were also usually trol are usually effective for mucocutaneous lesions also. When the
within the first few years of disease. These suggest that BS ‘burns only problem is mucocutaneous lesions, the treatment decision
out’ after approximately 40 years of age, with the exception of cen- should be made according to patients’ preferences, depending on
tral nervous system involvement and major-vessel disease. They can how bothersome these lesions are and how frequently they recur.
have late onset (5–10 years after the disease starts). The disease was For mild and infrequent oral ulcers, genital ulcers, and papulopus-
less severe among females for almost all of its manifestations. There tular lesions topical measures can be tried first. Systemic measures
were no female patients with arterial aneurysms in this cohort. The are needed if topical measures are not sufficient, the lesions tend
mortality rate was found to be similarly high in a French survey to recur frequently, or erythema nodosum is the dominant lesion.
(Saadoun et al. 2010a). The survey found that 41 (5%) in a cohort of A trial of topical interferon (Hamuryudan et al. 1991) and of top-
817 patients with BS had died after a median follow-up of 7.7 years. ical ciclosporin (Ergun et al. 1997) did not produce improvement
Among those who died, the mean age at death was 34.8 years and in the number, size, and healing time of oral ulcers. The topical
95% of these patients were men. The standardized mortality ratios measures that can be used for oral ulcers are steroid preparations,
compared to the matched general population was highest among lidocaine gel, chlorhexidine, and sucralfate suspension (Hatemi
the 15–24 years age group (2.99) followed by the 25–34 years age et al. 2008c; Alpsoy et al. 1999). Intralesional injection of recom-
group (2.9). The main causes of death were major vessel involve- binant human granulocyte–macrophage colony stimulating factor
ment such as arterial aneurysms and Budd–Chiari syndrome in (Alli et al. 1997) may be of some value. Patients should be encour-
44%, malignancies in 15%, central nervous system involvement in aged to maintain a good oral hygiene because this was shown to
12%, and sepsis in 12%. Multivariate analysis showed that male sex, be associated with the severity of mucocutaneous lesions (Mumcu
arterial involvement, and the number of flares were associated with et al. 2004).
mortality. Colchicine is frequently used for the management of mucocu-
Another study evaluated the long-term outcome in a group of taneous lesions. Randomized controlled trials (RCTs) and obser-
men who had no major organ involvement during the early years vational studies have shown that it is effective for nodular lesions
of their disease (Hamuryudan et  al. 2010). They suggested that and for genital ulcers, especially in women (Aktulga et  al. 1980;
age at onset of the disease, but not the initial clinical presentation, Yurdakul et al. 2001). The question whether a subset of BS patients
determined the long-term prognosis of men with BS. Young-onset benefit from colchicine for oral ulcers awaits a placebo-controlled
(<24 years of age) patients developed significantly more compli- withdrawal study.
cations requiring treatment with immunosuppressives during Depot glucocorticoids were useful in controlling erythema
follow-up compared with old-onset (≥25 years of age) patients. nodosum among females in a RCT, but were not effective in males
(Mat et al. 2005).
For resistant cases, azathioprine, mycophenolate sodium, thalido-
Management mide, IFN-α (Alpsoy et al. 2002), and in most resistant cases TNF-α
The aims of management in BS are rapid and effective suppression antagonists, can be used. Thalidomide was shown to be beneficial
of inflammatory exacerbations in the eyes, vascular system, central for oral and genital ulcers and papulopustular lesions in an RCT,
nervous system, and gastrointestinal system to prevent irreversible but the number of nodular lesions increased (Hamuryudan et al.
damage and prevention of exacerbations in these organs as well as 1998). Caution is required with its potential adverse events such as
skin, mucosa, and joint lesions, which can seriously affect the qual- teratogenicity and peripheral neuropathy (Ochonisky et al. 1994).
ity of life. Treatment strategies need to be customized according to IFN-α should also only be used in selected cases due to its cost and
prognostic factors such as age, sex, and disease duration, keeping in various side-effects. The efficacy of etanercept for mucocutaneous
mind that men with an early age at disease onset carry a higher risk lesions has been shown in an RCT and there are several case reports
of developing major organ involvement mandating more aggressive with infliximab and adalimumab (Melikoglu et al. 2005).
treatment strategies (Hamuryudan et al. 2010; Yazici et al. 1984a). Various antibiotics have also been claimed to be effective in sup-
It is important to follow a multidisciplinary approach due to the pressing mucocutaneous lesions and arthritis with the rationale of
multisystemic nature of this syndrome (Hatemi et al. 2008c). There eradicating streptococci implicated in the pathogenesis. A prospec-
is no widely accepted way of assessing disease activity, although a tive study reported favourable results with penicillin (Calgüneri
disease activity scheme has been prepared by international effort et al. 1996). The combination of benzathine benzylpenicillin with
(Hamuryudan et al. 1999). A recent physician survey suggested that colchicine has been reported to be superior to colchicine alone in
current treatment of BS became more intensive in terms of immu- managing mucocutaneous and arthritic episodes of BS (Al-Waiz
nosuppressive use when compared to 10–20  years ago (Turkstra et al. 2005). Minocycline was also claimed to be effective in treat-
et  al. 2012). However, there was a rather low agreement among ing mucocutaneous symptoms based on an uncontrolled study
the physicians. The approach to the management of the systemic (Kaneko et al. 1997). Beneficial results have also been reported with
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 481

rebapimide and dapsone (Matsuda et al. 2003; Sharquie et al. 2002). symptoms constitute its side-effects but they can usually be man-
Controlled trials with antibiotics are required before their use can aged by lowering the dose of the drug.
be recommended. IFN-α was observed to provide improved visual acuity, rep-
erfusion of occluded retinal vessels, regression of retinal neovas-
Joint involvement cularization, and high remission rates, long-lasting even after its
The arthritis of BS is usually self-limited (Yurdakul et  al. 1983). discontinuation (Kotter et al. 2004; Kotter et al. 2010; Deuter et al.
Non-steroidal anti-inflammatory drugs and local glucocorticoid 2010). Cytopenia, which is especially pronounced when interferon
injections are not very beneficial, but colchicine seems to be help- is used together with azathioprine (Hamuryudan et al. 2002), and
ful in both men and women (Yurdakul et al. 2001). Brief courses of depression can limit its use. Most of its adverse events are dose
oral glucocorticoids may be given in patients who have protracted dependent. Although the frequently used starting dose is 5 MU/
symptoms. In the occasional patient with prolonged episodes or day, a recent open study showed good results with 3 MU/day for 2
frequent recurrences of arthritis that are resistant to colchicine, weeks followed by 3MU three times a week, and escalating the dose
azathioprine may be added. IFN-α and TNF-α antagonists may be if necessary.
also used. There is growing experience with TNF-α antagonists, espe-
cially infliximab 5mg/kg every 6–8 weeks for eye involvement
Ocular involvement (Ohno et al. 2004; Sfikakis et al. 2004; Tugal-Tutkun et al. 2005).
Azathioprine was one of the first drugs extensively evaluated for Improvement in visual acuity, retinal vasculitis, retinitis, vitri-
ophthalmitis of BS. It is generally well tolerated with occasional tis, refractory cystoid macular oedema, optic disc oedema, and
bone marrow suppression and transaminase elevation. One group a decrease in the frequency of uveitis attacks and corticosteroid
showed that it was effective in controlling eye disease at a dose of need was reported. Moreover, in acute panuveitis attacks a faster
2.5 mg/kg/day (Yazici et al. 1990). It was also superior to placebo in response was observed compared to that of corticosteroids, with
preventing progression from unilateral to bilateral eye disease and clearance of inflammation within 24 hours (Markomichelakis et al.
in decreasing the use of glucocorticoids in patients who had estab- 2011). Improvement in uveoretinitis was observed in 92% of the
lished eye disease. Patients enrolled in the initial study with azathi- patients with 1-year follow-up in a study from Japan (Okada et al.
oprine were reanalysed with the aim of determining the long-term 2012). Concomitant use of other immunosuppressives such as aza-
effects of the drug (Hamuryudan et al. 1997). After 8 years, patients thioprine and ciclosporin is common. Relapses are common when
who were on azathioprine were still doing better (in terms of emer- it is discontinued. Intravitreal infliximab injections may also be
gence of blindness, drop in visual acuity, and extraocular com- beneficial (Markomichelakis et al. 2012). Caution is required for
plications) than patients who had received placebo. The outcome increased infection and especially tuberculosis risk. Adalimumab
was even better for patients allocated to azathioprine within the is also a promising agent and success has been reported in patients
first 2 years of eye involvement. This raises the question whether who switched from infliximab due to inefficacy or adverse events
young male patients without eye disease should be treated with (Olivieri et al. 2011). Other biologicals, such as gevokizumab and
this drug without waiting for its emergence. A  recent retrospec- canakinumab, are new candidates for the management of eye
tive survey showed that 7% of BS patients with eye involvement did involvement (Gul et al. 2012; Ugurlu et al. 2012).
not respond to azathioprine after a mean follow-up of 71.5 months Many ophthalmologists use glucocorticoids but there are no con-
while 41% had a partial response (Saadoun et al. 2010b). trolled trials evaluating their efficacy in eye disease. Glucocorticoids
Retinal vasculitis and severe visual loss at baseline were the should be used as briefly as possible and should be tapered as soon
predictors of an incomplete response. The presence of such poor as an immunosuppressive is added to the regimen. Local corticoid
prognostic factors may mandate the addition of ciclosporin to aza- eye drops combined with mydriatics are also used during acute
thioprine or the sole use of IFN-α right from the beginning. These attacks with the aim of preventing synechiae.
treatment modalities and TNF-α antagonists are also preferred in Vitrectomy and cataract surgery do not seem to add much to
patients who do not have an adequate response to azathioprine, the maintenance of vision in BS. Reports on the beneficial effects
whose attacks continue to recur, and visual acuity continues to of photocoagulation (Atmaca et  al. 1996)  certainly need further
drop. Ciclosporin 4–5mg/kg/day was proven to be beneficial in evaluation.
three RCTs, providing a rapid improvement in visual acuity and
decreasing the frequency and severity of ocular attacks (BenEzra Major vessel disease
et  al. 1988; Ozyazgan et  al. 1992; Masuda et  al. 1989). Patients An important clinical point concerning vascular disease is the
should be followed for hypertension and nephrotoxicity. It may relative absence of embolic phenomena in spite of the thrombotic
be preferable to refrain from using ciclosporin in BS patients with episodes. The role of anticoagulation in deep vein thrombosis has
CNS involvement, because ciclosporin, which may be neurotoxic, not been evaluated in a controlled study, but one group does not
was suggested to potentiate neurological involvement in patients use routine anticoagulation other than low-dose aspirin in elderly
with BS (Kotake et  al. 1999; Kotter et  al. 2006; Kato et  al. 2001; patients. Azathioprine is given to young patients based on the
Akman-Demir et al. 2008a). results of the long-term follow-up of patients in an azathioprine trial
Tacrolimus (FK506), a potent immunosuppressive macrolide (Hamuryudan et al. 1997) and the association of life-threatening
antibiotic, has been evaluated in patients with posterior uveitis pulmonary arterial aneurysms with deep vein thrombosis in this
resistant to ciclosporin. There is no controlled study of its efficacy age group (Hamuryudan et al. 1994; Hamuryudan et al. 2004a).
but open studies claim that it is effective in preserving visual acu- Venous thrombosis in BS results from inflammation of the
ity in cases of refractory posterior uveitis (Sloper et al. 1999). Mild vessel wall. Systemic immunosuppressives prevent relapses and
increases in serum creatinine, hyperglycaemia, and neurological post-thrombotic syndrome. A  retrospective study showed that
482 SECTION 5 vasculitic diseases and syndromes and related disorders

patients who are prescribed only anticoagulants have a signifi- topical and/or systemic antibiotics as needed. The vasculitic ulcers
cantly higher recurrence rate than those who receive additional usually require systemic immunosuppressives in addition to these
immunosuppressives (Desbois et al. 2012). Moreover, in another measures.
retrospective study it was observed that using anticoagulants with-
out immunosuppressives does not decrease the recurrence rate of Central nervous system disease
venous thrombosis (Ahn et  al. 2008). Considering the frequent There are no controlled studies evaluating the management of
association of venous thrombosis with arterial aneurysms, anti- CNS disease in BS. Thrombosis of the dural sinuses and increased
coagulants are not recommended in BS patients with deep vein intracranial pressure is empirically treated with 1 g of methylpred-
thrombosis in recent EULAR guidelines (Hatemi et  al. 2008c). nisolone administration intravenously daily for 3–7 days, followed
However, a recent physician survey showed that anticoagulants by oral maintenance doses. Parenchymal involvement, which
were still used by a majority of rheumatologists from Israel and manifests itself as varying degrees of cognitive, pyramidal, and
USA and 40% of the Turkish rheumatologists (Tayer-Shifman cerebellar symptoms, is usually treated with cyclophosphamide or
et  al. 2012). Azathioprine 2.5 mg/kg/day is usually preferred for azathioprine combined with glucocorticoids. Immunosuppressives
deep vein thrombosis in the extremities, but ciclosporin or IFN-α may improve the outcome and prevent recurrences. Long-term
may be used alternatively. Moderate doses of corticosteroids may use of azathioprine seems to improve the prognosis (Kurtuncu
be used during acute exacerbations. In more serious cases, such as et al. 2008). The use of ciclosporin should be avoided in CNS dis-
superior vena cava thrombosis or Budd–Chiari syndrome, cyclo- ease because of its potential neurotoxicity (Kato et al. 2001; Kotter
phosphamide may be preferred. There have been rather unsuc- et  al. 2006; Akman-Demir et  al. 2008a). Chlorambucil, claimed
cessful attempts with infliximab (Seyahi et al. 2007). Thrombi in beneficial by some (O’Duffy et al. 1984), has not found wide use,
the pulmonary vessels, which may resemble pulmonary emboli in mainly due to its myelotoxicity. An open trial in a small number of
imaging studies, should also be treated with immunosuppressives. patients suggests that low-dose, oral methotrexate (7.5–12.5 mg/
Limited experience with fibrinolytic therapy has been mostly dis- week) stops the progression of chronic neurological involvement
appointing. One study evaluated the role of side-to-side portocaval (Hirohata et al. 1998), but larger studies are needed to confirm this
shunts in the treatment of the dreaded Budd–Chiari syndrome, finding.
which has a reported mortality rate of 61% (Orloff and Orloff Cyclophosphamide and INF-α may be alternatives for more
1999). The procedure resulted in a substantial increase in survival severe and resistant cases. A few case reports suggest that the TNF-α
(80% at a mean of 10.6 years of follow-up), coupled with a decrease blocker infliximab is beneficial in treating parenchymal neurologi-
in ascites and diuretic use. cal disease refractory to other medications (Licata et al. 2003).
Pulmonary artery aneurysms, which may be life-threatening, Tocilizumab has also been used with beneficial results (Shapiro
should be treated aggressively with immunosuppressives and cor- et al. 2012). Dural sinus thrombosis, which usually presents with
ticosteroids. Pulse corticosteroids, usually 1 g intravenous methy- headache and increased intracranial pressure, is also treated with
prednisolone every other day for three doses, is started at the corticosteroids.
same time as monthly 1-g intravenous cyclophosphamide pulses.
Following this, oral prednisolone 1 mg/kg/day is started and tapered Gastrointestinal disease
slowly. This approach has significantly decreased the mortality of The treatment of gastrointestinal involvement is quite similar to that
pulmonary aneurysm in BS (Hamuryudan et al. 1994; Hamuryudan of Crohn’s disease. acetylsalicylic acid (5-ASA) derivatives or aza-
et al. 2004a). Cyclophosphamide is usually continued for 2 years, thioprine, together with corticosteroids during acute exacerbations
followed by azathioprine 2 mg/kg/day. Infliximab was found to be is usually sufficient to control gastrointestinal involvement of BS
effective in refractory cases with pulmonary artery involvement (Jung et al. 2012; Choi et al. 2000). Thalidomide and TNF-α antag-
(Tolosa-Vilella et al. 2011; Schreiber et al. 2011; Adler et al. 2012). onists have been used with success in resistant cases. Infliximab
In emergency situations, endovascular embolization may be tried; was beneficial in healing ulcers and fistulae, improving abdominal
however, this may be problematic due to coexisting venous thrombi. pain and rectal bleeding rapidly, and providing long-term remis-
For peripheral aneurysms surgery is usually required. Aorto-bi- sion. Perforation of the deep penetrating ulcers, which is somewhat
iliac by-pass may be preferred when the aneurysm is located in the more common than in Crohn’s disease, and severe bleeding may
infrarenal part of aorta and synthetic graft insertion in extremity require emergency operations such as ileocolectomy or hemicolec-
aneurysms (Tuzun et al. 2012). Concomitant immunosuppressives tomy. Recurrences after gastrointestinal surgery have been reported
are necessary to prevent relapses. Intra-arterial embolization has and inspection of the entire bowel during the operation is highly
been attempted (Cantasdemir et al. 2002; Mouas et al. 1996) but the recommended (Lee et al. 1997). It is important to maintain immu-
value of this procedure is not clear. There are various reports that nosuppressives such as azathioprine in these patients to improve
suggest endovascular surgery may be beneficial in major arterial the outcome and prevent relapses. A  younger age at diagnosis
and venous disease (Castelli et al. 2005). (<35 years), higher CRP levels, and higher disease activity index
Occasionally, patients present with cardiac disease such as score for intestinal involvement at baseline were shown to predict
mitral and aortic insufficiency and intracavitary thrombi. Surgical relapses (Jung et al. 2012).
thrombectomy (Kirali et al. 1998) and valve replacement (Zakaria In conclusion, management of eye, vascular, neurological, and
et al. 1998; Lee et al. 2008) have been reported with variable success. gastrointestinal involvement needs to be more aggressive with
Management of leg ulcers depends on the cause. For the immunosuppressives and corticosteroids to prevent damage. On
post-thrombophlebitic ulcers caused by venous stasis in BS patients the other hand, mucocutaneous and joint lesions can be man-
with deep vein thrombosis, the recommended measures are rest, aged with a more conservative approach, keeping safety in mind.
elevation, topical zinc preparations, keeping the ulcer clean, and Differences in the reimbursement of newer biologicals across
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 483

health-care systems in different countries also shape treatment Behçet’s disease: A double blind study by independent observers. Acta
strategies. The EULAR recommendations for the management of Dermato-Venereologica, 62, 158–9.
BS underline the lack of controlled evidence, especially for vascular, Altiparmak, M.R., Tanverdi, M., Pamuk, O.N., Tunc, R., and Hamuryudan,
V. (2002). Glomerulonephritis in Behçet’s disease: report of seven cases
neurological, and gastrointestinal involvement (Hatemi et al. 2009).
and review of the literature. Clinical Rheumatology, 21, 14–8.
Even for eye and mucocutaneous involvement, although there are
Al-Waiz, M.M., Sharquie, K.E., A-Qaissi, M.H., and Hayani, R.K. (2005).
RCTs, head to head comparison of different treatment modalities Colchicine and benzathine penicillin in the treatment of Behçet dis-
is lacking. Another shortcoming is the lack of widely accepted ease: a case comparative study. Dermatology Online Journal, 11, 3.
outcome measures to evaluate disease activity, damage, response Apaydin, S., Erek, E., Ulku, U., Hamuryudan, V., Yazici, H., and Sariyar, M.
to treatment, and definitions of flare for various types of organ (1999). A successful renal transplantation in Behçet’s syndrome. Annals
involvement to be used in drug trials. of the Rheumatic Diseases, 58, 719.
Arai, Y., Kohno, S., Takahashi, Y., et al. (2006). Autopsy case of neuro-Behçet’s
References disease with multifocal neutrophilic perivascular inflammation.
Adler, S., Baumgartner, I., and Villiger, P.M. (2012). Behçet’s dis- Neuropathology, 26, 579–85.
ease: Successful treatment with infliximab in 7 patients with severe vas- Arkin, C.R., Rothschild, B.M., Florendo, N.T., and Popoff, N. (1980). Behçet’s
cular manifestations. A retrospective analysis. Arthritis Care Research, syndrome with myositis. A case report with pathologic findings. Arthritis
64, 607–11. and Rheumatism, 23, 600–4.
Aguiar de Souza, D., Mestre, T., and Ferro, J.M. (2011). Cerebral venous Atmaca, L.S., Batioglu, F., and Idil, A. (1996). Retinal and disc neovas-
thrombosis in Behçet’s disease: a systematic review. Journal of Neurology, cularization in Behçet’s disease and efficacy of laser photocoagula-
258, 719–27. tion. Graefes Archives of Clinical and Experimental Ophtalmology,
Ahn, J.K., Lee, Y.S., Jeon, C.H., Koh, E.M., and Cha, H.S. (2008). Treatment 234, 94–9.
of venous thrombosis associated with Behcet’s disease: immunosuppres- Avci, O., Kutluay, E., Argon, N., Erdem, S., and Tahsin, G.A. (1998).
sive therapy alone versus immunosuppressive therapy plus anticoagula- Subclinical cerebral involvement in Behçet’s disease:  A  SPECT study.
tion. Clinical Rheumatology, 27, 201–5. European Journal of Neurology, 5, 49–53.
Akman-Demir, G., Ayranci, O., Kurtuncu, M., Vanli, E.N., Mutlu, M., and Aykutlu, E., Baykan, B., Akman-Demir, G., et al. (2006). Headache in Behçet’s
Tugal-Tutkun, I. (2008a). Cyclosporine for Behçet’s uveitis: is it associ- disease. Cephalalgia, 26, 180–6.
ated with an increased risk of neurological involvement? Clinical and Azizlerli, G., Kose, A.A., Sarica, R., et  al. (2003). Prevalence of Behçet’s
Experimental Rheumatology, 26, S84–90. disease in Istanbul, Turkey. International Journal of Dermatology,
Akman-Demir, G., Baykan-Kurt, B., Serdaroglu, P., et al. (1996). Seven-year 42, 803–6.
follow-up of neurologic involvement in Behçet’s syndrome. Archives of Azizlerli, G., Ozarmagan, G., Ovul, C., Sarica, R., and Mustafa, S.O. (1992).
Neurology, 53, 691–4. A new kind of skin lesion in Behçet’s disease: extragenital ulcers. Acta
Akman-Demir, G., Serdaroglu, P., and Tasçi, B. (1999). Clinical patterns of Dermato-Venereologica, 72, 286.
neurological involvement in Behçet’s disease: evaluation of 200 patients. Bang, D., Hur, W., Lee, E.S., and Lee, S. (1995). Prognosis and clinical rel-
The Neuro-Behçet Study Group. Brain, 122, 2171–82. evance of recurrent oral ulceration in Behçet’s disease. Journal of
Akman-Demir, G., Tuzun, E., Icoz, S., et  al. (2008b). Interleukin-6 in Dermatology, 22, 926–9.
neuro-Behçet’s disease: association with disease subsets and long-term Barnes, C.G. and Yazici, H. (1999). Behçet’s syndrome. Rheumatology
outcome. Cytokine, 44, 373–6. (Oxford), 38, 1171–4.
Akpolat, T., Diri, B., Oguz, Y., Yilmaz, E., Yavuz, M., and Dilek, M. (2003). Bayraktar, Y., Balkanci, F., Bayraktar, M., et  al. (1997). Budd-Chiari syn-
Behçet’s disease and renal failure. Nephrology Dialysis Transplantation, drome: a common complication of Behçet’s disease. American Journal of
18, 888–91. Gastroenterology, 92, 858–62.
Aktulga, E., Altaç, M., Müftüoglu, A., et al. (1980). A double blind study of Behçet, H. (1937). Ueber rezidivierende aphtose durch ein virus verursachte
colchicine in Behçet’s disease. Haematologica, 65, 399–402. geschwuere am mund, am auge und an den genitalien. Dermatologische
Al Dalaan, A., AlBalla, S., Alsukait, M., et  al. (1996). The prevalence of Woschenschrift, 105, 1152–7.
Behçet’s disease in al. Quassim region of Saudi Arabia. Revue de Ben-Chetrit, E. and Yazici, H. (2002). Thoughts on the proposed links
Rhumatisme (English Edition), 63, 538. between Behçet’s disease and familial Mediterranean fever. Clinical and
Alli, N., Karakayalı, G., Kahraman, I., and Artuz, F. (1997). Local intrale- Experimental Rheumatology, 20 (Suppl. 26), S1–2.
sional therapy with rhGM-CSF for a large genital ulcer in Behçet’s dis- BenEzra, D. and Cohen, E. (1986). Treatment and visual prognosis in Behçet’s
ease. British Journal of Dermatology, 136, 639–40. disease. British Journal of Ophthalmology, 70, 589–92.
Alpsoy, E., Dönmez, L., Önder, M., et al. (2007). Clinical features and natu- BenEzra, D., Cohen, E., Chajek, T., et al. (1988). Evaluation of conventional
ral course of Behçet’s disease in 661 cases. a multicenter study. British therapy versus cyclosporine A  in Behçet’s syndrome. Transplantation
Journal of Dermatology, 157, 901–6. Proceedings, 20, 136–43.
Alpsoy, E., Durusoy, C., Yilmaz, E., et al. (2002). Interferon alfa-2a in the Ben Hmida, M., Hachicha, J., Kaddour, N., et al. (1997). ANCA in Behçet’s
treatment of Behçet disease:  a randomized placebo-controlled and disease. Nephrology Dialysis Transplantation, 12, 2465–6.
double-blind study. Archives of Dermatology, 138, 467–71. Budak-Alpdogan, T., Demircay, Z., Alpdogan, O., et  al. (1997). Behçet’s
Alpsoy, E., Elpek, G.O., Yilmaz, F., et al. (2005). Androgen receptor levels of disease in patients with chronic myelogenous leukemia:  possible role
oral and genital ulcers and skin pathergy test in patients with Behçet’s of interferon-alpha treatment in the occurrence of Behçet’s symptoms.
disease. Dermatology, 210, 31–5. Annals of Haematology, 74, 45–8.
Alpsoy, E., Er, H., Durusoy, C., and Yılmaz, E. (1999). The use of sucralfate Budak-Alpdogan, T., Demircay, Z., Alpdogan, O., et al. (1998). Skin hyper-
suspension in the treatment of oral and genital ulceration of Behçet dis- reactivity of Behçet’s patients (pathergy reaction) is also positive in
ease: a randomized, placebo-controlled, double-blind study. Archives of interferon alpha-treated chronic myeloid leukaemia patients, indicating
Dermatology, 135, 529–32. similarly altered neutrophil functions in both disorders. British Journal
Al-Rawi, Z.S. and Neda, A.H. (2003). Prevalence of Behçet’s disease among of Rheumatology, 37, 1148–51.
Iraqis. Advances in Experimental and Medical Biology, 528, 37–41. Cakir, N., Dervis, E., Benian, O., et al. (2004). Prevalence of Behçet’s disease
Altaç, M., Tuzun, Y., Yurdakul, S., Binyildiz, P., and Yazici, H. (1982). in rural western Turkey: a preliminary report. Clinical and Experimental
The validity of pathergy test (non-specific skin hypereactivity) in Rheumatology, 22, S53–55.
484 SECTION 5 vasculitic diseases and syndromes and related disorders

Cakir, N., Yazici, H., Chamberlain, M.A., et al. (1991). Response to intra- Deniz, E., Guc, U., Buyukbabani, N., and Gul, A. (2010). HSP 60 expres-
dermal injection of monosodium urate crystals in Behçet’s syndrome. sion in recurrent oral ulcerations of Behçet’s disease. Oral Surgery Oral
Annals of the Rheumatic Diseases, 50, 634–6. Medicine and Oral Pathology, 110, 196–200.
Calguneri, M., Kiraz, S., Ertenli, I., Benekli, M., Karaarslan, Y., and Celik, I. Desbois, A.C., Wechsler, B., Resche-Rigon, M., et  al. (2012).
(1996). The effect of prophylactic penicillin treatment on the course of Immunosuppressants reduce venous thrombosis relapse in Behçet’s dis-
arthritis episodes in patients with Behçet’s disease. A randomized clini- ease. Arthritis and Rheumatism, 64, 2753–60.
cal trial. Arthritis and Rheumatism, 39, 2062–5. Deuter, C.M., Zierhut, M., Möhle, A., Vonthein, R., Stöbiger, N., and Kötter,
Cantasdemir, M., Kantarci, F., Mihmanli, I., et al. (2002). Emergency endo- I. (2010) Long-term remission after cessation of interferon-α treatment
vascular management of pulmonary artery aneurysms in Behçet’s dis- in patients with severe uveitis due to Behçet’s disease. Arthritis and
ease: report of two cases and a review of the literature. Cardiovascular Rheumatism, 62, 2796–805.
and Interventional Radiology, 25, 533–7. Dilsen, E., Konice, M., Aral, O., et  al. (1988). Behçet’s disease associated
Carletto, A., Pacor, M.L., Biasi, D., et  al. (1997). Changes of neutrophil with amyloidosis in Turkey and in the world. Annals of the Rheumatic
migration without modification of in vitro metabolism and adhesion in Diseases, 47, 157–63.
Behçet’s disease. Journal of Rheumatology, 24, 1332–6. Dinc, A., Takafuta, T., Jiang, D., Melikoglu, M., Saruhan-Direskeneli, G., and
Castelli, P., Caronno, R., Piffaretti, G., et al. (2005). Endovascular treatment Shapiro, S.S. (2003). Anti-endothelial cell antibodies in Behçet’s disease.
for superior vena cava obstruction in Behçet disease. Journal of Vascular Clinical and Experimental Rheumatology, 21 (Suppl. 30), S27–30.
Surgery, 41, 548–51. Direskeneli, H., Eksioglu-Demiralp, E., Kibaroglu, A., Yavuz, S., Ergun, T.,
Celik, A.F., Hatemi, I., Hatemi, G., et  al. (2010). A comparative study of and Akoglu, T. (1999). Oligoclonal T cell expansions in patients with
clinical, endoscopic, and histologic findings in patients with gastroin- Behçet’s disease. Clinical and Experimental Immunology, 117, 166–70.
testinal Behçet’s disease and Crohn’s disease. Clinical and Experimental Direskeneli, H., Fujita, H., and Akdis, C.A. (2011). Regulation of TH17 and
Rheumatology, 28, S111–2. regulatory T cells in patients with Behçet disease. Journal of Allergy and
Cetinel, B., Akpinar, H., Tufek, I., Uygun, N., Solok, V., and Yazici, H. Clinical Immunology, 128, 665–6.
(1999). Bladder involvement in Behçet’s syndrome. Journal of Urology, Direskeneli, H., Keser, G., D’Cruz, D., et al. (1995). Anti-endothelial cell anti-
161, 52–6. bodies, endothelial proliferation and von Willebrand factor antigen in
Cetinel, B., Obek, C., Solok, V., Yaycioglu, O., and Yazici, H. (1998). Urologic Behçet’s disease. Clinical Rheumatology, 14, 55–61.
screening for men with Behcet’s syndrome. Urology, 52, 863–5. Direskeneli, H. and Saruhan-Direskeneli, G. (2003). The role of heat shock
Chamberlain, M.A. and Robertson, R.J. (1993). A controlled study of sac- proteins in Behçet’s disease. Clinical and Experimental Rheumatology,
roiliitis in Behçet’s disease. British Journal of Rheumatology, 32, 693–8. 21, S44–8.
Chambers, J.C., Haskard, D.O., and Kooner, J.S. (2001). Vascular endothelial Diri, E., Mat, C., Hamuryudan, V., Yurdakul, S., Hizli, N., and Yazici, H.
function and oxidative stress mechanisms in patients with Behçet’s syn- (2001). Papulopustular skin lesions are seen more frequently in patients
drome. Journal of the American College of Cardiology, 37, 517–20. with Behçet’s syndrome who have arthritis:  a controlled and masked
Chang, H.K. and Kim, J.W. (2002). The clinical features of Behçet’s disease study. Annals of the Rheumatic Diseases, 60, 1074–6.
in Yongdong district: analysis of a cohort followed from 1997 to 2001. Duymaz-Tozkır, J., Uyar, A., Norman, P.J., et al. (2008). Distribution of killer
Journal of Korean Medical Science, 17, 784–9. immunoglobulin like receptor 3DL1/3DS1 alleles in Behçet’s disease.
Choi, I.J., Kim, J.S., Cha, S.D., et al. (2000). Long-term clinical course and Arthritis and Rheumatism, 58, S855.
prognostic factors in intestinal Behçet’s disease. Diseases of the Colon Efthimiou, J., Johnston, C., Spiro, S.G., and Turner-Warwick, M. (1986).
and Rectum, 43, 692–700. Pulmonary disease in Behçet’s syndrome. Quarterly Journal of Medicine,
Chun, S.I., Su, W.P., and Lee, S. (1990). Histopathologic study of cutaneous 227, 259–80.
lesions in Behçet’s syndrome. Journal of Dermatology, 17, 333–41. Erdogru, T., Kocak,T., Serdaroglu, P., Kadioglu, A., and Tellaloglu, S. (1999).
Coban, O., Bahar, S., Akman-Demir, G., et al. (1999). Masked assessment of Evaluation and therapeutic approaches of voiding and erectile dysfunc-
MRI findings: is it possible to differentiate neuro-Behçet’s disease from tion in neurological Behçet’s syndrome. Journal of Urology, 162, 147–53.
other central nervous system. Neuroradiology, 41, 255–60. Ergun, T., Gürbüz, O., Dogusoy, G., Mat, C., and Yazici, H. (1998).
Coskun, S., Seyahi, E., Mat, C., and Yazici, H. (2004). Female Behçet’s Histopathologic features of the spontaneous pustular lesions of Behçet’s
syndrome patients have more severe oral ulcerations. Clinical and syndrome. International Journal of Dermatology, 37, 194–6.
Experimental Rheumatology, 22, S–86, abs A16. Ergun, T., Gurbuz, O., Yurdakul, S., Hamuryudan, V., Bekiroglu, N., and
Crivelli, M.R., Aguas, S., Adler, I., Quarracino, C., and Berque, P. (1988). Yazici, H. (1997). Topical cylosporine-A for treatment of oral ulcers of
Influence of socioeconomic status on oral mucosa lesion prevalence in Behçet’s syndrome. International Journal of Dermatology, 36, 720.
schoolchildren. Community Dentistry and Oral Epidemiology, 16, 58–60. Ergun, T., Ince, U., Eksioglu-Demiralp, E., et al. (2001). HSP 60 expression
Davatchi, F., Jamshidi, A.R., Banihashemi, A.T., et al. (2008). WHO-ILAR in mucocutaneous lesions of Behçet’s disease. Journal of the American
COPCORD Study (Stage 1, Urban Study) in Iran. Journal of Academy of Dermatology, 45, 904–9.
Rheumatology, 35, 1384. Erkan, D., Yazici, Y., Sanders, A., Trost, D., and Yazici, H. (2004). Is
de Craen, A.J., Posthuma, D., Remarque, E.J., van den Biggelaar, A.H., Hughes-Stovin syndrome Behçet’s disease? Clinical and Experimental
Westendorp, R.G.,and Boomsma, D.I. (2005). Heritability estimates of Rheumatology, 22, S64–8.
innate immunity: an extended twin study. Genes and Immunity, 6, 167–70. Esin, S., Gül, A., Hodara, V., et al. (1997). Peripheral blood T cell expansions
De Menthon, M., Lavalley, M.P., Maldini, C., et al. (2009). HLAB51/B5 and in patients with Behçet’s disease. Clinical and Experimental Immunology,
the risk of Behçet’s disease:  a systematic review and meta-analysis of 107, 520–7.
case control genetic association studies. Arthritis and Rheumatism, 61, Espinosa, G., Font, J., Tassies, D., et  al. (2002). Vascular involvement in
1287–96. Behçet’s disease: relation with thrombophilic factors, coagulation acti-
Demirhindi, O., Yazici, H., Binyildiz, P., et al. (1981). Silivri Fener koyu ve vation and thrombomodulin. American Journal of Medicine, 112, 37–43.
yoresinde Behçet hastaligi sikligi ve bu hastaligin toplum icinde tarana- Farah, S., Al-Shubaili, A., Montaser, A., et al. (1998). Behçet’s syndrome: a
bilmesinde kullanabilecek bir yontem. Cerrahpasa Tip Dergisi, 12, report of 41 patients with emphasis on neurological manifestations.
509–14. Journal of Neurology Neurosurgery and Psychiatry, 64, 382–4.
Demirkesen, C., Tuzuner, N., Mat, C., et al. (2001). Clinicopathologic evalua- Fei, Y., Webb, R., Cobb, B.L., et al. (2009). Identification of a novel genetic
tion of nodular cutaneous lesions of Behçet syndrome. American Journal susceptibility loci for Behçet’s disease using a genome-wide association
of Clinical Pathology, 116, 341–6. study. Arthritis Research and Therapy, 11, R66.
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 485

Ferrante, A., Ciccia, F., Principato, A., Giardina, A.R., Impastato, R., Peralta, Hamuryudan, V., Fresko, I., Direskeneli, H., et al. (1999). Evaluation of the
S., and Triolo, G. (2010). A Th1 but not a Th17 response is present Turkish translation of a disease activity form for Behçet’s syndrome.
in the gastrointestinalinvolvement of Behçet’s disease. Clinical and Rheumatology (Oxford), 38, 734–6.
Experimental Rheumatology, 28, S27–30. Hamuryudan, V., Hatemi, G., Tascilar, K., et al. (2010). Prognosis of Behcet’s
Firestein, G.S., Gruber, H.E., Weisman, M.H., Zvaifler, N.J., Barber, J., and syndrome among men with mucocutaneous involvement at disease
O’Duffy, J.D. (1985). Mouth and genital ulcers with inflamed carti- onset:  long-term outcome of patients enrolled in a controlled trial.
lage: MAGIC syndrome. Five patients with features of relapsing poly- Rheumatology (Oxford), 49, 173–7.
chondritis and Behçet’s disease. American Journal of Medicine, 79, 65–72. Hamuryudan, V., Hatemi, G., Sut, N., Ugurlu, S., Yurdakul, S., and Yazici,
Frassanito, M.A., Dammacco, R., Cafforio, P., and Dammacco, F. (1999). H. (2012). Frequent oral ulceration during early disease may predict
Th1 polarization of the immune response in Behçet’s disease: a puta- a severe disease course in males with Behçet’s syndrome. Clinical and
tive pathogenetic role of interleukin-12. Arthritis and Rheumatism, 42, Experimental Rheumatology, 30 (Suppl. 72), S32–4.
1967–74. Hamuryudan, V., Mat, C., Saip, S., et al. (1998). Thalidomide in the treatment
Fresko, I. (2012). 15th International Congress on Behçet’s Disease. Clinical of mucocutaneous lesions of the Behçet’s syndrome. A  randomized,
and Experimental Rheumatology, 30 (Suppl. 72), S118–28. double-blind, placebo-controlled trial. Annals of Internal Medicine, 128,
Fresko, I., Ugurlu, S., Ozbakir, F., et al. (2005). Anti-Saccharomyces cerevi- 443–50.
siae antibodies (ASCA) in Behçet’s syndrome. Clinical and Experimental Hamuryudan, V. and Melikoglu, M. (2010). Vascular involvement in Behçet’s
Rheumatology, 23 (Suppl. 38), S67–70. syndrome. In Behçet’s syndrome, 1st edn (ed. Y. Yazici and H. Yazici), pp.
Fukuda, Y. and Watanabe, I. (1979). Pathological studies on intestinal 115–34. Springer, New York.
Behçet’s (entero-Behçet’s) disease. In Behçet’s disease. Proceedings of an Hamuryudan, V., Oz, B., Tuzun, H., and Yazici, H. (2004b). The menac-
International Symposium on Behçet’s Disease (ed. Dilsen, N., Konice, M., ing pulmonary artery aneurysms of Behçet’s syndrome. Clinical and
and Ovul, C.), pp. 90–5. Excerpta Medica, Amsterdam. Experimental Rheumatology, 22, S1–3.
George, R.K., Chan, C.C., Whitcup, S.M., and Nussenblatt, R.B. (1997). Hamuryudan, V., Ozyazgan, Y., Fresko, Y., Ma, T.C., Yurdakul, S., and Yazici,
Ocular immunopathology of Behçet’s disease. Survey of Ophthalmology, H. (2002). Interferon alfa combined with azathioprine for the uveitis of
42, 157–62. Behçet’s disease:  an open study. Israel Medical Association Journal, 4,
Geri, G., Terrier, B., Rosenzwajg, M., et al. (2011). Critical role of IL-21 in 928–30.
modulating TH17 and regulatory T cells in Behçet disease. Journal of Hamuryudan, V., Ozyazgan, Y., Hizli, N., et  al. (1997). Azathioprine in
Allergy and Clinical Immunology, 128, 655–64. Behçet’s syndrome:  Effects on long term prognosis. Arthritis and
Geri, G., Wechsler, B., Thi Huong Du, L., et al. (2012). Spectrum of cardiac Rheumatism, 40, 769–74.
lesions in Behçet disease: a series of 52 patients and review of the litera- Hamuryudan, V., Yurdakul, S., Rosenkaimer, F., and Yazici, H. (1991).
ture. Medicine (Baltimore), 91, 25–34. Inefficacy of topical alpha interferon in the treatment of oral ulcers in
Gogus, F., Fresko, I., Elbir, Y., Eksioglu-Demiralp, E., and Direskeneli, Behçet’s syndrome: a randomized, double blind trial. British Journal of
H. (2005). Oxidative burst response to monosodium urate crys- Rheumatology, 30, 395–6.
tals in patients with Behçet’s syndrome. Clinical and Experimental Hamuryudan, V., Yurdakul, S., Moral, F., et al. (1994). Pulmonary arterial
Rheumatology, 23 (Suppl. 38), S81–5. aneurysms in Behçet’s syndrome: A report of 24 cases. British Journal of
Gonzalez-Gay, M.A., Garcia-Porrua, C., Branas, F., Lopez-Lazaro, L., and Rheumatology, 33, 48–51.
Olivieri, I. (2000). Epidemiologic and clinical aspects of Behçet’s dis- Hamzaoui, K. (2011). Th17 cells in Behçet’s disease: a new immunoregula-
ease in a defined area of Northwestern Spain, 1988-1997. Journal of tory axis. Clinical and Experimental Rheumatology, 2011, 29, S71–6.
Rheumatology, 27, 703–7. Hatemi, G., Bahar, H., Uysal, S., et al. (2004). The pustular skin lesions in
Goto, H., Mochizuki, M., Yamaki, K., et al. (2007). Epidemiological survey of Behçet’s syndrome are not sterile. Annals of the Rheumatic Diseases, 63,
intraocular inflammation in Japan. Japanese Journal of Ophthalmology, 1450–2.
51, 41–4. Hatemi, G., Fresko, I., Tascilar, K., and Yazici, H. (2008b). Increased
Grattan, C.E. and Scully, G. (1986). Oral ulceration: a diagnostic problem. enthesopathy among Behçet’s syndrome patients with acne and arthri-
British Medical Journal, 292, 1093–4. tis: an ultrasonography study. Arthritis and Rheumatism, 58, 1539–45.
Griffin, J.W.Jr, Harrison, H.B., Tedesco, F.J., and Mills, L.R. 4th (1982). Hatemi, G., Fresko, I., Yurdakul, S., et al. (2010). Reply to letter by Priori
Behçet’s disease with multiple sites of gastrointestinal involvement. et al commenting on whether Behçet’s syndrome patients with acne and
Southern Medical Journal, 75, 1405–8. arthritis comprise a true subset. Arthritis and Rheumatism, 62, 305–6.
Gul, A. (2005). Behçet’s disease as an autoinflammatory disorder. Current Hatemi, G., Silman, A., Bang, D, et al. (2008c). EULAR Expert Committee.
Drug Targets Inflammation and Allergy, 4, 81–3. EULAR recommendations for the management of Behçet disease.
Gul, A., Esin, S., Dilsen, N., Konice, M., Wigzell, H., and Biberfeld, P. (1995). Annals of the Rheumatic Diseases, 67, 656–62.
Immunohistology of skin pathergy reaction in Behçet’s disease. British Hatemi, G., Silman, A., Bang, D., et al. (2009). Management of Behçet dis-
Journal of Dermatology, 132, 901–7. ease:  a systematic literature review for the European League Against
Gul, A., Inanc, M., Ocal, L., Aral, O., and Konice, M. (2000). Familial aggre- Rheumatism evidence-based recommendations for the management of
gation of Behçet’s disease in Turkey. Annals of the Rheumatic Diseases, Behçet disease. Annals of the Rheumatic Diseases, 68, 1528–34.
59, 622–5. Hatemi, I., Hatemi, G., Celik, A.F., et al. (2008a). Frequency of pathergy phe-
Gul, A. and Ohno, S. (2010). Genetics of Behçet’s disease in Behçet’s syn- nomenon and other features of Behçet’s syndrome among patients with
drome. In Behçet’s syndrome, 1st edn (ed. Y. Yazici and H. Yazici), pp. inflammatory bowel disease. Clinical and Experimental Rheumatology,
265–76. Springer, New York. 26, S91–5.
Gul, A., Tugal-Tutkun, I., Dinarello, C.A., et al. (2012). Interleukin-1β-regulating Hirohata, S. (2008). Histopathology of central nervous system lesions in
antibody XOMA 052 (gevokizumab) in the treatment of acute exacerba- Behçet’s disease. Journal of Neurological Sciences, 267, 41–7.
tions of resistant uveitis of Behcet’s disease: an open-label pilot study. Hirohata, S., Arai, H., and Matsumoto, T. (2003). Immunohistological stud-
Annals of the Rheumatic Disease, 71, 563–6. ies in neuro-Behçet’s disease. Advances in Experimental Medicine and
Gunaydin, I., Ustundag, C., Kaner, G., et al. (1994). The prevalence of Sjögren’s Biology, 528, 385–7.
syndrome in Behçet’s syndrome. Journal of Rheumatology, 21, 1662–4. Hirohata, S., Suda, H., and Hashimoto, T. (1998). Low-dose methotrexate for
Hamuryudan, V., Er, T., Seyahi, E., et al. (2004a). Pulmonary artery aneu- progressive neuropsychiatric manifestations in Behçet’s disease. Journal
rysms in Behçet syndrome. American Journal of Medicine, 117, 867–70. of the Neurological Sciences, 159, 181–5.
486 SECTION 5 vasculitic diseases and syndromes and related disorders

Hou, S., Shu, Q., Jiang, Z., et al. (2012). Replication study confirms the associ- Kidd, D. (2006). The prevalence of headache in Behçet’s syndrome.
ation between UBAC2 and Behçet’s disease in two independent Chinese Rheumatology, 45, 621–3.
sets of patients and controls. Arthritis Research and Therapy, 14, R70. Kim, J., Park, J.A., Lee, E.Y., Lee, Y.J., Song, Y.W., and Lee, E.B. (2010).
Houng, D.I., Wechsler, B., Papo, T., et al. (1997). Endomyocardial fibrosis in Imbalance of Th17 to Th1cells in Behçet’s disease. Clinical and
Behçet’s disease. Annals of the Rheumatic Diseases, 56, 205–8. Experimental Rheumatology, 28, S16–9.
Ideguchi, H., Suda, A., Takeno, M., Ueda, A., Ohno, S., and Ishigatsubo, Kingston, M., Ratcliffe, J.R., Alltree, M., and Merendino, K.A. (1979).
Y. (2011a) Characteristics of vascular involvement in Behçet’s dis- Aneurysm after arterial puncture in Behçet’s disease. British Medical
ease in Japan: a retrospective cohort study. Clinical and Experimental Journal, 11, 1766–7.
Rheumatology, 29, S47–53. Kirali, K., Civelek, A., Daglar, B., et al. (1998). An uncommon complication
Ideguchi, H., Suda, A., Takeno, M., Ueda, A., Ohno, S., and Ishigatsubo, Y. of Behçet’s disease: intracardiac thrombosis needing surgical treatment.
(2011b) Behçet disease: evolution of clinical manifestations. Medicine Thoracic and Cardiovascular Surgeon, 46, 102–5.
(Baltimore), 90, 125–32. Kobayashi, M., Ito, M., Nakagawa, A., et al. (2000). Neutrophil and endothe-
Idil, A., Gurler, A., Boyvat, A., et al. (2002).The prevalence of Behçet’s dis- lial cell activation in the vasa vasorum in vasculo-Behçet disease.
ease above the age of 10 years. The results of a pilot study conducted at Histopathology, 36, 362–71.
the Park Primary Health Care Center in Ankara, Turkey. Ophthalmic Kobayashi, T., Kikawada, T., Shima, K., and Fukuda, O. (1982). Ulceration
Epidemiology, 9, 325–31. and stenosis of the hypopharynx and its surgical management. Head and
Iida, M., Kobayashi, H., Matsumoto, T., et al. (1993). Intestinal Behçet’s dis- Neck Surgery, 5, 66–9.
ease: serial changes at radiography. Radiology, 188, 65–9. Kocer, N., Islak, C., Siva, A., et al. (1999). CNS involvement in neuro-Behçet
Imamura, Y., Kurokawa, M.S., Yoshikawa, H., et al. (2005). Involvement of syndrome:  an MR study. American Journal of Neuroradiology, 20,
Th1 cells and heat shock protein 60 in the pathogenesis of intestinal 1015–24.
Behcet’s disease. Clinical and Experimental Immunology, 139, 371–8. Kone-Paut, I., Geisler, B., and Wechsler, A., et al. (1999). Familial aggregation
International Study Group for Behçet’s Disease (1990). Criteria for diagnosis in Behçet’s disease: high frequency in siblings and parents of pediatric
of Behçet’s disease. Lancet, 335, 1078–80. probands. Journal of Pediatrics, 135, 89–93.
International Study Group for Behçet’s Disease (1992). Evaluation of diag- Kone-Paut, I., Yurdakul, S., Bahabri, S.A., et al. (1998). Clinical features of
nostic (classification) criteria in Behçet’s disease—towards internation- Behçet’s disease in children: an international colloborative study of 86
ally agreed criteria. British Journal of Rheumatology, 31, 299–308. cases. Journal of Pediatrics, 132, 721–5.
Jung, Y.S., Hong, S.P., Kim, T.I., Kim, W.H., and Cheon, J.H. (2012). Korman, U., Cantasdemir, M., Kurugoglu, S., et al. (2003). Enteroclysis find-
Long-term clinical outcomes and factors predictive of relapse after ings of intestinal Behçet disease: a comparative study with Crohn dis-
5-aminosalicylate or sulfasalazine therapy in patients with intestinal ease. Abdominal Imaging, 28, 308–12.
Behcet disease. Journal of Clinical Gastroenterology, 46, e38–45. Kotake, S., Higashi, K., Yoshikawa, K., Sasamoto, Y., Okamoto, T., and
Kaklamani, G., Vaiopoulos, G., and Kaklamanis, P.G. (1998). Behçet’s dis- Matsuda, H. (1999). Central nervous system symptoms in patients
ease. Seminars in Arthritis and Rheumatism, 27, 197–217. with Behçet disease receiving cyclosporine therapy. Ophthalmolog,
Kaklamani, V.G., Tzonou, A., Markomichelakis, N., Papazoglou, S., and 106, 586–9.
Kaklamanis, P.G. (2003). The effect of smoking on the clinical features of Kotter, I., Günaydin, I., Batra, M., et al. (2006). CNS involvement occurs more
Adamantiades-Behçet’s disease. Advances in Experimental Medicine and frequently in patients with Behçet’s disease under cyclosporin A (CSA)
Biology, 528, 323–7. than under other medications—results of a retrospective analysis of 117
Kaneko, F., Oyama, N., and Nishibu, A. (1997). Streptococcal infection in the cases. Clinical Rheumatology, 25, 482–6.
pathogenesis of Behçet’s disease and clinical effects of minocycline on Kotter, I., Gunaydin, I., Zierhut, M., and Stubiger, N. (2004). The use of
the disease symptoms. Yonsei Medical Journal, 38, 444–54. interferon alpha in Behçet disease: review of the literature. Seminars in
Kaneko, F., Tojo, M., Sato, M., and Isogai, E. (2003). The role of infectious Arthritis and Rheumatism, 33, 320–35.
agents in the pathogenesis of Behçet’s disease. Advances in Experimental Kotter, I., Hamuryudan, V., Oztürk, Z.E., and Yazici, H. (2010). Interferon
Medicine and Biology, 528, 181–3. therapy in rheumatic diseases:  state-of-the-art. Current Opinion in
Kansu, E., Sahin, G., Sahin, F., Sivri, B., Sayek, I., and Batman, F. (1986). Rheumatology, 22, 278–83.
Impaired prostacyclin synthesis by vessel walls in Behçet’s disease. Krause, I., Leibovici, L., Guedj, D., Molad, Y., Uziel, Y., and Weinberger, A.
Lancet, 2, 1154. (1999). Disease patterns of patients with Behçet’s disease demonstrated
Karaca, M., Hatemi, G., Sut, N., and Yazici, H. (2012). The papulopustular by factor analysis. Clinical and Experimental Rheumatology, 17, 347–50.
lesion/arthritis cluster of Behçet’s syndrome also clusters in families. Krause, I., Molad, Y., Mitrani, M., and Weinberger, A. (2000). Pathergy reac-
Rheumatology (Oxford), 51, 1053–60. tion in Behçet’s disease: lack of correlation with mucocutaneous mani-
Karasneh, J.A., Hajeer, A.H., Silman, A., Worthington, J., Ollier, W.E., and festations and systemic disease expression. Clinical and Experimental
Gul, A. (2005a). Polymorphisms in the endothelial nitric oxide syn- Rheumatology, 18, 71–4.
thase gene are associated with Behçet’s disease. Rheumatology (Oxford), Krause, I., Monselise, Y., Milo, G., and Weinberger, A. (2002).
44, 614–7. Anti-Saccharomyces cerevisiae antibodies–a novel serologic marker for
Karatas, G.K., Onder, M., and Meray, J. (2002). Autonomic nervous system Behçet’s disease. Clinical and Experimental Rheumatology, 20 (Suppl.
involvement in Behçet’s disease. Rheumatology International, 22, 155–9. 26), S21–4.
Kasahara, Y., Tanaka, S., Nishino, M., Umemura, H., Shiraha, S., and Kulahli, I., Balci, K., Koseoglu, E., Yuce, I., Cagli, S., and Senturk, M.(2005).
Kuyama, T. (1981). Intestinal involvement in Behçet’s disease: Review Audio-vestibular disturbances in Behçet’s patients: report of 62 cases.
of 136 surgical cases in Japanese literature. Diseases of the Colon and Hearing Research, 203, 28–31.
Rectum, 24, 103–6. Kural-Seyahi, E., Fresko, I., and Seyahi, N. (2003). The long-term mortal-
Kato, Y., Numaga, J., Kato, S., Kaburaki, T., Kawashima, H., and Fujino, Y. ity and morbidity of Behçet syndrome:  a 2-decade outcome survey
(2001). Central nervous system symptoms in a population of Behçet’s of 387 patients followed at a dedicated center. Medicine (Baltimore),
disease patients with refractory uveitis treated with cyclosporine A. 82, 60–76.
Clinical and Experimental Ophthalmology, 29, 335–6. Kurhan-Yavuz, S., Direskeneli, H., Bozkurt, N., et  al. (2000). Anti-MHC
Kazokoglu, H., Onal, S., Tugal-Tutkun, I. et  al. (2008). Demographic and autoimmunity in Behçet’s disease: T cell responses to an HLA-B-derived
clinical features of uveitis in tertiary centers in Turkey. Ophthalmic peptide cross-reactive with retinal-S antigen in patients with uveitis.
Epidemiology, 15, 285–93. Clinical and Experimental Immunology, 120, 162–6.
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 487

Kurosawa, M., Inaba, Y., Nishibu, A., et al. (2004). Nationwide epidemiologi- Matsuda, T., Ohno, S., Hirohata, S., et  al. (2003). Efficacy of rebamip-
cal survey of Behçet’s disease in 2003 in Japan. Clinical and Experimental ide as adjunctive therapy in the treatment of recurrent oral aphthous
Rheumatology, 22, S84. ulcers in patients with Behçet’s disease:  a randomised, double-blind,
Kurtuncu, M., Tuzun, E., Mutlu, M., et al. (2008). Clinical patterns and course placebo-controlled study. Drugs in R&D, 4, 19–28.
of Neuro-Behcet’s disease: analysis of 354 patients comparing cases pre- Mege, J.L., Dilsen, N., Sanguedolce, V., et  al. (1993). Overproduction of
sented before and after 1990. Clinical and Experimental Rheumatology, monocyte derived tumor necrosis factor alpha, interleukin (IL) 6, IL-8
26, S17. and increased neutrophil superoxide generation in Behçet’s disease.
Lakhanpal, S., Tani, K., Lie, J.T., Katoh, K., Ishigatsubo, Y., and Ohokubo, T. A comparative study with familial Mediterranean fever and healthy sub-
(1985). Pathologic features of Behçet’s syndrome. A review of Japanese jects. Journal of Rheumatology, 20, 1544–9.
autopsy registry data. Human Pathology, 16, 790–5. Meguro, A., Inoko, H., Ota, M., et al. (2012). Genetics of Behçet’s disease inside
Lee, I., Park, S., Hwang, I., et al. (2008). Cardiac Behçet disease presenting and outside the MHC. Annals of the Rheumatic Diseases, 69, 747–54.
as aortic valvulitis/aortitis or right heart inflammatory mass: a clinico- Melikoglu, M., Altiparmak, M.R., Fresko, I., et al. (2001). A reappraisal of
pathologic study of 12 cases. American Journal of Surgical Pathology, amyloidosis in Behçet’s syndrome. Rheumatology (Oxford), 40, 212–5.
32, 390–8. Melikoglu, M., Fresko, I., Mat, C., et al. (2005). Short-term trial of etanercept
Lee, K.H., Chung, H.S., Kim, H.S., et al. (2003). Human alpha-enolase from in Behçet’s disease: a double blind, placebo controlled study. Journal of
endothelial cells as a target antigen of anti-endothelial cell antibody in Rheumatology, 32, 98–105.
Behçet’s disease. Arthritis and Rheumatism, 48, 2025–35. Melikoglu, M., Ugurlu, S., Tascilar, K., et al. (2008). Large vessel involvement
Lee, K.S., Kim, S.J., Lee, B.C., Yoon, D.S., Lee, W.J., and Chi, H.S. (1997). in Behçet’s syndrome:a retrospective survey. Annals of the Rheumatic
Surgical treatment of intestinal Behçet’s disease. Yonsei Medical Journal, Diseases, 67, 67.
38, 455–60. Melikoglu, M., Uysal, S., Kaplan, G., et  al. (2002). Leukocyte phenotype
Lehner, T. (1997). The role of heat shock protein, microbial and autoim- and cytokine expression at the skin pathergy reaction in Behçet’s dis-
mune agents in the etiology of Behçet’s disease. International Review of ease: evidence for a type I immune response. Arthritis and Rheumatism,
Immunology, 14, 21–32. 46, S191.
Lehner, T. and Batchelor, J.R. (1979). Classification and an immunogenetic Mizuki, N., Meguro, A., Ota, M., et  al. (2010). Genome-wide association
basis of Behçet’s syndrome. In Behçet’s disease (ed. T. Lehner and C.G. studies identify IL23R-IL12RB2 and IL10 as Behçet’s disease susceptibil-
Barnes,), pp. 13–32. Excerpta Medica, London. ity loci. Nature Genetics, 42, 703–6.
Leiba, M., Seligsohn, U., Sidi, Y., et  al. (2004). Thrombophilic factors are Mizuki, N., Meguro, A., Tohnai, I., Gül, A., Ohno, S., and Mizuki, N. (2007).
not the leading cause of thrombosis in Behçet’s disease. Annals of the Association of major histocompatibility complex class I chain-related
Rheumatic Diseases, 63, 1445–9. gene A  and HLA-B alleles with Behçet’s disease in Turkey. Japanese
Licata, G., Pinto, A., and Tuttolomondo, A. (2003). Anti-tumour necro- Journal of Ophthalmology, 51, 431–6.
sis factor alpha monoclonal antibody therapy for recalcitrant cerebral Mogulkoc, N., Burgess, M.I., and Bishop, P.W. (2000). Intracardiac thrombus
vasculitis in a patient with Behçet’s syndrome. Annals of the Rheumatic in Behçet’s disease: a systematic review. Chest, 118, 479–87.
Diseases, 62, 280–1. Molinari, N., Kone Paut, I., Manna, R., Demaille, J., Daures, J.P., and Touitou,
Mahesh, S.P., Li, Z., Buggage, R., et  al. (2005). Alpha tropomyosin as a I. (2003). Identification of an autosomal recessive mode of inheritance
self-antigen in patients with Behçet’s disease. Clinical and Experimental in paediatric Behçet’s families by segregation analysis. American Journal
Immunology, 140, 368–75. of Medical Genetics Part A, 122, 115–8.
Mahr, A., Belarbi, L.,Wechsler, B., et al. (2008). Population-based prevalence Monastero, R., Camarda, C., Pipia, C., et al. (2004). Cognitive impairment
study of Behçet’s disease: differences by ethnic origin and low variation in Behçet’s disease patients without overt neurological involvement.
by age at immigration. Arthritis and Rheumatism, 58, 3951–9. Journal of the Neurological Sciences, 220, 99–104.
Mahr, A., Guillevin, L., Poissonnet, M., and Aymé, S. (2004). Prevalences of Mori, S., Yoshihara, A., Kavamura, H., Takeuchi, A., Hashimoto, T., and
polyarteritis nodosa, microscopic polyangiitis, Wegener’s granulomato- Inaba, G. (1983). Esophageal involvement in Behçet’s disease. American
sis, and Churg-Strauss syndrome in a French urban multiethnic popula- Journal of Gastroenterology, 78, 548–53.
tion in 2000: a capture-recapture estimate. Arthritis and Rheumatism, Mouas, H., Lorthorary, O., Lacombe, P., et al. (1996). Embolization of mul-
51, 92–9. tiple pulmonary arterial aneurysms in Behçet’s disease. Scandinavian
Markomichelakis, N., Delicha, E., Masselos, S., Fragiadaki, K., Kaklamanis, Journal of Rheumatology, 25, 58–60.
P., and Sfikakis, P.P. (2011). A single infliximab infusion vs corticos- Muftuoglu, A.U., Yazici, H., Yurdakul, S., et  al. (1986). Behçet’s disease.
teroids for acute panuveitis attacks in Behçet’s disease: a comparative Relation of serum C-reactive protein and erythrocyte sedimentation
4-week study. Rheumatology (Oxford), 50, 593–7. rates to disease activity. International Journal of Dermatology, 25, 235–9.
Markomichelakis, N., Delicha, E., Masselos, S., and Sfikakis, P.P. (2012). Muller, W. and Lehner, T. (1982). Quantitative electron microscopical analy-
Intravitreal infliximab for sight-threatening relapsing uveitis in Behçet sis of leucocyte infiltration in oral ulcers of Behçet’s syndrome. British
disease: a pilot study in 15 patients. American Journal of Ophthalmology, Journal of Dermatology, 106, 535–44.
154, 534–41. Mumcu, G., Ergun, T., Inanc, N., et  al. (2004). Oral health is impaired in
Masatlioglu, S., Seyahi, E., Tahir-Turanli, E., et al. (2010). A twin study in Behçet’s disease and is associated with disease severity. Rheumatology
Behçet’s syndrome. Clinical and Experimental Rheumatology, 28, S62–6. (Oxford), 43, 1028–33.
Masuda, K., Nakajima, A., Urayama, A., Nakae, K., Kogure, M., and Inaba, Nara, K., Kurokawa, M.S., Chiba, S., Yoshikawa, H., Tsukikawa, S., Matsuda,
G. (1989). Double-masked trial of cyclosporin versus colchicine and T., and Suzuki, N. (2008). Involvement of innate immunity in the
long-term open study of cyclosporin in Behçet’s disease. Lancet, 1, pathogenesis of intestinal Behçet’s disease. Clinical and Experimental
1093–6. Immunology, 152, 245–51.
Mat, C., Goksugur, N., Engin, B., Yurdakul, S., and Yazici, H. (2006). The fre- Nobili, F., Cutolo, M., Sulli, A., Vitali, P., Vignola, S., and Rodriguez, G.
quency of scarring after genital ulcers in Behçet’s Syndrome: A prospec- (2002). Brain functional involvement by perfusion SPECT in systemic
tive study. International Journal of Dermatology, 45, 554–6. sclerosis and Behçet’s disease. Annals of the New  York Academy of
Mat, C., Nazarbaghi, M., Tuzun, Y., et al. (1998). Wound healing in Behçet’s Sciences, 966, 409–14.
syndrome. International Journal of Dermatology, 37, 120–3. Ochonisky, S., Verroust, J., Bastuji-Garin, S., Gherardi, R., and Revuz, J. (1994).
Mat, C., Yurdakul, S., Uysal, S., et al. (2005). A double-blind trial of depot glu- Thalidomide neuropathy incidence and clinico-electrophysiologic find-
cocorticoids in Behçet’s syndrome. Rheumatology (Oxford), 45, 245–7. ings in 42 patients. Archives of Dermatology, 130, 66–9.
488 SECTION 5 vasculitic diseases and syndromes and related disorders

O’Duffy, J.D. (1994). Behçet’s disease. Current Opinion in Rheumatology, Saadoun, D., Wechsler, B., Terrada, C., et al. (2010b). Azathioprine in severe
6, 39–43. uveitis of Behçet’s disease. Arthritis Care Research (Hoboken), 62, 1733–8.
O’Duffy, J.D., Robertson, D.M., and Goldstein, N.P. (1984). Chlorambucil Saatci, I., Ozmen, M., Balkanci, F., Akhan, O., and Senaati, S. (1993). Behçet’s
in the treatment of uveitis and maningoencephalitis of Behçet’s disease. disease in the etiology of Budd-Chiari disease. Angiology, 44, 392–8.
American Journal of Medicine, 76, 75–84. Saip, S., Siva, A., Altintas, A., et al. (2005). Headache in Behçet’s syndrome.
Oflaz, H., Mercanoglu, F., Karaman, O., et  al. (2005). Impaired Headache, 45, 911–9.
endothelium-dependent flow-mediated dilation in Behçet’s dis- Sakaguchi, T., Ibe, M., Miwa, K., et al. (1997). Predominant role of N-terminal
ease: more prominent endothelial dysfunction in patients with vascular residue of nonamer peptides in their binding to HLA-B* 5101 mole-
involvement. International Journal of Clinical Practice, 59, 777–81. cules. Immunogenetics, 46, 245–8.
Oguz, A., Serdaroglu, S., Tuzun, Y., Erdogan, N., Yazici, H., and Savaskan, H. Salvarani, C., Pipitone, N., Catanoso, M.G., et  al. (2007). Epidemiology
(1992). Acute febrile neutrophilic dermatosis associated with Behçet’s and clinical course of Behçet’s disease in the Reggio Emilia area of
syndrome. International Journal of Dermatology, 31, 645–6. Northern Italy: a seventeen-year population-based study. Arthritis and
Ohno, E., Ohtsuka, E., Watanabe, K., et al. (1997). Behçet’s disease associ- Rheumatism, 57, 171–8.
ated with myelodysplastic syndromes. A case report and a review of the Saruhan-Direskeneli, G., Uyar, F.A., Cefle, A., et al. (2004). Expression of KIR
literature. Cancer, 79, 262–8. and C-type lectin receptors in Behçet’s disease. Rheumatology (Oxford),
Ohno, S., Nakamura, S., Hori, S., et al. (2004). Efficacy, safety, and pharma- 43, 423–7.
cokinetics of multiple administration of infliximab in Behçet’s disease Sawalha, A.H., Hughes, T., Nadig, A., et al. (2011). A putative functional vari-
with refractory uveoretinitis. Journal of Rheumatology, 31, 1362–8. ant within the UBAC2 gene is associated with increased risk of Behçet’s
Okada, A.A., Goto, H., Ohno, S., and Mochizuki, M. (2012). Ocular Behçet’s disease. Arthritisand Rheumatism, 63, 3607–12.
Disease Research Group Of Japan. Multicenter study of infliximab for Saylan, T. (1997). Life story of Dr Hulusi Behçet. Yonsei Medical Journal, 38,
refractory uveoretinitis in Behçet disease. Archives of Ophthalmology, 327–32.
130, 592–8. Saylan, T., Mat, C., Fresko, I., and Melikoglu, M. (1999). Behçet’s disease in
Olivieri, I., Leccese, P., D’Angelo, S., et  al. (2011). Efficacy of adalimumab the Middle East. Clinics in Dermatology, 17, 209–23.
in patients with Behçet’s disease unsuccessfully treated with infliximab. Schreiber, B.E., Noor, N., Juli, C.F., and Haskard, D.O. (2011). Resolution of
Clinical and Experimental Rheumatology, 29, S54–7. Behçet’s syndrome associated pulmonary arterial aneurysms with inf-
Olivieri, I., Salvarini, C., and Cantini, F. (1997). Is Behçet’s disease part of the liximab. Seminars in Arthritis and Rheumatism, 41, 482–7.
spondyloartritis complex? Journal of Rheumatology, 34, 1870–1. Serdaroglu, P. (1998). Behçet’s disease and nervous system. Journal of
Orem, A., Vanizor, B., Cimsir, G., Kiran, E., Deger, O., and Malkoc, M. Neurology, 245, 197–205.
(1999). Decreased nitric oxide production in patients with Behçet’s dis- Serdaroglu, P., Yazici, H., Ozdemir, C., Yurdakul, S., Bahar, S., and Aktin,
ease. Dermatology, 198, 33–6. E. (1989). Neurologic involvement in Behçet’s syndrome. A prospective
Orloff, L.A. and Orloff, M.J. (1999). Budd-Chiari syndrome caused by study. Archives of Neurology, 46, 265–69.
Behçet’s disease: treatment by side-to-side portocaval shunt. Journal of Seyahi, E., Baskurt, M., Melikoglu, M., et al. (2011). The estimated pulmo-
the American College of Surgeons, 188, 396–407. nary artery pressure can be elevated in Behçet’s syndrome. Respiratory
Ozarmagan, G., Saylan, T., Azizlerli, G., Ovul, C., and Aksungur, V.L. Medicine, 105, 1739–47.
(1991). Re-evaluation of the pathergy test in Behçet’s disease. Acta Seyahi, E., Hamuryudan, V., Hatemi, G., et al. (2007). Infliximab in the treat-
Dermato-Venereologica, 71, 75–6. ment of hepatic vein thrombosis (Budd-Chiari syndrome) in three
Ozbakir, F., Yazici, H., Mat, C., Tuzun, Y., Yurdakul, S., and Yilmazer, S. (1987). patients with Behcet’s syndrome. Rheumatology (Oxford), 46, 1213–4.
HLA antigens in reccurrent oral ulcer:  evidence against a common Seyahi, E., Melikoglu, M., Akman, C., et  al. (2012). Pulmonary artery
disease spectrum with Behçet’s syndrome. Clinical and Experimental involvement and associated lung disease in Behçet disease: a series of 47
Rheumatology, 5, 263–5. patients. Medicine (Baltimore), 91, 35–48.
Ozen, S., Karaaslan, Y., Ozdemir, O., et  al. (1998). Prevalence of juvenile Seyahi, E. and Ozdogan, H. (2010). Juvenile Behçet’s syndrome. In Behçet’s
chronic arthritis and familial Mediterranean fever in Turkey:  a field syndrome, 1st edn (ed. Y. Yazici and H .Yazici), pp. 205–14. Springer,
study. Journal of Rheumatology, 25, 2445–9. New York.
Ozyazgan, Y., Yurdakul, S., Yazici, H., et  al. (1992). Low dose cylosporin Seyahi, E., Tahir Turanlı, E., Mangan, M.S., et al. (2010). The prevalence of
A  versus pulsed cyclophosphamide in Behçet’s syndrome:  a single Behçet’s syndrome, familial Mediterranean fever, HLA B51 and MEVF
masked trial. British Journal of Ophthalmology, 4, 241–3. gene mutations among ethnic Armenians living in Istanbul, Turkey.
Pazarli, H., Ozyazgan, Y., Bahcecioglu, H., et al. (1986). Ocular involvement Clinical and Experimental Rheumatology, 28, S67–75.
in Behçet’s syndrome in Turkey. In Behçet’s disease, Vol. 103 (ed. T. Seyahi, E. and Yazici, H. (2007). Atherosclerosis in Behçet’s syndrome.
Lehner and C.G. Barnes), pp. 267–8. Royal Society of Medicine Services, Clinical and Experimental Rheumatology, 25, S1–5.
London. Seyahi, E. and Yurdakul, S. (2011). Behçet’s syndrome and thrombo-
Poon, W., Verity, D.H., Larkin, G.L., Graham, E.M., and Stanford, M.R. sis. Mediterranean Journal of Hematology and Infectious Diseases, 3,
(2003). Behçet’s disease in patients of west African and Afro-Caribbean e2011026.
origin. British Journal of Ophthalmology, 87, 876–8. Sfikakis, P.P., Kaklamanis, P.H., Elezoglou, A., et al. (2004). Infliximab for recur-
Remmer, E.F., Cosan, F., Kirino, Y. et  al. (2010). Genome-wide asso- rent, sight-threatening ocular inflammation in Adamantiades-Behçet
ciation study identifies variants in the MHC class  I, IL10 and disease. Annals of Internal Medicine, 140, 404–6.
IL-23R-IL12RB2 regions associated with Behçet’s disease. Nature Shapiro, L.S., Farrell, J., Haghighi, A.B. (2012). Tocilizumab treatment
Genetics, 42, 698–702. for neuro-Behcet's disease, the first report. Clinical Neurology and
Rodriguez, A., Calonge, M., Pedroza-Serez, M., et al. (1996). Referral pat- Neurosurgery, 114, 297–8.
terns of uveitis in a tertiary eye care center. Archives of Ophthalmology, Sharquie, K.E., Najim, R.A., and Abu-Raghif, A.R. (2002). Dapsone in
114, 593–9. Behçet’s disease: a double-blind, placebo-controlled, cross-over study.
Saadoun, D., Asli, B., Wechsler, B., et  al. (2012). Long-term outcome of Journal of Dermatology, 29, 267–79.
arterial lesions in Behçet disease:  a series of 101 patients. Medicine Shi, H.-L. and Huang, Z.-J. (1993). Study on cutaneous lesions in Behçet’s dis-
(Baltimore), 91, 18–24. ease and meanings of relative laboratory parameters. In Behçet’s Disease.
Saadoun, D., Wechsler, B., Desseaux, K., et al. (2010a). Mortality in Behçet’s Proceedings of the 6th International Conference on Behçet’s Disease (ed. F.
disease. Arthritis and Rheumatism, 62, 2806–12. Godeau and B. Weschler), p. 325. Excerpta Medica, Amsterdam.
CHAPTER 34 behçet’s syndrome: pathogenesis, clinical manifestations, and treatment 489

Shimizu, T., Ehrlich, G.E., Inaba, G., and Hayashi, K. (1979). Behçet’s disease. Tunc, R., Uluhan, A., Melikoglu, M., Ozyazgan, Y., Ozdogan, H., and Yazici,
Seminars in Arthritis and Rheumatism, 8, 223–60. H. (2001). A reassessment of the International Study Group criteria
Shohat-Zabarski, R., Kalderon, S., Klein, T., and Weinberger, A. (1992). Close for the diagnosis (classification) of Behçet’s syndrome. Clinical and
association of HLA–B5a in persons with recurrent aphtous stomatitis. Experimental Rheumatology, 19 (Suppl. 24), S45–7.
Oral Surgery Oral Medicine and Oral Pathology, 74, 455–8. Turkstra, F., van Vugt, R.M., Dijkmans, B.A.C., Yazici, Y., and Yazici, H.
Siva, A., Altintas, A., and Saip, S. (2004). Behçet’s syndrome and the nervous (2012). Results of a questionnaire on the treatment of patients with
system. Current Opinion in Neurology, 17, 347–57. Behçet’s syndrome: a trend for more intensive treatment. Clinical and
Siva, A. and Saip, S. (2009). The spectrum of nervous system involvement in Experimental Rheumatology, 30 (Suppl. 72), S10–3.
Behçet’s syndrome and its differential diagnosis. Journal of Neurology, Tuzun, B., Tuzun, Y., Yurdakul, S., Hamuryudan, V., Yazici, H., and Ozyazgan,
256, 513–29. Y. (1999). Neutrophil chemotaxis in Behçet’s syndrome. Annals of the
Sloper, C.M., Powell, R.J., and Dua, H.S. (1999). Tacrolimus (FK 506) in the Rheumatic Diseases, 58, 658.
treatment of posterior uveitis refractory to cylosporine. Opthalmology, Tuzun, H., Hamuryudan, V., Yildirim, S., et al. (1996). Surgical therapy of
106, 723–8. pulmonary arterial aneurysms in Behçet’s syndrome. Annals of Thoracic
Soy, M., Erken, E., Konca, K., and Ozbek, S. (2000). Smoking and Behçet’s Surgery, 61, 733–5.
disease. Clinical Rheumatology, 19, 508–9. Tuzun, H., Besirli, K., and Sayin, A. (1997). Management of aneurysms in
Soysal, M., Denizci, U., Alhan, S., et al. (1990). Splenic size in Behçet’s syn- Behçet’s syndrome: an analysis of 24 patients. Surgery, 121, 150–6.
drome. British Journal of Rheumatology, 29, 497–8. Tuzun, H., Seyahi, E., Arslan, C., Hamuryudan, V., Besirli, K., and Yazici,
Stamatoyannopoulos, J.A. (2012). What does our genome encode? Genome H. (2012). Management and prognosis of nonpulmonary large arterial
Research, 22, 1602–11. disease in patients with Behçet disease. Journal of Vascular Surgery, 55,
Stanford, M., Whittall, T., Bergmeier, L.A., et al. (2004). Oral tolerization with 157–63.
peptide 336-351 linked to cholera toxin B subunit in preventing relapses Tuzun, Y., Altac, M., Yazici, H., et al. (1980). Nonspecific skin hyperreactivity
of uveitis in Behçet’s disease. Clinical and Experimental Immunology, in Behçet’s disease. Haematologica, 65, 395–8.
137, 201–8. Ugurlu, S., Seyahi, E., and Yazici, H. (2008). Prevalence of angina, myocardial
Sugioka, Y., Ohe, H., and Tanaka, T. (1966). Two cases of hip joint defor- infarction and intermittent claudication assessed by Rose Questionnaire
mation associated with Behçet’s syndrome. Orthopedic Surgery and among patients with Behcet’s syndrome. Rheumatology (Oxford),
Traumatology, 15, 131. 47, 472–5.
Takeno, M., Kariyone, A., Yamashita, N., et al. (1995). Excessive function of Ugurlu, S., Ucar, D., Seyahi, E., Hatemi, G., and Yurdakul, S. (2012).
peripheral blood neutrophils from patients with Behçet’s disease and Canakinumab in a patient with juvenile Behcet’s syndrome with refrac-
from HLAB51 transgenic mice. Arthritis and Rheumatism, 38, 426–33. tory eye disease. Annals of the Rheumatic Diseases, 71, 1589–91.
Takeuchi, A., Haraoka, H., and Hashimoto, T. (1989). Increased serum alka- Uluduz, D., Kürtüncü, M., Yapıcı, Z., et al. (2011). Clinical characteristics of
line phosphatase activity in Behçet’s disease. Clinical and Experimental pediatric-onset neuro-Behçet disease. Neurology, 77, 1900–5.
Rheumatology, 7, 19–21. Ural, A.U., Yalcin, A., Beyan, C., Isimer, A., and Bayhan, H. (1994).
Takeuchi, A., Kobayashi, K., Mori, M., and Mizushima, Y. (1981).The mecha- Plasma endothelin-1 concentrations in patients with Behçet’s disease.
nism of hyperchemotaxis in Behçet’s disease. Journal of Rheumatology, Scandinavian Journal of Rheumatology, 23, 322–5.
8, 40–4. Uziel, Y., Brik, R., Padeh, S., et al. (1998). Juvenile Behçet’s disease in Israel.
Tasci, B., Direskeneli, H., Serdaroglu, P., Akman-Demir, G., Eraksoy, M., The Pediatric Rheumatology Study Group of Israel. Clinical and
and Saruhan-Direskeneli, G. (1998). Humoral immune response to Experimental Rheumatology, 16, 502–5.
mycobacterial heat shock protein (hsp)65 in the cerebrospinal fluid Yavuz, S., Fresko, I., Hamuryudan, V., Yurdakul, S., and Yazici, H. (1998).
of neuro-Behçet patients. Clinical and Experimental Immunology, Fibromyalgia in Behçet’s syndrome. Journal of Rheumatology, 25,
113, 100–4. 2219–20.
Tayer-Shifman, O.E., Seyahi, E., Nowatzky, J., and Ben-Chetrit, E. (2012). Yazici, H. (1997). The place of Behçet’s syndrome among the autoimmune
Major vessel thrombosis in Behçet’s disease: the dilemma of anticoagu- diseases. International Review of Immunology, 14, 1–10.
lant therapy—the approach of rheumatologists from different countries. Yazici, H., Chamberlain, M.A., Tuzun, Y., Yurdakul, S., and Muftuoglu,
Clinical and Experimental Rheumatology, 30, 735–40. A. (1984b). A comparative study of the pathergy among Turkish and
Tokay, S., Direskeneli, H., Yurdakul, S., and Akoglu, T. (2001). Anticardiolipin British patients with Behçet’s disease. Annals of the Rheumatic Diseases,
antibodies in Behçet’s disease: a reassessment. Rheumatology (Oxford), 43, 74–5.
40, 192–5. Yazici, H. and Fresko, I. (2005). Behçet’s disease and other autoinflammatory
Tolosa-Vilella, C., Capela, C.A., Monteagudo-Jiménez, M., and Marí-Alfonso, conditions: what’s in a name? Clinical and Experimental Rheumatology,
B. (2011). Infliximab for life-threatening pulmonary artery aneurysms in 23 (Suppl. 38), S1–2.
Behçet’s disease. A case report. Clinical and Experimental Rheumatology, Yazici, H., Mat, C., Deniz, S., et al. (1987). Sebum production is increased in
29, S94–5. Behçet’s syndrome and even more so in rheumatoid arthritis. Clinical
Tugal-Tutkun, I., Mudun, A., Urgancioglu, M., et al. (2005). Efficacy of inflixi- and Experimental Rheumatology, 5, 371–4.
mab in the treatment of uveitis that is resistant to treatment with the com- Yazici, H., Pazarli, H., Barnes, C.G., et al. (1990). A controlled trial of aza-
bination of azathioprine, cyclosporine, and glucocorticoids in Behçet’s thioprine in Behçet’s syndrome. New England Journal of Medicine,
disease: an open-label trial. Arthritis and Rheumatism, 52, 2478–84. 322, 281–5.
Tugal-Tutkun, I., Onal, S., Altan-Yaycioglu, R., Huseyin Altunbas, H., and Yazici, H., Tuzlaci, M., and Yurdakul, S. (1981a). A controlled survey of sac-
Urgancioglu, M. (2004). Uveitis in Behçet disease:  an analysis of 880 roiliitis in Behçet’s disease. Annals of the Rheumatic Disease, 40, 558–9.
patients. American Journal of Ophthalmology, 138, 373–80. Yazici, H., Tuzuner, N., Tuzun, Y., and Yurdakul, S. (1981b). Localized myosi-
Tunc, R., Keyman, E., Melikoglu, M., Fresko, I., and Yazici, H. (2002). Target tis in Behçet’s disease. Arthritis and Rheumatism, 24, 636.
organ associations in Turkish patients with Behçet’s disease: a cross sec- Yazici, H., Tuzun, Y., Pazarli, H., et al. (1984a). Influence of age of onset and
tional study by exploratory factor analysis. Journal of Rheumatology, 29, patient’s sex on prevalence and severity of manifestations of Behçet’s
2393–6. syndrome. Annals of the Rheumatic Disease, 43, 783–9.
Tunc, R., Saip, S., Siva, A., and Yazici, H. (2004). Cerebral venous thrombosis Yazici, H., Tuzun, Y., Tanman, A.B., et al. (1985). Male patients with Behçet’s
is associated with major vessel disease in Behçet’s syndrome. Annals of syndrome have stronger pathergy reactions. Clinical and Experimental
the Rheumatic Diseases, 63, 1693–4. Rheumatology, 3, 137–41.
490 SECTION 5 vasculitic diseases and syndromes and related disorders

Yazici, H., Tuzun, Y., Pazarli, H., et al. (1986). Sex factor and Behçet’s syn- Yurdakul, S., Mat, C., Tuzun, Y., et al. (2001). A double blind trial of colchi-
drome. In Recent advances in Behcet’s disease, Vol. 013 (ed. T. Lehner and cine in Behçet’s syndrome. Arthritis and Rheumatism, 44, 2686–92.
C.G. Barnes), pp. 205–6. Royal Society of Medicine Services, London. Yurdakul, S., Tuzuner, N., Yurdakul, I., Hamuryudan, V., and Yazici, H.
Yazici, H., Ugurlu, S., and Seyahi, E. (2012). Behçet’s syndrome: is it one con- (1990). Amyloidosis in Behçet’s syndrome. Arthritis and Rheumatism,
dition? Clinical Reviews in Allergy and Immunology, 43, 275–80. 33, 1586–9.
Yazici, Y. and Yazici, H. (eds) (2010). Behçet’s syndrome, 1st edn, pp. 115–34. Yurdakul, S., Tuzuner, N., Yurdakul, I., Hamuryudan, V., and Yazici, H.
Springer, New York. (1996). Gastrointestinal involvement in Behçet’s syndrome: a controlled
Yesilot, N., Bahar, S., Yilmazer, S., et al. (2009). Cerebral venous thrombosis study. Annals of the Rheumatic Diseases, 55, 208–10.
in Behçet’s disease compared to those associated with other etiologies. Yurdakul, S., Yazici, H., Tuzun, Y., et al. (1983). The arthritis of Behçet’s dis-
Journal of Neurology, 256, 1134–42. ease: a prospective study. Annals of the Rheumatic Disease, 42, 505–15.
Yesilot, N., Shehu, M., Oktem-Tanor, O., et  al. (2006). Silent neuro- Zakaria, M., Sasaki, S., Matsui, Y., Shiiya, N., Murashita, T., and Yasuda,
logical involvement in Behçet’s disease. Clinical and Experimental K. (1998). Successful repair of cardio-Behçet’s disease with aortic and
Rheumatology, 24, S65–S70. mitral regurgitation. Journal of Cardiovascular Surgery, 39, 803–5.
Yurdakul, S., Gunaydin, I., Tuzun, Y., et al. (1988). The prevalence of Behçet’s Zierhut, M., Mizuki, N., Ohno, S., et al. (2003). Immunology and functional
syndrome in a rural area in northern Turkey. Journal of Rheumatology, genomics of Behçet’s disease. Cellular and Molecular Life Sciences, 60,
15, 820–2. 1903–22.
Yurdakul, S., Hekim, N., Soysal, T., et  al. (2005). Fibrinolytic activity Zouboulis, C.C., Djawari, D., Kirch, D., et al. (1996). Adamantiades-Behçet’s
and d-dimer levels in Behçet’s syndrome. Clinical and Experimental disease in Germany:  Data of the German Registry in 1996. Revue de
Rheumatology, 23 (Suppl. 38), S53–8. Rheumatism (English Edition), 63, 538.
CHAPTER 35

Juvenile Behçet’s syndrome


Gülsevim Azizlerli and Rifkiye Sarica-Kucukoglu

Introduction Although it has been reported that the disease begins between
age 4 and 16, the most frequent age of onset for JBS lies between 9
Behçet’s syndrome (BS) is a chronic, relapsing, multisystemic and 16 years.
inflammatory disease of unknown aetiology. It was first described There was no difference in the incidence of oral and genital
in 1937 by Hulusi Behçet as a trisymptom complex comprised ulcerations between adult BS and JBS, but erythema nodosum and
of oral and genital ulcerations and iridocyclitis with hypopyon pseudofolliculitis were more frequent in JBS. It is noteworthy that
(Behçet 1937). BS has been reported all over the world but with non-aphthous ulceration occurred in 3% of adult BS patients as
a distinct geographical distribution. The prevalence of BS varies opposed to 1.8% in the JBS group. While eye involvement and deep
both between countries and between different regions of the same vein thrombosis were slightly more common in the JBS patients,
country. These regional differences are probably due to ethnic as this difference was not significant. The incidence of arthritis,
well as geographical factors. The prevalence of BS is much higher in epididymitis, and neurological signs were nearly the same in JBS
countries along the ancient Silk Road, extending from Japan to the patients as in adults.
Mediterranean and Middle-Eastern countries, including Turkey, To evaluate factors affecting the course of the disease, the patients
than in northern Europe and the USA (Sakane et al. 1999; Verity were divided into two groups, one showing severe disease with sys-
et al. 1999). Field surveys in Turkey have found prevalence rates in temic involvement, the other with mild disease mostly limited to
the range of 8–42/10 000 population, which identified Turkey as the mucocutaneous involvement. The severe disease group consisted
country with the highest rate of BS in the world (Demirhindi et al. of 35 patients (25 male,10 female) and the mild disease group 31
1981; Yurdakul et al. 1988; Idil et al. 2002; Azizlerli et al. 2003). male and 43 female. There was more systemic involvement (severe
The disease most frequently affects adults and is rare in children. disease) among males (71.4%) in both JBS and adult BS than in
The term ‘juvenile Behçet’s syndrome’ (JBS) is applicable to those females (38.6%). In our 109 JBS patients, a mild course was noted in
patients who fulfil the criteria for diagnosis before the age of 16. 67.8%, which was similar to that of our adult patient group of 1127
The course of the disease is influenced by sex and age in adult BS individuals (Sarica et al. 1996).
(Lang et al. 1990) and the finding that it is more aggressive in young Sixty of the 109 JBS patients were followed for between 5 and
males who develop the disease before age 25 has led to increased 25 years. We grouped the long-term follow-up patients into 5-year
interest in JBS (Sarica et al. 1996; Yazici et al. 1984). Among 3471 BS intervals and analysed the symptoms as mucocutaneous, ocular,
patients diagnosed according to International Study Group Criteria arthritic, vascular, and neurological (Table 35.2). Similar to adult
and followed at the Department of Dermatology BS Outpatient BS, JBS is observed to follow a predominantly mucocutaneous
Clinic of the University of Istanbul Faculty of Medicine, 109 had course (67.8%) during the first 5-year period. Most ocular involve-
JBS (International Study Group for Behçet’s Disease 1990). The ment was also apparent within the first 5 years of the disease and
prevalence of JBS was 3.14% in this series in comparison to 1.6% was more common in males (41.07%) than in females (28.3%).
(42/2519) in a Japanese study (Maeda and Nakae 1977), and 3% Only three of the 52 patients had new ocular disease after the first
(67/2175) in a study from Iran (Shafaie et al. 1993). In subsequent 5-year period. During the entire study, three JBS patients with eye
reports from Turkey, the prevalence was given as 2% (44/1921) involvement developed blindness.
(Özdoğan 1996; Yurdakul et al. 1993) and 3.3% (110/3382) (Atmaca In patients with JBD, the incidence of vascular involvement
et al. 2011). Kone-Paut et al. identified 17 JBS patients diagnosed in was 13.76% (12 male, three female). The most common vascular
France (Kone-Paut and Bernard 1993) and an Israeli study reported abnormality during the first 5  years was venous system disease.
19 JBS patients and 34 adult patients (Krause et al. 1999). Based on Deep vein thrombosis (DVT) was seen in 13 patients whereas
these studies, the prevalence of JBS is estimated to be 1.6–3% of BS pulmonary arterial aneurysm was detected in only two. Arthritis
patients. was diagnosed in 19 males and 11 females (27.5%). In summary,
The clinical manifestations of the 109 JBS patients from our arthritis, ocular, and vascular involvements were mainly observed
clinic are given in Table 35.1. The initial symptom was aphthous among male patients during the first 5 years of the disease. Ocular
ulceration in 90.8% of patients. The second lesion develops within and vascular lesions were more common in JBS than adult BS and
a mean interval of 2.57 ± 2.54 years, and most common is genital the incidence of arthritis was the same within the first 5 years.
ulceration. Systemic involvement ensues with a mean of 3.52 ± 4.72 Fifty-two of the 109 patients were followed during the second
(range 0–22) years after the detection of the initial symptom. 5-year period. In this period all the patients had mucocutaneous
492 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 35.1 Clinical features of JBS and adult Behçet’s syndrome in lesions. Organ involvement was observed in six patients (three
University of Istanbul, Istanbul Faculty of Medicine, Dermatology Clinic male, three female) and included eye (two males, one with loss
‘Behçet’s Disease Study Group’ of visual acuity), arthritis (one male, one female), vascular (two
males), neurological (one male and one female), and gastrointes-
Clinical feature Adult BS (n = 1127) Juvenile BS (n = 109) tinal (one female) involvement. Eye, vascular, arthritic, and neu-
% of patients % of patients rological involvement were present in one individual. The rest of
Male/female 610/517 56/53 the patients (88.46 %) had only a mild course with mucocutaneous
lesions.
Aphthous ulceration 100 100
In the third 5-year follow-up period, all the patients had mild
Genital ulceration 87.4 88.7 mucocutaneous involvement except one patient who showed severe
Pseudofolliculitis 58.1 64.2 mucocutaneous exacerbation; 24% had organ involvement (eye in
one male and one female patient, vascular involvement in three
Erythema nodosum 45.9 50.4
males, and neurological involvement in one male). In the fourth
Extragenital ulceration 3.1 1.8 5-year period, there were data for only 11 patients; only one patient
Eye involvement 29.1 37.6 had pulmonary arteritis besides mucocutaneous involvement. In
the fifth 5-year period, there were data for only six patients, includ-
Arthritis 28.9 28.4
ing one male patient with eye involvement and loss of visual acuity.
Superficial thrombophlebitis 15.8 10 In the sixth 5-year period, there were only three patients, who were
Deep vein thrombosis 3.9 7.3 clinically inactive without any organ involvement.
Based on these findings, the overall characteristics of JBS can be
Pulmonary involvement 2.1 1.8
summarized as in Table 35.3.
Epididymitis 2.5 Our long-term follow-up data suggest that JBS follows a clini-
Neurological involvement 3.0 3.6 cal course with exacerbations and remissions, and with a predomi-
nantly mucocutaneous involvement similar to the clinical course of
GI symptoms 0.8 0.9
the adult type.
Pathergy positivity 74.6 70.1
HLA B5 positivity 82.4 60
Clinical manifestations
Family history 6.1 13.7
Mucocutaneous lesions
Age of onset 27.2 ± 7.5 years 12.23 ± 2.879 years
(range 17–60) (range 9–16) The initial lesion(s) in JBS is oral aphthae, just as in the adult. Minor
aphthae are more frequent than major or herpetiform types. There
Interval between first and 2.2 ± 3.8 years 2.57 ± 2.54 years
is a time interval between the first and the second mucocutaneous
second symptom
lesion, and in our study the mean interval was 2.57 ± 2.54 years,

Table 35.2 Classification of 109 JBS cases in long-term follow-up according to clinical features and years

Year Year Year Year Year Year


0–5 5–10 10–15 15–20 20–25 25+
(n=109) (n=52) (n=26) (n=11) (n=6) (n=3)

F=53 M=56 F=28 M=24 F=15 M=11 F=6 M=5 F=2 M=4 F=2 M=1
Mucocutaneous 53 56 + + 1+ + + + + + + +
findings (aphthous
ulceration and other
skin findings)
Eye involvement 15 23 2(1a) 1 1a
Loss of visual acuity 1a 1a 1a
Arthritis 11 19 1 1a
Vascular involvement 2 11 2(1a) 3(1a) 2
Veins 1 1 1
Arteries pulmonary pulmonary pulmonary
CNS involvement 1 1 1a 1
GIS 1
a Indicates two or more different sites of involvement in the same patient.

+ All of the patients had mucocutaneous involvement though all showed a mild course; one had severe mucocutaneous involvement in the 13th year.
CNS, central nervous system; GIS, gastrointestinal.
CHAPTER 35 juvenile behçet’s syndrome 493

Table 35.3 The overall characteristic features of juvenile Behçet’s contrast, the frequency of arthritis in JBS was similar to adult BS
syndrome in our study. The incidence of arthritis in BS differs in different
geographical regions. For example, the rate was 21% in Tunisia,
JBS has generally a benign course just as in the adult type (67.8%). 69% in France, and 13% in Iran (Hamza et al. 1993; Kone-Paut and
Ocular involvement is most prominent within the first 5 years of the disease Bernard 1993; Shafaie et al. 1993).
in all of the patients with JBS.
Vascular involvement
Erythema nodosum and pseudofolliculitis are more frequent in JBS.
BS disease of veins, arteries or both, is a serious prognostic sign,
Familial involvement is more common in juvenile-onset BS.
being one of the main causes of death in BS. Peripheral veins of
Vascular involvement is more frequently seen in JBS compared to adults and is the calf, superior and inferior vena cava, and hepatic veins may be
prominent in males. Arterial involvement occurs only rarely. involved and more than one vascular territory may be affected in the
same patient. DVT is sometimes the first manifestation of BS but
is very rare in children (Leiba et al. 2004). In our series, DVT was
which was not significantly different from the time interval seen found in eight patients (7.3%), seven of whom were male. In these
in adult BS (2.2  ± 3.8). Genital ulcerations are the second most patients, as well as those from other sites in Turkey and Tunisia,
frequent lesion in JBS, and these lesions have a reportedly female vascular involvement in JBS almost exclusively affected males
predilection. Both Shafaie et al. (1993) and Krause et al. (1999) have (Hamza 1993; Yurdakul et al. 1993). Superficial thrombophlebitis is
reported that aphthae and genital ulcerations were less frequent in slightly less common in JBS (10%) compared with adult BS (15.8%).
JBS compared with adult BS. Yurdakul et al. (1993) found that the Moreover, arterial lesions, which usually represent late-stage com-
frequency of genital ulceration is lower in the prepubertal age and plications, were rarely observed in our JBS patients (Table 35.2).
increases after puberty. Our data, and the study by Kone-Paut et al. In summary, vascular complications are less frequent in JBS than
(1998), demonstrate no difference between JBS and adult BS in the in adults, more often occur in males, and can be life-threatening
frequency of oral–genital ulcerations. Scar tissue formation in heal- (Kone-Paut et al. 1998).
ing of the genital ulceration is typical.
Gastrointestinal involvement
Other skin lesions
Gastrointestinal symptoms in JBS, typically abdominal pain and
Pseudofolliculitis can be encountered in JBS, with acneiform gastric upset (Kone-Paut et al. 1998), are rare and minor in chil-
lesions developing mostly with puberty. Both pseudofolliculitis dren. More major problems or lesions such as severe ileocolitis
and erythema nodosum occur more often in JBS patients than haves not been reported in JBS.
in the adult BS. Extragenital aphthous ulcerations are less com-
mon in JBS, and may heal with scar formation (Azizlerli et  al. Neurological involvement
1992). These lesions are seen in 3% of BS patients and 1.8% in JBS Central nervous system disease is another leading cause of mortal-
patients. In addition, there was no statistical difference in the fre- ity in BS. Headache and symptoms attributable to raised intracra-
quency of positive pathergy test between JBS and adult BS, sexes, nial pressure are typical. The two major lesions are parenchymal
or ages. involvement and cerebral venous sinus thrombosis. The frequency
of neurological involvement in adult BS differs in different geo-
Eye involvement
graphical regions (O’Neill et al. 1993); it has been reported to vary
Eye disease is one of the most serious afflictions of BS; in the adult between 10 and 25% in northern America and Europe, but only
BS it is more typically seen in males (Sarica et al. 1996). Its fre- around 3–5% in Turkey. Similar differences are noted in JBS; for
quency is higher in the JBS group than in adults, but the differ- example the incidence of neurological involvement was reported to
ence is not statistically significant. The frequency of uveitis in JBS be 18% in France but 6–7% in Turkey and Tunisia (Table 35.4). In
is 14% in Tunisia, 47% in France, 31% in Iran, 47.4% in Israel, our JBS experience, there was no difference between sexes in terms
and 37.6% in our clinics (Table 35.4). JBS uveitis was more com- of neurological involvement, but the frequency of neurological
mon among males. Bilateral panuveitis with retinal vasculitis was involvement was slightly higher in the JBS group (3.6%) compared
the most common form of ocular involvement. These findings with adult BS (3%).
are similar to those seen in adult BS (Tugal-Tutkun et al. 2003).
Matsuo and Itami have described a female patient with onset at
a young age showing skin lesions, oral and genital ulcerations, Immunogenetics
and eye involvement. This involvement may have been a single Familial aggregation has been well documented in BS, and the sib-
incident and was not recorded at subsequent recurrence (Matsuo ling recurrence risk ratio for BS was found to be between 11.4 and
and Itami 2005). 52.5 in Turkey (Gul et al. 2000). Analysis of a small group of mul-
ticase families did not show any particular Mendelian inheritance
Arthritis pattern (Bird 1986). The HLA alleles appear to be one of the most
Small joints of the hands and feet, knees, and elbows are most prominent genomic regions associated with disease tendency in BS
frequently involved in BS. Arthritis is usually monoarticular, and (Gul et al. 2001). A close association of BS with the HLA B51 allele
articular destruction is uncommon. There is no predilection for in the Class 1 MHC region was well defined. Remmers et al. (2010)
males or females. Arthralgias were slightly more common in JBS performed a genome-wide association study in 1215 individuals
than in adults BS in the Israeli series. The rate of joint involvement with BS from Turkey and identified IL-10, IL23R, and IL23RB2 as
was 47.4% in JBS versus 17.6%, in adults (Krause et al. 1999). In disease susceptibility loci in addition to confirming the association
494 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 35.4 Clinical features and systemic involvement of juvenile BS patients from different centres

France Tunisia Iran Israel Turkey Turkey Turkey


Kone-Paut 1993 Hamza, Shafaie, Krause, Yurdakul, Atmaca, Azizlerli,
(n = 17) 1993 1993 1999 1993 2011 2012
(n = 14) (n = 67) (n = 19) (n = 44) (n = 110) (n = 109)
Oral ulcers 94 100 77 100 100 100 100
Genital ulcers 53 60 26 31.6 70 82.7 88.07
Skin lesions 92 86 61 89.5 – 76 –
Pseudofolliculitis – 71 – – 77 – 64.2
Erythema nodosum – 14 – 36.8 59 – 50.4
Eye involvement 47 14 31 47.4 48 30.9 37.6
Arthritis 69 21 13 31.6 34 22.7 28.4
CNS involvement 18 7 – 26.3 6 3.6 2.7
Vascular involvement 6 33 – 10.5 12 3.6 17.3
Pathergy 60 65 – 41.2 68 45.5 70.1
HLA B5 54 30 – 56.3 52 – 60
Family – – – 13.1

Numbers shown are percentages of patients. Dashes indicate findings that were not available.

of HLA-B51 with BS. They suggested that the disease-associated obtained using the transmission disequilibrium test. Arthritis and
IL10 variants could facilitate the development of BS via inducing a Rheumatism, 44, 239–40.
disruption in the control of the inflammatory response, resulting in Gul, A., Inanc, M., Ocal, L., Aral, O., and Konice, M. (2000). Familial aggregation
an inflammation-prone state. These findings may have a significant of Behçet’s disease in Turkey. Annals of the Rheumatic Diseases, 59, 622–5.
impact on the understanding of mechanisms as well as future treat- Hamza, M. (1993). Juvenile Behçet’s disease. In Behçet’s disease (ed. P. Godeau
and B. Wechsler), pp. 337–80. Elsevier, Amsterdam.
ment choices in BS.
İdil, A., Gürler, A., Boyvat, A., et al. (2002). The prevalence of Behçet’s dis-
ease above the age of 10 years. The results of a pilot study conducted at
Management the Park Primary Health Care Center in Ankara, Turkey. Ophthalmic
Epidemiology, 9, 325–31.
Colchicine is effective in the treatment of erythema nodosum, arthri- International Study Group for Behçet’s Disease (1990). Criteria for diagnosis
tis, and genital ulcerations. Local application of antiseptic solutions of Behçet’s disease. Lancet, 335, 1078–80.
and topical glucocorticoids, combined with periodontal care, reduces Kone-Paut, I. and Bernard, J.L. (1993). Behçet’s disease in children. A French
the frequency of aphthous lesions. Systemic glucocorticoids alone or nation wide survey. In Behcet’s disease (ed. P. Godeau and B. Wechsler),
combined with immunosuppressive drugs can also be used. pp. 385–9. Elsevier, Amsterdam.
Kone-Paut, I., Yurdakul, S., Bahabri, S.A., et al. (1998). Clinical features of
Behçet’s disease in children: An international collaborative study of 86
References cases. Journal of Pediatrics, 132, 721–5.
Atmaca, L., Bovyat, A., Yalçındağ, F.N., Atmaca-Sonmez, P., and Gurler, Krause, I., Uziel, Y., Guedj, D., et al. (1999). Childhood Behçet’s disease: clini-
A. (2011). Behcet’s disease in children. Ocular Immunology and cal features and comparison with adult-onset disease. Rheumatology, 38,
Inflammation, 19, 103–7. 457–62.
Azizlerli, G., Akdag Köse, A., Sarica, R., et al. (2003). Prevalence of Behçet’s Lang, B.A., Laxer, R.M., Thorner, P., Greenberg, M., and Silverman, E.D.
disease in Istanbul, Turkey. International Journal of Dermatology, (1990). Pediatric onset of Behçet’s syndrome with myositis. Case
42, 803–6. report and literature review illustrating unusual features. Arthritis and
Azizlerli, G., Özarmagan, G., Övül, C., Sarica, R., and Mustafa, S.O. (1992). A Rheumatism, 33, 418–25.
new kind of skin lesion in Behçet’s disease, extragenital ulcerations. Acta Leiba, M., Seligsohn, U., Sidi, Y., et  al. (2004). Thrombophilic factors are
Dermatologica Venerologica, 72, 286. not the leading cause of thrombosis in Behcet’s disease. Annals of the
Behçet, H. (1937). Über rezidivierende apthöse, durch ein Virus verursachte Rheumatic Diseases, 63, 1445–9.
Geschwüre am Mund, am Auge, und an dem Genitalien. Dermatologische Maeda, K. and Nakae, K. (1977). Recent epidemiological review on Behçet’s
Wochenschrift, 105, 1152–7. disease in Japan. Asian Medical Journal, 20, 568–82.
Bird, S.J.A. (1986). Genetic analysis of families of patients with Behçet’s syn- Matsuo, T. and Itami, M. (2005). Recurrent versus non-recurrent or no eye
drome: Data incompatible with autosomal recessive inheritance. Annals involvement Behçet’s disease. Ocular Immunology and Inflammation,
of the Rheumatic Diseases, 45, 265–8. 13, 73–7.
Demirhindi, O., Yazıcı, H., Binyıldız, P., et al. (1981). Silivri Fener Köyü ve yöres- O’Neill, T.W., Rigby, A.S., McHugh, S., Silman, A.J., and Barnes, C. on
inde Behçet Hastalığı sıklığı ve bu hastalığın toplum içinde taranmasında behalf of the International Study Group for Behçet’s Disease (1993).
kullanılabilecek bir yöntem. Cerrahpaşa Tıp Dergisi, 12, 509–14. Regional differences in clinical manifestations of Behçet’s disease. In
Gul, A., Hajeer, A.H., Worthington, J., Barrett, J.H., Ollier, W.E., and Silman, Behçet’s disease (ed. P. Godeau and B. Wechsler), pp. 159–63. Elsevier,
A.J. (2001). Evidence for linkage of the HLA–B locus in Behçet’s disease, Amsterdam.
CHAPTER 35 juvenile behçet’s syndrome 495

Özdoğan, H. (1996). Behçet’s syndrome in children. In Vasculitides (ed. B.M. Tugal-Tutkun, I. and Urgancıoglu, M. (2003). Childhood-onset uveitis
Ansell, P.A. Bacon, J.T. Lie, and H. Yazici), pp. 419–24. Chapman and in Behcet disease:  a descriptive study of 36 cases. American Journal
Hall, London. Ophthalmology, 136, 1114–9.
Remmers, E.F., Coşan, F., Kirino, Y., et al. (2010). Genome-wide association Verity, D.H., Marr, J.E., Ohno, S., Wallace, G.R., and Stanford, M.R. (1999).
study identifies variants in the MHC class 1, IL10, and IL 23R/IL 12RB2 Behcet’s disease, the Silk Road and HLA-B51: historical and geographi-
regions associated with Behçet’s disease. Nature Genetics, 42, 698–702. cal perspectives. Tissue Antigens, 54, 213–20.
Sakane, T., Takeno, M., Suzuki, N., and Inaba, G. (1999). Behçet’s disease. Yazici, H., Tüzün, Y., Pazarli, H., et al. (1984). Influence of age of onset and
New England Journal of Medicine, 341, 1284–91. patient’s sex on the prevalence and severity of manifestations of Behçet’s
Sarica, R., Azizlerli, G., Akdag Köse, A., et al. (1996). The course of disease activity syndrome. Annals of the Rheumatic Diseases, 83, 783–9.
among 1127 Turkish adult Behçet’s patient. In Behçet’s disease 7th Congress Yurdakul, S., Günaydın, I., Tüzün, Y., et al. (1988). The prevalence of Behcet’s syn-
on Behçet’s Disease Proceedings (ed. M. Hamza), pp. 157–9. Adhoua, Tunis. drome in a rural area in northern Turkey. Journal of Rheumatology, 15, 820–2.
Shafaie, N., Shahram, F., Davatchi, F., et al. (1993). Behçet’s disease in chil- Yurdakul, S., Özdogan, H., Kasapçopur, Ö., et  al. (1993). Behçet’s syn-
dren. In Behçet’s disease (eds. P. Godeau and B. Wechsler), pp. 385–9. drome with juvenile onset: Report of 44 patients. Clinical Experimental
Elsevier, Amsterdam. Rheumatology, 11, 71.
CHAPTER 36

Vasculitis of the central


nervous system
Rula A. Hajj-Ali, George F. Duna,
and Leonard H. Calabrese

Introduction was published in the mid to late 1980s; many of these lacked patho-
logical confirmation. In the early 1980s, a small series of PACNS
Vasculitis affecting the central nervous system (CNS) remains one successfully treated with a combination of cyclophosphamide and
of the most elusive forms of vascular inflammatory disease. Factors glucocorticoids increased the enthusiasm for diagnosing PACNS.
contributing to our relative lack of understanding of CNS vasculitis It became standard practice to treat all cases aggressively with this
include its rarity, the lack of an efficient non-invasive test, a paucity combination of drugs regardless of the method of diagnosis or clin-
of pathological material to study, and the absence of animal models ical picture (Cupps et al. 1983). In the mid-1990s, several investiga-
simulating the disease. Despite these limitations, the past 15 years tors noted that not all cases of PACNS, diagnosed by angiography,
have witnessed significant progress in our understanding of the appeared to have the inexorable and fatal course of those reported
clinical heterogeneity of these disorders and their subclassification earlier and they proposed that benign variants may exist (Calabrese
into clinical subsets. et al. 1993). As our understanding of PACNS progressed, it became
Vasculitis affecting the CNS can be classified into primary and very clear that these angiographically defined variants represent
secondary forms. Primary angiitis of the CNS (PACNS) is vasculi- a reversible cerebral angiopathy and not a true vasculitis (Singhal
tis confined to the CNS, including brain and spinal cord, and their 2004). In 2007, we and others (Calabrese et  al. 2007)  proposed
coverings. Secondary vasculitis of the CNS implies vascular inflam- the term ‘reversible cerebral vasoconstriction syndromes’ (RCVS)
mation of CNS as part of a larger process such as an infection, con- to encompass a clinicoradiological syndromes that closely mimic
nective tissue disease, or other systemic disorder. This chapter will PACNS. RCVS comprise a group of diverse conditions, all charac-
outline the epidemiological, clinical, diagnostic, and therapeutic terized by reversible, multifocal narrowing of the cerebral arteries;
aspects of both primary and secondary forms of CNS vasculitis. clinically, patients present with sudden (thunderclap), severe head-
aches with or without associated neurological deficits. Currently,
Primary angiitis of the CNS (PACNS) RCVS is considered one of the major mimics of PACNS and will
be discussed separately. Finally, variants of PACNS have been
Classification and diagnostic criteria described including those associated with cerebral amyloid, and
The modern era of PACNS began in the late 1950s with the elegant mass-like lesions, as well as others, which will be described in detail
descriptive report of a few patients with an isolated, progressive, (Calabrese et al. 1997).
fatal form of vasculitis affecting the CNS (Cravioto and Feigin In 1988, Calabrese and Mallek proposed three working criteria
1959). Upon pathological examination of these cases all were for the diagnosis of PACNS (Calabrese and Mallek 1988). Despite
found to be granulomatous in nature, leading to the alternative changes in our view of PACNS, particularly with the appreciation
term granulomatous angiitis of the CNS (GACNS). Following this of its broadening clinical spectrum and the increasing number of
early description, others were slow to be reported, with less than 40 mimics, these criteria still serve as a useful starting place for diag-
total cases by the mid 1980s (Calabrese and Mallek 1988). Recently, nosis (Calabrese 2009).
the disease has become more recognized and number of cases has The criteria are:
dramatically escalated with over 500 cases described world-wide 1. a history of a clinical finding of an acquired neurological defi-
by 2007. The increase in reporting is attributed to awareness and cit that remains unexplained after a thorough initial basic
recognition, and the application of advanced and aggressive neu- evaluation;
rodiagnostic approaches in the modern era. CNS vasculitis was
found to be one of the common misdiagnoses of patients with spo- 2. either classic (high probability) angiographic evidence or histo-
radic Creutzfeldt–Jakob disease, when a brain biopsy was used in pathological demonstration of angiitis within the CNS;
the work up of dementia (Paterson et al. 2012). With the advent of 3. no evidence of systemic vasculitis or any other condition to
cerebral angiography as a diagnostic tool for vasculitis, combined which the angiographic or pathological condition could be
with its spreading clinical application, a flurry of new case reports attributed (see Table 36.1).
498 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 36.1 Mimics of PACNS but clinically important distinct presentations (spinal cord, mass
lesion), warranting specialized clinical approaches to diagnosis or
Systemic vasculitides and inflammatory diseases treatment, which justifies their distinction. Finally, as our knowl-
Polyarteritis nodosa edge increases, new forms, such as lymphocytic vasculitis and amy-
Microscopic polyangiitis loid angiitis, are distinguished on pathological grounds.
Hypersensitivity vasculitis (cutaneous leukocytoclastic vasculitis) and
related disorders Clinical subsets of PACNS
Vasculitis of connective tissue diseases
Granulomatosis with polyangiitis (Wegener’s)
Granulomatous angiitis of the CNS
Eosinophilic granulomatosis with polyangiitis (Churg–Strauss syndrome) While there are no formal diagnostic criteria established for
Behçet’s syndrome GACNS, the essential features of this syndrome are outlined in
Lymphomatoid granulomatosis Table 36.2. Within the spectrum of PACNS, it is difficult to estimate
Cogan’s syndrome the percentage of patients who fulfil these criteria because they
Systemic lupus erythematosus have not been applied prospectively to any large series of patients.
Sjögren’s syndrome We estimate that no more than one-third of patients diagnosed
Sarcoidosis with PACNS would fulfil such criteria.
Infections The epidemiology of the disease is poorly understood, but
Viral it does appear to be a male-predominant disorder, occurring at
Bacterial virtually any age (Calabrese et al. 1997; Calabrese 1995). In 1997,
Fungal Younger and colleagues (Younger and Kass 1997)  reviewed the
Rickettsial cumulative literature on GACNS and analysed 136 reported cases
Neoplasms and one new case. Fifty-one of these patients had associated condi-
Angioimmunoproliferative disorders tions, including lymphoproliferative diseases, varicella zoster viral
Carcinomatous meningitis (VZV) infection, amyloidosis, sarcoidosis, or one of several other
Infiltrating glioma conditions. Whether such cases are equivalent to GACNS occur-
Malignant angioendotheliomatosisLymphoma ring de novo is unknown, but this analysis remains the basis of
Vasospastic disorders much of the clinical appreciation for this relatively rare disorder.
Reversible cerebral vasoconstriction syndrome (RCVS) These data emphasize the need to rule out secondary causes such
Phaeochromocytoma as malignancies and infections before attributing the pathology
Aneurysmal subarachnoid haemorrhage to PACNS.
Other vasculopathies and mimicking conditions The most common clinical manifestations in GACNS are head-
Fibromuscular dysplasia ache and mental changes. These features generally evolve over
Moya moya disease weeks to months and are followed by focal cerebral signs, including
Thrombotic thrombocytopenic purpura seizure, aphasia, hemiparesis, or tetraparesis, in the vast majority of
Sickle cell anaemia patients. Thus, the combination of focal and non-focal neurological
Neurofibromatosis dysfunction remains the hallmark of GACNS.
Cerebrovascular atherosclerosis The onset of GACNS may occasionally be acute, but is more com-
Demyelinating disease monly subacute. While fulminate presentations have been reported, an
Intracranial arterial dissection insidious onset is much more common. Neurological signs and symp-
Radiation vasculopathy
toms may predate the diagnosis by months or even years. Curiously,
Retinal vasculopathy with cerebral leukodystrophy (RVCL)
Emboli (subacute bacterial endocarditis, cardiac myxoma, paradoxical
emboli)
Acute posterior placoid pigment epitheliopathy and cerebral vasculitis Table 36.2 Key features of GACNS
Antiphospholipid antibody syndrome
Susac’s syndrome (encephalopathy, branch retinal artery occlusions and Clinical and laboratory features
sensorineural hearing loss) Variable onset but most frequently a prolonged prodrome of 3–6 or
MELAS (mitochondrial myopathy, encephalopathy, lactic acidosis, more months
and stroke-like episodes) Mixture of focal and non-focal neurological signs
CADASIL (cerebral autosomal dominant arteriopathy with subcortical CSF analysis abnormal in 90% (aseptic meningitis findings)
infarcts and leukoencephalopathy)
Radiographic
Neuroimaging reveals signs of multifocal ischaemia of varying ages
Variable presence of leptomeningeal enhancement
Angiography normal in approximately 90%
While these criteria can be used to secure a diagnosis of PACNS,
Pathology
further categorization into one of several currently recognizable Vasculitis of small and medium vessels of leptomeninges and underlying
clinical subsets is essential to determine prognosis and prescribe cortex with variable degrees of granulomatous changes
optimal therapy. Among the clinical forms of PACNS, a few may Giant cells may or may not be present
represent distinct nosological entities, for example granulomatous
Exclusions
angiitis of the CNS, with distinct histology and possibly unique
Diseases and conditions in Table 36.1
pathobiology. Other subsets may merely represent alternative
CHAPTER 36 vasculitis of the central nervous system 499

headache, the most common symptom, may be extraordinarily severe this clinicopathological subset were older at diagnosis, predomi-
or relatively mild, but never thunderclap in nature. The headache may nantly male, had a more acute onset, a higher frequency of cognitive
wax and wane, with periods of prolonged symptom-free remission dysfunction, and showed prominent gadolinium-enhanced lep-
unassociated with therapy (Calabrese 1995). Thunderclap headaches tomeningeal lesions with MRI, as compared to patients with PACNS
are extremely rare in GACNS in contrast to RCVS. without histological evidence of amyloid deposition (Scolding et al.
A variety of less common clinical manifestations of GACNS have 2005; Salvarani et al. 2008a). There is apparent increased frequency of
also been reported, including cranial neuropathies, visual changes, ML in the amyloid-related angiitis subset of PACNS; this may relate
ataxia, and coma. Patients with GACNS may rarely present with in part to publication bias, and to the greater likelihood of a brain
cerebral haemorrhage. biopsy being performed in an elderly patient with a mass lesion as
As noted, non-focal neurological deficits occur in this disor- compared to those presenting with other manifestations of CAA.
der and include an acute or subacute encephalopathy (Scolding Nevertheless, this finding emphasizes the importance of staining for
et al. 1997), which rarely manifests as dementia alone. A decreas- amyloid in patients presenting with ML-PACNS.
ing or fluctuating level of consciousness is more common, how- The spectrum of amyloid deposition in the cerebral tissue include
ever. A  pseudomultiple sclerosis-like presentation has also been CAA with perivascular inflammatory infiltrates, including multi-
described (Scolding et al. 1997). nucleated giant cells, in the absence of true vasculitis; this has been
The signs and symptoms of systemic vasculitis, such as fever, referred to as ‘CAA-related inflammation’ (Eng et al. 2004). This
weight loss, rashes, peripheral neuropathy, and visceral end-organ suggests the existence of a spectrum of severity of inflammatory
dysfunction, are only rarely part of GACNS. Wasting and low-grade reaction to the presence of amyloid β, from little or no inflamma-
fever may be observed following prolonged neurological dysfunc- tion, to perivascular infiltrates, to marked granulomatous angiitis.
tion, but these are not part of the primary disorder. The inflammatory infiltrate in GACNS and cerebral amyloid
angiopathy (Anders et al. 1997) is somewhat atypical for classical
Spinal cord vasculitis GACNS in that the mononuclear cells are found only in the outer
Several other distinctive clinical presentations deserve comment for portion of the vessels and surrounding brain parenchyma and are
they often pose diagnostic difficulties. Spinal cord involvement is well not transmural (Anders et al. 1997). The presence of Aβ peptide
documented but uncommon in PACNS (Goertz et al. 2010; Ropper deposition primarily in the thickened media of vessels damaged by
et al. 2003; Campi et al. 2001). Spinal cord involvement may or may amyloid and within the cytoplasm of multinucleated giant cells may
not coincide with cortical involvement. When isolated it may pose indicate that the granulomatous angiitis represents a foreign body
a formidable diagnostic challenge. Most cases reported in the litera- response to amyloid proteins, causing secondary destruction of the
ture were diagnosed post-mortem and only recently has ante-mortem vessel wall. Therapy for this variant of PACNS in unsettled. From
diagnosis become more common. The symptoms and signs of spinal their experience, Salvarani et al. (2008a) reported that most of these
cord involvement are non-specific and are similar to any other vascular patients responded promptly to therapy; further, they concluded
or inflammatory myelopathy. Treatment is similar to that of GACNS. that the vasculitis influences the clinical findings to a greater degree
Mass lesion presentation than the presence of amyloid deposits in the vessels.
Less well appreciated is the presentation of PACNS as a mass lesion Lymphocytic PACNS
(ML) in about 5% of reported cases (Salvarani et al. 2008b). The clini- This subset represents patients with PACNS diagnosed on the basis
cal findings in this setting are non-specific, but headache and a vari- of pathology but with the presence of lymphocytic rather than
ety of focal and diffuse abnormalities are common. Angiography is granulomatous angiitis on CNS biopsy. In general, the favoured
generally abnormal, with a ‘mass effect’. These vasculitic mass lesions approach to such patients is combination immunosuppressive
have no distinctive features to differentiate them from other patho- therapy with glucocorticoids and azathioprine or mycophenolate,
logical lesions, so they must all be approached as potentially neoplas- with cyclophosphamide being reserved for cases refractory to the
tic. Molloy et al. (2008), from their cohort and the medical literature, favoured therapies (Hajj-Ali et al. 2011).
identified a total of 38 cases of CNS vasculitis who presented with ML.
Twenty-nine per cent of these lesions were positive for amyloid-related Angiographically defined PACNS
angiitis. Poorer outcomes were reported in the amyloid group, with This PACNS subset is diagnosed by abnormal cerebral angiography
five deaths. Of the non-amyloid group, better outcomes were seen in and cerebrospinal fluid (CSF) examination but normal CNS biopsy.
the group treated with corticosteroids and cyclophosphamide as com- The absence of pathological documentation of vasculitis in this
pared with the group treated with corticosteroids alone. Biopsy evi- subset requires a high degree of ongoing scrutiny for an alternative
dence of angiitis is required for diagnosis; specimens should routinely diagnosis, even when no such alternative diagnosis is identified at
be stained for amyloid. While excision of the lesion may be curative initial presentation. Glucocorticoid monotherapy may be sufficient
(Berger et al. 1995), aggressive immunosuppressive therapy is associ- in these cases, with additional immunosuppressive therapies added
ated with favourable outcomes and may obviate the need for surgery. in patients that relapse (Hajj-Ali et al. 2011).

Amyloid β-related angiitis


Cerebral amyloid angiopathy (CAA) is a well-established cause of cer- Secondary forms and mimics
ebral haemorrhage with half of such cases associated with progression of CNS vasculitis
to Alzheimer’s disease. In recent years, it has increasingly been recog-
nized that CAA may be associated with angiocentric inflammatory Infections
cell infiltrates indistinguishable from PACNS; this has been referred In the evaluation of CNS vasculitis, it is imperative to search for
to as Aβ-related angiitis (ABRA) (Scolding et al. 2005). Patients with infection through microbiological analysis of CSF and biopsy
500 SECTION 5 vasculitic diseases and syndromes and related disorders

material. This cannot be overemphasized because the clinical majority of brain lesions (59%). Of note, the coexistence of multiple
and angiographic presentations of infection-related cerebral arte- pathological processes in a brain was characteristic in this series
ritis mimic those of PACNS (Calabrese et  al. 1997; Mandell and (Table 36.3). A peculiar variant of isolated CNS vasculitis associ-
Calabrese 1998). Furthermore, the underlying infection may be ated with microaneurysm has been reported in HIV-infected chil-
occult when the neurovascular complication occurs. Suspicion of dren (Ake et al. 2006; Bonkowsky and Pavia 2007). This illustrates
specific pathogens should be guided by epidemiological features the complexities of the relationship between HIV infection and
and individual risk factors. Screening for infectious agents, includ- CNS vasculitis. The clinical outcome of infection-associated CNS
ing human immunodeficiency virus (HIV), VZV, hepatitis C virus vasculitis is variable but it has been reported to respond to combi-
(HCV), and syphilis, should be considered standard (Mandell nation antiretroviral therapy (Cutfield et al. 2009).
and Calabrese 1998). Infectious vasculitis of the CNS can affect
small vessels, as seen in the case of VZV in immunocompromised Lymphoproliferative diseases
hosts (Gilden et al. 2009) as well as in vasculitis affecting large and Vasculitis of the CNS has been reported in association with
medium-sized vessels (Katchanov et al. 2010). The range of organ- Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, and angioim-
isms is broad and includes viruses as noted previously as well as munolymphoproliferative lesions (AIL) (Calabrese et  al. 1997).
bacteria, fungi, and parasites. Of note, the anatomic location of the lymphoproliferative disease
is not necessarily within the CNS. Although the clinical presenta-
PACNS Associated with varicella zoster infection tion is generally similar to that of PACNS, mass lesions, spinal cord
While PACNS associated with infection is generally considered a involvement, and CNS haemorrhage should raise suspicion for an
secondary form of arteritis, the relationship between PACNS and underlying lymphoproliferative disease (Calabrese et al. 1997). It is
VZV is perplexing enough to warrant a separate discussion. The important to realize that a biopsy of the CNS lesion may only reveal
association of VZV infection with CNS vascular disease is well the angiitis without evidence of malignancy. On the other hand,
known (Gilden et al. 2009). Several syndromes have been described a lymphocytic angiitis may itself be the malignant lesion of AIL,
well, especially that of contralateral hemiplegia in the setting of a diagnosis established by detailed immunohistochemistry, T-cell
herpes zoster ophthalmicus or VZV infection of the trigeminal receptor analysis, and B-cell immunoglobulin studies. Finally, one
nerve (Sigal 1987). The clinical features of this syndrome include an should also look for concomitant infections, particularly VZV.
ischaemic CNS event that occurs several weeks to months follow- Treatment is generally directed at the underlying lymphoprolifera-
ing VZV infection. The ischaemia relates to vasculitis of the middle tive disease, be it within or outside the CNS. This generally consists
cerebral artery or several of its branches and occasionally the inter- of combination chemotherapy and irradiation.
nal carotid artery. Vasculitis follows, from retrograde spread of VZV More commonly, malignancy within the CNS can serve to mimic
via anastomotic branches of the Gasserian ganglion to the cerebral PACNS and this may occur in two different situations. Primary CNS
circulation (Gilden et al. 2009; Gilden 2002; Gilden et al. 2002). In lymphoma can closely mimic the clinical and neuroradiographic
general, the arteritis remains anatomically localized and monopha- appearance of PACNS because of its angiotropic predisposition.
sic in course, and we do not view this as true PACNS, from which it Biopsies that lack clear cellular atypia but are purely lymphocytic
is readily differentiated on clinical and angiographic grounds. deserve careful examination by immunohistochemistry and gene
A more complex VZV-associated syndrome of CNS vascular rearrangement studies. The other neoplastic variant that is the
disease indistinguishable from classic GACNS has been described closest mimic of PACNS is intravascular B-cell lymphoma, which
in patients who frequently have altered host defences (Lie 1992b; may be either systemic or confined to the CNS. In this disorder
Gilden et al. 2009). This disease may evolve in the setting of VZV there may be subacute to chronic neurological decline, modest CSF
infection of the trigeminal nerve or a somatic dermatome, or even abnormalities, and even highly suspicious angiographic changes,
in the absence of clinical VZV infection. For example, Gilden and all suggesting an inflammatory disease. CSF cytology is often
colleagues (Gilden et al. 2009; Gilden 2002; Gilden et al. 2002) have unrewarding and only biopsy can secure this diagnosis (Lie 1992a;
described GACNS where VZV was identified, via molecular tech- Lozsadi et al. 2005).
niques, in a patchy distribution throughout the inflamed cerebral
vessels in an elderly patient with no clinical evidence of preced-
ing VZV infection. The treatment for such cases is problematic
Table 36.3 HIV-associated CNS disease
but probably requires a combination of antiviral and immunosup-
pressive drugs, such as a brief course of glucocorticoids as recom- Pathologic findings
mended by Gilden and colleagues (Gilden et al. 2009), There are no
Encephalitis
controlled trials, however, on which to base therapy.
Leptomeningitis
CNS Vasculitis in HIV infection Vasculitis
Evaluating patients with HIV infection for possible CNS vasculi- Vacuolar myelopathy
tis is particularly challenging. An array of CNS vascular diseases, Leukoencephalopathy
both inflammatory and non-inflammatory, has been associated Aetiologies
with HIV infection. In an analysis of CNS specimens from 100 HIV
cases of acquired immunodeficiency syndrome (AIDS), pathologi- Opportunistic infections
cal changes were detected in 87 (Mossakowski and Zelman 1997). Lymphoproliferative diseases
Encephalitis, leptomeningitis, or vasculitis were described in 35%
Drug-associated vasculopathy
of cases. Opportunistic CNS infections, however, accounted for the
CHAPTER 36 vasculitis of the central nervous system 501

Systemic vasculitides and connective tissue diseases vasoconstrictive syndromes (RCVS) (Calabrese et al. 2007). RCVS
Vasculitis of the CNS may occur with any of the systemic vasculitides, include a group of conditions all sharing similar reversible cerebral
but is most commonly reported in polyarteritis nodosa (PAN), micro- vasoconstriction and each presenting with angiographic findings
scopic polyangiitis (MPA), Behçet’s syndrome (BS), granulomatosis indistinguishable from CNS arteritis.
with polyangiitis (GPA, Wegener), and eosinophilic granulomatosis Patients with RCVS have historically presented to different spe-
with polyangiitis (Churg–Strauss syndrome) (Calabrese et al. 1997; cialists such as neurologists, headache specialists, obstetricians,
Hajj-Ali 2010; Salvarani et  al. 2007). The true prevalence of CNS and rheumatologists, depending on their symptoms. These inves-
arteritis in systemic vasculitides is difficult to estimate because the tigators have imparted their own biases in nomenclature, theories
diagnosis is most often presumed on clinical grounds when neuro- of pathogenesis, and clinical approaches. Only in recent years have
logical events occur in the setting of systemic disease (pre-mortem investigators begun to define the unifying features of those disor-
pathological evidence of CNS vasculitis is rarely sought). Alternative ders that we now consider under the umbrella of RCVS (Calabrese
explanations should be considered initially in patients with systemic et al. 2007; Singhal et al. 2011; Ducros et al. 2007; Chen et al. 2010;
vasculitis presenting with symptoms or signs of CNS dysfunction Ducros 2012).
(Salvarani et al. 2012; Hajj-Ali et al. 2011). Treatment of CNS disease In the field of neurology, the concept of cerebral vasospasm has
is directed at the underlying systemic vasculitis and generally con- long been understood in the setting of delayed vasoconstriction fol-
sists of high-dose glucocorticoids. Cytotoxic drugs are added in MPA lowing subarachnoid haemorrhage, and in rare patients described
and GPA as well as in other serious systemic vasculitides in which with what was once thought of as migrainous vasospasm (Solomon
permanent CNS damage may occur. et al. 1990). In 1988, Call and colleagues reported 19 patients with
CNS involvement is not uncommon in connective tissue dis- ‘reversible cerebral arterial segmental vasoconstriction syndrome’
eases, mainly systemic lupus erythematosus (SLE) and Sjögren’s (Call et al. 1988). These patients were predominantly women who
syndrome (SS) (Hajj-Ali et  al. 2011). In SLE, brain lesions most had acute headaches with or without focal neurological deficits,
often represent vasculopathy, with small-vessel hyalinization, occurring either spontaneously or in association with a variety of
thickening, intramural platelet deposition, and thrombus forma- conditions, such as vasoactive drug exposure, trauma, and oth-
tion (Jennekens and Kater 2002). Frank CNS vasculitis occurs in ers. CSF analysis was largely benign. While the pathophysiology
less than 7% of cases of SLE (Ellis and Verity 1979). In Sjögren’s was unknown, migrainous vasospasm (vasospasm secondary to
syndrome, CNS manifestations may be caused by a mononuclear migraine headache) was considered likely. The eponym Call–
inflammatory vasculopathy involving the small vessels of the cortex Fleming syndrome has endured in neurology and is still applied to
and meninges (Alexander 1993). Angiographic abnormalities con- patients with reversible cerebral vasoconstriction.
sistent with vasculitis are seen uncommonly. In rheumatology, the 1970s witnessed an increasing use of cer-
ebral angiography for the diagnosis of PACNS in the absence of
Miscellaneous conditions that confirming histopathology (Calabrese et al. 1997). It is of interest
can mimic PACNS that many of these patients with RCVS documented by angiogra-
A variety of other disorders are capable of mimicking the clinical phy were women who had benign outcomes and normal or near
and angiographic picture of CNS vasculitis, while others are associ- normal CSF. In 1993, Calabrese and co-workers (Calabrese et  al.
ated with true CNS arteritis (Table 36.1) (Calabrese 1995; Singhal 1993)  reviewed these cases and proposed the term ‘benign angi-
2012). These include a variety of embolic and hypercoagulable dis- opathy of the central nervous system’ (BACNS) to define a subset
orders. The antiphospholipid antibody (aPL) syndrome is of partic- of patients with PACNS defined on the basis of their acute presenta-
ular interest since it is clearly associated with CNS ischaemic events tion, angiographic abnormalities, normal spinal fluid examinations,
related to thrombosis. Other arteriopathies include common dis- and monophasic course. While patients fitting this picture often had
orders such as atherosclerosis, small-vessel disease due to diabetes, favourable outcomes and thus were benign (Snyder and McClelland
dyslipedaemia, and hypertension, Moya moya disease, intracranial 1978) compared to those PACNS patients with the granulomatous
artery dissection, and arteriopathy secondary to remote cranial variant, BACNS patients could also have more severe outcomes such
irradiation (Singhal 2012). as stroke. The term ‘angiopathy’ was employed to emphasize that in
The presence of retinal disease and or inner ear disease sugges- the absence of histopathological confirmation there was consider-
tive of cochlear dysfunction should raise the spectrum of retino- able uncertainty with regards to vessel wall status.
cochleocerebral vasculopathy and in particular Susac’s syndrome In 2002, we reported (Hajj-Ali et al. 2002) that in a series of 16
(Martinet et al. 2007), which carries specific prognostic and thera- BACNS patients studied serially by neurovascular imaging there
peutic implications. Lastly, a family history of unexplained central was dramatic resolution of angiographic abnormalities in 4 to 12
nervous system disease should prompt a search for a number of weeks without intensive immunosuppressive therapy. Several of
genetic variants, including cerebral autosomal dominant arteriopa- these patients received either calcium channel blockers alone or
thy with subcortical infarcts and leukoencephalopathy (CADASIL), were merely observed. In two patients biopsies were obtained,
its autosomal recessive variant (CARASIL), retinal vasculopathy revealing no evidence of vasculitis. Based upon these data, it
with cerebral leukodystrophy (RVCL), and others (Singhal 2012). became increasingly apparent that for most patients presenting
with what was recognized as BACNS the underlying vascular lesion
Reversible cerebral vasoconstrictive was reversible vasoconstriction rather than arteritis.
Similar cases of RCVS have been described in the obstetrical lit-
syndromes erature under the labels post-partum angiopathy (Singhal 2004),
Perhaps the greatest single advance in CNS vasculitis has been the post-partum angiitis (Farine et al. 1984), benign isolated angiitis of
recognition of its major angiographic mimic, the reversible cerebral the CNS (Sugiyama et al. 1997), and puerperal vasospasm.
502 SECTION 5 vasculitic diseases and syndromes and related disorders

Currently, the term RCVS is well accepted in the literature to Table 36.5 Features distinguishing between PACNS and RCVS
encompass syndromes that share clinical, laboratory, and radio-
logical phenotypes. Essential elements for the diagnosis of RCVS PACNS RCVS
have been proposed by Calabrese et  al. and are summarized in Sex predominance Male Female
Table 36.4. The pathogenesis of RCVS is thought to be due to vaso-
constriction of the cerebrovasculature. More than half the cases Onset Long prodrome; Acute; hours to
3–6 months days
occur post partum or after exposure to adrenergic or serotoner-
gic drugs. The syndrome is monophasic and a majority of cases Symptoms and signs Chronic headache, Thunderclap
have a benign outcome. However, several haemorrhagic and focal, and non-focal headache
ischaemic strokes, brain oedema, and death can occur. Infarctions ± focal event
occur mainly in arterial watershed regions of the cerebral hemi- CSF analysis Chronic meningitis findings Normal
spheres, often between the posterior circulation and the carotid
Angiogram Normal in 40% Abnormal in 100%
territories. Parenchymal haemorrhages are more frequently single
than multiple and lobar than deep, and more often associated with MRI Abnormal in 100% Normal in 15%
another type of stroke. Convexity subarachnoid haemorrhages are Lobar haemorrhages Very rare Common
non-aneurysmal, usually mild, unilateral or bilateral, and mani-
Convexity subarachnoid Very rare Common
fest as a hyperintense signal on fluid-attenuated inversion recovery
haemorrhage
(FLAIR) MRI and a hypointense signal on T2-weighted MRI in
a few sulcal spaces near the convexity (Ducros 2012). Reversible Biopsy Vasculitis inin 75–80% No evidence of
brain oedema is an early manifestation of RCVS and more fre- vasculitis
quently associated with at least one variety of strokes than iso-
lated. Oedema usually totally reverses within 1 month of clinical
onset, much earlier than does vasoconstriction (Ducros 2012). To inclusion of RCVS, and many patients with RCVS may have been
diagnose RCVS, direct (transfemoral) or indirect (CT or magnetic subjected to the risks of brain biopsy and chronic immunosup-
resonance) cerebral angiography is needed to show segmental nar- pressive therapy. PACNS and RCVS have important differences
rowing and dilatation of multiple vessel beds. Calibre irregularities at multiple levels. Clinically, RCVS is characterized by an acute
can affect the anterior and the posterior circulation, and are mostly onset of recurrent thunderclap headaches and associated fac-
bilateral and diffuse. To secure the diagnosis of RCVS a repeat cer- tors such as vasoconstrictive drug exposure or recent childbirth,
ebral vascular study is need to show resolution of the vasoconstric- which are present in about half of the cases. This contrasts with the
tion in about 60 days. chronic presentation of PACNS, where the headaches are chronic
and progressive. The CSF is normal or near normal in RCVS in
Differentiating RCVS from PACNS contrast to abnormal values seen in PACNS. At the radiological
The differentiation of RCVS from PACNS is of vital clinical level PACNS is rarely associated with normal brain parenchymal
importance because their therapies differ greatly (Table  36.5). imaging or haemorrhagic lesions. In contrast, patients with RCVS
Misdiagnosis between the two entities is common due to overlap- can have severe vasoconstriction without parenchymal lesions;
ping clinical and imaging features, such as headaches, seizures, lobar and cortical surface subarachnoid haemorrhages as well as
strokes, and cerebral angiographic abnormalities. Indeed, many reversible oedematous lesions similar to the lesions of the ‘pos-
published reports of PACNS may have been confounded by the terior reversible leukoencephalopathy syndrome’ are common.
Finally, in RCVS, the angiographic abnormalities are dynamic,
more severe, and typically resolve over a period of weeks to
3  months. Nevertheless, in some cases differentiating PACNS
Table 36.4 Key features of reversible vasoconstrictive from RCVS remains challenging, given the false-negative brain
syndromes (RCVS) biopsy in PACNS and the absence of confirmatory serological
tests for PACNS. Ongoing research using high-resolution MRI
A diagnosis of RCVS requires all of the following criteria: shows promise in distinguishing the various causes of cerebral
1. A severe and acute headache with or without other associated arteriopathies based on the presence and the pattern (e.g. concen-
neurological signs and symptoms tric vs. eccentric) of vessel wall contrast enhancement (Mandell
et al. 2012).
2. No evidence of aneurismal subarachnoid haemorrhage
3. Normal or near normal cerebrospinal fluid analysis
a. Protein <70 mg% Diagnostic studies for CNS vasculitis
b. White blood cell count <20 cells per/mm3 Several tools are available to assist in the diagnosis of PACNS.
c. Normal glucose The main challenge remains to critically assess their diagnostic
d. No evidence of subarachnoid haemorrhage sensitivity and specificity in the diagnosis of PACNS. Diagnostic
4. Reversible cerebral segmental vasoconstriction involving arteries of Circle of studies for PACNS include routine laboratory testing, CSF, neu-
Willis (see Figure 36.3) roimaging studies, cerebral angiography, and biopsy of CNS tis-
a. Documented by serial angiography, CT, or MR angiography or sues. Screening laboratory studies performed on blood have little
flow-related vascular technique (transcranial Doppler) within 12 weeks positive predictive value because there are no tests of sufficient
after onset specificity to secure a diagnosis but various tests for chemistries,
CHAPTER 36 vasculitis of the central nervous system 503

autoantibodies, and cultures and other investigations are useful for Brain biopsy remains the gold standard for the diagnosis of
ruling out the conditions listed in Table 36.1. PACNS. Obtaining tissue for the diagnosis is of great value in
Acute-phase reactants, such as erythrocyte sedimentation rate positively identifying angiitis, in cases where vascular imaging is
(ESR) and C-reactive protein (CRP), are usually normal in patients inconclusive for a diagnosis and in identifying mimics of PACNS
with PACNS. However, these markers are important to obtain as (Parisi and Moore 1994). Two key messages are of great impor-
assessment for any systemic inflammatory or infectious process. tance in interpreting the biopsy results: first, when confronted
Serological tests, performed in several autoimmune and autoin- by a biopsy demonstrating vasculitis, special stains and cultures
flammatory diseases, are negative in PACNS; however, these are are still necessary to confirm that the lesion does not represent
performed to exclude mimics. The extent of an infectious work up a secondary form of vasculitis, such as CNS lymphoma or infec-
is determined by the epidemiological factors and the host immune tions; second, a non-diagnostic or negative biopsy does not rule
defence. out the diagnosis of PACNS. False-negative biopsies can be as
Cerebrospinal fluid analysis is an important part of the work up high as 25% of autopsy-documented cases (Parisi and Moore
in PACNS. CSF findings are non-specific for PACNS, but the central 1994)  due to the patchy involvement or inaccessibility of the
role of CSF analysis is to exclude infectious or malignant diseases. lesions.
In pathologically documented cases of PACNS, CSF abnormalities The gross neuropathological findings in GACNS consist of
are present in the majority of the patients; the characteristic CSF multiple small foci of infarction or haemorrhage, but occasion-
findings are those of elevated protein and pleocytosis. The white ally larger areas have been involved. Microscopically, GACNS
count is rarely over 250 cells/mm³ and protein rarely exceeds 500 is a segmental necrotizing and granulomatous vasculitis of the
mg% (Calabrese et al. 1992; Younger and Kass 1997). small- and medium-sized arteries, predominantly in the lep-
Magnetic resonance imaging is the central neuroradiographic tomeninges and cortex. Veins are affected in about half of the
modality in the work up of patients with suspected PACNS. The cases. The predominant inflammatory cell is the small lympho-
sensitivity of MRI in PACNS has been reported as low as 75% or cyte with an admixture of epitheloid cells, plasma cells, mac-
approaching 100% (Hajj-Ali et al. 2011). Interpretation of figures rophages, and giant cells of both the Langhans and foreign body
derived from older series is limited because most series were con- types. Not all of these features are present in all patients, and the
founded by inclusion of many patients with RCVS. Infarcts are the abnormalities have a patchy distribution throughout the CNS.
most common lesions, occurring in up to 53% of PACNS (Hajj-Ali Frank, well-formed granulomas may be seen, but are relatively
et al. 2011); infarcts are often multiple, affecting the cortex and the rare (Figure  36.1). More commonly, there is loose granuloma
subcortex. Non-specific high-intensity T2WI/FLAIR lesions in formation with giant cells (Figure  36.2). Lastly, up to 20% of
white matter are common (Hajj-Ali et al. 2011). Mass lesions occur cases are lymphocytic forms without prominent granulomatous
in around 5% of patients with PACNS. Intracranial haemorrhage features.
is rare, occurring in 8% of the patients. Collectively, abnormali- When biopsy is performed, it should include both brain and
ties of CSF and MRI are noted in close to 100% of patients. Thus, leptomeninges. The preferred site for brain biopsy is a radio-
when normal, these studies (CSF studies and MRI) generate a high graphically abnormal area; if this is not amenable to biopsy, the
negative predictive value, making the diagnosis of GACNS unlikely temporal tip of the non-dominant hemisphere is an alternative
(Stone et al. 1994). site (Parisi and Moore 1994). An infratentorial approach should
Direct or indirect angiography is often used in the diagnostic be strongly considered in patients suspected of having conditions
work up of PACNS. The typical angiographic findings of PACNS such as tuberculosis or sarcoidosis, which preferentially involve
include alternating areas of stenosis and dilatation, referred to as the basilar meninges.
beading, which can be smooth or irregular and typically occur
bilaterally but can also involve single vessels (Stone et  al. 1994;
Pomper et al. 1999). It is very important to note that these angi-
ographic findings are not specific and are widely encountered in
non-vasculitic disorders such as atherosclerosis, radiation vascu-
lopathy, fibromuscular dysplasia, infection, and in particular RCVS
(Stone et al. 1994; Pomper et al. 1999). The low specificity of cere-
bral angiograms in the diagnosis of PACNS, as low as 30%, suggests
the need for further work-up to rule out other mimicking condi-
tions. Other angiographic findings include tapering of the vessel
lumen of a single vessel or multiple vessels.
Cerebral angiograms are more sensitive than MRA but their spa-
tial resolution remains limited in vessels smaller than 500 mm in
diameter; thus the high false-negative rate when disease is limited
to small vessels such as in GACNS.
Advances in MRI techniques, such as the high-resolution mul-
ticontrast wall and lumen imaging, are a promising modalities.
This technique provides information about the wall structure of the Fig. 36.1 Marked mononuclear inflammatory infiltrates in a cortical vessel with a
intracranial blood vessels, which luminal imaging cannot, and it is well-formed paravascular granuloma (haematoxylin and eosin stain, magnification
hoped that this will enhance the test specificity in CNS vasculopa- × 200). Reprinted from: Vasculitis of the central nervous system. 21, Calabrese, L.H.
thies (Mandell et al. 2012). 1059–76. Copyright (1995), with permission from Elsevier.
504 SECTION 5 vasculitic diseases and syndromes and related disorders

(a) (b) Younger and colleagues (Younger and Kass 1997)  analysed out-
come in 50 reported cases and noted that 70% of patients treated
with glucocorticoids with or without cyclophosphamide survived,
although often with significant sequelae. We believe that the major-
ity of patients with documented GACNS should be treated with
combined therapy at least initially, in a similar approach to the
systemic small vessel vasculitides algorithm (Molloy and Langford
2006), until the disease has been controlled, usually between 3
and 6 months. Attempts should then be made to limit exposure
to the alkylating agent by switching to maintenance therapy with
an antimetabolite such as mycophenolate mofetil or azathioprine
(Hajj-Ali et  al. 2011). There are no long-term follow-up studies
that can shed light on the duration of treatment. In the author’s
experience, immunosuppressive maintenance therapy is used
for the long term in the absence of contraindications or adverse
effects. All patients should receive prophylaxis for Pneumocystis
Fig. 36.2 (a) Brain biopsy findings of granulomatous vasculitis in PACNS side by jirovecii infection and special attention to their bone health in view
side with a polyarteritis nodosa-type necrotizing vasculitis (open arrow) and two of their long-term steroid intake.
normal arterioles (short arrows). (b) Close-up showing foreign body (short arrows) Following disease activity in PACNS can be problematic. Clinical
and Langhans’ (long arrow) giant cells in the granulomatous inflammation. symptoms and signs may be slow to change, as in any ischaemic
(Hematoxylin and eosin stain, magnification × 64 and × 400). Reproduced insult to the brain. MRI changes should improve, but are likely
with permission from Calabrese, L.H., Duna, G.F. and Lie, J.T. 1997. Vasculitis in
to leave residual scar. A clear discussion with the patient should
the central nervous system. Arthritis Rheum, 40, 1189–201. Copyright © 1997
American College of Rheumatology. occur initially to educate them about disease damage versus dis-
ease progression, to realistically set expectations. Following clinical
stability, serial MRI examinations at 3 to 6-month intervals should
be performed during the tapering phase of immunosuppressive
therapy. Relapses do occur and may appear after many years of
clinical stability. As with long-term treatment of any vasculitis with
immunosuppressive drugs, vigilance to reduce treatment-related
morbidity from infections, osteoporosis, and other complications
is of paramount importance (Hajj-Ali et al. 2011).

References
Ake, J.A., Erickson, J.C., and Lowry, K.J. (2006). Cerebral aneurysmal arte-
riopathy associated with HIV infection in an adult. Clinical Infectious
Diseases, 43, e46–50.
Alexander, E.L. (1993). Neurologic disease in Sjogren’s syndrome: mononu-
clear inflammatory vasculopathy affecting central/peripheral nervous
system and muscle. A clinical review and update of immunopathogen-
esis. Rheumatic Diseases Clinics of North America, 19, 869–908.
Anders, K.H., Wang, Z.Z., Kornfeld, M., Gray, F., Soontornniyomkij, V., Reed,
L.A., Hart, M.N., Menchine, M., Secor, D.L., and Vinters, H.V. (1997).
Giant cell arteritis in association with cerebral amyloid angiopathy: immu-
nohistochemical and molecular studies. Human Pathology, 28, 1237–46.
Berger, J.R., Romano, J., Menkin, M., and Norenberg, M. (1995). Benign focal
cerebral vasculitis: case report. Neurology, 45, 1731–4.
Fig. 36.3 An angiogram demonstrating numerous areas of focal stenosis and Bonkowsky, J. and Pavia, A. (2007). Cerebral vasculopathy with aneurysm for-
ectasia from a patient with RCVS. mation in HIV-infected young adults. Neurology, 68, 623; author reply 623.
Calabrese, L.H. (1995). Vasculitis of the central nervous system. Rheumatic
Diseases Clinics of North America, 21, 1059–76.
Calabrese, L.H. (2009). Primary angiitis of the central nervous sys-
Treatment and prognosis tem: reflections on 20 years of investigation. Clinical and Experimental
There are no controlled clinical trials in PACNS. Therapeutic Rheumatology, 27, S3–4.
guidelines are informal and have been based upon a collective Calabrese, L.H., Dodick, D.W., Schwedt, T.J., and Singhal, A.B. (2007).
experience suggesting that PACNS is progressive and uniformly Narrative review:  reversible cerebral vasoconstriction syndromes.
Annals of Internal Medicine, 146, 34–44.
fatal if untreated (Calabrese et al. 1997). More recent reviews of
Calabrese, L.H., Duna, G.F., and Lie, J.T. (1997). Vasculitis in the central
this topic have questioned whether this observation is accurate. nervous system. Arthritis and Rheumatism, 40, 1189–201.
These indicate that the reported mortality reflect historical bias, Calabrese, L.H., Furlan, A.J., Gragg, L.A., and Ropos, T.J. (1992). Primary
with the majority of untreated cases discovered at post mortem angiitis of the central nervous system: diagnostic criteria and clinical
examination (Calabrese et  al. 1997; Younger and Kass 1997). approach. Cleveland Clinic Journal of Medicine, 59, 293–306.
CHAPTER 36 vasculitis of the central nervous system 505

Calabrese, L.H., Gragg, L.A., and Furlan, A.J. (1993). Benign angiopathy: a intensive care unit: a clinico-radiological study. Neurocritical Care, 12,
distinct subset of angiographically defined primary angiitis of the cen- 369–74.
tral nervous system. Journal of Rheumatology, 20, 2046–50. Lie, J.T. (1992a). Malignant angioendotheliomatosis (intravascular lympho-
Calabrese, L.H. and Mallek, J.A. (1988). Primary angiitis of the central nerv- matosis) clinically simulating primary angiitis of the central nervous
ous system. Report of 8 new cases, review of the literature, and proposal system. Arthritis and Rheumatism, 35, 831–4.
for diagnostic criteria. Medicine (Baltimore), 67, 20–39. Lie, J.T. (1992b). Primary (granulomatous) angiitis of the central nervous
Call, G.K., Fleming, M.C., Sealfon, S., Levine, H., Kistler, J.P., and Fisher, system: a clinicopathologic analysis of 15 new cases and a review of the
C.M. (1988). Reversible cerebral segmental vasoconstriction. Stroke, 19, literature. Human Pathology, 23, 164–71.
1159–70. Lozsadi, D.A., Wieshmann, U., and Enevoldson, T.P. (2005). Neurological
Campi, A., Benndorf, G., Filippi, M., Reganati, P., Martinelli, V., and Terreni, presentation of intravascular lymphoma: report of two cases and discus-
M.R. (2001). Primary angiitis of the central nervous system: serial MRI sion of diagnostic challenges. European Journal of Neurology, 12, 710–4.
of brain and spinal cord. Neuroradiology, 43, 599–607. Mandell, B.F. and Calabrese, L.H. (1998). Infections and systemic vasculitis.
Chen, S.P., Fuh, J.L., Wang, S.J., Chang, F.C., Lirng, J.F., Fang, Y.C., Shia, B.C., Current Opinion in Rheumatology, 10, 51–7.
and Wu, J.C. (2010). Magnetic resonance angiography in reversible cer- Mandell, D.M., Matouk, C.C., Farb, R.I., Krings, T., Agid, R., Terbrugge, K.,
ebral vasoconstriction syndromes. Annals of Neurology, 67, 648–56. Willinsky, R.A., Swartz, R.H., Silver, F.L., and Mikulis, D.J. (2012). Vessel
Cravioto, H. and Feigin, I. (1959). Noninfectious granulomatous angiitis with wall MRI to differentiate between reversible cerebral vasoconstriction
a predilection for the nervous system. Neurology, 9, 599–609. syndrome and central nervous system vasculitis:  preliminary results.
Cupps, T.R., Moore, P.M., and Fauci, A.S. (1983). Isolated angiitis of the cen- Stroke, 43, 860–2.
tral nervous system. Prospective diagnostic and therapeutic experience. Martinet, N., Fardeau, C., Adam, R., Bodaghi, B., Papo, T., Piette, J.C., and
American Journal of Medicine, 74, 97–105. Lehoang, P. (2007). Fluorescein and indocyanine green angiographies in
Cutfield, N.J., Steele, H., Wilhelm, T., and Weatherall, M.W. (2009). Successful Susac syndrome. Retina, 27, 1238–42.
treatment of HIV associated cerebral vasculopathy with HAART. Journal Molloy, E.S. and Langford, C.A. (2006). Advances in the treatment of small ves-
of Neurology Neurosurgery and Psychiatry, 80, 936–7. sel vasculitis Rheumatic Diseases Clinics of North America, 32, 157–72, x.
Ducros, A. (2012). Reversible cerebral vasoconstriction syndrome. Lancet Molloy, E.S., Singhal, A.B., and Calabrese, L.H. (2008). Tumour-like mass
Neurology, 11, 906–17. lesion: an under-recognised presentation of primary angiitis of the cen-
Ducros, A., Boukobza, M., Porcher, R., Sarov, M., Valade, D., and Bousser, tral nervous system. Annals of the Rheumatic Diseases, 67, 1732–5.
M.G. (2007). The clinical and radiological spectrum of reversible cer- Mossakowski, M.J. and Zelman, I.B. (1997). Neuropathological syndromes in
ebral vasoconstriction syndrome. A  prospective series of 67 patients. the course of full blown acquired immune deficiency syndrome (AIDS)
Brain, 130, 3091–101. in adults in Poland (1987–1995). Folia Neuropathol, 35, 133–43.
Ellis, S.G. and Verity, M.A. (1979). Central nervous system involvement Parisi, J.E. and Moore, P.M. (1994). The role of biopsy in vasculitis of the
in systemic lupus erythematosus:  a review of neuropathologic find- central nervous system. Seminars in Neurology, 14, 341–8.
ings in 57 cases, 1955—1977. Seminars in Arthritis and Rheumatism, Paterson, R.W., Torres-Chae, C.C., Kuo, A.L., Ando, T., Nguyen, E.A., Wong,
8, 212–21. K., Dearmond, S.J., Haman, A., Garcia, P., Johnson, D.Y., Miller, B.L.,
Eng, J.A., Frosch, M.P., Choi, K., Rebeck, G.W., and Greenberg, S.M. (2004). and Geschwind, M.D. (2012). Differential diagnosis of Jakob-Creutzfeldt
Clinical manifestations of cerebral amyloid angiopathy-related inflam- disease. Archives of Neurology, 69, 1578–82.
mation. Annals of Neurology, 55, 250–6. Pomper, M.G., Miller, T.J., Stone, J.H., Tidmore, W.C., and Hellmann, D.B.
Farine, D., Andreyko, J., Lysikiewicz, A., Simha, S., and Addison, A. (1984). (1999). CNS vasculitis in autoimmune disease:  MR imaging findings
Isolated angiitis of brain in pregnancy and puerperium. Obstetrics and and correlation with angiography. American Journal of Neuroradiology,
Gynecology, 63, 586–8. 20, 75–85.
Gilden, D., Cohrs, R.J., Mahalingam, R., and Nagel, M.A. (2009). Varicella Ropper, A.H., Ayata, C., and Adelman, L. (2003). Vasculitis of the spinal cord.
zoster virus vasculopathies: diverse clinical manifestations, laboratory Archives of Neurology, 60, 1791–4.
features, pathogenesis, and treatment. Lancet Neurology, 8, 731–40. Salvarani, C., Brown, R.D. Jr, Calamia, K.T., et al. (2007). Primary central
Gilden, D.H. (2002). Varicella zoster virus vasculopathy and disseminated nervous system vasculitis: analysis of 101 patients. Annals of Neurology,
encephalomyelitis. Journal of the Neurological Sciences, 195, 99–101. 62, 442–51.
Gilden, D.H., Mahalingam, R., Cohrs, R.J., Kleinschmidt-Demasters, B.K., Salvarani, C., Brown, R.D., JR., Calamia, K.T., Christianson, T.J., Huston, J.
and Forghani, B. (2002). The protean manifestations of varicella-zoster 3rd, Meschia, J.F., Giannini, C., Miller, D.V., and Hunder, G.G. (2008a).
virus vasculopathy. Journal of Neurovirology, 8 (Suppl. 2), 75–9. Primary central nervous system vasculitis: comparison of patients with
Goertz, C., Wegner, C., Bruck, W., and Berlit, P. (2010). Primary angiitis of and without cerebral amyloid angiopathy. Rheumatology (Oxford), 47,
the CNS with pure spinal cord involvement:  a case report. Journal of 1671–7.
Neurology, 257, 1762–4. Salvarani, C., Brown, R.D., JR., Calamia, K.T., Christianson, T.J., Huston, J. 3rd,
Hajj-Ali, R.A. (2010). Primary angiitis of the central nervous system:  dif- Meschia, J.F., Giannini, C., Miller, D.V., and Hunder, G.G. (2008b). Primary
ferential diagnosis and treatment. Best Practice and Research Clinical CNS vasculitis with spinal cord involvement. Neurology, 70, 2394–400.
Rheumatology, 24, 413–26. Salvarani, C., Brown, R.D., JR., and Hunder, G.G. (2012). Adult primary
Hajj-Ali, R.A., Furlan, A., Abou-Chebel, A., and Calabrese, L.H. (2002). central nervous system vasculitis:  an update. Current Opinion in
Benign angiopathy of the central nervous system: cohort of 16 patients Rheumatology, 24, 46–52.
with clinical course and long-term followup. Arthritis and Rheumatism, Scolding, N.J., Jayne, D.R., Zajicek, J.P., Meyer, P.A., Wraight, E.P., and
47, 662–9. Lockwood, C.M. (1997). Cerebral vasculitis—recognition, diagnosis
Hajj-Ali, R.A., Singhal, A.B., Benseler, S., Molloy, E., and Calabrese, L.H. and management. QJM, 90, 61–73.
(2011). Primary angiitis of the CNS. Lancet Neurology, 10, 561–72. Scolding, N.J., Joseph, F., Kirby, P.A., Mazanti, I., Gray, F., Mikol, J., Ellison,
Jennekens, F.G. and Kater, L. (2002). The central nervous system in sys- D., Hilton, D.A., Williams, T.L., Mackenzie, J.M., Xuereb, J.H., and
temic lupus erythematosus. Part 2. Pathogenetic mechanisms of clini- Love, S. (2005). Abeta-related angiitis: primary angiitis of the central
cal syndromes: a literature investigation. Rheumatology (Oxford), 41, nervous system associated with cerebral amyloid angiopathy. Brain,
619–30. 128, 500–15.
Katchanov, J., Siebert, E., Klingebiel, R., and Endres, M. (2010). Infectious vas- Sigal, L.H. (1987). The neurologic presentation of vasculitic and rheumato-
culopathy of intracranial large- and medium-sized vessels in neurological logic syndromes. A review. Medicine (Baltimore), 66, 157–80.
506 SECTION 5 vasculitic diseases and syndromes and related disorders

Singhal, A.B. (2004). Postpartum angiopathy with reversible posterior leu- Stone, J.H., Pomper, M.G., Roubenoff, R., Miller, T.J., and Hellmann, D.B.
koencephalopathy. Archives of Neurology, 61, 411–6. (1994). Sensitivities of noninvasive tests for central nervous system
Singhal, A.B. (2012). Diagnostic challenges in RCVS, PACNS, and other cer- vasculitis:  a comparison of lumbar puncture, computed tomogra-
ebral arteriopathies. Cephalalgia, 31, 1067–70. phy, and magnetic resonance imaging. Journal of Rheumatology, 21,
Singhal, A.B., Hajj-Ali, R.A., Topcuoglu, M.A., Fok, J., Bena, J., Yang, D., 1277–82.
and Calabrese, L.H. (2011). Reversible cerebral vasoconstriction syn- Sugiyama, Y., Muroi, A., Ishikawa, M., Tsukamoto, T., and Yamamoto, T.
dromes: analysis of 139 cases. Archives of Neurology, 68, 1005–12. (1997). A benign form of isolated angiitis of the central nervous system
Snyder, B.D. and McClelland, R.R. (1978). Isolated benign cerebral vasculitis. in puerperium: an identical disorder to postpartum cerebral angiopa-
Archives of Neurology, 35, 612–4. thy? Internal Medicine, 36, 931–4.
Solomon, S., Lipton, R.B., and Harris, P.Y. (1990). Arterial stenosis in Younger, D.S. and Kass, R.M. (1997). Vasculitis and the nervous system.
migraine: spasm or arteriopathy? Headache, 30, 52–61. Historical perspective and overview. Neurologic Clinics, 15, 737–58.
CHAPTER 37

Thromboangiitis obliterans
(Buerger’s disease)
Michael Frank and Jean-Noel Fiessinger

Introduction to date being small and results being related to the country of origin
(Papa et al. 1992).
Physicians have been aware of cases of gangrene of the limbs in young TAO was initially thought to affect men almost exclusively; how-
patients for a long time. In 1879, Felix von Winiwarter reported the ever, in the most recent studies, the proportion of female patients sig-
case of a 57-year-old man who had his leg amputated for gangrene nificantly increased, ranging from 11 to 23% (Dehaine-Bamberger
by Billroth; the observed pathological findings described arterial et al. 1993; Olin et al. 1990). This increased incidence in women is
and venous lesions that were chiefly the result of inflammatory pro- closely related to changing smoking habits in the last decades, with
liferation of the intima (Lie et al. 1979). He pointed out that this an increase in tobacco abuse in women in Western countries (Olin
case was very similar to that published by Lariviere in the Journal et al. 1990). This link has not been evidenced in Japan and Asia,
de Bordeaux in 1866. Thirty-nine years later, Leo Buerger described despite similar changes in smoking habits in Asian women.
a new clinical entity, to which he gave the name of thromboangiitis The young age of the patients is another key characteristic of
obliterans (TAO), on the basis of the pathology of 11 amputated TAO; the symptoms almost always begin before the age of 40 years.
limbs (Buerger 1908). The clinical picture involved distal arterial The strong link with smoking is one of the unique features of
occlusions of the upper and, more often, of the lower limbs accom- TAO. Almost all the patients affected are heavy smokers. Once the
panied by migrating thrombophlebitis in young Jewish male smok- disease has become established, stopping smoking is the only effec-
ers. The histological picture was that of a segmental inflammatory tive way to prevent evolution of the disease and its recurrence. The
disease involving small and medium-sized arteries and veins. resumption of smoking, even years later, may trigger new flare-ups
As pointed out by Wessler over 50 years ago (Wessler et al. 1960), of the disease. This strong association suggests that tobacco abuse
no clinical symptoms/signs, radiological or laboratory abnormali- plays a role in the pathogenesis of the disease. It has been suggested
ties, or pathological features were clearly specific for Buerger’s that something undefined in nicotine may be involved. All types
disease. Thus, in the absence of any aetiological factor, Wessler rec- of tobacco have been implicated and a few cases have even been
ommended that the term of Buerger’s disease should be discarded. described among users of smokeless tobacco (O’Dell et al. 1987). At
Nevertheless, there appears to be a consensus to recognize throm- the present time, a majority of our patients with TAO are smokers
boangiitis obliterans as a distinct clinical and pathological entity and users of cannabis. Consideration of a triggering role for can-
(Olin 2000), characterized by an inflammatory occlusive vasculitis nabis has resulted in the description of a specific cannabis arteritis,
of the small and medium-size arteries and veins that affects young which is very similar to Buerger’s disease clinically, and the reported
adult smokers. differences in pathological lesions are not convincing (Combemale
et al. 2005; Karila et al. 2004). Thus tobacco and cannabis should be
considered as co-factors strongly contributing to the pathogenesis
Epidemiology and pathogenesis of TAO (Martin-Blondel et al. 2011).
In the absence of specific diagnostic criteria, it is difficult to deter- Less than 5% of TAO patients are non-smokers. These cases might
mine the exact prevalence of TAO. Although the disease is present be triggered by cold and/or frostbite, and sustained microtrauma of
world-wide, its prevalence seems to be higher in the Middle East, the extremities. Abuse of sympathomimetic drugs (Dezellus et al.
Asia, and in the Far East (Olin 2000; Shionoya 1993), than in Europe 1982; Hagen and Lohse 1984; Kjeldsen and Mozes 1969) has also
or North America. In Europe, TAO accounts for 0.5–5% of patients been reported in patients with TAO.
hospitalized for arterial occlusive disease (Dehaine-Bamberger Several clotting abnormalities have been described in TAO but
et al. 1993). Despite the fact that Buerger’s disease was previously there is no evidence to suggest that antiphospholipid antibodies,
considered to occur almost exclusively in Jews, it is now known to hypercoagulability, or fibrinolytic disorders play a major patho-
affect individuals of all races. However, a higher incidence of TAO genic role (Adar et al. 2000; Siguret et al. 1997). In a prospective
has been reported in Israel, among Ashkenazim as compared with study (Darnige et al. 2010), we compared circulating platelet micro-
non-Ashkenazim (Kjeldsen and Mozes 1969). An increased inci- particle levels of 15 TAO patients with those of a control group of
dence of HLA-A9 and HLA-B5 has been reported (McLoughlin 15 patients with peripheral artery disease. The results suggested an
et al. 1976), yet these findings are conflicting, the published series association between platelet activation and exacerbation of TAO;
508 SECTION 5 vasculitic diseases and syndromes and related disorders

however, platelet microparticle levels are probably more likely to


be a marker of the evolution of the disease rather than a risk factor
of TAO.
Endothelial dysfunction may be involved in the pathophysiol-
ogy of thromboangiitis obliterans. In ten consecutive patients
with TAO and acute lower limb ischaemic ulcers, we reported
(Azizi et  al. 2010)  that flow-mediated vasodilation (FMD) and
endothelium-independent vasodilatation (EID) of the brachial
artery in patients in an active phase of TAO were not impaired.
Moreover, our findings suggested that TAO is associated with an
increase in vasomotor tone of arteries that cannot be accounted for
by endothelial dysfunction alone. The beneficial effects of vasodila-
tor treatments, especially of prostanoids, could thus be explained
by a pharmacological reversal of this increase in vasomotor tone.
Various investigations have been carried out with the aim of
identifying an autoimmune mechanism responsible for TAO (Adar
et al. 1983; Gulati et al. 1979). Hypersensitivity to type I and type
Fig. 37.1 Ischaemic ulceration with necrosis on the distal portion of the great left
III collagen, associated with the presence of anticollagen or antie- toe in a young male patient with thromboangiitis obliterans.
lastin antibodies, has been described but these abnormal findings
have proved to be non-specific, related to inflammatory modifica-
tions rather than being the cause of the vascular lesions (Smolen the presenting symptom is rarely a secondary Raynaud’s phenom-
et al. 1983). Increased levels of antiendothelial cell antibodies have enon associated with persistent signs of peripheral ischaemia. More
been reported in patients with an active disease (Eichhorn et al. commonly, the patients present with distal digital necrosis accom-
1998) but the specificity of such findings remains to be established. panied by excruciating pain. Notably, Buerger’s disease has been
described as being the main cause of digital gangrene in young
males (Vayssairat et al. 1979). The pathogenic relationship between
Clinical presentation old diffuse Raynaud’s phenomenon and the onset of thromboangii-
There are two essential features in the clinical presentation of tis is uncertain (Long and Fiessinger 1989), but may suggest a pos-
TAO:  the involvement of small and medium-sized arteries and sible role of vasospasm in the pathophysiology of TAO.
veins of the limbs, and its characteristic evolution marked by acute Superficial thrombophlebitis is present in 40 to 60% of cases.
exacerbations and long remissions. In contrast, deep vein thrombosis is unusual and is suggestive of
Ischaemia of the lower limbs is the commonest presenting symp- other diagnoses, such as Behçet’s disease. Typically, the superficial
tom. Foot or plantar claudication is virtually specific of Buerger’s phlebitis migrates, involving segments of the upper and lower limb
disease (Hirai and Shionoya 1978). The patient complains on exer- superficial veins. Lesions usually develop acutely as red, raised,
tion of tiredness, persistent cramp, or squeezing pain involving the slightly indurated and tender cords. Thrombophlebitis in TAO is
sole of the foot; however, typically, diagnosis of ischaemic pain is not confined to varicose veins and often mimics erythema nodo-
delayed because abnormal distal pulses are often misinterpreted or sum. The diagnosis of superficial venous thrombosis is confirmed
not recognized at first hand. Moreover, blood pressures at the ankle by biopsy showing venous obstruction and the inflammatory
are often normal. This delay in diagnosis has harmful consequences nature of the lesion. This migrating phlebitis or ‘phlebitis saltans’ in
in terms of functional outcome of the patient and must be avoided a young patient is highly suggestive of thromboangiitis (Bollinger
by proper education of physicians and podiatrists. As a result, et al. 1979; Mills 1994). Sometimes there is a topographic relation-
as many as 70% of patients present with critical ischaemia when ship between the inflammatory venous process and the occurrence
the diagnosis of TAO is finally made. Rest pain is associated with of arterial occlusions.
objective evidence of severe ischaemia of the forefoot. The clinical Visceral arterial involvement has been described during active
course of ischaemic lesions follows a typical pattern: an excruciat- TAO; however, such arterial lesions are unusual and it is necessary
ingly painful vesicle appears on the pulp of one toe with intense to exclude another vasculitis or an association with atherosclerotic
hyperaemia and hypersensitivity of the surrounding skin. This lesions. Since Buerger’s original description of mesenteric involve-
vesicle progresses into a small punched-out painful ulcer, which ment in four patients, anecdotal reports of intestinal involvement
enlarges and involves the nail-bed, without showing any tendency have been published (Adem et al. 2002; Arkkila et al. 2001; Buerger
to heal. Lesions then become infected and may evolve into distal 1908; Iwai 1998; Sauvaget et al. 1996). Visceral artery occlusions
gangrene and tissue loss (Figure 37.1). In contrast to the limited, may occur at any time in the course of the disease and may even
almost benign, appearance of the initial lesion, rest pain is excru- be the first manifestations. Digestive ischaemia may manifest as
ciatingly intense. It persists for hours and is increased by lying flat. abdominal pain, diarrhoea, weight loss, or melena. Intestinal per-
Symptomatic involvement of the upper limbs is found in 40 foration and mesenteric infarction may occur. Histopathological
to 50% of patients (Olin et al. 1990). An abnormal Allen test in a findings are usually necessary to diagnose TAO, thus the diagnosis
young smoker with ischaemic lesions of the lower limbs is sugges- is difficult and the prognosis poor (Cho et al. 2003). Hence unex-
tive of TAO (Olin and Shih 2006). Routine angiographic evaluation plained abdominal pain, even at its onset and when isolated, should
(Hirai and Shionoya 1979) showed that arterial occlusions of the evoke the possibility of Buerger’s disease in young male smokers
upper limbs were present in 90% of patients. As for the lower limbs, (Adem et al. 2002). Transient ischaemic attacks or ischaemic stroke
CHAPTER 37 thromboangiitis obliterans (buerger’s disease) 509

may occur (No et  al. 2005), with angiographic lesions that are most important diagnostic criterion is the smooth and regular,
identical to those present in the limbs. Coronary involvement is non-atherosclerotic nature of the arterial wall both at the site of,
extremely rare, if it exists (Fiessinger 1996). and proximally to arterial occlusions (Hagen and Lohse 1984).
The small and medium-sized arteries are affected in a segmen-
Systemic symptoms tal and often bilateral manner. In the legs, infrapopliteal lesions
predominate, affecting one or several vascular beds, but particu-
In 1999, we reported, from a retrospective study, the prevalence and larly the anterior and posterior tibial arteries. In the upper limbs,
nature of articular involvement in patients with TAO (Puéchal et al. the lesions primarily affect the radial and cubital arteries, the
1999). Arthritis was found in 11 of 88 patients (12.5%). Usually, palmar arcades, and the digital arteries (Figure 37.2) (Dimmick
rheumatic manifestations occur in the preocclusive phase preced- et al. 2012).
ing the first ischaemic symptoms by a mean of ten years. Patients Angiography remains the diagnostic gold standard. Distal arte-
present with recurrent episodes of non-erosive arthritis of the large rial occlusions, which may be segmental, are seen with normal
joints, with transient, migratory, single-joint episodes accompanied arteries proximal to the occlusion sites. Typically, ‘corkscrew’
by local inflammatory signs. The wrists and knees are the most fre- shaped collaterals develop around areas of occlusion (Figure 37.3)
quently involved joints, and the duration of symptoms ranges from (Piazza and Creager 2010). Corkscrew collaterals are not pathog-
two to 14 days. To recognize TAO at this stage is a challenge for the nomonic of TAO (Dimmick et al. 2012), but their presence may
rheumatologist. However, a diagnosis of TAO should be considered be useful in predicting the occurrence of ischaemic ulcers (Fujii
in otherwise healthy young smokers with intermittent or palindro- et al. 2010).
mic rheumatism, and distal pulses should be palpated regularly
(Puéchal and Fiessinger 2007). Arthritis disappears definitively
with the appearance of limb ischaemia. A few cases of undifferenti- Laboratory findings
ated HLA-B27 spondyloarthropathies have also been reported in TAO cannot be diagnosed by clinical laboratory testing. Indeed, it
patients with TAO (Puéchal et al. 1999). Nevertheless, in cases in is the normal or negative nature of biological tests that is valuable in
which intermittent joint manifestations are observed before the eliminating other conditions. Acute-phase reactants are normal or
appearance of ischaemic signs, autoimmune diseases, and other slightly elevated, even during active vasculitis except with extensive
types of vasculitis should be ruled out first. A search for the cardi- trophic lesions. In persons with diabetes mellitus, Buerger’s disease
nal signs of autoimmune diseases, elevated acute-phase reactants, may usually be excluded whereas modest hyperlipidaemia does not
and specific immunological tests generally lead to the diagnosis. necessarily exclude the diagnosis. Complete blood count is needed
to exclude a myeloproliferative syndrome. Screening for hyperco-
Diagnostic investigations agulability is negative. An increased prevalence of anticardiolipin
antibodies associated with increased morbidity has been reported
The diagnosis of TAO is rendered difficult by the lack of specific (Maslowski et  al. 2002); however, the relationship between TAO
clinical, radiological, and biological features, the rarity of histo- and anticardiolipin antibodies is unclear and needs to be confirmed
pathological evidence of inflammatory vascular lesions (Olin 2000; (Olin 2002; Puéchal et al. 1993).
Wessler et  al. 1960), and the lack of validated or internationally
accepted diagnostic criteria. The diagnosis is therefore made at the
end of investigations aiming to eliminate differential diagnoses, and
to search for other evocative signs of the disease. The distal nature
of the thromboangiitis and the involvement of the upper limbs are
two major arguments for differentiating TAO from atherosclerotic
arterial disease.

Vascular evaluation
Ankle systolic blood pressures are often normal for arterial occlu-
sions below the level of the popliteal artery; however, a gradient
between ankle and toe systolic pressures is generally present, indi-
cating occlusion of foot arteries. Measurement of toe systolic pres-
sure and of transcutaneous oxygen pressure in the ischaemic area
confirm the existence of critical ischaemia and may be of prognostic
value (European Working Group on Critical Leg Ischemia 1991).
Detailed assessment of the vascular bed is needed for the diag-
nosis and treatment of patients with TAO. Duplex ultrasonography,
intra-arterial contrast angiography, magnetic resonance angiogra-
phy, and computed tomography angiography may be performed
to exclude proximal atherosclerotic lesions, a potential source of
embolism, and to define the anatomy and extent of vascular lesions
(Collins et al. 2007). Fig. 37.2 Angiogram of a left hand showing multiple arterial occlusions: radial
Arterial occlusions almost always occur in more than one artery, deep palmar arch, and several digital arteries. On the fifth finger, arterial
limb, and often in all four, even if not clinically apparent. The occlusions are segmental and on the index finger a corkscrew collateral is visible.
510 SECTION 5 vasculitic diseases and syndromes and related disorders

(a) (b)

Fig. 37.4 Histological section (Masson’s trichrome staining) of a radial artery


with chronic occlusion in a patient with thromboangiitis obliterans showing
signs of intraluminal neovascularization. The media and internal elastic lamina are
preserved and almost normal. (Courtesy of Professor P. Bruneval, Department of
Pathology, HEGP, APHP, Paris, France.)

vasculitides (Bruneval and Fiessinger 1999). The disease affects the


(c) small and medium-sized (1 to 5 mm in diameter) arteries and veins
in a segmental and multifocal manner. Macroscopically, occluded
vessel segments appear to be contracted and indurated and inter-
spersed among normal vessel segments, with relatively clear-cut
demarcation of both involved and normal segments. Arteries and
veins may be bound into a rather firm cord. In most cases, the
microscopic appearance is that of an organized and recanalized
thrombus in a muscular artery. The media and internal elastic
lamina are close to normal, without cellular infiltration. Focal cal-
cifications may be present. Fibrosis of the adventitia may extend
around the artery and envelop veins and nerves. The lesions are
not specific and vary according to the duration of evolution of
the disease. In the active phase (Figure 37.5), arterial and venous
lesions are characterized by an association of thrombosis and
inflammation. These thrombi are often occlusive and sometimes
display moderate non-specific inflammatory infiltrates, consisting

Fig. 37.3 Angiogram of the right limb (a, b) and foot (c), in a patient with
TAO. Segmental occlusions (arrows) of the tibial and peroneal arteries are visible
with a normal proximal popliteal artery (a). Extensive tortuous ‘corkscrew’
collaterals have developed to bypass the arterial occlusions (b). A lateral oblique
angiographic projection of the foot (c) shows important distal arterial defects: the
plantar arteries, the dorsalis pedis artery, and the pedal–plantar loop are occluded.
A patent segment of the plantar artery is fed and bypassed by musculocutaneous
collaterals originating from the posterior leg.

Pathological findings
TAO is an inflammatory disease of the arteries and veins charac-
Fig. 37.5 Histological arterial section (Masson’s trichrome staining) showing
terized by highly cellular and inflammatory occlusive thrombi,
intense angiitis with thrombosis (the arterial lumen is occluded by a cellular
primarily of the distal extremities. However, the architecture of thrombus). Notably, the inflammatory process is spreading to the vessel wall
the vascular wall is preserved and there is no associated vascular without altering its architecture. (Courtesy of Professor P. Bruneval, Department of
wall necrosis (Figure  37.4), in contrast to all of the large-vessel Pathology, HEGP, APHP, Paris, France.)
CHAPTER 37 thromboangiitis obliterans (buerger’s disease) 511

mostly of polymorphonuclear leucocytes, mononuclear cells, Table 37.2 Point scoring system in the diagnosis of thromboangiitis
and rare multinucleated giant cells. Arterial infiltration is mostly obliterans, as proposed by Papa et al. (1996)
observed in the intima and the thrombus. Macrophages express-
ing HLA-DR and dendritic cells are preferentially located in the Positive points
intima (Kobayashi et al. 1999). T lymphocytes infiltrate the vascu- Age at onset Less than 30/30–40 years +2/+1
lar wall and immunophenotyping demonstrated that CD4+ cells Foot or arch claudication Present/history of +2/+1
were the predominant cell type, and were located in the intima and Upper extremity Symptomatic/asymptomatic +2/+1
Migrating superficial vein Present/history of +2/+1
the outer media of occluded arteries. These findings suggest that
thrombosis
endothelial cell injury is related to early arterial occlusion through
Raynaud’s phenomenon Present/history of +2/+1
an immunological-based mechanism (Lee et  al. 2003). Linear Angiography; biopsy If typical both/either +2/+1
deposits of immunoglobulins and complement are found along the
length of the internal elastic lamina, which, like the media, retains Negative points
its integrity. No fibrinoid necrosis of the arterial wall is observed. Age at onset 45–50/more than 50 years −1/−2
Sex, smoking Female, non smoker −1/−2
The appearance of panvasculitis described by Buerger (1908), in
Location Single limb/no LE involved −1/−2
which inflammatory infiltrate extends to all the layers of the vessel
Absent pulses Brachial/femoral −1/−2
wall without mutilation, is rarely observed in the arteries. Biopsy Arteriosclerosis, Discovered after diagnosis −1/−2
of a subcutaneous nodule can be used to confirm the presence of diabetes, hypertension, 5.1–10 years/2.1– 5 years later
superficial thrombophlebitis during the active phase, with the same hyperlipidaemia
occlusive thrombotic lesions and various degrees of inflammation
extending to the entire venous wall. Clinical and non-clinical weighted characteristics of TAO are combined with other
characteristics and conditions that may question the diagnosis. Results are expressed as
levels of probability of TAO.
Diagnostic criteria LE, lower extremity.

The diagnosis of TAO is usually founded on a probabilistic


approach. In 1970, Mozes (Mozes et al. 1970) described such cri- also included female patients and improved the specificity of diag-
teria for diagnosing Buerger’s disease:  a patient was considered nosis. There were six clinical and histopathological elements that
with a low probability to have thromboangiitis if he was a young contributed to a positive score and five negative clinical points. In a
male smoker with distal arterial occlusions and with no evidence young patient with distal lower limb ischaemia with no evidence of
of arteriosclerosis, hyperlipidaemia, diabetes, collagen disease, arterioclerosis, hyperlipidaemia, diabetes collagen disease, haema-
haematological disorder, or a potential source of proximal emboli. tological disorder, or a potential source of proximal emboli, scoring
However, when any one of the three recognized hallmarks of the was completed after all points were assigned. The probability of the
disease (superficial vein phlebitis (phlebitis saltans), involvement diagnosis of thromboangiitis was then obtained by the arithmetic
of the upper limb distal arteries, and Raynaud’s phenomenon) was sum of positive and negative points (Table 37.3). The possibility to
present, the diagnosis of Buerger’s disease was considered probable, include patients with exclusion criteria developing after diagnosis
and it was considered definite if two or more of the hallmark clini- needs a regular re-evaluation of the initial diagnosis. More recently,
cal signs were present. In 1996, we proposed (Fiessinger 1996) to Adar et  al. suggested a redefinition of these diagnostic criteria,
use Mozes criteria regardless of the sex of the patient (Table 37.1) including also thrombophilic states (Adar et al. 2000). Olin’s algo-
with an important supplemental exclusion criterion:  the entrap- rithm (Olin and Shih 2006) allows a more pragmatic approach to
ment syndrome of the popliteal artery. More recently, Adar’s group the diagnosis of Buerger’s disease founded on exclusion diagnoses.
proposed a point score system (Table 37.2) (Papa et al. 1996). This Arteriosclerosis obliterans: while a diagnosis of insulin-
dependent diabetes clearly excludes Buerger’s disease, the relation-
ship between dyslipidaemia and the atheromatous nature of arterial
Table 37.1 Thromboangiitis obliterans: diagnostic criteria (Fiessinger lesions is not clear. Thus, moderate hyperlipidaemia probably
1996; Mozes et al. 1970) remains compatible with a diagnosis of thromboangiitis. Rather
than ruling out patients with atherosclerosis risk factors, the key
A young smoker with distal lower limb ischaemia point is to confirm the atheromatous nature of lesions. Frank ath-
with no evidence of: erosclerotic lesions are usually easily found by arterial evaluation.
diabetes mellitus
atheromatous lesions
Table 37.3 Point scoring system as proposed by Papa et al. (1996)
potential source of embolism
popliteal entrapment syndrome
autoimmune disease Number of points Probability of diagnosis
hypercoagulability 0–1 Excluded
with 2 or more of the 3 following symptoms:
2–3 Suspected, probability low
superficial thrombophlebitis
arterial upper limb involvement 4–5 Probable, probability medium
Raynaud’s phenomenon
6 or more Definite, probability high
has a definite Buerger’s disease
If 1 only of the 3 symptoms is present the diagnosis is probable The likelihood of TAO is determined by adding positive and subtracting negative points
obtained in Table 37.2. The final score indicates level of probability of diagnosis of TAO.
512 SECTION 5 vasculitic diseases and syndromes and related disorders

The main risk is failure to identify proximal lesions, which could of new ischaemic lesions is generally linked to continued smok-
be sources of distal emboli. Thus investigation of the subrenal aorta ing (Dehaine-Bamberger et al. 1993; Olin 2000; Piazza and Creager
is always necessary. Protruding plaques in the thoracic aorta have 2010). In a retrospective series of 110 patients, 19% of those who
been reported as a potential source of emboli (Amarenco et  al. continued smoking required a major amputation, versus none of
1992; Tunick et al. 2002) and could explain distal arterial occlusions those that stopped (Ohta et al. 2004).
both in the lower and upper limbs. Consequently, transoesopha- The requirement of amputations in other published series is simi-
geal echocardiography, which provides good visualization of the lar. At the end of a 5-year follow-up period, one-quarter of patients
thoracic aorta, should be considered in patients with unexplained are likely to have had an amputation. After 10 years, almost half
distal arterial occlusions. of patients had undergone one or more amputations (Mills 1994;
Embolism of cardiac origin: there are numerous cardiac sources Ohta and Shionoya 1988). The risk of a major amputation (above
of peripheral embolism. Transoesophageal echocardiography is or below the knee or even the hand) remains high, up to 20% at
superior to transthoracic echocardiography in the detection of a 10  years. Remarkably, the risk of amputation is continuingly
cardiac sources of embolism (Cujec et al. 1991). increased in ongoing smokers, but disappeared by 8  years after
Popliteal vascular entrapment syndrome is a rare congeni- smoking cessation (Cooper et al. 2004).
tal disorder resulting from an abnormal relationship between the
popliteal artery and vein, and adjacent muscles (Sinha et al. 2011). Treatment
The syndrome is often bilateral and can cause distal arterial emboli
and venous thrombosis. The diagnosis can be suspected on the Conservative therapy
basis of clinical symptoms and by sonography. Duplex sonogra- The major role played by tobacco abuse in the development, pro-
phy (Cormier et  al. 1985)  and angiogram are very suggestive of gression, and prognosis of TAO accounts for the requirement of
the diagnosis when they show characteristic medial deviation or discontinuation of tobacco use as the cornerstone of therapeutic
compression of the artery. MR imaging (Fermand et al. 1990) is the management of patients with TAO. All possible means should be
method of choice for demonstrating the abnormal band of muscle used to encourage patients to give up the use of tobacco in all its
crossing the vessels. forms, completely and definitively. Patient education is important,
Autoimmune diseases: vasculitis of small vessels, Raynaud’s but only 43–70% of cases manage to give up smoking (Fiessinger
phenomenon, and venous thrombosis are well-known manifesta- 1996; Olin 2000). Patients should be reassured that if they manage
tions of vascular diseases such as scleroderma and systemic lupus to give up smoking completely, the disease will go into remission
erythematosus. Usually, the clinical picture and immune abnormal- and amputation will be avoided. Psychological help may be useful
ities enable diagnosis (Shionoya 1993). In contrast, the exclusion of in certain cases (Hofer-Mayer et al. 1995).
vascular Behçet’s disease is often far more difficult but it frequently Local care is the other main component of therapeutic man-
comprises (Matsumoto et  al. 1991)  venous thrombosis, arterial agement (Fiessinger 1996). The aim is to obtain cleansing of the
aneurysm (aorta), and occlusions of large arteries. Nevertheless, wound with the creation of sufficient new tissue to enable heal-
we have seen patients with Behçet’s disease with only superficial ing. Various dressings, ointments, and healing agents should be
thrombophlebitis and distal arterial occlusions. applied with extreme care to these atonic, extremely painful lesions.
Hypercoagulability syndromes: myeloproliferative disorders in Wound cleansing with a scalpel and/or a curette, and Vaseline com-
young adults may cause arterial and venous occlusion. For example, presses form the basis of local treatment. Major factors in proper
the risk of thrombosis is increased in patients carrying a homozy- care include adequate analgesia in order to allow dressings to be
gous JAK2 mutation (De Stefano et al. 2008; De Stefano et al. 2010), done, and bed rest to reduce leg oedema. It is often useful, in this
and thrombotic symptoms may be manifestations of erythromela- context, to carry out the first dressing under general analgesia
lgia. Antiphospholipid syndrome, and antithrombin III, protein C, (Lassalle-Fontaine et  al. 1990). When there are clinical signs of
and protein S deficits have also been associated with venous and wound infection, systemic antibiotics are necessary. Topical antibi-
sometimes arterial occlusion (Eldrup-Jorgensen et al. 1989). otics should be avoided.
Disturbance to the immune system described during the course
of the disease (Adar et al. 1983) and the inflammatory nature of
Outcome and prognosis the vascular lesions could justify the use of immunosuppressive
The clinical course of TAO is characterized by acute exacerbation therapy. Bollinger et al. (1979) attempted the use of high doses of
phases separated by periods of remission which may last several aspirin (3 g/day) or, when this regimen failed, corticosteroids or
years. The trophic lesions progress during flare-ups, either by immunosuppressive agents. However, this strategy rarely seems
being more proximal or by affecting a limb that was not previously justified in a disease in which inflammatory manifestations are
affected. The need for repeated amputations is a marker of severity often lacking or only secondary to cutaneous lesions. Non-steroidal
of the disease. The life-expectancy of patients with TAO has been anti-inflammatory drugs are the treatment of choice for superficial
reported as normal (McPherson et al. 1963), with a survival rate of venous thrombosis.
85% up to 25 years (Ohta et al. 2004). However, an excessive late Despite the constant finding of both arterial and venous thrombo-
mortality was ascertained in a study of 111 patients with a mean sis, relationship between coagulation abnormalities and the patho-
follow-up of 15 years (Cooper et al. 2004), especially for persons genesis of TAO have never been clearly defined (Adar et al. 2000).
who did not discontinue tobacco use. Thus, the question of the use of antithrombotic drugs remains
Tobacco abuse plays a key role in the progression and prognosis unanswered in the treatment of Buerger’s disease (Olin 2002).
of the disease. Flare-ups may resolve and ulcers may heal only after The role of tobacco together with the frequency of vasomotor
the patient has stopped any tobacco or nicotine use. The occurrence events preceding the disease have led to consideration of the role
CHAPTER 37 thromboangiitis obliterans (buerger’s disease) 513

of spasm in the pathophysiology of TAO (Long and Fiessinger need for an analgesic at the end of follow-up. High-dose oral ilo-
1989; Gore and Burrows 1958). Silbert (1930), using daily intrave- prost was not significantly more effective than placebo.
nous injections of hypertonic saline solution, reported a dramatic Calcium inhibitors are often advocated even though there is no
improvement in prognosis in comparison with an untreated his- proof of their efficacy (Olin and Shih 2006).
torical series. These findings were the basis of induced hypervol- In a clinical pilot study, 12 patients were treated with bosentan,
aemia in these patients (Amtoft 1973; Housset and Leduc 1963). a competitive antagonist of endothelin-1, with encouraging results.
Infusion of 500 ml of intravenous solutions with dextran function Clinical improvement was observed in 12 of the 13 extremities (De
or 3 litres of normal saline over 3 hours, increases perfusion pres- Haro et al. 2012), associated with a radiologically assessed increase
sure and blood flow in the ischaemic limb. It may also provide a in distal flow and improved endothelial function outcome.
useful vector for subsequent antibiotic treatment.
Epidural spinal cord stimulation has been proposed in patients Surgical treatment
who have exhausted all other pain relieving therapeutic options Sympathectomy has often been advocated as the treatment of
(Swigris et al. 1999). In a retrospective study of 29 patients with spi- choice in thromboangiitis obliterans. However, its value in pre-
nal cord stimulation (Donas et al. 2005), significant microcircula- venting amputation is difficult to assess (Mills 1994; Olin and Shih
tory and clinical benefits were reported, despite continuing tobacco 2006). In the series of Nakata et al. (1975), blood flow in the ante-
exposure in more than 80% of patients over a 4-year follow-up rior tibial muscles did not increase within 5 weeks postsympathec-
period. These findings are still awaiting confirmation in a prospec- tomy. At the present time, sympathectomy may be considered after
tive randomized setting. failure of medical treatment before, or associated with, an amputa-
Prostacycline derivatives have been evaluated in case–control tion (Mills 1994). Surgical revascularization for Buerger’s disease
studies. In the first (Nizankowski et  al. 1985), 30 patients with is rarely possible due to the diffuse character of vascular damage
ischaemic ulcers were treated by continuous intra-arterial infusion and the distal nature of arterial lesions. For popliteal occlusions,
for 72 hours, with either with placebo or epoprostenol (PGI2); 15 despite poor long-term patencies of infrapopliteal bypass grafts,
of the patients had Buerger’s disease. The treated group showed a short-term patency is usually sufficient to allow healing of distal
significant improvement in trophic disorders and pain at both the tissue loss (Nishikimi 2000; Sayin et al. 1993) and may thus be dis-
fourth and sixth week following infusion. In another randomized cussed in some cases. More recently, endovascular distal arterial
trial (Fiessinger et al. 1990), 152 patients with Buerger’s disease and recanalization was reported in 17 consecutive patients (20 limbs)
pain at rest, with or without tissue loss, received a daily perfusion of with Buerger’s disease (Graziani et al. 2012). Endovascular treat-
iloprost versus placebo plus aspirin (Table 37.4). After 21 to 28 days ment was a technical success in 95% of the treated limbs with a limb
of perfusion, the trophic lesions had healed, or the pain had disap- salvage rate of 100% at 2-years follow-up. This case series suggests
peared in 85% of the patients on iloprost and 17% of the patients on that endovascular procedures could be an interesting alternative to
aspirin. At 6 months, 88% of the patients treated with iloprost had surgical revascularization.
responded to treatment, versus only 21% of those treated with aspi-
rin. Furthermore, amputation was necessary in 18% of the patients Angiogenesis therapy
in the aspirin group and in only 6% of the patients in the iloprost A new therapeutic approach was introduced with the use of gene
group. The results of a European study, which compared two doses transfer to induce therapeutic angiogenesis in patients with periph-
of oral iloprost to placebo, were less impressive (The European TAO eral artery disease (De Haro et al. 2009; Isner et al. 1995). In 1998,
Study Group 1998). No significant difference was found between Isner et  al. (1998) published preliminary encouraging results in
groups for the primary end-point, which was total healing of lesions six patients with Buerger’s disease receiving intramuscular injec-
at any time point. Nevertheless, low-dose iloprost was significantly tions of vascular endothelial growth factor (VEGF). However, these
more effective than placebo in relieving pain at rest without the results were not confirmed and a recent review concluded that no
clinical study has shown significant benefit of gene therapy over
placebo (Mughal et al. 2011).
Table 37.4 Early and late results of intravenous perfusion of iloprost Treatment of peripheral arterial disease using stem and progeni-
versus placebo combined with low-dose aspirin in 133 patients with tor cell therapy seems a more promising tool for the treatment of
thromboangiitis obliterans included in the Milestone’s study (Fiessinger patients with critical limb ischaemia (Lawall et al. 2011). In 2002,
et al. 1990) (analysis on intention-to-treat) Tateishi-Yayuma et  al. published impressive results in patients
with critical ischaemia treated by autologous bone marrow cell
Iloprost Aspirin transplantation (Tateishi-Yuyama et  al. 2002). Yet, there have
been only a few studies in patients with TAO. Kim et al. (2011),
At day 28 (n=68) (n=65)
in a preliminary study of seven patients with TAO, suggest that
Responders 58 (85%) 11 (17%) human cord blood-derived mononuclear cells might provide a
Total relief of rest pain 43 (63%) 18 (28%)
Complete healing of all trophic changes safe source of stem cells. Motukuru et al. treated 38 patients with
18/52 (35%) 6/46 (13%)
Amputations (minor) TAO by injection of bone marrow mononuclear cells into the
2 2
calf muscles (Motukuru et al. 2008). At 6 months, three patients
At 6 months from start of treatment (n=51) (n=44) underwent a major amputation, the others had improvement in
Responders 45 (88%) 12 (27%) their ulcer healing, an increase of the ankle-brachial index, and
Minor amputations 0 1
an improvement in transcutaneous oxygen release (tcpO2). Of the
Major amputations 1 (thigh) 5
eight patients treated with bone marrow cell transplantation and
Responders were defined by ulcer healing or relief of ischaemic pain. followed for a mean 684  days, four experienced serious clinical
514 SECTION 5 vasculitic diseases and syndromes and related disorders

adverse events (Miyamoto et al. 2006). One patient died of sudden Combemale, P., Consort, T., Denis-Thelis, L., Estival, J.L., Dupin, M., and
death 20 months after transplantation, three patients had worsen- Kanitakis, J. (2005). Cannabis arteritis. British Journal of Dermatology,
ing or recurrence of ischaemic symptoms, and the fourth devel- 152, 166–9.
oped a transient arteriovenous shunt by suspected inappropriate Cooper, L.T., Tse, T.S., Mikhail, M.A., McBane, R.D., Stanson, A.W., and
Ballman, K.V. (2004). Long-term survival and amputation risk in
angiogenesis. Thus, more studies with careful long-term monitor- thromboangiitis obliterans (Buerger’s disease). Journal of the American
ing are necessary to demonstrate safety and efficacy of these new College of Cardiology, 44, 2410–11.
therapeutic approaches. Cormier, J.M., Laurian, C.L., Fichelle, J.M., Bardi, K., Franceschi, C.L., and
Luizy, F. (1985). Artére poplitée piègée. Apport de l’exploration ultra-
Conclusion sonographique. Presse Médicale, 14, 2183–5.
Cujec, B., Polasek, P., Voll, C., and Shuaib, A. (1991). Transesophageal echo-
Thromboangiitis obliterans (Buerger’s disease) is currently cardiography in the detection of potential cardiac source of embolism in
defined as a vasculitis that most commonly affects the small and stroke patients. Stroke, 22, 727–33.
medium-sized arteries and veins of the extremities. It affects Darnige, L., Helley, D., Fischer, A.M., Emmerich, J., Smadja, D.M., and
young smokers and cannabis addiction is often a cofactor asso- Fiessinger, J.N. (2010). Platelet microparticle levels:  A  biomarker of
ciated with the use of tobacco. The aetiology of TAO remains thromboangiitis obliterans (Buerger’s disease) exacerbation. Journal of
undetermined; however, some clinical and haemodynamic data Cellular and Molecular Medicine, 14, 449–51.
suggest an increase in vasomotor tone. The diagnostic criteria of Dehaine-Bamberger, N., Amar, R., Touboul, C., Emmerich, J., and Fiessinger,
TAO are not validated and, thus, the diagnosis of TAO remains J.-N. (1993). Maladie de Buerger, aspects cliniques et pronostiques 83
observations. Presse Médicale, 22, 945–8.
founded on exclusion criteria. Discontinuing tobacco abuse is
De Haro, J., Acin, F., Bleda, S., Varela, C., and Esparza, L. (2012). Treatment
the key of the prognosis of the disease, and in spite of new thera- of thromboangiitis obliterans (Buerguer’s disease) with bosentan. BMC
peutic approaches, the prevalence of major amputations remains Cardiovascular Disorders, 12, 1–5.
unacceptably high. De Haro, J., Acin, F., Lopez-Quintana, A., Florez, A., Martinez-Aguilar, E.,
and Varela, C. (2009). Meta-analysis of randomized, controlled clinical
References trials in angiogenesis: gene and cell therapy in peripheral arterial dis-
Adar, R., Papa, M.Z., Halpern, Z., et al. (1983). Cellular sensitivity to col- ease. Heart Vessels, 24, 321–8.
lagen in thromboangiitis obliterans. New England Journal of Medicine, De Stefano, V., Za, T., Rossi, E., et al. (2008). Recurrent thrombosis in patients
308, 1113–6. with polycythemia vera and essential thrombocythemia: incidence, risk
Adar, R., Papa, M.Z., and Schneiderman, J. (2000). Thromboangiitis oblit- factors, and effect of treatments. Haematologica, 93, 372–80.
erans:  an old disease in need of a new look. International Journal of De Stefano, V., Za, T., Rossi, E., et  al. (2010). Increased risk of recurrent
Cardiology, 75, S167–70. thrombosis in patients with essential thrombocythemia carrying the
Adem, C., Benamouzig, R., Royer, I., Chapel, N., and Martin, A. (2002). homozygous JAK2 V617F mutation. Annals of Hematology, 89, 141–6.
[Buerger’s disease or thromboangiitis obliterans revealed by an enteric Dezellus, P.H., Capron, L., Fiessinger, J.-N., and Housset, E. (1982).
ischemia. Case report and literature review]. Gastroentérologie Clinique Artériopathie juvénile et sympathomimètiques nasaux. Presse Médicale,
et Biologique, 26, 409–11. 11, 2231.
Amarenco, P., Duyckaerts, C., Tzourio, C., Hénin, D., Bousser, M.G., and Dimmick, S.J., Goh, A.C., Cauzza, E., et al. (2012). Imaging appearances of
Hauw, J.J. (1992). The prevalence of ulcerated plaques in the aortic arch Buerger’s disease complications in the upper and lower limbs. Clinical
in patients with stroke. New England Journal of Medicine, 326, 221–5. Radiology, 67, 1207–11.
Amtoft, A. (1973). The effect of destran-40 on capillary blood flow and capil- Donas, K.P., Schulte, S., Ktenidis, K., and Horsch, S. (2005). The role of
lary diffusion capacity in the anterior tibial muscle of subjects with inter- epidural spinal cord stimulation in the treatment of Buerger’s disease.
mittent claudication. Scandinavian Journal of Clinical and Laboratory Journal of Vascular Surgery, 41, 830–6.
Investigation, 31, 263–8. Eichhorn, J., Sima, D., Lindschau, C., et al. (1998). Antiendothelial cell anti-
Arkkila, P.E.T., Kahri, A., and Farkkila, M. (2001). Intestinal type of throm- bodies in thromboangiitis obliterans. American Journal of the Medical
boangiitis obliterans (Buerger disease) preceding symptoms of severe Sciences, 315, 17–23.
peripheral arterial disease. Scandinavian Journal of Gastroenterology, 36, Eldrup-Jorgensen, J., Flanigan, P., Brace, L., et al. (1989). Hypercoagulable
669–72. states and lower limb ischemia in young adults. Journal of Vascular
Azizi, M., Boutouyrie, P., Bura-Riviere, A., Peyrard, S., Laurent, S., and Surgery, 9, 334–41.
Fiessinger, J.N. (2010). Thromboangiitis obliterans and endothelial European Working Group on Critical Leg Ischemia (1991). Second European
function. European Journal of Clinical Investigation, 40, 518–26. consensus document on chronic critical leg ischemia. Circulation, 84
Bollinger, A., Hollmann, B., Schneider, E., and Fontana, A. (1979). (Suppl. IV), IV-1–IV-26.
Thromboangiitis obliterans:  Diagnose und Therapie im Lichtneuer Fermand, M., Houlle, D., Fiessinger, J.-N., and Cormier, J.M. (1990).
immunologischer Befunde. Schweizerische Medizinische Wochenschrift, Entrapement of the popliteal artery: MR findings. American Journal of
109, 537–43. Roentgenology, 154, 425–6.
Bruneval, P. and Fiessinger, J.-N. (1999). Vascularite des gros troncs. Annals Fiessinger, J.-N. (1996). Buerger’s disease or thromboangiitis obliterans. In A
of Pathology, 19, 223–37. textbook of vascular medicine (ed. J.E. Tooke and G.D. Lowe), pp. 275–
Buerger, L. (1908). Thrombo-angiitis obliterans:  a study of the vascular 86. Arnold, London, Sydney, Auckland.
lesions leading to presenile spontaneous gangrene. American Journal of Fiessinger, J.-N., Schäfer, M., for the TAO Study (1990). Trial of iloprost
the Medical Sciences, 139, 567–80. versus aspirin treatment for critical limb ischaemia of thromboangiitis
Cho, Y.P., Kwon, Y.M., Kwon, T.W., and Kim, G.E. (2003). Mesenteric obliterans. Lancet, 335, 555–7.
Buerger’s disease. Annals of Vascular Surgery, 17, 221–3. Fujii, Y., Soga, J., Nakamura, S., et al. (2010). Classification of corkscrew col-
Collins, R., Burch, J., Cranny, G., et al. (2007). Duplex ultrasonography, mag- laterals in thromboangiitis obliterans (Buerger’s disease). Circulation
netic resonance angiography, and computed tomography angiography Journal, 74, 1684–8.
for diagnosis and assessment of symptomatic, lower limb peripheral Gore, I. and Burrows, S. (1958). Reconsideration of the pathogenesis of
arterial disease: systematic review. BMJ, 334, 1257–65. Buerger’s disease. American Journal of Clinical Pathology, 29, 319–30.
CHAPTER 37 thromboangiitis obliterans (buerger’s disease) 515

Graziani, L., Morelli, L., Parini, F., et  al. (2012). Clinical outcome after McPherson, J.R., Juergens, J.L., and Gifford, R.W. (1963). Thromboangiitis
extended endovascular recanalization in Buerger’s disease in 20 con- obliterans and arteriosclerosis obliterans. Clinical and prognostic differ-
secutive cases. Annals of Vascular Surgery, 26, 387–95. ences. Annals of Internal Medicine, 59, 288–96.
Gulati, S.M., Singh, K.S., Thusoo, T.K., and Saha, K. (1979). immunological Mills, J.L. (1994). Buerger’s disease: current status. Vascular Medicine Review,
studies in thromboangiitis obliterans. Journal of Surgical Research, 27, 5, 139–50.
287–93. Miyamoto, K., Nishigami, K., Nagaya, N., et al. (2006). Unblinded pilot
Hagen, B. and Lohse, S. (1984). Clinical and radiologic aspects of Buerger’s study of autologous transplantation of bone marrow mononuclear
disease. Cardiovascular and Interventional Radiology, 7, 283–93. cells in patients with thromboangiitis obliterans. Circulation, 114,
Hirai, M. and Shionoya, S. (1978). Intermittent claudication in the foot and 2679–84.
Buerger’s disease. British Journal of Surgery, 65, 210–3. Motukuru, V., Suresh, K.R., Vivekanand, V., Raj, S., and Girija, K.R. (2008).
Hirai, M. and Shionoya, S. (1979). Arterial obstruction of the upper limb Therapeutic angiogenesis in Buerger’s disease (thromboangiitis oblit-
in Buerger’s disease: its incidence and primary lesion. British Journal of erans) patients with critical limb ischemia by autologous transplanta-
Surgery, 66, 124–8. tion of bone marrow mononuclear cells. Journal of Vascular Surgery, 48
Hofer-Mayer, T., Adler, R.H., Minder, C., Mahler, F., and Radvila, A. (1995). (Suppl.), 53S–60S.
Clinical speculation: is specificity back? this time thromboangiitis oblit- Mozes, M., Cahansky, G., Doitsch, V., and Adar, R. (1970). The association of
erans. Psychosomatic Medicine, 57, 195–201. atherosclerosis and Buerger’s disease: a clinical and radiological study.
Housset, E. and Leduc, X. (1963). Le traitement des artérites des membres Journal of Cardiovascular Surgery, 11, 52–9.
inférieurs par les perfusions intraveineuses de sérum physiologique. Mughal, N.A., Russell, D.A., Ponnambalam, S., and Homer-Vanniasinkam,
Presse Médicale, 71, 33–335. S. (2011). Gene therapy in the treatment of peripheral arterial disease.
Isner, J.M., Baumgartner, I., Rauh, G., et  al. (1998). Treatment of throm- British Journal of Surgery, 99, 6–15.
boangiitis obliterans (Buerger’s disease) by intramuscular gene trans- Nakata, Y., Suzuki, S., Kawai, S., et al. (1975). Effects of lumbar sympathec-
fer of vascular endothelial growth factor: Preliminary clinical results. tomy on thromboangiitis obliterans. Journal of Cardiovascular Surgery,
Journal of Vascular Surgery, 28, 964–73. 16, 415–25.
Isner, J.M., Walsh, K., Symes, J., et  al. (1995). Arterial gene therapy for Nishikimi, N. (2000). Fate of limbs with failed vascular reconstruction
therapeutic angiogenesis in patients with peripheral artery disease. in Buerger’s disease patients. International Journal of Cardiology, 75,
Circulation, 91, 2687–92. S183–5.
Iwai, T. (1998). Buerger’s disease with intestinal involvement. International Nizankowski, R., Krolikowski, W., Bielatowicz, J., and Szezeklik, A. (1985).
Journal of Cardiology, 66, S257–63. Prostacyclin for ischemia ulcers in peripheral arterial disease. A random
Karila, L., Danel, T., Coscas, S., Chambon, J.P., and Reynaud, M. (2004). assignement, placebo controlled study. Thrombosis Research, 37, 21–8.
[L’artérite progressive cannabique, un sous-type clinique de la throm- No, Y.J., Lee, E.M., Lee, D.H., and Kim, J.S. (2005). Cerebral angiographic
boangéite oblitérante?] Presse Médicale, 33, 21–3. findings in thromboangiitis obliterans. Neuroradiology, 47, 912–5.
Kim, A.K., Kim, M.H., Kim, S., et al. (2011). Stem-cell therapy for peripheral O’Dell, J.R., Linder, J., Markin, R.S., and Moore, G.F. (1987). Thromboangiitis
arterial occlusive disease. European Journal of Vascular and Endovascular obliterans (Buerger’s disease) and smokeless tobacco. Arthritis and
Surgery, 42, 667–75. Rheumatism, 30, 1054–6.
Kjeldsen, K. and Mozes, M. (1969). Buerger’s disease in Israel. Acta Chirurgica Ohta, T., Ishioashi, H., Hosaka, M., and Sugimoto, I. (2004). Clinical and
Scandinavica, 135, 495–8. social consequences of Buerger disease. Journal of Vascular Surgery, 39,
Kobayashi, M., Ito, M., Nakagawa, A., Nishikimi, N., and Nimura, Y. (1999). 176–80.
Immunohistochemical analysis of arterial wall cellular infiltration in Ohta, T. and Shionoya, S. (1988). Fate of ischaemic limb in Buerger’s disease.
Buerger’s disease (Endarteritis obliterans). Journal of Vascular Surgery, British Journal of Surgery, 75, 259–62.
29, 451–8. Olin, J.W. (2000). Current concepts:  thromboangiitis obliterans (Buerger’s
Lassalle-Fontaine, C., Cousin, M.T., and Fiessinger, J.-N. (1990). disease). New England Journal of Medicine, 343, 864–9.
[Amélioration des soins des troubles trophiques en pathologie vascu- Olin, J.W. (2002). Are anticardiolipin antibodies really important in throm-
laire. L’anesthésie en milieu médical.] Presse Médicale, 19, 473. boangiitis obliterans (Buerger’s disease)? Vascular Medicine, 7, 257–8.
Lawall, H., Bramlage, P., and Amann, B. (2011). Treatment of peripheral arte- Olin, J.W. and Shih, A. (2006). Thromboangiitis obliterans (Buerger’s dis-
rial disease using stem and progenitor cell therapy. Journal of Vascular ease). Current Opinion in Rheumatology, 18, 18–24.
Surgery, 53, 445–53. Olin, J.W., Young, J.R., Graor, R.A., Ruschhaupt, W.F., and Bartholomew, J.R.
Lee, T., Seo, J.W., Sumpio, B.E., and Kim, S.J. (2003). Immunobiologic analy- (1990). The changing clinical spectrum of thromboangiitis obliterans
sis of arterial tissue in Buerger’s disease. European Journal of Vascular (Buerger’s) disease. Circulation, 82 (Suppl. IV), IV-3–IV-8.
and Endovascular Surgery, 25, 451–7. Papa, M., Bass, A., Adar, R., et al. (1992). Autoimmune mechanism in throm-
Lie, J.T., Mann, R.J., and Ludwig, J. (1979). The brothers von Winiwarter, boangiitis obliterans (Buerger’s disease): The role of tobacco antigen and
Alexander (1848–1917) and Felix (1852–1931), and thromboangiitis the major histocompatibility complex. Surgery, 111, 527–31.
obliterans. Mayo Clinic Proceedings, 54, 802–7. Papa, M.Z., Rabi, I., and Adar, R. (1996). A point scoring system for the
Long, A. and Fiessinger, J.-N. (1989). Maladie de Buerger associée au phé- clinical diagnosis of Buerger’s disease. European Journal of Vascular and
nomène de Raynaud chez la femme. Presse Médicale, 18, 1438. Endovascular Surgery, 11, 335–9.
Martin-Blondel, G., Koskas, F., and Cacoub, P.D. (2011). Is thromboangii- Piazza, G. and Creager, M.A. (2010). Thromboangiitis obliterans. Circulation,
tis obliterans presentation influenced by cannabis addiction? Annals of 121, 1858–61.
Vascular Surgery, 25, 469–73. Puéchal, X. and Fiessinger, J.N. (2007). Thromboangiitis obliterans or
Maslowski, L., McBane, R., Alexewicz, P., and Wysokinski, W.E. (2002). Buerger’s disease:  challenges for the rheumatologist. Rheumatology
Antiphospholipid antibodies in thromboangiitis obliterans. Vascular (Oxford), 46, 192–9.
Medicine, 7, 259–64. Puéchal, X., Fiessinger, J.N., Kahan, A., and Menkes, C.J. (1999). Rheumatic
Matsumoto, T., Uekusa, T., and Fukuda, Y. (1991). Vasculo-Behcet’s disease: a manifestations in patients with thromboangiitis obliterans (Buerger’s
pathologic study of eight cases. Human Pathology, 22, 45–51. disease). Journal of Rheumatology, 26, 1764–8.
McLoughlin, G.A., Helsby, C.R., Evans, C.C., and Chapman, D.M. (1976). Puéchal, X., Job-Deslandre, C., Menkès, C.J., and Fiessinger, J.-N. (1993).
Association of HLA-A9 and HLA-B5 with Buerger’s disease. British Buerger’s disease and antiphospholipid syndrome. Annals of the
Medical Journal, 2(6045), 1165–6. Rheumatic Diseases, 52, 766.
516 SECTION 5 vasculitic diseases and syndromes and related disorders

Sauvaget, F., Debray, M., Hervé de Sigalony, J.P., et al. (1996). Colonic ischemia Swigris, J.J., Olin, J.W., and Mekhail, N.A. (1999). Implantable spinal cord
reveals thromboangiitis obliterans (Buerger’s disease). Gastroenterology, stimulator to treat the ischemic manifestations of thromboangiitis oblit-
110, 900–3. erans (Buerger’s disease). Journal of Vascular Surgery, 29, 928–35.
Sayin, A., Bozkurt, A.K., Tuzun, H., Vural, F.S., Erdog, G., and Ozer, M. Tateishi-Yuyama, E., Matsubara, H., Murohara, T., et al. (2002). Therapeutic
(1993). Surgical treatent of Buerger’s disease:  experience with 216 angiogenesis for patients with limb ischaemia by autologous transplan-
patients. Cardiovascular Surgery, 1, 377–80. tation of bone-marrow cells: a pilot study and a randomised controlled
Shionoya, S. (1993). Buerger’s disease:  diagnosis and management. trial. Lancet, 360, 427–35.
Cardiovascular Surgery, 1, 207–14. The European TAO Study Group (1998). Oral iloprost in the treatment of
Siguret, V., Alhenc-Gelas, M., Aiach, M., Fiessinger, J.N., and Gaussem, P. thromboangiitis obliterans (Buerger’s disease):  a double-blind, ran-
(1997). Response to DDAVP stimulation in thirteen patients with domised, placebo-controlled trial. European Journal of Vascular and
Buerger’s disease. Thrombosis Research, 86, 85–7. Endovascular Surgery, 15, 300–7.
Silbert, S. (1930). Thromboangiitis-onliterans (Buerger) V. Results of treat- Tunick, P.A., Nayar, A.C., Goodkin, G.M., et al. (2002). Effect of treatment
ment with repeated injections of hypertonic salt solution. JAMA, 94, on the incidence of stroke and other emboli in 519 patients with severe
1730–3. thoracic aortic plaque. American Journal of Cardiology, 90, 1320–5.
Sinha, S., Houghton, J., Holt, P.J., Thompson, M.M., Loftus, I.M., and Vayssairat, M., Fiessinger, J.-N., and Housset, E. (1979). Phénomène de
Hinchliffe, R.J. (2011). Popliteal entrapment syndrome. Journal of Raynaud étude prospective de 100 cas. Nouvelle Presse Médicale, 8,
Vascular Surgery, 55, 252–62. 2177–80.
Smolen, J.S., Weidinger, P., and Menzel, E.J. (1983). Sensitivity to colla- Wessler, S., Ming, S.-C., Gurewich, V., and Freiman, D.G. (1960). A critical
gen in thromboangiitis obliterans. New England Journal of Medicine, evaluation of thromboangiitis obliterans. The case against Buerger’s dis-
209, 857–8. ease. New England Journal of Medicine, 262, 1149–60.
CHAPTER 38

Cutaneous small-vessel
vasculitis
Anna Haemel, Lindy Fox, and M. Kari Connolly

Introduction medium, or large) together with clinical, laboratory, imaging,


and/or histological data to classify disease subtype (Fiorentino
Cutaneous small-vessel vasculitis (CSVV) is synonymous with 2003). The American College of Rheumatology criteria for CSVV,
cutaneous leukocytoclastic angiitis, hypersensitivity angiitis or also known as ‘hypersensitivity vasculitis,’ as initially defined by
vasculitis, and cutaneous necrotizing venulitis (Devore and Jorizzo Hunder et  al., are outlined in Table  38.2 (Hunder et  al. 1990).
2008; Shinkai and Fox 2012). CSVV signifies a small-vessel vascu- These criteria demonstrate poor sensitivity and specificity (71%
litis whose primary target is the postcapillary venules in the skin. and 84%, respectively) in distinguishing ‘hypersensitivity vasculi-
The most common clinical manifestation is symmetric palpable tis’ from other vasculitic syndromes (Hunder et al. 1990; Hunder
purpura of the lower extremities. The histopathological correlates 1998) and are of limited diagnostic utility (Rao et al. 1998). The
demonstrated by skin biopsy are perivascular neutrophilic infil- Chapel Hill nomenclature system for vasculitis was updated in
trate, neutrophilic debris (‘leukocytoclasia’), and fibrin deposition 2012 (Jennette et  al. 2013). This new classification system pro-
in the postcapillary venules (Weedon 2010). vides a separate category for single-organ vasculitis affecting
The term CSVV implies disease limited to the skin, with little the skin, defined as ‘cutaneous leukocytoclastic angiitis’. This is
to no systemic involvement and the absence of features diagnos- a provisional classification in that patients must be reclassified
tic for the other systemic vasculitic syndromes. However, the same if signs of a systemic vasculitis syndrome ensue (Jennette et al.
clinical features of palpable purpura with histopathological find- 2013). This system also includes separate categories for patients
ings of leukocytoclastic vasculitis are also found in other vasculitic with specific types of CSVV, including hypocomplementaemic
syndromes that involve the small (e.g. IgA vasculitis) or small and urticarial vasculitis, and for patients with CSVV and a suspected
medium-sized vessels (e.g. ANCA associated vasculitis, cryoglob- trigger (i.e. ‘drug-associated immune complex vasculitis’ and
ulinaemic vasculitis). CSVV lacks clinical and laboratory features ‘cancer-associated vasculitis’). In spite of these developments,
of these vasculitic syndromes. which are more pathology based, the simplest diagnostic crite-
Because CSVV is an immune-complex-mediated disease, the ria for CSVV might include palpable purpura clinically with the
direct immunofluorescence (DIF) test demonstrates the deposition histology of leukocytoclastic vasculitis, in the absence of systemic
of immunoglobulin and complement in the small blood vessels of small or medium-vessel vasculitis.
the skin (Carlson et al. 2005). The DIF can be helpful in diagnos-
ing IgA vasculitis (Henoch–Schönlein purpura), a distinct subset
of CSVV (Table 38.1) with IgA deposition in the small blood ves-
sels on DIF; it occurs more often in children, has a greater degree Table 38.1 Small-vessel vasculitis subsets
of renal and gastrointestinal involvement, and is discussed in
Chapter  39. Similarly, cryoglobulinaemic vasculitis is a systemic Primary:
vasculitis with prominent cutaneous manifestations that is dis- Cutaneous small-vessel vasculitis
cussed in Chapter 40. While systemic involvement should be con- Urticarial vasculitis
sidered and investigated for in patients with CSVV, classic CSVV Erythema elevatum diutinum
carries a better prognosis than the other vasculitides and may spon- Henoch-Schönlein purpura
taneously resolve. Treatment should be tailored to the severity and Cryoglobulinaemia
extent of the disease. Secondary:
Classification Drugs
Infections
There is ongoing controversy regarding the optimal vasculitis Connective tissue diseases
classification system, as outlined in Chapter 1 of this book and Inflammatory conditions
elsewhere (Fauci et al. 1978; Jorizzo 1993; Jennette et al. 2013).
Malignancy
Most schemes use some combination of vessel size (small,
518 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 38.2 American College of Rheumatology criteria for Table 38.3 Associations with cutaneous small-vessel vasculitis (Sams
hypersensitivity vasculitis (Hunder et al. 1990) and Sams 2001; Lotti et al. 1998)

1. Age at disease onset >16 years Drugs: 10% Allopurinol, aminosalicylic acid, beta-blockers,


herbicides, hydantoins, insecticides, insulin, iodides,
2. Medication at disease onset
oral contraceptives, penicillin, petroleum products,
3. Palpable purpura phenacetin, phenothiazines, phenylbutazone,
4. Maculopapular rash propylthiouracil, quinine, quinidine, serum,
streptokinase, streptomycin, sulfonamides, tamoxifen,
5. Biopsy including arteriole and venule with histological changes showing thiazides, vaccines (influenza), vitamins
granulocytes in a perivascular or extravascular location
Infections: 20% Candida albicans, cytomegalovirus, Epstein–Barr virus,
Hypersensitivity vasculitis is diagnosed if a patient meets 3 or more criteria. hepatitis A, B, and C, herpes simplex, histoplasmosis,
Specificity of 83.9% and sensitivity of 71%. influenza, HIV, Mycobacterium leprae, Mycobacterium
tuberculosis, Onchocerca volvulus, Plasmodium malariae,
Schistosoma haematobium, Schistosoma mansoni,
Staphylococcus aureus, Streptococcus group A
Epidemiology Malignancies: 5% Adult T-cell leukaemia, chronic lymphocytic leukaemia,
The incidence of CSVV is about 15 per million (Carlson et  al. hairy cell leukaemia, Hodgkin’s disease, lymphosarcoma,
2005). This makes it one of the most common vasculitides. Among multiple myeloma, mycosis fungoides, solid tumours
patients presenting to rheumatology clinics with a diagnosis of (breast, colon, head and neck, lung, prostate, and renal)
vasculitis, 9% have CSVV (Devore and Jorizzo 2008; Hunder et al. Other Behçet’s syndrome, bowel bypass syndrome, chronic
1990). Both genders can be affected, with a slight female predomi- conditions: 15 to active hepatitis, cryoglobulinaemia, cystic fibrosis,
nance (Carlson et al. 2005). CSVV affects all age groups, though 20% haemolytic anaemia, HIV/AIDS, hyperglobulinaemic
90% of cases occur in the adult population (Lotti et al. 1998). Once states, primary biliary cirrhosis, rheumatoid arthritis,
present, disease may persist for months or longer. In a study of 160 Sjögren’s syndrome, ulcerative colitis
patients with CSVV, disease resolved in <6 months in 46.9% of the
patients; however, at the end of the 3-year study, 28.8% of patients
still had disease. Median disease duration was 3.7  months (Sais
et al. 1998). Features predicting persistence of disease may include myeloma) is more commonly implicated than solid organ tumours
absence of fever and presence of arthralgia (Sais et al. 1998). (Watts et al. 2005). However, solid organ tumours, especially lung,
prostate, breast, colon, or renal cancers (Kurzrock et  al. 1994;
Aetiology Podjasek et al. 2012), may be present with CSVV and may demon-
strate perivascular IgA deposition on direct immunofluorescence.
While there is regional variation in the underlying causes of Neoplasia may trigger CSVV through a variety of mechanisms,
CSVV, an estimate is that ≥40% of cases remain idiopathic, 20% including impaired clearance of proteins, abnormal production
are secondary to infection, 10% secondary to drugs, 15–20% sec- of immunoglobulins, and/or shared antigens on tumour cells and
ondary to underlying autoimmune/ autoinflammatory disease, endothelial cells (Fortin 1996). In general, recalcitrant CSVV not
and 5% secondary to neoplasm (Ekenstam and Callen 1984; Sais responding as expected to treatment or the presence of IgA on DIF
et al. 1998) (Table 38.3). Taken together, the underlying aetiologies in an adult patient should raise concern for underlying neoplasm,
of CSVV are best thought of as antigens to which antibodies are malignant transformation of an indolent disease (e.g. myelodys-
generated, with subsequent immune complex deposition, comple- plastic syndrome to acute myelogenous leukaemia), or recurrence
ment activation, neutrophilic inflammatory infiltration, and dam- or metastasis of a malignancy thought to be in remission (Watts
age to the vessel wall. All types of infections have been implicated et al. 2005).
as causing CSVV, though the most common causes are bacterial
(e.g. β-haemolytic streptococcus) and viral infections (e.g. upper
respiratory infections, viral hepatitis, and HIV disease) (Carlson Pathogenesis
and Chen 2006; Hautmann et al. 1999). Among the drug-related CSVV is caused by immune-complex deposition in the postcapil-
cases, some of the most implicated agents include propylthioura- lary venules (Braverman and Yen 1975; Yancey and Lawley 1984).
cil, hydralazine, colony-stimulating factors, allopurinol, cefaclor, The circulating immune-complexes consist of bound foreign
penicillin, sulfonamides, minocycline, D-penicillamine, pheny- antigens and antibodies (IgG, IgA, and IgM) that, due to size and
toin, isotretinoin, methotrexate, and quinidine (Carlson and Chen solubility, attach to the endothelium of small blood vessels. As dis-
2006; Hautmann et al. 1999) as well as TNF- inhibitors (Hawryluk cussed in the Section Aetiology, and listed in Table 38.3, the list
et al. 2012). CSVV may present within 7–10 days of the antecedent of potential triggers is long and includes drugs, infectious agents,
exposure. autoimmune disease, or malignancy (Sais et al. 1998). Often a spe-
With regards to CSVV related to underlying autoimmune/ auto- cific antigen is not identified. Under normal conditions, circulating
inflammatory disorders, a variety of disorders may be implicated, foreign antigen–antibody complexes are cleared without sequelae.
ranging from Sjögren’s syndrome to inflammatory bowel dis- However, under certain pathological conditions of antigen excess,
ease. CSVV may also occur either before or concurrent with the immune complexes are made that are not readily cleared from the
clinical presentation of neoplastic disease (Podjasek et  al. 2012). circulation, become insoluble, and deposit in the small blood ves-
Haematological malignancy (e.g. lymphoproliferative disorders, sels (Lotti et al. 1998).
CHAPTER 38 cutaneous small-vessel vasculitis 519

Deposition of immune complex leads to activation of the comple-


ment cascade, production of complement fragments C3a and C5a
(Boom et al. 1989), and endothelial damage from the membrane
attack complex. C3a and C5a directly recruit neutrophils, basophils,
and mast cells to the site (Boom et al. 1989). C5a and other inflam-
matory signals in CSVV (e.g. thrombin, IL-1, TNF- ) (Claudy
1998; Lotti et al. 1998; Sais et al. 1997) also promote up-regulation
of adhesion molecules (i.e. selectins, integrins, and members of the
immunoglobulin superfamily) on endothelial cells and leukocytes
(Foreman et al. 1994; Sais et al. 1997). Up-regulation of adhesion
molecules helps to recruit inflammatory cells from the circulation,
promote rolling/ adhesion, and allow migration into sites of inflam-
mation (Kevil and Bullard 1999; Tonnesen 1989). E-selectin (Sais
et al. 1997) and β2 integrins may be of particular importance in
CSVV (Kevil and Bullard 1999).
Resulting tissue injury leads to ‘danger signals’ (Maugeri et al.
2009), resulting in release of additional cytokines (e.g. IL-8)
(Maugeri et al. 2009). IL-8 further promotes neutrophil migration
and primes the respiratory burst (Metzner et al. 1995; Tonnesen
1989). Recruited PMNs ultimately degranulate, releasing proteo-
lytic enzymes and free oxygen radicals which cause further damage
to blood vessel walls. Recruited basophils and mast cells promote
histamine release, causing increased vascular permeability (Claudy
1998). The damaged, leaky vessels allow additional deposition of
immune complexes and translocation of other proteins and inflam- Fig. 38.1 Palpable purpura over the lower extremities. Note small, 1 to
matory cells, perpetuating the inflammatory cycle (Claudy 1998). 2 mm, red-purple papules that coalesce into larger macules. This photograph
While CSVV was initially believed to be mediated solely by demonstrates the pressure effect with accentuation at the stocking edge. (Photo
PMNs, lymphocytes are also recruited, particularly in more long- courtesy of Dr. Richard Odom.)
standing disease (Lotti et  al. 1998). Lymphocytes probably react
to autoantigens exposed by the ongoing cellular damage and play
present in up to 90% of patients (Sais et al. 1998), clinical variants
a role in chronic disease activity (Ghersetich et  al. 1999). Fibrin
as described in this section are not uncommon. Lesions may occur
degradation products (Tosca and Stratigos 1988) and perivascular
at sites of trauma or friction (i.e. Koebner phenomenon) (Sais et al.
expression of HLA-DR and vascular cell adhesion molecule-1 (Sais
1998) and show a predilection for the dependent areas of the lower
et al. 1997) are also associated with chronicity.
extremities. The overall presentation often depends on the severity
Changes in flow and coagulation factors are intertwined with
and/or duration of disease.
the inflammatory pathways implicated in CSVV. TNF-α released
The earliest lesion of CSVV may be a pink macule or papule
by inflammatory cells may stimulate release of plasminogen activa-
or may be petechial in nature. Classic lesions then progress to
tor inhibitor, leading to reduced fibrinolysis (Jordan et al. 1987).
This may perpetuate fibrin deposition, necrosis, and microvascu-
lar clots, promoting a vicious cycle (Jordan et al. 1987). Activated
platelets also release inflammatory mediators such as leukotrienes
and adhesion molecules; this cross-talk between platelets and neu-
trophils may further promote thrombotic activity (Maugeri et al.
2009) and tissue compromise. Sites of slow flow and vessel bifur-
cations may be particularly prone to immune complex deposition
and subsequent damage (Lotti et al. 1998).

Clinical features
Palpable purpura refers to raised (palpable) lesions, which are
usually red-purple in colour and range from 2 to 5  mm in size
(Figures 38.1 and 38.2). The raised nature as well as the erythema
are attributable to the inflammatory response in the skin and con-
trast with the flat, bland appearance of petechiae (Stone and Nousari
2001), which also tend to be smaller (1–2mm). Lesions are gener-
ally non-blancheable, due to associated vessel damage and resulting
haemorrhage (Stone and Nousari 2001). Lesions may sting/ burn Fig. 38.2 Small, discreet, sharply demarcated, annular, non-blanching
(30%), itch (41%) (Sais et  al. 1998), or be asymptomatic. While erythematous papules and macules (<1 cm) over the lower extremities. (Photo
palpable purpura is the classic presentation of CSVV and may be courtesy of Dr. Matthew Goldberg.)
520 SECTION 5 vasculitic diseases and syndromes and related disorders

Fig. 38.5 With sufficient vascular destruction, necrosis ensues, often with


irregular borders. (From Sams, H.H. and Sams, W.M. Jr (2002). Cutaneous
leukocytoclastic vasculitis. In Vasculitis (Ball, G.V. and Bridges, S.L. Jr, eds), 1st edn,
pp. 467–75. Oxford University Press, New York.)

from medium-vessel vasculitis, which more typically presents as


stellate ulcers, clinically challenging.
Fig. 38.3 Punctate, haemorrhagic, papules on soles, some with focal erosion.
(Photo courtesy of Dr. Matthew Goldberg.)
Differential diagnosis
become palpable purpura. Alternatively, lesions may be urticaria As discussed in Section Clinical Features, CSVV presents with a
like (oedematous pink papules or small plaques) and resolve with variety of lesional morphologies, from which the clinical differen-
dusky patches or may be targetoid in appearance. Later lesions of tial diagnosis is generated. For lesions of typical palpable purpura,
CSVV may include pustules, vesicles/ bullae, erosions, or ulcers the differential diagnosis includes infectious entities (e.g. rickettsial
(Figure 38.3). Specifically, if the inflammatory response is particu-
larly brisk or longstanding, a sufficient number of neutrophils may
be recruited to form a pustule or turbid vesicle/ bulla, generally
overlying the centre of a lesion of palpable purpura (Figure 38.4).
Longstanding lesions may erode or ulcerate in the centre, often
leaving a central haemorrhagic eschar. In addition, lesions of CSVV
may coalesce to form larger ulcers with a stellate or angulated
appearance (Figures 38.5 and 38.6); this can make differentiation

Fig. 38.4 Severe form with haemorrhagic pustules that become confluent over Fig. 38.6 Severe ulcers may form in CSVV. (From Sams, H.H. and Sams, W.M. Jr
the ankles and dorsum of the feet. There is a tense, haemorrhagic blister over the (2002). Cutaneous leukocytoclastic vasculitis. In Vasculitis (Ball, G.V. and Bridges,
right, 2nd toe. (Photo courtesy of Dr. Matthew Goldberg.) S.L. Jr, eds), 1st edn, pp. 467–75. Oxford University Press, New York.
CHAPTER 38 cutaneous small-vessel vasculitis 521

urticarial vasculitis may, in fact, have associated SLE or Sjögren’s


syndrome.
Urticarial vasculitis can be distinguished from ordinary urticaria
in that individual lesions persist for >24 hours (usually for 48–72
hours). To assess this, it can be helpful to have patients circle a few
lesions and record how long they last. Overall lesion size may also
be a bit smaller than classic urticaria, measuring 1 to 2 centimetres
typically. In addition, lesions may resolve with purpura or hyper-
pigmentation of the skin, neither of which is characteristic of ordi-
nary urticaria. Patients may describe lesions as having a burning or
painful quality (Davis and Brewer 2004), which is also not generally
observed in ordinary urticaria.
McDuffie et al. first described the syndrome of hypocomplemen-
taemic urticarial vasculitis (HUV) (McDuffie et al. 1973). Patients
with this specific syndrome may have a systemic disease resembling
SLE. Distinguishing features in HUV include the presence of chronic
obstructive pulmonary disease-like symptoms and uveitis, as these
features are not usually seen in SLE. Hypocomplementaemia in
HUV is best demonstrated, and should be checked for, when skin
lesions are active.
The diagnostic criteria for hypocomplementaemic urticarial vas-
culitis syndrome as described by Schwartz are as follows:  major
criteria include urticarial skin lesions for more than 6 months and
Fig. 38.7 Urticarial vasculitis, presenting with urticaria-like, oedematous low complement (all components); minor criteria (two required)
pink plaques. A: photo courtesy of Dr. Agne Naujokas include dermal venulitis, arthralgia or arthritis, episcleritis or uvei-
tis, glomerulonephritis, recurrent abdominal pain, or C1q precipi-
diseases including Rocky Mountain Spotted fever, bacteraemia tin (IgG autoantibody) with a suppressed C1q level (Schwartz et al.
such as meningococcaemia, and viral infections such as parvovirus 1982). Other manifestations may include angioedema, chronic
B19), inflammatory disorders (e.g. chilblains), haemorrhage into obstructive pulmonary disease-like symptoms, and neurological
another eruption (e.g. drug exanthem or arthropod bites), or rarely manifestations (Jara et al. 2009; Kroshinsky et al. 2011). Of note,
haematological malignancy. some patients may have both hypocomplementaemia and urticarial
For petechial lesions or macular purpura, the differential diag- vasculitis but not meet the criteria for the hypocomplementaemic
nosis includes haemorrhage (e.g. platelet dysfunction). Also on the urticarial vasculitis syndrome, as this syndrome refers to a particu-
differential is the benign condition ‘pigmented purpura’ or capil- lar constellation of features as detailed above in this section. For
laritis, which tends to present as tiny (<1 mm) cayenne pepper-like further discussion of these entities, please see the excellent review
macules over the lower legs. Some of the infections discussed in the by Davis and Brewer (2004).
previous paragraph should also be entertained when petechiae are Erythema elevatum diutinum: erythema elevatum diutinum
present, depending on the acuity and overall picture. (EED) is a unique subset of chronic, low-grade CSVV. Unlike most
For urticaria-like lesions (Figure 38.7), the differential diagnosis lesions of CSVV, which resolve without residua or with scars limited
includes urticarial vasculitis (specifically discussed in the Section to areas of prior ulceration, EED lesions tend to persist for years. The
Special Subtypes) but also urticaria itself, Sweet’s syndrome, Still’s chronicity, in turn, leads to clinical and histological fibrosis (LeBoit
disease, serum sickness, rheumatic fever, drug reactions, and viral et  al. 1986). Early lesions begin as purple papules or plaques that
exanthems. For targetoid-appearing lesions, diagnostic considera- evolve into persistent yellowish fibrotic plaques or nodules, generally
tions include erythema multiforme and other related disorders. over the extensor extremities. EED can be associated with arthral-
With regards to pustular lesions, infection should be considered, gias, keratitis, and constitutional symptoms but is otherwise a purely
including bacterial infections and disseminated fungal infections. cutaneous process (LeBoit et al. 1986). While EED does not itself lead
Lesions that aggregate to form stellate ulcers will prompt workup to systemic disease, it has been associated with an array of underlying
for processes involving medium vessels. conditions including autoimmune disease, haematological abnor-
malities (e.g. paraproteinaemia, IgA monoclonal gammopathy), and
infection (e.g. HIV) (Wahl et al. 2005; Yiannias et al. 1992).
Special subtypes EED can be difficult to distinguish from a related condition called
Urticarial vasculitis: urticarial vasculitis is a clinicopathological granuloma faciale in which red brown plaques occur on the face;
entity comprised of clinical urticarial lesions that histopathologi- underlying disease and systemic symptoms are generally absent in
cally demonstrate leukocytoclastic vasculitis. In addition to the granuloma faciale. Some authors have posited that EED and granu-
immunoreactants within vessel walls as observed in typical CSVV, loma faciale actually represent a spectrum of disorders occurring
patients may have granular deposits of immunoglobulins in the at different anatomical sites (Ziemer et al. 2011). The diagnosis of
small blood vessels. Less commonly, there may be deposition granuloma faciale may be favoured by a predominance of eosino-
along the basement membrane zone reminiscent of the changes phils in the inflammatory infiltrate among other subtle differences
seen in systemic lupus erythematosus (SLE), and patients with (Ziemer et al. 2011).
522 SECTION 5 vasculitic diseases and syndromes and related disorders

Systemic involvement Histopathology


Patients with CSVV may experience systemic symptoms, including A punch biopsy (generally 4 mm or larger) should be taken from the
flu-like symptoms, myalgias, and arthralgias with disease attacks centre of a fresh lesion, ideally within 24–48 hours of its appearance.
(Lotti et al. 1998). Both the presence of fever and absence of pain Adequate depth, including at least the superficial fat, is necessary
within skin lesions may be risks for systemic disease (Sais et  al. to rule out medium-vessel involvement. A separate lesion is biop-
1998). However, the severity of clinical and histological findings sied for direct immunofluorescence testing; this must be planned
in the skin does not predict whether there will be internal organ at the time of biopsy and sent in the appropriate special medium
involvement (Sais et al. 1998). While the majority of patients with (Stone and Nousari 2001). We recommend biopsy for both routine
CSVV will have skin-limited disease (Lotti et  al. 1998), any site histology and direct immunofluorescence testing for all patients
where immune complex could be filtered and deposited other than suspected to have CSVV. If there is concern for infection, tissue may
skin should be considered as a potential disease target (Devore and also be sent for bacterial, fungal, and mycobacterial cultures.
Jorizzo 2008). Therefore, all patients with CSVV should receive a On routine histology, early CSVV demonstrates neutrophilic
thorough evaluation for end-organ disease (Table  38.4) and the infiltration into the postcapillary venule and extending into the
presence of an underlying systemic vasculitis syndrome. perivascular zone (Figure 38.8a) (Weedon 2010). The neutrophils
undergo degeneration or ‘leukocytoclasis’, resulting in deposition
Evaluation of nuclear dust in the vicinity. There is also fibrin deposition (‘fibri-
The laboratory evaluation of patients with CSVV can be divided noid necrosis’) within the blood vessel wall. Oedema and extrava-
into:  (1)  skin biopsy to confirm the diagnosis and evaluate for sation of erythrocytes are commonly observed. Over time, the
immunoprecipitants (discussed in Section Histopathology), infiltrate may become more mixed, including eosinophils, lympho-
(2) evaluation for underlying aetiology, and (3) evaluation for sys- cytes, and macrophages. Drug-induced forms of CSVV may have a
temic involvement. higher ratio of eosinophils.
The evaluation for underlying aetiology includes the follow- Direct immunofluorescence (DIF) is positive 80% of the time in
ing: (1) historical elements, including any recent acute infections or early CSVV (Carlson et al. 2005). For optimal results, DIF should
travel (including tick exposures), medication changes, status of rou- be completed within 24–48 hours of the lesion’s appearance. The
tine cancer screening, and history or symptoms of connective tis- test shows IgG, IgA, IgM, complement factor 3, and/or fibrinogen
sue disease, chronic infections (e.g. HBV, HCV, HIV), malignancy, in small dermal blood vessels. IgA positivity is seen in adult patients
or coagulopathy; (2) examination findings suggestive of underly- with Henoch–Schönlein Purpura (HSP), also known as IgA vascu-
ing systemic disease (e.g. stigmata of liver disease or inflammatory litis, which can be otherwise indistinguishable from CSVV. Given
arthropathy); and (3) further laboratory work-up to include: ANA, that adult HSP can be associated with a higher risk of renal disease
ANCAs, C3/4, rheumatoid factor; antistreptolysin-O, quantiferon and solid organ malignancy, it is important to make this distinc-
gold assay for tuberculosis, or tuberculin purified protein deriva- tion. IgM is seen in cryoglobulinaemia, which is discussed else-
tive; HBV/HCV serologies and cryoglobulins; HIV test; urine toxi- where in this text. C3 is the most common non-specific finding in
cology screen; and serum protein electrophoresis. For hospitalized CSVV, as shown in Figure 38.8b.
patients or if the concern for infection is high, blood cultures are In addition to HSP and cryoglobulinaemic vasculitis, the his-
sent and an echocardiogram may be indicated. topathological differential diagnosis of CSVV includes ‘inciden-
The evaluation for systemic involvement includes a thorough tal vasculitis’, which can occur in association with a longstanding
review of systems (Table 38.4) and laboratory workup, to include ulcer or in entities such as Sweet’s syndrome (Carlson and Chen
CBC with differential, BUN/ creatinine, LFTs, UA, stool for occult 2006). When the biopsy is superficial (i.e., does not include the
blood, and chest radiograph. Inflammatory markers such as ESR dermal–subcutaneous junction and some fat or does not demon-
and CRP may be obtained and followed over time. strate medium vessels), it is also important to maintain suspicion
for mixed small and medium-vessel vasculitis. The differential
diagnosis for these mixed processes includes ANCA-associated
Table 38.4 Systemic manifestations of cutaneous small-vessel vasculitis processes, cryoglobulinaemic vasculitis, septic vasculitis, IgA vas-
(Lotti et al. 1998)
culitis (Carlson and Chen 2006), and rarely PAN. Cocaine can
also induce a mixed small and medium-vessel picture, particularly
Joints Arthralgias and arthritis
when combined with levamisole, with an associated neutropenia
Pulmonary Cough and haemoptysis or asymptomatic and an unusual pattern of ANCAs including anti-PR3 and antihu-
Cardiac Myocardial angiitis and pericarditis man neutrophil elastase (Waller et al. 2010). These patients typically
have additional findings of non-inflammatory vessel thrombosis
Gastrointestinal Nausea, vomiting, diarrhoea, colicky pain,
haematemesis, melena, and pancreatitis
(thrombotic vasculopathy). The finding of a true small-vessel vas-
culitis and true thrombotic vasculopathy in the same specimen is
Renal Nephritis with microscopic haematuria and highly suggestive of levamisole-induced skin disease. Lastly, long-
proteinuria, acute and chronic renal failure
standing thrombotic lesions can recruit inflammation and appear
Ophthalmological Retinal vasculitis, conjunctivitis, keratitis, and histologically indistinguishable from CSVV.
pseudotumour cerebri Some patients will not present with new or active lesions but fur-
Nervous system Headache, diplopia, hypoesthesia, paraesthesia ther characterization of their CSVV process is still desired. Within
about 24 to 48 hours, immunoprecipitants wash out of vessels and
Constitutional Fever
vessel thrombosis becomes the predominate feature, presenting
CHAPTER 38 cutaneous small-vessel vasculitis 523

(a) (b)

Fig. 38.8 (a) H&E demonstrating neutrophil infiltration into vessel wall, fibrinoid necrosis, and nuclear dust. (Photo courtesy of Dr Agne Naujokas.)
(b) DIF demonstrating deposition of C3 immunoreactants within vessel wall.

a diagnostic dilemma (Braverman and Yen 1975). In these cases,


the histamine trap test can be pursued (Braverman and Yen 1975; Table 38.5 Therapeutic ladder for the treatment of CSVV (Barham
Jorizzo et al. 1988). In this test, 0.05 ml of histamine solution (1 mg/ et al. 2004)
ml) is injected intradermally and the resulting wheel outlined. At
Skin lesions alone
3–4 hours later a punch biopsy is obtained for direct immunofluo-
Supportive therapyc
rescence studies and/or routine histology. This test reproduces the
Antihistaminesc
histology of an early CSVV lesion with both immunoprecipitants Non-steroidal anti-inflammatory drugsb
and recruitment of neutrophils (Braverman and Yen 1975; Jorizzo Pentoxiphyllinec
et  al. 1988); a more typical-appearing clinical CSVV lesion may Hydroxychloroquinec
ensue within 18–24 hours (Braverman and Yen 1975). Colchicinea
Dapsoneb
Treatment Ulcerative skin lesions alone
Thalidomidec
The therapeutic ladder for treatment of CSVV is presented in Low-dose weekly methotrexatec
Table  38.5 (Barham et  al. 2004; Devore and Jorizzo 2008). The Prednisoneb
patient should be evaluated for any specific triggers, and identifiable
Systemic disease
culprits (e.g. medications) should be eliminated. The disease may
Prednisoneb
also respond to treatment of underlying disorders (Tsampau et al.
Azathioprine 1–2.5 mg/kg/day p.o. as single dose or divided
2012). Beyond this, therapeutic decisions for patients with CSVV twice a dayb
depend on the severity of disease and co-morbidities. There are few Cyclophosphamide pulsed dosing regimen, 40–50 mg/kg i.v. in
double-blind placebo controlled trials to guide treatment decisions, divided doses over 2–5 days or 10–15 mg/kg i.v. every 7–10 days or
and current treatment is based largely on small trials, case series, 3–5 mg/kg
and expert opinion (Jorizzo et al. 1988; Tsampau et al. 2012). i.v. twice weeklyb
For patients with the mild, non-ulcerating disease localized to Mycophenolate mofetil 500–2000 mg p.o. b.i.d.c
skin only, a 2 to 4-week course of medium-potency topical steroid Methotrexate 7.5–15 mg once a week
ointment such as triamcinolone may be sufficient. Ointment is pre- Ciclosporin 2.5 mg/kg/day p.o. divided twice daily; after 4 weeks,
dose may be increased 0.5 mg/kg/day at 2-week intervals; maximum
ferred over cream as it is more soothing, and the occlusive property
of 4 mg/kg/dayc
augments the effect. NSAIDs can also be helpful provided there Interferon-α and ribavirin (if hepatitis C-associated) 3 million units,
is no renal impairment. Hydroxyzine 25–75 mg nightly, titrated 3 times a week, and 1000 mg/day, respectivelya
as allowed by sedation, can also be a useful steroid-sparing agent Intravenous gammaglobulinc
by reducing histamine-mediated gaps between endothelial cells Extracorporeal immunomodulationc
(Tsampau et al. 2012). For patients with more extensive or ulcerat- Biological agents (TNF- inhibitors), infliximab, etanerceptc
ing disease and in whom infection is believed to be absent, pred- a Evidence from double-blind studies.
nisone can be initiated at 0.5–1 mg/kg. This can then be tapered b Evidence from case series.
over 6–10 weeks, depending on response. Slow taper helps to pre- c Evidence from case reports.
vent rebound/ relapse (Tsampau et al. 2012).
524 SECTION 5 vasculitic diseases and syndromes and related disorders

In patients with recurrent episodes or persistent/ severe disease, internal manifestations is required to rule out systemic disease or
colchicine 0.6 mg twice to three times daily by mouth, as tolerated an underlying vasculitis syndrome. Despite recent advances in our
by gastrointestinal upset, can help to reduce neutrophil infiltration understanding of this disease and its classification (Jennette et al.
(Callen 1985). Dapsone is an additional neutrophil-targeted ther- 2013), much remains to be elucidated in regard to underlying
apy that can be substituted or added to this regimen for additional mechanisms and optimal treatment (Callen 1998).
effect. A G6PD level, baseline CBC, and LFTs are generally checked
before starting this agent; CBC with differential should be moni- References
tored weekly for 4 weeks and then monthly; this is repeated with any Atzori, L., Ferreli, C., and Biggio, P. (1999). Less common treatment in cuta-
dosage increase. Dapsone is usually initiated at 50 mg/day and then neous vasculitis. Clinics in Dermatology, 17, 641–7.
increased by 50 mg/day at weekly intervals until a dose of 150–200 Barham, K.L., Jorizzo, J.L., Grattan, B., et al. (2004). Vasculitis and neutro-
mg is reached or effect is achieved. Alternative agents for recalcitrant philic vascular reactions. In Rook’s textbook of dermatology, 7th edn (ed.
T. Burns), pp. 49.1–49.46. Blackwell, Oxford.
skin disease might include antimalarials and pentoxifylline (Atzori
Boom, B.W., Mommaas, M., Daha, M.R., et al. (1989). Complement-mediated
et al. 1999), as well as agents targeting the fibrinolytic system (e.g.
endothelial cell damage in immune complex vasculitis of the skin: ultra-
stanazol) (Lotti et al. 1998). For pregnant patients or patients unable structural localization of the membrane attack complex. Journal of
to tolerate the above regimens, IVIg may be an option. Investigative Dermatology, 93 (Suppl.), 68S–72S.
In patients with recurrent CSVV in whom identifiable trig- Braverman, I.M. and Yen, A. (1975). Demonstration of immune com-
gers have been eliminated, long-term suppressive therapy may be plexes in spontaneous and histamine-induced lesions and in normal
required. Maintenance colchicine and/or dapsone may be helpful. skin of patients with leukocytoclastic angitis. Journal of Investigative
Scheduled hydroxyzine can be a helpful adjunct as noted (Tsampau Dermatology, 64, 105–112.
et al. 2012). For patients with more recalcitrant chronic disease or Callen, J.P. (1985). Colchicine is effective in controlling chronic cutane-
ous leukocytoclastic vasculitis. Journal of the American Academy of
internal involvement, immunosuppression would be warranted
Dermatology, 13, 193–200.
(Atzori et al. 1999). For these patients, options may include every
Callen, J.P. (1998). Cutaneous vasculitis: what have we learned in the past
other day low-dose (<10 mg) prednisone, methotrexate up to 25 20 years? Archives of Dermatology, 134, 355–7.
mg/week, azathioprine 2 mg/kg/day (Callen 1985), or mycopheno- Carlson, J.A. and Chen, K.R. (2006). Cutaneous vasculitis update:  small
late mofetil 2–3 grams per day (Haeberle et al. 2012). Rituximab vessel neutrophilic vasculitis syndromes. American Journal of
has also been reported to be helpful in intractable cases of CSVV Dermatopathology, 28, 486–506.
(Chung et al. 2006). Carlson, J.A., Ng B.T., and Chen K.R. (2005). Cutaneous vasculi-
Wound care for patients with ulcerating lesions includes cul- tis update:  diagnostic criteria, classification, epidemiology, etiol-
tures of any areas that appear clinically infected (discharge, odour). ogy, pathogenesis, evaluation and prognosis. American Journal of
Dermatopathology, 27, 504–28.
We treat Staphylococcus aureus colonizing these lesions as a true
Chung, L. Funke, A.A., Chakravarty, E.F., et al. (2006). Successful use of ritux-
pathogen with the potential to impair healing. Treatment includes
imab for cutaneous vasculitis. Archives of Dermatology, 142, 1407–10.
mupirocin ointment with or without a course of oral antibiotics. Claudy, A. (1998). Pathogenesis of leukocytoclastic vasculitis. European
It is prudent to avoid over-the-counter antibiotic ointments such Journal of Dermatology, 8, 75–9.
as bacitracin and neomycin due to the high incidence of allergic Davis, M.D.P. and Brewer, J.D. (2004). Urticarial vasculitis and hypocom-
contact dermatitis associated with these agents. For malodorous plementemic urticarial vasculitis syndrome. Immunology and Allergy
wounds, metronidazole 0.75% gel applied to the ulcer bed can help Clinics of North America, 24, 183–213, vi.
with odour and also exert an anti-inflammatory effect (Finlay et al. Devore, A. and Jorizzo, J. (2008). Cutaneous small vessel vasculitis. In
1996). Dilute bleach baths (prepared by adding no more than a Vasculitis, 2nd edn (ed. S. Louis Bridges and Gene V. Ball), pp. 503–10.
quarter cup of bleach into a full bathtub of water) can also be help- Oxford University Press, New York.
ful in broadly reducing bacterial carriage in these lesions. Ekenstam, E. and Callen, J.P. (1984). Cutaneous leukocytoclastic vasculitis.
Clinical and laboratory features of 82 patients seen in private practice.
Keeping the wound environment clean and moist promotes heal- Archives of Dermatology, 120, 484–9.
ing. Dressing care may include Vaseline gauze applied to the ulcer Fauci, A.S. Haynes, B., and Katz, P. (1978). The spectrum of vasculitis: clini-
bed; a non-stick dressing such as Telfa may be applied on top and cal, pathologic, immunologic and therapeutic considerations. Annals of
gently secured in place with gauze roll bandages such as Kerlix. Internal Medicine, 89, 660–76.
Light compression may be achieved with elastic bandages placed Finlay, I.G., Bowszyc, J., Ramlau, C., et al. (1996). The effect of topical 0.75%
over the top of these dressings (provided that arterial supply is suf- metronidazole gel on malodorous cutaneous ulcers. Journal of Pain and
ficient); compression and elevation enhance healing by control- Symptom Management, 11, 158–62.
ling oedema (Tsampau et al. 2012). A multidisciplinary approach Fiorentino, D.F. (2003). Cutaneous vasculitis. Journal of the American
including rheumatology, dermatology, and wound care clinics may Academy of Dermatology, 48, 311–40.
Foreman, K.E. Vaporciyan, A.A., Bonish B.K., et  al. (1994). C5a-induced
be required for non-healing wounds.
expression of P-selectin in endothelial cells. Journal of Clinical
Investigation, 94, 1147–55.
Summary Fortin, P.R. (1996). Vasculitides associated with malignancy. Current Opinion
in Rheumatology, 8, 30–3.
CSVV is a reactive process mediated by immune complexes sec-
Ghersetich, I. Comacchi, C., Katsambas, A., et al. (1999). Cellular steps in
ondary to identifiable and unidentified antigen triggers, resulting pathogenesis of cutaneous necrotizing vasculitis. Clinics in Dermatology,
in palpable purpura clinically and leukocytoclastic vasculitis patho- 17, 597–601.
logically. It is distinct from the other vasculitic syndromes by being Haeberle, M.T., Adams, W.B., Callen, J.P., et al. (2012). Treatment of severe
ANCA negative and not having significant internal organ involve- cutaneous small-vessel vasculitis with mycophenolate mofetil. Archives
ment. It often remains limited to the skin; however, a workup for of Dermatology, 148, 887–8.
CHAPTER 38 cutaneous small-vessel vasculitis 525

Hautmann, G., Campanile, G., and Lotti, T.M. (1999). The many faces of Rao, J.K., Allen, N.B., Pincus, T., et  al. (1998). Limitations of the 1990
cutaneous vasculitis. Clinics in Dermatology, 17, 515–31. American College of Rheumatology classification criteria in the diagno-
Hawryluk, E.B., Linskey, K.R., Duncan, L.M., et al. (2012). Broad range of sis of vasculitis. Annals of Internal Medicine, 129, 345–52.
adverse cutaneous eruptions in patients on TNF-alpha antagonists. Sais, G., Vidaller, A., Jucglá, A., et al. (1997). Adhesion molecule expression
Journal of Cutaneous Pathology, 39, 481–92. and endothelial cell activation in cutaneous leukocytoclastic vasculi-
Hunder, G.G. (1998). The use and misuse of classification and diagnostic cri- tis. An immunohistologic and clinical study in 42 patients. Archives of
teria for complex diseases. Annals of Internal Medicine, 129, 417–8. Dermatology, 133, 443–50.
Hunder, G.G., Arend, W. P., Bloch, D.A., et al. (1990). The American College Sais, G., Vidaller, A., Jucgla, A., et al. (1998). Prognostic factors in leukocy-
of Rheumatology 1990 criteria for the classification of vasculitis. toclastic vasculitis: a clinicopathologic study of 160 patients. Archives of
Introduction. Arthritis and Rheumatism, 33, 1065–7. Dermatology, 134, 309–15.
Jara, L.J., Navarro, C., Medina, G., et al. (2009). Hypocomplementemic urti- Sams, H. and Sams, W. (2001). Cutaneous leukocytoclastic vasculitis. In
carial vasculitis syndrome. Current Rheumatology Reports, 11, 410–5. Vasculitis (ed. Gene V. Ball and S. Louis Bridges), pp. 467–75. Oxford
Jennette, J., Falk, R.J., Bacon, P.A., et al. (2013). 2012 Revised International University Press, New York.
Chapel Hill Consensus Conference Nomenclature of Vasculitides. Schwartz, H.R., McDuffie, F.C., Black, L.F., et al. (1982). Hypocomplementemic
Arthritis and Rheumatism, 65, 1–11. urticarial vasculitis:  association with chronic obstructive pulmonary
Jordan, J.M., Allen, N.B., and Pizzo, S.V. (1987). Defective release of tissue disease. Mayo Clinic Proceedings, 57, 231–8.
plasminogen activator in systemic and cutaneous vasculitis. American Shinkai, K. and Fox, L. (2012). Cutaneous vasculitis. In Dermatology, 3rd edn
Journal of Medicine, 82, 397–400. (ed. J. Bolognia, et al.), pp. 385–410. Elsevier.
Jorizzo, J.L. (1993). Classification of vasculitis. Journal of Investigative Stone, J.H. and Nousari, H.C. (2001). ‘Essential’ cutaneous vasculitis: what
Dermatology, 100, 106S–110S. every rheumatologist should know about vasculitis of the skin. Current
Jorizzo, J.L., Solomon, A.R., Zanolli, M.D., et al. (1988). Neutrophilic vascular Opinion in Rheumatology, 13, 23–34.
reactions. Journal of the American Academy of Dermatology, 19, 983–1005. Tonnesen, M.G. (1989). Neutrophil-endothelial cell interactions:  mecha-
Kevil, C.G. and Bullard, D.C. (1999). Roles of leukocyte/endothelial cell nisms of neutrophil adherence to vascular endothelium. Journal of
adhesion molecules in the pathogenesis of vasculitis. American Journal Investigative Dermatology, 93 (Suppl.), 53S–58S.
of Medicine, 106, 677–87. Tosca, N. and Stratigos, J.D. (1988). Possible pathogenetic mechanisms in
Kroshinsky, D., Hoang, M.P., and Hasserjian, R.P. (2011). Case records of the allergic cutaneous vasculitis. International Journal of Dermatology,
Massachusetts General Hospital. Case 22-2011. A 79-year-old man with 27, 291–6.
a rash, arthritis, and ocular erythema. New England Journal of Medicine, Tsampau, D., Buggiani, G., Hercogova, J., et al. (2012). Cutaneous necrotiz-
365, 252–62. ing vasculitis: a rational therapeutic approach. Dermatologic Therapy,
Kurzrock, R., Cohen, P.R., and Markowitz, A. (1994). Clinical manifestations 25, 335–9.
of vasculitis in patients with solid tumors. A case report and review of Wahl, C.E., Bouldin, M.B., Gibson, L.E., et al. (2005). Erythema elevatum
the literature. Archives of Internal Medicine, 154, 334–40. diutinum:  clinical, histopathologic, and immunohistochemical char-
LeBoit, P.E., Yen, T.S., and Wintroub, B. (1986). The evolution of lesions in acteristics of six patients. American Journal of Dermatopathology, 27,
erythema elevatum diutinum. American Journal of Dermatopathology, 397–400.
8, 392–402. Waller, J.M., Feramisco, J.D., Alberta-Wszolek, L., et  al. (2010).
Lotti, T., Ghersetich, I., Comacchi, C., et al. (1998). Cutaneous small-vessel Cocaine-associated retiform purpura and neutropenia: is levamisole the
vasculitis. Journal of the American Academy of Dermatology, 39, 667–87; culprit? Journal of the American Academy of Dermatology, 63, 530–5.
quiz 688–90. Watts, R.A., Lane, S., and Scott, D.G. (2005). What is known about the
Maugeri, N., Rovere-Querini, P., Baldini, M., et  al. (2009). Translational epidemiology of the vasculitides? Best Practice and Research Clinical
mini-review series on immunology of vascular disease: mechanisms of Rheumatology, 19, 191–207.
vascular inflammation and remodelling in systemic vasculitis. Clinical Weedon, D. (2010). Leukocytoclastic (hypersensitivity). vasculitis. In
and Experimental Immunology, 156, 395–404. Weedon’s skin pathology, 3rd edn, pp. 208–11. Elsevier.
McDuffie, F.C., Sams, W.M. Jr, Maldonado, J.E., et  al. (1973). Yancey, K.B. and Lawley, T.J. (1984). Circulating immune complexes: their
Hypocomplementemia with cutaneous vasculitis and arthritis. Possible immunochemistry, biology, and detection in selected dermatologic and
immune complex syndrome. Mayo Clinic Proceedings, 48, 340–8. systemic diseases. Journal of the American Academy of Dermatology, 10,
Metzner, B., Barbisch, M., Parlow, F., et al. (1995). Interleukin-8 and GRO 711–31.
alpha prime human neutrophils for superoxide anion production Yiannias, J.A., el-Azhary R.A., and Gibson L.E. (1992). Erythema elevatum
and induce up-regulation of N-formyl peptide receptors. Journal of diutinum: a clinical and histopathologic study of 13 patients. Journal of
Investigative Dermatology, 104, 789–91. the American Academy of Dermatology, 26, 38–44.
Podjasek, J.O., Wetter, D.A., Pittelkow, M.R., et  al. (2012). Cutaneous Ziemer, M., Koehler M.J., and Weyers W. (2011). Erythema elevatum diuti-
small-vessel vasculitis associated with solid organ malignancies:  the num—a chronic leukocytoclastic vasculitis microscopically indistin-
Mayo Clinic experience, 1996 to 2009. Journal of the American Academy guishable from granuloma faciale? Journal of Cutaneous Pathology, 38,
of Dermatology, 66, e55–65. 876–83.
CHAPTER 39

IgA vasculitis (Henoch-


Schönlein purpura)
Miguel A. González-Gay, Ricardo Blanco, and
Trinitario Pina

Editors’ note pertaining to the title of the chapter as IgA Vasculitis: clinical triad of IgAV is palpable purpura, joint symptoms, and
The International Chapel Hill Consensus Conference 2012 abdominal pain, the most serious complication is renal involve-
(CHCC2012) of 28 participants from 12 countries was convened ment. IgAV is characterized histologically by infiltration of the
to revise and improve the CHCC1994 nomenclature of vasculitis. small blood vessels with polymorphonuclear leukocytes and the
The conference considered the use of eponyms and deliberated to presence of leukocytoclasia. Immunofluorescence staining usu-
determine a non-eponymous replacement term if there was ade- ally reveals the presence of IgA-dominant immune deposits in the
quate understanding of the pathophysiology of a vasculitis. The walls of the small vessels (capillaries, venules, or arterioles) and
consensus of participants therefore favoured replacing the eponym in the renal glomeruli (Giancomo and Tsai 1977; Jennette et  al.
Henoch–Schönlein purpura with IgA vasculitis (IgAV) based on 1994). IgAV is essentially a childhood disease. The paediatric form
the compelling literature which indicted abnormal IgA deposits in of IgAV has been widely described and it is generally considered
vessel walls as the defining pathophysiological feature. (This was a benign, self-limited disorder (Calviño et  al. 2001; Cassidy and
related to reduced terminal glycosylation in the Hinge region of Petty 1995; Trapani et al. 2005). IgAV in adults is less common and
IgA 1 in serum and tissue deposits; furthermore, there were data is generally associated with a worse outcome (Garcia-Porrúa and
identifying circulating abnormally glycosylated IgA1 and IgG anti- González-Gay 1999a; Garcia-Porrúa et al. 2002).
bodies that form IgA1–IgG anti-IgA1 immune complexes.) The
terminology used throughout this text reflects recommendations Epidemiology
of the consensus conference (Jennette et al. 2013).
IgAV is the most common vasculitis in children (González-Gay
and Garcia-Porrúa 2001). Although it may occur from 6 months of
Introduction age through adulthood, 50% of the cases occur in children less than
In the first half of the nineteenth century, Schönlein (1837) named 5 years of age, and 75% of patients are less than 10 years old. Indeed,
the association between arthralgia and purpuric cutaneous lesions the disease is observed predominantly in children between the ages
in a child ‘purpura rheumatica’. His pupil Edward-Heinrich Henoch of 2 and 10, and the median age of onset is 4 years.
(1874) described a syndrome of purpura, severe abdominal colic, In some reports, girls and boys were affected equally. A lower fre-
and melena. It was not until the end of the nineteenth century that quency of boys (46%) than girls was observed in paediatric IgAV
this author referred to nephritis as a complication of this syndrome from the Lugo region of north-western Spain (Calviño et al. 2001).
(Henoch 1895). The first description of a child with the syndrome In contrast, in Charlottesville, Virginia, USA, boys outnumbered
that we currently know as HSP was by Heberden (1801) in his trea- girls (57 of 100) (Saulsbury 1999). Previous studies also found a
tise ‘On cutaneous disease’. A child with joint pain and painful sub- higher incidence in boys than girls (Allen et  al. 1960; Robson
cutaneous oedema, abdominal pain, vomiting, bloody stools and and Leung 1994; Sterky and Thilén 1960). In Taiwan, Yang et al.
urine, and ‘bloody points’ over the skin of his legs was described. In reported a slightly higher annual incidence in males before the age
1915, Frank called this syndrome ‘anaphylactoid purpura’ (Frank of 10 that became lower with increasing age (Yang et al. 2005).
1915). This term is not commonly used, however, because an aller- Classic studies described an annual incidence rate of IgAV in chil-
gic aetiology has not been substantiated. dren between 135 and 180 per million (Nielsen 1988; Robson and
Leung 1994; Stewart et al. 1988). Nielsen (1988) showed that while
the average annual incidence rate in the county of Copenhagen was
Definition 180 per million for children aged 0–14, it was only 8 per million for
IgA vasculitis (IgAV) is a systemic vasculitis characterized by the population 15 years and older. European epidemiological studies
purpuric skin lesions unrelated to any underlying coagulopathy, yielded an estimated annual incidence of 61 per million children in
abdominal pain and bleeding, arthritis, and renal involvement the Netherlands (Aalberse et al. 2007), 102 per million in the Czech
(González-Gay et al. 2005; Saulsbury 2001). Although the classic Republic (Dolezalova et al. 2004), 126 per million in the Lugo region
528 SECTION 5 vasculitic diseases and syndromes and related disorders

of north-western Spain (Calviño et al. 2001), and 204 per million Table 39.1 Genetic polymorphisms associated with disease severity in
in the West Midlands, UK (Gardner-Medwin et  al. 2002). In the IgAV patients from north-western Spain
Middle East countries an annual incidence of 67 per million has
been reported among Saudi children (Abdel-Al et al. 1990). A gen- Authors Gene polymorphism Type of complication
eral population-based nationwide survey among Taiwanese children
Amoli et al. (2001a) Codon 469 of ICAM-1 Gastrointestinal
estimated an annual incidence of 129 per million (Yang et al. 2005).
The incidence of IgAV is much lower in adults than in children. Amoli et al. (2002a) HLA-B35 Susceptibility to nephritis
Based on the American College of Rheumatology (ACR) classifi- Amoli et al. (2002b) IL1RN Nephrotic syndrome and/or
cation criteria for IgAV (Mills et  al. 1990), the annual incidence renal insufficiency
of IgAV in an English population of patients older than 16 years Amoli et al. (2002c) Interleukin-8 Susceptibility to nephritis
was 13.0 per million (Watts et al. 1998). In north-western Spain,
the annual incidence rate of biopsy-proven IgAV in a population Amoli et al. (2004) Interleukin-1beta Nephrotic syndrome and/or
renal insufficiency
21  years of age and older was 14.3 per million (Garcia-Porrúa
and González-Gay 1999a). Previous reports described a similar Rueda et al. (2006) VEGF Susceptibility to nephritis
incidence among men and women (Clauvel et  al. 1972; Debray
ICAM-1, intercellular adhesion molecule-1 locus; IL1RN, interleukin 1 receptor antagonist
et  al. 1971), but an epidemiological study from north-western gene polymorphism; VEGF, vascular endothelial growth factor.
Spain disclosed a higher incidence in men than women (2:1 ratio)
(Garcia-Porrúa and González-Gay 1999a).

Topaloglu et  al. (2001) reported that vascular endothelial


Genetic factors growth factor (VEGF) could play a crucial role in the inflamma-
IgAV seems to be a polygenic disease and it has been described tory reaction in IgAV. Reports from Spanish and Chinese series
in parents, offspring, and siblings of affected individuals. suggest that VEGF-634G/C gene polymorphism may be associ-
Environmental factors may explain these cases (Knight 1990; Cakir ated with the development of IgAV nephritis and that C allele may
et  al. 2004); however, the familial occurrence of IgAV in differ- be a susceptible gene of IgAV nephritis (Rueda et al. 2006; Zeng
ent episodes separated by several years supports a genetic predis- et al. 2009).
position (Lofters et al. 1973). Genetic susceptibility to IgAV may Renin–angiotensin system (RAS) gene polymorphism has also
be conferred by the interaction of a number of loci, including the been associated with susceptibility and severity of IgAV. The inser-
major histocompatibility complex (MHC). Amoroso et al. (1997) tion and deletion polymorphism of angiotensin-converting enzyme
analysed 152 Italian patients with IgAV (105 with renal disease) (ACE I/D) within intron 16 has been studied in this context, as
and found that HLA-DRB1*01 and DRB1*11 were more common well as angiotensinogen (AGT) and angiotensin receptor (AGTR)
among IgAV patients than controls, while HLA DRB1*07 was less genes. Özkaya et al. (2006) found that patients with ID/DD ACE
common in IgAV patients than in controls. These authors did not genotypes had a higher risk of developing IgAV, and patients with
find significant differences in DRB, DQB, and DQA alleles between the MT/TT genotype of the AGT gene M235T had susceptibility
patients with or without renal disease. In a cohort of 110 Turkish to IgAV and nephritis. Liu et al. (2010) reported similar results. In
patients (Soylemezoglu et al. 2008), the frequency of HLA DRB1 contrast, other authors found no association (Dudley et al. 2000;
11/14 was higher in patients with IgAV compared to controls. IgAV Amoroso et al. 1998).
in a population from north-west Spain was significantly associated Association of IgAV with Familial Mediterranean Fever has been
with HLA-DRB1*01 (Amoli et al. 2001b). Another study disclosed reported by several authors, with a frequency of MEFV mutations
a weak genetic association of IgAV with HLA B35 and DR4 (Knight in Mediterranean children with IgAV ranging from 10 to 44%
1990). HLA B35 was associated with the severity of renal involve- (Gershoni et al. 2003; Bayram et al. 2011).
ment in a Spanish cohort (Amoli et al. 2002a). In contrast, Peru Genetic studies in the Lugo region of north-western Spain have
et al. (2008) observed that HLA B35 was related to disease suscep- examined the role of ICAM-1, HLA-B35, IL1RN, interleukin (IL)-
tibility but not with nephritis in a Turkish cohort. 8, IL-1β, and VEGF in disease severity (Table 39.1).
In individuals from north-western Spain, the multiallelic
(CCTTT)n polymorphism in the promoter region of the gene
encoding inducible nitric oxide synthase (NOS2A) was associated
Precipitating factors
with susceptibility to IgAV (Martin et al. 2005). The aetiology of IgAV remains unknown. Seasonal increases in
Both IgAV nephritis and IgA nephropathy have been associated the incidence of IgAV, with peaks in the spring, fall, and winter,
with deficiencies in the second and fourth components of comple- have been reported, but these findings are controversial. While in
ment (C2 and C4), and with deletion of C4 genes (Ault et al. 1990; Jin children from north-west Spain IgAV appeared more commonly
et al. 1992; Sussman et al. 1973). Jin et al. (1996) showed that locus II in fall and winter, summer and winter were the most common sea-
deletion of C4 is a risk factor for these diseases, and the deleted gene sons of onset in adults from the same region (Garcia-Porrúa et al.
can be either C4A or C4B. An increased frequency of DQA1*0301 2002). This may be related to a higher frequency of upper respira-
was present among these patients, suggesting that this allele could also tory tract infections (URI) in children, in particular during the
be a genetic risk factor for these diseases. More recently, Stefansson coldest months of the year. The site of infection varies: pharyngitis,
Thors et al. (2005) reported that C4 null alleles were found in 66% of rhinopharyngitis, and tracheobronchitis are frequently observed.
56 IgAV patients from Iceland compared with only 41% of 98 blood Other foci of infection, such as the skin, have been considered
donors used as controls. This difference was due to an increased fre- uncommon. In north-western Spain, URIs in children with IgAV
quency of C4B*Q0 allele in the group of IgAV patients. were almost twice as frequent as in adults (Garcia-Porrúa et  al.
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 529

2002). In some series, infection was a presumed precipitant in more without proteinuria, compared with only 39% of children without
than 50% of cases. A URI preceding the onset of the disease was URI (González-Gay et al. 2004). Helicobacter pylori (H. pylori) has
reported to occur in 32 (41%) of 78 children with IgAV in the Lugo also been implicated in the gastrointestinal and extragastrointesti-
region of north-western Spain (González-Gay et al. 2004). Among nal manifestations of IgAV. IgG antibodies to H. pylori were pre-
them, 15 children had tonsillitis, two had tracheitis, and 15 had sent, mostly in acute IgAV compared with controls. Ratios of IgA/
a history of influenza with pharyngitis or rhinitis (González-Gay IgG antibodies to H. pylori were significantly higher in the remit-
et al. 2004). In adults, Debray et al. (1971) reported an infectious ting phase (Novak et al. 2003).
precipitating event in 27% of patients. In north-western Spain Some drugs, especially penicillin, ampicillin and erythromycin,
(Garcia-Porrúa and González-Gay 1999a), underlying URIs occur- paracetamol, and non-steroidal anti-inflammatory drugs have
ring within 7 days before the onset of the disease were observed in been thought to precipitate IgAV (Blanco et al. 1997b; Calviño et al.
four of 27 adults (18.5%) with IgAV. In a controlled study, Farley 2001; Garcia-Porrúa and González-Gay 1999a; Garcia-Porrúa et al.
et  al. (1989) observed throat infections in 52% of patients with 1999; González-Gay et al. 2004). A history of use of these drugs
IgAV, but in only 22% of controls; almost 60% of the throat infec- was reported to occur in 30% children. These drugs were generally
tions were streptococcal. prescribed because of the presence of URI. A history of medica-
In general, an infection precedes the onset of the disease by sev- tion use was not associated with worse outcome (González-Gay
eral days to weeks. The organism most frequently isolated is the et al. 2004). Exposures to cold, insect bites, a variety of vaccines,
beta-haemolytic streptococcus group A (pyogenes) (Ballard et al. and food allergens have also been implicated as potential triggers.
1970; Farley et  al. 1989). Other organisms implicated in IgAV One report describes the potential role of pregnancy as a trigger for
include:  other streptococci, Staphylococcus aureus, Escherichia IgAV or its recurrence (Cummins et al. 2003).
coli, Mycobacterium tuberculosis, yersinia species, legionella spe-
cies, Mycoplasma pneumoniae, Epstein–Barr virus, hepatitis B Clinical manifestations
virus, varicella, adenovirus, cytomegalovirus, and parvovirus B19
(Cimolai and MacNab 1991; Pacheco 1991; Szer 1994; Somer and Cutaneous
Finegold 1995). Vaccinations against typhoid, paratyphoid A and B, A rash of erythematous papules followed by non-thrombocytopenic
measles, cholera, and yellow fever have also been implicated in the palpable purpura constitutes the most common initial manifesta-
development of IgAV (reviewed in Mormile et al. 2004). tion of IgAV. The rash occurs in all cases, but it is not always the
Mesangial deposition of nephritis-associated plasmin receptor, a presenting sign (Szer 1994; Calviño et al. 2001). The rash is more
group A streptococcal antigen, has been detected in the glomer- commonly petechial or purpuric with contiguous erythematous
uli of children with IgAV with nephritis (Masuda et al. 2003). In plaques, often associated with macular, papular, or vesicular ele-
north-western Spain, approximately 72% of children with IgAV ments. In the series from Lugo (Calviño et  al. 2001), besides
and URIs had renal involvement, manifested by haematuria with or the typical palpable purpura (Figure  39.1a), other skin lesions,

(a)

(b)

Fig. 39.1 (a) Typical palpable purpura in dependent areas of the lower extremities. (b) The purpuric rash does not blanch (go white) when pressure is applied, for
example by the glass test.
530 SECTION 5 vasculitic diseases and syndromes and related disorders

In young children, facial involvement and subcutaneous oedema


of the hands, feet, scalp, and ears are observed as early manifesta-
tions in up to 45% of cases (Dillon and Ansell 1995; Szer 1994).
Adults may experience skin necrosis in areas of severe haemor-
rhage that are subjected to significant pressure, such as dependent
areas (Cream et al. 1970).

Gastrointestinal
Gastrointestinal (GI) symptoms are common; the main gastrointes-
tinal complications include abdominal pain, nausea and vomiting,
and GI bleeding manifest as melena or haematemesis, sometimes
with massive haemorrhage and shock. Children may suffer acute
intussusception. Other GI manifestations include bowel infarc-
tion and perforation; duodenojejunal stenosis; malabsorption and
exudative enteropathy; and intestinal serositis and haemorrhagic
Fig. 39.2 A bedridden boy with palpable purpura in a dependent and ascites.
pressure-bearing area of the buttocks. Abdominal pain constitutes the second most frequent clini-
cal manifestation of IgAV, occurring in approximately 60–75% of
patients (Bailey et al. 1998; Calviño et al. 2001; Kraft et al. 1998).
generally macular, papular, or more rarely urticarial or vesicular, In children from north-western Spain, abdominal pain was often
were observed in 44% of children with IgAV. In all cases, how- the initial manifestation, preceding the onset of purpura by 1 to
ever, palpable purpura was the predominant cutaneous lesion. 10 days (mean 3.3 days), in 17% of the cases (Calviño et al. 2001).
The purpuric eruption evolves from red to purple, then becomes In a Chinese cohort of 115 adult patients, abdominal pain preceded
rust-coloured with a brownish hue and then fades. The rash does skin rash in 24.1% of the cases (Zhang and Huang 2008). Abdominal
not blanch when pressure is applied (Figure 39.1b). Purpura may pain is frequently colicky, sometimes severe, and is associated with
be preceded by a pruritic, urticarial rash. Palpable purpura occurs nausea and vomiting. It occurs in waves, returning as the purpu-
in dependent and pressure-bearing areas such as the lower limbs ric skin lesions spread. The pain is commonly periumbilical; it may
and buttocks (Figure 39.2). The distribution is roughly symmetrical sometimes mimic an acute abdomen. The pain is due to peritoneal
(Figure 39.3); it typically appears on the lower extremities and then or visceral purpura leading to submucosal and mucosal extravasa-
spreads to the thighs and buttocks. The upper extremities and the tion of blood and oedema fluid, which may lead to ulceration of the
abdomen are involved less frequently. The rash is usually intermit- bowel mucosa. There may eventually be bleeding into the lumen,
tently progressive and is exacerbated by prolonged standing. with occult blood in the stool. Melena and haematemesis have
been reported in up to 50% and 15% of patients, respectively (Allen
et al. 1960). Interestingly, the frequency of GI bleeding in two epi-
demiological studies of unselected children with IgAV from Lugo
(Calviño et al. 2001) and Charlottesville Virginia, USA (Saulsbury
1999)  was similar (31% in Lugo and 33% in Charlottesville). In
patients with haematemesis or melena, upper GI endoscopy usu-
ally discloses erythema, petechiae, and mucosal ulcerations and
erosions, especially in the second part of duodenum (Kato et al.
1992). Submucosal haemorrhages have been observed in the stom-
ach, duodenum, and sigmoid colon (Goldman and Lindenberg
1981) (Figure 39.4). Colonoscopy may disclose petechial colonic
lesions in the descending colon.
Acute intussusception may be observed in 1–5% of children
(Allen et al. 1960; Cull et al. 1990; Robson and Leung 1994). This
is usually ileoileal and less commonly ileocolic and jejunojejunal
(MacPherson 1974). Perforation is a rare, but serious, complica-
tion. Severe cases may proceed to haemorrhage and shock or even
to duodenojejunal stenosis (Allen et al. 1960; Boyer et al. 1978).
Other uncommon complications such as malabsorption, exuda-
tive enteropathy, intestinal serositis, and haemorrhagic ascites have
been reported (Archimandritis et al. 1994; Jones et al. 1966; Roy
1972; Santiago et al. 1996).
Radiological changes include thickening of mucosal folds, pro-
longation of transit time, and increased distance between loops
of small intestine. Abdominal ultrasonography may be the imag-
ing modality of choice to investigate intestinal manifestations of
Fig. 39.3 Symmetrical distribution of cutaneous lesions in legs. IgAV, such as thickening in the bowel wall, reduced peristalsis,
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 531

Fig. 39.4 An adult with IgAV manifested as erythema, oedema, and haemorrhagic lesions involving the duodenal mucosa.

intussusception, and free peritoneal fluid (Connolly and O’Halpin 90% of cases during 6-month follow-up. In most cases, the pattern
1994). Ozdemir et al. (1995) reported the use of sonography in the of joint involvement was oligoarthritis involving feet and ankles or
diagnosis and follow-up of children with IgAV. They observed dila- knees and, less commonly, upper extremity joints (Calviño et al.
tation of intestinal segments, hypomotility, eccentric thickening of 2001; Jauhola et al. 2010b). Although Duquesnoy (1991) reported
the wall of the small intestine, centrally located intestinal contents, that joint manifestations were more common in adults than in
and intramural haemorrhage. Ultrasonography may also be useful children, we have observed arthritis more commonly in children
in differentiating IgAV from other non-haemorrhagic GI diseases (64%) than in adults (23%) (Garcia-Porrúa et al. 2002). Joint mani-
(Ozdemir et  al. 1995). Jeong et  al. (1997) have emphasized the festations are generally transient; permanent deformities are rarely
importance of computed tomography (CT) scan as another tool to seen. Patients may present with arthralgias without objective signs
delineate GI involvement in IgAV. CT scans may disclose multifocal of synovitis. In our series of adults with IgAV, arthralgia without
bowel wall thickening, mesenteric oedema, and vascular engorge- clinically evident synovitis was twice as common as joint inflam-
ment. Given the negative results of other studies, the wireless video mation (Garcia-Porrúa et al. 2002). In this region of Spain, arthral-
capsule endoscopy technique could be useful in the imaging diag- gias were reported in 42% of adults and in only 11% of children.
nosis of small bowel complications (Stancanelli et al. 2006; Daniel Arthralgias or arthritis of the knees and ankles (Garcia-Porrúa
et al. 2006; De Angelis et al. 2007). Hepatosplenomegaly has also and González-Gay 1999a) may be associated with oedema; upper
been reported in patients with IgAV (Kraft et al. 1998). extremity joints such as elbows and wrists may also be involved
(Garcia-Porrúa and González-Gay 1999a; Calviño et al. 2001).
Joints
Joint manifestations have been reported to precede the development Renal
of palpable purpura by several days in up to 25% of cases (Piette Renal involvement constitutes the most serious feature of IgAV,
1994). Farley et al. (1989) observed in some cases a delay between and occurs in 20–80% of patients (Chang et al. 2005; Garcia-Porrúa
the onset of joint manifestations and the occurrence of cutaneous et  al. 2002; Kaku et  al. 1998), most commonly in older children
lesions ranging from 1–75  days, with an average of 5  days. The and adults. In unselected patients with IgAV from north-western
proportion of patients reported to have joint involvement during Spain the incidence of nephritis, manifest as haematuria, with
the course of the disease varies between 43 and 95% (Chang et al. or without proteinuria, was similar in children (54%) (Calviño
2004; Huber et al. 2004). In a prospective study of 223 children et al. 2001) and adults (~50%) (Garcia-Porrúa and González-Gay
with IgAV, Jauhola et al. (2010b) observed joint manifestations in 1999a). Renal sequelae, characterized by nephrotic syndrome or
532 SECTION 5 vasculitic diseases and syndromes and related disorders

renal insufficiency, were more common in adults (Garcia-Porrúa In most cases, the severity of disease at onset appears to predict
et al. 2002). Renal complications are infrequent in children younger outcome. The presence of both proteinuria and haematuria at the
than 2  years, where it occurs in no more than 25% of the cases. onset of the disease may be linked to progression to renal insuffi-
Renal disease may be more common than is clinically apparent, as ciency (Kraft et al. 1998). In 69 children from Lugo, Spain, develop-
minor degrees of focal mesangial proliferation have been reported ment of nephrotic syndrome within the first 3 months of disease was
in the absence of urinary abnormalities (White 1994). the best predictor of renal sequelae during the extended follow-up
Renal involvement generally occurs within the first 3  months (Calviño et al. 2001). Coppo et al. (2006) reported that the risk for
after clinical onset of the disease (Calviño et al. 2001); however, it progression of IgAV nephritis in children and adults was associated
may be observed later, generally in the setting of relapses of palpa- with increasing mean proteinuria levels during follow-up. More
ble purpura. In keeping with these observations, in a Finnish pro- recently, Wakaki et al. (2011) retrospectively analysed 42 children
spective study of 223 IgAV children, Jauhola et al. (2010a) reported with nephrotic state at disease onset. They found that a nephrotic
that 87% of patients developing nephritis experienced this compli- state lasting for more than 3 months was an independent predictor
cation within the first month after the onset of disease, and 14% of poor long-term outcome. In a long-term study of 78 children with
and 2% after 1 and 2 months, respectively. Nephropathy in patients IgAV with nephritis, Goldstein et al. (1992) observed that 44% of
with IgAV usually presents with gross haematuria lasting a few days those with nephritic syndrome, nephrotic syndrome, or both at the
(although it may persist for several weeks), followed by microscopic onset of the disease had long-term impairment of renal function. In
haematuria that may persist for months or even years. Recurrences another study of 114 children with IgAV nephritis the presence of
of gross haematuria may be observed in association with relapses of nephrotic syndrome, decreased factor XIII activity, hypertension,
skin lesions. Recurrences of haematuria long after other manifesta- and renal failure at onset were more frequently observed in those
tions of IgAV have resolved are less common. Haematuria may be who had a bad outcome at last follow-up (Kawasaki et al. 2003).
isolated or associated with proteinuria. Based on the morphological classification of IgAV glomerulone-
Persistent, severe proteinuria accompanied by microscopic hae- phritis in the International Study for Kidney Disease in Children
maturia suggests the presence of glomerulonephritis. Initial haema- (ISKDC), in which renal biopsy findings may be graded from grade
turia may be accompanied by hypertension and elevation of serum I to VI (Counahan et al. 1977), Goldstein et al. (1992) correlated
creatinine (Meadow et al. 1972). In addition to nephrotic syndrome outcomes with the severity of the lesions on initial renal biopsy.
(Niaudet and Habib 1989), the most severe clinical presentation is In this study, 58% of the children in whom more than half of the
that of mixed nephritic and nephrotic syndromes, with haematu- glomeruli were affected by crescent formation (corresponding to
ria, hypertension and renal insufficiency, severe proteinuria, and grades IV and V of the ISKDC classification) had a poor outcome.
hypoalbuminaemia. In IgAV nephritis the glomerular filtration Of those with crescents in less than one-half of glomeruli (grade
rate is moderately reduced early and is more impaired in patients III) or with no crescents (grades I and II), the outcome was poor
with proteinuria and more advanced histopathological changes on in only 17%. Other studies have confirmed that children with
kidney biopsy (Halling et al. 2005). glomerular crescents have a high chance of developing end-stage
The long-term morbidity and mortality of IgAV are almost com- renal disease (Lanzkowsky et al. 1992; Shenoy et al. 2007). Kaku
pletely due to renal involvement. Many early reports documented et al. (1998) studied factors associated with the progression of renal
a relatively high frequency of chronic renal failure in children and involvement in children with IgAV. Renal disease appeared in 63
adults, but these suffered from selection bias, as these patients were of their 194 children between 3 days and 17 months after onset of
usually referred because of kidney dysfunction (Counahan et  al. IgAV. The probability of renal involvement in IgAV was influenced
1977; Fogazzi et al. 1989; Lee et al. 1986; Meadow et al. 1972; Roth by the presence of severe abdominal symptoms, persistent pur-
et al. 1985). Garcia-Porrúa et al. (2002) observed that the frequency pura, and decreased activity of coagulation factor XIII. In contrast,
of severe renal manifestations or renal insufficiency during the treatment with glucocorticoids reduced the risk of nephritis. An
course of the disease was significantly lower in children than adults. increased risk of developing renal insufficiency was associated with
Other studies (Blanco et al. 1997b; Uthman et al. 1998) also found age at onset of more than 7 years, the presence of persistent pro-
a higher frequency of severe renal involvement in adults. The final teinuria, and decreased activity of coagulation factor XIII. In the
outcome in both age groups is good in most cases. After 6 years’ Finnish series (Jauhola et al. 2010a), an age over 8 years at onset,
median follow-up, complete recovery was observed in 65 of 73 chil- abdominal pain, and recurrence of IgAV disease were independent
dren; however, after 5  years’ median follow-up almost 40% of 31 risk factors for developing nephritis. Furthermore, accumulating
adults had persistent haematuria and three of them (10%) had renal two or three risks factors increased the risk of nephritis.
insufficiency, two of whom required haemodialysis (Garcia-Porrúa Data on adults with IgAV nephritis differ from one series to
et al. 2002). another. In Italian adults with nephritis, Fogazzi et  al. (1989)
The urine should be routinely tested for blood and protein at observed a high rate of renal function deterioration (11 of 16 adults
onset and during the follow-up until systemic signs have resolved. with a mean follow-up of 90.5  months). Although patients who
Proteinuria, if present, should be quantified. Narchi (2005) reviewed developed renal insufficiency had a higher percentage of crescents
12 studies that included 1133 children with IgAV to assess the risk at the disease onset, no clinical features at presentation predicted
of long-term renal impairment. Haematuria and proteinuria were the course of the disease (Fogazzi et al. 1989). In fact, in this series
found in 34% of the children, occurring in 85% of cases within 4 most adults with IgAV who had normal renal function at onset sub-
weeks of the diagnosis of IgAV, in 91% within 6 weeks, and in 97% sequently developed renal insufficiency. In a Spanish series of 28
within 6 months. Thus, if there are no abnormalities on the urinaly- adults with long-term follow-up of IgAV, haematuria at the onset of
sis within the first 6 months of the disease, urinalysis testing need the disease, or renal manifestations during the course of the disease
not be performed thereafter (Narchi 2005). (frequently observed in patients with relapses), were the main risk
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 533

factors for renal sequelae (Garcia-Porrúa et al. 2001). Rauta et al. Laboratory investigation
(2002) reported 38 Finnish adults with IgAV nephritis. Renal dete-
rioration occurred in 21%. Proteinuria greater than 1 g/24 hours Elevation of acute phase reactants, moderate leukocytosis, and
was the only factor related to progression. Pillebout et  al. (2002) thrombocytosis are common during the active phase of disease.
reported a retrospective 250 French adult series with IgAV nephritis The haemoglobin is generally normal unless severe GI or pul-
and a mean follow-up period of 14.8 years. Renal function was nor- monary bleeding occurs. A pathogenic role for IgA is supported
mal in 68% at presentation; however, only 20% achieved remission. by its presence in cryoprecipitate (see following paragraph),
Noteworthy, 26% died and only 11% developed end-stage renal fail- (Garcia Fuentes et al. 1977) and the finding of an increased num-
ure. Independent risk factors for severe renal failure were creatinine ber of circulating IgA-secreting cells in patients with active IgAV
and proteinuria levels at presentation and the proportions of inter- (Casanueva et al. 1983; Levinsky and Barratt 1979; Trygstad and
stitial fibrosis, glomerular fibrinoid necrosis, and glomerular sclero- Stiehm 1971). IgA-dominant immune deposits are observed in
sis on renal biopsy. In UK adults with IgAV nephritis, Shrestha et al. many cases of IgAV (Hené et al. 1986), and their presence has been
(2006) observed a high rate of end-stage renal failure (27%) with included in the definition adopted by the Consensus Conference
also a high rate of remissions (43%). Proteinuria >1 g/24 h during on the Nomenclature of Systemic Vasculitides (Jennette et al. 1994).
follow-up was a predictor of end-stage renal failure. Although IgA levels in the serum are reportedly increased in 50%
of children in acute phases of disease (Dillon and Ansell 1995), they
Other clinical manifestations were increased in only 5.6% of the tested patients in the American
Other features of IgAV, which are rare or under-reported, are College of Rheumatology (ACR) study database (Mills et al. 1990).
summarized in this section. Extrarenal genitourinary problems In Spain, increased values of serum IgA were only observed in 10
sometimes precede the onset of purpuric lesions (Szer 1994). of 27 (37%) adults and 20 of 37 (54%) children (Garcia-Porrúa
Acute scrotal swelling due to inflammation and haemorrhage was et al. 2002).
observed in 32% of the series of boys reported by Chamberlain and Circulating IgA-containing immune complexes and cryoglobu-
Greenberg (1992). These authors emphasized the need to differ- lins have also been found (Blockmans and Bobbaers 1998; Levinsky
entiate this complication from testicular torsion of the spermatic and Barratt 1979; Trygstad and Stiehm 1971). Serum C3, C4, and
cord, as it too may be the initial manifestation of IgAV. In patients CH50 are generally normal; however, in a series of patients with
with testicular torsion, both Doppler flow and radionuclide scan IgAV and IgA nephropathy the measurement of plasma anaphyla-
are abnormal, while in children with scrotal swelling due to toxins C3a and C4a showed significant correlation with plasma cre-
IgAV, Doppler and radionuclide scan show normal or increased atinine and urea values (Abou-Ragheb et al. 1992). This suggests a
blood flow. Orchitis has also been reported in IgAV (Gibson and role for plasma anaphylatoxin determination as a sensitive indica-
Su 1995). tor of complement activation and a useful parameter in monitoring
Neurological complications are probably more common in IgAV IgAV activity. Complement activation through both the alternative
patients than are generally described. Central nervous system and lectin pathways has been found in patients with IgAV nephritis
(CNS) involvement usually causes headache, which Ostergaard (Hisano et al. 2005).
and Storm (1991) reported in 31% of 26 cases. Other neurologi- Antineutrophil cytoplasmic antibodies (ANCA) have been iden-
cal problems include: behavioural changes due to encephalopathy, tified in a wide variety of vasculitic disorders, but the presence of
changes in mental status, apathy, hyperactivity, mood swings, and ANCA in sera of patients with IgAV is controversial. Van den Wall
seizures (Fielding et al. 1998; Woolfenden et al. 1998). Subdural Bake et  al. (1987) suggested a role for ANCA in the pathogene-
haematomas, intracranial haemorrhage, intraparenchymal bleed- sis of IgAV, as they detected IgA ANCA in 55% of their patients.
ing, and non-haemorrhagic vasculitis of the cerebral parenchyma O’Donoghue et al. (1992) did not confirm that observation, as none
have been documented less often (Chiaretti et  al. 1995; Ha and of 30 children with early IgAV were ANCA-positive. Coppo et al.
Cha 1996; Misra et al. 2004). In a literature review (Garzoni et al. (1997) suggested that the conflicting reports on IgA ANCA might
2009) altered level of consciousness was the commonest presenta- be due to atypical characteristics of the reaction in some ELISA
tion of CNS involvement in IgAV, followed by convulsions, focal assays. Ozaltin et al. (2004) undertook a prospective study to estab-
neurological deficits, visual abnormalities, and verbal disability. lish the presence of ANCA IgA subclass in IgAV. IgA ANCA in a
Peripheral nervous system lesions may appear as mononeuropa- cytoplasmic pattern was detected in a higher percentage of chil-
thy involving the facial, ulnar, femoral, sciatic, or peroneal nerves; dren diagnosed with IgAV (82% of 35) in the acute phase compared
polyneuropathy such as Guillain–Barré syndrome, polyradiculo- to children with other vasculitides (38% of 13). In the resolution
neuropathy, and brachial plexopathy have been reported (Robson phase of IgAV, IgA ANCA was negative in 88% of patients. No
and Leung 1994). relationship was found between disease severity of IgAV and IgA
Interstitial pulmonary disease is common, but it is generally ANCA. Accordingly, IgA ANCA testing might be useful to confirm
asymptomatic. Impairment of lung diffusion capacity has been the diagnosis of IgAV in children (Ozaltin et al. 2004). High titres
reported in the majority of children during the active phase of of serum antiphospholipid antibodies have been described in adult
the disease (Chaussain et  al. 1992). A  few cases of pulmonary patients with IgAV. IgA anticardiolipin antibodies and antiphos-
haemorrhage consisting of diffuse alveolar haemorrhage, lead- phatidylserine–prothrombin complex showed a significant corre-
ing in some cases to death, or more rarely interstitial pneumonia, lation with serum IgA, C-reactive protein levels, and proteinuria
have been described (Olson et al. 1992; Paller et al. 1997; Nadrous (Kawakami et al. 2006; Kawakami et al. 2008). However, the patho-
et al. 2004). Other manifestations such as malaise and low-grade genic role of antiphospholipid antibodies in IgAV is not clear.
fever are observed in half of the patients with IgAV (Robson and As with other vasculitides, and as a consequence of vascular
Leung 1994). endothelial damage, high concentrations of von Willebrand factor
534 SECTION 5 vasculitic diseases and syndromes and related disorders

have been observed (Ates et al. 1994; De Mattia et al. 1995), suggest- confirmed complement activation through both the alternative and
ing its possible use as a marker of disease severity. Elevated levels lectin pathways in patients with IgAV nephritis and they showed
of serum thrombomodulin, derived from damaged endothelium, that complement activation is promoted in situ in the glomerulus.
have also been observed in patients with IgAV nephritis (Fujieda In IgAV patients with nephritis, complement activation through
et al. 1998). The prothrombin time and partial thromboplastin time the lectin pathway may play a role in the development of advanced
are typically normal. Reduced factor XIII activity has been found in glomerular injuries and prolonged urinary abnormalities (Hisano
patients with severe GI complications (De Mattia et al. 1995). et  al. 2005). Mast cells and eosinophil activation could also play
a role in the pathogenesis of IgAV nephritis. Children with active
Pathophysiology IgAV nephritis have higher levels of serum eosinophil cationic
protein in comparison with those in remission and controls, and
In IgAV, IgA-dominant immune deposits are observed in the wall kidney infiltration of mast cells correlated with renal histological
of the small vessels and in the renal glomeruli. IgA1 accounts for severity (Chen et al. 2006). Alpha smooth muscle actin (α-SMA)
80–90% of serum IgA. Secretory IgA is composed of roughly equal predominates within vascular smooth muscle cells and plays an
proportions of IgA1 and IgA2 (Saulsbury 1999). Aberrant glycosyla- important role in fibrogenesis. Increased renal expression of α
tion of the hinge region of IgA1 may be responsible for the clinical -SMA has also been associated with progression of renal injury in
and histopathological features of IgAV (Saulsbury 1999; Saulsbury patients with IgAV nephritis (Kawasaki et al. 2008). Finally, proin-
2001). Although the pathogenic role of IgA is widely accepted, the flammatory cytokines seems to be involved in the pathogenesis of
diagnostic significance of IgA vascular immune deposits detected nephritis in IgAV. In the acute phase of the disease, serum TNF-α
by direct immunofluorescence is not completely defined, as similar levels were significantly higher in IgAV with proteinuria compared
immune deposits have been observed in patients with other types to those without renal involvement. These observations suggest
of cutaneous vasculitis (Sams et al. 1975). Helander et al. (1995) that increased TNF-α levels in the serum are involved in the acute
found that the presence of vascular deposits of IgA was sensi- phase of glomerular disease, and may correlate with disease activity
tive, but not specific for diagnosis. Nevertheless, the presence of of IgAV in patients with severe renal involvement (Ha 2005).
IgA immune deposits, when combined with selected clinical data
such as GI involvement, previous history of URI, or age less than
20  years, improved the diagnostic accuracy of IgAV (Helander Histology
et al. 1995). Skin
Increased IgA may be due to increased production or decreased Consonant with other small-sized blood vessel vasculitides involv-
clearance. An unknown antigen may stimulate IgA production, ing the skin (González-Gay et al. 2004; González-Gay et al. 2005),
activating pathways leading to necrotizing vasculitis. In addi- cutaneous lesions of patients with IgAV generally involve capil-
tion to elevations in serum IgA concentration, there may be laries, postcapillary venules, and non-muscular arterioles (less
IgA-containing immune complexes and IgA isotype autoantibod- than 50 µ in diameter) found mainly within the superficial papil-
ies (rheumatoid factor, ANCA, and antiendothelial cell antibodies) lary dermis (Figure 39.5). Biopsy samples of the cutaneous lesions
(Tizard 1999). IgA deposits are present in both skin and glomeruli. in patients with IgAV show a leukocytoclastic vasculitis of small
The IgA immune complexes are capable of activating comple- blood vessels with infiltration of neutrophils within and around
ment, leading to the formation of chemotactic factors such as C5a, vessel walls; leukocytoclasia; fibrinoid necrosis of the damaged
which in turn recruit polymorphonuclear leukocytes to the site vessel walls; necrosis, swelling, and proliferation of the endothe-
of deposition. The release of lysosomal enzymes due to the inges- lial cells; and red cell extravasation (Figure  39.6). Eosinophils
tion of immune complexes by the polymorphonuclear leukocytes may also be present. Necrosis of small blood vessels and platelet
results in vessel damage. The membrane-attack complex (MAC)
is also required for the endothelial damage; serum concentration
of the C5b-9 complex has been found to be significantly elevated
in many patients at the time of disease flare. The MAC has been
found along with IgA and C3 on the vessel walls of the skin and on
the capillary walls and mesangium of patients with IgAV nephri-
tis (Kawana and Nishiyama 1992). Measurement of the terminal
complement complex is another possibly useful tool for monitoring
disease activity in IgAV patients. Although serum C3 and C4 levels
are generally normal in most patients with IgAV, anaphylatoxins
C3a and C4a are sensitive indicators of complement activation and
indicators of disease activity in some patients with IgAV and IgA
nephritis (Abou-Ragheb et al. 1992). Complement abnormalities
such as C2 deficiency, the homozygous null C4 phenotype, and
C4B deficiency have been detected in some patients (Tizard 1999).
Although high C3d concentrations and glomerular C3 and proper-
din deposition in the acute phase of disease activity have suggested
complement activation, a study of three multimolecular comple- Fig. 39.5 Skin biopsy of a patient with IgAV showing dermal vascular neutrophilic
ment activation protein complexes failed to support complement inflammatory infiltration, leukocytoclasia, vessel wall fibrinoid necrosis, and
activation in the disease (Smith et al. 1997). Hisano et al. (2005) haematic extravasation (haematoxylin and eosin × 100).
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 535

RCE

ES

FN

Fig. 39.6 Skin biopsy disclosing small-sized blood vessel vasculitis with a Fig. 39.7 Kidney biopsy showing glomerular sclerosis and crescent formation
vessel showing fibrinoid necrosis (FN), endothelial swelling (ES), and red cell (Masson × 400).
extravasation (RCE) consistent with leukocytoclastic vasculitis (haematoxylin and
eosin × 400).

to circumferential forms. Large cellular crescents cause extensive


thrombi in capillaries and venules are common findings. IgA, C3, obliteration of the urinary spaces. In the acute phase, crescents are
and fibrin may deposit within the walls of the dermal vessels as predominantly cellular, but become fibrous after being infiltrated
well as in the connective tissue of the upper dermis of the purpuric with collagen. Adherent glomerular capillaries become sclerotic.
skin lesions (Giancomo and Tsai 1977). IgA deposits have also been The extent of tubular atrophy and interstitial cellular infiltration
found, albeit less often, in biopsy specimens of non-purpuric skin and fibrosis is a measure of the severity of the glomerular lesions
(Giancomo and Tsai 1977). (White 1994).
Immunofluorescence microscopy almost invariably reveals dif-
Gastrointestinal tract fuse distribution of mesangial deposits of IgA (Figure 39.9), which
are generally associated with minimal amounts of IgG, IgM, C3, and
Non-specific inflammatory infiltrates of lymphocytes and IgA
properdin. In patients with more severe disease, IgA deposits may
deposits in the capillaries of the stomach and duodenal mucosa
extend into the capillary walls; fibrin, which could have a role in
have been reported (Kato et al. 1992) and mild inflammation and
crescent formation, may also be seen (Heaton et al. 1977). Electron
endothelial cell proliferation have been observed in colonic biop-
microscopy shows that some deposits are found in the mesangial
sies (Cappell and Gupta 1990).
regions, especially in the perimesangium. Smaller deposits are also
Kidney observed in the subendothelial zone of the capillary walls adjacent
to the mesangiocapillary junction, and to a minor extent in the sub-
The early lesion is an endocapillary, focal or diffuse, prolifera-
epithelial region (White 1994).
tive glomerulonephritis involving both endothelial and mesan-
gial cells (Robson and Leung 1994). The glomeruli are infiltrated
with polymorphonuclear and mononuclear cells. Proliferation of
extracapillary cells, including epithelial cells and Bowman’s cap-
sule cells, may result in variable degrees of crescent formation,
due to adhesion of the proliferating cells to the glomerular tuft
(Figure 39.7). Interstitial inflammation and tubular changes with
atrophy and tubular casts are common. Disease of renal arte-
rioles occurs infrequently; therefore the brunt of the disease is
borne by the glomeruli, which characteristically show two basic
lesions: mesangial proliferative and epithelial crescent formation
(White 1994) (Figure 39.8).
Renal biopsy findings may be graded according to the classifi-
cation of the ISKDC (see Section Renal), which is based on the
percentage of glomeruli showing crescents and segmental lesions
(Counahan et  al. 1977). Renal biopsies may reveal minimal to
severe glomerulonephritis indistinguishable from IgA nephropa-
thy. When renal involvement is minimal, glomeruli appear normal
on light microscopy (Yoshikawa et al. 1981). Mesangial prolifera-
Fig. 39.8 Renal biopsy showing two glomeruli. One of them has an extracapillary
tion may be either diffuse or focal and segmental. Associated fea- proliferative crescent that obliterates the glomerular tuft. The other glomerulus
tures include thrombosis and necrosis. Epithelial crescents vary in shows diffuse mesangial proliferative involvement. (Haematoxylin and
size from small segmental wedges, affecting one or two lobules, eosin × 100.)
536 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 39.2 Criteria for differentiating IgAV from hypersensitivity


vasculitis (traditional format)

Criterion Definition
1. Palpable purpura Slightly elevated purpuric rash over one or more
areas of the skin not related to thrombocytopenia
2. Bowel angina Diffuse abdominal pain worse after meals or bowel
ischemia usually including bloody diarrhoea
3. Gastrointestinal Gastrointestinal bleeding, including melena,
bleeding haematochezia or positive test for occult blood in
the stool
4. Haematuria Gross haematuria or microhaematuria
5. Age at onset <20 years Development of first symptoms at age 20 or less
6. No medications Absence of any medications at onset of disease,
which may have been a precipitating factor

The presence of any three or more of the six criteria yields a correct classification of IgAV
cases of 87.1%. Two or fewer criteria yield a corrected classification of hypersensitivity
vasculitis cases of 74.2%.
(Data from Michael et al. (1992), with permission from Journal of Rheumatology.)

Fig. 39.9 Glomerulus from a patient with IgAV and proliferative vasculitides involving preferentially the skin, which share many
mesangial glomerulonephritis with granular mesangial deposits of IgA (IgA clinical and pathological features such as leukocytoclasia and pal-
immunofluorescence × 400). pable purpura. Furthermore, the overall sensitivity of the ACR
criteria for classification of ‘hypersensitivity’ vasculitis was low
(Calabrese et al. 1990).
Lung A considerable overlap between the ACR 1990 criteria for IgAV
In pulmonary specimens obtained at autopsy, IgA deposits have and that for leukocytoclastic vasculitis was reported. In fact, in pre-
been found along the alveolar septa (Kathuria and Cheifec 1982). vious classifications such as that proposed by Fauci et al. (1978),
IgAV was classified as a subgroup of hypersensitivity vasculitis.
Likewise, Gilliam and Smiley (1976) classified IgAV within the
Diagnosis group of leukocytoclastic, hypersensitivity, or allergic vasculitis.
The clinical diagnosis of IgAV is supported by finding leukocyto- In 1992, to compare the characteristics of leukocytoclastic vascu-
clastic vasculitis on skin biopsy, with IgA immunofluorescence in litis and IgAV as separate and definable clinical syndromes, Michel
small vessels of the skin and in the renal glomeruli. Elevation of et al. using the ACR database, investigated which clinical criteria
serum IgA may suggest a diagnosis of IgAV. Its histological picture best differentiated these two vasculitides (Michel et al. 1992). They
is not specific, since IgAV is characterized by a leukocytoclastic vas- reported that requiring the presence of three or more criteria, from
culitis that is indistinguishable from leukocytoclastic (hypersensi- a list of six, yielded correct classification of 87.1% of IgAV cases. Two
tivity) vasculitis and other systemic vasculitides involving small or fewer criteria from the same list of six criteria yielded a percentage
blood vessel of the skin (González-Gay et al. 2004; González-Gay of correctly classified leukocytoclastic vasculitis cases of 74.2%. In
et al. 2005). Table 39.2, the traditional format of criteria for differentiating IgAV
As no absolute diagnostic test is available, classification criteria from ‘hypersensitivity’ vasculitis is shown. In 1994, the Consensus
and definitions are commonly used. Classification of vasculitides is Conference on the Nomenclature of Systemic Vasculitides required
often confusing and controversial. In 1990, the American College the presence of IgA-dominant immune deposits for the defini-
of Rheumatology proposed classification criteria for IgAV (Hunder tion of IgAV (Jennette et al. 1994). Immunofluorescence, however,
et al. 1990). The criteria were presented in two forms, a traditional was not routinely performed in the series of systemic vasculitis
format and a tree format. The traditional format for the classifica- reported by Watts et al. (1998) and in that of González-Gay and
tion of IgAV included the following four features: palpable purpura García-Porrúa (1999). As expected, when Chapel Hill Consensus
defined by the presence of slightly raised skin lesions unrelated to Conference definitions were applied to some series of systemic
thrombocytopenia; age at the onset of the vasculitis younger than vasculitides (González-Gay and García-Porrúa 1999), many of the
20  years old; bowel angina consisting of diffuse abdominal pain patients classified as IgAV according to the ACR and Michel’s clas-
or bowel ischaemia, with or without bloody diarrhoea; and histo- sification criteria, fell into the category of microscopic polyangiitis.
logical changes showing granulocytes in the walls of arterioles or Furthermore, the requirement of immunofluorescence data would
venules on skin biopsy (Mills et al. 1990). For the classification of have underestimated the number of IgAV patients reported in
IgAV at least two of these four criteria were required to be present. other series (Blanco et al. 1997b).
However, both IgAV and hypersensitivity vasculitis (for which the The 1990 ACR criteria for the classification of IgAV were devel-
preferred term is leukocytoclastic) are small-sized blood vessel oped by comparing 85 patients with IgAV with 772 patients with
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 537

other forms of vasculitis. No children were included in the study. cutaneous vasculitis has traditionally been considered an impor-
In 2008, the European League Against Rheumatism (EULAR), tant criterion for differentiating leukocytoclastic vasculitis (Michel
the Paediatric Rheumatology International Trials Organisation et  al. 1992)  from IgAV. A  history of prior drug use is common
(PRINTO), and the Paediatric Rheumatology European Society in leukocytoclastic vasculitis; antecedent medication at disease
(PRES), proposed together a new classification criteria for child- onset was adopted by the ACR Subcommittee on Classification of
hood vasculitides, based on a population of 1398 children (827 with Vasculitis as a classification criterion for ‘hypersensitivity’ vascu-
IgAV) from 36 different countries (Ozen et al. 2006; Ruperto et al. litis (Calabrese et al. 1990). Although found at a lower frequency
2010; Ozen et al. 2010). According to these new criteria, to classify in IgAV than in patients with ‘hypersensitivity’ vasculitis, a history
a child as having IgAV required the presence of a typical palpable of recent drug intake has also been implicated in its pathogenesis
purpura or petechiae with predominant lower limb involvement (Blanco et al. 1997b; Garcia-Porrúa et al. 1999).
plus the presence of one of the following four criteria:  abdomi- As with leukocytoclastic vasculitis and mixed cryoglobulinae-
nal pain, defined as an acute onset, diffuse, colic pain; arthritis or mia, IgAV has been related to bacterial and viral infections (Somer
arthralgia of acute onset; renal involvement defined by the presence and Finegold 1995). In the majority of series, Gram-positive cocci
of proteinuria (> 0.3 g/24h), or haematuria (> 5 red blood cells/ (staphylococcus and streptococcus species) (Saulsbury 1999;
high power field); and histopathology showing leukocytoclastic Calviño et  al. 2001)  and Neisseria meningitidis and gonorrhoeae
vasculitis or proliferative glomerulonephritis, with predominant were the most common bacteria implicated in this process; how-
IgA deposits. However, when skin lesion had no typical distribu- ever, IgAV has been reported in the context of infections with
tion a biopsy was mandatory to establish the diagnosis. Applying Gram-negative bacteria, anaerobes, mycobacteria, and brucella
these set of criteria the authors found a sensitivity and specificity of (Pacheco 1991). Parvovirus B19 has been related to IgAV in anec-
100% and 87%, respectively. dotal reports, but this association was uncommon in a case–control
study (Ferguson et al. 1996). If some cases of IgAV are caused by
infection, responsible mechanisms might include immune com-
Conditions associated with IgAV plex formation and activation initiated by antigen products of the
Drugs (Calviño et  al. 2001; Garcia-Porrúa et  al. 1999), malig- responsible organisms, or abnormal immunoregulation related to
nancies (Garcia-Porrúa and González-Gay 1998), and infections the infectious disease (Garcia-Porrúa and González-Gay 1999b).
(Garcia-Porrúa and González-Gay 1999b; González-Gay and
Garcia-Porrúa 1999) have been associated with IgAV. Cutaneous Differences between IgAV
vasculitis, including IgAV, may be associated with malignant dis-
orders and behave as a paraneoplastic syndrome (González-Gay in children and adults
et al. 2000); however, there is no consensus about the actual pro- A few studies have discussed clinical and epidemiological differ-
portion of patients with IgAV and malignancy. Gibson and Su ences of IgAV according to the age of onset (Blanco et al. 1997b;
(1995) reported a frequency of 8% of cutaneous vasculitis associ- Coppo et al. 1997; Garcia-Porrúa et al. 2002; Ilan and Naparstek
ated with malignancy. Cutaneous vasculitis may antedate the dis- 1991; Lin et  al. 1998; Uthman et  al. 1998). Ilan and Naparstek
covery of the neoplasia, coincide, or appear after the malignancy (1991) did not find significant differences in clinical and laboratory
has already been recognized; it may also indicate recurrence of findings between children and adult patients with IgAV from Israel.
cancer (Fortin 1996; García-Porrúa and González-Gay 1998; In their series, renal failure was observed in two of the 15 adults and
González-Gay et al. 2000; Mertz and Conn 1992; Sánchez-Guerrero in five of the 46 children during the course of the disease, but none
et al. 1990). Haematological disorders constituted the most com- developed chronic renal failure. Coppo et al. (1997) compared the
mon group of neoplasms associated with cutaneous vasculitis long-term evolution of IgAV nephritis in adults and children. No
(Fortin 1996; García-Porrúa and González-Gay 1998; Mertz and differences in the frequency of impaired renal function at onset,
Conn 1992). Cutaneous vasculitis associated with solid tumours or in the long-term renal outcome were observed. In contrast,
often fulfils criteria for the diagnosis of IgAV (Kurzrock et al. 1994; other investigators reported a more severe disease in adults. In a
Sánchez-Guerrero et  al. 1990; Garcia-Porrúa and González-Gay retrospective study of patients with IgAV classified according to
1998; Weiler-Bisig et  al. 2005). In a retrospective cohort of 250 the criteria proposed by Michel et al. (1992), Blanco et al. (1997b)
IgAV adults with a mean follow-up of 14.8 years, Pillebout et al. observed more frequent and severe renal disease in patients older
(2002) reported cancer as the leading cause of mortality (27% of than 20  years than in those younger than 20. Similarly, Uthman
deaths). et al. (1998) found a statistically significant difference in the inci-
There are several possible mechanisms accounting for either dence of nephropathy between adults and children, with two of 10
IgAV or other cutaneous vasculitis in the setting of malignancy adults (age more than 20) developing renal failure compared with
(González-Gay et al. 2000), such as impaired clearance of normally none of 35 children. A Chinese study also found that renal involve-
produced immune complexes. Alternatively, there may be abnor- ment was more common and severe in adults with IgAV (Lin and
mal production of immunoglobulins that react either with vascular Huang 1998). To establish clear differences in terms of age at dis-
antigens (causing formation of in situ immune complexes) or with ease onset in Spain, individuals older than 20 years (considered as
a circulating antigen (forming circulating immune complexes that adults) were compared with those younger than 14 years. In this
then deposit in the vessel walls). Finally, there may be production series adults complained of arthralgias more often than children
of immunoglobulins directed to the abnormal tumour cells as well but swelling in the joints was observed more frequently in chil-
as normal endothelium. dren. The frequency of GI manifestations was similar in both age
Drugs are often implicated in the development of cutaneous groups. During the course of disease, six of the 31 adults (19%)
vasculitis. A  history of drug exposure prior to the onset of the from Spain had severe renal manifestations and another four (13%)
538 SECTION 5 vasculitic diseases and syndromes and related disorders

renal insufficiency, whereas in 73 children from the same region to occur more commonly in the setting of a flare of the disease
the frequency of severe renal manifestations was 8% and of renal (González-Gay and García-Porrúa 1999).
insufficiency 3%. After a median follow-up period of approximately A careful clinical history in patients presenting with palpable
6 years, complete recovery was observed in 89% of the 73 children. purpura may be useful in differentiating vasculitic skin lesions from
None of the children developed chronic renal insufficiency and the those produced by trauma, drug reactions, or physical trauma such
few who developed renal insufficiency during the course of the dis- as that seen in child abuse (González-Gay et  al. 2004). Purpuric
ease had improved renal function at last follow-up. After 5 years’ rash may also be due to sepsis, bacterial endocarditis, meningococ-
median follow-up almost 40% of adults had persistent haematuria, caemia, Rocky Mountain spotted fever, or other infections (Stevens
three (10%) had renal insufficiency, of whom two required haemo- et  al. 1995). Thrombotic disorders, such as the antiphospholipid
dialysis (Garcia-Porrúa et al. 2002). Relapses were also more com- antibody syndrome or thromboembolism, thrombocytopenia,
mon in adults but the difference was not statistically significant pseudovasculitic lesions such as atheroembolic disease, neoplasms
(Garcia-Porrúa et al. 2002). Complete recovery was reported in 107 such as the cardiac myxoma, or scurvy, may also cause purpuric
of the 114 children and 33 of the 38 adults studied by Blanco et al. cutaneous lesions and mimic vasculitis (González-Gay et al. 1999;
(1997b). Among these, persistent microscopic haematuria alone González-Gay et al. 2004).
was observed in six children and mild renal insufficiency in four Abdominal pain, particularly in children, in which it often
adults (Blanco et al. 1997b). precedes cutaneous lesions, prompts the consideration of acute
Adults from the Lugo region of Spain developed the disease dur- abdomen (Glasier et  al. 1981). IgA glomerulonephritis is not
ing the summer months more frequently than did children. The pathognomonic of IgAV; rheumatic diseases other than SLE (seron-
paediatric disease occurred more commonly in the coldest months egative spondyloarthropathy), dermatological diseases (psoriasis
of the year (Garcia-Porrúa et  al. 2002). These observations were or dermatitis herpetiformis), neoplasms, and intestinal disorders
in keeping with the data reported in Israel by Ilan and Naparstek (celiac sprue or hepatic cirrhosis) have been infrequently associ-
(1991). ated with this type of glomerulonephritis. These conditions can be
clearly differentiated from IgA glomerulonephritis in the context
of IgAV, or from idiopathic IgA glomerulonephritis by clinical fea-
Differential diagnosis tures (Galla 1995).
Diagnosis of IgAV is straightforward in children presenting with Finally, in infants younger than 2 years there is a syndrome called
the classic triad of palpable purpura, arthritis, and GI manifesta- acute haemorrhagic oedema that has been reported most often
tions or haematuria. IgAV is more common in children, and leu- from Europe. It is characterized by a cutaneous leukocytoclastic
kocytoclastic vasculitis is more common in adults (González-Gay vasculitis and oedema, and ecchymotic, target-like purpura on the
et  al. 2004; González-Gay et  al. 2005). IgAV in adults tends to limbs and face. In many of the infants with this syndrome there is
involve organs more extensively, and the prognosis appears to be a history of recent illness, use of drugs, or immunizations. Visceral
worse than in those with leukocytoclastic vasculitis (García-Porrúa involvement is the exception, and spontaneous recovery occurs
and González-Gay 1999a). The clinical criteria proposed by Michel generally within 1–3 weeks. It is not clear whether this syndrome
et  al. are useful in differentiating IgAV from leukocytoclastic should be considered a benign infantile form of IgAV or an inde-
vasculitis. pendent disease within the spectrum of conditions presenting with
As in IgAV, other systemic vasculitides such as microscopic leukocytoclastic vasculitis (Athreya 1996; Ince et al. 1995).
polyangiitis (MPA) and granulomatosis with polyangiitis (GPA,
Wegener’s granulomatosis) may present with skin lesions and cres-
centic glomerulonephritis. In addition to other clinical features, Treatment
the presence of ANCA may be useful in differentiating these con- There is no specific treatment for IgAV. Indeed, IgAV in most cases
ditions. Perinuclear ANCA (pANCA) is found frequently in MPA is a self-limiting disease (Garcia-Porrúa et al. 2002). Treatment is
while cytoplasmic (cANCA) is more commonly observed in GPA largely supportive and, besides bed rest, includes adequate hydra-
(Gross et al. 1993; Guillevin et al. 1996; Rao et al. 1995; Specks and tion and monitoring of vital signs. Non-steroidal anti-inflammatory
Homburger 1994). drugs can improve joint pain, but they should be avoided in patients
Patients with mixed cryoglobulinaemia and cryoglobulinaemic with renal insufficiency.
vasculitis may present with recurrent episodes of purpuric skin There is no consensus on the efficacy of glucocorticoid therapy to
lesions, frequently associated with visceral involvement, for exam- prevent severe complications in children with IgAV and use of glu-
ple glomerulonephritis and neuropathy. Thus, patients should cocorticoids in relapses is controversial (González-Gay and Llorca
be tested for cryoglobulin, C4 and C3 complement serum levels, 2005). Glucocorticoids have been used, although not without con-
rheumatoid factor, and hepatitis C and B virus (González-Gay troversy, in patients with severe GI manifestations, such as abdomi-
et al. 2004). nal pain (Szer 1996). Abdominal pain improves in some children
Connective tissue diseases, in particular systemic lupus ery- independent of glucocorticoids (Glasier et  al. 1981; Rosenblum
thematosus (SLE), may present with vasculitic skin lesions and Winter 1987). Rosenblum and Winter, however, described
(González-Gay and Garcia-Porrúa 1999) and systemic symptoms. improvement of abdominal pain within the first 24 hours of glu-
Although antinuclear antibodies (ANA) have been reported in cocorticoid therapy in almost 50% of children, compared with a
some patients with IgAV (Saulsbury 1986), ANA and anti-dsDNA spontaneous resolution of only 14%. Many clinicians use glucocor-
antibodies, and low levels of serum C3 and C4, differentiate ticoids in all cases of IgAV with abdominal pain. It appears that the
patients with SLE from those with IgAV. Furthermore, leukocyto- risk of intussusception in children with abdominal pain is lower
clastic cutaneous vasculitis in patients with SLE has been reported in those treated with glucocorticoids. Children and adults with
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 539

IgAV with severe abdominal pain, melena, and massive haemor- In most cases the absence of randomized, placebo-controlled
rhage are routinely and, in general, successfully treated with these studies makes valid conclusions difficult. Based on a series of 20
drugs (Blanco et al. 1997b; Calviño et al. 2002; García-Porrúa and patients with IgAV nephritis, Shin et  al. (2005b) suggested that
González-Gay 1999a). Marked improvement of abdominal pain combined therapy of azathioprine and glucocorticoids might be
was observed in a patient immediately after the administration of beneficial in improving histopathological features and the clini-
factor XIII concentrate (Shimomura et al. 2005). cal course of IgAV patients with severe nephritis. An uncontrolled
Another important point of contention is the role of glucocorti- study of combined glucocorticoids, immunosuppressive drugs,
coids in the prevention of nephritis. Mollica et al. (1992) reported a and anticoagulants in patients with rapidly progressive glomeru-
prospective, randomized, controlled study on glucocorticoid pre- lonephritis due to various causes, including IgAV, suggested that
vention of renal disease in children with IgAV. One hundred and deterioration of renal function could be retarded if the patient was
sixty-eight children with IgAV without nephritis were randomized not severely oliguric (Brown et al. 1974). Niaudet and Habib (1998)
to receive either prednisone (1 mg/kg per day orally for 2 weeks) evaluated the efficacy of methylprednisolone pulse therapy on the
or no glucocorticoids. None of the treated group (n = 84), but 10 of outcome of nephropathy in 38 children with severe forms of IgAV.
the control group (n = 84), developed nephropathy within 6 weeks. All patients were treated with high-dose methylprednisolone pulse
Two other controls developed nephropathy at 24 and 72 weeks. The therapy for 3 days, followed by oral prednisone for 3 months, and
results of this study suggested a role for prednisone in preventing followed for a mean of 67 months. Based on clinical symptoms and
IgAV nephritis; however, a retrospective study based on 50 chil- histopathological changes, the investigators suggested that methyl-
dren with IgAV without nephritis at the time of diagnosis, showed prednisolone pulse therapy should be limited to IgAV patients with
a similar frequency of nephritis in those patients treated or not nephritis who are at risk of progressive renal disease, especially
treated with glucocorticoids (Saulsbury 1993). Kaku et al. (1998) those with nephrotic syndrome or crescentic glomerulonephritis.
reported results of a multivariate analysis and found that gluco- They considered this therapy useful when it was started early in the
corticoid treatment reduced the risk of nephritis and advocated its disease, before crescents became fibrosed.
use in patients with risk factors for renal involvement. Huber et al. The prognosis is poor if patients present with rapidly progres-
(2004) performed a randomized, placebo-controlled study of 40 sive glomerulonephritis. Tarshish et al. (2004) found no differences
children with IgAV to assess whether glucocorticoid administra- in outcome between 56 patients with histopathologically severe
tion within 7 days of disease onset could reduce the rate of renal IgAV nephritis randomized to receive supportive therapy with or
or GI complications. Twenty-one children were treated with oral without cyclophosphamide, 90 mg/m2/day for 42 days. Neither did
prednisone, 2 mg/kg/day for 1 week, with weaning over a second Pillebout et al. (2010) in a prospective randomized study compar-
week, while the remaining 19 received placebo. In this study, early ing 54 adults to receive steroids (n=29) or steroids plus i.v. pulse
prednisone therapy did not reduce the risk of renal involvement cyclophosphamide (n=25; 0.6 mg/m2 week 0, 2, 4, 8, 12, 16). In an
at 1 year, or the risk of acute GI complications (Huber et al. 2004). uncontrolled study of 12 children with IgAV and biopsy-proven,
Ronkainen et al. (2006) performed a prospective, double-blinded, rapidly progressive crescentic glomerulonephritis, Oner et  al.
placebo-controlled trial of 171 children to evaluate the effect of (1995) suggested that a combination of methylprednisolone for
early prednisone on IgAV. The treatment group (n = 84) received 3 days (30 mg/kg/day) followed by oral prednisone at 45 mg/m2/
oral prednisone 1 mg/kg/day for 2 weeks, then 0.5 mg/kg/day day tapered over 2 months, oral cyclophosphamide at 2 mg/kg/day
for 1week, and then 0.5 mg/kg/day every other day for 1 week. for 2 months, and dipyridamole at 5 mg/kg/day for 6 months, may
Thirty-six patients in the prednisone group and 35 in the placebo be effective in normalizing glomerular filtration. Only one patient
group had renal symptoms, but these resolved more rapidly in the from this small series developed persistent nephropathy with
prednisone group. The authors concluded that prednisone was nephrotic syndrome, gross haematuria, and renal failure. Another
effective in altering, but not preventing, the course of renal dis- study with a longer follow-up (mean 7.5 years) of 14 children with
ease. In this study prednisone was also effective in reducing the severe nephritis and crescent formation corresponding to grades
intensity of abdominal and joint pain. More recently, Chartapisak IV and V of the ISKDC iterated the benefit from intensive combi-
et al. (2009) performed a meta-analysis of four randomized con- nation therapy (prednisolone, cyclophosphamide, heparin/ warfa-
trolled trials (669 children) evaluating prednisone therapy at rin, and dipyridamole) (Iijima et al. 1998). Kawasaki et al. (2004b)
onset of IgAV. They did not find any difference in the risk of per- studied 37 patients with IgAV who had been diagnosed with glo-
sistent renal disease with prednisone compared with placebo. In merulonephritis of at least grade IVb. Twenty of them were treated
2010, Weiss et al. reported a retrospective series including 1985 with methylprednisolone and urokinase pulse therapy (group A),
new-onset IgAV hospitalized children. Corticosteroid treatment and the remaining 17 (group B) were treated with methylpred-
initiated during the first 2 days of hospitalization reduced the need nisolone and urokinase pulse therapy combined with cyclophos-
for abdominal surgery, endoscopy, and abdominal imaging (Weiss phamide. After 6 months of treatment, the mean urinary protein
et al. 2010). excretion in group B was significantly lower than those in Group A,
The management of IgAV nephritis is also highly controversial. and on the second kidney biopsy, the chronicity index of Group B
Patients with severe nephritis have been treated with glucocorti- was lower than that of Group A. These observations support the use
coids (oral or pulse therapy) alone, or in combination with immu- of methylprednisolone and urokinase pulse therapy combined with
nosuppressive agents such as cyclophosphamide, azathioprine, cyclophosphamide in IgAV patients with severe nephritis.
chlorambucil, ciclosporin, or mycophenolate; plasmapheresis; In an uncontrolled, retrospective study, Shin et  al. (2005a)
high-dose intravenous immunoglobulin therapy; danazol; and fish reported a beneficial effect of ciclosporin in IgAV children with
oil (Kawasaki et al. 2004a; Kawasaki et al. 2004b; Szer 1996; Shin nephrotic syndrome. In a prospective randomized clinical trial,
et al. 2005a; Shin et al. 2005b; Tarshish et al. 2004; White 1994). Jauhola et al. (2011) compared ciclosporin and methylprednisolone
540 SECTION 5 vasculitic diseases and syndromes and related disorders

pulses in the treatment of paediatric IgAV with nephrotic-range observed and when the graft function was seriously impaired,
proteinuria or III–VI grade of the ISKDC classification on biopsy. other systemic manifestations, including typical purpura, tended to
At 3 months after treatment onset, all ciclosporin patients achieved recur. Recurrences of the disease appear to be strongly associated
remission of nephrotic-range proteinuria in comparison with only with living related donor transplants (Baliah et al. 1974; Hasegawa
46% in the methylprednisolone pulses group. Furthermore, renal et al. 1989; Nast et al. 1987). The significance of this association is
survival rate after 6 years was 100% in the ciclosporin group com- unknown, but it may suggest a genetic predisposition for the devel-
pared with 85% in methylprednisolone pulses group. Noteworthy, opment of the disease.
there were no differences in the biopsy outcomes at 2  years.
Mycophenolate mofetil (MMF) has also been reported to be useful
in severe IgAV (Du et al. 2012; Nikibakhsh et al. 2010). Du et al.
Clinical course and outcome
(2012) reported the successful treatment of 12 steroid-resistant Most patients with IgAV have a self-limited disease (Garcia-Porrúa
children with nephrotic-range proteinuria treated with MMF. All 12 et al. 2002). On occasion, hormone therapy has been reported to be
patients achieved normal urine protein levels within 15 months of associated with remission in some patients (Lahita 1997). The aver-
starting MMF. High-dose intravenous immunoglobulin (IVIg) has age duration of the disease is 4 weeks (Allen et al. 1960); however,
proved to be effective in the treatment of several immune-mediated in both children and adults, relapses of the disease are common.
diseases, including systemic vasculitis (Dwyer 1992; Jayne et  al. They are characterized by flares of purpuric skin lesions which in
1991). Rostoker et  al. (1994, 1995)  reported that both low- and some patients are associated with renal and GI complications.
high-dose IVIg may be effective in treating either moderate or Allen et al. (1960) reported relapses in 40% of children. Blanco
severe nephritis in IgAV and IgA nephropathy. There has been con- et al. (1997b) described relapses in 43% of IgAV patients younger
cern about renal deterioration following such treatment in cases of than 20  years. In children from Spain, Calviño et  al. (2001)
systemic vasculitis and SLE (Barron et al. 1992; Pasatiempo et al. reported relapses (defined as an IgAV patient asymptomatic for at
1994; Schifferli et al. 1991). Although the occurrence of acute renal least 1 month presenting with a flare of skin lesions or other sys-
failure after high-dose IVIg seems to be rare, persistent deteriora- temic complications) in 15% of 69 children followed for at least
tion was observed in renal function following this therapy in an 12 months. In three studies of IgAV in adults, relapses occurred
adult with IgAV and focal proliferative glomerulonephritis with in 41% (Shrestha et al. 2006), 36% (Blanco et al. 1997b), and 21%
mesangial IgA and C3 deposits (Blanco et al. 1997a). (García-Porrúa and González-Gay 1999a) of patients. Although the
Outcomes of treatment with plasma exchange combined with prognosis is not as good for adults as for children, the overall out-
glucocorticoids, anticoagulants, cyclophosphamide, and azathio- come in IgAV is better than that observed in polyarteritis nodosa,
prine (in different combinations) in children with crescentic glo- granulomatosis with polyangiitis, or eosinophilic granulomato-
merulonephritis related to IgAV (and various other conditions) sis with polyangiitis (Churg–Strauss syndrome) (González-Gay
were retrospectively examined by Jardim et  al. (1992). In 30 and García-Porrúa 1999; Hunder 1996). Comparisons among
patients, they observed a progression to end-stage renal failure in different series are tenuous because most epidemiological stud-
50% of the children. The interval between disease onset and com- ies (Blanco et al. 1997b; García-Porrúa and González-Gay 1999a;
mencement of therapy was an important prognostic factor for the Garcia-Porrúa et al. 2002; Trapani et al. 2005) have been based on
outcome. A retrospective study of plasma exchange in children with unselected patients classified according to the 1990 ACR classifica-
rapidly progressive nephritis or cerebral vasculitis associated with tion criteria or those of Michel et al. (Michel et al. 1992; Mills et al.
IgAV was reported by Gianviti et  al. (1996). These investigators 1990), or on the basis of IgA deposits in the dermal vessel walls
described 17 IgAV patients treated with plasma exchange and com- (Tancrede-Bohin et al. 1997). In contrast, some have been based on
binations of glucocorticoids, cyclophosphamide, and azathioprine. selected patient populations, usually those with kidney dysfunction
Fourteen of them had severe glomerulonephritis (30–100% of glo- (Fogazzi et al. 1989; Lee et al. 1986; Roth et al. 1985). Some stud-
meruli with crescents) or were dialysis-dependent, and three had ies have included patients with several different types of cutaneous
cerebral vasculitis. Recovery was observed in all three children with vasculitis, and some patients with underlying diseases (Cream et al.
cerebral vasculitis. In those with severe nephritis, the time of onset 1970). Thus, different methods of selection of patients may explain
of therapy was a determinant of outcome, as all nine with renal a discrepancy in reported outcomes of the disease.
vasculitis who started plasma exchange within 1 month of disease The long-term morbidity and mortality of IgAV are predomi-
onset had significant improvement in renal function. In contrast, nantly attributable to renal involvement. IgAV has been reported
five of six children with IgAV nephritis treated later in the disease to account for between 5 and 15% of end-stage renal failure in
developed end-stage renal failure. No relapses of vasculitis were children (Bunchman et al. 1988; Meadow 1978). Bunchman et al.
observed after treatment with plasma exchange. Recently, Augusto (1988) observed that a failure to reach a creatinine clearance higher
et al. (2012) retrospectively analysed 11 adult patients with severe than 70 ml/min/1.73 m2 by 3  years after the onset of IgAV pre-
IgAV treated with plasma exchange and steroids. Two patients died dicted progression to end-stage renal failure. In an unselected
at 1 and 2 years of follow-up, respectively. In both, plasma exchange series of 141 children with IgAV, Koskimies et al. (1981) reported
treatment was stopped prematurely. Kidney function improved in persistence of abnormal urinary sediment for at least 1 month in
the remaining nine patients and no relapse was noted after a 6-year 28% of their patients. After a mean follow-up of 7.2 years, only one
mean follow-up. These results suggest that plasma exchange asso- child progressed to end-stage renal failure (0.7%) and two (1.4%)
ciated with immunosuppressive therapy may be useful in cases of developed chronic glomerular disease. In another unselected series
severe renal or extrarenal IgAV. of 270 children, Stewart et al. (1988) observed evidence of renal
Renal transplantation may be considered for the few patients involvement in 55 (20%). Re-examination at an average of 8.3 years
who progress to end-stage renal disease. Recurrences have been after the onset of the disease showed a good overall prognosis, with
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 541

mortality less than 1% overall and long-term morbidity of only Amoli, M.M., Calviño, M.C., Garcia-Porrúa, C., Llorca, J., Ollier, W.E., and
1.1% (Stewart et  al. 1988). Unlike adults, none of 73 unselected González-Gay, M.A. (2004). Interleukin 1beta gene polymorphism asso-
children with IgAV from Spain who were followed for at least 1 year ciation with severe renal manifestations and renal sequelae in Henoch–
Schönlein purpura. Journal of Rheumatology, 31, 295–8.
developed end-stage renal failure (Garcia-Porrúa et al. 2002). All
Amoli, M.M., Mattey, D.L., Calviño, M.C., et  al. (2001a). Polymorphism
these studies (Garcia-Porrúa et  al. 2002; Koskimies et  al. 1981;
at codon 469 of the intercellular adhesion molecule-1 locus is associ-
Stewart et al. 1988) support a better prognosis for IgAV nephritis ated with protection against severe gastrointestinal complications in
in children than the majority of published estimates. In extensive Henoch–Schönlein purpura. Journal of Rheumatology, 28, 1014–8.
surveillance for 23.4 years of 78 of 99 children with IgAV nephri- Amoli, M.M., Thomson, W., Hajeer, A.H., et  al. (2001b). HLA-DRB1*01
tis (Goldstein et al. 1992), the severity of the clinical presentation association with Henoch–Schönlein purpura in patients from northwest
and the initial findings on the renal biopsy correlated well with Spain. Journal of Rheumatology, 28, 1266–70.
the outcome. In 16 of 44 girls, pregnancies were complicated by Amoli, M.M., Thomson, W., Hajeer, A.H., et al. (2002a). HLA-B35 association with
proteinuria and hypertension, even in the absence of active renal nephritis in Henoch–Schönlein purpura. Journal of Rheumatology, 29, 948–9.
disease. Because of that, Goldstein et al. (1992) suggested that IgAV Amoli, M.M., Thomson, W., Hajeer, A.H., et  al. (2002b). Interleukin 1
receptor antagonist gene polymorphism is associated with severe renal
nephritis in children should be followed long-term, and especially
involvement and renal sequelae in Henoch–Schönlein purpura. Journal
during pregnancy. of Rheumatology, 29, 1404–7.
As discussed in Section Renal, severe renal involvement is more Amoli, M.M., Thomson, W., Hajeer, A.H., et al. (2002c). Interleukin-8 gene
common in adults than in children (Blanco et al. 1997b; Garcia-Porrúa polymorphism is associated with increased risk of nephritis in cutane-
et al. 2002; Uthman et al. 1998) but in the unselected series of Blanco ous vasculitis. Journal of Rheumatology, 29, 2367–70.
et al. (1997b), the final outcome was equally good in both age groups. Amoroso, A., Berrino, M., Canale, L., Coppo, R., Cornaglia, M., Guarrera, S.,
For patients older than 15 who were studied based on the presence et al. (1997). Immunogenetics of Henoch–Schönlein disease. European
of cutaneous IgA deposits and purpura, Tancrede-Bohin (1997) Journal of Immunogenetics, 24, 323–33.
observed a good outcome. In an unselected series of 27 IgAV adults Amoroso A., Danek G., Vatta S., Crovella S., Berrino M., et  al. (1998).
(older than 20 years) with biopsy-proven leukocytoclastic vasculitis, Polymorphisms in angiotensin-converting enzyme gene and sever-
ity of renal disease in Henoch-Schonlein patients. Nephrology Dialysis
García-Porrúa and González-Gay (1999a) observed the development
Transplantation, 13, 3184–88.
of renal insufficiency in 8.3% of patients after a median follow-up of
Archimandritis, A., Kalos, A., Pantzos, A., Sakellariou, D., Malamas, N., and
3 years. These data suggest that the prognosis in adults with IgAV is Fertakis, A. (1994). Hemorrhagic ascites in a patient with anaphylactoid
not as good as that reported in children. purpura. Journal of Clinical Gastroenterology, 18, 257–8.
Ates, E., Bakkaloglu, A., Saatci, U., Soylemezoglu, O. (1994). Von Willebrand
Conclusions factor antigen compared with other factors in vasculitic syndromes.
Archives of Disease in Childhood, 70, 40–3.
IgAV is an IgA-mediated disease affecting small blood vessels.
Athreya, B.H. (1996). Vasculitis in children. Current Opinion in Rheumatology,
It is the most common vasculitis in children and an infrequent 8, 477–84.
condition in adults. It may follow an intercurrent illness, usu- Augusto, J.F., Sayegh J., Delapierre L., Croue A., et al. (2012). Addition of
ally infection of the upper respiratory tract. Skin, joints, gut, plasma exchange to glucocorticosteroids for the treatment of severe
and kidneys are the organs most commonly involved. It is a Henoch-Schönlein purpura in adults: A case series. American Journal
benign and self-limited condition in the vast majority of chil- Of Kidney Diseases, 59, 663–9.
dren and most adults, but a small percentage, especially adults, Ault, B.H., Stapleton, F.B., Rivas, M.L., Waldo, F.B., Roy, S., McLean, R.H.,
progress to renal insufficiency. For this reason, close follow-up et al. (1990). Association of Henoch–Schönlein purpura glomerulone-
with repeated urinalysis is advisable in those who present renal phritis with C4B deficiency. Journal of Pediatrics, 117, 753–5.
manifestations during the first 6  months after the onset of the Bailey, M., Chapin, W., Licht, H., and Reynolds, J.C. (1998). The effects of
vasculitis on the gastrointestinal tract and liver. Gastroenterology Clinics
disease. Pharmacological treatments are controversial and so far of North America, 27, 747–82.
no management of IgAV has been declared optimal in patients Baliah, T., Kim, K., Anthone, S., Anthone, R., Montes, M., and Andres, G.
with renal disease or severe GI complications. Reports on pro- (1974). Recurrence of Henoch–Schönlein purpura glomerulonephritis
phylactic glucocorticoid therapy to prevent renal disease are con- in transplanted kidneys. Transplantation, 18, 343–6.
tradictory. Long-term follow-up studies are required for a better Ballard, H.S., Eisinger, R.P., and Gallo, G. (1970). Renal manifestations of the
understanding of this vasculitis. Henoch–Schönlein syndrome in adults. American Journal of Medicine, 79,
328–35.
References Barron, K.S., Sher, M.R., and Silverman, E.D. (1992). Intravenous immuno-
Aalberse, J., Dolman, K., Ramnath, G., Rodrigues Pereira, R., and Davin, J.- globulin therapy:  Magic or black magic. Journal of Rheumatology, 19
C. (2007). Henoch-Schönlein purpura in children: an epidemiological (Suppl. 33), 94–7.
study among Dutch paediatricians on incidence and diagnostic criteria. Bayram, C., Demircin, G., Erdogan, O., and Bulbul, M. (2011). Prevalence of
Annals of the Rheumatic Diseases, 66, 1648–50. MEFV gene mutations and their clinical correlations in Turkish children
Abdel-Al, Y.K., Hejazi, Z., and Majjed, H.A. (1990). Henoch Schonlein pur- with Henoch-Schonlein purpura. Acta Paediatrica, 100, 745–9.
pura in Arab children. Analysis of 52 cases. Tropical and Geographical Blanco, R., González-Gay, M.A., Ibañez, D., Sanchez-Andrade, A., and
Medicine, 42, 52–7. González-Vela, C. (1997a). Paradoxical and persistent renal impair-
Abou-Ragheb, H.H.A., Williams, A.J., Brown, C.B., and Milford-Ward, A. ment in Henoch–Schönlein purpura after high-dose immunoglobulin
(1992). Plasma levels of the anaphylatoxins C3a and C4a in patients with therapy. Nephron, 76, 247–8.
IgA nephropathy/Henoch–Schönlein nephritis. Nephron, 62, 22–6. Blanco, R., Martinez-Taboada, V.M., Rodriguez-Valverde, V., García-Fuentes,
Allen, D., Diamond, L.K., and Howell, D.A. (1960). Anaphylactoid pur- M., and González-Gay, M.A. (1997b). Henoch–Schönlein purpura in
pura in children (Schönlein-Henoch syndrome). American Journal of the adulthood and in the childhood:  two different expressions of the
Diseases of Children, 99, 833–54. same syndrome. Arthritis and Rheumatism, 40, 859–64.
542 SECTION 5 vasculitic diseases and syndromes and related disorders

Blockmans, D. and Bobbaers, H. (1998). Inflammation phenomena in vas- (IgA-ANCA): new insight into a debated issue. Italian Group of Renal
culitis:  from immune complexes to less immune forms. Acta Clinica Immunopathology Collaborative Study on Henoch–Schönlein purpura
Belgica, 53, 83–91. in adults and children. Nephrology Dialysis Transplantation, 12, 2269–76.
Boyer, J., Baudet, P., Ben Bouali, A., Ronceray, J., and Joubaud, F. (1978). Counahan, R., Winterborn, M.H., White, R.H.R., Heaton, J.M., Meadow,
Unusual manifestations during rheumatoid purpura. Apropos of 2 cases. S.R., Bluett, N.H., et al. (1977). Prognosis of Henoch–Schönlein purpura
La Semaine des Hôpitaux de Paris, 54, 298–302. in children. British Medical Journal, 2, 11–14.
Brown, C.B., Wilson, D., Turner, D., Cameron, J.S., Ogg, C.S., Chantler, C., Cream, J.J., Gample, J.M., and Peachey, R.D.G. (1970). Schönlein-Henoch
et  al. (1974). Combined immunosuppression and anticoagulation in purpura in the adult. Quarterly Journal of Medicine, 39, 461–82.
rapidly progressive glomerulonephritis. Lancet, 2, 1166–72. Cull, D.L., Rosario, V., Lally, K.P., Ratner, I., and Mahour, G.H. (1990).
Bunchman, T.E., Mauer, S.M., Sibley, R.K., and Vernier, R.L. (1988). Surgical implications of Henoch–Schönlein purpura. Journal of Pediatric
Anaphylactoid purpura: characteristics of 16 patients who progressed to Surgery, 25, 741–3.
renal failure. Pediatric Nephrology, 2, 393–7. Cummins, D.L., Mimouni, D., Rencic, A., Kouba, D.J., and Nousari, C.H.
Cakir, N., Pamuk, O.N., and Donmez, S. (2004). Henoch–Schönlein purpura (2003). Henoch–Schönlein purpura in pregnancy. British Journal of
in two brothers imprisoned in the same jail: presentation two months Dermatology, 149, 1282–5.
apart. Clinical and Experimental Rheumatology, 22, 235–7. Daniel, L., Sonia, M., Cathy, H., Tonya, M., and Adam, G. (2006).Use of wire-
Calabrese, L.H., Michel, B.A., Bloch, D.A., Arend, W.P., Edworthy, S.M., less capsule endoscopy in the management of severe Henoch-Schonlein
Fauci, A.S., et al. (1990). The ACR 1990 criteria for the classification of purpura. Pediatrics, 118, e904–6.
hypersensitivity vasculitis. Arthritis and Rheumatism, 33, 1108–13. De Angelis, G.L., Fornaroli, F., De Angelis, N., Magiteri, B., and Bizzarri, B.
Calviño, M.C., Llorca, J., Garcia-Porrúa, C., Fernandez-Iglesias, J.L., (2007). Wireless capsule endoscopy for pediatric small-bowel diseases.
Rodriguez-Ledo, P., and González-Gay, M.A. (2001). Henoch–Schönlein American Journal of Gastroenterology, 102, 1749–57.
purpura in children from northwestern Spain: a 20-year epidemiologic Debray, J., Krulik, M., and Giorgi, H. (1971). Le purpura rhumatoide de
and clinical study. Medicine (Baltimore), 80, 279–90. l’adulte (syndrome de Schönlein-Henoch). A propos de 22 observations.
Cappell, M.S. and Gupta, A.M. (1990). Colonic lesions associated with Semaine des Hopitaux de Paris, 47, 1805–19.
Henoch–Schönlein purpura. American Journal of Gastroenterology, 85, De Mattia, D., Penza, R., Giordano, P., Del Vecchio, G.C., Aceto, G., Altomare,
1186–8. M., et al. (1995). Von Willebrand factor and factor XIII in children with
Casanueva, B., Rodriguez-Valverde, V., Merino, J., Arias, M., and Henoch–Schönlein purpura. Pediatric Nephrology, 9, 603–5.
García-Fuentes, M. (1983). Increased IgA-producing cells in the blood Dillon, M.J. and Ansell, B.M. (1995). Vasculitis in children and adolescents.
of patients with active Henoch–Schönlein purpura. Arthritis and Rheumatic Diseases Clinics of North America, 21, 1115–36.
Rheumatism, 26, 854–60. Dolezalova, P., Telekesova, P., Nemcova, D., and Hoza, J. (2004). Incidence of
Cassidy, J.T. and Petty, R.E. (1995). Henoch–Schönlein purpura. In Textbook vasculitis in children in the Czech Republic: 2-year prospective epidemi-
of pediatric rheumatology (ed. J.T. Cassidy and R.E. Petty), 3rd edn, pp. ology survey. Journal of Rheumatology, 31, 2295–9.
384–8. W.B. Saunders Company, Philadelphia. Du, Y., Hou, L., Zhao, C., and Han, M. (2012). Treatment of children with
Chamberlain, R.S. and Greenberg, L.W. (1992). Scrotal involvement in Henoch-Schönlein purpura nephritis with mycophenolate mofetil.
Henoch–Schönlein purpura. A  case report and review of literature. Pediatric Nephrology, 27, 765–71.
Pediatric Emergency Care, 8, 213–5. Dudley, J., Afifi, E., Gardner, A., Tizard, E., and McGraw, M. (2000).
Chang, W.L., Yang, Y.H., Lin, Y.T., and Chiang, B.L. (2004). Gastrointestinal Polymorphism of the ACE gene in Henoch-Schönlein purpura nephri-
manifestations in Henoch-Schönlein purpura: a review of 261 patients. tis. Pediatric Nephrology, 14, 218–20.
Acta Paediatrica, 93, 1427–31. Duquesnoy, B. (1991). Henoch–Schönlein purpura. Bailliere’s Clinical
Chang, W.L., Yang, Y.H., Wang, L.C., Lin, Y.T., and Chiang, B.L. (2005). Renal Rheumatology, 5, 253–61.
manifestations in Henoch–Schönlein purpura: a 10-year clinical study. Dwyer, J.M. (1992). Manipulating the immune system with immune globu-
Pediatric Nephrology, 20, 1269–72. lin. New England Journal of Medicine, 326, 107–16.
Chartapisak, W., Opastiraku, S., Willis, N.S., Craig, J.C., and Hodson, E.M. Farley, T.A., Gillespi, S., Rasoulpour, M., Tolentino, N., Hadler, J.L., and
(2009). Prevention and treatment of renal disease in Henoch-Schönlein Hurwitz, E. (1989). Epidemiology of a cluster of Henoch–Schönlein
purpura: a systematic review. Archives of Disease in Childhood, 94, 132–7. purpura. American Journal of Diseases of Children, 143, 798–803.
Chaussain, M., De Boissieu, D., Kalifa, G., Epelbaum, S., Niaudet, P., Badoual, Fauci, A.S., Haynes, B.F., and Katz, P. (1978). The spectrum of vasculitis: clin-
J., et  al. (1992). Impairment of lung diffusion capacity in Henoch– ical, pathologic, immunologic, and therapeutic considerations. Annals of
Schönlein purpura. Journal of Pediatrics, 121, 12–6. Internal Medicine, 89, 660–76.
Chen, Y., Zhou, J.H., Wu, H.S., and Wang, H.W. (2006). Role of mast cells and Ferguson, P.J., Saulsbury, F.T., Dowell, S.F., Török, T.J., Erdman, D.A., and
eosinophil cationic protein in the pathogenesis of Henoch-Schonlein Anderson, L.A. (1996). Prevalence of human parvovirus B19 infection
purpura nephritis. Zonghua Er Ke Za Zhi, 44, 407–10. in children with Henoch–Schönlein purpura. Arthritis and Rheumatism,
Chiaretti, A., Scommegna, S., Castorina, M., Piastra, M., Tortorolo, L., Villali, 39, 880–2.
A., et  al. (1995). Intracranial hemorrhage in Henoch–Schönlein syn- Fielding, R.E., Hawkins, C.P., Hand, M.F., Heath, P.D., and Davies, S.J. (1998).
drome. Pediatria Medica e Chirurgica, 17, 177–9. Seizure complicating Henoch–Schönlein purpura. Nephrology Dialysis
Cimolai, N. and MacNab, A. (1991). Henoch–Schönlein purpura and Transplantation, 13, 761–2.
Streptococcus equisimilis. British Journal of Dermatology, 125, 403. Fogazzi, G.B., Pasquali, S., Moriggi, M., Casanova, S., Damilano, I., Mihatsch,
Clauvel, J.P., Touraine, R., and Bernard, J. (1972). Rheumatoid purpura in adults. M.J., et al. (1989). Long-term outcome of Schönlein–Henoch nephritis
Apropos of 22 cases. Nouvelle Revue Française d’Hématologie, 12, 117–22. in the adult. Clinical Nephrology, 31, 60–6.
Connolly, B. and O’Halpin, D. (1994). Sonographic evaluation of the abdo- Fortin, P.R. (1996). Vasculitides associated with malignancy. Current Opinion
men in Henoch–Schönlein purpura. Clinical Radiology, 49, 320–3. in Rheumatology, 8, 30–3.
Coppo, R., Andrulli, S., Amore, A., Gianoglio, B., Conti, G., et al. (2006). Frank, E. (1915). Die essentielle thrombopenie. Berliner Klinische
Predictors of outcome in Henoch-Schönlein nephritis in children and Wochenschrift, 52, 454.
adults. American Journal of Kidney Diseases, 47, 993–1003. Fujieda, M., Oishi, N., Naruse, K., Hashizume, M., Nishiya, K., Kurashige,
Coppo, R., Cirina, P., Amore, A., Sinico, R.A., Radice, A., and Rollino, C. T., et al. (1998). Soluble thrombomodulin and antibodies to bovine glo-
(1997). Properties of circulating IgA molecules in Henoch–Schönlein pur- merular endothelial cells in patients with Henoch–Schönlein purpura.
pura nephritis with focus on neutrophil cytoplasmic antigen IgA binding Archives of Disease in Childhood, 78, 240–4.
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 543

Galla, J.H. (1995). IgA nephropathy. Kidney International, 47, 377–87. González-Gay, M.A., García-Porrúa, C., Afonso, E., Basanta, D., and
García Fuentes, M., Chantler, C., and Williams, D.G. (1977). Moreno-Lugris, C. (1999). Scurvy: a condition mimicking cutaneous vascu-
Cryoglobulinaemia in Henoch–Schönlein purpura. British Medical litis. Report of three cases. Revue du Rhumatisme (English Edition), 66, 360–1.
Journal, 2, 163–5. González-Gay, M.A., Garcia-Porrúa, C., and Pujol, R.M. (2005). A clinical
Garcia-Porrúa, C., Calvino, M.C., Llorca, J., Couselo, J.M., and González-Gay, approach to cutaneous vasculitis. Current Opinion in Rheumatology,
M.A. (2002). Henoch–Schönlein purpura in children and adults: clinical 17, 56–61.
differences in a defined population. Seminars Arthritis Rheumatism, 32, González-Gay, M.A., Garcia-Porrúa, C., Salvarani, C., and Hunder, G.G
149–56. (2000). Cutaneous vasculitis and cancer:  a clinical approach. Clinical
Garcia-Porrúa, C. and González-Gay, M.A. (1998). Cutaneous vasculitis as and Experimental Rheumatology, 18, 305–7.
a paraneoplastic syndrome in adults. Arthritis and Rheumatism, 41, González-Gay, M.A. and Llorca, J. (2005). Controversies on the use of
1133–5. corticosteroid therapy in children with Henoch–Schönlein purpura.
Garcia-Porrúa, C. and González-Gay, M.A. (1999a). Comparative clinical Seminars in Arthritis and Rheumatism, 35, 135–7.
and epidemiologic study of hypersensitivity vasculitis versus Henoch– Gross, W.L., Schmitt, W.H., and Csernok, E. (1993). ANCA and associated
Schönlein purpura in adults. Seminars in Arthritis and Rheumatism, 28, diseases:  immunodiagnostic and pathogenetic aspects. Clinical and
404–12. Experimental Immunology, 91, 1–12.
Garcia-Porrúa, C. and González-Gay, M.A. (1999b). Bacterial infection Guillevin, L., Lhote, F., Amouroux, J., Gherardi, R., Callard, P., and Casassu,
presenting as cutaneous vasculitis in adults. Clinical and Experimental P. (1996). Antineutrophil cytoplasmic antibodies, abnormal angiograms
Rheumatology, 17, 471–3. and pathological findings in polyarteritis nodosa and Churg-Strauss
Garcia-Porrúa, C., González-Gay, M.A., and Lopez-Lazaro, L. (1999). syndrome:  indications for the classification of the vasculitides of the
Drug-associated cutaneous vasculitis in adulthood. Clinical and epide- polyarteritis nodosa group. British Journal of Rheumatology, 35, 958–64.
miological associations in a defined population of Northwestern Spain. Ha, T.S. (2005). The role of tumor necrosis factor-alpha in Henoch–Schönlein
Journal of Rheumatology, 26, 1942–4. purpura. Pediatric Nephrology, 20, 149–53.
Garcia-Porrúa, C., González-Louzao, C., Llorca, J., and González-Gay, M.A. Ha, T.S. and Cha, S.H. (1996). Cerebral vasculitis in Henoch–Schönlein
(2001). Predictive factors for renal sequelae in adults with Henoch– purpura:  a case report with sequential magnetic resonance imaging.
Schönlein purpura. Journal of Rheumatology, 28, 1019–24. Pediatric Nephrology, 10, 634–6.
Gardner-Medwin, J.M., Dolezalova, P., Cummins, C., and Southwood, T.R. Halling, S.F., Soderberg, M.P., and Berg, U.B. (2005). Henoch–Schönlein
(2002). Incidence of Henoch-Schönlein purpura, Kawasaki disease, nephritis:  clinical findings related to renal function and morphology.
and rare vasculitides in children of different ethnic origins. Lancet, 360, Pediatric Nephrology, 20, 46–51.
1197–202. Hasegawa, A., Kawamura, T., Ito, H., Hasegawa, O., Ogawa, O., Honda, M.,
Garzoni, L., Vanoni, F., Rizzi, M., Simonetti, G.D., et al. (2009). Nervous sys- et al. (1989). Fate of renal grafts with recurrent Henoch–Schönlein pur-
tem dysfunction in Henoch-Schönlein syndrome: systematic review of pura nephritis in children. Transplantation Proceedings, 21, 2130–3.
the literature. Rheumatology, 48, 1524–9. Heaton, J.M., Turner, D.R., and Cameron, J.S. (1977). Localization of glomer-
Gershoni-Baruch, R., Broza, Y., and Brik, R. (2003). Prevalence and sig- ular deposits in Henoch–Schönlein nephritis. Histopathology, 1, 93–104.
nificance of mutations in the Familial Mediterranean Fever gene in Heberden, W. (1801). Commentarii de Marlbaun. Historia et curatione.
Henoch-Schonlein purpura. Journal of Pediatrics, 143, 658-61. London, Payne.
Giancomo, J. and Tsai, C.C. (1977). Dermal and glomerular deposition of Helander, S.D., de Castro, F.R., and Gibson, L.E. (1995). Henoch–Schönlein
IgA in anaphylactoid purpura. American Journal of Diseases of Children, purpura:  Clinicopathologic correlation of cutaneous vascular IgA
131, 981–3. deposits and the relationship to leukocytoclastic vasculitis. Acta
Gianviti, A., Trompeter, R.S., Barratt, T.M., Lythgoe, M.F., and Dillon, D.J. Dermato-Venereologica, 75, 125–9.
(1996). Retrospective study of plasma exchange in patients with idi- Hené, R.J., Velthuis, P., van de Wiel, A., Klepper, D., Mees, E.J.D., and Kater,
opathic rapidly progressive glomerulonephritis and vasculitis. Archives L. (1986). The relevance of IgA deposits in vessel walls of clinically nor-
of Disease in Childhood, 75, 186–90. mal skin. Archives of Internal Medicine, 146, 745–9.
Gibson, L.E. and Su, W.F. (1995). Cutaneous vasculitis. Rheumatic Diseases Henoch, E.H.H. (1874). Über eine eigentümliche Form von Purpura. Berliner
Clinics of North America, 21, 1097–113. Klinische Wochenschrift, 2, 641–3.
Gilliam, J.N. and Smiley, J.D. (1976). Cutaneous necrotizing vasculitis and Henoch, E.H.H. (1895). Vorlesungen über Kinderkrankheiten. Berlin,
related disorders. Annals of Allergy, 37, 328–39. Hirschwald.
Glasier, C.M., Siegel, M.J., McAlister, W.H., and Shackelford, G.D. (1981). Hisano, S., Matsushita, M., Fujita, T., and Iwasaki, H. (2005). Activation of
Henoch–Schönlein syndrome in children:  gastrointestinal manifesta- the lectin complement pathway in Henoch–Schönlein purpura nephri-
tions. American Journal of Roentgenology, 136, 1081–5. tis. American Journal of Kidney Diseases, 45, 295–302.
Goldman, L.P. and Lindenberg, R.L. (1981). Henoch–Schönlein pur- Huber, A.M., King, J., McLaine, P., Klassen, T., and Pothos, M. (2004).
pura:  gastrointestinal manifestations with endoscopic correlation. A randomized, placebo-controlled trial of prednisone in early Henoch–
American Journal of Gastroenterology, 75, 357–60. Schönlein Purpura. BioMed Central Medicine, 2, 7.
Goldstein, A.R., White, R.H., Akuse, R., and Chantler, C. (1992). Long-term Hunder, G. (1996). Vasculitis: Diagnosis and therapy. American Journal of
follow-up of childhood Henoch–Schönlein nephritis. Lancet, Medicine, 100 (Suppl. 2A), 37S–45S.
339, 280–2. Hunder, G.G., Arend, W.P., Bloch, D.A., Calabrese, L.H., Fauci, A.S., Fries,
González-Gay, M.A., Calviño, M.C., Vazquez-Lopez, M.E., Garcia-Porrúa, J.F., et al. (1990). The ACR 1990 criteria for the classification of vasculi-
C., Fernandez-Iglesias, J.L., Dierssen, T., et al. (2004). Implications of tis. Arthritis and Rheumatism, 33, 1065–7.
upper respiratory tract infections and drugs in the clinical spectrum Iijima, K., Ito-Kariya, S., Nakamura, H., and Yoshikawa, N. (1998). Multiple
of Henoch–Schönlein purpura in children. Clinical and Experimental combined therapy for severe Henoch–Schönlein nephritis in children.
Rheumatology, 22, 781–4. Pediatric Nephrology, 12, 244–8.
González-Gay, M.A. and García-Porrúa, C. (1999). Systemic vasculitis in Ilan, Y. and Naparstek, Y. (1991). Schönlein-Henoch syndrome in adults and
adults in Northwestern Spain, 1988–1997: Clinical and epidemiologic children. Seminars in Arthritis and Rheumatism, 21, 103–9.
aspects. Medicine (Baltimore), 78, 292–308. Ince, E., Mumcu, Y., Suskan, E., Yalcincaya, F., Tumer, N., and Cin, S. (1995).
González-Gay, M.A. and Garcia-Porrúa, C. (2001). Epidemiology of the vas- Infantile acute hemorrhagic edema: a variant of leukocytoclastic vascu-
culitides. Rheumatic Diseases Clinics of North America, 27, 729–49. litis. Pediatric Dermatology, 12, 224–7.
544 SECTION 5 vasculitic diseases and syndromes and related disorders

Jardim, H.M., Leake, J., Risdon, R.A., Barrat, T.M., and Dillon, M.J. (1992). Kawasaki, Y., Suzuki, J., Sakai, N., Nemoto, K., Nozawa, R., Suzuki, S., et al.
Crescentic glomerulonephritis in children. Pediatric Nephrology, (2003). Clinical and pathological features of children with Henoch–
6, 231–5. Schönlein purpura nephritis: risk factors associated with poor progno-
Jauhola, O., Ronkainen, J., Autio-Harmainen, H., Koskimies, O., sis. Clinical Nephrology, 60, 153–60.
Ala-Houhala, M., et  al. (2011). Cyclosporine A  vs. methylpredniso- Kawasaki, Y., Suzuki, J., and Suzuki, H (2004b). Efficacy of methylpredni-
lone for Henoch-Schönlein nephritis:  a randomized trial. Pediatric solone and urokinase pulse therapy combined with or without cyclo-
Nephrology, 26, 2159–66. phosphamide in severe Henoch–Schönlein nephritis:  a clinical and
Jauhola, O., Ronkainen, J., Koskimies, O., Ala-Houhala, M., et al. (2010a). histopathological study. Nephrology Dialysis Transplantation, 19,
Renal manifestations of Henoch-Schönlein purpura in a 6-month pro- 858–64.
spective study of 223 children. Archives of Disease in Childhood, 95, Knight, J.F. (1990). The rheumatic poison: a survey of some published inves-
877–82. tigations of the immunopathogenesis of Henoch–Schönlein purpura.
Jauhola, O., Ronkainen, J., Koskimies, O., Ala-Houhala, M., et al. (2010b). Pediatric Nephrology, 4, 533–41.
Clinical course of extrarenal symptoms in Henoch-Schönlein purpura: a Koskimies, O., Mir, S., Rapola, J., and Vilska, J. (1981). Henoch–Schönlein
6-month prospective study. Archives of Disease in Childhood, 95, 871–6. nephritis:  long-term prognosis of unselected patients. Archives of
Jayne, D.R.W., Davies, M.J., Fox, C.J.V., Black, C.M., and Lockwood, C.M. Disease in Childhood, 56, 482–4.
(1991). Treatment of systemic vasculitis with pooled intravenous immu- Kraft, D.A., McKee, D., and Scott, C. (1998). Henoch–Schönlein pur-
noglobulin. Lancet, 337, 1137–9. pura: A review. American Family Physician, 58, 405–8.
Jennette, J.C., Falk, R.J., Andrassy, K., Bacon, P.A., Churg, J., Gross, W.L., et al. Kurzrock, R., Cohen, P., and Markowitz, A. (1994). Clinical manifestations
(1994). Nomenclature of systemic vasculitides: proposal of an interna- of vasculitis in patients with solid tumors. Archives of Internal Medicine,
tional consensus conference. Arthritis and Rheumatism, 37, 187–92. 154, 334–40.
Jennette, J.C., Falk, R.J., Bacon, P.A., Basu, N., Cid, M.C., Ferrario, F., Lahita, R.G. (1997). Influence of age on Henoch–Schönlein purpura. Lancet,
Flores-Suarez, L.F., Gross, W.L., Guillevin, L., Hagen, E.C., Hoffman, 350, 1116–7.
G.S., Jayne, D.R., Kallenberg, C.G., Lamprecht, P., Langford, C.A., Lanzkowsky, S., Lanzkowsky, L., and Lanzkowsky, P. (1992). Henoch–
Luqmani, R.A., Mahr, A.D., Matteson, E.L., Merkel, P.A., Ozen, S., Schönlein purpura. Pediatrics in Review, 13, 130–7.
Pusey, C.D., Rasmussen, N., Rees, A.J., Scott, D.G., Specks, U., Stone, Lee, H.S., Koh, H.I., Kim, M.J., and Rha, H.Y. (1986). Henoch–Schönlein
J.H., Takahashi, K., and Watts, R.A. (2013). 2012 revised International nephritis in adults:  a clinical and morphological study. Clinical
Chapel Hill Consensus Conference nomenclature of vasculitides. Nephrology, 26, 125–30.
Arthritis and Rheumatism, 65, 1–11. Levinsky, R.J. and Barratt, T.M. (1979). IgA immune complexes in Henoch–
Jeong, Y.K., Hu, H.K., Yoon, C.H., et  al. (1997). Gastrointestinal involve- Schönlein purpura. Lancet, 2, 1100.
ment in Henoch–Schönlein syndrome: CT findings. American Journal Lin, S.J. and Huang, J.L. (1998). Henoch–Schönlein purpura in Chinese
of Roentgenology, 168, 965–8. children and adults. Asian Pacific Journal of Allergy and Immunology,
Jin, D.K., Kohsaka, T., Jun, A., and Kobayashi, N. (1992). Complement 4 16, 21–5.
gene deletion in patients with IgA nephropathy and Henoch–Schönlein Liu, D., Lu, F., Zhai, S., Wei, L., Ma, S., et al. (2010). Renin-angiotensin sys-
nephritis. Child Nephrology and Urology, 12, 208–11. tem gene polymorphisms in children with Henoch-Schönlein purpura
Jin, D.K., Kohsaka, T., Koo, J.W., Ha, I.S., Cheong, H.I., and Choi, Y. (1996). in West China. Journal of the Renin-Angiotensin-Aldosterone System, 11,
Complement 4 locus II gene deletion and DQA1*0301 gene: genetic risk 248–55.
factors for IgA nephropathy and Henoch–Schönlein nephritis. Nephron, Lofters, W.S., Pineo, G.F., Luke, K.H., and Yaworsky, R.G. (1973). Henoch–
73, 390–5. Schönlein purpura occurring in three members of a family. Canadian
Jones, N.F., Creamer, B., and Gimlette, T.M.D. (1966). Hypoproteinemia in Medical Association Journal, 109, 46–8.
anaphylactoid purpura. British Medical Journal, 2, 1166–8. MacPherson, R.I. (1974). The radiologic manifestations of Henoch–
Kaku, Y., Nohara, K., and Honda, S. (1998). Renal involvement in Henoch– Schönlein purpura. Journal of the Canadian Association of Radiologists,
Schönlein purpura: a multivariate analysis of prognostic factors. Kidney 25, 275–81.
International, 53, 1755–9. Martin, J., Paco, L., Ruiz, M.P., et al. (2005). Inducible nitric oxide synthase
Kathuria, S. and Cheifec, G. (1982). Fatal pulmonary Henoch–Schönlein polymorphism is associated with susceptibility to Henoch–Schönlein
syndrome. Chest, 82, 654–6. purpura in Northwestern Spain. Journal of Rheumatology, 32, 1081–5.
Kato, S., Shibuya, H., Naganuma, H., and Nakagawa, H. (1992). Masuda, M., Nakanishi, K., Yoshizawa, N., Iijima, K., and Yoshikawa, N.
Gastrointestinal endoscopy in Henoch–Schönlein purpura. European (2003). Group A streptococcal antigen in the glomeruli of children with
Journal of Pediatrics, 151, 482–4. Henoch–Schönlein nephritis. American Journal of Kidney Diseases, 41,
Kawakami, T., Watabe, H., Mizoguchi, M., and Soma, Y. (2006). Elevated 366–70.
serum IgA anticardilopin antibody levels in adult Henoch-Schönlein Meadow, S.R. (1978). The prognosis of Henoch–Schönlein nephritis. Clinical
purpura. British Journal of Dermatology, 155, 983–7. Nephrology, 9, 87–90.
Kawakami, T., Watabe, H., Mizoguchi, M., and Soma, Y. (2008). High titer of Meadow, S.R., Glasgow, E.F., Whire, R.H.R., Moncrieff, M.W., Cameron, J.S.,
serum antiphospholipid antibody levels in adult Henoch-Schönlein pur- and Ogg, C.S. (1972). Schönlein-Henoch nephritis. Quarterly Journal of
pura and cutaneous leukocytoclastic angiitis. Arthritis and Rheumatism, Medicine, 41, 241–58.
59, 561–7. Mertz, L.E. and Conn, D.L. (1992). Vasculitis associated with malignancy.
Kawana, S. and Nishiyama, S. (1992). Serum SC5b-9 (terminal complement Current Opinion in Rheumatology, 4, 39–46.
complex) level, a sensitive indicator of disease activity in patients with Michel, B.A., Hunder, G.G., Bloch, D.A., and Calabrese, L.H. (1992).
Henoch–Schönlein purpura. Dermatology, 184, 171–6. Hypersensitivity vasculitis and Henoch–Schönlein purpura: A compari-
Kawasaki, Y., Imaizumi, T., Matsuura, H., Ohara, S., Takano, K., Suyama, son between the 2 disorders. Journal of Rheumatology, 19, 721–8.
K., et  al. (2008). Renal expression of alpha-smooth muscle actin and Mills, J.A., Michel, B.A., Bloch, D.A., Calabrese, L.H., Hunder, G.G., Arend,
c-Met in children with Henoch-Schönlein purpura nephritis. Pediatric W.P., et  al. (1990). The ACR 1990 criteria for the classification of
Nephrology, 23, 913–9. Henoch–Schönlein purpura. Arthritis and Rheumatism, 33, 1114–21.
Kawasaki, Y., Suzuki, J., Murai, M., Takahashi, A., Isome, M., Nozawa, R., Misra, A.K., Biswas, A., Das, S.K., Gharai, P.K., and Roy, T. (2004). Henoch–
et al. (2004a). Plasmapheresis therapy for rapidly progressive Henoch– Schönlein purpura with intracerebral haemorrhage. Journal of
Schönlein nephritis. Pediatric Nephrology, 19, 920–3. Association of Physicians India, 52, 833–4.
CHAPTER 39 iga vasculitis (henoch-schönlein purpura) 545

Mollica, F., Li Volti, S., Garozzo, R., and Russo, G. (1992). Effectiveness Paller, A.S., Kelly, K., and Sethi, R. (1997). Pulmonary hemorrhage:  an
of early prednisone treatment in preventing the development of often fatal complication of Henoch–Schönlein purpura. Pediatric
nephropathy in anaphylactoid purpura. European Journal of Pediatrics, Dermatology, 14, 299–302.
15, 140–4. Pasatiempo, A.M.G., Kroser, J.A., Rudnick, M., and Hoffman, B.I. (1994).
Mormile, R., D’Alterio, V., Treccagnoli, G., and Sorrentino, P. (2004). Acute renal failure after intravenous immunoglobulin therapy. Journal
Henoch–Schönlein purpura with antiphospholipid antibodies after of Rheumatology, 21, 347–9.
influenza vaccination: how fearful is it in children? Vaccine, 23, 567–8. Peru, H., Soylemezoglu, O., Gonen, S., Cetinyurek, A., et al. (2008). HLA class
Nadrous, H.F., Yu, A.C., Specks, U., and Ryu, J.H. (2004). Pulmonary involve- 1 associations in Henoch Schonlein purpura: increased and decreased
ment in Henoch–Schönlein purpura. Mayo Clinic Proceeding, 79, frequencies. Clinical Rheumatology, 27, 5–10.
1151–7. Piette, W.W. (1994). The differential diagnosis of purpura from a morpho-
Narchi, H. (2005). Risk of long term renal impairment and duration of fol- logic perspective. Advances in Dermatology, 9, 3–23.
low up recommended for Henoch–Schönlein purpura with normal or Pillebout, E., Alberti, C., Guillevin, L., Ouslimani, A., and Thervet, E.
minimal urinary findings:  a systematic review. Archives of Disease in (2010). Addition of cyclophosphamide to steroids provides no ben-
Childhood, 90, 916–20. efit compared with steroids alone in treating adult patients with severe
Nast, C.C., Ward, H.J., Kyole, M.A., and Cohen, A.H. (1987). Recurrent Henoch-Schönlein purpura. Kidney International, 78, 495–502.
Henoch–Schönlein purpura following renal transplantation. American Pillebout, E., Thervet, E., Hill, G., Alberti, C., Vanhille, P., and Nochy, D.
Journal of Kidney Diseases, 9, 39–43. (2002). Henoch-Schönlein purpura in adults: outcome and prognostic
Niaudet, P. and Habib, R. (1989). Nephropathie du purpura rhumatoïde. factors. Journal of the American Society of Nephrology, 13, 1271–8.
Presse Medicale, 18, 99–101. Rao, J.K., Weinberger, M., Oddone, E.Z., Allen, N.B., Landsman, P., and
Niaudet, P. and Habib, R. (1998). Methylprednisolone pulse therapy in the Feussner, J.R. (1995). The role of anti-neutrophil cytoplasmic antibody
treatment of severe forms of Schönlein-Henoch purpura nephritis. (c-ANCA) testing in the diagnosis of Wegener granulomatosis. A litera-
Pediatric Nephrology, 12, 238–43. ture review and meta-analysis. Annals of Internal Medicine, 123, 925–32.
Nielsen, H.E. (1988). Epidemiology of Schönlein–Henoch purpura. Acta Rauta, V., Tornroth, T., and Gronhagen-Riska, C. (2002). Henoch-Schoenlein
Paediatrica Scandinava, 77, 125–31. nephritis in adults-clinical features and outcomes in Finnish patients.
Nikibakhsh, A.A., Mahmoodzadeh, H., Karamyyar, M., Hejazi, S., et  al. Clinical Nephrology, 58, 1–8.
(2010). Treatment of complicated henoch-schonlein purpura with Robson, W.L.M. and Leung, A.K.C. (1994). Henoch–Schönlein purpura.
mycophenolate mofetil: a retrospective case series report. International Advances in Pediatrics, 41, 163–94.
Journal of Rheumatology, 2010, 254316. Ronkainen, J., Koskimies, O., Ala-Houhala, M., Antikainen, M., et al. (2006).
Novak, J., Szekanecz, Z., Sebesi, J., et  al. (2003). Elevated levels of Early prednisone therapy in Henoch-Schönlein purpura: a randomized,
anti-Helicobacter pylori antibodies in Henoch–Schönlein purpura. double-blind, placebo-controlled trial. Journal of Pediatrics, 149, 241–7.
Autoimmunity, 36, 307–11. Rosenblum, N.D. and Winter, H.S. (1987). Steroid effects on the course of
O’Donoghue, D.J., Nusbaum, P., Noel, L.H., Halbwachs-Mecarelli, L., and abdominal pain in children with HSP. Pediatrics, 79, 1018–21.
Lesavre, P.H. (1992). Antineutrophil cytoplasmic antibodies in IgA Rostoker, G., Desvaux-Belghiti, D., Pilatte, Y., Petit-Phar, M., Philippon,
nephropathy and Henoch–Schönlein purpura. Nephrology Dialysis C., Deforges, L., et  al. (1994). Immunoglobulin therapy for severe
Transplantation, 7, 534–8. IgA nephropathy and Henoch–Schönlein purpura. Annals of Internal
Olson, J.C., Kelly, K.J., Pan, C.G., and Wortmann, D.W. (1992). Pulmonary Medicine, 120, 476–84.
disease with hemorrhage in Henoch–Schönlein purpura. Pediatrics, 89, Rostoker, G., Desvaux-Belghiti, D., Pilatte, Y., Petit-Phar, M., Philippon, C.,
1177–81. Deforges, L., et al. (1995). Immuno-modulation with low-dose immu-
Oner, A., Tinaztepe, K., and Erdogan, O. (1995). The effect of triple ther- noglobulins for moderate IgA nephropathy and Henoch–Schönlein pur-
apy on rapidly progressive Henoch–Schönlein nephritis. Pediatric pura. Nephron, 69, 327–34.
Nephrology, 9, 6–10. Roth, R., Wilz, D.R., and Theil, G.B. (1985). Schönlein–Henoch syndrome in
Ostergaard, J.R. and Storm, K. (1991). Neurologic manifestations of adults. Quarterly Journal of Medicine, 55, 145–59.
Schönlein-Henoch purpura. Acta Paediatrica Scandinava, 80, 339–42. Roy, S. (1972). Steatorrhoea in Henoch’s syndrome. British Medical Journal,
Ozaltin, F., Bakkaloglu, A., Ozen, S., Topaloglu, R., Kavak, U., Kalyoncu, M., 3, 682.
and Besbas, N. (2004). The significance of IgA class of antineutrophil Rueda, B., Perez-Armengol, C., Lopez-Lopez, S., Garcia-Porrúa, C., Martin,
cytoplasmic antibodies (ANCA) in childhood Henoch–Schönein pur- J., and González-Gay, M.A. (2006). Association between functional hap-
pura. Clinical Rheumatology, 23, 426–9. lotypes of vascular endothelial growth factor and renal complications in
Ozdemir, H., Isik, S., Buyan, N., and Hasanoglu, E. (1995). Sonographic dem- Henoch–Schönlein purpura. Journal of Rheumatology 33, 69–73.
onstration of intestinal involvement in Henoch–Schönlein syndrome. Ruperto, N., Ozen, S., Pistorio, A., Dolezalova, P., Brogan, P., Cabral, D.A.,
European Journal of Radiology, 20, 32–4. et  al. (2010). EULAR/PRINTO/PRES criteria for Henoch-Schönlein
Ozen, S., Pistorio, A., Lusan, S.M., Bakkaloglu, A., Herlin, T., et al. (2010). purpura, childhood polyarteritis nodosa, childhood Wegenerr granulo-
EULAR/PRINTO/PRES criteria for Henoch-Schönlein purpura, child- matosis and childhood Takayasu arteritis: Ankara 2008. Part I: Overall
hood polyarteritis nodosa, childhood Wegener granulomatosis and methodology and clinical characterization. Annals of the Rheumatic
childhood Takayasu arteritis: Ankara 2008. Part II: Final classification Diseases, 69, 790–7.
criteria. Annals of the Rheumatic Diseases, 69, 798–806. Sams, W.M., Claman, H.N., Kholer, P.F., McIntosh, R.M., Small, P., and Mass,
Ozen, S., Ruperto, N., Dillon, M.J., Bagga, A., Barron, K., et al. (2006). P.S. (1975). Human necrotizing vasculitis: immunoglobulins and com-
EULAR/PReS endorsed consensus criteria for the classification plement in vessel walls of cutaneous lesions and normal skin. Journal of
of childhood vasculitides. Annals of the Rheumatic Diseases, 65, Investigative Dermatology, 64, 441–5.
936–41. Sánchez-Guerrero, J., Gutiérrez-Ureña, S., Vidaller, A., Reyes, E., Iglesias, A.,
Özkaya, O., Soylemezoglu, O., Gonen, S., Misirhoglu, M., et  al. (2006). and Alarcón-Segovia, D. (1990). Vasculitis as a paraneoplastic syndrome.
Renin-angiotensin system gene polymorphisms: association with sus- Report of 11 cases and review of the literature. Journal of Rheumatology,
ceptibility to Henoch-Schonlein purpura and renal involvement. Clinical 17, 1458–62.
Rheumatology, 25, 861–5. Santiago, J., Blanco, R., González-Gay, M.A., Mateos, A., Roses, L., Andrade,
Pacheco, A. (1991). Henoch–Schönlein vasculitis and tuberculosis. Chest, A.S., et al. (1996). Henoch–Schönlein purpura with hemorrhagic ascites
100, 293–4. and intestinal serositis. Gastrointestinal Endoscopy, 44, 624–5.
546 SECTION 5 vasculitic diseases and syndromes and related disorders

Saulsbury, F.T. (1986). Case report:  antinuclear antibody in Henoch– Szer, I.S. (1994). Henoch–Schönlein purpura. Current Opinion in
Schönlein purpura. American Journal of the Medical Science, 291, 180–2. Rheumatology, 6, 25–31.
Saulsbury, F.T. (1993). Corticosteroid therapy does not prevent nephritis in Szer, I.S. (1996). Henoch–Schönlein purpura: when and how to treat. Journal
Henoch–Schönlein purpura. Pediatric Nephrology, 7, 69–71. of Rheumatology, 23, 1661–5.
Saulsbury, F.T. (1999). Henoch–Schönlein purpura in children. Report of 100 Tancrede-Bohin, E., Ochonisky, S., Vignon-Pennamen, M.D., Flageul,
patients and review of the literature. Medicine (Baltimore), 78, 395–409. B., Morel, P., and Rybojad, M. (1997). Schönlein-Henoch purpura
Saulsbury, F.T. (2001). Henoch–Schönlein purpura. Current Opinion in in adult patients: predictive factors for IgA glomerulonephritis in
Rheumatology, 13, 35–40. a retrospective study of 57 cases. Archives of Dermatology, 133,
Schifferli, J., Leski, M., Favre, H., Imbach, P., Nydegger, U., and Davies, K. 438–42.
(1991). High-dose intravenous IgG treatment and renal function. Tarshish, P., Bernstein, J., and Edelmann, C.M. Jr. (2004). Henoch–Schönlein
Lancet, 337, 457–8. purpura nephritis: course of disease and efficacy of cyclophosphamide.
Schönlein, J.L. (1837). Allgemeine und specielle Pathologie und Therapie, Vol. Pediatric Nephrology, 19, 51–6.
2, 3rd edn. Wurzburg, Herisau. Tizard, E.J. (1999). Henoch–Schönlein purpura. Archives Diseases in
Shenoy, M., Bradbury, M.G., Lewis, M.A., and Webb, N.J.A. (2007). Outcome Children, 80, 380–3.
of Henoch-Schönlein purpura nephritis treated with long-term immu- Topaloglu, R., Sungur, A., Baskin, E., Besbas, N., Saatci, L., and Bakkalogllu,
nosuppression. Pediatric Nephrology, 22, 1717–22. A. (2001).Vascular endothelial growth factor in Henoch-Schonlein pur-
Shimomura, N., Kawai, K., Watanabe, S., Katsuumi, K., and Ito, M. (2005). pura. Journal of Rheumatology, 28, 2269–73.
Henoch–Schönlein purpura with severe abdominal pain treated with Trapani, S., Micheli, A., Grisolia, F., Resti, M., Chiappini, E., Falcini, F., et al.
dapsone and factor XIII concentrate. Journal of Dermatology, 32, 124–7. (2005). Henoch Schönlein purpura in childhood: epidemiological and
Shin, J.I., Park, J.M., Shin, Y.H., Kim, J.H., Kim, P.K., Lee, J.S., et al. (2005a). clinical analysis of 150 cases over a 5-year period and review of litera-
Cyclosporin A  therapy for severe Henoch–Schönlein nephritis with ture. Seminars Arthritis Rheumatism, 35, 143–53.
nephrotic syndrome. Pediatric Nephrology, 20, 1093–7. Trygstad, C.W. and Stiehm, E.R. (1971). Elevated serum IgA globulin in ana-
Shin, J.I., Park, J.M., Shin, Y.H., Kim, J.H., Lee, J.S., Kim, P.K., et al. (2005b). phylactoid purpura. Pediatrics, 47, 1023–8.
Can azathioprine and steroids alter the progression of severe Henoch– Uthman, I., Kassak, K., and Nasr, F.W. (1998). Henoch–Schönlein purpura in
Schönlein nephritis in children? Pediatric Nephrology, 20, 1087–92. adulthood and childhood. Arthritis and Rheumatism, 41, 1518.
Shrestha, S., Sumingan, N., Tan, J., Althous, H., McWilliam, L., and Van den Wall Bake, A., Lobatto, S., and Jonges, L. (1987). IgA antibodies
Ballardie, F. (2006). Henoch Schönlein purpura with nephritis in directed against cytoplasmic antigens of polymorphonuclear leuko-
adults: adverse prognostic indicators in a UK population. Quarterly cytes in patients with Henoch–Schönlein purpura. Nephrology Dialysis
Journal of Medicine, 99, 253–65. Transplantation, 7, 1238–41.
Smith, G.C., Davidson, J.E., Hughes, D.A., Holme, E., and Beattie, T.J. (1997). Wakaki, H., Ishikura, K., Hataya, H., Hamasaki, Y., et  al. (2011).
Complement activation in Henoch–Schönlein purpura. Pediatric Henoch-Schönlein purpura nephritis with nephrotic state in chil-
Nephrology, 11, 477–80. dren: predictors of poor outcome. Pediatric Nephrology, 26, 921–5.
Somer, T. and Finegold, S.M. (1995). Vasculitides associated with infections, Watts, R.A., Jolliffe, V.A., Grattan, C.E.H., Elliot, J., Lockwood, M., and Scott,
immunizations and antimicrobial drugs. Clinical Infectious Diseases, 20, D.G.I. (1998). Cutaneous vasculitis in a defined population: clinical and
1010–36. epidemiological associations. Journal of Rheumatology, 25, 920–4.
Soylemezoglu, O., Peru, H., Gonen, S., Cetinyurek, A., and Buyan, N. (2008). Weiler-Bisig, D., Ettlin, G., Brink, T., Arnold, W., Glatz-Krieger, K., and
HLA-DRB1 alleles and Henoch-Schönlein purpura: susceptibility and Fischer, A. (2005). Henoch–Schönlein purpura associated with esopha-
severity of disease. Journal of Rheumatology, 35,1165–8. gus carcinoma and adenocarcinoma of the lung. Clinical Nephrology,
Specks, U. and Homburger, H.A. (1994). Laboratory medicine and pathol- 63, 302–4.
ogy. Anti-neutrophil cytoplasmic antibodies. Mayo Clinic Proceedings, Weiss, P., Klink, A., Localio, R., Hall, M., Hexem, K., et  al. (2010).
69, 1197–8. Corticosteroids may improve clinical outcomes during hospitalization
Stancanelli, B., Vita, A., Vinci, M., Magnano, A., and Purrello, F. (2006). for henoch-Schönlein purpura. Pediatrics, 126, 674–81.
Bleeding of small bowel in Henoch-Schonlein syndrome: the success- White, R.H.R. (1994). Henoch–Schönlein nephritis. A disease with signifi-
ful diagnostic role of video capsule endoscopy. American Journal of cant late sequelae. Nephron, 68, 1–9.
Medicine, 119, 82–4. Woolfenden, A.R., Hukin, J., Poskitt, K.J., and Connolly, M.B. (1998).
Stefansson Thors, V., Kolka, R., Sigurdardottir, S.L., Edvardsson, V.O., Arason, Encephalopathy complicating Henoch–Schönlein purpura:  reversible
G., and Haraldsson, A. (2005). Increased frequency of C4B*Q0 alleles MRI changes. Pediatric Neurology, 19, 74–7.
in patients with Henoch–Schönlein purpura. Scandinavian Journal of Yang, Y.-H., Hung, C.-F., Hsu, C.-R., Wang, L.-C., Chuang, Y.-H., Lin, Y.-
Immunology, 61, 274–8. T., and Chiang, B.-L. (2005). A nationwide survey on epidemiological
Sterky, G. and Thilén, A. (1960). A study on the onset and prognosis of acute characteristics of childhood Henoch-Schönlein purpura in Taiwan.
vascular purpura (the Schönlein-Henoch syndrome) in children. Acta Rheumatology, 44, 618–22.
Paediatrica, 49, 217–29. Yoshikawa, N., White, R.H.R., and Cameron, A.H. (1981). Prognostic sig-
Stevens, G.L., Adelman, H.M., and Wallach, P.M. (1995). Palpable pur- nificance of the glomerular changes in Henoch–Schönlein nephritis.
pura: an algorithmic approach. American Family Physician, 52, 1355–62. Clinical Nephrology, 16, 223–9.
Stewart, M., Savage, J.M., Bell, B., and McCord, B. (1988). Long term renal Zeng, H., Xiong, X., Chen, Y., and Luo, X. (2009). Gene polymorphism of
prognosis of Henoch–Schönlein purpura in an unselected childhood vascular endothelial growth factor in children with Henoch-Schonlein
population. European Journal of Pediatrics, 147, 113–5. purpura nephritis. Chinese Journal of Contemporary Pediatrics, 11,
Sussman, M., Jones, J.H., Almeida, J.D., and Lachmann, P.J. (1973). 417–21.
Deficiency of second component of complement associated with ana- Zhang, Y. and Huang, X. (2008). Gastrointestinal involvement in
phylactoid purpura and presence of mycoplasma in the serum. Clinical Henoch-Schönlein purpura. Scandinavian Journal of Gastroenterology,
and Experimental Immunology, 14, 531–9. 43, 1038–43.
CHAPTER 40

Cryoglobulinaemic
vasculitis
Massimo Galli, Salvatore Sollima, Francesco
Saccardo, and Giuseppe Monti

Professor Angelo Monteverde, MD (1930–2001), and Professor et al. 1977; Galli et al. 1980; Popp et al. 1980), this hypothesis influ-
Fulvio Invernizzi, MD (1929–2007), who participated in the prepa- enced research until, 13 years later, Pascual et al. (1990) demon-
ration of the previous editions, are no longer with us, but remain strated an association between a large majority of EMC and the
in our thoughts through their teaching. The present edition of this newly discovered HCV, thus opening up new research perspectives
chapter is dedicated to their memory. and suggesting new therapeutic opportunities.

Definition Epidemiology
Cryoglobulinaemic vasculitis (CV) is a systemic vasculitis affecting Monoclonal (type I) CV is a rare disorder, occurring in a minor-
small and medium-sized vessels that is caused by immune com- ity of cases of patients suffering from lymphoproliferative diseases,
plexes of cryoprecipitating immunoglobulins and is sustained by such as Waldenström’s macroglobulinaemia, multiple myeloma
benign B-cell clonal proliferation. The most frequent pathological (Payet et  al. 2013), or, less frequently, chronic lymphocytic leu-
trigger is hepatitis C virus (HCV). Palpable purpura is the clinical kaemia. It accounted for 6–15% of total cases in the largest case
hallmark of the disease. series of symptomatic cryoglobulinaemias (Monti et  al. 1995a;
Dammacco et al. 2001).
MC represents the large majority of reported cases, with differ-
Historical note ences in the percentage of MC type II and type III in the differ-
The formation of a cryoprecipitate (a sugar-like deposit at the ent studies. For example, type II accounted for 62% and type III
bottom of a test tube of serum at temperatures of less than 37°C) for 31% of a large series of 694 cryoglobulinaemic patients (Monti
(Figure 40.1) was first observed in 1933 in relation to a patient with et al. 1995a), but the MC type II accounted for only 26.8% of the
multiple myeloma (Wintrobe and Buell 1933). The term ‘cryoglo- 265 cases described in another study (Tissot et al. 1994). In Europe,
bulin’ was introduced in 1947 by Lerner and Watson, who dem- CV is included in the list of rare diseases (i.e. among the diseases
onstrated the reversibility of the phenomenon when the sera were whose prevalence in the general population is lower than 5/10 4).
heated to 37°C. In 1954, Petermann and Braunsteiner reported Nevertheless, in a population-based study performed in Northern
cryoprecipitates of immunoglobulins with different sedimentation Italy, we estimated an overall prevalence of HCV-related CV not
rates, thus introducing the concept of ‘mixed cryoglobulinaemia’ less than 8.5/104 (Monti et al. unpublished data).
(MC). At about the same time, Steinhardt and Fisher (1955) first It is likely that the prevalence of HCV infection in different geo-
described cryoglobulinaemias apparently unassociated with lym- graphical areas is decisive in influencing differences in the local CV
phoproliferative diseases as ‘essential’. The role of a rheumatoid fac- incidence. Similarly, the prevalence of HCV in the general popula-
tor (RF) in cryoprecipitation was first demonstrated by Lospalluto tion determines the rate of HCV-related CV out of the total num-
et al. in 1962. ber of the observed cases of CV. In fact, in published case series,
The classic triad of symptoms (purpura, asthenia, and arthralgia) the percentages of HCV-related CV ranges from 30 to nearly 100%,
was initially described by Meltzer and Franklin (1966), who applied with the lowest prevalence found in Northern Europe and North
the term ‘essential MC’ (EMC) to the cases unassociated with lym- America and the highest in the Mediterranean area (Sansonno
phoproliferative or autoimmune diseases. Some years later, Brouet et al. 2007; Ramos-Casals et al. 2012). This reflects quite closely the
et al. (1974) proposed their immunochemical classification of mon- different geographical prevalence of HCV infection in the elderly
oclonal cryoglobulin (type I), mixed cryoglobulins with a mono- (Cornberg et al. 2011).
clonal component (type II), and mixed polyclonal cryoglobulins On the other hand, the percentage of HCV-infected patients
(type III). found to be positive for circulating mixed cryoglobulins (CMC)
In 1977, Levo et al. suggested hepatitis B virus (HBV) as the pos- ranges from 10 to 70%, and is higher in Southern Europe than
sible aetiological agent in EMC. Although not confirmed (Dienstag in Northern Europe and North America (Kayali et al. 2002). The
548 SECTION 5 vasculitic diseases and syndromes and related disorders

Causes of cryoglobulin production


and diseases associated with CV
Infections
Ever since the first studies of cryoglobulins (Wager et al. 1969), it has
been hypothesized that the IgG component represents antibodies
against bacterial or viral antigens. Circulating cryoglobulins have
been found in many acute and chronic infections due to a variety of
pathogens (Galli et al. 1986) (Table 40.1). Generally, cryoglobulins
disappear from circulation following symptom regression and true
CV occurs in only a few cases. Therefore, with the exceptions of
HCV, probably HBV, and perhaps a few other agents responsible
for chronic infections and with propensity for stimulation of B-cell
compartment, it is unclear whether the majority of infections in the
course of which CMC are found are truly capable of causing CV or
if this finding is simply a transient phenomenon of modest and/or
uncertain clinical significance. It is also conceivable that genetically
predisposed people can produce CMC more efficiently and develop
frank vasculitis following stimulation caused by an infection. In
this case, characteristics of the infectious agent acting as a trigger
might be relatively unimportant.
Hepatitis C virus
Before the discovery of HCV a number of authors (Florin-
Christensen et al. 1974; Jori and Buonanno 1972; Levo et al. 1977;
Fig. 40.1 Cryoglobulin precipitation in a cryocrit tube. McIntosh et al. 1976; Realdi et al. 1974; Galli et al. 1982) suggested
that hepatotropic viruses might play a role in evoking cryoglobulin
production.
annual incidence rate of cmc in hcv-positive patients has been The association between HCV and cryoglobulinaemic vasculitis
reported to be 3% (Adinolfi et  al. 1996)  And cryoglobulins can became evident in the early 1990s, when active HCV infection was
be detected in more than 50% of hcv-infected italian patients, but found in the vast majority of patients with EMC (Pascual et al. 1990;
overt cv was seen in only about 5% of them (Ferri et  al. 2004). Casato et al. 1991; Disdier et al. 1991; Durand et al. 1991; Ferri et al.
In contrast, in the united states, the prevalence of cmc has been 1991; Galli et al. 1992b; Agnello et al. 1992). The demonstration
reported to be 10–50%, with a much lower prevalence of vasculitis of HCV in immune complexes in the kidney, cutaneous vasculi-
(Giordano et al. 2007). tis lesions, and vessel walls of cryoglobulinaemic patients (Agnello
One should consider that the variability of results obtained 1997; Agnello and Abel 1997; Agnello et al. 1992; Sansonno et al.
in the studies on mc might also be attributable to selection 1995; Sansonno et al. 1997) provided further support for a causa-
biases. For example, Lunel et al. (1994) Found that hcv-infected tive role of HCV. CV is therefore now considered a common and
cmc-positive patients had a duration of hcv infection almost definitely validated extrahepatic manifestation of HCV infection
twice longer than hcv-infected cmc-negative patients, yield- (Agnello and De Rosa 2004).
ing information lacking in other investigations. Additionally, HCV is a single-stranded positive-sense RNA virus, which
the laboratory procedures for cmc detection are prone to belongs to the Flaviviridae family of RNA viruses and has a genome
false-negative results, and differences in the procedure adopted consisting of about 9500 nucleotides. Six major genotypes have
might explain a large part of the discrepancies in the results been identified. The virus mutates rapidly and generates quasispe-
(Charles and Dustin 2009). Moreover, many studies do not dis- cies, a series of related but immunologically distinct variants.
tinguish between the mere detectability of cmc and the presence One of the most intriguing questions is why only some
of overt vasculitis, causing confusion in interpreting the data HCV-infected patients develop CV. In fact, CMC are frequently
and comparing results of different reports. detectable in patients with chronic hepatitis C, although they
A significant limitation of epidemiological studies has been the are usually present in small quantities and classifiable as type III,
lack of validated classification criteria for cv. Several attempts at while their presence is associated with clinical manifestations in
creating a clinical classification system have been made since the no more than 1% of HCV-infected patients, according to Meyers
1990s (Invernizzi et al. 1995; Ferri et al. 2002; Ferri 2008), but none et al. (2003). Lunel et al. (1994) found that the prevalence of MC
of them were based on appropriate statistical support. The gisc increases with the progression of chronic hepatitis C, and Kayali
(gruppo italiano per lo studio delle crioglobulinemie) has com- et  al. (2002) found symptomatic cryoglobulinaemia in 34% of
pleted a large co-operative international study for the definition of patients with cirrhosis due to HCV.
classification criteria of cv according to standard methodologies Like other RNA viruses, HCV is characterized by a high degree
(De vita et al. 2012). The performance of the gisc criteria for cv was of genetic heterogeneity (Ogata et al. 1991; Okamoto et al. 1992).
also assessed comparing hcv-positive and hcv-negative patients The possibility that particular strains of HCV have a propen-
with cmc (Quartuccio et al. 2012). sity for inducing cryoglobulin production, which may trigger a
CHAPTER 40 cryoglobulinaemic vasculitis 549

Table 40.1 Diseases responsible of mixed cryoglobulinaemias

Infections RNA viruses Hepatitis C virus; HIV-1; HTLV-1; hepatitis A virus; hepatitis E virus; Chikungunya virus; Dengue
virus; hantavirus
DNA viruses Hepatitis B virus; Epstein–Barr virus; cytomegalovirus; parvovirus B-19; Adenovirus
Chlamydiae and Rickettsiae Chlamydia psittaci; Rickettsia conorii
Bacteria Streptococcus spp.; Brucella spp.; Coxiella spp.; Klebsiella spp.; Proteus mirabilis; Mycobacterium
tuberculosis; Mycobacterium leprae; Leptospira icterohaemorrhagiae; Borrelia burgdorferi; Treponema
pallidum
Fungi Coccidioides immitis
Protozoa Toxoplasma gondii; Leishmania donovani; Plasmodium falciparum; Entamoeba histolytica
Helminths Echinococcus granulosus; Schistosoma mansoni
Vaccinations Influenza vaccination; hepatitis B vaccination; intravesical BCG
Autoimmune diseases Sjögren’s syndrome; systemic lupus erythematosus; rheumatoid arthritis; systemic sclerosis;
antiphospholipid syndrome; inflammatory myopathies; adult-onset Still’s disease; polyarteritis
nodosa; giant-cell arteritis; Takayasu’s arteritis; ANCA-associated vasculitis; autoimmune hepatitis
Cancers B-cell lymphoma; multiple myeloma; Waldenström’s disease; Hodgkin’s lymphoma; chronic
lymphocytic leukaemia; chronic myeloid leukaemia; myelodysplasia; hepatocellular carcinoma;
papillary thyroid cancer; lung adenocarcinoma; renal cell carcinoma; nasopharyngeal carcinoma
Other causesa Alcoholic cirrhosis; jejunoileal bypass for morbid obesity; graft dysfunction; myocardial infarction;
Moya moya disease
a Co-trimoxazole, interferon-α, intravenous radiographic contrast administration, and cocaine use have been reported to be responsible of cryoglobulinaemic exacerbation in some
patients.
ANCA, antineutrophil cytoplasmic antibodies.
Items in bold refer to the leading causes of mixed cryoglobulinaemias.

vasculitic syndrome, has been examined in several studies. No sig- women aged more than 50  years, with often no risk factors for
nificant association of CV with particular HCV genotypes has been HCV infection. It is probable that genetic factors, with different
observed (Zehender et al. 1995; Zignego et al. 1996), although a distribution across populations of Caucasian ethnicity, can influ-
Brazilian study (Vigani et al. 2008) reported a significantly greater ence individual responses to HCV infection and consequently the
incidence of cryoglobulinaemia in patients infected with HCV incidence of CV. In fact, HLA studies in HCV-related MC observed
genotype 3 than in those with genotype 1.  The greater tendency that HLA-DR11 is more frequent in patients with type II MC than
of genotype 3 than other genotypes to cause liver fibrosis (Cholet in those without it, but no correlation with the presence or absence
et al. 2004), which might be associated with greater immune system of vasculitis was found (Cacoub et al. 2001). Moreover, an Italian
stimulation by impaired clearance of antigens of intestinal origin, study suggested that DR5 and DQ3 HLA class  II clusters and a
might explain these findings. However, we have detected genotype higher frequency of HLA homozygosity may play a role in the clini-
3 only in a minority of HCV-related CV. cal outcome of type II symptomatic HCV-related MC (De Re et al.
A single amino acid insertion/ deletion at codons 384–385 of the 2007). Patients with HCV-related CV also showed a significant
hypervariable region 1 (HVR1) of HCV was found in one-third increase in serum B-cell activating factor (BAFF) concentrations
of patients with CV (Gerotto et al. 2001). Nevertheless, only one (Toubi et al. 2006; Fabris et al. 2007; Sène et al. 2007), and a higher
of our patients was infected by a virus showing a five-amino acid prevalence of −871 T/T homozygosity (31% versus 16%; P = 0.001)
insertion at codon 385–389 of the HVR (Zehender et al. 2005), and and a greater frequency of T alleles of the BAFF promoter (80%
Bianchettin et al. (2007) found insertions at position 385 of HVR1 versus 57%; P = 0.004) when compared with HCV-infected patients
in samples from cryoglobulinaemic patients less frequently (6.2%) without CMC (Gragnani et al. 2011).
than in CMC-negative controls (9.2%). In summary, cryoglobulins are a frequent finding in patients
In comparison with controls, the HCV in CV patients was less with chronic HCV infection, and their detection (particularly when
heterogeneous and more frequently compartmentalized, with the they are produced in small amounts) depends on the accuracy of
plasma, cryoprecipitates, and peripheral blood mononuclear cells the method, the maintenance of a temperature of 37°C or greater
(PBMCs) showing different quasispecies content (Zehender et al. during sample handling, and the amount of blood processed. In
2005). Reduced HCV heterogeneity has been observed in patients line with this, the percentage of chronically HCV-infected patients
with long-lasting HCV infections (Allain et al. 2000) and in immu- producing cryoglobulins is probably underestimated, but cryo-
nocompromised hosts (Toyoda et al. 1997; Booth et al. 1998), but globulins have no clinical significance in the majority of cases.
it is still unclear whether HCV quasispecies compartmentalization On the contrary, the HCV-infected patients developing frank CV
plays a role in the pathogenesis of CV. represent a minority of probably genetically predisposed subjects,
The most likely explanation of cryoglobulin overproduction in and a minority of the HCV-infected patients in whom cryoglo-
a minority of patients probably lies in the host rather than in the bulins are at least occasionally isolated. The origin of the inves-
virus. Patients with symptomatic cryoglobulinaemia are mainly tigated cases (rheumatology or haematology versus hepatology
550 SECTION 5 vasculitic diseases and syndromes and related disorders

or gastroenterology units) may lead to selection biases, and the of vasculitis symptoms in association with the decline of CD4 cell
reported variability in the prevalence of CV among HCV-infected count (Antinori et al. 1995).
patients (Agnello and De Rosa 2004). Chikungunya virus is an alphavirus causing a febrile disease fre-
quently followed by chronic rheumatism, sometimes lasting for
Other viral infections years. Studies have suggested a relevant role for cryoprecipitating
On the basis of serological data, Levo et al. (1977) claimed that immune complexes in the pathogenesis of the arthritis associated
the majority of EMCs were caused by HBV, but the real signifi- with this disease. MC, prevalently of type II, was found in 94% of
cance of these findings was subsequently questioned and attrib- patients positive for anti-Chikungunya virus IgM antibodies. Over
uted to confounding variables, such as age, geographical origin, 90% of them had concomitant arthralgias. Significant amounts of
socioeconomic status, and frequent hospitalizations (Dienstag CMC were also found 6  months after the onset of symptoms of
et al. 1977; Galli et al. 1980; Popp et al. 1980). In particular, only a acute infection (Oliver et al. 2009). Interestingly, purpura was not
slight difference in the prevalence of serological markers of HBV seen in these patients, and it was thought to be due to the relatively
was found when a large series of cryoglobulinaemic patients was low median levels (8 mg/l) of serum cryoglobulins.
compared with hospitalized patients or blood donors resident in The finding of CMC in a patient with acute Dengue virus infec-
the same areas (Galli 1991; Galli et al. 1992a). Critical rereading tion causing a complex pattern of autoimmune disorders (Jardim
of the paper by Levo et al. and subsequent refutations with cur- et al. 2012) suggests that this flavivirus may play a role in evoking
rent knowledge raises the possibility of occult HBV infection. cryoglobulin production.
The role of an underlying occult HBV infection in the produc- EBV is a good inducer of polyclonal stimulation of B lympho-
tion of cryoglobulins has never been investigated. It must be said, cytes, and has also been considered an obvious candidate among
however, that almost all patients whose data were included in our the possible causes of cryoglobulinaemia (Fiorini et  al. 1988).
papers in order to refute the hypothesis of Levo et al. (Galli 1991; Despite this, the evidence of its involvement remains sparse. A case
Galli et al. 1992a) subsequently had chronic infections with HCV. of cryoglobulinaemia with fever, anaemia, numbness of the lower
Nevertheless, HBV probably plays a major role in causing CV in limbs, pleural effusion, thrombocytopenia, and liver dysfunction
some patients, and findings suggest that it might be a significant was reported in an elderly woman diagnosed with chronic active
cause of symptomatic cryoglobulinaemia in children (Liou et al. EBV infection (Ichinose et al. 2013).
2013). CMC can be detected in patients with acute and chronic Since 1993, the association of MC with parvovirus B19 acute
HBV infections, but are generally absent in chronic carriers not infection has been described in no more than four case reports. The
showing clinical or laboratory evidence of liver damage. In the two most recent reports consisted of adult patients (Chiche et al.
last survey of HCV-negative symptomatic cryoglobulinaemias 2010; Gorse et al. 2011), the second of which had a frank purpura.
performed by GISC, chronic HBV infection was reported in 4% The vast majority of the other reports of CMC in the course of
of cases of CV (Galli et al. unpublished data), which imputed less viral infections other than HCV listed in Table 40.1 should be con-
than 0.3% of symptomatic cryoglobulinaemias observed in Italy sidered transient phenomena of uncertain clinical significance.
to HBV.
A sizable transient cryoglobulin production has also been Other infectious agents
described during the acute phase of hepatitis A (Galli et al. 1986; CV is rarely observed in patients with bacterial infections, although
Shalit et  al. 1982), and polyclonal IgM (including antihepatitis CMC production is a common finding.
A  virus-specific IgM) have been found in cryoprecipitates, but Renal failure, purpura, and peritonitis were described in a man
they rapidly decrease with symptom remission (Galli et al. 1986; with acute brucellosis and type III cryoglobulinaemia (Hermida
Galli 1995). It is worth mentioning that liver damage induced by Lazcano et al. 2005). In this patient the cryocrit reached 1.4%, but
hepatotropic viruses itself may have a significant role in triggering CMC and cutaneous vasculitis disappeared when the infection
the production of CMC, through impaired clearance of antigens of was cured. The authors reviewed some other cases, including two
intestinal origin. reported by us (Galli et al. 1986), thus suggesting that Brucella spp.
Variable but generally small amounts of cryoglobulins were found infection can trigger the production of ‘pathogenic’ cryoglobulins
in human immunodeficiency virus (HIV)-1 infected patients (Galli more efficiently than other bacterial diseases. Even in this case,
et al. 1986; Matsuda et al. 1994; Taillan et al. 1993), but there is some however, it was a transient phenomenon, which does not determine
doubt as to whether they are attributable to HIV-1 or co-infections the onset of chronic or relapsing vasculitis.
with viruses such as Epstein–Barr virus (EBV) or HCV. Scotto et al. CV with purpuric manifestations subsiding after antiparasitic
(2006) found a prevalence of CMC of 6% in HIV-monoinfected therapy was also observed in patients suffering from visceral leish-
patients and 14.2% in HIV–HCV co-infected patients. In spite of maniasis (Casato et al. 1999; Rizos et al. 2005).
this high prevalence, cryoglobulinaemia is rarely symptomatic Other infections in the course of which CMC have been
(Bonnet et al. 2003), although neurological, dermatological, renal, described, in most cases without evidence of frank CV, are listed
or rheumatological complications have sometimes been described in Table 40.1.
(Saadoun et  al. 2006a). The occurrence of symptomatic MC has
been described during HIV primary infection (Genet et al. 2011). Autoimmune diseases
A decline in the prevalence of cryoglobulinaemia in HIV-infected Systemic autoimmune diseases are probably the second leading
patients has been reported following the introduction of highly cause of CMC production and CV after infections in general and
active antiretroviral therapy (Kosmas et  al. 2006). Interestingly, HCV in particular. In the large series of GISC published in 1995
the occurrence of HIV infection in a patient who had been diag- (Monti et  al. 1995), symptomatic cryoglobulinaemias associated
nosed with HCV-related CV some years before led to remission with connective tissue disorders were 49 out of 891 (5.5%), but in
CHAPTER 40 cryoglobulinaemic vasculitis 551

a later revision (unpublished data) about half of these cases were in the cryocomplexes throughout the entire period of observation
found to be infected with HCV. In a more recent GISC study (Galli (Konstantinova et al. 2011). The phenomenon seems to be ascribed
et al. unpublished data), autoimmune diseases accounted for 32% to the acute inflammatory process associated with heart attack
(41/125) of total HCV-negative cases (i.e. about 1.6% of total CV rather than to the pre-existence of underlying CV.
attended by GISC Centres):  18 patients suffered from primary
Sjögren’s syndrome and 12 from systemic lupus erythematosus. Essential cryoglobulinaemias
In a French survey (Terrier et al. 2012), connective tissue diseases After the discovery of HCV as the aetiological agent of the major-
were the cause of CV in 73/242 patients (30.2%), 61 of whom had ity of CV, the number of cases that still elude any known asso-
primary Sjögren’s syndrome. ciation has thinned so much that they have almost disappeared.
Among autoimmune disorders, primary Sjögren’s syndrome is Nevertheless, 60% of HCV-negative CV in the GISC study (Galli
the most frequently associated with frank CV, while cryoglobulinae- et al. unpublished data) and 48% in the French series did not show
mia in Sjögren’s syndrome patients is associated with extraglandu- any association with autoimmune, lymphoproliferative, or chronic
lar involvement, enhanced risk of developing B-cell lymphoma, and infectious diseases.
poor survival (Baimpa et al. 2009; Brito-Zerón et al. 2007; Tzioufas A possible role of latent HCV infection in causing at least some
et al. 1996). Cryoglobulins can be detected in 25% of patients with cases of EMC might be suggested by the findings of Casato et al.
systemic lupus erythematosus (García-Carrasco et al. 2001), and (2003), who reported the appearance of HCV RNA in the serum
in a wide range of other autoimmune disorders (Table 40.1), but and/or cryoprecipitate of three previously HCV RNA-negative
cryocrit values are frequently low. and persistently anti-HCV antibodies-negative subjects with
In the case of autoimmune diseases there is some confusion gen- type II MC.
erated by insufficient distinction between simple presence of CMC,
which is quite frequent, and frank CV, which is more unusual. Pathogenesis
Moreover, in patients with connective tissue diseases, especially
those in whom arthralgias are prominent, it is often difficult to B-cell clonal expansion
distinguish the cases in which CMC are an epiphenomenon with- Clonal expansion of B cells, triggered by persistent immune stimu-
out clinical significance from those in which the cryoglobulins are lation or in the context of lymphoproliferative disorders, is the key
frankly pathogenic. factor for cryoglobulin production. The model in which the mecha-
nisms responsible for the production of cryoglobulins have been
Malignancies extensively studied is HCV infection, while there are few data on
As mentioned in Section Epidemiology, type I cryoglobulinaemia non-HCV-related MC. The E2 HCV envelope protein binds human
can occur in patients with B-cell lymphoproliferative diseases, CD81, a tetraspanin expressed by various cell types, including
such as Waldenström’s macroglobulinaemia, multiple myeloma, or hepatocytes and B lymphocytes (Pileri et al. 1998) and is consid-
chronic lymphocytic leukaemia (Dammacco et al. 2007). ered to be the receptor involved in the HCV-mediated activation
The association of MC with B-cell lymphomas was clearly shown of B cells that leads to their polyclonal proliferation and expansion
in the first published large case series (Invernizzi et al. 1983) and (Rosa et al. 2005). HCV lymphotropism was identified in the early
subsequently confirmed also in HCV-negative cases. In the 1990s with the demonstration of viral segments in peripheral blood
GISC HCV-negative symptomatic MC series, 7% of patients had mononuclear cells (Zignego et al. 1992), and it was postulated to
non-Hodgkin’s lymphoma (NHL) as the only possible cause of cry- be the first step of B-cell clonal expansion (Ferri et al. 2007). HCV
oglobulinaemia (Galli et al. unpublished data), while in a French RNA has been detected by polymerase chain reaction in the periph-
series B-cell NHL was associated with cryoglobulinaemia in 22% of eral blood mononuclear cells and bone marrow of patients with MC
cases (Terrier et al. 2012). (Ferri et al. 1993; Galli et al. 1995; Zignego et al. 1992) and in CD34
Cryoglobulins can also be detected in patients with solid cancers haemopoietic progenitor cells (Sansonno et al. 1998), and studies of
(Rullier et al. 2009), but their pathogenic significance is unclear. fractioned subpopulations have detected it almost exclusively at the
B-cell level (Zehender et al. 1997). Moreover, immunohistochemi-
Other causes of CV cal and in situ hybridization studies have revealed HCV proteins in
CMC are frequently found in non-virus-related chronic liver dis- the cytoplasm of lymphoid cells (Sansonno et al. 1996b; Sansonno
orders, including alcoholic cirrhosis (Jori et al. 1977; Breitschwerdt et al. 1996c).
et al. 1999); however, an association with real CV would be quite Lymphoid aggregates with pseudofollicular features and mono-
exceptional. clonal B-cell restriction (Monteverde et al. 1988; Monteverde et al.
A syndrome resembling CV develops in patients undergoing 1995; Monteverde et al. 1997; Sansonno et al. 1998) are detectable
jejunoileal bypass for morbid obesity (Stein et  al. 1981; Gamble in the bone marrow (Figure 40.2a) and liver (Figure 40.2b) of cryo-
et al. 1982). globulinaemic patients, particularly those with type II cryoglob-
Type III cryoglobulin-related graft dysfunction has been ulinaemia (Sansonno et al. 2004; Quartuccio et al. 2007; Charles
described in HCV-negative renal transplant patients (Basse et al. et al. 2008), and are the probable source of cryoglobulin produc-
2006). CMC were found in 30–45% of kidney transplant patients tion (Gabrielli et al. 1994; Galli et al. 1995; Monteverde et al. 1997;
with negative HCV serology (Sens et  al. 2005; Faguer et  al. Sansonno et al. 1996a; Sansonno et al. 1998). In fact, B lympho-
2008)  and in about one-third of HCV-negative liver transplant cytes isolated from hepatic follicles produced RF that most fre-
patients (Garrouste et al. 2008). quently display the WA cross-reactive idiotype (Agnello et al. 1995;
Type III cryoglobulins were reported by a Russian group in Sansonno et al. 1998), previously evidenced in subjects with type
patients with acute myocardial infarction. Fibronectin was detected II cryoglobulinaemia (Mageed et al. 1988). Sansonno et al. (2003b)
552 SECTION 5 vasculitic diseases and syndromes and related disorders

(a) (b)

Fig. 40.2 (a) Bone marrow biopsy of a patient with type II HCV-related cryoglobulinaemia showing a nodular infiltrate consisting of small lymphocytes,
lymphoplasmacytoid elements, and plasma cells (Giemsa, 250 × magnification). (b) Nodular infiltrate in the liver of a patient with HCV-related CV.

have subsequently found cryoprecipitates and purified cryoglo- of anergy in a large proportion of them (Charles et al. 2011); thus,
bulins with IgM RF bearing the same WA cross idiotype that were the authors concluded that, although HCV-infected patients with
linked to anti-HCV core protein IgGs. Moreover, intrahepatic B MC have expanded peripheral B cells capable of differentiating into
cell clonalities were reported to be associated with extrahepatic RF-secreting plasmablasts, these cells do not have transcriptional
manifestations of HCV infection, including high serum levels of RF features of neoplastic transformation, and a significant proportion
activity, cryoglobulins, monoclonal gammopathy of undetermined of this clonal population may be refractory to ongoing antigenic
significance (MGUS), and frank B-cell NHL (Sansonno et al. 2004). stimulation. Another study confirmed that B cell homeostasis in
A role of HCV antigens in the induction of both CV and lym- chronic HCV-related MC is maintained through naïve B cell apop-
phoma was suggested by De Re et  al. (2000a, 2000b), who sub- tosis. Moreover, the total number of B cells was surprisingly lower
sequently showed that the B-cell receptor of the monoclonal, in cryoglobulin-positive than in cryoglobulin-negative patients
over-expanded B-cell population as well as the IgM-RF-positive (Holz et al. 2012).
component of the cryoprecipitate were cross-reactive against HCV In conclusion, HCV could induce lymphoproliferation by anti-
NS3 antigen in patients with CV (De Re et al. 2006b). Interestingly, genic stimulation, direct interaction, or entering in B lymphocytes.
the N-terminal region of E2, the HCV protein that binds CD81 However, the similarity of the N-terminal region of E2 with human
receptor on B cells, is antigenically and structurally similar to Ig variable domains suggests that this may be a main mechanism
human Ig variable domains (Hu et al. 2005), thus representing a for the induction of proliferation of RF-producing clones and their
possible target for RF. Furthermore, the analysis of the complemen- selection toward monoclonality, and might explain the particular
tary determining region (CDR)3 sequences of the IgM-RF-positive aptitude of HCV for inducing RF production. While it now seems
purified from the cryoprecipitate identified HCV-E2 as the antigen established that lymphoproliferation in cryoglobulinaemias has no
driving the production of IgM-RF (Ferri et al. 2006). features of malignancy, it is clear that a minority of HCV-related
The relationship between type II and type III cryoglobulins is MCs evolve into frank lymphoma (see Section Clinical Aspects of
another key point for understanding the evolution of the cryoglo- Cryoglobulinaemic Vasculitis).
bulin disease. De Re et al. (2006a) showed, by GeneScan analysis of
clonal B cells, that type II MC is an oligo rather than a mono B-cell Biochemical characteristics of cryoglobulins
disorder. Landau et al. (2007) subsequently hypothesized that the and mechanism of cryoprecipitation
oligoclonal type II/III cryoglobulins are an intermediate form The biochemical mechanisms of cryoprecipitation are not fully
towards the selection of a single monoclonal IgM-RF, and type III understood. The role of temperature as the only factor for the
cryoglobulins would represent the initial step of this process. precipitation of immmunoglobulins is questioned. The thermal
The possibility that MCs, in particular type II, are smouldering threshold of cryoprecipitation is not the same in all cases and it
lymphomas has long been debated. An oligoclonal non-neoplastic can be assumed that it is in vivo sometimes just under 37°C and
B-cell expansion was shown to be the key feature of type II MC often higher than 4°C, which represents the standard temperature
(De Vita et al. 2000; Quartuccio et al. 2007). Hypermutated, mar- for the in vitro search of CMC. Indeed, if it is conceivable that cold
ginal zone-like IgM+CD27+ B cells, a predominantly RF-encoding exposure could be a contributing factor to clinical manifestations
VH1-69/JH4 and Vκ3-20 gene segment-restricted cell popula- of CV in the distal extremities, it is more difficult to explain, on the
tion, were found to be clonally expanded in certain subjects with basis of temperature changes alone, the involvement of the internal
HCV-related MC (Charles et al. 2008). These data, together with organs. Protein solubility also depends on a range of physical and
those of other investigations (De Re et al. 2006b; Fazi et al. 2010; biochemical factors, which are different in monoclonal and mixed
Ohtsubo et al. 2009) also exclude a prominent role of CD5+ B cells cryoglobulins (Sargur et al. 2010).
as a source of cryoglobulin-related RF, which was suggested in pre-
vious studies (Monteverde et  al. 1997). Contrary to the authors’ Monoclonal cryoglobulins
expectations, B-cell clones revealed a transcriptional profile sug- Monoclonal (type I) cryoglobulins usually consist of IgM, IgG,
gestive of anergy and apoptosis, with immunophenotypic features IgA, or, very rarely, Bence-Jones proteins. A non-immunologically
CHAPTER 40 cryoglobulinaemic vasculitis 553

mediated self-association of monoclonal immunoglobulins gen- immune complex deposition and subsequent vascular inflamma-
erating vasculitis-inducing cryoglobulins was shown by Spertini tion (Sansonno et al. 2009).
et  al. (1988), after the intraperitoneal injection in mice of an Local complement activation might be insufficient to disso-
IgG3-producing hybridoma. The cryoprecipitation of single mono- ciate the aggregates in the presence of IgM RF. Many factors are
clonal paraproteins seems to be strictly linked to the quaternary involved in the failure to clear circulating immune complexes in
structure of the immunoglobulins, because the separated Fc and CV. According to Madi et al. (1991), IgM-RF depletes complement
Fab fragments usually lose their cold-induced precipitability. proteins, enhances immune aggregation reactions, and inhibits
Hydrophobic interactions were also indicated as a factor influenc- the opsonization of immune complexes. A failure in macrophage
ing cryoprecipitation (Hall and Abraham 1984;Wang 1988). Kuroki clearance also contributes to the deposition of immune aggregates.
et al. (2002) demonstrated that the cryoglobulin activity is primar- Patients with active nephritis have impaired liver uptake of immune
ily determined by the galactose content of CH2 oligosaccharide complexes and cryoglobulin processing (Roccatello et al. 1997).
chains, with the secondary involvement of VH chains, and Kuroda An important role in the pathogenesis of CV is probably also
et al. (2005) showed in a murine model that the content of nega- played by cytokines and chemokines (Mathsson et al. 2005). High
tively charged sialic acids in oligosaccharide side chains is one of concentrations of soluble TNF-α receptors (sTNFR1 and sTNFR2)
the critical factors to determine IgG3 cryoglobulin activity, along were observed in patients with severe visceral vasculitis (Kaplanski
with amino acid sequences of the IgG3 variable regions. Otani et  al. 2002). Patients with HCV-related CV were subsequently
et al. (2012) reported that nephritogenicity of IgG3 cryoglobulins found to have significantly increased serum levels of BAFF (Toubi
is determined by sialylation and Wang et al. (2012) demonstrated et  al. 2006; Fabris et  al. 2007; Sène et  al. 2007), B-cell-attracting
that monoclonal cryoglobulins from patients with multiple mye- chemokine-1 (BCA-1, CXCL13), with the highest levels strongly
loma can form crystals and that crystallization can occur at quite associated with active cutaneous vasculitis (Sansonno et al. 2008),
low cryoglobulin concentrations. The morphology of cryoprecipi- and CCL2 and CXCL10 chemokines, which suggested a preva-
tates and kinetics of their formation are strongly associated with the lence of the Th1 immune response in the acute phase of the disease
supersaturation of cryoglobulins. (Antonelli et al. 2008; Antonelli et al. 2009). Boyer et al. (2004) also
reported a defect in CD4+/CD25+ regulatory T cells.
Mixed cryoglobulins Vascular occlusion, due to cryoglobulin precipitation in the
In MC, cryoprecipitation depends mainly on the IgM fraction microcirculation, is responsible of cold-induced acral necrosis.
(Williams and Malone 1994; Schott et al. 1998; Sutton et al. 2000). Often related to the so-called hyperviscosity syndrome, it is gener-
Lower temperatures increase the number of binding sites of the ally due to high cryoglobulin levels. For this reason, it is frequently
RF complexes (Brandau et al. 1986). In HCV-related CV, IgM RF observed in type I cryoglobulinaemia.
does not react with the virus, but is responsible for the second-
ary reaction inducing cryoprecipitation of the viral RNA or solu-
ble HCV proteins found in plasma, which have formed IC with
Clinical aspects of
specific anti-HCV antibodies (Sansonno et al. 2003b; Sansonno cryoglobulinaemic vasculitis
and Dammacco 2005).The occupancy of the Fc portion of IgG The manifestations of CV are affected by age, cryocrit, type of
by IgM RF is probably a key factor in impairing the clearance of cryoglobulin, and associated diseases. The triad of symptoms first
CMC. After the interaction of IgM and IgG, the solubility of the described by Meltzer and Franklin (1966) is considered to be the
immune complexes is significantly less than that of their indi- classical presentation of the disease (purpura, asthenia, and arthral-
vidual components. Thus, the precipitation of mixed cryoglobu- gia), and has been reported in up to or more than 80% of cases at
lins is influenced more by the size of the complexes than by small disease onset (Ferri et al. 2004; Trejo et al. 2001); however, asthenia
increases in the binding constant of an IgM cryoglobulin at lower and arthralgia are less specific symptoms than purpura, which is
temperatures. the typical clinical marker of the syndrome. Moreover, asthenia has
Occasionally, IgG in cryoprecipitates belong to a specific iso- been variably evaluated and reported in different studies (Poynard
type, but cryoprecipitates generally contain polyclonal IgG. In the et al. 2002), and arthralgia is less frequently observed than purpura.
cryoprecipitates of patients with hepatitis C, HCV RNA and spe- Flares of CV can be accompanied by fever. In some patients the
cific anti-HCV antibodies enriched compared to the supernatant, onset of the disease manifests as a fever of unknown origin. Livedo
accompanied by an overwhelming quantity of non-HCV-related reticularis can be seen in some cases. Fibromyalgia and other endo-
polyclonal IgG and antigens. It can be concluded that in MC the crine processes, such as hypothyroidism, have often been described
cryoprecipitate represents a sampling of circulating antigens and in patients with CV (Antonelli et  al. 2004a). Diabetes, which is
antibodies, with a relative prevalence of those related to the current quite common in chronic HCV infection (Mason et al. 1999), is
dominant immune stimulation. also not unusual in CV (Antonelli et al. 2004b).

Vasculitis and tissue injury Purpura


In HCV-associated MC, cryoprecipitates include C1q, and mol- Purpura is the most evident manifestation of CV, and is due to the
ecules of the lectin complement pathway (Ohsawa et  al. 2001). precipitation of immune complexes in the small vessels of the der-
Sansonno and Dammacco (2005) have shown substantially mis and (less frequently) subcutaneous tissue (Figure  40.3). The
enriched C1q protein and C1q binding activity in cryoprecipi- lesions are infiltrating, palpable, polymorphic, and mainly localized
tates, contrasting with a reduction in C3 and C4 proteins. The to the legs (Figure 40.4), although the thighs and less frequently the
large complexes containing HCV core protein can specifically trunk may also be involved. Bullous or vesicular lesions are not com-
bind the C1q receptor expressed by endothelial cells, facilitating mon. Prolonged standing, exercise, chronic venous insufficiency,
554 SECTION 5 vasculitic diseases and syndromes and related disorders

Fig. 40.3 Immune complex deposits at the level of the basal membrane of


dermal vessels. Immunofluorescence staining with anti-IgM antiserum of a
cutaneous purpuric lesion (320 × magnification).

depilation, and low temperatures facilitate the appearance of the


purpuric manifestations. Purpura is generally intermittent, partic-
ularly at the onset of the disease, when it is often the only symptom,
and recovers spontaneously in a short time (1–2 weeks). Recurrent
episodes are frequent and the hallmark of relapsing, long-lasting Fig. 40.5 Recurrent lower limb cryoglobulinaemic purpura and ochraceous
cryoglobulinaemic purpura is an ochraceous pigmentation due to pigmentation due to haemosiderin deposits in the involved areas.
haemosiderin deposits in the involved areas (Figure 40.5).

Cutaneous ulcers are due to coalescence of vasculitic lesions, and usually result from
the involvement of medium-sized vessels (Bryce et al. 2006; Della
Skin ulcers, mainly around the malleoli, are relatively frequent Rossa et al. 2001; Ferri et al. 2004). The occurrence of ulcers is a sig-
and often have a chronic, disabling course (Figure  40.6). Ulcers nificant index of disease activity, a criterion for starting an aggres-
sive treatment, and a significant risk factor for infection, sepsis, and
death (Ramos-Casals et al. 2012).

Fig. 40.6 Malleolar ulcer in a patient with cutaneous type III HCV-related


Fig. 40.4 Lower-limb purpuric lesions in a patient with type II HCV-related cryoglobulinaemia. The surrounding skin shows areas of ochraceous pigmentation
cryoglobulinaemia. due to longstanding cryoglobulinaemic purpura.
CHAPTER 40 cryoglobulinaemic vasculitis 555

Raynaud’s phenomenon found at diagnosis in 40–60% (Beddhu et al. 2002; Monteverde et al.
Raynaud’s phenomenon is observed in 20% of cryoglobulinaemic 1996). Nephrotic or nephritic syndromes at CV diagnosis account
patients and more frequently in females. Sometimes it represents for 21% and 14% of cases, respectively (Roccatello et  al. 2007).
the first clinical manifestation of the syndrome, generally triggered When present at onset, acute nephritic or nephrotic syndromes
by low temperatures. The endothelial damage caused by precipitat- may be rapidly progressive; in a series of 105 cryoglobulinaemic
ing immune complexes is involved in its pathogenesis and facili- glomerulonephritis, 14% progressed to chronic renal failure after a
tates relapses. Raynaud’s phenomenon is particularly frequent in mean follow-up of 6 years (Tarantino et al. 1986). Relapsing renal
MC associated with connective tissue diseases (particularly sclero- impairment is frequent and generally associated with poor progno-
derma), in which its prevalence reaches 37% (Monti et al. 1995a). sis (Invernizzi et al. 1990).
The typical pathological form of cryoglobulinaemic nephropa-
Arthralgias/arthritis thy is membranoproliferative glomerulonephritis (MPGN), which
is characterized by immune complex deposits in the mesangium
Arthralgias usually affect the hands, knees, and wrists. They are and subendothelial space (Figure  40.7). Its particular features
generally intermittent and migratory, without any typical pattern, are:  (1)  abundant endocapillary proliferation with heavy mono-
and not associated with local evidence of inflammation (Della nuclear cell infiltration, mainly represented by monocytes that are
Rossa et al. 2001; Ferri et al. 2004). The synovial membranes do thought to have the function of scavenger cells; (2)  amorphous
not demonstrate pathological changes even in individuals with deposits within the glomerular capillaries that appear to consist of
long-standing pain. IgM, IgG, and C3; and (3) a split capillary basement membrane.
Arthritis is present in about 10% of cases (Monti et al. 1995b), Electron microscopy reveals subendothelial electron-dense depos-
may have an acute or chronic course, and does not generally give its in the glomerular capillary walls, with a finely fibrillar pattern
rise to erosive lesions (Monteverde et al. 1996). identical to that observed in precipitated cryoglobulins in vitro
Myalgias (mainly involving the shoulder girdle) or rheumatic (Monga et al. 1986). More than one-third of the patients under-
polymyalgia-like patterns may be present in a minority of cases going renal biopsy or pathological examination of renal tissue at
(Monteverde et al. 1996). Radiographic evidence of bone erosion autopsy have evidence of vasculitis of the small and medium-sized
is generally lacking and anticitrullinated antibodies are negative, arteries, with fibrinoid necrosis and monocyte infiltration. CMC
unlike in rheumatoid arthritis (Wener et al. 2004). were reported in 50–70% of the HCV-positive patients with MPGN,
with the majority of them also being RF positive and having low
Neuropathy serum C4 levels (Meyers et al. 2003).
Peripheral neuropathy has been found in up to 69% of patients
with CV, with large variations in different studies (Della Rossa et al. Liver disease
2010; Ferri et al. 2004; Sène et al. 2004). It can be the first CV mani- In a meta-analysis of 19 studies, Kayali et al. (2002) concluded that
festation. The most common form is sensory neuropathy (76%), the risk of liver cirrhosis was significantly higher in HCV-positive
followed by sensorimotor polyneuropathy (15%) and mononeuritis patients with MC than those without; however, cryoglobulins were
multiplex (9%) (Gemignani et al. 2005). typed in only 11 of these studies, seven of which reported type III
The most frequent symptoms are paraesthesia, painful dysaes- cryoglobulins in the majority of cases. This finding conflicts with the
thesia, and myalgias in the lower limbs, often worsening at night. results of the largest series of patients with CV and raises questions
The deep tendon reflexes are generally impaired. Cases of monon- on the comparability of the studies included in the meta-analysis.
europathy are rare. Our clinical series did not show any significant differences in liver
Electrophysiological studies have revealed axonal damage attrib- damage or disease progression between the patients with and
utable to epineural vasculitis caused by immune complexes, with
subsequent ischaemic damage and possible alterations in blood
flow (Ferri et al. 1992). Cell-mediated damage is suggested by the
finding of Th1 cytokines and chemokines in the inflammatory vas-
cular lesions (Saadoun 2005). No direct involvement of HCV has
been demonstrated. Peripheral neuropathy may be associated with
autonomic neuropathy (Migliaresi et al. 1999).

Nephropathy
Renal involvement has been reported in approximately 20% of
cases of CV at the time of diagnosis, with prevalence variations
influenced by the patient selection criteria (Della Rossa et al. 2010;
Ferri et al. 2004; Ferri 2008; Monteverde et al. 1996; Roccatello et al.
2007), and is probably more frequent in type II cryoglobulinaemias
(Agnello and De Rosa 2004; Monti et al. 1995a). The clinical picture
ranges from acute nephritic syndrome with haematuria, proteinu-
ria, and altered renal function to a mild and slowly developing form
with urinary abnormalities, observed in the majority of cases. Fig. 40.7 Kidney biopsy in a patient with type II HCV-related cryoglobulinaemia
Hypertension has been described in 42–70% of patients, and showing membranoproliferative glomerulonephritis with mononuclear cell
abnormal serum creatinine concentrations (>1.5 mg/dl) have been infiltration and intraluminal thrombi (PAS, 320 × magnification).
556 SECTION 5 vasculitic diseases and syndromes and related disorders

without cryoglobulinaemia over 6  years’ follow-up (Monteverde CV patients with gastrointestinal involvement did not show poorer
et al. 1996). overall survival than those without (Terrier et al. 2010b).
Although this issue has not been formally investigated in the
context of studies, our experience and the opinions of other experts Central nervous system involvement
suggest that the onset of CV is generally not preceded by clinical Central nervous system involvement has been observed in up to
evidence of acute hepatitis. Given the relatively late onset of CV 6% of patients (Ramos-Casals et al. 2012). Although stroke is the
(the fifth decade of life or later), HCV infection may have been pre- most frequently reported event, diffuse cerebral involvement has
sent for many years in most patients, and this should be considered been described in some patients (Retamozo et al. 2011), together
when selecting control groups for clinical studies. An independent with isolated cases of recurrent transverse myelitis and ischaemic
association between cryoglobulins and steatosis as well as advanced spinal cord disorders (Morgello 2005).
fibrosis was reported by Saadoun et al. (2006b). Liver cancer is the
second most frequently diagnosed neoplasia in cryoglobulinae- Pulmonary involvement
mic patients after non-Hodgkin’s lymphoma, and is attributable to Interstitial lung disease and pulmonary alveolitis, that in few
the close association of CV with HCV (Ferri et al. 2004; Saccardo patients manifest as acute alveolar haemorrhage, are the pulmonary
et al. 2007). disorders most frequently observed in CV, affecting about 5% of
The liver is clearly involved in the clonal B-cell expansion patients (Amital et al. 2005; Ramos-Casals et al. 2005; Retamozo
observed in CV, as lymphocytic infiltrates with a pseudofollicular et al. 2011). Patients with interstitial fibrosis present with dyspnoea
appearance are the site of the clonal B-cell proliferation that pro- and dry cough, with a predominance of macrophages in bronchoal-
duces IgMk-RF bearing the Wa cross-reactive idiotype, particularly veolar lavage (Ferri et al. 1997). Rare cases of pleural effusions have
in type II cryoglobulinaemia (Monteverde et al. 1997). been described (Rieu et al. 2002).

Cardiovascular abnormalities Multiorgan failure


Congestive heart failure, particularly in patients with renal impair- A ‘catastrophic’, life-threatening syndrome, involving the kidney,
ment, is the most prevalent cardiovascular manifestation in cryo- lung, central nervous system, and gastrointestinal tract, may occur
globulinaemic patients (Gorevic et al. 1980), and vasculitic lesions in some patients with CV and is sometimes the first manifesta-
of the myocardium have been reported upon post-mortem exami- tion of the disease. On autopsy, these patients show vasculitis of
nation in at least one patient who died of congestive heart disease the cholecystic, pancreatic, gastrointestinal, and coronary vessels
(Saccardo et  al. 1986). Au et  al. (2005) described structural aor- (Au et al. 2005; Ramos-Casals et al. 2006; Saccardo et al. 2007). No
tic abnormalities, including multiple dissections, aneurysms, and specific risk factors for developing acute multiorgan failure have
congenital aortic arch abnormalities in four HCV-negative Chinese been found.
patients with cryoglobulinaemia (three patients had underlying
malignancies, and the fourth was HBV-positive). Although there Non-hodgkin’s lymphoma
are less than 10 reports of myocardial vasculitis in cryoglobuli- HCV infection is associated with an increased risk of NHL (Giordano
naemic patients described in the literature (Ramos-Casals et  al. et  al. 2007). As expected, B-cell lymphoma is the most frequent
2012)  cardiovascular events represent the third leading cause of neoplastic complication of HCV-related CV, reported in 5–22% of
death after liver disease and sepsis (12–17%) in patients with CV patients with MC (Ferri et al. 2004; Geri et al. 2010; Monti et al.
(Saccardo et al. 2007). In fact, GISC data suggest that the contain- 2005; Trejo et al. 2003). Through the analysis of 1255 HCV-positive
ment of catastrophic events and the increased life expectancy due patients with symptomatic cryoglobulinaemia followed up for
to therapy on one hand and the high frequency of risk factors, 8928 person-years, Monti et al. (2005) found 59 incident cases of
such as renal impairment and diabetes, on the other, have resulted NHL, only 20 of which had the histological characteristics of highly
in increased mortality rates due to cardiovascular disease in CV malignant lymphomas. The overall risk for developing aggres-
patients. sive NHL was about 12 times higher than in the general popula-
tion, with a cumulative incidence of 1.6%; more than 90% of the
Gastrointestinal abnormalities patients who developed NHL had type II cryoglobulins. The over-
Abdominal pain, which is sometimes severe, is described in about all median time between the diagnoses of CV and aggressive NHL
5–20% of patients and vasculitic gastrointestinal involvement, was 3.5 years (range 0.5–23 years). MALT lymphomas were also
sometimes requiring surgical procedures, is reported in up to 6% of more frequent than in the general population. Lymphoplasmocytic
cases (Della Rossa et al. 2010; Monteverde et al. 1996; Quartuccio proliferation exclusively involving bone marrow, as reported in the
et al. 2010; Ramos-Casals et al. 2005; Terrier et al. 2010b). At the study by Monti et al. (2005) and in other studies, more probably
onset, gastrointestinal vasculitis causes acute abdominal pain represents the CV-associated monoclonal lymphoproliferation
and general malaise, accompanied by fever and bloody stools in described in Section Pathogenesis (Figure 40.2a) rather than true
one-third of patients. CV involving the biliary tract can mimic chol- lymphomas, although the possibility of transitional forms evolving
ecystitis or pancreatitis (Terrier et al. 2010b). Some patients present into overt lymphoma cannot be excluded.
with intestinal perforation and shock. Intestinal ischaemia should Both CV and NHL are probably multiple-step, antigen-driven
be suspected in patients presenting with severe abdominal pain. lymphoproliferative disorders, as suggested in HCV-related lym-
Mortality rates associated with intestinal ischaemia are very high phoproliferation by the IgH ongoing mutation process and the
(>80%) (Ramos-Casals et al. 2006), and intestinal involvement in expression of an Ig antigen receptor significantly homologous to
CV is generally considered high risk for poor outcome; however, in an anti-HCV antibody (De Re et al. 2000b). Similar to Helicobacter
the only large study specifically addressing this topic, HCV-related pylori-related lymphomagenesis (Sagaert et al. 2010), independence
CHAPTER 40 cryoglobulinaemic vasculitis 557

from the antigen-driven mechanism may progressively develop,


possibly due to the occurrence of chromosomal translocations or
other genetic aberrations (Zignego et al. 2000; Zignego et al. 2012).
Hypogammaglobulinaemia has been shown to be a marker
for impending lymphomagenesis (Geri et  al. 2010)  and has also
been confirmed to be a correlate of increased risk for NHL in
HCV-negative cryoglobulinaemic patients (Saadoun et al. 2006c).
Moreover, two different studies showed that genetic factors may
influence the progression of HCV-related type II cryoglobulinae-
mia to malignant lymphoma. Polymorphisms of the fibronectin
(FN) gene, particularly the DD-MspI and the AA-HaeIIIb homozy-
gosis, appeared significantly associated with the development of
B-cell NHL in CV patients. No association between FN plasma lev-
els and FN genotypes was found (Fabris et al. 2008).
There are no data from trials on the treatment of lymphomas in
CV patients, who usually receive standard chemotherapy regimens
and rituximab.

Hyperviscosity syndrome Fig. 40.8 Necrosis of the finger in a patient with type I cryoglobulinaemia and
hyperviscosity syndrome.
Hyperviscosity syndrome is frequent in type I monoclonal cryo-
globulinaemias, in which the amount of circulating cryoglobulins
is often high, but is relatively rare in MC, where it is reported in (i.e. at diagnosis or during follow-up). In the GISC series patients
less than 3% of cases (Della Rossa et  al. 2010; Ferri et  al. 2004). with CV associated with autoimmune diseases presented less fre-
Symptoms usually occur at viscosity measurements over 4.0 cen- quently with purpura or the complete triad of Meltzer and Franklin
tipoise (Treon 2009)  and include headache, confusion, blurry when compared with patients with CV associated with B-cell lym-
vision, visual and hearing loss, and epistaxis. Acute or rapidly pro- phomas and Sjögren’s disease or classified as ‘essential’. On the other
gressive renal failure, due to massive intratubular cryoprecipitation, hand, apart from the cases associated with autoimmune diseases,
has been reported. Ischaemia in the distal regions (fingers, lips, the other non-HCV-related CV cases had a clinical picture very
ears, nose, and, sometimes, perimalleolar regions), due to cryopre- similar to that observed in HCV-related CV.
cipitation in small vessels, can cause extended necrotic phenomena
(Figure 40.8). The role of hyperviscosity syndrome in increasing Diagnosis
cardiovascular risk of cryoglobulinaemic patients still needs to be
assessed. Funduscopy to rule out hyperviscosity-related retinal Laboratory diagnosis
changes, especially haemorrhages, should be included in the moni- The clinical diagnosis of CV must be confirmed by the detection of
toring of patients with CV (Ramos-Casals et al. 2012). Symptomatic cryoglobulins in the peripheral blood, but a negative finding may
hyperviscosity should be urgently treated with plasma exchange or not be sufficient to exclude the presence of CV. Because the proce-
other apheresis procedures. dures of drawing and handling of blood samples are crucial, even
minor deviations may strongly affect the reliability of the result
Clinical features of non-HCV-related CV (Bakker et  al. 2003). To obtain cryoprecipitates, patients should
In type I cryoglobulinaemia, the large amount of circulating mono- be acclimatized to room temperature (20–25°C) for 15–30 min-
clonal immunoglobulins can cause a hyperviscosity syndrome and utes, and at least 15 ml of blood should be collected in prewarmed
cutaneous manifestations, such as purpura, ulcers, and, sometimes, syringes and tubes and immediately incubated at 37°C for 2 hours.
extended necrotic phenomena in the distal body regions more fre- It is crucial to ensure that the temperature of the tubes never falls
quently than in MC, as already reported in this Section. A retro- below 37°C during transport to the laboratory. The clotted samples
spective study (Terrier et al. 2013) confirmed more frequent severe should then be centrifuged twice at 1500 rpm for 3 minutes at 37°C,
cutaneous involvement (i.e. necrosis and ulcers) in type I  CV. and the serum cooled in a graduated tube at 4°C for 7 days before
Interestingly, B-cell malignancies were diagnosed in only approxi- being centrifuged at 4°C for 15 minutes at 1700 rpm. Because type
mately half of the patients, while the others presented benign lym- I cryoprecipitate may appear as early as 24 hours, but mixed cryo-
phoproliferation, that is monoclonal gammopathy of unknown globulins may precipitate after several days, the samples should be
significance (MGUS). considered negative only after 7 days of observation (Motyckova
The true percentage of non-HCV-related cases in the total and Murali 2011). Due to the possibility of false-negative results
numbers of CV observed is not yet well assessed, but probably caused by improper handling of the sample or fluctuation of CMC
ranges between 1 and 5%. Clinical and laboratory characteris- concentration, serial determinations should be recommended
tics of HCV-negative symptomatic MC have been investigated before excluding CV when the clinical picture is highly suggestive
by the French CryoVas survey (Terrier et al. 2012) and the GISC. of the disease (Bakker et al. 2003; Sargur et al. 2010). On the other
Table 40.2 shows a comparison of the symptoms in HCV-negative hand, the presence of CMC, particularly at low concentrations in
and HCV-positive CV as reported in the different studies. Some asymptomatic patients, does not allow a diagnosis of CV.
of the differences observed may depend on the different composi- The cryocrit is determined as the percentage of total serum vol-
tion of case studies or the inclusion of data at different time points ume and represents the reference test for monitoring the response
558 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 40.2 Clinical features in patients with cryoglobulinaemic vasculitis

Type I Mixed, HCV-negative Mixed, HCV-positive

Terrier et al. 2013 Huart et al. 2012 GISC Studya Terrier and Ferri C et al. 2004b
Authors
Cacoub 2013
Number of patients 64 242 125 165 231
Mean age (years) 65 63 55c 60 56
Women (%) 56 69 82 54 66
Purpura (%) 69 75 55 71 81
Arthralgia/ arthritis (%) 28 40 67 53 72
Raynaud’s phenomenon (%) 30 26 30 – 36
Ulcers (%) 27 16 13 4 11
Livedo (%) 13 2 – 4 –
Renal abnormalities (%) 30 35 21 34 20
GI abnormalities (%) – 5 0 7 –
Peripheral neuropathy (%) 44 52 41 74 58
a GISC study (Galli et al. unpublished data)
b 92% anti-HCV-positive at the last follow-up.
c median.

to treatment. Cryocrit levels generally correlate with the severity of 2011). In the first stage, a questionnaire was validated to identify
symptoms, although in MC it is not uncommon to observe serious the screening queries with the highest sensitivity and specificity
clinical pictures at low cryocrit levels. Cryoglobulin concentrations for CV. In the second stage, it was used a standard methodology
can also be assayed indirectly by measurements of the total pro- for classification studies, also including the questions previously
tein concentrations within the cryoprecipitates. To characterize the selected. Table  40.3 reports the identified classification criteria
cryoglobulins, the cryoprecipitates should be resuspended in three for CV, the performance of which was also assayed by comparing
volumes of icy saline and centrifuged three times at 3500 rpm for HCV-positive and HCV-negative patients with CMC (Quartuccio
10 minutes at 4°C (discarding the supernatant after each washing). et al. 2012).
The cryoprecipitates should then be dissolved by incubation in
three volumes of saline at 37°C for 1 hour (with periodic shaking). Differential diagnosis
Immunofixation is currently considered the technique of choice for The differential diagnosis of CV includes diseases associated
typing cryoprecipitates. The samples are placed on thin-layer aga- with leukocytoclastic vasculitis, such as IgA vasculitis (Henoch–
rose gels which, after electrophoretic migration and the antiserum Schönlein purpura), polyarteritis nodosa, and microscopic poly-
reaction, are washed three times at 37°C in a 10% NaCl solution angiitis. RF positivity implies the differential diagnosis with
before the plates are stained. A more sensitive technique is immu- rheumatoid arthritis, which is quite easy when considering the
noblotting, which allows the typing of low-concentration (10–80 peculiar features of CV, such as the presence of a cryoprecipitate,
mg/l) cryoprecipitates. HCV-positivity, low C4 levels, and palpable purpura. CV can be
Low C4 levels and raised titres of serum RF are the most impor- associated with autoimmune diseases, including Sjögren’s syn-
tant laboratory hallmarks of CV, besides the presence of CMC, drome and systemic lupus erythematosus among the most frequent.
and can be useful in monitoring the disease activity and response
to treatment. HCV testing (antibodies and serum HCV RNA) is
mandatory (Ramos-Casals et al. 2012), and testing for other viruses Prognosis
(HBV, HIV) and autoimmune diseases (antinuclear, anti-DNA, The assessment of the prognosis of CV is affected by the differ-
anti-Ro/La, anticyclic citrullinated peptide antibodies) is also ences in the composition of case studies and diagnostic criteria. In
recommended, even in patients known to have HCV (Sargur an early large case series, more than 70% of the patients were alive
et al. 2010). 10 or more years after the diagnosis (Monteverde et al. 1996). In
another Italian series, in which more than 90% were HCV-positive,
Diagnostic criteria one-third of patients had moderate-to-severe disease, with chronic
Although the diagnosis of CV is relatively easy in patients present- renal failure or cirrhosis, and nearly 15% presented with sudden
ing with all the hallmarks of the diseases, the lack of validated clas- life-threatening disease (Ferri et  al. 2004). The 10-year survival
sification criteria has caused several problems in clinical practice rates in patients with glomerulonephritis is reported to be 33–49%
and in the comparison of the results of different studies. The GISC (Ferri et  al. 2004; Ramos-Casals et  al. 2006), although improve-
has completed a large co-operative international two-stage study ment in therapeutic management may account for the increased
for the definition of classification criteria for CV (De Vita et  al. survival rate up to the 80% reported in another study (Roccatello
CHAPTER 40 cryoglobulinaemic vasculitis 559

Table 40.3 Classification criteria for cryoglobulinaemic vasculitis and progression of liver disease, with the contemporaneous pres-
ence of renal and liver impairment associated with the worst out-
Satisfied if at least two of the three items (questionnaire, clinical, laboratory ) come. Male sex, age more than 60 years, gastrointestinal, renal, or
are positive. pulmonary involvement, and type II cryoglobulinaemia were also
The patient must be positive for serum cryos in at least 2 determinations at reported as negative prognostic factors (Della Rossa et  al. 2010;
≥12-week interval Ferri et al. 2004; Monti et al. 1995b; Ramos-Casals et al. 2006; Huart
et al. 2012). Peripheral neuropathy impairs the patient’s quality of
(1) Questionnaire item: at least two out of the following:
life, but its role as a predictor of a poor outcome is unclear.
Do you remember one or more episodes of small red spots on your skin,
particularly involving the lower limbs?
At present, the leading causes of death are liver impairment (i.e.
end-stage liver disease and/or hepatocellular carcinoma) in patients
Have you ever had red spots on your lower extremities which leave a
brownish colour after their disappearance? with HCV-related MC (Saccardo et al. 2007; Terrier et al. 2011) and
serious infections, mainly bacterial, in patients with non-infectious
Has a doctor ever told you that you have viral hepatitis?
MC (Huart et al. 2012). Lymphomas are a further important cause
(2) Clinical item : at least three out of the following four (present or past): of death (Saccardo et al. 2007).
Constitutional symptoms The outcome of type I CV has been assessed by Terrier et al.
Fatigue (2013). In this study, 1-year, 2-year, 5-year, and 10-year overall
Low grade fever (37–37.9°C, >10 days, no cause) survival rates were reported to be 97, 94, 94 and 87%, respec-
Fever (>38°C, no cause) tively. Therefore, the long-term survival seems to be better
Fibromyalgia than that in mixed CV, although malignant haemopathy was
Articular involvement observed in 56% of the patients included in the study. As found
Arthralgias in non-HCV-related mixed CV, severe infections represented the
Arthritis main cause of death.
Vascular involvement
Purpura Treatment
Skin ulcers
CV has long been considered an immune disorder requiring immu-
Necrotizing vasculitis
nosuppressive treatment, but the discovery of the aetiological role
Hyperviscosity syndrome of HCV, increasing knowledge of the pathogenesis of the disease,
Raynaud’s phenomenon and the availability of new drugs have radically changed therapeu-
Neurologic involvement tic strategies. Based on the best available evidence from clinical tri-
Peripheral neuropathy als and expert opinion, any rational therapeutic approach in CV
Cranial nerve involvement should have three main objectives: to remove the causes (HCV or
Vasculitic CNS involvement other underlying diseases); to block the pathogenetic mechanisms
(3) Laboratory item : at least two out of the following three (present): (suppressing B-lymphocyte proliferation); and to reduce the symp-
toms, including pain.
Reduced serum C4 Because the majority of studies have been carried out in
Positive serum rheumatoid factor HCV-related cryoglobulinaemias, the therapeutic suggestions
Positive serum M component reported in this section refer to these, unless otherwise indicated.

Interferon and antiviral drugs


The treatment of patients with HCV-related CV has followed the
et  al. 2007). In HCV-positive patients, the 1-year, 3-year, 5-year, evolution of chronic hepatitis C treatment, although interferon-α
and 10-year survival rates were reported to be 96, 86, 75, and 63%, (IFN-α) as an antiproliferative agent was used to treat MC even
respectively, and the main factors associated with a poor prog- before the identification of HCV (Bonomo et al. 1987).
nosis were the presence of severe liver fibrosis and heart involve- The combination of pegylated IFN (Peg-IFN) and ribavirin (RBV)
ment at CV diagnosis (Terrier et al. 2011). A study by Huart et al. is currently the standard of care for HCV treatment and leads to
(2012) found similar survival rates in patients with HCV-positive 41–80% sustained virological responses (SVRs), according to viral
and HCV-negative MC. It can therefore be concluded that the genotype (Ghany et al. 2009). It has been shown that HCV RNA
clinical course of the disease is relatively benign or at least slowly clearance following treatment with Peg-IFN plus RBV is associated
evolving in the majority of patients fully meeting the diagnostic with the disappearance of the clinical and laboratory manifesta-
criteria for CV and is rapidly evolving or devastating in a minor- tions of the disease in the majority of patients with HCV-related
ity of cases. However, the outcome is strongly conditioned by the CV (Mazzaro et al. 2005; Cacoub et al. 2005). Furthermore, in a
co-morbidities, and the 10-year survival rate of patients with cryo- large retrospective study, antiviral treatment was associated with a
globulinaemia is lower than in the general population (Della Rossa good prognosis, unlike immunosuppressive treatment, even after
et al. 2010; Ferri et al. 2004). adjustment for the severity of vasculitis (Terrier et al. 2011). Despite
No prospective investigations have sought to define prognostic this, an Italian open-label study (Mazzaro et al. 2011) determined
criteria. On the basis of retrospective study results, the main risk that, even if most patients (88.5%) achieved a complete and persis-
factors for poor outcome are the severity and frequency of purpura tent recovery from CV clinical symptoms after antiviral treatment,
relapses, the presence of cutaneous ulcers or MPGN, the severity SVR rate was lower than that observed in HCV-infected patients
560 SECTION 5 vasculitic diseases and syndromes and related disorders

without CV. The presence of co-morbidities and the older age of CV RTX has also been studied in combination with antiviral therapy
patients may explain these results, even if another study performed in HCV-related CV, with encouraging results (Saadoun et al. 2010;
in China also found that MC is an independent factor negatively Dammacco et al. 2010).
associated with SVR (Fan et al. 2012). Randomized controlled tri- The use of RTX is currently recommended in patients with seri-
als with adequate statistical power and an appropriate follow-up ous CV complications. RTX may induce severe reactivation of HBV
are needed to clarify whether better SVR rates can be obtained infection; therefore, irrespective of the presence of HBV DNA, it
by extending treatment duration in virological non-responders should only be used in HBsAg-positive patients when strictly
who show clinical and laboratory improvements after a standard needed, and in combination with antiviral therapy. The same pol-
course of Peg-IFN plus RBV. However, the recent advent of the new icy should be adopted in the case of potential occult HBV carri-
direct-acting antiviral agents, such as HCV protease inhibitors, will ers (HBsAg-negative/anti-HBc-positive patients) (Pietrogrande
probably open up new opportunities for the treatment of CV, as et al. 2011).
suggested by anecdotal reports and preliminary results, and a radi-
cal change in HCV treatment approaches should be expected in the Interleukin-2
near future. The effects of the administration of low-dose interleukin-2
An attempt at viral eradication should be considered the first (1.5 million IU per day for 5 days, followed by three 5-day courses
therapeutic option in patients with mild–moderate HCV-related of 3 million IU per day at weeks 3, 6, and 9) was investigated in
CV; however, taking into account the slow and uncertain response a prospective open-label, phase 1–phase 2a study in ten patients
to antiviral therapy, severe and rapidly progressive CV complica- with HCV-related CV who were refractory to conventional antivi-
tions require prompter and more aggressive treatment; in such ral therapy, RTX therapy, or both, and who were not receiving glu-
settings, antiviral therapy may be used after or, when possible, cocorticoid or immunosuppressant therapy (Saadoun et al. 2011).
concomitantly with more rapid, immunosuppressive regimens. Recovery of regulatory T-cell levels was observed in all ten cases, a
Moreover, it should be remembered that some vasculitic mani- reduction in cryoglobulinaemia in nine patients, and improvement
festations (including peripheral neuropathy and skin ulcers) of vasculitis in eight of ten patients. No significant side-effects were
may be worsened by IFN-based antiviral therapy (Pietrogrande reported, but long-term outcome data on these patients are not yet
et al. 2011). available.
Finally, it is important to note that some patients occasionally
have persistence of cryoglobulins with or without CV symptoms Cytotoxic agents
even after the clearance of HCV RNA (Landau et al. 2008; Levine Cyclophosphamide is the only cytotoxic agent whose use in CV is
et al. 2005). These observations further support the hypothesis that supported by sufficient literature data (L’Abbate et al. 1985; Frankel
some cryoglobulin-producing cell clones can proliferate indepen- et  al. 1992). It is usually used in combination with apheresis in
dently or even in the absence of the antigenic stimulus that origi- the case of serious CV complications, when other therapeutic
nally triggered their proliferation. approaches fail or are contraindicated (Pietrogrande et al. 2011).
Cyclophosphamide may increase plasma HCV RNA levels and
Rituximab cause liver dysfunction.
Rituximab (RTX) is an anti-CD20 monoclonal antibody that has
proved to be beneficial in various clinical manifestations of CV, Plasmapheresis
including fatigue, arthralgias, skin lesions, glomerulonephritis Plasmapheresis is postulated to rapidly reduce the levels of circu-
(in about 90% of cases), peripheral neuropathy (in about 75% of lating immune complexes and inflammation mediators, modify
cases), hyperviscosity syndrome, and gastrointestinal vasculitis the quality of the immune complexes and their solubilization, and
(Zaja et al. 2003; Sansonno et al. 2003a; Quartuccio et al. 2010; Ferri restore reticuloendothelial system function (Tavoni et al. 1995). It
et al. 2011; Sneller et al. 2012); in such settings, RTX was shown remains the treatment of choice for life-threatening hyperviscosity
to be more effective than conventional immunosuppressive treat- syndrome, but is also indicated in patients with severe cryoglob-
ments (De Vita et al. 2012). Clinical responses occur within a few ulinaemic manifestations unresponsive to, or ineligible for, other
months after the end of RTX treatment (375 mg/m2 in four weekly therapies. In uncontrolled studies of small series of patients, plas-
infusions), and are long term (more than 1 year) in the majority mapheresis, alone or in combination with cytotoxic drugs, gluco-
of patients. A study has evaluated the efficacy of a lower dosage of corticoids, or intravenous high-dose immunoglobulins, has proved
RTX (250 mg/m2 given twice, at 1-week interval), obtaining a 79% to be effective in improving acute renal disease, neuritis, and ulcers
overall response rate and a mean time to relapse comparable with (Geltner et al. 1981; L’Abbate et al. 1985; Ferri et al. 1986; Frankel
the studies with high-dose RTX; in addition, side-effects were simi- et al. 1992; Pietrogrande et al. 1999). There is some evidence that
lar to those seen in patients treated with high-dose RTX (Visentini plasmapheresis synchronized with the intravenous administra-
et al. 2011). tion of high-dose immunoglobulins can be used to treat ulcers and
It has also been observed that RTX activity is associated with CV-related peripheral neuropathy (Ramunni et al. 2008), but this
serum cryoglobulin and RF level decrease, C4 level increase, the may also have a considerable immunosuppressive effect.
disappearance of bone marrow B-cell clonal expansion (Quartuccio Plasma exchange and double filtration are now considered
et al. 2008), and normalization of the size of all B-cell subsets and the best apheretic approaches, with many experts preferring
the T1/T2-ratio (Holz et al. 2012). In HCV-related CV patients, the plasma exchange in the case of life-threatening complications
response to RTX was related to F allele homozygosity of FCGR3A, (Pietrogrande et al. 2011). Cryoglobulin production may increase
a polymorphism of the low-affinity Fcγ receptor (Gragnani after cessation of apheresis, and immunosuppressive drugs (usually
et al. 2011). cyclophosphamide for up to 6 weeks) are added with the aim of
CHAPTER 40 cryoglobulinaemic vasculitis 561

partially abolishing the postpheresis rebound of cryoglobulins and the second includes serotonin-norepinephrine reuptake inhibitors,
immune complexes (Ramos-Casals et al. 2012). such as duloxetine and venlafaxine; and the third comprises opi-
oids, analgesics, or μ-opioid receptor agonists, such as tramadol and
Glucocorticoids tapentadol (Scarpato et al. unpublished data). Clinical conditions
Unlike in the past, glucocorticoids (GCs) have limited use in CV or co-morbidities must always be considered before starting these
treatment today. Among experts there is general agreement on the therapies. Caution is recommended when NSAIDs are administered
use of high-dose (1–10 mg/kg) pulse GC therapy in critical con- to patients with liver or kidney impairment.
ditions, such as renal, neurological, or hyperviscosity syndromes,
alone or in combination with other drugs (De Vecchi et al. 1983; Treatment of non-HCV-related CV
Auzerie et al. 2003). On the contrary, the effectiveness of long-term The treatment of non-HCV-related CV is fundamentally that of
administration of low–medium GC doses (0.1–0.5 mg/kg/day) is the underlying disease. In essential CV, therapy should aim to
questioned and there is some evidence from controlled studies improve the clinical manifestations and limit the proliferation of
against its use (Pietrogrande et al. 2011). Moreover, chronic therapy cryoglobulin-producing B-cell clones; however, data on treat-
with GCs should also be discouraged for its side-effects, which can ment of HCV-negative cryoglobulinaemias are very limited and
be serious and irreversible. derive from case reports and retrospective surveys. RTX seemed
to be highly effective in 23 patients with non-viral CV included
Colchicine in the French AutoImmunity and Rituximab registry, but relevant
The rationale underlying the use of colchicine to treat CV is based side-effects were seen in almost half of patients, including severe
on the drug’s activity in reducing Ig secretion (Invernizzi and Monti infections in 26%, with a rate of 14.1 per 100 patient-years (com-
1993). Despite the limited available data and the absence of con- pared with 5.0 per 100 patient-years in rheumatoid arthritis patients
trolled trials supporting its use, colchicine is quite widely used, at in the same registry) (Terrier et al. 2010a). A retrospective survey
least in Italy (Pietrogrande et al. 2011), in patients with mild–mod- of 242 cases revealed that RTX and GCs showed greater therapeutic
erate CV who have failed or cannot receive antiviral or RTX treat- efficacy compared with GCs alone and alkylating agents plus GCs
ment. An uncontrolled retrospective study of 17 patients treated to achieve complete clinical, renal, and immunological responses
with colchicine 1 mg/day for 6–48  months showed that it had and a prednisone dosage <10 mg/day at 6 months; however, this
favourable effects on purpura, weakness, leg ulcers, and CV-related regimen was also associated with severe infections, particularly
laboratory abnormalities. Gastrointestinal side-effects may occur, when high doses of GCs were used, whereas death rates did not
and prolonged treatment can cause haematological abnormalities differ between the therapeutic regimens (Terrier et al. 2012). It is
(Monti et  al. 1995c). Myotoxicity is a rare adverse effect, mainly conceivable that treatment with RTX, possibly at the dosage used
occurring in patients with renal impairment, in whom dosage in HCV-related CV, is the best approach currently available, and its
adjustment may be needed (Wilbur and Makowsky 2004). use in patients not heavily pretreated with GCs or alkylating drugs
might reduce the risk of infection.
Low-antigen-content diet Rituximab plus fludarabine and cyclophosphamide resulted in
A low-antigen-content (LAC) diet can improve phagocyte activity effective treatment in a small case series of refractory MC associ-
and modify the composition of immune complexes. CV patients ated with lymphoma (Saadoun et al. 2013).
who strictly follow such a diet experience a significant reduction in A flow chart of suggested therapeutic approaches according to
purpura and pain within 4–8 weeks (Ferri et al. 1989; Bombardieri severity and aetiology of CV is shown in Figure 40.9.
and Ferri 1992). LAC diet is safe and inexpensive, and can be con-
sidered as supportive treatment in all CV patients. Its only limita-
Treatment of type I CV
tion is patient compliance, which is often insufficient. In malignant haemopathy-related type I  CV the treatment coin-
cides with that of the underlying haemopathy, sometimes associated
with plasma exchange, particularly in patients with hyperviscosity
Analgesic and non-steroidal syndrome.
anti-inflammatory treatment In type I CV related to monoclonal gammopathy of undetermined
Several clinical manifestations of CV can provoke either nocic- significance, the use of RTX is controversial (Nehme-Schuster
eptive, neuropathic, or mixed pain, which often severely limits the et al. 2005; Pandrangi et al. 2008). In fact, exacerbations of vascu-
patients’ quality of life. Interventions aiming at controlling pain litis and increases in cryoglobulin levels after RTX infusion have
are frequently necessary, but the use of different classes of drugs been observed (Nehme-Schuster et al. 2005), together with lack
is still based on empirical criteria; therefore, the management of of efficacy, which might be due to the absence of CD20 expres-
pain should be individually tailored and based on the drugs that sion on proliferating B cells clones in these patients. The French
have proved to be effective in controlling pain due to other vascu- nationwide CryoVas survey reported a response to RTX in 80%
litides and neuropathies (Pietrogrande et al. 2011). Short courses of of patients on first- or second-line treatment, with vasculitis flares
non-steroidal anti-inflammatory drugs (NSAIDs) or paracetamol within 48 hours following infusion in 13% of cases (Terrier et al.
may be administered to control nociceptive pain, but combined 2013). In the same study a bortezomib-based regimen was shown
analgesic plus opioid treatment may be necessary when pain become to be effective in 86% of patients, while a thalidomide- and/or
severe. The GISC suggests a three-step approach for pain control lenalinomide-based regimen was effective in 83%, with a good
in CV: the first contemplates the use of secondary amine tricyclic tolerance profile for both regimens. These drugs should probably
antidepressants, such as nortriptyline and desipramine, and calcium be avoided in patients with CV-related peripheral neuropathy
channel alpha 2-delta ligands, such as gabapentin and pregabalin; (Terrier and Cacoub 2013).
562 SECTION 5 vasculitic diseases and syndromes and related disorders

ASYMPTOMATIC or MODERATE- MODERATE- RAPIDLY


MILD CV, SEVERE CV, SEVERE CV, PROGRESSIVE CV,
HCV+*/HCV- HCV+ HCV+/HCV- HCV+/-

monitoring Peg-IFN + RBV +/- RTX PE + CPX


DAAS**
or or
colchicine high-dose GCs
and/or
LAC diet

SVR NR or PR and/or NON RESPONSE


PERSISTENT CV

COMPLETE CV RESPONSE RESPONSE


REMISSION

monitoring HCV+CV
Peg-IFN + RBV +/- DAAS**

Fig. 40.9 Flow chart of suggested therapeutic approaches in cryoglobulinaemic vasculitis (CV). HCV +/−, hepatitis C virus positive/ negative; LAC, low-antigen-content;
Peg-IFN, pegylated interferon; RBV, ribavirin; DAA, direct-acting antiviral agent; RTX, rituximab; PE, plasmapheresis; CPX, cyclophosphamide; GCs, glucocorticoids; SVR,
sustained virological response; NR, null (virological) response; PR, partial (virological) response. * Antiviral treatment may also be considered in some cases. ** According
to HCV treatment guidelines.

References Antonelli, A., Ferri, C., Fallahi, P., et  al. (2004a). Thyroid involvement in
patients with overt HCV-related mixed cryoglobulinaemia. Quarterly
Adinolfi, L.E., Utili, R., Attanasio, V., et  al. (1996). Epidemiology, clinical Journal of Medicine, 97, 499–506.
spectrum and prognostic value of mixed cryoglobulinaemia in hepatitis
Antonelli, A., Ferri, C., Fallahi, P., et al. (2004b). Type 2 diabetes in hepatitis
C virus patients: a prospective study. Italian Journal of Gastroenterology,
C-related mixed cryoglobulinaemia patients. Rheumatology (Oxford),
28, 1–9.
43, 238–40.
Agnello, V. (1997). The etiology and pathophysiology of mixed cryoglobu- Antonelli, A., Ferri, C., Fallahi, P., et al. (2008). High values of CXCL10 serum
linemia secondary to hepatitis C virus infection. Springer Seminars in levels in mixed cryoglobulinemia associated with hepatitis C infection.
Immunopathology, 19, 111–29. American Journal of Gastroenterology, 103, 2488–94.
Agnello, V. and Abel, G. (1997). Localization of hepatitis C virus in cutaneous Antonelli, A., Ferri, C., Fallahi, P., et al. (2009). CXCL10 and CCL2 serum lev-
vasculitic lesions in patients with type II cryoglobulinemia. Arthritis and els in patients with mixed cryoglobulinaemia and hepatitis C. Digestive
Rheumatism, 40, 2007–15. and Liver Disease, 41, 42–8.
Agnello, V., Chung, R.T., and Kaplan, L.M. (1992). A role for hepatitis C virus Au, W.Y., Kwok, J.S., Chu, K.M., and Ma, E.S. (2005). Life-threatening cryo-
infection in type II cryoglobulinemia. New England Journal of Medicine, globulinemia in HCV-negative Southern Chinese and a novel association
327, 1490–5. with structural aortic abnormalities. Annals of Hematology, 84, 95–8.
Agnello, V. and De Rosa, F.G. (2004). Extrahepatic disease manifestations of Auzerie, V., Chiali, A., Bussel, A., et al. (2003). Leg ulcers associated with
HCV infection, some current issues. Journal of Hepatology, 40, 341–52. cryoglobulinemia: clinical study of 15 patients and response to treat-
Agnello, V., Zhang, Q.X., Abel, G., and Knight, G.B. (1995). The associa- ment. Archives of Dermatology, 139, 391–3.
tion of hepatitis C virus infection with monoclonal rheumatoid factors Baimpa, E., Dahabreh, I.J., Voulgarelis, M., and Moutsopoulos, H.M. (2009).
bearing the WA cross-idiotype, implications for the etiopathogenesis Hematologic manifestations and predictors of lymphoma development
and therapy of mixed cryoglobulinemia. Clinical and Experimental in primary Sjögren syndrome:  clinical and pathophysiologic aspects.
Rheumatology, 13 (Suppl. 13), S101–4. Medicine (Baltimore), 88, 284–93.
Allain, J.P., Dong, Y., Vandamme, A.M., Moulton, V., and Salemi, M. (2000). Bakker, A.J., Slomp, J., de Vries, T., et  al. (2003). Adequate sampling in
Evolutionary rate and genetic drift of hepatitis C virus are not correlated cryoglobulinaemia:  recommended warmly. Clinical Chemistry and
with the host immune response: studies of infected donor-recipient clus- Laboratory Medicine, 41, 85–9.
ters. Journal of Virology, 74, 2541–9. Basse, G., Ribes, D., Kamar, N., et al. (2006). Rituximab therapy for mixed
Amital, H., Rubinow, A., and Naparstek Y. (2005). Alveolar hemorrhage cryoglobulinemia in seven renal transplant patients. Transplantation
in cryoglobulinemia:  an indicator of poor prognosis. Clinical and Proceedings, 38, 2308–10.
Experimental Rheumatology, 23, 616–20. Beddhu, S., Bastacky, S., and Johnson, J.P. (2002). The clinical and mor-
Antinori, S., Galimberti, L., Rusconi, S., Zehender, G., Esposito, R., and Galli, phologic spectrum of renal cryoglobulinaemia. Medicine (Baltimore),
M. (1995). Disappearance of cryoglobulins and remission of symptoms 81: 398–409.
in a patient with HCV-associated type II mixed cryoglobulinemia after Bianchettin, G., Bonaccini, C., Oliva, R., et al. (2007). Analysis of hepatitis C
HIV-1 infection. Clinical and Experimental Rheumatology, 13 (Suppl. 13), virus hypervariable region 1 sequence from cryoglobulinemic patients
S157–9. and associated controls. Journal of Virology, 81, 4564–71
CHAPTER 40 cryoglobulinaemic vasculitis 563

Bombardieri, S. and Ferri, C. (1992). Low antigen content diet in the manage- Dammacco, F., Lauletta, G., Montrone, M., and Sansonno, D. (2007). Mixed
ment of immunomediated diseases. Israel Journal of Medical Sciences, cryoglobulinaemia: a model of virus-related disease in internal medi-
28, 117–20. cine. Digestive and Liver Disease, 39 (Suppl. 1), S8–12.
Bonnet, F., Pineau, J.J., Taupin, J.L., et al. (2003). Prevalence of cryoglobuline- Dammacco, F., Sansonno, D., Piccoli, C., Tucci, F.A., and Racanelli, V.
mia and serological markers of autoimmunity in human immunodefi- (2001). The cryoglobulins, an overview. European Journal of Clinical
ciency virus infected individuals: a cross-sectional study of 97 patients. Investigation, 31, 628–38.
Journal of Rheumatology, 30, 2005–10. Dammacco, F., Tucci, F.A., Lauletta, G., et al. (2010). Pegylated interferon- alpha,
Bonomo, L., Casato, M., Afeltra, A., and Caccavo, D. (1987). Treatment of ribavirin, and rituximab combined therapy of hepatitis C virus-related
idiopathic mixed cryoglobulinemia with alpha interferon. American mixed cryoglobulinemia: a long-term study. Blood, 116, 343–53.
Journal of Medicine, 83, 726–30. Della Rossa, A., Tavoni, A., Baldini, C., and Bombardieri, S. (2001). Mixed
Booth, J.C., Kumar, U., Webster, D., Monjardino, J., and Thomas, H.C. (1998). cryoglobulinemia and hepatitis C virus association: ten years later. Israel
Comparison of the rate of sequence variation in the hypervariable region Medical Association Journal, 3, 430–4.
of E2/NS1 region of hepatitis C virus in normal and hypogammaglobu- Della Rossa, A., Tavoni, A., D’Ascanio, A., et al. (2010). Mortality rate and
linemic patients. Hepatology, 27, 223–7. outcome factors in mixed cryoglobulinaemia: the impact of hepatitis C
Boyer, O., Saadoun, D., Abriol, J., et al. (2004). CD4+CD25+ regulatory T-cell virus. Scandinavian Journal of Rheumatology, 39, 167–70.
deficiency in patients with hepatitis C-mixed cryoglobulinemia vasculi- De Re, V., Caggiari, L., De Vita, S., et al. (2007). Genetic insights into the dis-
tis. Blood, 103, 3428–30. ease mechanisms of type II mixed cryoglobulinemia induced by hepati-
Brandau, D.T., Trautman, P.A., Steadman, B.L., Lawson, E.Q., and Middaugh, tis C virus. Digestive and Liver Disease, 39 (Suppl. 1), S65–71.
C.R. (1986). The temperature-dependent stoichiometry of mixed cryo- De Re, V., De Vita, S., Marzotto, A., et al. (2000a). Pre-malignant and malig-
immunoglobulins. Journal of Biological Chemistry, 26, 16385–91. nant lymphoproliferations in an HCV-infected type II mixed cryoglobu-
Breitschwerdt, B., Goeser, T., Tissot, J.D., Trendelenburg, M., Sis-Kluzik, J.M., linemic patient are sequential phases of an antigen-driven pathological
and Andrassy, K. (1999). Hepatitis virus-related and ethanol-induced process. International Journal of Cancer, 87, 211–6.
chronic liver disease with or without cryoglobulins: is there a difference De Re, V., De Vita, S., Marzotto, A., et  al. (2000b). Sequence analysis of
concerning clinical or laboratory manifestation? Infection, 27, 248–51. the immunoglobulin antigen receptor of hepatitis C virus-associated
Brito-Zerón, P., Ramos-Casals, M., Bove, A., Sentis, J., and Font, J. (2007). non-Hodgkin lymphomas suggests that the malignant cells are derived
Predicting adverse outcomes in primary Sjögren’s syndrome: identifica- from the rheumatoid factor-producing cells that occur mainly in type II
tion of prognostic factors. Rheumatology (Oxford), 46, 1359–62. cryoglobulinemia. Blood, 96, 3578–84.
Brouet, J.C., Clauvel, J.P., Danon, F., Klein, M., and Seligmann, M. (1974). De Re, V., De Vita, S., Sansonno, D., et al. (2006a). Type II mixed cryoglob-
Biologic and clinical significance of cryoglobulins. A report of 86 cases. ulinaemia as an oligo rather than a mono B-cell disorder: evidence from
American Journal of Medicine, 57, 775–88. GeneScan and MALDI-TOF analyses. Rheumatology (Oxford), 45, 685–93.
Bryce, A.H., Kyle, R.A., Dispenzieri, A., and Gertz, M.A. (2006). Natural his- De Re, V., Sansonno, D., Simula, M.P., et al. (2006b). HCV-NS3 and IgG-Fc
tory and therapy of 66 patients with mixed cryoglobulinaemia. American crossreactive IgM in patients with type II mixed cryoglobulinemia and
Journal of Hematology, 81, 511–8. B-cell clonal proliferations. Leukemia, 20, 1145–54.
Cacoub, P., Renou, C., Kerr, G., et al. (2001). Influence of HLA-DR pheno- De Vecchi, A., Montagnino, G., Pozzi, C., Tarantino, A., Locatelli, F., and
type on the risk of hepatitis C virus-associated mixed cryoglobulinemia. Ponticelli, C. (1983). Intravenous methylprednisolone pulse therapy
Arthritis and Rheumatism, 44, 2118–24. in essential mixed cryoglobulinemia nephopathy. Clinical Nephrology,
Cacoub, P., Saadoun, D., Limal, N., Sene, D., Lidove, O., and Piette, J.C. (2005). 19, 221–7.
Pegylated interferon alfa-2b and ribavirin treatment in patients with De Vita, S., De Re, V., Gasparotto, D., et al. (2000). Oligoclonal non-neoplastic B
hepatitis C virus-related systemic vasculitis. Arthritis and Rheumatism, cell expansion is the key feature of type II mixed cryoglobulinemia: clinical
52, 911–5. and molecular findings do not support a bone marrow pathologic diag-
Casato, M., de Rosa, F.G., Pucillo, L.P., et al. (1999). Mixed cryoglobuline- nosis of indolent B cell lymphoma. Arthritis and Rheumatism, 43, 94–102.
mia secondary to visceral leishmaniasis. Arthritis and Rheumatism, 42, De Vita, S., Quartuccio, L., and Isola, M. (2012). A randomized controlled
2007–11. trial of rituximab for the treatment of severe cryoglobulinemic vasculi-
Casato, M., Lilli, D., Donato, G., et al. (2003). Occult hepatitis C virus infec- tis. Arthritis and Rheumatism, 64, 843–53.
tion in type II mixed cryoglobulinemia. Journal of Viral Hepatitis, De Vita, S., Soldano, F., Isola, M., et  al. (2011). Preliminary classification
10, 455–9. criteria for the cryoglobulinemic vasculitis. Annals of the Rheumatic
Casato, M., Taliani, G., Pucillo, L.P., Goffredo, F., Lagana, B., and Bonomo, L. Diseases, 70, 1183–90.
(1991). Cryoglobulinaemia and hepatitis C virus. Lancet, 337, 1047–8. Dienstag, J.L., Wands, J.R., and Isselbacher, K.J. (1977). Hepatitis B and
Charles, E.D., Brunetti, C., Marukian, S., et  al. (2011). Clonal B cells in essential mixed cryoglobulinemia. New England Journal of Medicine,
patients with hepatitis C virus–associated mixed cryoglobulinemia con- 297, 946–7.
tain an expanded anergic CD21 low B-cell subset. Blood, 117, 5425–37. Disdier, P., Harle, J.R., and Weiller, P.J. (1991). Cryoglobulinaemia and hepa-
Charles, E.D. and Dustin, L.B. (2009). Hepatitis C virus-induced cryoglobu- titis C infection. Lancet, 338, 1151–2.
linemia. Kidney International, 76, 818–24. Durand, J.M., Lefevre, P., Harle, J.R., Boucrat, J., Vitviski, L., and Soubeyrand,
Charles, E.D., Green, R.M., Marukian, S., et al. (2008). Clonal expansion of J. (1991). Cutaneous vasculitis and cryoglobulinaemia type II associated
immunoglobulin M+CD27+ B cells in HCV-associated mixed cryoglo- with hepatitis C virus infection. Lancet, 337, 499–500.
bulinaemia. Blood, 111, 1344–56. Fabris, M., Quartuccio, L., Sacco, S., et al. (2007). B-Lymphocyte stimula-
Chiche, L., Grados, A., Harlé, J.R., and Cacoub, P. (2010). Mixed cryoglobu- tor (BLyS) up-regulation in mixed cryoglobulinaemia syndrome and
linemia: a role for parvovirus B19 infection. Clinical Infectious Diseases, hepatitis-C virus infection. Rheumatology (Oxford), 46, 37–43.
50, 1074–5. Fabris, M., Quartuccio, L., Salvin, S., et al. (2008). Fibronectin gene poly-
Cholet, F., Nousbaum, J., Richecoeur, M., et  al. (2004). Factors associ- morphisms are associated with the development of B-cell lymphoma
ated with liver steatosis and fibrosis in chronic hepatitis C patients. in type II mixed cryoglobulinemia. Annals of the Rheumatic Diseases,
Gastroentérologie Clinique et Biologique, 28, 272–8. 67, 80–3.
Cornberg, M., Razavi, H.A., Alberti, A., et al. (2011). A systematic review Faguer, S., Kamar, N., Boulestin, A., et al. (2008). Prevalence of cryoglobu-
of hepatitis C virus epidemiology in Europe. Liver International, 31 linemia and autoimmunity markers in renal-transplant patients. Clinical
(Suppl. 2), S30–60. Nephrology, 69, 239–43.
564 SECTION 5 vasculitic diseases and syndromes and related disorders

Fan, X.H., Wu, C.H., Wang, L.F., et al. (2012). Cryoglobulinemia is an inde- Galli, M., Invernizzi, F., Chemotti, M., et al. (1986). Cryoglobulins and infec-
pendent factor negatively associated with sustained virological response tious diseases. La Ricerca in Clinica e Laboratorio, 16, 301–13.
in chronic hepatitis C patients. Chinese Medical Journal, 125, 4014–17. Galli, M., Monti, G., Invernizzi, F., et al. (1992a). Hepatitis B virus-related mark-
Fazi, C., Dagklis, A., Cottini, F., et al. (2010). Monoclonal B cell lymphocyto- ers in secondary and in essential mixed cryoglobulinemias, a multicentric
sis in hepatitis C virus infected individuals. Cytometry. Part B, Clinical study of 596 cases. The Italian Group for the Study of Cryoglobulinemias
Cytometry, 78 (Suppl. 1), S61–8. (GISC). Annali Italiani di Medicina Interna, 7, 209–14.
Ferri, C. (2008). Mixed cryoglobulinemia. Orphanet Journal of Rare Galli, M., Monti, G., Monteverde, A., et al. (1992b). Hepatitis C virus and
Diseases, 3, 25. mixed cryoglobulinaemia. Lancet, 339, 989.
Ferri, C., Antonelli, A., Mascia, M.T., et  al. (2007). HCV-related autoim- Galli, M., Zehender, G., Monti, G., et al. (1995). Hepatitis C virus RNA in
mune and neoplastic disorders: the HCV syndrome. Digestive and Liver the bone marrow of patients with mixed cryoglobulinemia and in sub-
Disease, 39 (Suppl. 1), S13–21. jects with non-cryoglobulinemic chronic hepatitis type C. Journal of
Ferri, C., Cacoub, P., Mazzaro, C., et al. (2011). Treatment with rituximab in Infectious Diseases, 171, 672–5.
patients with mixed cryoglobulinemia syndrome: results of multicenter Gamble, C.N., Kimchi, A., Depner, T.A., and Christensen, D. (1982). Immune
cohort study and review of the literature. Autoimmunity Reviews, 11, 48–55. complex glomerulonephritis and dermal vasculitis following intestinal
Ferri, S., Dal Pero, F., Bortoletto, G., et al. (2006). Detailed analysis of the bypass for morbid obesity. American Journal of Clinical Pathology, 77,
E2-IgM complex in hepatitis C-related type II mixed cryoglobulinae- 347–52.
mia. Journal of Viral Hepatitis, 13, 166–76. García-Carrasco, M., Ramos-Casals, M., Cervera, R., et  al. (2001).
Ferri, C., Greco, F., Longombardo, G., Palla, P., Marzo, E., and Moretti, A. Cryoglobulinaemia in systemic lupus erythematosus:  prevalence and
(1991). Hepatitis C virus antibodies in mixed cryoglobulinemia. Clinical clinical characteristics in a series of 122 patients. Seminars in Arthritis
and Experimental Rheumatology, 9, 95–6. and Rheumatism, 30, 366–73.
Ferri, C., La Civita, L., Cirafisi, C., et al. (1992). Peripheral neuropathy in Garrouste, C., Kamar, N., Boulestin, A., et  al. (2008). Prevalence of cryo-
mixed cryoglobulinemia, clinical and electrophysiologic investigations. globulinemia and autoimmune markers in liver transplant patients.
Journal of Rheumatology, 19, 889–95. Experimental and Clinical Transplantation, 6, 184–9.
Ferri, C., La Civita, L., Fazzi, P., et al. (1997). Interstitial lung fibrosis and Geltner, D., Kohn, R.W., Gorevic, P., and Franklin, E.C. (1981). The effect of
rheumatic disorders in patients with hepatitis C virus infection. British combination therapy (steroids, immunosuppressives, and plasmapher-
Journal of Rheumatology, 36, 360–5. esis) on 5 mixed cryoglobulinemia patients with renal, neurologic, and
Ferri, C., Monti, M., La Civita, L., et al. (1993). Infection of peripheral blood vascular involvement. Arthritis and Rheumatism, 24, 1121–7.
mononuclear cells by hepatitis C virus in mixed cryoglobulinemia. Gemignani, F., Brindani, F., Alfieri, S., et  al. (2005). Clinical spectrum of
Blood, 82, 3701–4. cryoglobulinaemic neuropathy. Journal of Neurology, Neurosurgery, and
Ferri, C., Moriconi, L., Gremignai, G., et al. (1986). Treatment of the renal Psychiatry, 76, 1410–4.
involvement in mixed cryoglobulinemia with prolonged plasma Genet, P., Courdavault, L., Wifaq, B., and Gerbe, J. (2011). Symptomatic
exchange. Nephrology, 43, 246–53. mixed cryoglobulinemia during HIV primary infection: a case report.
Ferri, C., Pietrogrande, M., Cecchetti, R., et al. (1989). Low-antigen-content Case Reports in Infectious Diseases, 2011, 525841.
diet in the treatment of patients with mixed cryoglobulinemia. American Geri, G., Terrier, B., Semoun, O., et al. (2010). Surrogate markers of B cell
Journal of Medicine, 87, 519–24. non-Hodgkin’s lymphoma in patients with hepatitis C virus-related
Ferri, C., Sebastiani, M., Giuggioli, D., et al. (2004). Mixed cryoglobuline- cryoglobulinaemia vasculitis. Annals of the Rheumatic Diseases, 69,
mia: demographic, clinical, and serologic features and survival in 231 2177–80.
patients. Seminars in Arthritis and Rheumatism, 33, 355–74. Gerotto, M., Dal Pero, F., Loffreda, S., Bianchi, F.B., Alberti, A., and Lenzi, M.
Ferri, C., Zignego, A.L., and Pileri, S.A. (2002). Cryoglobulins. Journal of (2001). A 385 insertion in the hypervariable region 1 of hepatitis C virus
Clinical Pathology, 55, 4–13. E2 envelope protein is found in some patients with mixed cryoglobu-
Fiorini, G.F., Sinico, R.A., Winearls, C., Custode, P., De Giuli-Morghen, linemia type 2. Blood, 98, 2657–63.
C., and D’Amico, G. (1988). Persistent Epstein-Barr virus infection Ghany, M.G., Strader, D.B., Thomas, D.L., and Seeff, L.B. (2009). Diagnosis,
in patients with type II essential mixed cryoglobulinemia. Clinical management, and treatment of hepatitis C: an update. Hepatology, 49,
Immunology and Immunopathology, 47, 262–9 1335–74.
Florin-Christensen, A., Roux, M.E., and Arana, R.M. (1974). Cryoglobulins in Giordano, T.P., Henderson, L., Landgren, O., et  al. (2007). Risk of
acute and chronic liver diseases. Clinical and Experimental Immunology, non-Hodgkin lymphoma and lymphoproliferative precursor diseases in
16, 599–605. US veterans with hepatitis C virus. JAMA, 297, 2010–7.
Frankel, A.H., Singer, D.R., Winearls, C.G., Evans, D.J., Rees, A.J., and Pusey, Gorevic, P.D. (2012). Rheumatoid factor, complement, and mixed cryoglobu-
C.D. (1992). Type II essential mixed cryoglobulinaemia, presentation, linemia. Clinical and Developmental Immunology, 2012, 439018.
treatment and outcome in 13 patients. Quarterly Journal of Medicine, Gorevic, P.D., Kassab, H.J., Levo, Y., et  al. (1980). Mixed cryoglobuline-
82, 101–24. mia: clinical aspects and long-term follow-up of 40 patients. American
Gabrielli, A., Manzin, A., Candela, M., et al. (1994). Active hepatitis C virus Journal of Medicine, 69, 287–308.
infection in bone marrow and peripheral blood mononuclear cells from Gorse, A., Boileau, J., Goetz, J., Stoll-Keller, F., and Martin, T. (2011).
patients with mixed cryoglobulinaemia. Clinical and Experimental Parvovirus B19-induced type II mixed cryoglobulinemia. American
Immunology, 97, 87–93. Journal of Medicine, 124, e7–8.
Galli, M. (1991). Cryoglobulinaemia and serological markers of hepatitis Gragnani, L., Piluso, A., Giannini, C., et  al. (2011). Genetic determinants
viruses. Lancet, 338, 758–9. in hepatitis C virus-associated mixed cryoglobulinemia: role of poly-
Galli, M. (1995). Viruses and cryoglobulinemia. Clinical and Experimental morphic variants of BAFF promoter and Fcγ receptors. Arthritis and
Rheumatology, 13 (Suppl. 13), S63–70. Rheumatism, 63, 1446–51.
Galli, M., Caredda, F., D’Arminio Monforte, A., Fiorenza, A.M., Messina, K., Hall, C.G. and Abraham, G.N. (1984). Reversible self-association of a human
and Invernizzi, F. (1982). Mixed cryoglobulinemias and liver diseases. myeloma protein. Thermodynamics and relevance to viscosity effects
Minerva Medica, 73, 1159–60. and solubility. Biochemistry, 23, 5123–9.
Galli, M., Caredda, F., D’Arminio, A., Monti, G., Messina, K., and Invernizzi, Hermida Lazcano, I., Sáez Méndez, L., and Solera Santos, X. (2005). Mixed
F. (1980). Hepatitis B virus and essential mixed cryoglobulinaemia. cryoglobulinemia with renal failure, cutaneous vasculitis and peritonitis
Lancet, 1, 1093. due to Brucella melitensis. Journal of Infection, 51, e257–9.
CHAPTER 40 cryoglobulinaemic vasculitis 565

Holz, LE., Yoon, J.C., Raghuraman, S., Moir, S., Sneller, M.C., and Rehermann, (cold precipitate serum globulin). American Journal of Medical Sciences,
B. (2012). B cell homeostasis in chronic hepatitis C virus-related 214, 410–5.
mixed cryoglobulinemia is maintained through naïve B cell apoptosis. Levine, J.W., Gota, C., Fessler, B.J., Calabrese, L.H., and Cooper, S.M. (2005).
Hepatology, 56, 1602–10. Persistent cryoglobulinemic vasculitis following successful treatment of
Hu, Y.W., Rocheleau, L., Larke, B., et al. (2005). Immunoglobulin mimicry by hepatitis C virus. Journal of Rheumatology, 32, 1164–7.
hepatitis C Virus envelope protein E2. Virology, 332, 538–49. Levo, Y., Gorevic, P.D., Kassab, H.J., Zucker-Franklin, D., and Franklin, E.C.
Huart, A., Bonnet, F., Le Guenno, G., et al. (2012). Prognostic factors of sur- (1977). Association between hepatitis B virus and essential mixed cryo-
vival in patients with non-infectious mixed cryoglobulinaemia vascu- globulinemia. New England Journal of Medicine, 296, 1501–4.
litis: data from 242 cases included in the CryoVas survey. Annals of the Liou, Y.T., Huang, J.L., Ou, L.S., et al. (2013). Comparison of cryoglobuline-
Rheumatic Diseases,72, 374–80. mia in children and adults. Journal of Microbiology, Immunology, and
Ichinose, K., Origuchi, T., Tashiro, N., et  al. (2013). An elderly patient Infection, 46, 59–64.
with chronic active Epstein–Barr virus infection with mixed cryo- Lospalluto, J., Dorward, B., Miller, W. Jr, and Ziff, M. (1962). Cryoglobulinemia
globulinemia and review of the literature. Modern Rheumatology, 23, based on interaction between a gamma macroglobulin and 7S gamma
1022–8. globulin. American Journal of Medicine, 32, 142–7.
Invernizzi, F., Galli, M., Serino, G., et  al. (1983). Secondary and essential Lunel, F., Musset, L., Cacoub, P., et al. (1994). Cryoglobulinemia in chronic
cryoglobulinemias. Frequency, nosological classification, and long-term liver diseases: role of hepatitis C virus and liver damage. Gastroenterology,
follow-up. Acta Haematologica, 70, 73–82. 106, 1291–300.
Invernizzi, F. and Monti, G. (1993) Colchicine and mixed cryoglobulinemia. Madi, N., Steiger, G., Estreicher, J., and Schifferli, J.A. (1991). Defective
Arthritis and Rheumatism, 3, 722–3. immune adherence and elimination of hepatitis B surface antigen/anti-
Invernizzi, F., Monti, G., and Zanussi, C. (1990). The clinical spectrum of body complexes in patients with mixed essential cryoglobulinemia type
mixed cryoglobulinemia. In Immunological aspects of malignant lympho- II. Journal of Immunology, 147, 495–502.
mas and cryoglobulinemia (ed. F. Dammacco), pp. 221–34. Edi-Ermes, Mageed, R.A., Carson, D., and Jefferis, R. (1988). Analysis of rheumatoid fac-
Milano. tor autoantibodies in patients with essential mixed cryoglobulinaemia
Invernizzi, F., Pietrogrande, M., and Sagramoso, B. (1995). Classification and rheumatoid arthritis. Scandinavian Journal of Rheumatology, 75
of the cryoglobulinemic syndrome. Clinical and Experimental (Suppl.), 172–8.
Rheumatology, 13 (Suppl. 13), S123–8. Mason, A.L., Lau, J.Y., Hoang, N., et al. (1999). Association of diabetes mel-
Jardim, D.L., Tsukumo, D.M., Angerami, R.N., Carvalho Filho, M.A., and litus and chronic hepatitis C virus infection. Hepatology, 29, 328–33.
Saad, M.J. (2012). Autoimmune features caused by Dengue fever: a case Mathsson, L., Tejde, A, Carlson, K., et  al. (2005). Cryoglobulin-induced
report. Brazilian Journal of Infectious Diseases, 16, 92–5. cytokine production via FcγRIIa: inverse effects of complement block-
Jori, G.P. and Buonanno, G. (1972). Chronic hepatitis and cirrhosis on the ade on the production of TNF-α and IL-10. Implications for the growth
liver in cryoglobulinemic. Gut, 13, 610–3. of malignant B-cell clones. British Journal of Haematology, 129, 830–8.
Jori, G.P., Buonanno, G., D’Onofrio F., Tirelli, A., Gonnella, F., and Gentile, S. Matsuda, J., Tsukamoto, M., Gohchi, K., Saitoh, N., and Gotoh, M. (1994).
(1977). Incidence and immunochemical features of serum cryoglobulin Hepatitis C virus (HCV) RNA and human immunodeficiency virus
in chronic liver disease. Gut, 18, 245–9. (HIV) p24 antigen in the cryoglobulin of hemophiliacs with HIV and/
Kaplanski, G., Marin, V., Maisonobe, T., et al. (2002). Increased soluble p55 or HCV infection. Clinical Infectious Diseases, 18, 832–3.
and p75 tumour necrosis factor-alpha receptors in patients with hepa- Mazzaro, C., Monti, G., Saccardo, F., et  al. (2011). Efficacy and safety of
titis C-associated mixed cryoglobulinaemia. Clinical and Experimental peginterferon alfa-2b plus ribavirin for HCV-positive mixed cryoglo-
Immunology, 127, 123–30. bulinemia:  a multicentre open-label study. Clinical and Experimental
Kayali, Z., Buckwold, V.E., Zimmerman, B., and Schmidt, W.N. (2002). Rheumatology, 29, 933–41.
Hepatitis C, cryoglobulinemia, and cirrhosis, a meta-analysis. Mazzaro, C., Zorat, F., Caizzi, M., et al. (2005). Treatment with peg-interferon
Hepatology, 36, 978–85. alfa-2b and ribavirin of hepatitis C virus-associated mixed cryoglobu-
Konstantinova, N.A., Yeremin, I.I., Konstantinova, E.V., Karandashov, E.N., and linemia, a pilot study. Journal of Hepatology, 42, 632–8.
Shostak, N.A. (2011). Cryoglobulin composition in patients with myocar- McIntosh, R.M., Koss, M.N., and Gocke, K.J. (1976). The nature and inci-
dial infarction. Bulletin of Experimental Biology and Medicine, 150, 412–5. dence of cryoproteins in hepatitis B antigen (HBsAg) positive patients.
Kosmas, N., Kontos, A., Panayiotakopoulos, G., Dimitrakopoulos, A., and Quarterly Journal of Medicine, 45, 23–38.
Kordossis, T. (2006). Decreased prevalence of mixed cryoglobulinemia Meltzer, M. and Franklin, E.C. (1966). Cryoglobulinemia–a study of
in the HAART era among HIV-positive, HCV-negative patients. Journal twenty-nine patients. I. IgG and IgM cryoglobulins and factors affecting
of Medical Virology, 78, 1257–61. cryoprecipitability. American Journal of Medicine, 40, 828–36.
Kuroda, Y., Kuroki, A., Kikuchi, S., Funase, T., Nakata, M., and Izui, S. (2005) Meyers, C.M., Seeff, L.B., Stehman-Breen, C.O., and Hoofnagle, J.H. (2003).
A critical role for sialylation in cryoglobulin activity of murine IgG3 Hepatitis C and renal disease, an update. American Journal of Kidney
monoclonal antibodies. Journal of Immunology, 175, 1056–61. Diseases, 42, 631–57.
Kuroki, A., Kuroda, Y., Kikuchi, S., et  al. (2002). Level of galactosylation Migliaresi, S., Amendola, A., Di Iorio, G., et al. (1999). Autonomic dysfunc-
determines cryoglobulin activity of murine IgG3 monoclonal rheuma- tion assessed by sympathetic skin response in patients with HCV-related
toid factor. Blood, 99, 2922–8. mixed cryoglobulinemia. Annals of Rheumatic Diseases, 113, 440.
L’Abbate, A., Maggiore, Q., Caccamo, A., et al. (1985). Long term effects of Monga, G., Coppo, R., Piccoli, G., and Coda, R. (1986). Glomerular findings
cryoapheresis and cytostatic treatment in essential mixed cryoglobu- in mixed IgG-IgM cryoglobulinemic. Light, electron microscopi, immu-
linemia. International Journal of Artificial Organs, 8 (Suppl. 2), S19–22. nofluorescence and histochemical correlation. Virchows Archiv. B: Cell
Landau, D.A., Saadoun, D., Calabrese, L.H., and Cacoub, P. (2007). The Pathology, 20, 185–96.
pathophysiology of HCV induced B-cell clonal disorders. Autoimmunity Monteverde, A., Ballare, M., and Pileri, S. (1997). Hepatic lymphoid aggre-
Reviews, 6, 581–7. gates in chronic hepatitis C and mixed cryoglobulinemia. Springer
Landau, D.A., Saadoun, D., Halfon, P., et al. (2008). Relapse of hepatitis C Seminars in Immunopathology, 19, 99–110.
virus-associated mixed cryoglobulinemia vasculitis in patients with sus- Monteverde, A., Invernizzi, F., Pilieri, S., Galli, M., Monti, G., and Ballarè,
tained viral response. Arthritis and Rheumatism, 58, 604–11. M. (1996). Le crioglobulinemie miste. In Atti dei Congressi della Società
Lerner, A.B. and Watson, C.J. (1947). Studies on cryoglobulins. Unusual pur- Italiana di Medicina Interna; 97° Congresso, Venezia, 15–19 ottobre, pp.
pura associated with the presence of a high concentration of cryoglobulin 1–80. Luigi Pozzi Editore, Roma.
566 SECTION 5 vasculitic diseases and syndromes and related disorders

Monteverde, A., Sabattini, E., Poggi, S., et al. (1995). Bone marrow findings Pileri, P., Uematsu, Y., Campagnoli, S., et al. (1998). Binding of hepatitis C
further support the hypothesis that essential mixed cryoglobulinemia virus to CD81. Science, 282, 938–41.
type II is characterized by a monoclonal B-cell proliferation. Leukemia Popp, J.W. Jr, Dienstag, J.L., Wands, J.R., and Bloch, K.J. (1980). Essential
and Lymphoma, 20, 119–24. mixed cryoglobulinemia without evidence for hepatitis B virus infec-
Monti, G., Galli, M., Invernizzi, F., et  al. (1995a). Cryoglobulinaemias, a tion. Annals of Internal Medicine, 92, 379–83.
multi-centre study of the early clinical and laboratory manifestations Poynard, T., Cacoub, P., Ratziu, V., et  al. (2002). Fatigue in patients with
of primary and secondary disease. Quarterly Journal of Medicine, 88, chronic hepatitis C. Journal of Viral Hepatitis, 9, 295–303.
115–26. Quartuccio, L., Fabris, M., Salvin, S., et al. (2007). Bone marrow B-cell clonal
Monti, G., Pioltelli, P., Saccardo, F., et al. (2005). Incidence and character- expansion in type II mixed cryoglobulinaemia: association with nephri-
istics of non-Hodgkin lymphomas in a multicenter casefile of patients tis. Rheumatology (Oxford), 46, 1657–61.
with hepatitis C virus-related symptomatic mixed cryoglobulinemias. Quartuccio, L., Isola, M., Corazza, L., et al. (2012). Performance of the prelim-
Archives of Internal Medicine, 165, 101–5. inary classification criteria for cryoglobulinaemic vasculitis and clinical
Monti, G., Saccardo, F., Pioltelli, P., and Rinaldi, G. (1995b). The natural his- manifestations in hepatitis C virus-unrelated cryoglobulinaemic vascu-
tory of cryoglobulinemia, symptoms at onset and during follow-up. litis. Clinical and Experimental Rheumatology, 30 (1 Suppl. 70), S48–52.
A report by the Italian Group for the Study of Cryoglobulinemias (GISC). Quartuccio, L., Petrarca, A., Mansutti, E., et al. (2010). Efficacy of rituximab in
Clinical and Experimental Rheumatology, 13 (Suppl. 13), S129–33. severe and mild abdominal vasculitis in the course of mixed cryoglobu-
Monti, G., Saccardo, F., Rinaldi, G., Petrozzino, M.R., Gomitoni, A., and linemia. Clinical and Experimental Rheumatology, 28 (Suppl. 57), S84–7.
Invernizzi, F. (1995c). Colchicine in the treatment of mixed cryoglo- Quartuccio, L., Salvin, S., Fabris, M., Sacco, S., and De Vita, S. (2008).
bulinemia. Clinical and Experimental Rheumatology, 13 (Suppl.  13), Disappearance of bone marrow B cell clonal expansion in patients with
S197–9. type II hepatitis C virus-related cryoglobulinemic glomerulonephri-
Morgello, S. (2005). The nervous system and hepatitis C virus. Seminars in tis after clinical efficient rituximab therapy. Annals of the Rheumatic
Liver Disease, 25, 118–21. Diseases, 67, 1494–5.
Motyckova, G. and Murali, M. (2011). Laboratory testing for cryoglobulins. Ramos-Casals, M., Brito-Zerón, P., Nardi, N., et al. (2005). Life-threatening
American Journal of Hematology, 86, 500–2. cryoglobulinemic vasculitis. Clinical presentation and outcome in a
Nehme-Schuster, H., Korganow, A.S., Pasquali, J.L., and Martin, T. (2005). series of 560 patients with cryoglobulinemia. Arthritis and Rheumatism,
Rituximab inefficiency during type I cryoglobulinaemia. Rheumatology 52 (Suppl.), S645–6.
(Oxford), 44, 410–1. Ramos-Casals, M., Robles, A., Brito-Zerón, P., et al. (2006) Life-threatening
Ogata, N., Alter, H.J., Miller, R.H., and Purcell, R.H. (1991). Nucleotide cryoglobulinemia:  clinical and immunological characterization of 29
sequence and mutation rate of the H strain of hepatitis C virus. cases. Seminars in Arthritis and Rheumatism, 36, 189–96.
Proceedings of the National Academy of Sciences of USA, 88, 3392–6. Ramos-Casals, M., Stone, J.H., Cid, M.C., and Bosch, X. (2012). The cryoglo-
Ohsawa, I., Ohi, H., and Tamano, M.(2001). Cryoprecipitate of patients with bulinaemias. Lancet, 379, 348–60.
cryoglobulinemic glomerulonephritis contains molecules of the lectin Ramunni, A., Lauletta, G., Brescia, P., et al. (2008). Double-filtration plasma-
complement pathway. Clinical Immunology, 101, 59–66. pheresis in the treatment of leg ulcers in cryoglobulinemia. Journal of
Ohtsubo, K., Sata, M., Kawaguchi, T., et al. (2009). Characterization of the Clinical Apheresis, 23, 118–22.
light chain-restricted clonal B cells in peripheral blood of HCV-positive Realdi, G., Alberti, A., Rigoli, A., and Tremolada, F. (1974). Immune-complexes
patients. International Journal of Hematology, 89, 452–9. and Australia antigen in cryoglobulinemic sera. Zeitschrift für
Okamoto, H., Kojima, M., Okada, S., et al. (1992). Genetic drift of hepatitis Immunitätsforschung, Experimentelle und Klinische Immunologie, 147,
C virus during an 8.2-year infection in a chimpanzee: variability and 114–26.
stability. Virology, 190, 894–9. Retamozo, S., Diaz-Lagares, C., Bosch, X., De Vita, S., and Ramos-Casals,
Oliver, M., Grandadam, M., and Marimoutou, C. (2009). Persisting mixed M. (2011). Life-threatening cryoglobulinemia. In Autoimmune dis-
cryoglobulinemia in Chikungunya infection. PLoS Neglected Tropical eases:  acute and complex situations (ed. M.A. Khamashta and M.
Diseases, 3, e374. Ramos-Casals), pp. 133–62. Springer-Verlag, London.
Otani, M., Kuroki, A., Kikuchi, S., et al. (2012). Sialylation determines the Rieu, V., Cohen, P., André, M.H., et al. (2002). Characteristics and outcome
nephritogenicity of IgG3 cryoglobulins. Journal of the American Society of 49 patients with symptomatic cryoglobulinaemia. Rheumatology
of Nephrology, 23, 1869–78. (Oxford), 41, 290–300.
Pandrangi, S., Singh, A., Wheeler, D.E., and Rossi, N.F. (2008). Rituximab Rizos, E., Dimos, G., Liberopoulos, E.N., Elisaf, M.S., and Drosos, A.A. (2005).
treatment for a patient with type I cryoglobulinemic glomerulonephri- Cryoglobulinemic purpura in visceral leishmaniasis. Rheumatology
tis. Nature Clinical Practice. Nephrology, 4, 393–7. International, 25, 469–71.
Pascual, M., Perrin, L., Giostra, E., and Schifferli, J.A. (1990). Hepatitis C Roccatello, D., Fornasieri, A., Giachino, O., et al. (2007). Multicenter study
virus in patients with cryoglobulinemia type II. Journal of Infectious on hepatitis C virus related cryoglobulinaemic glomerulonephritis.
Diseases, 162, 569–70. American Journal of Kidney Diseases, 49, 69–82.
Payet, J., Livartowski, J., Kavian, N., et al. (2013). Type I cryoglobulinemia Roccatello, D., Morsica, G., Picciotto, G., et  al. (1997). Impaired hepatos-
in multiple myeloma, a rare entity:  analysis of clinical and biological plenic elimination of circulating cryoglobulins in patients with essen-
characteristics of seven cases and review of the literature. Leukemia and tial mixed cryoglobulinaemia and hepatitis C virus (HCV) infection.
Lymphoma, 54, 767–77. Clinical and Experimental Immunology, 110, 9–14.
Petermann, M.L. and Braunsteiner, H. (1954). A cryoglobulin of high Rosa, D., Saletti, G., De Gregorio, E., et  al. (2005). Activation of naïve B
sedimentation rate (macroglobulin) from human serum. Archives of lymphocyties via CD81, a pathogenetic mechanism for hepatitis C
Biochemistry, 53, 491–500. virus-associated B lynphocite disorders. Proceedings of the National
Pietrogrande, M., Marelli, F., Dilani, S., Pessina, I., and Invernizzi, F. (1999). Academy of Sciences of USA, 102, 18544–9.
Treatment of severe cryoglobulinemic syndrome with plasma exchange Rullier, P., Le Quellec, A., and Cognot, C. (2009). Cryoglobulins not
followed by high dose intravenous immunoglobulins, preliminary HCV-related and solid tumors: retrospective analysis from a series of
results. Journal of Autoimmunity, 12 (Suppl.), S–95. 493 patients. European Journal of Internal Medicine, 20, e158.
Pietrogrande, M., De Vita, S., Zignego, A.L., et al. (2011). Recommendations Saadoun, D., Aaron, L., Resche-Rigon, M., et al. (2006a). Cryoglobulinaemia
for the management of mixed cryoglobulinemia syndrome in hepatitis C vasculitis in patients coinfected with HIV and hepatitis C virus. AIDS,
virus-infected patients. Autoimmunity Reviews, 10, 444–54. 20, 871–7.
CHAPTER 40 cryoglobulinaemic vasculitis 567

Saadoun, D., Asselah, T., Resche-Rigon, M., et al. (2006b) Cryoglobulinemia situ hybridization in bone marrow and peripheral blood mononuclear
is associated with steatosis and fibrosis in chronic hepatitis C. Hepatology, cells of chronically HCV-infected patients. Clinical and Experimental
43, 1337–45. Immunology, 103, 414–21.
Saadoun, D., Bieche, I., Maisonobe, T., et  al. (2005). Involvement of Sansonno, D., Lauletta, G., De Re, V., et al. (2004). Intrahepatic B cell clonal
chemokines and type 1 cytokines in the pathogenesis of hepatitis expansions and extrahepatic manifestations of chronic HCV infection.
C virus-associated mixed cryoglobulinemia vasculitis neuropathy. European Journal of Immunology, 34, 126–36.
Arthritis and Rheumatism, 52, 2917–25. Sansonno, D., Lauletta, G., Nisi, L., et  al. (2003b) Non-enveloped HCV
Saadoun, D., Pineton de Chambrun, M., Hermine, O., et al. (2013). Using core protein as constitutive antigen of cold-precipitable immune com-
rituximab plus fludarabine and cyclophosphamide as a treatment for plexes in type II mixed cryoglobulinaemia. Clinical and Experimental
refractory mixed cryoglobulinemia associated with lymphoma. Arthritis Immunology, 133, 275–82.
Care and Research, 65, 643–7. Sansonno, D., Lotesoriere, C., Cornacchiulo, V., et  al. (1998). Hepatitis C
Saadoun, D., Resche-Rigon, M., and Sene, D. (2010). Rituximab plus virus infection involves CD34(+) hematopoietic progenitor cells in hep-
Peg-interferon-alpha/ ribavirin compared with Peg-interferon-alpha/rib- atitis C virus chronic carriers. Blood, 92, 3328–37.
avirin in hepatitis C-related mixed cryoglobulinemia. Blood, 116, 326–34. Sansonno, D., Tucci, F.A., Ghebrehiwet, B., et al. (2009). Role of the receptor
Saadoun, D., Rosenzwajg, M., Joly, F., et  al. (2011). Regulatory T-cell for the globular domain of C1q protein in the pathogenesis of hepatitis
responses to low-dose interleukin-2 in HCV-induced vasculitis. New C virus-related cryoglobulin vascular damage. Journal of Immunology,
England Journal of Medicine, 65, 2067–77. 183, 6013–20.
Saadoun, D., Sellam, J., Ghillani-Dalbin, P., Crecel, R., Piette, J.C., and Sansonno, D., Tucci, F.A., Troiani, L., et al. (2008). Increased serum levels
Cacoub, P. (2006c). Increased risks of lymphoma and death among of the chemokine CXCL13 and up-regulation of its gene expression are
patients with non-hepatitis C virus-related mixed cryoglobulinaemia. distinctive features of HCV-related cryoglobulinemia and correlate with
Archives of Internal Medicine, 166, 2101–8. active cutaneous vasculitis. Blood, 112, 1620–7.
Saccardo, F., Massaro, P., Monti, G., Angelopulos, N., and Galli, M. (1986). Sargur, R., White, P., and Egner, W. (2010). Cryoglobulin evaluation:  best
Causes of death in essential mixed cryoglobulinemia. La Ricerca in practice? Annals of Clinical Biochemistry, 47, 8–16.
Clinica e in Laboratorio, 16, 389–91. Schott, P., Polzien, F., Müller-Issberner, A., Ramadori, G., and Hartmann, H.
Saccardo, F., Novati, P., Sironi, D., Castelnovo, L., Rinaldi, G., and Monti, G. (1998). In vitro reactivity of cryoglobulin IgM and IgG in hepatitis C
(2007). Causes of death in symptomatic cryoglobulinemia: 30 years of virus-associated mixed cryoglobulinemia. Journal of Hepatology, 28, 17–26.
observation in a Department of Internal Medicine. Digestive and Liver Scotto, G., Cibelli, D.C., Saracino, A., et al. (2006). Cryoglobulinemia in sub-
Disease, 39 (Suppl. 1), S52–4. jects with HCV infection alone, HIV infection and HCV/HIV coinfec-
Sagaert, X., Van Cutsem, E., De Hertogh, G., Geboes, K., and tion. Journal of Infection, 52, 294–9.
Tousseyn, T. (2010). Gastric MALT lymphoma:  a model of chronic Sène, D., Ghillani-Dalbin, P., Thibault, V., et al. (2004). Longterm course of
inflammation-induced tumor development. Nature Reviews. mixed cryoglobulinaemia in patients infected with hepatitis C virus.
Gastroenterology and Hepatology, 7, 336–46. Journal of Rheumatology, 31, 2199–206.
Sansonno, D., Carbone, A., De Re, V., and Dammacco, F. (2007) Hepatitis C Sène, D., Limal, N., Ghillani-Dalbin, P., Saadoun, D., Piette, J.C., and Cacoub,
virus infection, cryoglobulinemia and beyond. Rheumatology (Oxford), P. (2007). Hepatitis C virus-associated B-cell proliferation-the role of
46, 572–8. serum B lymphocyte stimulator (BLyS/BAFF). Rheumatology (Oxford),
Sansonno, D., Cornacchiulo, V., Iacobelli, A.R., Di Stefano, R., Lospalluti, M., 46, 65–9.
and Dammacco, F. (1995). Localization of hepatitis C virus antigens in Sens, Y.A., Malafronte, P., Souza, J.F., et al. (2005). Cryoglobulinemia in
liver and skin tissues of chronic hepatitis C virus-infected patients with kidney transplant recipients. Transplantation Proceedings, 37, 4273–5.
mixed cryoglobulinemia. Hepatology, 21, 305–12. Shalit, M., Wollner, S., and Levo, Y. (1982). Cryoglobulinemia in acute type-A
Sansonno, D. and Dammacco, F. (2005). Hepatitis C virus, cryoglobuli- hepatitis. Clinical and Experimental Immunology, 47, 613–6.
naemia, and vasculitis, immune complex relations. Lancet Infectious Sneller, M.C., Hu, Z., Langford, C.A. (2012). A randomized controlled
Diseases, 5, 227–36. trial of rituximab following failure of antiviral therapy for hepatitis C
Sansonno, D., De Re, V., Lauletta, G., Tucci, F.A., Boiocchi, M., and virus-associated cryoglobulinemic vasculitis. Arthritis and Rheumatism,
Dammacco, F. (2003a). Monoclonal antibody treatment of mixed cry- 64, 835–42.
oglobulinemia resistant to interferon alpha with an anti-CD20. Blood, Spertini, F., Gyotoku, Y., Shibata, T., Izui, S., and Lambert, P.H. (1988).
101, 3818–26. Experimental model of murine cryoglobulinemia induced by mono-
Sansonno, D., De Vita, S., Cornacchiulo, V., Carbone, A., Boiocchi, M. clonal antibodies and modulation by anti-idiotypic antibodies. Springer
and Dammacco, F. (1996a). Detection and distribution of hepatitis C Seminars in Immunopathology, 10, 91–101.
virus-related proteins in lymph nodes of patients with type II mixed Stein, H.B., Schlappner, O.L., Boyko, W., Gourlay, R.H., and Reeve, C.E.
cryoglobulinemia and neoplastic or non-neoplastic lymphoprolifera- (1981). The intestinal bypass:  arthritis-dermatitis syndrome. Arthritis
tion. Blood, 88, 4638–45. and Rheumatism, 24, 684–90.
Sansonno, D., De Vita, S., Iacobelli, A.R., Cornacchiulo, V., Boiocchi, M., Steinhardt, M.J. and Fisher, G.S. (1955). Essential cryoglobulinemia.
and Dammacco, F. (1998). Clonal analysis of intrahepatic B cells from American Journal of Medicine, 65, 848–58.
HCV-infected patients with and without mixed cryoglobulinemia. Sutton, B., Corper, A., Bonagura, V., and Taussig, M. (2000) The structure
Journal of Immunology, 160, 3594–601. and origin of rheumatoid factors. Immunology Today, 21, 177–83.
Sansonno, D., Gesualdo, L., Manno, C., Schena, F.P., and Dammacco, F. Taillan, B., Garnier, G., Pesce, A., et al. (1993). Cryoglobulinemia related to
(1997). Hepatitis C virus-related proteins in kidney tissue from hepatitis hepatitis C virus infection in patients with human immunodeficiency
C virus-infected patients with cryoglobulinemic membranoproliferative virus infection. Clinical and Experimental Rheumatology, 11, 350.
glomerulonephritis. Hepatology, 25, 1237–44. Tarantino, A., Montagnino, G., Baldassarri, A., et al. (1986). Prognostic fac-
Sansonno, D., Iacobelli, A.R., Cornacchiulo, V., et  al. (1996b). tors in essential mixed cryoglobulinemia nephropathy. In Antiglobulins,
Immunochemical and biomolecular studies of circulating immune com- cryoglobulins and glomerulonephritis (ed. C. Ponticelli, L. Minetti, G.
plexes. European Journal of Clinical Investigation, 26, 465–75. D’Amico), pp. 219–32. Martinus Nijoff, Dordrecht.
Sansonno, D., Iacobelli, A.R., Cornacchiulo, V., Iodice, G., and Dammacco, Tavoni, A., Mosca, M., Ferri, C., et al. (1995). Guidelines for the manage-
F. (1996c). Detection of hepatitis C virus (HCV) proteins by immu- ment of essential mixed cryoglobulinemia. Clinical and Experimental
nofluorescence and HCV RNA genomic sequences by non-isotopic in Rheumatology, 13 (Suppl. 13), S191–5.
568 SECTION 5 vasculitic diseases and syndromes and related disorders

Terrier, B. and Cacoub, P. (2013). Cryoglobulinemia vasculitis: an update. cryoglobulinemia secondary to hepatitis C virus infection. Autoimmunity
Current Opinion in Rheumatology, 25, 10–8. Reviews, 10, 714–9.
Terrier, B., Karras, A., Kahn, J.E., et al. (2013). The spectrum of type I cry- Wager, O., Rasanen, J.A., and Sihvonen, T. (1969). Immunological studies
oglobulinemia vasculitis:  new insights based on 64 cases. Medicine on mixed cryoglobulinemia. In Human anti-human gammaglobulins
(Baltimore), 92, 61–8. (ed. R. Grubb and G. Samuelsson), pp. 161–71. Pergamon Press, Oxford,
Terrier, B., Krastinova, E., Marie, I., et al. (2012). Management of noninfec- New York.
tious mixed cryoglobulinemia vasculitis: data from 242 cases included Wang, A.C. (1988). Molecular basis for cryoprecipitation. Springer Seminars
in the CryoVas survey. Blood, 119, 5996–6004. in Immunopathology, 10, 21–34.
Terrier, B., Launay, D., Kaplanski, G., et al. (2010a). Safety and efficacy of Wang, Y., Lomakin, A., Hideshima, T., et al. (2012). Pathological crystalliza-
rituximab in nonviral cryoglobulinemia vasculitis: data from the French tion of human immunoglobulins. Proceedings of the National Academy
Autoimmunity and Rituximab registry. Arthritis Care and Research, 62, of Sciences USA, 109, 13359–61.
1787–95. Wener, M.H., Hutchinson, K., Morishima, C., and Gretch, D.R. (2004).
Terrier, B., Saadoun, D., Sène, D., Scerra, S., Musset, L., and Cacoub, P. Absence of antibodies to cyclic citrullinated peptide in sera of patients
(2010b). Presentation and outcome of gastrointestinal involvement in with hepatitis C virus infection and cryoglobulinaemia. Arthritis and
hepatitis C virus-related systemic vasculitis: a case-control study from a Rheumatism, 50, 2305–8.
single-centre cohort of 163 patients. Gut, 59, 1709–15. Wilbur, K. and Makowsky, M. (2004). Colchicine myotoxicity: case reports
Terrier, B., Semoun, O., Saadoun, D., et  al. (2011). Prognostic factors in and literature review. Pharmacotherapy, 24, 1784–92.
patients with hepatitis C virus infection and systemic vasculitis. Arthritis Williams, R.C. Jr and Malone, C.C. (1994). Rheumatoid-factor-reactive
and Rheumatism, 63, 1748–57. sites on CH2 established by analysis of overlapping peptides of primary
Tissot, J.D., Schifferli, J.A., Hochstrasser, D.F., et al. (1994). Two-dimensional sequence. Scandinavian Journal of Immunology, 40, 443–56.
polyacrylamide gel electrophoresis analysis of cryoglobulins and iden- Wintrobe, M.M. and Buell, M.V. (1933). Hyperproteinemia associated
tification of an IgM-associated peptide. Journal of Immunological with multiple myeloma, with report of a case in which an extraordi-
Methods, 173, 63–75. nary hyperproteinemia was associated with thrombosis of retinal veins
Toubi, E., Gordon, S., Kessel, A., et al. (2006). Elevated serum B-Lymphocyte and symptoms suggesting Raynaud’s disease. Bulletin of John Hopkins
activating factor (BAFF) in chronic hepatitis C virus infection: associa- Hospital, 52, 156–72.
tion with autoimmunity. Journal of Autoimmunity, 27, 134–9. Zaja, F., De Vita, S., Mazzaro, C., et al. (2003). Efficacy and safety of rituximab
Toyoda, H., Fukuda, Y., Koyama, Y., Takamatsu, J., Saito, H., and Hayakawa, in type II mixed cryoglobulinemia. Blood, 101, 3827–34.
T. (1997). Effect of immunosuppression on composition of quasispecies Zehender, G., de Maddalena, C., Bernini, F., et  al. (2005).
population of hepatitis C virus in patients with chronic hepatitis C coin- Compartimentalization of Hepatitis C virus quasispecies in blood
fected with human immunodeficiency virus. Journal of Hepatology, 26, mononuclear cells of patients with mixed cryoglobulinemic sindrome.
975–82. Journal of Virology, 79, 9145–56.
Trejo, O., Ramos-Casals, M., García-Carrasco, M., et  al. (2001). Zehender, G., de Maddalena, C., Monti, G., et al. (1995). HCV genotypes in
Cryoglobulinemia: study of etiologic factors and clinical and immuno- bone marrow and peripheral blood mononuclear cells of patients with
logic features in 443 patients from a single center. Medicine (Baltimore), mixed cryoglobulinemia. Clinical and Experimental Rheumatology, 13
80, 252–62. (Suppl. 13), S87–90.
Trejo, O., Ramos-Casals, M., López-Guillermo, A., et al. (2003). Hematologic Zehender, G., Meroni, L., de Maddalena, C., Varchetta, S., Monti, G., and
malignancies in patients with cryoglobulinaemia:  association with Galli, M. (1997). Detection of hepatitis C virus RNA in CD19 periph-
autoimmune and chronic viral diseases Seminars in Arthritis and eral blood mononuclear cells of chronically infected patients. Journal of
Rheumatism, 33, 19–28. Infectious Diseases, 176, 1209–14.
Treon, S.P. (2009). How I treat Waldenström macroglobulinemia. Blood, 114, Zignego, A.L., Ferri, C., Giannini, C., et al. (1996). Hepatitis C virus geno-
2375–85. type analysis in patients with type II mixed cryoglobulinemia. Annals of
Tzioufas, A.G., Boumba, D.S., Skopouli, F.N., and Moutsopoulos, H.M. Internal Medicine, 124, 31–4.
(1996). Mixed monoclonal cryoglobulinaemia and monoclonal rheu- Zignego, A.L., Giannelli, F., Marrocchi, M.E., et al. (1997). Frequency of bcl-2
matoid factor cross-reactive idiotypes as predictive factors for the devel- rearrangement in patients with mixed cryoglobulinemia and HCV-positive
opment of lymphoma in primary Sjogren’s syndrome. Arthritis and liver diseases. Clinical and Experimental Rheumatology, 15, 711–2.
Rheumatism, 39, 767–72. Zignego, A.L., Giannelli, F., Marrocchi, M.E., et al. (2000). T(14;18) trans-
Vigani, A.G., Pavan, M.H., Tozzo, R., et  al. (2008). Comparative study of location in chronic hepatitis C virus infection. Hepatology, 31, 474–9.
patients with chronic hepatitis C virus infection due to genotype 1 and Zignego, A.L., Giannini, C., and Gragnani, L. (2012). HCV and lymphopro-
3 referred for treatment in southeast Brazil. BMC Infectious Diseases, liferation. Clinical and Developmental Immunology, 2012, 980942.
8, 164. Zignego, A.L., Macchia, D., Monti, M., et al. (1992). Infection of peripheral
Visentini, M., Ludovisi, S., Petrarca, A., et al. (2011). A phase II, single-arm mononuclear blood cells by hepatitis C virus. Journal of Hepatology,
multicenter study of low-dose rituximab for refractory mixed 15, 382–6.
CHAPTER 41

Miscellaneous forms
of vasculitis
Gim Gee Teng, Sumapa Chaiamnuay, and
W. Winn Chatham

For many patients presenting with clinical manifestations of Vasculitides induced by microbial
vasculitis, the combination of historical features, physical find-
ings, results of clinical laboratory tests, and histological exami- pathogens
nation will identify a readily classifiable vasculitic syndrome, Infections have long been implicated as triggers of primary vascu-
such as polyarteritis nodosa, or associated autoimmune dis- litides although causality is yet to be affirmed in most syndromes.
ease, such as systemic lupus erythematosus. In some patients, Nonetheless, certain infections are notorious mimickers of vas-
an established associated infectious trigger such as hepatitis C culitis. Inflammation of vessel walls due to direct or contiguous
virus or hepatitis B virus may be identified as the inciting factor. infection, type II or immune complex reaction, cell-mediated
Less commonly, comprehensive evaluation will dictate consid- hypersensitivity, or inflammation due to immune dysregulation
eration of either a rare disorder or one that, while not uncom- triggered by bacterial toxin and/or superantigen production, can
mon, has vasculitis as an uncommon feature. Recognition and compromise vascular integrity culminating in end-organ dam-
appreciation of the unique clinical and histopathological fea- age (Belizna 2009). Many organisms with propensity to involve
tures of uncommon causes of vasculitis is important not only blood vessels have been implicated in vascular inflammation
for optimal patient diagnosis and management but also for an (Table  41.1). Clinical manifestations overlap significantly with
understanding of the spectrum of vascular injury syndromes those of the primary vasculitides, ranging from non-specific
and their potential causes. These miscellaneous vasculitides constitutional symptoms, pyrexia, and a variety of skin lesions to
include: infarction of major organs. A high index of suspicion for infection
◆ Vasculitides induced by microbial pathogens as a potential cause of vasculitis is prudent in that appropriate
antimicrobial/ antiviral therapy can be directed at the underly-
◆ Vasculitis associated with immunodeficiency syndromes ing organism and immunosuppressive therapy administered in
Wiskott–Aldrich syndrome the absence of antimicrobial therapy is likely to increase morbid-
ity. This section considers pathogenesis of infection-related vas-
X-linked lymphoproliferative disorder
culitis, with a focus on described viral and bacterial aetiologies.
Purine nucleoside phosphorylase deficiency Localized suppuration of vessel wall related to foreign material
◆ Vasculitis associated with sarcoidosis (e.g. catheter insertion and bypass grafts) or invasive procedures
(e.g. angiography and vascular surgery) is beyond the scope of
◆ Cogan’s syndrome
this chapter.
◆ Degos’ disease
◆ Urticarial vasculitis Endovascular bacterial infection
◆ Erythema elevatum diutinum Bacterial endocarditis
◆ Familiar Mediterranean fever Bacterial endocarditis may trigger the development of systemic
vasculitis (Miranda-Filloy et  al. 2006). Vascular injury simulat-
◆ Hyperimmunoglobulinaemia D
ing vasculitis of various sized intracranial vessels has been dem-
◆ IgG4-related disease onstrated on routine cranial angiography and on cranial magnetic
◆ Paraneoplastic vasculitis syndromes resonance imaging studies in patients with bacterial endocarditis
and meningitis (Ferris et  al. 1968; Jan et  al. 2003), with mycotic
Myelodysplastic syndrome and myeloproliferative disorders aneurysm, retinal vasculitis, brain infarcts, and brain haemorrhage
Solid tumours. being dire consequences (Kastenbauer and Pfister 2003; Lee et al.
570 SECTION 5 vasculitic diseases and syndromes and related disorders

Table 41.1 Microbial pathogens implicated in vasculitis retroperitoneal collections, can also occur (Cook et al. 1989). In a
clincopathological study of 29 cases of surgical repair of infected
Viral aortic aneurysms, acute inflammation superimposed on severe
Parvovirus B19 chronic atherosclerosis was most prevalent, with approximately
Herpes virus: VZV, CMV, EBV one-third of the cases exhibiting only chronic inflammatory
Hepatitis viruses:Hep B, Hep C changes including lymphoplasmacytic inflammation, fibrosis,
Retroviruses: HTLV, HIV
healed pseudoaneurysm, and/or chronic atherosclerotic changes
without purulent inflammation. Acute inflammation tended to
Bacterial occur in patients with the shortest duration of symptoms and
Staphylococci antibiotic therapy. Blood cultures are positive in 50 to 85% and
Streptococci a microbe can be isolated from the excised aortic tissue in up to
Neisseria species 76% (Lopes et al. 2009), although in other published case series
Gram-negative enteric pathogens (salmonella)
bacterial culture from the aneurysm wall was noted to be positive
in less than half of surgical cases with the frequency of negative
Mycobacteria blood or tissue cultures ranging from 2 to 33% (Miller et al. 2004).
Spirochetes Endovascular infections, most commonly of the abdominal
Other aorta, are rare complications of non-typhoid salmonellosis (Cohen
et al. 1978; Lane et al. 1988; Wang et al. 1996). The onset is insidi-
Fungal ous and typically runs a subacute course with mean duration
Aspergillus of symptoms of 1  month (range 1  day to 6  months) in reported
Coccidioides cases (Soravia-Dunand et  al. 1999). The most common isolates
Candida are Salmonella  typhimurium (serogroup B), S.  enteritidis (sero-
group D1), and S. enterica serotype Choleraesuis (S. choleraesuis;
Mucormycetes
serogroup C1), in that order (Soravia-Dunand et al. 1999; Oskoui
Parasites et  al. 1993). Albeit extremely uncommon below the age of 50,
Cysticercus risks factors should be sought in younger patients with salmonella
Rickettsiae bacteraemia or arteritis, especially if recurrent. In a 20-year retro-
spective study of 373 cases of non-typhoid salmonellosis, systemic
Mycoplasma lupus erythematosus, liver cirrhosis, HIV infection, and solid organ
cancers were shown to be independent risk factors for salmonella
bacteraemia but the only independent positive predictor of endo-
vascular infection was atherosclerosis, contrary to the traditional
2005; Reddy 2008). In addition to directed antibiotic therapy, early
belief that chronic immunosuppression such as immunodeficiency
adjunctive use of dexamethasone in selected patients may reduce
and malignancies may be at play (Hsu et al. 2005).
vascular inflammation and neurological sequelae (McIntyre et al.
Other categories of infectious aortitis include syphilitic,
1997; De Gans et al. 2002).
tuberculous, and lepromatous. Important differential diagnoses
Infectious endarteritis and aortitis include several syndromes that affect the aorta and surround-
While Takayasu’s arteritis and giant cell arteritis are the most ing structures described under the label of chronic periaorti-
common types of vasculitis that cause aortitis, infective aor- tis, including idiopathic retroperitoneal fibrosis, mediastinal
titis should be suspected in patients, often older men with ath- fibrosis, and inflammatory aneurysm (see Chapter  18, includ-
erosclerosis, presenting with prolonged fever, abdominal or back ing Figures 18.20, 18.21, and 18.22) and IgG4-related systemic
pain, palpable abdominal mass, and leukocytosis, with or with- disease.
out positive blood cultures (Lopes 2009). Infection may arise as Computed tomographic angiography is the most useful imaging
(1) direct extension of a local infection, (2) traumatic contami- modality for evaluating suspected infectious aortitis as it is widely
nation, (3) septic embolism from infective endocarditis (IE), or available and fast. The role of leukocyte scintigraphic imaging is
(4) haematogenous seeding from a distant source of bacteraemia. controversial. In one report, an abnormal study led to a confirmed
Extension from contiguous infection (Bronze et al. 1999) occurs tissue diagnosis in >80% of studies (Macedo et al. 2004). Magnetic
less commonly. resonance imaging (MRI) with gadolinium enhancement should
Whereas streptococci predominated in the preantibiotic era as be considered when the use of iodinated contrast media is con-
the pathogens for IE- associated infectious aortitis, Staphylococcus traindicated. Infective aortitis is fatal if untreated. Optimal treat-
aureus and enteric Gram-negative bacteria (especially salmo- ment with early surgery, with excision of infected aorta and bypass
nella) are the main causative organisms seeding atheromatous grafting, in combination with prolonged antibiotic administration
vessels (Foote et  al. 2005; Lidar et  al. 2009). Micro-organisms have improved outcomes (Ting et al. 2005; Fernandez Guerreroet
including enterococci, Clostridium septicum, and Listeria mono- et al. 2004).
cytogenes, and fungal organisms such as Candida and Aspergillus,
are rare but increasing in immunosuppressed patients (Gornik Septic phlebitis
and Creager 2008). Mycotic aneurysm, aneurysm rupture, aor- Lemierre’s syndrome (postanginal sepsis) rarely complicates an
toduodenal fistula, and vertebral osteomyelitis are possible com- oropharyngeal infection in previously healthy adolescent and
plications. Rupture of non-aneurysmal infected aorta, cued by young adults, evolving as a septic thrombophlebitis of the internal
CHAPTER 41 miscellaneous forms of vasculitis 571

jugular vein with subsequent septicaemia and metastatic lesions to components of the coagulation cascade (Sahni and Rydkina 2009).
the lungs, musculoskeletal system, and other sites (Moreno 1989; Activation of NF-κB plays a crucial role by controlling the expres-
Karkos et al. 2009). The usual aetiological agent is Fusobacterium sion of several procoagulant and proinflammatory genes (Shi et al.
necrophorum, but may not be uniquely anaerobic. Staphylococcus 1998) and by protecting the host cells from apoptosis (Clifton et al.
aureus is an emerging pathogen in case reports (Chanin 2011). An 1998). Cell damage has been associated with rickettsial phospho-
inflammatory prothrombotic state characterized by antiphospho- lipase A2 and protease activities and free-radical-induced injury
lipid antibodies and elevated factor VIII levels has been suggested as (Walker 1989). The net effect is a lymphohistocytic response with
an attendant pathogenic mechanism in the syndrome (Goldenberg widespread vasculitis, most importantly in the brain and lungs,
et al. 2005). Early diagnosis, surgical drainage, and aggressive anti- causing increased vascular permeability, oedema, hypotension,
microbial therapies are usually curative while the role of antico- and multiorgan failure (Woods and Olano 2008). Hyponatraemia
agulation is controversial (Chirinos et al. 2002; Karkos et al. 2009). may be due to hypovolaemia from capillary leak or syndrome of
Pylephlebitis or septic thrombophlebitis of the portovenous inappropriate antidiuretic hormone secretion (Mouallem et  al.
system (e.g. portal vein, inferior and superior mesenteric vein) 1987). Interstitial pneumonia, myocarditis, perivascular glial nod-
begins from small veins that drain infected areas resulting from ules of the CNS and similar vascular nodules in the skin, GI tract,
a perforated viscus, most commonly diverticulitis, appendicitis, pancreas, liver, skeletal muscles, and kidneys have been observed
or abdominal sepsis from trauma/ surgery (Kanellopoulou et  al. (Walker and Ismail 2010).
2010). Bacteraemia (often polymicrobial) was present in most The non-specific presentation poses a diagnostic challenge,
patients with Bacteroides fragilis, Escherichia coli, and Streptococcus and often leads to delayed therapy. Symptoms usually commence
species being the most common blood isolates (Kanellopoulou 2–14 days after the tick bite with fever, headache, myalgias, nausea
et al. 2010). Detection of venous thrombus and venous gas by com- with or without vomiting, and abdominal pain (Kirk et al. 1990).
puted tomography is diagnostic and intrahepatic abscesses due to The characteristic rash appears after 3–5  days and evolves from
septic embolization may be seen (Kasper et al. 2005). In cases unre- macular to maculopapular to petechial, on the ankles and wrists and
lated to cirrhosis or malignancy, an inherited thrombophilia such then spreads centrally and to the palms and soles. Absence of the
as factor V Leiden mutation, prothrombin 20210 G/A mutation, typical rash (‘viscerotropic’ RMSF) frequently delays diagnosis and
and protein C deficiency is often an underlying predisposing factor is more commonly associated with fatal outcomes (Hattwick et al.
superimposed on local intra-abdominal infection (Kanellopoulou 1976). An eschar at the site of the tick bite may be evident on care-
et  al. 2010; Janssen et  al. 2000). Combined anticoagulation and ful history and physical examination (Walker et al. 1981; Shapiro
antibiotics appeared to aid in recanalization (Kanellopoulou et al. 2010). Meningoencephalitis and neurological involvement is gen-
2010). Septic ovarian thrombosis must be sought for in females erally ominous (Horney and Walker et al. 1988). Non-cardiogenic
with puerperal fever, pelvic pain, or following pelvic surgery as pulmonary oedema and adult respiratory distress may require
potentially fatal complications such as sepsis, septic pulmonary mechanical ventilation. Azotaemia, skin necrosis, thrombocyto-
thromboembolism, and thrombosis of the inferior vena cava and penia (true disseminated intravascular coagulation is rare), retinal
the renal veins can ensue (Hippach et al. 2000). vasculitis, and multifocal rhabdomyolysis are other complications.
In fulminant RMSF, death occurs within the first 5  days, and is
Rickettsial vasculitis more common with increasing age, male gender, and presence of
Rickettsiae are arthropod-borne (ticks), Gram-negative, obligate glucose-6-phosphate dehydrogenase deficiency (Hattwick et  al.
intracellular bacteria, which primarily infect endothelial cells. They 1976; Walker 1990).
cause systemic multiorgan diseases such as Rocky Mountain spot- It is advisable to inquire about travel history and tick exposures
ted fever (RMSF; caused by Rickettsia rickettsii), sporadic but poten- in the appropriate setting as early clinical diagnosis can be lifesav-
tially persistent boutonneuse fever (caused by Rickettsia conorii), ing, using empirical therapy with a tetracycline class antibiotic
as well as other spotted fevers (Walker and Ismail 2008; Richards (Kirkland et al. 1995). Culture is difficult for rickettsiae but new
2012). RMSF can be potentially fatal, even in young immunocom- genetic tools and use of cell culture assays have improved detec-
petent individuals, if not adequately treated with antibiotics early tion. Serological analysis confirms diagnosis retrospectively only
during the course of the illness. when antibodies appear during convalescence but is not use-
When Rickettsia are introduced into the skin, the organisms ful during initial management and antibody cross-reactivity may
spread via lymphatics into the circulation, infecting and replicating result in false-positive test results. Polymerase chain reaction
in the cytoplasm of target endothelial cells after escaping rapidly (PCR) of rickettsial DNA in blood samples is insensitive; however,
from phagosomes. The organisms then traverse cell-to-cell utiliz- RT- PCR analysis for genes of major surface proteins and immu-
ing RickA protein-induced actin polymerization for directional nohistochemical analysis of skin biopsy specimens for R. rickettsii
intracellular movements and intercellular spread (Heinzen 2003; antigen have increased sensitivity and provide timely diagnosis of
Gouin et al. 2004). The outer membrane proteins and cell surface emerging as well as established rickettsioses (Prakash et al. 2012;
antigens (e.g. Omps, Sca-2, Sca-4), purported immunogens capable Richards 2012).
of eliciting protective immune responses in experimental animals
(Walker 1989), facilitate attachment to and entry into the endothe- Viral-induced vasculitides
lial cell membrane via vinculin activation and actin organization Viruses are frequently considered to be causative agents in vas-
(Park et al. 2011; Cardwell and Martinez 2009). Following attach- culitis due to the frequent occurrence of circulating immune
ment and ‘induced phagocytosis’ of the pathogen into the cytosol, a complexes during acute viral infection and the tropism of some
myriad of host cell immune responses follow, including the expres- viruses for vascular cells. The associations of hepatitis B and hep-
sion of adhesion molecules, cytokines, chemokines, and regulatory atitis C with polyarteritis nodosa (PAN) and cryoglobulinaemia
572 SECTION 5 vasculitic diseases and syndromes and related disorders

are well recognized and are discussed in detail elsewhere (see Kawasaki’s disease remains speculative (Holm et  al. 1995; Nigro
Chapters  25 and 40). Vasculitis induced by herpes viruses, et al. 1994; Rowley et al. 1994).
including cytomegalovirus (CMV), varicella zoster virus (VZV), Parvovirus DNA has also been detected by PCR amplification in
and Epstein–Barr virus (EBV), has been reported predominantly temporal artery biopsy specimens of patients with GCA, suggesting
in the context of immunosuppression, with most reports involv- a possible role for the virus in the development of large-vessel vas-
ing transplant recipients, patients on chemotherapy for cancer, or culitis (Gabriel et al. 1999), particularly in those patients with high
patients with immunodeficiency due to HIV infection. Evidence viral load (Álvarez-Lafuente et al. 2005). However, results of other
for infection with various other viruses, including parvovirus studies did not corroborate this observation (Salvarani et al. 2002;
B19, in the clinical setting of vasculitis affecting small, medium, Rodriguez-Pla et al. 2004; Cooper 2008). Furthermore, a similar
or large-calibre vessels, has been documented in various case frequency of PVB19 DNA localization was found in arterial sur-
reports. As the role of viruses in vasculitis becomes better under- gical specimens obtained from age-matched controls with athero-
stood, treatment regimens incorporating therapies targeting sclerotic carotid/ aortic disease (Salvarani et al. 2002).
specific viruses are playing a greater role in the management of
Cytomegalovirus
vasculitis.
While CMV commonly produces invasive disease affecting multi-
Parvovirus B19 ple organs in the immunocompromised host, it is also capable of
Although usually asymptomatic, acute human PVB19 infection infecting endothelial cells, leading to a clinical presentation con-
may present with various clinical manifestations ranging from mild sistent with small to medium-sized vessel vasculitis (Golden et al.
self-limiting erythema infectiosum in immunocompetent children 1994). CMV DNA sequences and proteins have also been identi-
to lethal cytopenias (transient aplastic anaemia, chronic anaemia) fied in inflammatory lesions of the aorta, suggesting the virus may
in immunocompromised patients or those with haemoglobinopa- play a role in the pathogenesis of aortitis (Tanaka et al. 1994). The
thies, and fetal hydrops in pregnant women. In adults, there may lesions associated with CMV vasculitis are most common in the gut
be symmetric polyarthralgia and polyarthritis. PVB19 has tropism or the skin, but their appearance is frequently non-specific; neuro-
for cell types that possess the P blood group antigen globoside and logical deficits due to central or peripheral nervous system infarcts
α5β1 integrins, the main receptor complex for PVB19 entry into may also appear (Koeppen et al. 1981; Golden et al. 1994; Tatum
the cell (Slavov 2011). The target cells include endothelium as well et  al. 1989). Biopsy of lesions to identify cytomegalic endothelia
as erythroid progenitors, megakaryocytes, fetal myocytes, and pla- and macrophages with intranuclear inclusions is required to estab-
cental trophoblast. lish the diagnosis.
Acute PVB19 infection has been associated with clinical features CMV-induced vasculitis involving small or medium-sized
suggestive of vasculitis, including intestinal gangrene (Kishore arteries with enteric vasculitis, alveolar haemorrhage, and classic
et  al. 2010; Dyrsen et  al. 2008), papular–purpuric ‘glove and PAN, have been reported to occur in immunocompetent patients
stocking’ syndrome (Santonja 2011; Aguilar-Bernier et  al. 2006; (Fernandes et  al. 1999; Taniwaki et  al. 1997; Magro et  al. 2005).
Bilenchi et al. 2012), mononeuritis multiplex (Lenglet et al. 2011), Pulmonary septal capillary injury without classic inclusion body
immune-complex glomerulonephritis (Mori et al. 2002 Sakalli et al. cytopathic change, which is morphologically indistinguishable
2010), cutaneous infarction (Dyrsen et al. 2008), and macrophage from small-vessel vasculitis involving the lungs, has also been
activation syndrome (Soldo-Juresa et al. 2010). While mild cases described. In a recent meta-analysis, thrombosis (splanchnic vein
may resolve spontaneously, severe disease warrants use of IVIg and thrombosis, deep vein thrombosis, pulmonary embolism) asso-
occasionally corticosteroids. It should be recognized that acute par- ciated with CMV infection was more prevalent in immunocom-
vovirus infection may also complicate the clinical course of other petent hosts, particularly those with inherited thrombophilia in
medium and small-vessel vasculitides. The presence of parvovirus the setting of CMV mononucleosis (Justo et al. 2011). In a young
in reported cases of granulomatosis with polyangiitis appeared to patient (Factor V Leiden heterozygote) with multifocal superfi-
alter expected response to therapy wherein the manifestations of cial phlebitis in primary CMV infection, transmural lymphocytic
vasculitis responded poorly to treatment with glucocorticoids and inflammatory infiltrate filled with CD45 positive cells and cytotoxic
cytotoxic agents, only to resolve following courses of high-dose T cells and neovascularization were evident on blood vessel biopsy
IVIg (Finkel et al. 1994). without detectable CMV proteins on immunohistochemistry or
The tropism of PVB19 for endothelium has prompted numer- PCR (Jeanneret 2008). CMV has been implicated in the patho-
ous investigations examining the potential role of the virus in the genesis of arteriosclerotic plaques in cerebral arteries and in stroke
pathogenesis of the established vasculitides. Through serological, (Nagel 2010).
immunohistochemistry, and DNA amplification in blood samples, Chronic viral shedding and latency due to persistence of her-
skin tissue, and endothelial cells, human PVB19 infection has been pesvirus in otherwise immunocompetent hosts can develop in
implicated in the pathogenesis of polyarteritis nodosa and gran- overt reactivation of CMV when they become critically ill, espe-
ulomatosis with polyangiitis (Finkel et al. 1994), Behçet’s disease cially with sepsis (Cook 2011). CMV infection, primary or reacti-
(Baskan et al. 2007), and Henoch–Schönlein purpura (Cioc et al. vation, should also be suspected in a setting of clinical worsening
2002; Ferguson et al. 1996; Veraldi et al. 1999; Ohtsuka et al. 2005; despite traditional immunosuppression of presumed autoimmune
Moffat et  al. 1996). A  pathogenic role for PVB19 has also been vasculitis (Magro et al. 2005). A diagnostic and therapeutic chal-
implicated in the vasculopathy associated with systemic sclero- lenge arises when CMV-induced vasculitis occurs in patients who
sis (Zakrzewska et al. 2009), dermatomyositis (Magro et al. 2009; are on immunosuppressive therapy for treatment of other vascu-
Lewkonia et al. 1995) and systemic lupus erythematosus (Trapani litides, including granulomatosis with polyangiitis (Sackier et  al.
1999; Pavlovic 2010). The association of human PVB19 with 1991; Weiss et  al. 1993)  as manifestations of CMV infection in
CHAPTER 41 miscellaneous forms of vasculitis 573

such patients may mimic recrudescence of the underlying vascu- studies show pleocytosis and frequently red blood cells. Diagnosis
litis. Appropriate cultures, PCR studies, and mucosal biopsy speci- of VZV vasculopathy can be confirmed by presence of VZV DNA
mens to rule out CMV infection should be procured in previously or anti-VZV IgG (the most sensitive test) or IgM antibody in CSF
stable patients who develop new lesions involving the skin, lungs, or of anti-VZV IgM antibody in serum. VZV infection should be
glomeruli, gut, or CNS. In transplant recipients, CMV infection considered in the differential diagnosis of patients with idiopathic
may be difficult to differentiate from allograft rejection. In stud- primary angiitis of the CNS and all strokes of unclear aetiology, par-
ies of allograft biopsies, CMV infection is associated with a higher ticularly in HIV-positive and other immunocompromised patients.
prevalence of subintimal arteriolar allograft inflammation, and it According to expert/ consensus opinion, suspected cases should be
is postulated that this infection may promote the development of treated immediately with intravenous acyclovir for at least 2 weeks
atherosclerotic lesions in the allograft (Koskinen et al. 1994). and a short course of prednisone to control the inflammation of the
While necrotizing arteritis may require glucocorticoid treat- cerebral arteries (Gilden 2009).
ment initially, appropriate antiviral therapy should be instituted
concurrently and promptly in patients with CMV-associated vas- Human T-cell lymphotropic virus-1
culitis. Early recognition of CMV-induced vasculitis in immu- Human T-cell lymphotropic virus-1 (HTLV-1) is one of several ret-
nosuppressed patients is critical, even after apparently adequate roviruses capable of directly or indirectly inducing vascular lesions.
anti-CMV prophylaxis (Zandberg et al. 2005), as favourable out- In addition to inducing T-cell lymphomas, HTLV-1 has been iden-
comes require prompt institution of antiviral therapy (with gan- tified as a causative factor in a number of non-malignant inflam-
ciclovir or foscarnet) and a decrease in the immunosuppressive matory lesions, most notably a myelopathy classically described as
regimen (Golden et al. 1994). tropical spastic paraparesis in endemic areas. Vascular complica-
tions of HTLV-1 infection include inflammation of the uveal tract
Varicella zoster virus
with anterior uveitis and retinal vasculitis (Hayasaka et al. 1991;
Varicella zoster virus (VZV) vasculitis in the central nervous sys- Watanabe et al. 1997; Miyanaga 2009). HTLV-1 infection is diag-
tem (CNS) (an entity previously termed granulomatous CNS angii- nosed by the presence of elevated serum or CNS antibody titres to
tis) results from direct viral infection of large and small cerebral HTLV-1 in an appropriate clinical setting.
vessels and can follow either primary varicella infection (varicella)
or reactivation (zoster) (Nagel 2010). Cutaneous or ophthalmic Human immunodeficiency virus
manifestations of herpes zoster are frequently absent; notably, in Vasculitis associated with human immunodeficiency virus (HIV)
more than one-third of patients, CNS VZV vasculitis was not asso- is heterogeneous with regard to the clinical presentation and histo-
ciated with rash (Nau et al. 1998; Orme et al. 2007) and some may logical appearance (Guillevin 2008). Vascular inflammation in HIV
only have worsening post-herpetic neuralgia (zoster sine herpete) infection may occur as a result of host responses to HIV, opportun-
(Chang 2012). In an earlier case report, VZV DNA and antigens istic infection, reactivation of latent viral infections, co-infection
were identified in the affected arterial tissues of an elderly man with HCV and HBV, or hypersensitivity to antimicrobial and anti-
with segmental granulomatous angiitis of the CNS, in the absence viral drugs. Moreover, a state of immune activation is characteristi-
of cutaneous or ophthalmic zoster (Gilden et al. 1996). cally observed in HIV infection, creating a favourable milieu for
The disease runs a relatively subacute and protracted course, from the development and perpetuation of vascular inflammation. These
weeks to months after herpes zoster ophthalmicus or zoster that considerations emphasize the need to carefully consider all these
may have occurred anywhere in the body, with duration of disease aetiologies and HIV biology when formulating a diagnostic and
manifestations occurring up to 2  years before diagnosis (Gilden treatment approach to HIV patients who present with vasculopathy.
et  al. 1996; Gilden 2004; Silver et  al. 2012). VZV vasculopathy Small-calibre arterial vessels in dermal, muscular, and periph-
affects both immunocompetent as well as immunocompromised eral neural tissues are most commonly affected, with peripheral
patients including those with organ transplant, cancer or HIV/ neuropathy a common presenting manifestation (Calabrese 1991;
AIDS (see Section Human Immunodeficiency Virus) or on biologi- Gherardi et al. 1993). HIV-associated polyarteritis nodosa (PAN)
cal agents (Baek et al. 2012). Primary VZV infection (chicken pox) tends to have a less fulminant course compared to classic PAN
may precede acute hemiplegia suggesting VZV involvement in the (Gajera and Kais 2009). Mostly asymptomatic, mixed cryoglob-
pathogenesis of post-VZV focal cerebral arteriopathy of childhood ulinaemia is quite prevalent in HIV infection, with and without
(Amlie-Lefond and Jubelt 2009; Braun 2009). VZV vasculitis can be HCV co-infection (Scotto 2006). Kawasaki-like syndromes in adult
either unifocal or multifocal. Transient ischaemic attacks, various patients, usually with severe immunosuppression and a high HIV
stroke syndromes including intranuclear opthalmoplegia, rapidly viral load, present with a constellation of signs and symptoms very
progressive multi-infarct dementia (Silver et al. 2012), and cerebral similar to Kawasaki’s disease in children without HIV, except gas-
aneurysms predisposing to dissection and/or subarachnoid and trointestinal complaints are more common and cervical lymphad-
intracerebral haemorrhages have been reported (Bhayani 2008). In enopathy less pronounced (Johnson et al. 2001; Stankovic 2007).
addition, VZV meningitis and myelitis are often concurrent with Presence of IgA plasma cells and paucity of complements within
VZV vasculitis (Chang 2009). vasculitic lesions unique to paediatric Kawasaki’s disease has been
Ischaemic or haemorrhagic infarction with grey or white mat- found in coronary arteries of a young HIV patient succumbing to
ter lesions, particularly at grey–white matter junctions, on MRI acute myocardial infarction (Johnson et al. 2003).
provides a clue to the diagnosis of VZV vasculitis (Gilden 2009). The appearance of palpable purpura in HIV-infected patients
Angiography demonstrates segmental narrowing of a mix of large may be due to HIV-associated leukocytoclastic vasculitis (Grayson
and small-vessel involvement, rather than only small or only large 2008), but also merits consideration of other viral infections, reac-
arteries, often with post-stenotic dilatation (Nagel 2010). CSF tions to antiviral drugs, or antimicrobial drugs, most commonly
574 SECTION 5 vasculitic diseases and syndromes and related disorders

sulfonamides. Other histological patterns of HIV skin manifesta- strokes in patients with HIV merit consideration of VZV foremost
tions include lymphocytic vasculitis, neutrophilic vasculitis with (Picard et al. 1997; Nagel 2010; Ortiz and Gutierrez 2011), followed
vascular thrombosis and intradermal suppuration, vasculitis by Toxoplasma gondii and herpes simplex virus (De Girolami et al.
with palisaded neutrophilic and granulomatous dermatitis, and 1990; Huang and Chou 1988). Infiltration of pulmonary vessels by
large-vessel vasculitis affecting subcutaneous vessels (Grayson Pneumocystis carinii/ jirovecii has been associated with necrotizing
2008). Microvascular inflammation may occur in the early, acute vasculitis in the pulmonary parenchyma (Liu et al. 1989).
stage of HIV infection, but lesions in the exanthem are character- HIV associated vasculitis generally responds favourably to glu-
ized by perivascular infiltrates of mononuclear cells without evi- cocorticoids (Cohen et al. 1993; Libman et al. 1995; Massari et al.
dence of leukocytoclasis (Balslev et al. 1990; McMillan et al. 1989). 1996), and immunosuppressants may be needed for refractory
Henoch–Schönlein purpura with dermal vascular deposits of IgA cases. Complete recovery of neurological function in patients with
has also been reported in patients with early as well as late-stage severe peripheral neuropathy has been reported when initial gluco-
HIV infection (Gherardi et al. 1993). Included in the spectrum of corticoid therapy is followed by antiretroviral therapy concurrent
leukocytoclastic vasculitis that occurs in HIV patients is erythema with plasmapheresis (Cohen et al. 1993). Appropriate antiviral and
elevatum diutinum (Grayson 2008). prophylaxis for opportunistic infections may prevent secondary
HIV produces a necrotizing arteritis by deposition or in situ vasculitis from these pathogens. Anti CD25 has been used to treat
formation of immune complexes (Gupta and Licorish 1984) or by CNS immune reconstitution inflammatory syndrome in one anec-
direct cell invasion and injury by HIV or opportunistic viruses. HIV dotal report (Nieuwhof 2006).
antigens have been detected in the vascular or perivascular cells
of inflamed muscular and neural vessels and it has been suggested Epstein–barr virus
that vasculitis may occur as a consequence of T-cell-mediated By virtue of its tropism for, and transforming properties of lym-
host responses to HIV replicating within them (Chad et al. 1990; phoid cells, and its ability to trigger typical immune responses,
Gherardi et al. 1993). Since perivascular macrophages constitute Epstein–Barr virus (EBV) may be associated with an unusual vari-
reservoirs for latent HIV, and macrophage activation by cytokines ety of vascular and perivascular syndromes. EBV may infect cells
(e.g. tumour necrosis factor) can promote HIV replication (Fauci comprising the vasculature, demonstrated by studies of patients
1988; Hickey and Kimura 1988), local cytokine release within the with granulomatous angiitis involving medium-sized and large
vasculature in response to either deposition of immune complexes arteries, where EBV DNA sequences have been found in intimal
or direct infection of intimal cells by opportunistic viruses may and medial tissues (Ban et al. 1999; Murakami et al. 1998). Immune
promote HIV replication within these perivascular macrophages, complex vasculitis involving smaller-calibre vessels and renal glo-
perpetuating immune activation. meruli has also been reported to occur during acute EBV infection
T-cell responses to perivascular foci of HIV replication may (Lande et al. 1998). Chronic EBV infection is life threatening and
account for angiocentric lymphoproliferative lesions characterized may present with hepatosplenomegaly and HSP-like features of pal-
by perivascular accumulation and transmural infiltration of CD8+ pable purpura, haematuria, and arthritis (Guissa et al. 2010).
T cells (Calabrese 1991; Anders et al. 1989; Katsetos et al. 1999). Studies have demonstrated associations between infection with
Monocytes and macrophages within the infiltrate are immunoreac- EBV and coronary artery aneurysms and other inflammatory
tive to the p24 HIV core protein (Katsetos et al. 1999). Although lesions of the aorta reminiscent of Kawasaki’s disease (Kanegane
arterial necrosis is not observed, an occlusive vasculopathy may et al. 1994; Nakagawa et al. 1996). A significantly greater prevalence
occur, resulting in CNS or visceral organ compromise (Calabrese of EBV DNA sequences has been demonstrated in circulating mon-
1991); the lesions may potentially evolve into malignant lymphoma onuclear cells of patients with KD and coronary artery aneurysms
(Anders et al. 1989). These angiocentric lesions are immunologi- than among age-matched controls (Kikuta et al. 1991; Kikuta et al.
cally and pathologically distinct from the diffuse infiltrative CD8 1992). In addition, EBV infection has been implicated in the patho-
lymphocytosis syndrome involving salivary glands and other vis- genesis of giant cell arteritis, Behçet’s disease, polyarteritis nodosa,
ceral organs, in which vasculitis has not been a described feature and Henoch–Schönlein purpura, supported by anecdotal evidence
(Itescu et al. 1990). (Belizna 2009; Caldeira et al. 2007).
The immunocompromised nature of late-stage HIV infection Transformation of EBV-infected B-cell lymphocytes and NK
mandates that opportunistic infection be considered when lesions cells may result in vascular syndromes associated with angiocen-
suggestive of vasculitis appear in visceral organs or the CNS. To tric lymphoid proliferation, as seen in lymphomatoid granuloma-
complicate matters, after initiation of highly active antiretroviral tosis (LG), which leads to tissue necrosis. Due to its propensity
therapy (HAART), immune reconstitution inflammatory syn- for multiorgan involvement, most frequently the lungs, and the
drome as a result of disproportionate immune reaction to subclini- histological features of granulomatous angiitis, LG often mimics
cal or opportunistic infections may manifest as cerebral vasculitis systemic vasculitis, most notably granulomatosus with polyangiitis
(Van der Ven et al. 2002; Newsome and Nath 2009; Anderson et al. (GPA) (Liebow et al. 1972). LG occurs most commonly in immu-
2010), superficial thrombophlebitis, and panniculitis (Alcaraz et al. nodeficiency states, such as AIDS, Wiskott–Aldrich syndrome, and
2010). As noted in Section Cytomegalovirus, CMV should be con- post-transplantation immunodeficiency (Jaffe and Wilson 1997).
sidered in the setting of cutaneous, pulmonary, enteric, renal, or LG has also been associated with autoimmune disorders includ-
CNS vasculitis. Although CNS vasculopathy associated with AIDS ing Sjögren’s syndrome, rheumatoid arthritis, and systemic lupus
has been described (Saravanan and Turnbull 2009), there were erythematosus, perhaps related to immunosuppression (Beste
no cases of true vasculitis (distinct from vasculopathy, which is et al. 2005).
perivascular lymphocytic cuffing) in the Edinburgh HIV Autopsy The vascular lesions in LG constitute mature CD3/CD4 (and less
Cohort with cerebral infarction (Connor et  al. 2000). Therefore, prominent CD3/CD8) positive T cells responding to EBV-infected
CHAPTER 41 miscellaneous forms of vasculitis 575

B cells in and around the vasculature. Atypical neoplastic X-linked lymphoproliferative disorder
EBV-positive B cells may be evident. Such findings also serve to
EBV infection or reactivation within endothelial cells may pre-
distinguish LG from angiocentric T-cell lymphomas, where there
cipitate a cytotoxic T-lymphocyte (CTL)-mediated vasculitis in
is evidence of clonal T-cell expansion (Guinee et  al. 1994). By
male patients with X-linked lymphoproliferative (XLP) disease,
immunohistochemical studies, the atypical EBV-positive B cells
an immunodeficiency characterized by fulminant EBV-associated
within the lesions are CD20 positive, occasionally CD30 posi-
infectious mononucleosis, lymphoproliferative disorder, and dys-
tive, and CD15 negative, and, from in situ hybridization studies,
gammaglobulinaemia (Dutz et  al. 2001; Nallasamy 2011). Fatal
EBV-encoded RNA is invariably present (Alinari et al. 2012). EBV
CNS vasculitis with or without systemic vasculitis, retinal necro-
DNA sequences are enriched in lesional B cells and are not typically
sis, and retinal vasculitis have been reported and prognosis is
found in the large numbers of non-clonally related T cells (Guinee
generally grim. The diagnosis can be confirmed by genetic analy-
et al. 1994).
sis for mutated SH2D1A/DSHP/SLAM-associated protein (SAP).
EBV transformation of NK cells resulting in multisystem vascu-
Mutations in the protein confer severe immune deficiency to EBV
lar infiltration by clonally expanded large granular lymphocytes has
infection and EBV-transformed cells (Sharifi et al. 2004), perhaps
also been reported (Gelb et al. 1994), further adding to the spec-
via defective polarization of the lytic machinery of NK cells and
trum of EBV-associated vasculopathies. As with post-transplant
cytotoxic T lymphocytes (Dupre et  al. 2005). However, 12% of
lymphoproliferative disorder, reduction of immunosuppression is
patients in a XLP registry had no demonstrable evidence of EBV
advisable. Data on success with use of anti-CD 20, rituximab, are
infection (Sumegi 2000; Talaat et  al. 2009). Allogeneic stem cell
emerging (Castrale et al. 2011).
transplantation, which can correct the underlying immune defect
in NK and CD8+ T cells, is curative.
Vasculitis associated Purine nucleoside phosphorylase deficiency
with immunodeficiencies Cerebral vasculitis is a reported complication in patients with defi-
Wiskott–aldrich syndrome ciency of purine nucleoside phosphorylase (PNP), a rare autosomal
recessive condition accounting for approximately 5% of cases of
Autoimmune manifestations, including vasculitis syndromes, are
severe combined immunodeficiency (Markert 1991). Deficiency of
sometimes observed as a consequence of immune dysregulation
PNP results in accumulation of deoxyguanosine and deoxyinosine,
in patients with genetic immunodeficiencies. The Wiskott–Aldrich
the dGTP and dITP metabolites of which are highly toxic to devel-
syndrome (WAS) is an X-linked disorder manifested by thrombo-
oping T cells. Affected individuals experience frequent dissemi-
cytopenia, severe immunodeficiency, and eczema. The molecular
nated bacterial, fungal, and viral infections. It is conceivable that
basis of the disorder has been shown to reside in mutations of
the cerebral vasculitis observed in the context of PNP deficiency
WAS Protein (WASP), a protein mediating the transduction of sig-
may be due to infection with varicella zoster virus.
nals from the cell membrane to the actin cytoskeleton in haema-
topoietic cells. Altered expression or function of WASP results in
functional defects in all leukocytes, including impaired matura- Vasculitis associated
tion of thymocytes, neutrophil phagocytosis, T-cell antigen recep-
tor (TCR)-triggered proliferative responses, and IL-2 production, with sarcoidosis
and TCR-triggered apoptotic responses. Functions of NK cells, Sarcoidosis is a multisystem inflammatory disease characterized by
nTreg cells, and B cells are also affected (Zhang et al. 1999; Catucci the presence of non-caseating granulomas in affected tissues. The
et al. 2012). cause of sarcoidosis has remained elusive. Variants in the class I and
In addition to autoimmune haemolytic anaemia and IgA II human leukocyte antigen (HLA) locus, susceptibility loci for
nephropathy, a variety of vascular lesions have been reported in annexin A11, tumour necrosis factor-alpha (TNF-α), and several
WAS patients, most commonly leukocytoclastic vasculitis, cerebral co-stimulatory molecules on antigen-presenting cells strongly
angiitis, and aortitis. Aortitis with aortic aneurysm and stenotic supports a genetic predisposition (Sweiss 2010). Microbial patho-
lesions of aortic branch vessels akin to Takayasu’s arteritis have gens (most notably cell wall deficient L forms of mycobacteria and
been reported in children as well as adults (Lau et  al. 1992; van Propionibacterium) may play a role in triggering the granulomatous
Son et al. 1995; Al Abrawi et al. 2008). Aneurysmal dilatation of inflammation and other immune abnormalities seen in this disor-
medium-calibre splanchnic and renal vessels with histological der (Almenoff et al. 1996; Newman et al. 1997; Saidha et al. 2012).
evidence of necrotizing arteritis has also been reported, as has Epithelioid cells surrounded by large numbers of CD4+ T lympho-
pulmonary vasculitis histologically indistinguishable from lym- cytes exhibiting a Th1 phenotype characterize the immunopathol-
phomatoid granulomatosis (Ilowite et al. 1986; McCluggage et al. ogy of sarcoid granulomas (Hunninghake and Crystal 1981; Saidha
1999). Corticosteroids and ciclosporin are reported to be of ben- et al. 2012). Although the prevalence of sarcoidosis is as high as
efit in managing WAS-associated vasculitis (Dupuis-Jerod et  al. 60/100 000 in Caucasians of northern European descent (Sartwell
2003; Catucci et  al. 2012). It has been speculated that impaired 1976) and 107/100 000 in African-American females (Rybicki et al.
host defence against viral pathogens known to engender vascu- 1997), vasculitis is a rare manifestation.
lar inflammation (see Section Viral-Induced Vasculitides) may Sarcoidosis vascular complications may involve large, medium,
account for some of the vasculitis complications observed in WAS or small-calibre vessels. Large-vessel granulomatous vasculitis in
(Ilowite et al. 1986; Catucci et al. 2012); antiviral therapy and/or sarcoidosis is very rare but when present usually affects the aortic
IVIg may therefore be indicated as part of the treatment strategy arch, histologically indistinguishable from that of giant cell arteritis
in such cases. (GCA) or Takayasu’s arteritis (Marcussen and Lund 1989; Weiler
576 SECTION 5 vasculitic diseases and syndromes and related disorders

et al. 2000; Bejerano et al. 2012; Rose et al. 1990). GCA-like dis- complications successfully managed with the equivalent of 1.0 mg/
ease involving large vessels occurring in association with systemic kg prednisone administered daily. Although the evidence is anec-
non-necrotizing granulomatous disease has been seen most com- dotal, weekly dosing with methotrexate, azathioprine (2 mg/kg),
monly in children but has also been reported in adults (Fernandes and mizoribine (a new purine synthesis inhibitor like mycophe-
et al. 2000). Complications include aortic dissection, saccular aor- nolate mofetil) (10–25 mg) have been used with success as a
tic aneurysm, myocardial infarction, stroke, and limb ischaemia steroid-sparing agents (Baughman and Lower 1977; Baughman and
(Faye-Petersen et al. 1991; Numata et al. 2005; Murai et al. 1986). Lower 1999; Diri et al. 1999; Kawakami and Soma 2011). Cytotoxic
The reported cases of large-vessel vasculitis in patients with granu- therapy with cyclophosphamide may be required for management
lomatous inflammation involving other organs emphasize the need of sarcoid vasculitis (Kwong et al. 1994). While there are emerg-
to consider sarcoidosis in the differential diagnosis of large-vessel ing data showing efficacy of anti-TNF agents for treatment of sar-
vasculitis. Moreover, the possibility of large-vessel involvement coidosis (Judson et al. 2008; Cruz et al. 2007; Bejerano et al. 2012),
should be entertained in adult as well as paediatric patients with these agents may paradoxically induce a sarcoid-like disease (Tong
established sarcoidosis who present with visceral organ or limb et al. 2012).
ischaemia.
Pulmonary vascular lesions in sarcoidosis are characterized by
granulomatous angiitis involving veins or arteries. Published series Cogan’s syndrome
of open lung and transbronchial biopsies of patients with estab- Cogan’s syndrome is one of the autoimmune inner ear diseases
lished sarcoidosis indicate venous involvement in the vast majority typified by interstitial keratitis, vestibular dysfunction, and senso-
of cases, combined venous and arterial involvement in one-quarter rineural hearing loss within 2 years of symptom onset. Diagnosis
to one-third of patients, and isolated arterial involvement in 8–11% of atypical Cogan’s syndrome can also be made when (1) the usual
(Rosen et  al. 1977; Takemura et  al. 1991). Necrotizing sarcoid pattern of vestibuloauditory impairment occurs with inflamma-
granulomatosis (NSG) is a form of pulmonary vasculitis that may tory eye disease other than interstitial keratitis; (2)  interstitial
be a variant of nodular pulmonary sarcoidosis (Churg et al. 1979; keratitis is associated with atypical audiovestibular symptoms
Leavitt and Fauci 1986; Tauber et al. 1999; Lazzarini et al. 2008). within 2 years; or (3) when there is a interval of more than 2 years
Histologically, these lesions are characterized by foci of confluent between the onset of ophthalmological and vestibuloauditory
granulomas with variable amounts of tissue necrosis and hyalini- symptoms (Haynes et al. 1980). The disease occurs most frequently
zation, and a granulomatous vasculitis. Although the histological in young adults of either sex, but may develop between the ages
appearance of granulomatous vasculitis associated with sarcoido- of 5 to 63 years. Although ocular and vestibuloauditory symptoms
sis may be similar to that seen in GPA (Wegener’s), the prognosis predominate, constitutional symptoms are present in 30% and the
is much more favourable, with patients improving either without majority have systemic involvement (90% in atypical Cogan’s syn-
therapy or with a limited course of glucocorticoids (Churg et al. drome versus 70% in classic Cogan’s syndrome) (Fidler and Jones
1979; Tauber et al. 1999). Differentiating features include greater 1989; Grasland et al. 2004).
degrees of liquefactive necrosis, abundance of eosinophils, and pau- The cause of Cogan’s syndrome is unknown. A  prodrome of
city of perivascular lymphocytes and plasma cells in GPA lesions, upper respiratory infection is common (Grasland et al. 2004; Gluth
and the absence of antineutrophil cytoplasmic antibodies and gran- et al. 2006). Chlamydia species, herpes virus, and Borrelia burgdor-
ulocytic vascular inflammation in NSG (DeRemee 1994; Saldana feri have been implicated in triggering the disease but definitive evi-
et al. 1977; Popper et al. 2003). Pulmonary hypertension may occur dence for infection with such organisms has not been established
as a complication of granulomatous vasculitis or perivascular fibro- (Haynes et al. 1980; Darougar et al. 1978; Ljunstrom et al. 1997; St
sis (Shigemitsu 2007; Nunes et al. 2006). Clair and McCallum 1999; Fox et al. 1990).
Neurological complications of necrotizing or non-necrotizing The initial presentation of Cogan’s syndrome may consist of ocu-
granulomatous inflammation of small-calibre arteries may include lar symptoms in the absence of vestibuloauditory symptoms or vice
peripheral mononeuritis with foot drop, myelopathy, chronic versa. However, the majority of patients develop symptoms refera-
inflammatory demyelinating neuropathy, sensory-motor neurop- ble to both organ systems within a year. Symptoms of the interstitial
athy, painful neuropathy, or cranial neuropathy (Diri et al. 1999; keratitis include blurred vision, photophobia, perilimbal erythema,
Caplan et al. 1983; Vital et al. 2008). Circulating immune complexes and ocular pain. Subepithelial keratitis in the periphery of the ante-
may engender a leukocytoclastic vasculitis with various cutaneous rior corneal stroma is an early finding of slit-lamp examination
manifestations, including erythema annulare centrifugum and pal- (Cobo and Haynes 1984). Deeper, granular corneal infiltrates are
pable purpura (Aractingi et al. 1993; Branford et al. 1982; Cecchi characteristic of more advanced lesions. Less common ocular man-
and Giomi 1999; García-Porrua et al. 1998; Johnston and Kennedy ifestations that may occur in the absence or presence of interstitial
1984). These cutaneous complications occur most commonly in keratitis include conjunctivitis, episcleritis, anterior or posterior
patients with early, acute disease, with erythema nodosum as an scleritis, and retinal vasculitis (Haynes et al. 1980; Shah et al. 1994;
accompanying feature. Other reported manifestations of sarcoido- Gluth et  al. 2006). Vestibular symptoms typically begin abruptly
sis attributed to inflammatory lesions of small vessels include cuta- with vertigo, ataxia, nausea, or vomiting and vestibular responses
neous ulcers and digital gangrene (Gibson et al. 1994; Petri et al. are seen on electronystagmometry and caloric testing. Hearing loss
1988; Takemura et al. 1997), posterior uveitis, retinitis, and cranial typically presents as poor speech discrimination, with audiomet-
neuropathies (Babin et al. 1984; Caplan et al. 1983; Sivakumar and ric testing revealing evidence of sensorineural hearing loss at mid-
Chee 1998) and myositis (Prayson 1999). dle and high frequencies. Changes in hearing often coincide with
Manifestations of sarcoidosis due to vascular inflammation flares of disease activity, and permanent severe hearing loss occurs
usually respond to limited courses of glucocorticoids, with most frequently, in up to two-thirds of cases (Haynes et al. 1980; Gluth
CHAPTER 41 miscellaneous forms of vasculitis 577

et al. 2006). Long-term sequelae of bilateral vestibular injury may and may demonstrate findings consistent with cerebral vasculitis.
be characterized by complete peripheral vestibular organ dysfunc- Occasionally, enhancement of vestibular and cochlear structures in
tion with ataxia and/or oscillopsia (Gluth et al. 2006). acute stages or obliterations of vestibular and labyrinth structures
Histopathological studies of corneal and vestibuloauditory in well-established disease may be observed using gadolinium MR
organs from patients with early disease are limited. Corneal tis- imaging or CT scan (Majoor et al. 1993; Casselman et al. 1994; St
sues from patients with interstitial keratitis are remarkable for infil- Clair and McCallum 1999). FDG-PET (positron emission tomog-
trates of plasma cells and lymphocytes into the deeper layers of the raphy with 2-deoxy-2-[18F] fluoro-D-glucose) studies may have
cornea, with variable degrees of scarring and neovascularization. a role in screening for subclinical large-vessel vasculitis (Murphy
Histological sections of temporal bones obtained at autopsy have 2009). Antibodies directed against the cornea and inner ears have
revealed infiltration of the spiral ligament with lymphocytes and been demonstrated in some cases (Helmchen 1999; Lunardi 2002;
plasma cells, demyelination and atrophy of the vestibular and coch- Bonaguri et  al. 2007)  but the utility of assays for these remains
lear nerves, and degeneration of the organ of Corti, cochlea, and doubtful (Murphy et al. 2009).
vestibular apparatus (Fisher and Hellstrom 1961; Schuknecht and Prompt use of glucocorticoids constitutes the mainstay of ther-
Nadol 1994). Evidence of vasculitis has not been seen; however, the apy for acute flares and recurrences of the ocular, vestibuloaudi-
shortest interval from disease onset to autopsy in cases reported to tory and systemic manifestations of Cogan’s syndrome (Haynes
date has been 4 years. et al. 1981; Murphy et al. 2009). Keratitis and anterior uveitis are
Vasculitis involving the aorta, aortic arch vessels, or large usually responsive to topical prednisolone, whereas posterior
muscular arteries occurs in approximately 10% of patients with scleritis and retinitis require treatment with oral glucocorticoids
Cogan’s syndrome (Haynes et al. 1980; Gluth et al. 2006). Aortitis (McCallum 1996; St Clair and McCallum 1999). Azathioprine,
may produce thoracoabdominal aneurysm (Tseng et  al. 1999; ciclosporin, tacrolimus, and/or cyclophosphamide may be used as
Weissen-Plenz et  al. 2010; Gasparovic et  al. 2011), and involve- steroid-sparing agents or for steroid-resistant ocular inflammation
ment of the proximal ascending aorta as well as the valve cusps or vestibuloauditory dysfunction (Allen et  al. 1990; Richardson
may result in aortic valve regurgitation necessitating valve replace- 1994; Roat et al. 1991; Touma 2007). An open-label study suggested
ment (Cochrane and Tatoulis 1991; Hammer et al. 1994; Livingston efficacy of MTX in immune-mediated inner ear disease, of which
et al. 1992). Narrowing of the major branches of the aorta at their three had Cogan’s syndrome (Matteson et  al. 2001). Anti-TNF
orifice similar to that observed in Takayasu’s arteritis may result in and rituximab have been used with isolated success (Ghadban
upper extremity claudication (Cochrane and Tatoulis 1991; Raza et al. 2008; Beccastrini et al. 2010; Orsoni 2010) but an open-label
et al. 1998). The histological appearance of lesions in the aorta and study of etanercept did not alleviate hearing loss in patients with
large muscular arteries is similar to that observed in GCA, with Cogan’s syndrome (Matteson et al. 2005). The efficacy of long-term
lymphocytes, plasma cells, and neutrophils infiltrating the vessel immunosuppression in averting total vestibular failure and hearing
wall, intimal proliferation, disruption of the internal elastic lamina, remains controversial. Given the success of cochlear implantation
and multinucleate giant cells (Cochrane and Tatoulis 1991; Fisher as a relatively safe and effective intervention for restoration of func-
and Hellstrom 1961; Gelfand et al. 1972; Livingston et al. 1992). tional hearing (Pasanisi et al. 2003; Wang et al. 2010; Degos 1966;
Matrix deterioration of the vascular wall may be related to overpro- Kohlmeier 1941), many experts favour limiting corticosteroid use
duction of GM-CSF by resident macrophages and smooth muscle to those with systemic disease or severe eye disease refractory to
cells (Weissen-Plenz et al. 2010). topical corticosteroids (Gluth et al. 2006; Migliori et al. 2009).
Arteritis of medium-calibre vessels occurs in a smaller percent- Patients with aortitis respond favourably to oral glucocorticoids.
age of patients. Coronary arteries may be compromised due to Prescribed in a manner similar to that for the management of GCA
arteritis or involvement of the coronary ostia (Livingston et  al. or Takayasu’s arteritis, initial treatment with prednisone (1 mg/
1992). Reported complications include limb ischaemia, mesenteric kg/day) can be followed by slow tapering of the dose. Published
insufficiency, renal artery rupture, and renal artery stenosis with experience of other immunosuppressive agents to manage aor-
hypertension (Allen et al. 1990; Bastug et al. 1997; Ho et al. 1999; titis or large-vessel vasculitis associated with Cogan’s syndrome
Raza et al. 1998; Thomas 1992; Vella et al. 1997). Typical of arteritis is limited to case reports. Concomitant therapy with daily cyclo-
affecting medium-calibre arteries, the walls of affected vessels are phosphamide, azathioprine, or ciclosporin, or low-dose weekly
infiltrated with lymphocytes, plasma cells, and a smaller number of methotrexate may be used to minimize the duration of high-dose
eosinophils and neutrophils (Fisher and Hellstrom 1961). Observed glucocorticoid therapy (Allen et al. 1990; Raza et al. 1998; Tseng
involvement of smaller-calibre arteries as well as veins (Fisher and et al. 1999). Methotrexate has also been used to maintain remis-
Hellstrom 1961) may account for the non-corneal ocular manifes- sion of the vascular lesions following initial therapy with gluco-
tations and other reported complications such as meninoencepha- corticoids and cyclophosphamide (Reinte et  al. 1996; Raza et  al.
litis, brain lacunar infarcts, inflammatory arthritis, and pericarditis 1998). Necrotizing vasculitis involving medium-calibre vessels may
(Cheson et al. 1976; Karni et al. 1991; Pinals 1978; Pysden 2009). be severe, if not life-threatening, and usually warrants the use of
The diagnosis of Cogan’s syndrome rests on both ocular inflamma- daily cyclophosphamide (2 mg/kg) concomitant with initiation of
tion and evidence of vestibuloauditory dysfunction. Differentiation glucocorticoids.
from Ménière’s disease is important (Migliori et al. 2009). Although
evidence of vasculitis involving other organ systems is not required,
Cogan’s syndrome should be considered in any patient presenting Degos’ disease
with manifestations of systemic vasculitis accompanied by visual or Initially described in independent case reports published in the
vestibuloauditory symptoms. Magnetic resonance imaging helps to early 1940s by Kohlmeier (1941) and Degos (1966), Degos’ dis-
rule out stroke and neoplasm (e.g. cerebellopontine angle tumours) ease is a rare occlusive vasculopathy characterized by multiple
578 SECTION 5 vasculitic diseases and syndromes and related disorders

(a) (b)

(c) (d)

Fig. 41.1 Cutaneous lesions of Degos’ disease syndrome. (a) Skin lesions on the thigh of an affected individual. (b) Close-up view of lesions demonstrating pale yellow
papules with violaceous margins; lesion on the left has overlying scale. (c) Photomicrograph of skin lesion demonstrating oedematous, relatively acellular dermis.
(d) Involved arteriole and vein in subjacent pannicular tissue demonstrating perivascular lymphoid infiltrates, proliferation of endothelium, and arteriolar thrombus.
(Courtesy of Dr M.K. Abele.)

cutaneous, mesenteric, and central nervous system (CNS) infarcts. The mesenteric and CNS infarcts of the malignant atrophic
Degos’ disease affects predominantly young and middle-aged papulosis variant are responsible for the poor prognosis and fatal
adults but can occur in young infants and in the elderly (Cabre outcomes, usually 1–2 years after the appearance of skin lesions.
et  al. 1974; Magrinat et  al. 1989; Soter et  al. 1982). Cases of Haemorrhages and perforations complicated by peritonitis com-
Caucasian, African-American, Asian Indian, and Japanese prise the major serious intestinal complications of the vasculopathy.
patients have been reported (Naylor et al. 1960; Ogawa et al. 1967; Typical findings at laparotomy and autopsy include white infarcts
Premalatha et  al. 1980). Two clinical variants of Degos’ disease ranging from 0.5 to 3 cm in size on the bowel serosa, most com-
have been described. A  benign variant confined to the skin is monly involving the small intestine (Degos 1979; Fruhwirth et al.
referred to as benign atropic papulosis, localized Degos’ disease, 1997; Su et  al. 1985; Gonzalez Valverde et  al. 2003; Chung et  al.
or cutaneous Degos’ disease. A malignant (usually fatal) variant 2009). Other reported visceral complications include fibrous peri-
involves the skin followed months to years later by systemic organ carditis, pulmonary infarcts, and pleural reactions (Degos 1979; Su
involvement (most commonly the gastrointestinal tract and cen- et  al. 1985). Neurological complications include seizures, ptosis,
tral nervous system), referred to as malignant atrophic papulo- diplopia, optic neuritis, loss of visual fields, stroke syndromes, and
sis, progressive arterial mesenteric vascular disease, disseminated polyradiculoneuropathy (Degos 1979; Su et al. 1985; Subbiah et al.
intestinal and cutaneous thromboangiitis, or systemic Degos’ dis- 1996). CNS infarcts and haemorrhages with intravascular thrombi,
ease (Magro 2011). There is a hereditary type in which familial often in the absence of demonstrable vasculitis, are characteristic
clustering has been observed (Kisch and Bruyzeel 1984; Newton findings at autopsy (Subbiah et al. 1996; Slaviero et al. 2009).
and Black 1984; Katz 1997). Cutaneous lesions grossly and histologically indistinguish-
Skin lesions associated with Degos’ disease are distinctive, typi- able from Degos’ lesions have also been demonstrated in patients
cally appearing in crops of painless, minimally elevated papules less with systemic lupus erythematosus (atrophie blanche), dermato-
than 5 mm in diameter (Figure 41.1). Initially gray, pale yellow, or myositis, polymyositis, systemic sclerosis, rheumatoid arthritis,
rose in colour, the papules turn white and develop a depressed cen- Wegener’s granulomatosis, and following streptococcal infection
tre, giving rise to the prototypic ‘porcelain drop’ lesions (Tzanck (Black and Hudson 1976; Dubin and Stawiski 1974; Durie et  al.
et al. 1948). Margins of the papules are initially erythematous or 1969; Torok et al. 1993; Olmos and Laugier 1977; Demitsu et al.
violaceous, later resolving into a filiform border. A  detachable 1997; Liu et al. 2005; Guhl et al. 2009; Pati et al. 2011). These lesions
scale frequently develops over the top of the papule (Degos 1979). did not significantly impact on the course of the underlying disease
The non-pruritic, non-ulcerative cutaneous lesions appear pre- (Black and Hudson 1976; Dubin and Stawiski 1974). The presence
dominantly on the trunk and proximal extremities (Degos 1979). of other manifestations of lupus as well as granular basal membrane
CHAPTER 41 miscellaneous forms of vasculitis 579

deposits of IgM, IgG, and C3 at the dermoepidermal junction dis- documented in the sera of patients with Degos’ disease (Basset
tinguish SLE from Degos’ disease (Black and Hudson 1976; Dubin et  al. 1969; Degos et  al. 1967), but such antibodies lack disease
and Stawiski 1974; Torok et al. 1993). The observation of Degos’ specificity as they are found in patients with malignant hyperten-
lesions in other autoimmune disorders lends support to the conten- sion or atherosclerosis, and are not uncommon in normal subjects
tion that Degos’ lesions probably represent a common end point to (Cerilli et  al. 1985; Gudbrandsson et  al. 1981; Kristensen et  al.
a variety of vascular insults associated with autoimmune disorders 1984). Furthermore, deposits of immunoglobulin have not been
(High et al. 2004). consistently demonstrated in lesions (with the exception of those
Skin biopsies are characterized by pauci-inflammatory throm- associated with SLE) and there is generally lack of efficacy of immu-
bogenic microangiopathy with endothelial cell injury; extracuta- nosuppressive agents in the treatment of this disease (Hall-Smith
neous organs demonstrate fibromucinous occlusive arteriopathy. 1964; Muller and Landry 1974; Pallensen and Rasmussen 1979; Su
The characteristic histological feature of the cutaneous lesion is et al. 1985).
described as an ‘inverted cone of necrobiosis’, with an atrophic The demonstration of abundant deposits of C5b-9 (membrano-
hyperkeratotic epidermis at the base, a thrombosed dermal ves- lytic attack complex) within the cutaneous vasculature, gastroin-
sel at the apex, and accumulation of acid mucopolysaccharide or testinal tract, and brain in patients with Degos’ disease has led to
mucin in the oedematous, acellular cone (Degos 1952; Soter et al. the more recent consideration of Degos’ disease as a C5b-9 medi-
1982; Su et al. 1985). Endothelial swelling and proliferation is seen ated endotheliopathy syndrome (Magro et al. 2011). The successful
in the affected dermal vessels, some of which contain platelet–fibrin treatment of Degos’ disease with eculizumab, a monoclonal anti-
thrombi within their lumens. Lymphocytic cell infiltrate associated body directed against the complement protein C5, has lent fur-
with interstitial mucin deposition in a superficial and deep perivas- ther credence to the role of C5b-9 membrane attack complex in
cular, periadnexal, and perineural pattern as well as endothelial cell pathogenesis. Rather then an autoimmune defect, Degos’ disease
necrosis and infiltration of the vessel wall by macrophages have may occur as a consequence of complement-mediated endothelial
been observed (Magro et al. 2011). Disruptions of the internal elas- injury, akin to paroxysmal nocturnal haemoglobinuria, a disorder
tic lamina, infiltration by neutrophils, and leukocytoclasis are not also managed effectively with eculizumab (Scheinfeld 2011; Magro
usually observed. et al. 2011).
The aetiology of Degos’ disease remains unclear. The presence The identified membrane attack complex deposits in Degos’
of thrombi in dermal and mesenteric vessels and the paucity of lesions are also associated with high expression of interferon-α and
vascular inflammation have prompted investigations to establish an interferon gene signature in peripheral blood mononuclear cells
Degos’ syndrome primarily as a coagulopathy (Daniel et al. 1982; (Magro et al. 2011), but the inciting trigger(s) and potential under-
Magrinat et  al. 1989; Paramo et  al. 1985). Levels of fibrinogen, lying genetic predispositions remain to be determined. The iden-
antithrombin III, plasminogen, von Willibrand factor, and protein tification by electron microscopy of ‘virus-like’ inclusions in the
C, activity of antithrombin III, plasminogen, and α2-antiplasmin, endothelial cytoplasm of affected endothelial cells have historically
platelet aggregation times, activated partial thromboplastin times, merited consideration of a viral aetiology or trigger for Degos’ dis-
and fibrin assembly kinetics in patients with Degos’ syndrome dis- ease (Bleehen 1977; Howard and Nishida 1969; Olmos and Laugier
ease have generally been normal (Durie et al. 1969; Magrinat et al. 1977). However, the observation of similar cytoplasmic inclu-
1989; Muller and Landry 1974; Sotrel et al. 1983). The failure to sions in tumour cells, experimentally induced wounds, and in skin
demonstrate evidence of a systemic coagulopathy has suggested lesions of patients with SLE or dermatomyositis suggests that the
that the platelet–fibrin thrombi observed in Degos’ lesions form inclusions in endothelial cells of Degos’ lesions might be induced
secondary to microvasculature injury. The concomitant occur- by factors other than viruses (Eady and Odland; 1975; Olmos
rence of antiphospholipid antibodies with Degos’ lesions has been et  al. 1979; Schaff et  al. 1972; Uzman et  al. 1971). Recombinant
reported but the overall prevalence of antiphospholipid antibodies α- or β-interferons have been shown to induce tubuloreticular
among individuals with Degos’ syndrome is not known (Englert structures in endothelial cells and epidermal keratinocytes, and
et al. 1984; Stephansson et al. 1991; Hohwy 2006). interferon-secreting cells such as activated T lymphocytes might
Originally viewed as an epiphenomenon of the dermal injury well be involved in the early events triggering these observed mod-
and necrobiosis, the role of mucin accumulation in the evolution ifications of the endothelium (Rich 1981; Rich and Owens 1982;
of Degos’ lesions has been reconsidered (Magrinat et  al. 1989). Magro et  al. 2011). The demonstration of parvovirus B19 RNA
Mucin-related glycoproteins with thrombogenic potential are con- expression in the endothelial cells and tissues in some cases of
sistently found in early, as well as in established, Degos’ lesions and Degos’ disease begs the question of the pathogenic significance of
their accumulation in or around dermal vessels may promote for- the virus (Magro et al. 2011).
mation of dermal thrombi (Kyung-Whan et al. 1980; Magrinat et al. Until recently, there has been no effective treatment for Dego’s
1989). Since activated T cells have been shown to induce mucin disease. The response to treatment with glucocorticoids and other
production by follicular keratinocytes (Reed 1981), mucin produc- immunosuppressive regimens has been uniformly disappointing
tion in response to T-cell cytokines constitutes a possible mecha- (Englert et al. 1984; Hall-Smith 1969; Strole et al. 1967; Su et al.
nistic link between immunological activation in and around the 1985; Subbiah et al. 1996). The ubiquitous appearance of luminal
vasculature and the thrombosis with infarction observed in Degos’ platelet-fibrin thrombi in Degos’ lesions has prompted the use of
lesions. antiplatelet agents and anticoagulants to avoid thrombotic com-
A role for antibody-mediated alterations in triggering the plications. Cutaneous lesions may improve during treatment with
endothelial abnormalities observed in Degos’ disease has not been aspirin and dipyridamole (Su et  al. 1985)  but systemic/visceral
substantiated (Magro et al. 2011). Antibodies with specificity for organ manifestations do not respond to antiplatelet agents, fibrino-
arterial endothelium or other vessel wall components have been lytics, or plasma exchange. While the use of heparin appears to have
580 SECTION 5 vasculitic diseases and syndromes and related disorders

been of benefit in some cases, there is lack of evidence to support a Table 41.2 The similarities and differences between systemic lupus
role for chronic anticoagulation. The demonstrated effectiveness of erythematosus and hypocomplementaemic urticarial vasculitis
eculizumab in the malignant atrophic papulosis variant of Degos’ (Aydogan et al. 2006)
disease remains promising (Magro et al. 2011).
Systemic lupus Hypocomplementaemic
erythematosus (%) urticarial vasculitis (%)
Urticarial vasculitis
Urticaria <10 100
Chronic or recurrent urticaria is common, but only approximately
Arthralgia/ arthritis 100 95
10% of patients with chronic urticarial lesions have associated
vasculitis (Wisnieski 2000). The lesions in urticarial vasculitis dif- Angioedema <5 72
fer from common urticaria, typically of greater duration lasting Chronic obstructive lung Rare 65
between 6 and 72 hours and upon resolution often leaving residual disease
pigmentation or ecchymosis. Urticarial vasculitis lesions are often
Eye involvement 61 15
characterized by pain or burning rather than pruritus. Patients
with urticarial vasculitis may have signs and symptoms of systemic Renal involvement 36–50 50
disease, including arthralgia, fever, gastrointestinal pain, lymphad- Pericarditis/ pericardial 30 17
enopathy, and abnormal urine sediment. Urticarial vasculitis may effusion
be a manifestation of systemic lupus erythematosus (SLE), Sjögren’s Anti-C1q antibody 35 100
syndrome, or mixed cryoglobulinaemia.
Urticarial vasculitis is traditionally categorized according to serum Low C1q 22–47 100
complement levels (Dincy et al. 2008). Normocomplementaemic ANA 95 61–71
urticarial vasculitis (NUV) is typically a self-limited form of vascu-
litis without internal organ involvement that is generally idiopathic
and benign. Hypocomplementaemic urticarial vasculitis syndrome
(HUV) is a rare and a potential severe form of urticarial vasculi- cogent argument has been proffered that HUV represents a sub-
tis associated with persistent urticarial skin lesions, IgG antibodies set of SLE (Davis et  al. 1998; Trendelenburg et  al. 1999). With
reactive to the C1q subunit of the first component of complement, the possible exception of obstructive pulmonary disease, most of
and markedly low serum levels of C1q (Zeiss et al. 1980). In addi- the extracutaneous features observed in patients with HUV are
tion to low serum levels of C1q, patients with HUV may have not uncommonly clinical features observed in patients with SLE
modest to marked depletion of C4, C2, and/or C3. Levels of C1r, (Table 41.2). In addition to the presence of antibodies to C1q and
C1s, the fifth through ninth components of complement (C5–C9), a continuous band of immunoreactants along the epidermal base-
as well as factor B and factor D are generally normal (Zeiss et al. ment membrane, other immunological features shared with SLE
1980). Other clinical features include severe angioedema, laryngeal include a high prevalence of antiendothelial cell antibodies and
oedema, ocular inflammation, arthritis, obstructive lung disease, shared anti-C1q IgG antibody binding properties (D’Cruz et  al.
recurrent abdominal pain, and glomerulonephritis (Wisnieski et al. 1995; Martensson et al. 1992; Nurnberg et al. 1995; Wisnieski and
1995; Zeiss et al. 1980). Skin biopsy specimens of the urticarial and Jones 1992a; Wisnieski and Jones 1992b). In both HUV and SLE,
purpuric lesions characteristically reveal leukocytoclastic vasculitis the serum reactivity to C1q is primarily to either surface-bound
with granular deposition of C3, C1q, and immunoglobulin in the or complexed C1q rather than to native (fluid phase) C1q. Despite
basement membrane zone (Davis et al. 1998; Sanchez et al. 1982). common clinical expression and immunological features, differ-
The arthritis in patients with HUV is typically non-erosive and ences in the epitope binding specificities of C1q-reactive IgG in
deforming, but may evolve into a Jaccoud’s-like arthropathy with HUV and SLE sera suggest there may nonetheless be some differ-
reversible subluxation of the metacarpophalangeal joints (Palazzo ences in the pathogenetic mechanisms operative in SLE and HUV
et al. 1993; Sturgess and Littlejohn 1988). Mesangial, and membra- (Martensson et al. 1992).
nous, as well as membranoproliferative lesions have been noted on The vasculitis in HUV responds favourably to glucocorticoids,
renal biopsy (Fortson et al. 1986; Kobayashi et al. 1994; Wisnieski but many patients have an excellent response to dapsone (Eiser
et al. 1995). A high prevalence of obstructive pulmonary disease, et al. 1997; Fortson et al. 1986; Nurnberg et al. 1995). Combinations
causing substantial morbidity and mortality, has been reported in of dapsone (100 mg/day) and pentoxifylline (300 mg t.i.d.) have
HUV (Wisnieski et  al. 1995). Although deaths from respiratory been well tolerated and successful in resistant cases of HUV, as
failure have been observed primarily in patients who smoke ciga- have hydroxychloroquine (Lopez et  al. 1984; Nurnberg et  al.
rettes, significant obstructive lung disease has also been reported 1995), cyclophosphamide, ciclosporin, azathioprine, mycopheno-
in non-smoking patients (Eiser et al. 1997). Lung biopsies reveal late mofetil, and methotrexate (Wisnieski et al. 1995; Balsam et al.
severe emphysema with immunoglobulin deposition and thick- 2008; Schwartz et  al. 1982; Mehregan et  al. 1992). B-cell deple-
ening of blood vessel walls (Fortson et al. 1986). Less commonly tion with rituximab has been used with success in SLE-associated
reported clinical features include pericarditis, regurgitant val- urticarial vasculitis (Saigal et al. 2003). Potentially life-threatening
vular lesions, and vasculitis involving medium to large-calibre lung involvement with progressive COPD requires specific treat-
arteries (Babajanians et al. 1991; Palazzo et al. 1993; Sturgess and ment for COPD as well as immunosuppression. Plasmapheresis
Littlejohn 1988). and IVIg have been proposed as treatment for rapidly progres-
Because the presence of antibodies to C1q and low levels of C1q, sive glomerulonephritis (Balsam et al. 2008). Since HUV may be
C2, C3, and C4 are frequently observed in patients with SLE, a a presenting feature of SLE (Trendelenburg et al. 1999), expectant
CHAPTER 41 miscellaneous forms of vasculitis 581

observation for evolving SLE is prudent; the development of sig- of IgA (Chow et al. 1996). Fibrotic lesions may be less responsive
nificant proteinuria, renal dysfunction, or other visceral manifesta- to therapy.
tions should prompt consideration of and assessment for evolving
lupus, with management directed toward treatment of SLE as clini-
cally indicated. Familial mediterranean fever
Vasculitis is an important but not readily appreciated feature of
familial Mediterranean fever (FMF), an autosomal recessive disease
Erythema elevatum diutinum characterized by recurrent self-limited attacks of fever accompanied
Erythema elevatum diutinum (EED) is a rare, chronic, localized by peritonitis, pleuritis, and arthritis. The majority of FMF cases
form of cutaneous leukocytoclastic vasculitis. It is most frequently are attributable to mutations in the MEFV gene, presumed to be
observed in patients who are systemically ill with other defined an important regulator of inflammation. FMF-related MEFV gene
disorders. Haematological disorders and malignancies, particularly mutations favour TH1 polarization with enhanced production of
IgA monoclonal gammopathy, were reported in 30% of EED cases inflammatory cytokines, including IL-1β, IL-6, IL-18, IL-33, TNF-α,
and constitute the most commonly associated disorders (Chow and interferon-γ (Simsek et al. 2007; Aypar et al. 2003), which are
et  al. 1996; Hatzitolios et  al. 2008). Other disease associations capable of inducing endothelial dysfunction, leukocyte infiltration,
include solid malignancies and infectious diseases, especially HIV and fibrinoid necrosis within the arterial wall (Akdogan et al. 2006;
infection (Dronda et al. 1996; Muratori et al. 1999; Soni et al. 1998; Pober and Sessa 2007). FMF patients are at increased risk for the
Burnett and Burgin 2003). Rheumatological and autoimmune development of vasculitis syndromes, including Henoch–Schönlein
disorders reported in association with EED include rheumatoid purpura, polyarteritis nodosa (PAN), protracted febrile myalgia,
arthritis, relapsing polychondritis, systemic lupus erythematosus, Behçet’s disease, and leukocytoclastic vasculitis.
Sjögren’s syndrome, and juvenile idiopathic arthritis (Shimizu IgAV (HSP) is considered to be the most common vasculitis asso-
et al. 2008; Golmia et al. 2008; Chen et al. 2002; Collier et al. 1990; ciated with FMF. The FMF-Turkish Study Group reported that FMF
Hancox et al. 2004). EED may also be a cutaneous manifestation of patients experience attacks of HSP at a significantly higher rate than
celiac disease (Tasanen et al. 1997). that observed in the general Turkish population, with no observed
The lesions of EED usually appear as bilateral symmetric plaques differences in the clinical manifestations between FMF-associated
or nodules, often over extensor surfaces of joints or buttock, less Henoch–Schönlein purpura and classical Henoch–Schönlein pur-
commonly on the trunk, retroauricular region, palms, and soles. pura (Ozdogan et al. 1997; Tunca et al. 2005). PAN was also found
Lesions may initially appear purpuric and soft, later develop- more frequently (approximately 1%) in FMF patients than in the
ing a pink or yellow hue and become hard and firm, reflecting a general population (0.006%) (Ozen et  al. 2001; Ozdogan et  al.
tendency toward fibrosis. On visual inspection, EED lesions may 1997; Tekin et al. 2000; Tunca et al. 2005). Relative to PAN patients
be difficult to distinguish clinically from dermatofibroma, granu- without FMF, FMF patients with PAN tended to be younger, had
loma annulare, granuloma faciale, or dermatitis herpetiformis. In greater severity of myalgia, had less frequent HBsAg positivity, no
HIV patients, lesions are more nodular, palms and soles are more peripheral nervous system involvement, and more central nervous
frequently involved, and may be similar to those of Kaposi’s sar- system involvement (Hatemi et al. 2004; Ozen et al. 2001). Perirenal
coma or bacillary angiomatosis (Suarez et  al. 1998; LeBoit and haematoma has been reported frequently in patients with FMF and
Cockerell 1993). PAN; its presence in a patient with FMF accordingly warrants care-
Biopsy of EED lesions reveals distinct histopathological features ful evaluation for arteritis (Glikson et al. 1989).
consistent with an immune complex vasculitis. Early, acute lesions Protracted febrile myalgia (PFM) experienced by patients with
are characterized by dermal infiltrates of neutrophils and eosino- FMF is thought to be caused by vasculitis. Symptoms include
phils with perivascular nuclear dust and fibrin within aggregates weight loss, hypertension, severe crippling myalgia, high fever,
of spindled cells (LeBoit and Cockerell 1993; Sangueza et al. 1977; abdominal pain, abdominal pain, diarrhoea, joint pain, and tran-
Wilkinson et al. 1992). More established, chronic lesions are char- sient purpura (Bircan and Usluer 2008; Tufan and Demir 2010).
acterized by a dense fibrosis with proliferation of fibroblasts and Laboratory features include high sedimentation rate, hyperglob-
myofibroblasts; lymphocytes, macrophages, and histiocytes con- ulinaemia, normal creatine phosphokinase, and non-specific
taining cholesterol clefts and lipid droplets comprise the remainder myopathic changes on electromyography (Langevitz et al. 1994).
of the dermal infiltrate (Kanitakis et al. 1993; Lee et al. 1989). Because both FMF and Behçet’s disease are common in Turkey and
Dapsone has been reported to be effective in the treatment of other certain geographic areas, these disorders share genetic fea-
EED, inducing regression of lesions in patients without known dis- tures and it is still debatable whether mutations in the MEFV gene
ease associations as well as in patients with HIV or gammopathies confer an increased risk of Behçet’s disease.
(Dronda et al. 1996; Katz et al. 1977; LeBoit and Cockerell 1993; Unlike many other acute manifestations of FMF, vasculitis in
Suarez et al. 1998). Dapsone may also be of benefit in the treatment FMF responds favourably to glucocorticoids. In FMF-associated
of EED associated with celiac disease but adherence to a gluten-free PAN, glucocorticoids plus cyclophosphamide were efficacious and
diet may be required for a sustained remission (Rodriguez-Serna long-term immunosuppressive treatment was not necessary (Ozen
et al. 1993; Tasanen et al. 1997). If patients fail to respond to dap- et al. 2001).
sone, other therapeutic options employed with success include
colchicine, niacinamide plus tetracycline, and systemic corticos-
teroids (Rosa et  al. 2012). Plasma exchange in conjunction with Hyperimmunoglobulinaemia D
chlorambucil or cyclophosphamide has been successful in treating Among the patients with periodic fever syndromes, and distinct
patients with EED lesions associated with exceedingly high levels from those with familial Mediterranean fever or Still’s disease,
582 SECTION 5 vasculitic diseases and syndromes and related disorders

are those with significantly elevated levels of IgD who experi-


ence recurrent attacks of fever, arthritis, and cutaneous vasculitis.
Hyperimmunoglobulin D syndrome (HIDS) is found primarily in
Europe, particularly in France and the Netherlands, and is asso-
ciated with an autosomal recessive mutation in the mevalonate
kinase (MVK) gene on chromosome 12q24. The resulting enzyme
deficiency (activity of 5–15 % of normal) results in overproduction
of IL-1β. Levels of serum IgD in affected patients often exceed 100
U/ml with the median reported serum IgD level of 400 U/mL, and
have been shown to correlate with percentages of circulating B cells
expressing surface IgD (Kumano et  al. 1997). However, normal
serum levels of IgD do not exclude the diagnosis, as serum IgD lev-
els may be normal in up to 22% of affected patients (van der Hilst
et al. 2010). Circulating immune complexes containing IgD have
been observed in patients during and between attacks, and IgD
deposits have been identified by immunofluorescence in affected
skin lesions (Boom et al. 1990).
In reported HIDS case series (van der Hilst et al. 2010; Drenth
et al. 1994b), the median time of first attack is at the age of 6 months,
with immunizations not uncommonly precipitating attacks. High
fever, arthritis, lymphadenopathy, splenomegaly, diarrhoea, nau-
sea, and vomiting are common presenting manifestations. Skin
manifestations accompany attacks of fever in more than two-thirds
of patients, most commonly maculopapular rash but urticarial
rash, purpura, and tender erythematous nodules have also been
reported. Oral aphthous ulcers with or without accompanying gen-
ital ulcerations were reported in 48.5% of patients; some patients Fig. 41.2 Histopathology of IgG4-related thoracic aortic aneurysm. Reprinted
may be misdiagnosed with Behçet’s before the diagnosis of HIDS from The Lancet, 373(9673):1494, Khosroshahi, A., Stone, J.R., Pratt, D.S.,
Deshpande, V., Stone, J.H., Painless jaundice with serial multi-organ dysfunction,
is established (Drenth et al. 1994a). The arthritis is non-deforming,
Copyright (2009), with permission from Elsevier.
most commonly affecting the ankles, knees and wrists. About 3%
of patients develop amyloidosis, an incidence considerably lower
than that found in the other autoinflammatory syndromes (van der
Hilst et al. 2005). IgG4RD may account for up to 9% of thoracic aortitis, with a
Etanercept and anakinra are reported to be successful in the reported prevalence comparable to aortitis due to giant cell arteritis,
treatment of HIDS (Bodar et al. 2005; Takada et al. 2003). In rand- Takayasu’s arteritis, or rheumatoid disease (Stone et al. 2010). The
omized controlled trials using simvastatin (Simon et al. 2004) and ascending aorta and the aortic arch are most commonly involved,
thalidomide (Drenth et al. 2001) for treating hyperimmunoglob- usually presenting as an aneurysm, with chronic aortic dissection
ulin D the frequency and severity of attacks were not decreased; less common (Stone et al. 2011). Pathology reveals marked lym-
colchicine and other immunosuppressive agents also seem to be phoplasmacytic infiltrate with storiform fibrosis and lack of granu-
ineffective in treating hyperimmunoglobulin D syndrome (van der loma formation, as seen in giant cell arteritis, Takayasu’s arteritis, or
Hilst and Frenkel 2010). rheumatoid aortitis (Khosroshahi et al. 2009) (Figure 41.2). There
are also several reports of IgG4-related inflammatory abdominal
aortic aneurysm (Kasashima et al. 2008). As compared with tho-
IgG4-related disease racic aneurysms, abdominal aneurysms in IgG4RD appear to be
Immunoglobulin G (IgG) 4-related disease (IgG4RD) is a recently associated with less severe disease, lower fractions of infiltrat-
described clinical disease entity characterized by elevated serum ing plasma cells staining for IgG4, lower serum IgG4 levels, and
IgG4 in association with a variety of clinical manifestations, includ- less frequent involvement of the aortic media (Stone et al. 2011).
ing aortitis. The disorder usually presents subacutely, and most However, ruptured abdominal aortic aneurysm occurring with
patients are not constitutionally ill (Stone et al. 2012). Multiorgan IgG4RD periaortitis with IgG4-positive cells invading the intima
involvement can be present at the time of diagnosis but can also has been reported (Qian et al. 2009).
evolve slowly in an additive fashion over months to years in patients Up to 30% of patients with histologically confirmed IgG4RD
having disease initially confined for many years to a single organ. have normal serum IgG4 concentrations (Sah and Chari 2011),
Two common findings in IgG4RD are tumefactive lesions and and the majority of patients with serum elevations in IgG4 have
allergic manifestations such as atopy, asthma, eczema, and mod- been shown to have disorders other than IgG4RD; the diagnosis of
est peripheral blood eosinophilia (Umehara et al. 2012). There are IgG4RD is therefore best confirmed with biopsy of involved tissues/
very few data relevant to the global incidence and prevalence of organs. Because biopsy of involved aortic tissue is often not feasi-
IgG4RD. The majority of affected patients are men over the age of ble, recognition of the disease involving other organ systems may
50 (Stone et al. 2012); IgG4RD should therefore be considered as prove useful in establishing the diagnosis of IgG4RD-associated
a potential cause of aortitis or aortic dissection in this population. aortitis. Other common clinical features of IgG4RD include
CHAPTER 41 miscellaneous forms of vasculitis 583

involvement of the orbital and periorbital tissues (Plaza et al. 2011), organ or muscle involvement occurs less frequently (Fain et  al.
sinuses (Deshpande et al. 2011), salivary and lacrimal glands (Sato 2007; Sanchez-Guerrero 1990). The most common pathologi-
et al. 2008; Geyer and Deshpande 2011), lymph nodes (Sato et al. cal lesions reported in association with both haematological and
2010), pituitary gland and meninges (Leporati et al. 2011), thyroid solid malignancies is leukocytoclastic vasculitis, accounting for
(Dahlgren et  al. 2010), lung parenchyma and pleura (Zen et  al. 50–60% of paraneoplastic vasculitis (Fain et  al. 2007; Kurzrock
2009a), pancreas (Okazaki et al. 2011), bile ducts (Ghazale et al. et  al. 1995a; Solans-Laque 2008). While the clinical features of
2008), and kidney with either interstitial nephritis (Saeki et  al. malignancy-associated vasculitis are similar to those observed in
2007)  or membranous nephropathy (Saeki et  al. 2009). IgG4RD vasculitis patients without malignancy (Fain et al. 2007; Agha et al.
can cause idiopathic retroperitoneal fibrosis with or without aor- 2012), the index of suspicion for malignancy should be heightened
titis, usually presenting as a localized inflammatory mass involv- in older patients presenting with cutaneous vasculitis or vasculitic
ing the abdominal aorta, ureter, and the kidney rather than diffuse neuropathy without other identifiable cause or in patients who have
retroperitoneal involvement. IgG4RD-associated retroperitoneal a suboptimal response to treatments usually effective for vasculitis
fibrosis has thus far been exclusively reported in males (Zen et al. (Bachmeyer et al. 2005; Hutson and Hoffman 2000).
2009b; Stone et al. 2010), with most of the patients having multior- The true prevalence of vasculitis in cancer patients remains
gan disease with involvement of the pancreas, salivary glands, and/ unknown, but evidence of malignancy is reported in up to 2–5%
or lymph nodes (Stone et al. 2011). of individuals presenting with vasculitis (Sanchez-Guerrero et al.
Glucocorticoid therapy is the cornerstone of initial therapy 1990; Garcia-Porrua and Gonzalez-Gay 1998). Myelodysplastic
in IgG4RD. In general, glucocorticoid treatment is indicated in syndrome, haematological malignancies (notably hairy cell leukae-
patients who have autoimmune pancreatitis with either back pain, mia, chronic myelogenous leukaemia, and multiple myeloma) are
obstructive jaundice, or abdominal pain and patients who have reported most commonly, followed by lymphoid malignancies and
symptomatic extrapancreatic lesions or aortitis (Kamisawa et  al. solid tumours (Faine et al. 2007; Garcia-Porrua and Gonzalez-Gay
2010). A consensus statement from 17 referral centres in Japan sug- 1998). Vasculitis frequency during cancer has been estimated at 1
gested 0.6 mg/kg/day of prednisolone for 2–4 weeks, then tapering in 1800 for haematological malignancies and 1 in 80 800 for solid
the dose to 5 mg/day within 3–6 months, and continuing a dose tumours (Greer et al. 1988). Non-small cell lung cancer is the most
of 2.5–5 mg/day for up to 3 years (Kamisawa et al. 2010). Another prevalent among associated solid tumours (Kurzrock et al. 1994).
proposed regimen is a starting dose of 40 mg/day of prednisone, The pathogenesis of malignancy-associated vasculitis is probably
initiation of a taper within 7 weeks, with discontinuation by 11 multifactorial. Postulated mechanisms include:  (1)  direct effects
weeks; however, more than half of patients who received this regi- of tumour cells (notably hairy cells) on vascular walls; (2) release
men experienced disease relapses within a median time of 3 months of tumour angiogenic factors and/or cytokines, which induce
(Ghazale et  al. 2008). Despite long-term glucocorticoid mainte- endothelial damage and increase vascular permeability, inflam-
nance, one-quarter of patients experience disease flares while on mation, and fibrosis; and (3) increased cellular turnover leading to
low-dose prednisolone (Kamisawa et  al. 2009). Monitoring of generation of autoantibodies with formation of immune complexes
IgG4 concentrations can reflect relapse in some patients but some of tumour-associated antigen/ antibodies (Park and Ranganathan
patients have relapses despite normal IgG4 levels (Stone et al. 2012). 2011; Klima and Wadell 1984; Gabriel et al. 1986). Attribution of
Immunosuppressive agents have been employed in patients with vasculitis to a paraneoplastic syndrome should be undertaken with
refractory or recurrent disease. Azathioprine (2.0–2.5 mg/kg/day) caution (Park and Ranganathan 2011). Because there are other
or mycophenolate mofetil (750 mg twice daily) has been used in possible aetiologies of vasculitis in cancer patients, such as drug
patients who have failed prednisone taper. Rituximab has been used reaction, infection, or cryoglobulins, these other causes should
with success to treat refractory cases, with demonstrated clinical be considered and appropriately addressed. In one review of 23
and serological responses (Khosroshahi et al. 2012). Interestingly, patients with haematological malignancies with skin biopsy confir-
IgG4 appeared to be the only IgG subclass significantly affected mation of vasculitis, 39% of the patients had vasculitis due to other
by rituximab (Khosroshahi et  al. 2010). Bortezomib, a protea- identifiable causes (Bachmeyer et al. 2005).
some inhibitor approved for treatment of multiple myeloma, was
reported to successfully treat IgG4RD presenting with recurrent Vasculitis associated with
lung disease and orbital pseudotumour (Khan et al. 2010). haematological malignancy
Myelodysplastic, myeloproliferative, and lymphoproliferative dis-
orders account for up to two-thirds of all paraneoplastic vasculitis
Vasculitis associated with malignancy (Fain et al. 2007). Relative to vasculitis associated with other malig-
The association of vasculitis with malignancy may include malig- nancies, vasculitis associated with myelodysplastic syndrome is
nancies masquerading as vasculitis, such as occurs with angi- associated with a poorer prognosis, more renal involvement, more
ocentric T-cell lymphomas, or true paraneoplastic syndromes steroid dependence, and a lower likelihood of achieving remission
whereby vasculitis triggers immune responses culminating in (Fain et  al. 2007). Leukocytoclastic vasculitis is seen most com-
vascular inflammation. Although the latter syndromes are uncom- monly (Farrell et al. 1999), usually presenting as palpable purpura,
mon, it is important to recognize that in such cases the appear- although rarely erythema elevatum diutunum (EED) and urticarial
ance of vasculitis not infrequently antedates the appearance of vasculitis (UV) are reported (Liu et al. 2009; Strickland and Ware
other manifestations of malignancy. The vast majority of paraneo- 1995; Wilson et al. 2002). However, EED and UV are more com-
plastic vasculitis syndromes involve small vessels with palpable monly caused by drug exposures than by paraneoplastic processes.
purpura, urticaria, maculopapular lesions, erythema multiforme, Several observations pertinent to vasculitis associated with hae-
arthralgia, and neuropathy as predominant manifestations; visceral matological malignancy other than paraprotein-secreting B cell
584 SECTION 5 vasculitic diseases and syndromes and related disorders

lymphomas are noteworthy: (1) low complement levels are typically et  al. 1999). In reported case series, vasculitis occurs most often
not present; (2) skin lesions typically stain negative for immuno- prior to or concurrent with the diagnosis or relapse of tumour
globulins and complement products; and (3) cutaneous vasculitis (Kurzrock and Cohen 1995a).
in myelodysplastic syndromes or chronic leukaemia may antedate Small-vessel vasculitis involving skin, muscle, and/or periph-
bone marrow disease but frequently diminishes with disease pro- eral nerves and IgAV (Henoch–Schönlein purpura) are the most
gression (Sanchez-Guerrero et al. 1990; Greer et al. 1988; Enright commonly reported forms of solid tumour-associated vasculitis
and Miller 1997; Longley et al. 1986). (Kurzrock and Cohen 1995a; Pertuiset et al. 2000; Fain et al. 2007;
Paraneoplastic cryoglobulinaemia accounts for 15% of all cryo- Podjasek et  al. 2012). In one series of biopsy-confirmed nerve–
globulinaemia; however, it rarely causes vasculitis (Dammacco muscle vasculitis, malignancy was found in 14% of cases (Vincent
et  al. 2001). Cryoglobulinaemia can be found in Waldenstrom’s et al. 1986). Malignancy-associated vasculitis targeting nerve and
macroglobulinaemia, multiple myeloma, non-Hodgkin’s lym- or muscle is reported most often among patients with prostate
phoma, and chronic lymphocytic leukaemia, usually in association and lung cancer (small cell and adenocarcinoma) (Kurzrock et al.
with monoclonal or type 1 cryoglobulins. However, the type 1 cryo- 1994). Patients may present with either symmetric polyneuropa-
globulins seen with these disorders typically do not form immune thy or mononeuropathy multiplex; the lower extremities are more
complexes that activate the classical complement pathway, and are frequently affected and protein in the cerebrospinal fluid is usu-
only rarely associated with vasculitis (Bayer-Garner and Smoller ally elevated (Oh et al. 1991). Cutaneous or other systemic organ
2003; Cem Ar 2005; Kim et  al. 2011). More commonly, patients involvement is often absent (Matsumuro et al. 1994).
with type 1 cryoglobulinaemia present with hyperviscosity syn- Approximately one-third of adult cases of IgAV are associ-
drome associated with acrocyanosis, digital gangrene, and strokes ated with solid tumour malignancy, particularly carcinomas of
(Tedeschi et al. 2007). the lungs, urogenital, and gastrointestinal tract (Zurada et  al.
Polyarteritis nodosa has been reported in patients with chronic 2006). When compared with non-malignancy associated IgAV,
myeloid leukaemia and myelodysplastic syndrome (MDS) malignancy-associated IgAV has more renal involvement;
(Hamidou et al. 2001), but the most frequent paraneoplastic asso- biopsy-confirmed proliferative nephritis and necrotizing vasculitis
ciation is with hairy cell leukaemia, with arteritis most commonly affecting the kidney has been demonstrated in some patients with
developing following splenectomy or infections (Hasler et al. 1995; solid tumour-associated IgAV (Kurzrock and Cohen et al. 1995a;
Elkon et al. 1979; Komadina and Houk 1989). Polyarteritis nodosa Zurada et al. 2006). Risk factors for paraneoplastic HSP are older
in association with malignancy resembles classic polyarteritis age and male gender (Pertuiset et  al. 2000; Zurada et  al. 2006;
nodosa in terms of age of onset, male predominance, presence of Mitsui et al. 2009). Given the distinctly uncommon occurrence of
visceral involvement, and elevation of sedimentation rate. However, this syndrome in adults, its occurrence in men older than 40 years
reported distinguishing features of polyarteritis in this population of age without evidence of antecedent infection should create sus-
include: (1) the presence of peripheral aneurysms (in radial, ulnar, picion for malignancy (Pertuiset et al. 2000).
brachial, temporal, occipital, dorsal pedal arteries), which are rarely Paraneoplastic ANCA-associated vasculitis is quite uncommon
reported in classic polyarteritis nodosa, and (2) a lower occurrence and accounts for less than 5% of all paraneoplastic vasculitis. There
of ANA, rheumatoid factor, hepatitis B surface antigen, circulating is a report of 14 patients with granulomatous with polyangiitis and
immune complexes, and decreased complement levels than seen in a concurrent malignancy, although a temporal association was
classic polyarteritis nodosa (Komadina and Houk 1989). observed in only four of the reported patients (Tatsis et al. 1999).
IgAV (Henoch–Schönlein purpura) may be a presenting mani- Churg–Strauss vasculitis has been reported in a patient with meta-
festation of myelodyspastic syndrome (Blanco et al. 1997b). Up to static melanoma (Cupps and Fauci 1982). Microscopic polyangiitis
one-third of reported occurrences of malignancy-associated IgAV has been reported to be a paraneoplastic syndrome of gastrodu-
are associated with myelodysplastic syndrome or other myelo- odenal cancer (Abe et al. 2011). In a review of 200 patients with
proliferative disorders, including non-Hodgkin and Hodgkin ANCA-associated vasculitis, 20 patients had co-existing malig-
lymphoma and multiple myeloma (Pertuiset et al. 2000). Primary nancy (diagnosed within 6  months); solid tumours were most
angiitis of the central nervous system has been reported in asso- common, notably colon cancer, breast cancer, and gynaecological
ciation with both Hodgkin’s and non-Hodgkin’s lymphoma (Rosen cancer (Pankhurst et  al. 2004). Patients with ANCA-associated
et al. 2000; Sheehy et al. 2003). Temporal arteritis has been reported vasculitis have an increased risk of cancer as compared with
in a patient with lymphoma and hairy cell leukaemia (Webster et al. aged-matched controls (relative risk 6.0, 95% confidence interval
1986). Takayasu’s arteritis was recently reported in patients with 3.7–9.4); however, positive ANCA is not predictive of malignancy
MDS (Cohen et al. 2011). (Pankhurst et al. 2004).
Although reported more commonly in association with MDS
Vasculitis associated with solid tumours and haematological malignancies, there are reports of polyarte-
The most common solid tumours associated with vasculitis involve ritis nodosa occurring in the context of various solid tumours
the genitourinary tract (prostate and renal cell carcinoma), lungs including bladder, colorectal, gastric, lung, and liver (Hayem et al.
(small-cell and non-small cell lung carcinoma), gastrointestinal 1997; Paajanen et al. 1995). Temporal arteritis has been described
tract (colon cancer), and breast (Solans-Laque et  al. 2008; Agha in patients with renal cell carcinoma, brain, lung, uterine cancer,
et al. 2012). Vasculitis has also been reported in association with and adenocarcinoma of unknown origin (Liozon et  al. 2006).
tumours of the nasopharynx, ovary, stomach, bile duct, small However, the observed incidence of cancer in patients with tem-
bowel, ovary, and endometrium (Blanco et al. 1997a; Hayem et al. poral arteritis does not appear to be different from that observed
1997; Matsumuro et al. 1994; Mita et al. 1999; Miyachi et al. 1987; in an age-matched population (Kurzrock and Cohen et al. 1995a;
Oh et al. 1991; Pertuiset et al. 2000; Ponge et al. 1998; Stashower Myklebust et al. 2002).
CHAPTER 41 miscellaneous forms of vasculitis 585

Treatment Almenoff, P.L., Johnson, A., Lesser, M., and Mattman, L.H. (1996). Growth
of acid fast L forms from the blood of patients with sarcoidosis. Thorax,
Paraneoplastic vasculitis may resolve with immunosuppressive 51, 530–3.
agents alone (Podjasek et  al. 2012)  or following effective treat- Alvarez-Lafuente, R., Fernandez-Gutierrez, B., and Lamas, J.R., et al. (2005).
ment of the underlying malignancy (Solans-Laque et  al. 2008). Human parvovirus B19, varicella zoster virus, and human herpes virus
However, successful and sustained resolution of vasculitis usu- 6 in temporal artery biopsy specimens of patients with giant cell arte-
ally requires treatment of the underlying malignancy (Kurzrock ritis:  analysis with quantitative real time polymerase chain reaction.
and Cohen 1993; Kurzrock and Cohen 1995a; Greer and Panush Annals of the Rheumatic Diseases, 64, 780–2.
1988). In the majority of patients with vasculitis associated with Amlie-Lefond, C. and Jubelt, B. (2009). Neurologic manifestations of vari-
cella zoster virus infections. Current Neurology and Neuroscience
myelodyspastic syndrome, responses to prednisone were favour-
Reports, 9, 430–4.
able (Enright and Miller 1997; Enright et al. 1995; Longley et al.
Anders, K.H., Latta, H., Chang, B.S., et al. (1989). Lymphomatoid granulo-
1986). Polyarteritis associated with hairy cell leukaemia responds matosis and malignant lymphoma of the central nervous system in the
favourably to glucocorticoids and cyclophosphamide, and suc- acquired immuno-deficiency syndrome. Human Pathology, 20, 326–34.
cess has been reported following treatment with interferon alone Anderson, A.M., Fountain, J.A., Green, S.B., Bloom, S.A., and Palmore, M.P.
(Carpenter and West 1994). Prednisone is usually started at mod- (2010). Human immunodeficiency virus-associated cytomegalovirus
erate to high dose (20 mg/day to 1 mg/kg/day) and the majority of infection with multiple small vessel cerebral infarcts in the setting of
patients have some response within 1 week. Immunosuppressive early immune reconstitution. Journal of Neurovirology, 16, 179–84.
therapies such as cyclophosphamide have been used in the man- Aractingi, S., Cadranel, J., Milleron, B., Saiag, P., Malepart, M.J., and
agement of arteritis occurring in the context of other malignan- Dubertret, L. (1993). Sarcoidosis associated with leucocytoclastic vascu-
litis. A case report and review of the literature. Dermatology, 187, 50–3.
cies but such interventions should be done with caution since
Aydogan, K., Karadogan, S.K., Adim, S.B., and Tunali, S. (2006).
their use could enhance tumour growth by suppression of tumour Hypocomplementemic urticarial vasculitis: a rare presentation of sys-
immunosurveillance. Flares/ relapses of vasculitis may her- temic lupus erythematosus. International Journal of Dermatology, 45,
ald tumour recurrence or progression of underlying malignan- 1057–61.
cies (Solans-Laque et  al. 2008); for patients in whom vasculitis Aypar, E., Ozen, S., Okur, H., Kutluk, T., Besbas, N., and Bakkaloglu, A.
emerges after initial successful treatment of malignancy, a thor- (2003). Th1 polarization in familial Mediterranean fever. Journal of
ough worked up for recurrence is needed before institution of Rheumatology, 30, 2011–3.
immunosuppressive therapy. Babajanians, A., Chung-Park, M., and Wisnieski, J.J. (1991). Recurrent peri-
carditis and cardiac tamponade in a patient with hypocomplementemic
References urticarial vasculitis syndrome. Journal of Rheumatology, 18, 752–5.
Babin, R., Liu, C., and Aschenbrener, C. (1984). Histopathology of neuro-
Abe, H., Momose, S., Takeuchi, T. (2011). Microscopic polyangitis compli-
sensory deafness in sarcoidosis. Annals of Otology Rhinology and
cating double carcinoma of the stomach and duodenum: improvement
Laryngology, 93, 389–93.
after the resection of these carcinomas. Rheumatology International,
Bachmeyer, C., Wetterwald, E., and Aractingi, S. (2005). Cutaneous vasculitis
31, 105–8.
in the course of hematologic malignancies. Dermatology, 210, 8–14.
Agha, A., Bateman, H., Sterrett, A., and Valeriano-Marcet, J. (2012).
Myelodysplasia and malignancy-associated vasculitis. Current Baek, W., Lee, S.G., Kim, Y.S., Kim, J.H., Jun, J.B., and Kim, H.Y. (2012). Fatal
Rheumatology Reports, 14, 526–31. varicella-zoster virus vasculopathy associated with adalimumab therapy.
Archives of Neurology, 25, 1–4.
Aguilar-Bernier, M., Bassas-Vila, J., Torné-Gutiérrez, J.I., Martínez-García,
G., Aragoneses-Fraile, H., and Miranda-Romero, A. (2006). Presence of Balsam, L., Karim, M., Miller, F., and Rubinstein, S. (2008). Crescentic glo-
perineuritis in a case of papular purpuric gloves and socks syndrome merulonephritis associated with hypocomplementemic urticarial vascu-
associated with mononeuritis multiplex attributable to B19 parvovirus. litis syndrome. American Journal of Kidney Diseases, 52, 1168–73.
Journal of the American Academy of Dermatology, 54, 896–9. Balslev, E., Thomsen, H.K., and Weissman, K. (1990). Histopathology of
Akdogan, A., Calguneri, M., Yavuz, B., Arslan, E.B., Kalyoncu, U., Sahiner, acute human immunodeficiency virus exanthem. Journal of Clinical
L., Karadag, O., Ertenli, I., Kiraz, S., Aytemir, K., Akata, D., Tokgozoglu, Pathology, 43, 201–2.
L., and Oto, A. (2006). Are familial Mediterranean fever (FMF) patients Ban, S., Goto, Y., Kamada, K., Takahama, M., Watanabe, H., Iwahori, T., and
at increased risk for atherosclerosis? Impaired endothelial function Takeuchi, H. (1999). Systemic granulomatous arteritis associated with
and increased intima media thickness are found in FMF. Journal of the Epstein-Barr virus infection. Virchows Archives, 434, 249–54.
American College of Cardiology, 48, 2351–3. Baskan, E.B., Yilmaz, E., Saricaoglu, H., Alkan, G., Ercan, I., Mistik, R., Adim,
Al Abrawi, S., Fouillet-Desjonqueres, M., David, L., Barral, X., Cochat, P., and S.B., Goral, G., Dilek, K., and Tunali, S. (2007). Detection of parvovirus
Cimaz, R. (2008). Takayasu arteritis in children. Pediatric Rheumatology B19 DNA in the lesional skin of patients with Behçet’s disease. Clinical
Online Journal, 6, 17. and Experimental Dermatology, 32, 186–90.
Alcaraz, I., Revelles, J.M., Camacho, D., Haro, R., Fernández-Aceñero, M.J., Basset, A., Kurtz, D., Bergoend, H., et al. (1969). Paulose atrophiante maligne
Fernández-Guerrero, M., and Requena, L. (2010). Superficial thrombo- de Degos avec atteinte du systeme nerveux central et presence d’anticorps
phlebitis:  A  new clinical manifestation of the immune reconstitution antiartere. Bulletin de la Societe de Dermatologie et de Syphiligraphie,
inflammatory syndrome in a patient with HIV infection. American 76, 333–4.
Journal of Dermatopathology, 32, 846–9. Bastug, D.E., Dominic, A., Ortiz, O., DiBartolomeo, A.G., Kotzan, J.M., and
Alinari, L., Pant, S., McNamara, K., Kalmar, J.R., Marsh, W., Allen, C.M., and Abraham, F.M. (1997). Popliteal artery thrombosis in a patient with Cogan
Baiocchi, R.A. (2012). Lymphomatoid granulomatosis presenting with syndrome: treatment with thrombolysis and percutaneous transluminal
gingival involvement in an immune competent elderly male. Head and angioplasty. Cardiovascular and Interventional Radiology, 20, 57–9.
Neck Pathology, 6, 496–501. Baughman, R.P. and Lower, E.E. (1977). Alternatives to corticosteroids in
Allen, N.B., Cox, C.C., Cobo, M., Kisslo, J., Jacobs, M.R., McCallum, R.M., the treatment of sarcoidosis. Sarcoidosis, Vasculitis, and Diffuse Lung
and Haynes, B.F. (1990). Use of immunosuppressive agents in the treat- Diseases, 14, 121–30.
ment of severe ocular and vascular manifestations of Cogan’s syndrome. Baughman, R.P. and Lower, E.E. (1999). A clinical approach to the use of
American Journal of Medicine, 88, 296–301. methotrexate for sarcoidosis. Thorax, 54, 742–46.
586 SECTION 5 vasculitic diseases and syndromes and related disorders

Bayer-Garner, I.B. and Smoller, B.R. (2003). Leukocytoclastic (small vessel) Caplan, L., Corbett, J., Goodwin, J., Thomas, C., Shenker, D., and Schatz, N.
vasculitis in multiple myeloma. Clinical and Experimental Dermatology, (1983). Neuro-ophthalmologic signs in the angiitic form of neurosar-
28, 521–4. coidosis. Neurology, 33, 1130–5.
Beccastrini, E., Emmi, G., Squatrito, D., Vannucchi, P., and Emmi, L. (2010). Cardwell, M.M. and Martinez, J.J. (2009). The Sca2 autotransporter protein
Infliximab and Cogan’s syndrome. Clinical Otolaryngology, 35, 441–2. from Rickettsia conorii is sufficient to mediate adherence to and inva-
Bejerano, C., Blanco, R., González-Vela, C., Agüero, R., Carril, J.M., and sion of cultured mammalian cells. Infection and Immunity, 77, 5272–80.
González-Gay, M.A. (2012). Refractory polymyalgia rheumatica as pre- Carpenter, M.T. and West, S.G. (1994). Polyarteritis nodosa in hairy cell leu-
senting manifestation of large-vessel vasculitis associated to sarcoido- kemia: treatment with interferon-alpha. Journal of Rheumatology, 21,
sis. Successful response to adalimumab. Clinical and Experimental 1150–2.
Rheumatology, 30 (Suppl. 70), S94–7. Casselman, J.W., Majoor, M.H.J.M., and Albers, F.W. (1994). MR of the
Belizna, C.C. (2009). Infection and vasculitis. Rheumatology, 48, 475–82. inner ear in patients with Cogan’s syndrome. American Journal of
Beste, L.A., Ansari-Lari, A., Borowitz, M., and Flynn, J.A. (2005). Neuroradiology, 15, 131–8.
Lymphadenopathy, cough, and fever in a 51-year-old woman with sys- Castrale, C., El Haggan, W., Chapon, F., Reman, O., Lobbedez, T., Ryckelynck,
temic lupus erythematosus: case report of lymphomatoid granulomato- J.P., and Hurault de Ligny, B. (2011). Lymphomatoid granulomatosis
sis. Arthritis and Rheumatism, 53, 621–4. treated successfully with rituximab in a renal transplant patient. Journal
Bhayani, N., Ranade, P., Clark, N.M., and McGuinn, M. (2008). of Transplantation, 2011, 865957.
Varicella-zoster virus and cerebral aneurysm: case report and review of Catucci, M., Castiello, M.C., Pala, F., Bosticardo, M., and Villa, A. (2012).
the literature. Clinical Infectious Diseases, 47, e1–3. Autoimmunity in wiskott-Aldrich syndrome:  an unsolved enigma.
Bilenchi, R., De Paola, M., Poggiali, S., Acciai, S., Feci, L., Sansica, P., and Frontiers in Immunology, 3, 209.
Fimiani, M. (2012). Papular-purpuric ‘gloves and socks’ syndrome. Cecchi, R. and Giomi, A. (1999). Annular vasculitis in association with sar-
Giornale Italiano di Dermatologia e Venereologia, 147, 119–21. coidosis. Journal of Dermatology, 26, 334–6.
Bircan, Z. and Usluer, H. (2008). Protracted febrile myalgia mimicking pol- Cem Ar, M., Soysal, T., Hatemi, G., Salihoglu, A., Yazici, H., and Ulku, B.
yarteritis nodosa. Journal of Clinical Rheumatology, 14, 222–5. (2005). Successful management of cryoglobulinemia-induced leukocy-
Black, M.M. and Hudson, P.M. (1976). Atrophie blanche lesions closely toclastic vasculitis with thalidomide in a patient with multiple myeloma.
resembling malignant atrophic papulosis (Degos’ disease) in systemic Annals of Hematology, 84, 609–13.
lupus erythematosus. British Journal of Dermatology, 95, 649–52. Cerilli, J., Brasile, L., and Karmady, A. (1985). Role of the vascular endothelial
Blanco, R., González-Gay, M.A., Ibanez, D., Alba, C., and Perez de Llano, L.A. cell antigen system in the etiology of atherosclerosis. Annals of Surgery,
(1997a). Henoch-Schönlein purpura as a clinical presentation of small 202, 329–34.
cell lung cancer. Clinical and Experimental Rheumatology, 15, 545–7. Chad, D.A., Smith, T.W., Blumenfeld, A., Fairchild, P.G., and de Girolami,
Blanco, R., González-Gay, M.A., Ibanez, D., Lopez-Viana, A., Ferran, C., U. (1990). Human immunodeficiency virus (HIV)-associated myopa-
Regueira, A., and Gonzalez-Vela, C. (1997b). Henoch-Schönlein pur- thy: immunocytochemcal identification of an HIV antigen (gp41). in
pura as clinical presentation of a myelodysplastic syndrome. Clinical muscle macrophages. Annals of Neurology, 28, 579–82.
Rheumatology, 16, 626–8. Chang, C.C., McLean, C., Vujovic, O., Jenney, A.J., Short, M., Lyon, S., Storey,
Bleehen, S.S. (1977). Intra-endothelial tubular aggregates in malignant atrophic E, and Lewin, S.R. (2009). Fatal acute varicella-zoster virus hemorrhagic
papulosis (Degos’ disease). Clinical Experimental Dermatology, 2, 73–4. meningomyelitis with necrotizing vasculitis in an HIV-infected patient.
Bodar, E.J., van der Hilst, J.C., Drenth, J.P., van der Meer, J.W., and Simon, Clinical Infectious Diseases, 48, 372–3.
A. (2005). Effect of etanercept and anakinra on inflammatory attacks in Chang, G.Y. (2012). Clinical recognition of varicella zoster virus vasculopa-
the hyper-IgD syndrome: introducing a vaccination provocation model. thy. European Neurology, 67, 297–9.
Netherlands Journal of Medicine, 63, 260–4. Chanin, J.M., Marcos, L.A., Thompson, B.M., Yusen, R.D., Dunne, W.M.
Bonaguri, C., Orsoni, J.G., Zavota, L., Monica, C., Russo, A., Pellistri, Jr, Warren, D.K., and Santos, C.A. (2011). Methicillin-resistant
I., Rubino, P., Giovannelli, L., Manzotti, F., and Piazza, F. (2007). Staphylococcus aureus USA300 clone as a cause of Lemierre’s syndrome.
Anti-68 kDa antibodies in autoimmune sensorineural hearing loss. Journal of Clinical Microbiology, 49, 2063–6.
Autoimmunity, 40, 73–8. Chen, K.R., Toyohara, A., Suzuki, A., and Miyakawa S. (2002). Clinical
Boom, B.W., Daha, M.R., Vermeer, B.J., and van der Meer, J.W. (1990). IgD and histopathological spectrum of cutaneous vasculitis in rheumatoid
immune complex vasculitis in a patient with hyperimmunoglobuline- arthritis. British Journal of Dermatology, 147, 905–13.
mia D and periodic fever. Archives of Dermatology, 126, 1621–4. Cheson, B.D., Bluming, A.Z., and Alroy, J. (1976). Cogan’s syndrome: a sys-
Branford, W.A., Farr, P.M., and Porter, D.I. (1982). Annular vasculitis temic vasculitis. American Journal of Medicine, 60, 549–55.
of the head and neck in a patient with sarcoidosis. British Journal of Chirinos, J.A., Lichtstein, D.M., Garcia, J., and Tamariz, L.J. (2002). The evo-
Dermatology, 106, 713–6. lution of Lemierre syndrome: report of 2 cases and review of the litera-
Braun, K.P., Bulder, M.M., Chabrier, S., Kirkham, F.J., Uiterwaal, C.S., Tardieu, ture. Medicine (Baltimore), 81, 458–65.
M., and Sébire, G. (2009). The course and outcome of unilateral intracra- Chow, R.K., Benny, W.B., Coupe, R.L., Dodd, W.A., and Ongley, R.C. (1996).
nial arteriopathy in 79 children with ischaemic stroke. Brain, 132, 544–57. Erythema elevatum diutinum associated with IgA paraproteinemia
Bronze, M.S., Shirwany, A., Corbett, C., and Schaberg, D.R. (1999). Infectious successfully controlled with intermittent plasma exchange. Archives of
aortitis:  anuncommon manifestation of infection with Streptococcus Dermatology, 132, 1360–4.
pneumoniae. American Journal of Medicine, 107, 627–30. Chung, H.C., Trendell-Smith, N.J., Yeung, C.K., and Mok, M.Y. (2009).
Burnett, P.E. and Burgin, S. (2003). Erythema elevatum diutinum. Degos’ disease: a rare condition simulating rheumatic diseases. Clinical
Dermatology Online Journal, 9, 37. Rheumatology, 28, 861–3.
Cabre, J., Noguer-Debray, S., Bosch-Castane, J., et al. (1974). Papulose atro- Churg, A., Carrington, C.B., and Gupta, R. (1979). Necrotizing sarcoid gran-
phiante maligne de Degos chez un nourrisson. Bulletin de la Societe de ulomatosis. Chest, 76, 406–13.
Dermatologie et de Syphiligraphie, 81, 652–3. Cioc, A., Sedmak, D., Nuovo, G., Dawood, M., Smart, G., and Magro, C.
Calabrese, L.H. (1991). Vasculitis and infection with the human immunode- (2002). Parvovirus B19 associated adult Henoch Schönlein purpura.
ficiency virus. Rheumatic Diseases Clinic of North America, 17, 131–47. Journal of Cutaneous Pathology, 29, 602–7.
Caldeira, T., Meireles, C., Cunha, F., Valbuena, C., Aparício, J., and Ribeiro, A. Clifton, D.R., Goss, R.A., Sahni, S.K., van Antwerp D., Baggs, R.B., Marder,
(2007). Systemic polyarteritis nodosa associated with acute Epstein-Barr V.J., Silverman, D.J., and Sporn, L.A. (1998). NF-κB-dependent inhi-
virus infection. Clinical Rheumatology, 26, 1733–5. bition of apoptosis is essential for host cell survival during Rickettsia
CHAPTER 41 miscellaneous forms of vasculitis 587

rickettsii infection. Proceedings of the National Academy of Sciences of Degos, R. (1966). Malignant atrophic papulosis: A fatal cutaneo-intestinal
the USA, 95, 4646–51. syndrome. British Journal of Dermatology, 66, 304–7.
Cobo, M. and Haynes, B.F. (1984). Early corneal findings in Cogan’s syn- Degos, R. (1979). Malignant atrophic papulosis. British Journal of
drome. Ophthalmology, 91, 903–7. Dermatology, 100, 21–35.
Cochrane, A.D. and Tatoulis, J. (1991). Cogan’s syndrome with aortitis, aor- Degos, R., Lortat-Jacob, E., Daniel, F., et  al. (1967). Papulose atrophiante
tic regurgitation, and aortic arch vessel stenoses. Annals of Thoracic Maligne avec presence d’anticorps anti-artere. Bulletin de la Societe de
Surgery, 52, 1166–7. Dermatologie et de Syphiligraphie, 74, 715–17.
Cohen, M.J., Shyman, A., Klein, M., Ben-Yehuda, A., Rubinow, A., Or, R., and Demitsu, T., Kakurai, M., Murata, S., Kiyosawa, T., and Yaoita, H. (1997).
Goldschmidt, N. (2011). Large vessel (Takayasu’s) arteritis in a patient Degos’ disease associated with rheumatoid arthritis. Journal of
with myelodysplastic syndrome:  is there a common pathogenesis? Dermatology, 24, 488–90.
Clinical Lymphoma Myeloma and Leukemia, 11, 60–3. DeRemee, R.A. (1994). Sarcoidosis and Wegener’s granulomatosis: a com-
Cohen, P., Guillevin, L., Gayraud, M., Lhote, F., Jarrousse, B., Leon, A., and parative analysis. Sarcoidosis, 11, 7–18.
Gherardi, R. (1993). Successful treatment of HIV-related vasculitis with Deshpande, V., Khosroshahi, A., Nielsen, G.P., Hamilos, D.L., and Stone, J.H.
peripheral neuropathy with short-term steroids followed by the associa- (2011). Eosinophilic angiocentric fibrosis is a form of IgG4-related sys-
tion of zidovudine and plasmapheresis. Transfusion Science, 14, 383–9. temic disease. American Journal of Surgical Pathology, 35, 701–6.
Cohen, P.S., O’Brien, T.F., Schoenbaum, S.C., and Medeiros, A.A. (1978). Dincy, C.V., George, R., Jacob, M., Mathai, E., Pulimood, S., and Eapen, E.P.
The risk of endothelial infection in adults with salmonella bacteraemia. (2008). Clinicopathologic profile of normocomplementemic and hypoc-
Annals of Internal Medicine, 89, 931–2. omplementemic urticarial vasculitis: a study from South India. Journal
Collier, P.M., Neill, S.M., Branfoot, A.C., and Staughton R.C. (1990). of the European Academy of Dermatology and Venereology, 22, 789–94.
Erythema elevatum diutinum—a solitary lesion in a patient with rheu- Diri, E., Espinoza, C.G., and Espinoza, L.R. (1999). Spinal cord granuloma-
matoid arthritis. Clinical and Experimental Dermatology, 15, 394–5. tous vasculitis: an unusual clinical presentation of sarcoidosis. Journal of
Connor, M.D., Lammie, G.A., Bell, J.E., Warlow, C.P., Simmonds, P., and Rheumatology, 26, 1408–10.
Brettle, R.D. (2000). Cerebral infarction in adult AIDS patients: obser- Drenth, J.P., Boom, B.W., Toonstra, J., and Van der Meer, J.W. (1994a).
vations from the Edinburgh HIV autopsy cohort. Stroke, 31, 2117–26. Cutaneous manifestations and histologic findings in the hyperimmuno-
Cook, A.M. and Christopoulos, D. (1989). Rupture of a non-aneuysmal sal- globulinemia D syndrome. Archives of Dermatology, 130, 59–65.
monellainfected aorta. Clinical Radiology, 40, 605–6. Drenth, J.P., Haagsma, C.J., and van der Meer, J.W. (1994b).
Cook, C.H. and Trgovcich, J. (2011). Cytomegalovirus reactivation in criti- Hyperimmunoglobulinemia D and periodic fever syndrome. The clini-
cally ill immunocompetent hosts: a decade of progress and remaining cal spectrum in a series of 50 patients. Medicine (Baltimore), 73, 133–44.
challenges. Antiviral Research, 90, 151–9. Drenth, J.P., Vonk, A.G., Simon, A., Powell, R., and van der Meer, J.W. (2001).
Cooper, R.J. (2008). Infection and temporal arteritis:  a PCR-based study Limited efficacy of thalidomide in the treatment of febrile attacks of the
to detect pathogens in temporal artery biopsy specimens. Journal of hyper-IgD and periodic fever syndrome: a randomized, double-blind,
Medical Virology, 80, 501–5. placebo-controlled trial. Journal of Pharmacology and Experimental
Cruz, B.A., Reis, D.D., Araujo, C.A.; Minas Gerais Vasculitis Study Group Therapeutics, 298, 1221–6.
(2007). Refractory retinal vasculitis due to sarcoidosis successfully Dronda, F., Gonzalez-Lopez, A., Lecona, M., and Barros, C. (1996). Erythema
treated with infliximab. Rheumatology International, 27, 1181–3. elevatum diutinum in human immunodeficiency virus-infected
Cupps, T.R. and Fauci, A.S. (1982). Neoplasm and systemic vasculitis: a case patients—report of a case and review of the literature. Clinical and
report. Arthritis and Rheumatism, 25, 475–6. Experimental Dermatology, 21, 222–5.
Dahlgren, M., Khosroshahi, A., Nielsen, G.P., Deshpande, V., and Stone, J.H. Dubin, H.V. and Stawiski, M.A. (1974). Systemic lupus erythematosus resem-
(2010). Riedel’s thyroiditis and multifocal fibrosclerosis are part of the bling malignant atrophic papulosis. Archives of Internal Medicine,
IgG4-related systemic disease spectrum. Arthritis Care and Research 134, 321–3.
(Hoboken), 62, 1312–8. Dupre, L., Andolfi, G., Tangye, S.G., Clementi, R., Locatelli, F., Arico, M.,
Dammacco, F., Sansonno, D., Piccoli, C., Tucci, F.A., and Racanelli V. Aiuti, A., and Roncarolo, M.G. (2005). SAP controls the cytolytic activ-
(2001). The cryoglobulins:  an overview. European Journal of Clinical ity of CD8+ T cells against EBV-infected cells. Blood, 105, 4383–9.
Investigation, 31, 628–38. Dupuis-Girod, S., Medioni, J., Haddad, E., Quartier, P., Cavazzana-Calvo, M.,
Daniel, F., Foix, C., and Gray, J.M., et al. (1982). Malignant atrophic papulo- Le Deist, F., de Saint Basile, G., Delaunay, J., Schwarz, K., Casanova, J. L.,
sis with insufficiency of blood fibrinolysis. Annals of Dermatology and Blanche, S., and Fischer, A. (2003). Autoimmunity in Wiskott-Aldrich
Venereology, 109, 763–4. syndrome: risk factors, clinical features, and outcome in a single-center
Darougar, S., John, A.C., Viswalingam, M., Cornell, L., and Jones, B.R. (1978). cohort of 55 patients. Pediatrics 111, e622–7.
Isolation of Chlamydia psittaci from a patient with interstitial keratitis Durie, B.G.M., Stroud, J.D., and Kahn, J.K. (1969). Progressive systemic scle-
and uveitis associated with otological and cardiovascular lesions. British rosis with malignant atrophic papulosis. Archives of Dermatology, 100,
Journal of Ophthalmology, 62, 709–14. 575–81.
Davis, M.D., Daoud, M.S., Kirby, B., Gibson, L.E., and Rogers, R.S. (1998). Dutz, J.P., Benoit, L., Wang, X., Demetrick, D.J., Junker, A., de Sa, D., and
Clinicopathologic correlation of hypocomplementemic and normocom- Tan, R. (2001). Lymphocytic vasculitis in X-linked lymphoproliferative
plementemic urticarial vasculitis. Journal of the American Academy of disease. Blood, 97, 95–100.
Dermatology, 38, 899–905. Dyrsen, M.E., Iwenofu, O.H., Nuovo, G., and Magro, C.M. (2008). Parvovirus
D’Cruz, D.P., Wisnieski, J.J., Asherson, R.A., Khamashta, M.A., and Hughes, B19-associated catastrophic endothelialitis with a Degos-like presenta-
G.R. (1995). Autoantibodies in systemic lupus erythematosus and urti- tion. Journal of Cutaneous Pathology, 35 (Suppl. 1), 20–5.
carial vasculitis. Journal of Rheumatology, 22, 1669–73. Eady, R.A.J. and Odland, G.F. (1975). Intraendothelial tubular aggregates
De Gans, J. and van de Beek, D. (2002). Dexamethasone in adults with bacte- in experimental wounds:  Ultrastructural study. British Journal of
rial meningitis. New England Journal of Medicine, 347, 1549–56. Dermatology, 93, 165–73.
De Girolami, U., Smith, T.W., Henin, D., and Hauw, J.-J. (1990). Eiser, A.R., Singh, P., and Shanies, H.M. (1997). Sustained dapsone-induced
Neuropathology of the acquired immune deficiency syndrome. Archives remission of hypocomplementemic urticarial vasculitis—a case report.
of Pathology and Laboratory Medicine, 114, 643–55. Angiology, 48, 1019–22.
Degos, R. (1952). Papulose atrophiante maligne (syndrome cuatneo-intestal Elkon, K.B., Hughes, G.R., Catovsky, D., et al. (1979). Hairy-cell leukaemia
mortel). Annales de Dermatologie et de Syphiligraphie, 79, 410–17. with polyarteritis nodosa. Lancet, 2 (8137), 280–2.
588 SECTION 5 vasculitic diseases and syndromes and related disorders

Englert, H.J., Hawkes, C.H., Bowy, M.L., et  al. (1984). Degos’ dis- of possible pathogenic mechanisms. Journal of Rheumatology, 13,
ease: Association with anticardiolipin antibodies and the lupus antico- 1167–72.
agulant. British Medical Journal, 289, 576. Gabriel, S.E., Espy, M., Erdman, D.D., Bjornsson, J., Smith, T.F., and Hunder,
Enright, H., Jacob, H.S., Vercellotti, G., Howe, R., Belzer, M., and Miller, W. G.G. (1999). The role of parvovirus B19 in the pathogenesis of giant
(1995). Paraneoplastic autoimmune phenomena in patients with myelo- cell arteritis: a preliminary evaluation. Arthritis and Rheumatism, 42,
dysplastic syndromes: response to immunosuppressive therapy. British 1255–8.
Journal of Haematology, 91, 403–8. Gajera, A. and Kais, S. (2009). HIV polyarteritis nodosa-like vasculitis pre-
Enright, H. and Miller, W. (1997). Autoimmune phenomena in patients with senting as chronic abdominal pain. Clinical Rheumatology, 28, 869–72.
myelodysplastic syndromes. Leukemia and Lymphoma, 24, 483–9. Garcia-Porrua, C. and Gonzalez-Gay, M.A. (1998). Cutaneous vasculitis as
Fain, O., Hamidou, M., Cacoub, P., Godeau, B., Wechsler, B., Paries, J., a paraneoplastic syndrome in adults. Arthritis and Rheumatism, 41,
Stirnemann, J., Morin, A.S., Gatfosse, M., Hanslik, T., Belmatoug, N., 1133–5.
Blétry, O., Cevallos, R., Delevaux, I., Fisher, E., Hayem, G., Kaplan, G., García-Porrua, C., González-Gay, M.A., García-Pais, M.J., and Blanco, R.
Le Hello, C., Mouthon, L., Larroche, C., Lemaire, V., Piette, A.M., Piette, (1998). Cutaneous vasculitis: an unusual presentation of sarcoidosis in
J.C., Ponge, T., Puechal, X., Rossert, J., Sarrot-Reynauld, F., Sicard, D., adulthood. Scandinavian Journal of Rheumatology, 27, 80–2.
Ziza, J.M., Kahn, M.F., and Guillevin, L. (2007). Vasculitides associated Gasparovic, H., Djuric, Z., Bosnic, D., Petricevic, M., Brida, M., Dotlic, S.,
with malignancies: analysis of sixty patients. Arthritis and Rheumatism, and Biocina, B. (2011). Aortic root vasculitis associated with Cogan’s
57, 1473–80. syndrome. Annals of Thoracic Surgery, 92, 340–1.
Farrell, A.M., Stern, S.C., El-Ghariani, K., Frankel, A., Woodrow, D., Muller, Gelb, A.B., van de Rijn, M., Regula, D.P. Jr, Cornbleet, J.P., Kamel, O.W.,
B., and Cream, J.J. (1999). Splenic lymphoma with villous lymphocytes Horoupian, D.S., Cleary, M.L., and Warnke, R.A. (1994). Epstein-Barr
presenting as leucocytoclastic vasculitis. Clinical and Experimental virus-associated natural killer-large granular lymphocyte leukemia.
Dermatology, 24, 19–22. Human Pathology, 25, 953–60.
Fauci, A.S. (1988). The human immunodeficiency virus:  infectivity and Gelfand, M.L., Kantor, T., and Gorstein, F. (1972). Cogan’s syndrome with car-
mechanism of pathogenesis. Science, 239, 617–22. diovascular involvement: aortic insufficiency. Bulletin of the New York
Faye-Petersen, O., Frankel, S.R., Schulman, P.E., Raucher, H., Spiera, H., and Academy of Medicine, 48, 647–60.
Dische, M.R. (1991). Giant cell vasculitis with extravascular granulomas Geyer, J.T. and Deshpande, V. (2011). IgG4-associated sialadenitis. Current
in an adolescent. Pediatric Pathology, 11, 281–95. Opinion in Rheumatology, 23, 95–101.
Ferguson, P.J., Saulsbury, F.T., Dowell, S.F., Torok, T.J., Erdman, D.D., and Ghadban, R., Couret, M., and Zenone, T. (2008). Efficacy of infliximab in
Anderson, L.J. (1996). Prevalence of human parvovirus B19 infection in Cogan’s syndrome. Journal of Rheumatology, 35, 2456–8.
children with Henoch-Schönlein purpura. Arthritis and Rheumatism, Ghazale, A., Chari, S.T., Zhang, L., Smyrk, T.C., Takahashi, N., Levy, M.J.,
39, 880–1. Topazian, M.D., Clain, J.E., Pearson, R.K., Petersen, B.T., Vege, S.S.,
Fernandes, S.R., Bertolo, M.B., Rossi, C.L., Samara, A.M., Bonon, S.H., Lindor, K., and Farnell, M.B. (2008). Immunoglobulin G4-associated
Durante, P., and Costa, S.C. (1999). Polyarteritis nodosa and cytomegal- cholangitis: clinical profile and response to therapy. Gastroenterology,
ovirus: diagnosis by polymerase chain reaction. Clinical Rheumatology, 134, 706–15.
18, 501–3. Gherardi, R., Belec, L., Mhiri, C., Gray, F., Lescs, M.-C., Sobel, A., Guillevin,
Fernandes, S.R., Singsen, B.H., and Hoffman, G.S. (2000). Sarcoidosis and L., and Wechsler, J. (1993). The spectrum of vasculitis in human immu-
systemic vasculitis. Seminars in Arthritis and Rheumatism, 30, 33–46. nodeficiency virus-infected patients—a clinicopathologic evaluation.
Fernandez Guerrero, M.L., Aguado, J.M., Arribas, A., Lubreras, C., and de Arthritis and Rheumatism, 36, 1164–74.
Gorgolas, M. (2004). The spectrum of cardiovascular infections due to Gibson, L.E., el-Azhary, R.A., Smith, T.F., and Reda, A.M. (1994). The spec-
Salmonella enterica: a review of clinical features and factors determining trum of cutaneous granulomatous vasculitis: histopathologic report of
outcome. Medicine (Baltimore), 83, 123–38. eight cases with clinical correlation. Journal of Cutaneous Pathology, 21,
Ferris, E.J., Rudikoff, J.C., and Shapiro, J.H. (1968). Cerebral angiography of 437–45.
bacterial infection. Radiology, 90, 727–34. Gilden, D. (2004). Varicella zoster virus and central nervous system syn-
Fidler, H. and Jones, N.S. (1989). Late onset Cogan’s syndrome. Journal of dromes. Herpes, 11 (Suppl. 2), 89A–94A.
Laryngology, 103, 512–14. Gilden, D. (2009). Varicella zoster virus vasculopathies:  diverse clinical
Finkel, T.H., Torok, T.J., Ferguson, P.J., Durigon, E.L., Zaki, S.R., Leung, D.Y., manifestations, laboratory features, pathogenesis, and treatment. Lancet
et al. (1994). Chronic parvovirus B19 infection and systemic narcotiz- Neurology, 8, 731.
ing vasculitis: opportunistic infection or aetiological agent? Lancet, 343, Gilden, D., Kleinschmidt-DeMasters, B.K., Wellish, M., Hedley-Whyte, E.T.,
1255–8. Rentier, B., and Mahalingam, R. (1996). Varicella zoster virus, a cause
Fisher, E.R. and Hellstrom, H.R. (1961). Cogna’s syndrome and systemic vascu- of waxing and waning vasculitis: the New England Journal of Medicine
lar disease: analysis of pathologic features with reference to its relationship case 5-1995 revisited. Neurology, 47, 1441–6.
to thromboangiitis obliterans (Buerger). Archives of Pathology, 72, 572–92. Glikson, M., Galun, E., Schlesinger, M., Cohen, D., Haskell, L., Rubinow, A.,
Foote, E.A., Postier, R.G., Greenfield, R.A., and Bronze, M.S. (2005). and Eliakim, M. (1989). Polyarteritis nodosa and familial Mediterranean
Infectious aortitis. Current Treatment Options in Cardiovascular fever:  a report of 2 cases and review of the literature. Journal of
Medicine, 7, 89–97. Rheumatology, 16, 536–9.
Fortson, J.S., Zone, J.J., Hammond, M.E., and Groggel, G.C. (1986). Gluth, M., Baratz, K., Matteson, E., and Driscoll, C. (2006). Cogan syn-
Hypocomplementemic urticarial vasculitis syndrome responsive to dap- drome: a retrospective review of 60 patients throughout a half century.
sone. Journal of the Academy of Dermatology, 15, 1137–42. Mayo Clinic Proceedings, 81, 483–8.
Fox, G.M., Heilskov, T., and Smith, J.L. (1990). Cogan’s syndrome and serore- Golden, M.P., Hammer, S.M., Wanke, C.A., and Albrecht, M.A. (1994).
activity of Lyme borreliosis. Journal of Clinical Neuro-Ophthalmology, Cytomegalovirus vasculitis. Case reports and review of the literature.
10, 83–7. Medicine (Baltimore), 73, 246–55.
Fruhwirth, J., Mischinger, H.J., Werkgartner, G., Beham, A., and Pfaffenthaller, Goldenberg, N.A., Knapp-Clevenger, R., Hays, T., and Manco-Johnson, M.J.
E.C. (1997). Kohlmeier-Degos’s disease with primary intestinal manifes- (2005). Lemierre’s and Lemierre’s-like syndromes in children: survival
tation. Scandinavian Journal of Gastroenterology, 32, 1066–70. and thromboembolic outcomes. Pediatrics, 116, e543–8.
Gabriel, S.E., Conn, D.L., Phyliky, R.L., Pittelkow, M.R., and Scott, R.E. (1986). Golmia, A., Grinblat, B., Finger, E., Klieman, C., Assir, F., and Scheinberg, M.
Vasculitis in hairy cell leukemia: review of literature and consideration (2008). The development of erythema elevatum diutinum in a patient
CHAPTER 41 miscellaneous forms of vasculitis 589

with juvenile idiopathic arthritis under treatment with abatacept. as a first clinical sign of non-Hodgkin’s lymphoma: a novel association?
Clinical Rheumatology, 27, 105–6. Journal of Dermatology, 35, 297–300.
González Valverde, F.M., Menarguez Pina, F., Ruiz, J.A., Gómez Ramos, M.J., Hayasaka, S., Takatori, Y., Noda, S., Setogawa, T., and Hayashi, H. (1991).
Mauri Barbera, F., Luri Prieto, P., García Marcilla, J.A., and Vázquez Retinal vasculitis in a mother and her son with human T-lymphotropic
Rojas, J.L. (2003). Presentation of Degos syndrome as acute small-bowel virus type 1 associated myelopathy. British Journal of Ophthalmology,
perforation. Archives of Surgery, 138, 57–8. 75, 566–7.
Gornik, H.L. and Creager, M.A. (2008). Aortitis. Circulation,117, 3039–51. Hayem, G., Gomez, M.J., Grossin, M., Meyer, O., and Kahn, M.F. (1997).
Gouin, E., Egile, C., Dehoux, P., Villiers, V., Adams, J., Gertler, F., Li, R., and Systemic vasculitis and epithelioma. A report of three cases with a litera-
Cossart, P. (2004). The RickA protein of Rickettsia conorii activates the ture review. Revue du Rhumatisme (English), 64, 816–24.
Arp2/3 complex. Nature, 427, 457–61. Haynes, B.F., Kaiser-Kupfer, M.I., Mason, P., and Fauci, A.S. (1980). Cogan
Grasland, A., Pouchet, J., Hachulla, E., Bletry, O., Papo, T., and Vinceneux, P. syndrome:  studies in thirteen patients, long-term follow-up, and a
(2004). Typical and atypical Cogan’s syndrome: 32 cases and review of review of the literature. Medicine (Baltimore), 59, 426–41.
the literature. Rheumatology, 43, 1007–15. Haynes, B.F., Pikus, A., Kaiser-Kupfer, M., and Fauci, A.S. (1981). Successful
Grayson, W. (2008). The HIV-postive skin biopsy. Journal of Clinical treatment of sudden hearing loss in Cogan’s syndrome with corticoster-
Pathology, 61, 802–17. oids. Arthritis and Rheumatism, 24, 501–3.
Greer, J.M., Longley, S., Edwards, N.L., Elfenbein, G.J., and Panush, R.S. Heinzen, R.A. (2003). Rickettsial actin-based motility: behavior and involve-
(1988). Vasculitis associated with malignancy. Experience with 13 ment of cytoskeletal regulators. Annals of the New  York Academy of
patients and literature review. Medicine (Baltimore), 67, 220–30. Sciences, 990, 535–47.
Gudbrandsson, T., Hansson, L., Herlitz, H., et al. (1981). Immunological changes Helmchen, C., Arbusow, V., Jäger, L., Strupp, M., Stöcker, W., and Schulz, P.
in patients with previous malignant essential hypertension. Lancet, 1, 406–8. (1999). Cogan’s syndrome: clinical significance of antibodies against the
Guhl, G., Diaz-Ley, B., Delgado, Y., Dauden, E., Fraga, J., and Garcia-Diez, A. inner ear and cornea. Acta Oto-laryngologica, 119, 528–37.
(2009). Wegener’s granulomatosis: a new entity in the growing differen- Hickey, W.F. and Kimura, H. (1988). Perivascular microglia are bone marrow
tial diagnosis of Degos’ disease. Clinical and Experimental Dermatology, derived and present antigen in vivo. Science, 239, 290–2.
34, e1–3. High, W.A., Aranda J., Patel, S.B., Cockerel, C.J., and Costner M.I. (2004).
Guillevin, L. (2008). Vasculitides in the context of HIV infection. AIDS, 22 Is Degos’ disease a clinical and histological end point rather than a
(Suppl. 3), S27–S33. specific disease? Journal of the American Academy of Dermatology,
Guinee, D. Jr, Jaffe, E., Kingma, D., Fishback, N., Wallberg, K., Krishnan, J., 50, 895–9.
Frizzera, G., Travis, W., and Koss, M. (1994). Pulmonary lymphoma- Hippach, M., Meyberg, R., Villena-Heinsen, C., Mink, D., Ertan, A.K.,
toid granulomatosis. Evidence for a proliferation of Epstein-Barr virus Schmidt, W., and Hippach, M. (2000). Postpartum ovarian vein throm-
infected B-lymphocytes with a prominent T-cell component and vascu- bosis. Clinical and Experimental Obstetrics and Gynecology, 27, 24–6.
litis. American Journal of Surgical Pathology, 18, 753–64. Ho, A.C., Roat, M.I., Venbrux, A., and Hellmann, D.B. (1999). Cogan’s syn-
Guissa, V.R., Aragão, P.A., Marques, H.H., Jacob, C.M., and Silva, C.A. drome with refractory abdominal aortitis and mesenteric vasculitis.
(2010). Chronic active Epstein-Barr virus infection mimicking Journal of Rheumatology, 26, 1404–7.
Henoch-Schönlein purpura. Acta Reumatológica Portuguesa, 35, 513–7. Hohwy, T., Jensen, M.G., Tøttrup, A., Steiniche, T., and Fogh K. (2006).
Gupta, S. and Licorish, K. (1984). Circulating immune complexes in AIDS. A fatal case of malignant atrophic papulosis (Degos’ disease) in a
New England Journal of Medicine, 310, 1530–1. man with factor V Leiden mutation and lupus anticoagulant. Acta
Hall-Smith, P. (1969). Malignant atrophic papulosis (Degos’ disease): Two Dermato-Venereologica, 86, 245–7.
cases occurring in the same family. British Journal of Dermatology, 81, Holm, J.M., Hansen, L.K., and Oxhoj, H. (1995). Kawasaki disease associated
817–22. with parvovirus B19 infection. European Journal of Pediatrics, 154, 633–4.
Hall-Smith, S.P. (1964). Malignant atrophic papulosis (Degos’ syndrome). Horney, L.F. and Walker, D.H. (1988). Meningoencephalitis as a major manifesta-
Proceedings of the Royal Society of Medicine, 57, 519–21. tion of Rocky Mountain spotted fever. Southern Medical Journal, 81, 915–8.
Hamidou, M.A., Boumalassa, A., Larroche, C., El Kouri, D., Bletry, O., and Howard, R.O. and Nishida, S. (1969). A case of Degos’ disease with elec-
Grolleau, J.Y. (2001). Systemic medium-sized vessel vasculitis associ- tron microscopic findings. Transactions of the American Academy of
ated with chronic myelomonocytic leukemia Seminars in Arthritis and Ophthalmology and Otolaryngology, 73, 1097–112.
Rheumatism, 31, 119–26. Hsu, R.B. and Lin, F.Y. (2005). Risk factors for bacteraemia and endovascular
Hammer, M., Witte, T., Mugge, A., Wollenhaupt, J., Laas, J., Laszig, R., and infection due to non-typhoid salmonella: a reappraisal. QJM, 98, 821–7.
Zeidler, H. (1994). Complicated Cogan’s syndrome with aortic insuffi- Huang, T.E. and Chou, S.M. (1988). Occlusive hypertrophic arteritis as the
ciency and coronary stenosis. Journal of Rheumatology, 21, 552–5. cause of discrete necrosis in CNS toxoplasmosis in the acquired immune
Hancox, J.G., Wallace, C.A., Sangueza, O.P., and Graham, G.F. (2004). deficiency syndrome. Human Pathology, 19, 1210–14.
Erythema elevatum diutinum associated with lupus panniculitis in a Hunninghake, G.W. and Crystal, R.G. (1981). Pulmonary sarcoidosis: a dis-
patient with discoid lesions of chronic cutaneous lupus erythematosus. order manifest by excess helper T-lymphocyte activity at sites of disease
Journal of the American Academy of Dermatology, 50, 652–3. activity. New England Journal of Medicine, 305, 429–34.
Harvell, J.D., Williford, P.L., White, W.L., (2001). Benign cutaneous Degos’ Hutson, T.E. and Hoffman, G.S. (2000). Temporal concurrence of vasculitis
disease: a case report with emphasis on histopathology as papules and cancer: a report of 12 cases. Arthritis Care and Research, 13, 417–23.
chronologically evolve. Am J Dermatopathol, 23(2), 116–23. Ilowite, N.T., Fligner, C.L., Ochs, H.D., Brichacek, B., Harada, S., Haas, J.E.,
Hasler, P., Kistler, H., and Gerber, H. (1995). Vasculitides in hairy cell leuke- Purtilo, D.T., and Wedgwood, R.J. (1986). Pulmonary angiitis with
mia. Seminars in Arthritis and Rheumatism, 25, 134–42. atypical lymphoreticular infiltrates in Wiskott-Aldrich syndrome: pos-
Hatemi, G., Masatlioglu, S., Gogus, F., and Ozdogan, H. (2004). Necrotizing sible relationship of lymphomatoid granulomatosis and EBV infection.
vasculitis associated with familial Mediterranean fever. American Clinical Immunology and Immunopathology, 41, 479–84.
Journal of Medicine, 117, 516–9. Itescu, S., Brancato, L.J., Buxbaum, J., et al. (1990). A diffuse infiltrative CD8
Hattwick, M.A., O’Brien, R.J., and Hanson, B.F. (1976). Rocky Mountain lymphocytosis syndrome in human immunodeficiency virus (HIV)
spotted fever: epidemiology of an increasing problem. Annals of Internal infection: A host immune response associated with HLA-DR5. Annals
Medicine, 84, 732–9. of Internal Medicine, 112, 3.
Hatzitolios, A., Tzellos, T.G., Savopoulos, C., Tzalokostas, V., Kaiafa, G., and Jaffe, E.S. and Wilson, W.H. (1997). Lymphomatoid granulomatosis: patho-
Psomas, E. (2008). Erythema elevatum diutinum with rare distribution genesis, pathology and clinical implications. Cancer Survey, 30, 233–48.
590 SECTION 5 vasculitic diseases and syndromes and related disorders

Jan W., Zimmerman, R.A., Bilaniuk, L.T., Hunter, J.V., Simon, E.M., and Katsetos, C.D., Fincke, J.E., Legido, A., Lischner, H.W., de Chadarevain, J.P.,
Haselgrove, J. (2003). Diffusion-weighted imaging in acute bacterial Kaye, E.M., Platsoucas, C.D., and Oleszak, E.L. (1999). Angiocentric
meningitis in infancy. Neuroradiology, 45, 634–9. CD3(+). T-cell infiltrates in human immunodeficiency virus type
Janssen, H.L., Meinardi, J.R., Vleggaar, F.P., van Uum, S.H., Haagsma, E.B., 1-associated central nervous system disease in children. Clinical and
van Der Meer, F.J., van Hattum, J., Chamuleau, R.A., Adang, R.P., Diagnostic Laboratory Immunology, 6, 105–14.
Vandenbroucke, J.P., van Hoek, B., and Rosendaal, F.R. (2000). Factor Katz, S.I., Gallin, J.I., Hertz, K.C., Fauci, A.S., and Lawley, T.J. (1977).
V Leiden mutation, prothrombin gene mutation, and deficiencies in Erythema elevatum diutinum: skin and systemic manifestations, immu-
coagulation inhibitors associated with Budd-Chiari syndrome and por- nologic studies, and successful treatment with dapsone. Medicine
tal vein thrombosis: results of a case-control study. Blood, 96, 2364–8. (Baltimore), 56, 443–55.
Jeanneret, C. (2008). Multifocal thrombophlebitis and deep vein thrombo- Katz, S.K., Mudd, L.J., and Roenigk, H.H. Jr (1997). Malignant atrophic pap-
sis in primary cytomegalovirus infection—a histopathological analysis. ulosis (Degos’ disease) involving three generations of a family. Journal of
Thrombosis and Haemostasis, 99, 445–7. the American Academy of Dermatology, 37, 480–4.
Johnson, R.M., Barbarini, G., and Barbaro, G. (2003). Kawasaki-like syn- Kawakami, T. and Soma, Y. (2011). Successful use of mizoribine in a patient
dromes and other vasculitic syndromes in HIV-infected patients. AIDS, with sarcoidosis and cutaneous vasculitis. Acta Dermato-Venereologica,
17 (Suppl. 1), S77–82. 91, 582–3.
Johnson, R.M., Little, J.R., and Storch, G.A. (2001). Kawasaki-like syndromes Khan, M.L., Colby, T.V., Viggiano, R.W., and Fonseca, R. (2010). Treatment
associated with human immunodeficiency virus infection. Clinical with bortezomib of a patient having hyper IgG4 disease. Clinical
Infectious Diseases, 32, 1628–34. Lymphoma Myeloma and Leukemia, 10, 217–9.
Johnston, C. and Kennedy, C. (1984). Cutaneous leucocytoclastic vasculitis Khosroshahi, A., Bloch, D.B., Deshpande, V., and Stone J.H. (2010).
associated with acute sarcoidosis. Postgraduate Medical Journal, 60, Rituximab therapy leads to rapid decline of serum IgG4 levels and
549–50. prompt clinical improvement in IgG4-related systemic disease. Arthritis
Judson, M.A., Baughman, R.P., Costabel, U., Flavin, S., Lo, K.H., Kavuru, and Rheumatism, 62, 1755–62.
M.S., Drent, M.; Centocor T48 Sarcoidosis Investigators (2008). Efficacy Khosroshahi, A., Carruthers, M.N., Deshpande, V., Unizony, S., Bloch, D.B.,
of infliximab in extrapulmonary sarcoidosis: results from a randomised and Stone J.H. (2012). Rituximab for the treatment of IgG4-related
trial. European Respiratory Journal, 31, 1189–96. disease:  lessons from 10 consecutive patients. Medicine (Baltimore),
Justo, D., Finn, T., Atzmony, L., Guy, N., and Steinvil, A. (2011). Thrombosis 91, 57–66.
associated with acute cytomegalovirus infection:  a meta-analysis. Khosroshahi, A., Stone, J.R., Pratt, D.S., Deshpande, V., and Stone, J.H. (2009).
European Journal of Internal Medicine, 22, 195–9. Painless jaundice with serial multi-organ dysfunction. Lancet, 373, 1494.
Kamisawa, T., Okazaki, K., Kawa, S., Shimosegawa, T., and Tanaka, M. (2010). Kikuta, H., Matsumoto, S., and Osato, T. (1991). Kawasaki disease and
Japanese consensus guidelines for management of autoimmune pancre- Epstein-Barr virus. Acta Paediatrica Japonica, 33, 765–70.
atitis: III. Treatment and prognosis of AIP. Journal of Gastroenterology, Kikuta, H., Nakanishi, M., Ishikawa, N., Konno, M., and Matsumoto,
45, 471–7. S. (1992). Detection of Epstein-Barr virus sequences in patients
Kamisawa, T., Shimosegawa, T., Okazaki, K., Nishino, T., Watanabe, H., with Kawasaki disease by means of the polymerase chain reaction.
Kanno, A., Okumura, F., Nishikawa, T., Kobayashi, K., Ichiya, T., Intervirology, 33, 1–5.
Takatori, H., Yamakita, K., Kubota, K., Hamano, H., Okamura, K., Kim, Y.L., Gong, S.J., Hwang, Y.H., Joo, J.E., Cho, Y.U., Lee, J.A., Sung, S.A.,
Hirano, K., Ito, T., Ko, S.B., and Omata, M. (2009). Standard steroid Lee, S.Y., and Kim, N.Y. (2011). Waldenstrom macroglobulinemia with
treatment for autoimmune pancreatitis. Gut, 58, 1504–7. CD5+ expression presented as cryoglobulinemic glomerulonephropa-
Kanegane, H., Tsuji, T., Seki, H., Yachie, A., Yokoi, T., Miyawaki, T., and thy: a case report. Journal of Korean Medical Science, 26, 824–8.
Taniguchi, N. (1994). Kawasaki disease with a concomitant primary Kirk, J.L., Fine, D.P., Sexton, D.J., and Munchmore, H.G. (1990). Rocky
Epstein-Barr virus infection. Acta Paediatrica Japonica, 36, 713–6. Mountain spotted fever. A clinical review based on 48 confirmed cases,
Kanellopoulou, T. Alexopoulou, A., Theodossiades, G., Koskinas, J., and 1943–1986. Medicine (Baltimore), 69, 35–45.
Archimandritis, A.J. (2010). Pylephlebitis: an overview of non-cirrhotic Kirkland, K.B., Williamson, W.E., and Sexton, D.J. (1995). Therapeutic
cases and factors related to outcome. Scandinavian Journal of Infectious delay and mortality in cases of Rocky Mountain spotted fever. Clinical
Diseases, 42, 804–11. Infectious Diseases, 20, 1118–21.
Kanitakis, J., Cozzani, E., Lyonnet, S., and Thivolet, J. (1993). Ultrastructural Kisch, L.S. and Bruyzeel, D.P. (1984). Six cases of malignant atrophic
study of chronic lesions of erythema elevatum diutinum:  ‘extracellu- papulosis (Degos’ disease) occurring in one family. British Journal of
lar cholesterosis’ is a misnomer. Journal of the American Academy of Dermatology, 111, 469–71.
Dermatology, 29, 363–7. Kishore, J., Dash, N.R., Saxena, R., and Krishnani, N. (2010). Novel detec-
Karkos, P.D., Asrani, S., Karkos, C.D., Leong, S.C., Theochari, E.G., tion of parvovirus B19 DNA and IgM antibodies in patients with
Alexopoulou, T.D., and Assimakopoulos, A.D. (2009). Lemierre’s syn- non-occlusive gangrene of stomach and bowel. Indian Journal of
drome: A systematic review. Laryngoscope, 119, 1552–9. Medical Research, 131, 702–10.
Karni, A., Sadeh, M., Blatt, I., and Goldhammer, Y. (1991). Cogan’s syn- Klima, M. and Waddell, C.C. (1984). Hairy cell leukemia associated with
drome complicated by lacunar brain infarcts. Journal of Neurology, focal vascular damage. Human Pathology, 15, 657–9.
Neurosurgery, and Psychiatry, 54, 169–71. Kobayashi, S., Nagase, M., Hidaka, S., Arai, T., Ikegaya, N., Hishida, A., and
Kasashima, S., Zen, Y., Kawashima, A., Konishi, K., Sasaki, H., Endo, M., Honda, N. (1994). Membranous nephropathy associated with hypocom-
Matsumoto, Y., Kawakami, K., Kasashima, F., Moriya, M., Kimura, K., plementemic urticarial vasculitis: report of two cases and a review of the
Ohtake, H., and Nakanuma, Y. (2008). Inflammatory abdominal aor- literature. Nephron, 66, 1–7.
tic aneurysm: close relationship to IgG4-related periaortitis. American Koeppen, A.H., Lansing, L.S., Peng, S.K., and Smith, R.S. (1981). Central
Journal of Surgical Pathology, 32, 197–204. nervous system vasculitis in cytomegalovirus infection. Journal of
Kasper, D.L., Sahani, D., and Misdraji, J. (2005). A 40-year-old man with Neurological Science, 51, 395–410.
prolonged fever and weight loss, case 25-2005. New England Journal of Kohlmeier, W. (1941). Multiple Hautnekrosen beiThrombangiitis obliterans.
Medicine, 353, 713–22. Archiv für Dermatologie und Syphilis, 181, 783–92.
Kastenbauer, S. and Pfister, H.W. (2003). Pneumococcal meningitis in Komadina, K.H. and Houk, R.W. (1989). Polyarteritis nodosa presenting as
adulsts: spectrum of complications and prognostic factors in a series of recurrent pneumonia following splenectomy for hairy-cell leukemia.
87 cases. Brain, 126, 1015–25. Seminars in Arthritis and Rheumatism, 18, 252–7.
CHAPTER 41 miscellaneous forms of vasculitis 591

Koskinen, P., Lemstrom, K., Bruggeman, C., Lautenschlager, I., and Hayry, Libman, B.S., Quismorio, F.P. Jr, and Stimmler, M.M. (1995). Polyarteritis
P. (1994). Acute cytomegalovirus infection induces a subendothelial nodosa-like vasculitis in human immunodeficiency virus infection.
inflammation (endothelialitis) in the allograft vascular wall. A possible Journal of Rheumatology, 22, 351–5.
linkage with enhanced allograft arteriosclerosis. American Journal of Lidar, M., Lipschitz, N., Langevitz, P., and Shoenfeld, Y. (2009). The infectious
Pathology, 144, 41–50. etiology of vasculitis. Autoimmunity, 42, 432–8.
Kristensen, B.O., Andersen, P.L.M., Wiik, A., et al. (1984). Autoantibodies Liebow, A.A., Carrington, C.R., and Friedman, P.J. (1972). Lymphomatoid
and vascular events in essential hypertension: A five-year longitudinal granulomatosis. Human Pathology, 3, 457–558.
study. Journal of Hypertension, 2, 19–24. Liozon, E., Loustaud, V., Fauchais, A.L, Soria, P., Ly, K., Ouattara, B., Rhaiem,
Kumano, Y., Nagato, T., Kurihara, K., Kikukawa, H., Goto, M., Kawano, K., Nadalon, S., and Vidal, E. (2006). Concurrent temporal (giant cell)
Y., Ohnishi, Y., and Inomata, H. (1997). Hyperimmunoglobulinemia arteritis and malignancy: report of 20 patients with review of the litera-
D in idiopathic retinal vasculitis. Graefes Archives in Clinical and ture. Journal of Rheumatology, 33, 1606–14.
Experimental Ophthalmology, 235, 372–8. Liu, C.M., Harris, R.M., and Hansen, C.D. (2005). Lesions resembling malig-
Kurzrock, R. and Cohen, P.R. (1993). Vasculitis and cancer. Clinical nant atrophic papulosis in a patient with progressive systemic sclerosis.
Dermatology, 11, 175–87. Cutis, 75, 101–4.
Kurzrock, R. and Cohen, P.R. (1995a). Cutaneous paraneoplastic syndromes Liu, T.C., Chen, I.S., Lin, T.K., Lee, J.Y., Kirn, D., and Tsao, C.J. (2009). Erythema
in solid tumors. American Journal of Medicine, 99, 662–71. elevatum diutinum as a paraneoplastic syndrome in a patient with pulmo-
Kurzrock, R. and Cohen, P.R. (1995b). Mucocutaneous paraneoplastic mani- nary lymphoepithelioma-like carcinoma. Lung Cancer, 63, 151–3.
festations of hematologic malignancies. American Journal of Medicine, Liu, Y.C., Tomashefski, J.F., Tomford, J.W., et al. (1989). Necrotizing pneu-
99, 207–16. mocystis carinii vasculitis associated with acquired immunodeficiency
Kurzrock, R., Cohen, P.R., and Markowitz, A. (1994). Clinical manifestations syndrome. Archives of Pathology and Laboratory Medicine, 113, 494.
of vasculitis in patients with solid tumors. A case report and review of Livingston, J.Z., Casale, A.S., Hutchins, G.M., and Shapiro E.P. (1992).
the literature. Archives of Internal Medicine, 154, 334–40. Coronary involvement in Cogan’s syndrome. American Heart Journal,
Kwong, T., Valderrama, E., Paley, C., and Ilowite, N. (1994). Systemic 123, 528–30.
necrotizing vasculitis associated with childhood sarcoidosis. Seminars Ljunstrom, L., Franzen, C., Schlaug, M., Elowson, S., and Vidas, U. (1997).
in Arthritis and Rheumatism, 23, 388–95. Reinfection with chlamydia pneumonia may induce isolated and sys-
Kyung-Whan, M., Yorkey, F., and Sato, C. (1980). Mucin-producing aden- temic vasculitis in small and large vessels. Scandinavian Journal of
oscarcinomas and nonbacterial thrombotic endocarditis: Pathogenetic Infectious Diseases, 104 (Suppl.), 37–40.
role of tumor mucin. Cancer, 45, 2375–82. Longley, S., Caldwell, J.R., and Panush, R.S. (1986). Paraneoplastic vascu-
Lande, M.B., Mowry, J.A., Houghton, D.C., White, C.R. Jr, and Brozy, M.S. litis. Unique syndrome of cutaneous angiitis and arthritis associated
(1998). Immune complex disease associated with Epstein-Barr virus with myeloproliferative disorders. American Journal of Medicine, 80,
infectious mononucleosis. Pediatric Nephrology, 12, 651–3. 1027–30.
Lane, G.P., Cochrane, A.D., and Fore, D.R. (1988). Salmonella mycotic Lopes, R.J. (2009). Infectious thoracic aortitis: a literature review. Clinical
abdominal-aortic aneurysm. Medical Journal of Australia, 149, 95–7. Cardiology, 32, 488–90.
Langevitz, P., Zemer, D., Livneh, A., Shemer, J., and Pras, M. (1994). Lopez, L.R., Davis, K.C., Kohler, P.F., and Schocket, A.L. (1984). The hypoc-
Protracted febrile myalgia in patients with familial Mediterranean fever. omplementemic urticarial-vasculitis syndrome:  therapeutic response
Journal of Rheumatology, 21, 1708–9. to hydroxychloroquine. Journal of Allergy and Clinical Immunology,
Lau, Y.L., Wong, S.N., and Lawton, W.M. (1992). Takayasu’s arteritis associ- 73, 600–3.
ated with Wiskott-Aldrich syndrome. Journal of Paediatrics and Child Lunardi, C., Bason, C., Leandri, M., Navone, R., Lestani, M., Millo, E.,
Health, 28, 407–9. Benatti, U., Cilli, M., Beri, R., Corrocher, R., and Puccetti, A. (2002).
Lazzarini, L.C., de Fatima do Amparo Teixeira, M., Souza Rodrigues, R., and Autoantibodies to inner ear and endothelial antigens in Cogan’s syn-
Marcos Nunes Valiante, P. (2008). Necrotizing sarcoid granulomatosis in drome. Lancet, 360, 915–21.
a family of patients with sarcoidosis reinforces the association between Macedo, T.A., Stanson, A.W., Oderich, G.S., Johnson, C.M., Panneton, J.M.,
both entities. Respiration 76, 356–60. and Tie, M.L. (2004). Infected aortic aneurysms imaging findings.
Leavitt, R.Y. and Fauci, A.S. (1986). Pulmonary vasculitis. American Review Radiology, 231, 250–7.
of Respiratory Disease, 134, 149–66. Magrinat, G., Kerwin, K.S., and Gabriel, D.A. (1989). The clinical mani-
LeBoit, P.E. and Cockerell, C.J. (1993). Nodular lesions of erythema elevatum festations of Degos’ syndrome. Archives of Pathology and Laboratory
diutinum in patients infected with the human immunodeficiency virus. Medicine, 113, 354–62.
Journal of the American Academy of Dermatology, 28, 919–22. Magro, C., Ali, N., Williams, J.D., Allen, J.N., and Ross, P. Jr (2005).
Lee, A.Y., Nakagawa, H., Nogita, T., and Ishibashi, Y. (1989). Erythema ele- Cytomegalovirus-associated pulmonary septal capillary injury sine
vatum diutinum:  an ultrastructural case study. Journal of Cutaneous inclusion body change:  a distinctive cause of occult or macroscopic
Pathology, 16, 211–7. pulmonary hemorrhage in the immunocompetent host. Applied
Lee, S.B., Jones L.K., and Giannini, C. (2005). Brainstem infarcts as an early Immunohistochemistry and Molecular Morphology, 13, 268–72.
manifestation of Streptococcus anginosus meningitis. Neurocritical Magro, C.M., Iwenofu, O.H., Kerns, M.J., Nuovo, G.J., Dyrsen, M.E., and
Care, 3, 157–60. Segal, J.P. (2009). Fulminant and accelerated presentation of dermato-
Lenglet, T., Haroche, J., Schnuriger, A., Maisonobe, T., Viala, K., Michel, Y., myositis in two previously healthy young adult males: a potential role
Chelbi, F., Grabli, D., Seror, P., Garbarg-Chenon, A., Amoura, Z., and for endotheliotropic viral infection. Journal of Cutaneous Pathology,
Bouche, P. (2011). Mononeuropathy multiplex associated with acute 36, 853–8.
parvovirus B19 infection:  characteristics, treatment and outcome. Magro, C.M., Poe, J.C., Kim, C., Shapiro, L., Nuovo, G., Crow, M.K., and
Journal of Neurology, 258, 1321–6. Crow, Y.J. (2011). Degos disease: a C5b-9/interferon-α-mediated endo-
Leporati, P., Landek-Salgado, M.A., Lupi, I., Chiovato, L., and Caturegli, P. theliopathy syndrome. American Journal of Clinical Pathology, 135,
(2011). IgG4-related hypophysitis: a new addition to the hypophysitis spec- 599–610.
trum. Journal of Clinical Endocrinology and Metabolism, 96, 1971–80. Majoor, M.H.J.M., Albers, F.W.J., and Casselman, J.W. (1993). Clinical
Lewkonia, R.M., Horne, D., and Dawood, M.R. (1995). Juvenile dermatomy- relevance of magnetic resonance imaging and computed tomogra-
ositis in a child infected with human parvovirus B19. Clinical Infectious phy in Cogan’s syndrome. Acta Otolaryngologica (Stockholm), 113,
Diseases, 21, 430. 625–31.
592 SECTION 5 vasculitic diseases and syndromes and related disorders

Marcussen, N. and Lund, C. (1989). Combined sarcoidosis and disseminated NS1, of human parvovirus B19 induces activation of interleukin-6 gene
visceral giant cell vasculitis. Pathology Research in Practice, 184, 325–30. expression. Journal of Virology, 70, 8485–91.
Markert, M.L. (1991). Purine nucleoside phosphorylase deficiency. Molbak, K. (2005). Human health consequences of antimicrobial
Immunodeficiency Reviews, 3, 45. drug-resistant Salmonella and other foodborne pathogens. Clinical
Martensson, U., Sjoholm, A.G., Sturfelt, G., Truedsson, L., and Laurell, A.B. Infectious Diseases, 41, 1613–20.
(1992). Western blot analysis of human IgG reactive with the collagenous Moreno, S., Garcia Altozano, J., Pinilla, B., Lopez, J.C., de Quiros, B., Ortega,
portion of C1q: evidence of distinct binding specificities. Scandinavian A., and Bouza, E. (1989). Lemierre’s disease:  postanginal bacteremia
Journal of Immunology, 35, 735–44. and pulmonary involvement caused by Fusobacterium necrophorum.
Massari, M., Salvarani, C., Portioli, I., Ramazzotti, E., Gabbi, E., and Bonazzi, Reviews of Infectious Diseases, 11, 319–24.
L. (1996). Polyarteritis nodosa and HIV infection:  no evidence of a Mori, Y., Yamashita, H., Umeda, Y., Uchiyama-Tanaka, Y., Nose, A.,
direct role of HIV. Infection, 24, 159–61. Kishimoto, N., Kijima, Y., Nagata, T., Mori, M., Matsubara, H., Yoshida,
Matsumuro, K., Izumo, S., Umehara, F., Arisato, T., Maruyama, I., Yonezawa, H., and Iwasaka, T. (2002). Association of parvovirus B19 infection with
S., Shirahama, H., Sato, E., and Osame, M. (1994). Paraneoplastic vascu- acute glomerulonephritis in healthy adults: Case report and review of
litic neuropathy: immunohistochemical studies on a biopsied nerve and the literature. Clinical Nephrology, 2002, 57, 69–73.
postmortem examination. Journal of Internal Medicine, 236, 225–30. Mouallem, M., Friedman, E., Pauzner, R., Schwartz, E., and Rubenstein, E.
Matteson, E.L., Choi, H.K., Poe, D.S., Wise, C., Lowe, V.J., McDonald, T.J., (1987). Rickettsiosis-associated hyponatremia. Infection, 15, 315–6.
and Rahman, M.U. (2005). Etanercept therapy for immune-mediated Muller, S.A. and Landry, M. (1974). Exchange autografts in malignant
cochleovestibular disorders:  A  multi-center open-label, pilot study. atrophic papulosis (Degos’ disease). Mayo Clinic Proceedings,
Arthritis and Rheumatism, 53, 337–42. 49, 884–8.
Matteson, E.L., Fabry, D.A., Facer, G.W., Beatty, C.W., Driscoll, C.L., Strome, Murai, T., Imai, M., Inui, M., Watanabe, H., and Hosoda, Y. (1986).
S.E., and McDonald, T.J. (2001). Open trial of methotrexate as treatment Generalized granulomatous arteritis with aortic dissection.
for autoimmune hearing loss. Arthritis and Rheumatism, 45, 146–50. Zentralblatt fur Allgemeine Pathologie und Pathologische Anatomie,
McCallum, R.M. (1996). Cogan’s syndrome. In Current therapy of allergy, 132, 41–7.
immunology, and rheumatology, 5th edn (ed. L.M. Lichtenstein and A.S. Murakami, K., Ohsawa, M., Hu, S.X., Kanno, H., Aozasa, K., and Nose, M.
Fauci), pp. 255–60. Mosby, St Louis. (1998). Large-vessel arteritis associated with chronic active Epstein-Barr
McCluggage, W.G., Armstrong, D.J., Maxwell, R.J., Ellis, P.K., and virus infection. Arthritis and Rheumatism, 41, 369–73.
McCluskey, D.R. (1999). Systemic vasculitis and aneurysm formation in Muratori, S., Carrera, C., Gorani, A., and Alessi, E. (1999). Erythema eleva-
the Wiskott-Aldrich syndrome. Journal of Clinical Pathology, 52, 390–2. tum diutinum and HIV infection: a report of five cases. British Journal
McIntyre, P.B., Berkey, C.S., King, S.M., Schaad, U.B., Kilpi, T., and Kanra, of Dermatology, 141, 335–8.
G.Y. (1997). Dexamethasone as adjunctive therapy in bacterial menin- Murphy, G., Sullivan, M.O., Shanahan, F., Harney, S., and Molloy, M. (2009).
gitis. A meta-analysis of randomized clinical trials since 1988. JAMA, Cogan’s syndrome:  present and future directions. Rheumatology
278, 925–31. International, 29, 1117–21.
McMillan, A., Bishop, P.E., Aw, D., and Peutherer, J.F. (1989). Immunohistology Myklebust, G., Wilsgaard, T., Jacobsen, B.K., and Gran, J.T. (2002). No
of the skin rash associated with acute HIV infection. AIDS, 3, 309–12. increased frequency of malignant neoplasms in polymyalgia rheumatica
Mehregan, D.R., Hall, M.J., and Gibson, L.E. (1992). Urticarial vasculitis: a and temporal arteritis. A prospective longitudinal study of 398 cases and
histopathologic and clinical review of 72 cases. Journal of the American matched population controls. Journal of Rheumatology, 29, 2143–7.
Academy of Dermatology, 26, 441–8. Nagel, M.A. (2010). Virus vasculopathy and stroke:  an under-recognized
Migliori, G., Battisti, E., Pari, M., Vitelli, N., and Cingolani, C. (2009). A cause and treatment target. Infectious Disorders Drug Targets, 10,
shifty diagnosis: Cogan’s syndrome.A case report and review of the lit- 105–11.
erature. Acta Otorhinolaryngologica Italica, 29, 108–13. Nakagawa, A., Ito, M., Iwaki, T., Yatabe, Y., Asai, J., and Hayashi, K. (1996).
Miller, D.V., Oderich, G.S., Aubry, M.C., Panneton, J.M., and Edwards, W.D. Chronic active Epstein-Barr virus infection with giant coronary aneu-
(2004). Surgical pathology of infected aneurysms of the descending tho- rysms. American Journal of Clinical Pathology, 105, 733–6.
racic and abdominal aorta: Clinicopathologic correlations in 29 cases Nallasamy, S., Eagle, R.C. Jr, Rorke-Adams, L., Nichols, K.E., and Brucker,
(1976 to 1999).  Human Pathology, 35, 1112–20. A.J. (2011). Eye findings in X-linked lymphoproliferative disorder.
Miranda-Filloy, J.A., Veiga, J.A., Juarez, Y., Gonzalez-Juanatey, C., Retina, 31, 790–7.
Gonzalez-Gay, M.A., and Garcia-Porrua, C. (2006). Microscopic poly- Nau, R., Lantsch, M., Stiefel, M., et al. (1998). Varicella zoster virus-associated
angiitis following recurrent Staphylococcus aureus bacteremia and focal vasculitis without herpes zoster: recovery after treatment with acy-
infectious endocarditis. Clinical and Experimental Rheumatology, clovir. Neurology, 51, 914–5.
24, 705–6. Naylor, D., Mullins, J.F., and Gilmore, J.F. (1960). Papulosis atrophicans
Mita, T., Nakanishi, Y., Ochiai, A., Shimoda, T., Kato, H., Yamaguchi, H., and maligna (Degos’ syndrome): Report of the first United States case and
Toda, G. (1999). Paraneoplastic vasculitis associated with esophageal review of the literature. Archives of Dermatology, 81, 189–97.
carcinoma. Pathology International, 49, 643–7. Newman, L.S., Rose, C.S., and Maier, L.A. (1997). Medical progress:  sar-
Mitsui, H., Shibagaki, N., Kawamura, T., Matsue, H., and Shimada, S. coidosis. New England Journal of Medicine, 336, 1224–34.
(2009). A clinical study of Henoch-Schonlein Purpura associated with Newsome, S.D. and Nath, A. (2009). Varicella-zoster virus vasculopathy and
malignancy. Journal of the European Academy of Dermatology and central nervous system immune reconstitution inflammatory syndrome
Venereology, 23, 394–401. with human immunodeficiency virus infection treated with steroids.
Miyachi, H., Akizuki, M., Yamagata, H., Mimori, T., Yoshida, S., and Homma, Journal of NeuroVirology, 15, 288–91.
M. (1987). Hypertrophic osteoarthropathy, cutaneous vasculitis, and Newton, J. and Black, M.M. (1984). Familial malignant atrophic papulosis.
mixed-type cryoglobulinemia in a patient with nasopharyngeal carci- Clinical and Experimental Dermatology, 9, 298–9.
noma. Arthritis and Rheumatism, 30, 825–9. Nieuwhof, C.M., Damoiseaux, J., and Tervaert, J.W. (2006). Successful treat-
Miyanaga, M., Shimizu, K., Kawaguchi, T., Miyata, K., and Mochizuki, M. ment of cerebral vasculitis in an HIV-positive patient with anti-CD25
(2009). A clinical survey of uveitis in HTLV-1 endemic region. Ocular treatment. Annals of the Rheumatic Diseases, 65, 1677–8.
Immunology and Inflammation, 17, 335–41. Nigro, G., Zerbini, M., Krzysztofiak, A., Gentilomi, G., Porcaro, M.A., Mango,
Moffatt, S., Tanaka, N., Tada, K., Nose, M., Nakamura, M., Muraoka, O., T., and Musiani, M. (1994). Active or recent parvovirus B19 infection in
Hirano, T., and Sugamura, K. (1996). A cytotoxic nonstructural protein, children with Kawasaki disease. Lancet, 343, 1260–1.
CHAPTER 41 miscellaneous forms of vasculitis 593

Numata, S., Kanda, K., Tanda, S., Inoue T., Doi, K., Sasaki S., and Yaku, H. Park, H.J. and Ranganathan, P. (2011). Neoplastic and paraneoplastic vasculi-
(2005). Sarcoidosis with double saccular abdomainl aortic aneurysm. tis, vasculopathy, and hypercoagulability. Rheumatic Diseases Clinics of
Journal of Vascular Surgery, 41, 1065. North America, 37, 593–606.
Nunes, H., Humbert, M., Capron, F., Brauner, M., Sitbon, O., Battesti, J.P., Pasanisi, E., Vincenti, V., Bacciu, A., Guida, M., Berghenti, T., Barbot, A.,
Simonneau, G., and Valeyre, D. (2006). Pulmonary hypertension asso- Orsoni, J.G., and Bacciu, S. (2003). Cochlear implantation and Cogan
ciated with sarcoidosis:  mechanisms, haemodynamics and prognosis. syndrome. Otology and Neurotology, 24, 601–4.
Thorax, 61, 68–74. Pati, S., Muley, S.A., Grill, M.F., Coons, S., and Walker, R. (2011).
Nurnberg, W., Grabbe, J., and Czarnetzki, B.M. (1995). Urticarial vasculi- Post-streptococcal vasculopathy with evolution to Degos’ disease.
tis syndrome effectively treated with dapsone and pentoxifylline. Acta Journal of the Neurological Sciences, 300, 157–9.
Dermato-Venereologica, 75, 54–6. Pavlovic, M., Kats, A., Cavallo, M., and Shoenfeld, Y. (2010). Clinical and
Ogawa, K., Ohmori, M., Kobayashi, S., et  al. (1967). An autopsy case of molecular evidence for association of SLE with parvovirus B19. Lupus,
papulose atrophiante maligne: The first case in Japan. Acta Pathologica 19, 783–92.
Japonica, 17, 457–63. Pertuiset, E., Liote, F., Launay-Russ, E., Kemiche, F., Cerf-Payrastre, I.,
Oh, S.J., Slaughter, R., and Harrell, L. (1991). Paraneoplastic vasculitic neu- and Chesneau, A.M. (2000). Adult Henoch-Schönlein purpura asso-
ropathy: a treatable neuropathy. Muscle and Nerve, 14, 152–6. ciated with malignancy. Seminars in Arthritis and Rheumatism,
Ohtsuka, T. and Yamazaki, S. (2005). Prevalence of human parvovirus B19 29, 360–7.
component NS1 gene in patients with Henoch-Schonlein purpura Petri, M., Barr, E., Cho, K., and Farmer, E. (1988). Overlap of granuloma-
and hypersensitivity vasculitis. British Journal of Dermatology, 152, tous vasculitis and sarcoidosis: presentation with uveitis, eosinophilia,
1080–1. leg ulcers, sinusitis, and past foot drop. Journal of Rheumatology, 15,
Okazaki, K., Uchida, K., Koyabu, M., Miyoshi, H., and Takaoka, M. (2011). 1171–3.
Recent advances in the concept and diagnosis of autoimmune pan- Picard, O., Brunereau, L., Pelosse, B., Kerob, D., Cabane, J., and Imbert, J.C.
creatitis and IgG4-related disease. Journal of Gastroenterology, 46, (1997). Cerebral infarction associated with vasculitis due to varicella
277–88. zoster virus in patients infected with the human immunodeficiency
Olmos, L., Hunziker, N., and Laugier, P. (1979). Microcylinders of endoplas- virus. Biomedical Pharmacotherapy, 51, 449–54.
mic reticulum in histiocytes in patients suffering from Degos’ syndrome Pinals, R.S. (1978). Cogan’s syndrome with arthritis and aortic insufficiency.
and dermatomyositis. British Journal of Dermatology, 100, 137–45. Journal of Rheumatology, 5, 294–8.
Olmos, L. and Laugier, P. (1977). Ultrastructural study of a new case of Plaza, J.A., Garrity, J.A., Dogan, A., Ananthamurthy, A., Witzig, T.E.,
Degos’ disease, with a review of the literature. Annals of Dermatology and Salomao, D.R. (2011). Orbital inflammation with IgG4-positive
and Venereology, 104, 280–93. plasma cells:  manifestation of IgG4 systemic disease. Archives of
Orme, H.T., Smith, A.G., Nagel, M.A., Bert, R.J., Mickelson, T.S., and Gilden, Ophthalmology, 129, 421–8.
D.H. (2007). VZV Spinal cord infarction identified by diffusion-weighted Pober, J.S. and Sessa, W.C. (2007). Evolving functions of endothelial cells in
MRI (DWI). Neurology, 69, 398–400. inflammation. Nature Reviews Immunology, 7, 803–15.
Orsoni, J.G., Laganà, B., Rubino, P., Zavota, L., Bacciu, S, and Mora, P. (2010). Podjasek, J.O., Wetter, D.A., Pittelkow, M.R., and Wada D.A. (2012).
Rituximab ameliorated severe hearing loss in Cogan’s syndrome: a case Cutaneous small-vessel vasculitis associated with solid organ malignan-
report. Orphanet Journal of Rare Diseases, 5, 18. cies: the Mayo Clinic experience, 1996 to 2009. Journal of the American
Ortiz, G. and Gutierrez, J. (2011). HIV/AIDS patients with HIV vasculopathy Academy of Dermatology, 66, e55–65.
and VZV vasculitis a case series. Clinical Neuroradiology, 21, 145–51. Ponge, T., Boutoille, D., Moreau, A., Germaud, P., Dabouis, G., Baranger, T.,
Oskoui, R., Davis, W.A., and Gomes, M.N. (1993). Salmonella aortitis. and Barrier, J. (1998). Systemic vasculitis in a patient with small-cell
A report of a successfully treated case with a comprehensive review of neuroendocrine bronchial cancer. European Respiratory Journal, 12,
the literature. Archives of Internal Medicine, 153, 517–25. 1228–9.
Ozdogan, H., Arisoy, N., Kasapcapur, O., Sever, L., Caliskan, S., Tuzuner, N., Popper, H.H., Klemen, H., Colby, T.V., and Churg, A. (2003). Necrotizing
Mat, C., and Yazici, H. (1997). Vasculitis in familial Mediterranean fever. sarcoid granulomatosis—is it different from nodular sarcoidosis?
Journal of Rheumatology, 24, 323–7. Pneumonologie, 57, 268–71.
Ozen, S., Ben-Chetrit, E., Bakkaloglu, A., Gur, H., Tinaztepe, K., Calguneri, Prakash, J.A., Sohan Lal, T., Rosemol, V., Verghese, V.P., Pulimood, S.A.,
M., Turgan, C., Turkmen, A., Akpolat, I., Danaci, M., Besbas, N., Reller, M., and Dumler, J.S. (2012). Molecular detection and analysis of
and Akpolat, T. (2001). Polyarteritis nodosa in patients with Familial spotted fever group Rickettsia in patients with fever and rash at a ter-
Mediterranean Fever (FMF): a concomitant disease or a feature of FMF? tiary care centre in Tamil Nadu, India. Pathogens and Global Health,
Seminars in Arthritis and Rheumatism, 30, 281–7. 106, 40–5.
Paajanen, H., Heikkinen, M., Tarvainen, R., Vornanen, M., and Paakkonen, Prayson, R.A. (1999). Granulomatous myositis. Clinicopathologic study of
M. (1995). Anaplastic colon carcinoma associated with necrotizing vas- 12 cases. American Journal of Clinical Pathology, 112, 63–8.
culitis. Journal of Clinical Gastroenterology, 21, 168–9. Premalatha, S., Yesudian, P., Janaki, V.R., et al. (1980). Malignant atrophic
Palazzo, E., Bourgeois, P., Meyer, O., DeBandt, M., Kazatchkine, M., and papulosis (Degos’ syndrome): First case report from India. Clinical and
Kahn, M.F. (1993). Hypocomplementemic urticarial vasculitis syn- Experimental Dermatology, 5, 370–1.
drome, Jaccoud’s syndrome, valvulopathy: a new syndromic combina- Pysden, K.S., Long, V., and Colin, D. (2009). Cogan’s syndrome: a rare cause
tion. Journal of Rheumatology, 20, 1236–40. of meningoencephalitis. Journal of Child Neurology, 24, 753.
Pallensen, R.M. and Rasmussen, N.R. (1979). Malignant atrophic papulosis Qian, Q., Kashani, K.B., and Miller, D.V. (2009). Ruptured abdominal aor-
(Degos’ syndrome). Acta Chirurgica Scandinavica, 145, 279–83. tic aneurysm related to IgG4 periaortitis. New England Journal of
Pankhurst, T., Savage, C.O., Gordon, C., and Harper, L. (2004). Malignancy Medicine, 361, 1121–3.
is increased in ANCA-associated vasculitis. Rheumatology (Oxford), 43, Raza, K., Karokis, D., and Kitas, G.D. (1998). Cogan’s syndrome with
1532–5. Takayasu’s arteritis. British Journal of Rheumatology, 37, 369–72.
Paramo, J.A., Rocha, E., Cuesta, B., et al. (1985). Fibrinolysis in Degos’ dis- Reddy, U. P. (2008). Post-streptococcal vasculitis. Ocular Immunology and
ease. Thrombosis and Haemostasis, 54, 730. Inflammation, 16, 35–6.
Park, H., Lee, J.H., Gouin, E., Cossart, P., and Izard, T. (2011). The rickettsia Reed, R.J. (1981). The T-lymphocyte, the mucinous epithelial interstitium,
surface cell antigen 4 applies mimicry to bind to and activate vinculin. and immunostimulation. American Journal of Dermatopathology, 3,
Journal of Biological Chemistry, 286, 35096–103. 207–14.
594 SECTION 5 vasculitic diseases and syndromes and related disorders

Reinte, L., Taglione, E., and Berretini, S. (1996). Efficacy of methotrexate in Saldana, M.J., Patchefsky, A.S., Israel, H.I., and Atkinson, G.W. (1977).
Cogan’s syndrome. Journal of Rheumatology, 23, 1830–31. Pulmonary angiitis and granulomatosis. The relationship between histo-
Rich, S.A. (1981). Human lupus inclusions and interferon. Science, logical features, organ involvement, and response to treatment. Human
213, 772–5. Pathology, 8, 391–409.
Rich, S.A. and Owens, T.R. (1982). Inducibility of human lupus inclusions by Salvarani, C., Farnetti, E., Casal, B., Nicoli, D., Wenlan, L., Bajocchi, G.,
interferons. Journal of Cellular Biochemistry, S6, 279A. Macchioni, P., Lo Scocco, G., Grazia Catanoso, M., Boiardi, L., and
Richards, A.L. (2012). Worldwide detection and identification of new and Cantini, F. (2002). Detection of parvovirus B19 DNA by polymerase
old rickettsiae and rickettsial diseases. FEMS Immunology and Medical chain reaction in giant cell arteritis: a case-control study. Arthritis and
Microbiology, 64, 107–10. Rheumatism, 46, 3099–101.
Richardson, B. (1994). Methotrexate therapy for hearing loss in Cogan’s syn- Sanchez, N.B., Canedo, I.F., Garcia-Patos, P.E., de Unamuno Perez, P., Benito,
drome. Arthritis and Rheumatism, 37, 1559–61. A.V., and Pascual, A.M. (2004). Paraneoplastic vasculitis associated with
Roat, M.I., Thoft, R.A., Thomson, A.W., Jain, A., Funf, J.J., and Starzl, T.E. multiple myeloma. Journal of the European Academy of Dermatology
(1991). Treatment of Cogan’s syndrome with FK506:  a case report. and Venereology, 18, 731–5.
Transplantation Proceedings, 23, 3347. Sanchez, N.P., Winkelmann, R.K., Schroeter, A.L., and Dicken, C.H. (1982).
Rodriguez-Pla, A., Bosch-Gil, J.A., Echevarria-Mayo, J.E., Rossello-Urgell, J., The clinical and histopathologic spectrums of urticarial vasculitis: study
Solans-Laque, R., Huguet-Redecilla, P., Stone, J.H., and Vilardell-Tarres, of forty cases. Journal of the American Academy of Dermatology, 7,
M. (2004). No detection of parvovirus B19 or herpesvirus DNA in giant 599–605.
cell arteritis. Journal of Clinical Virology, 31, 11–5. Sanchez-Guerrero, J., Gutierrez-Urena, S., Vidaller, A., Reyes, E., Iglesias,
Rodriguez-Serna, M., Fortea, J.M., Perez, A., Febrer, I., Ribes, C., and Aliaga, A., and Alarcon-Segovia, D. (1990). Vasculitis as a paraneoplastic
A. (1993). Erythema elevatum diutinum associated with celiac dis- syndrome. Report of 11 cases and review of the literature. Journal of
ease: response to a gluten-free diet. Pediatric Dermatology, 10, 125–8. Rheumatology, 17, 1458–62.
Rosa, D.J., Machado, R.F., Fraga Rde, O., Scotton, A.S., de Souza, V.A., Sangueza, O.P., Pilcher, B., and Martin Sangueza, J. (1977). Erythema ele-
and Fernandes, B. (2012). Erythema elevatum diutinum as a differen- vatum diutinum: a clinicopathological study of eight cases. American
tial diagnosis of rheumatic diseases: case report. Revista Brasileira de Journal of Dermatopathology, 19, 214–22.
Reumatologia, 52, 291–4. Santonja, C. (2011). Immunohistochemical detection of parvovirus B19 in
Rose, C.D., Eichenfield, A.H., Goldsmith, D.P., and Athreya, B.H. (1990). ‘gloves and socks’ papular purpuric syndrome: direct evidence for viral
Early onset sarcoidosis with aortitis—juvenile systemic granulomatosis. endothelial involvement. Report of three cases and review of the litera-
Journal of Rheumatology, 17, 102–6. ture. American Journal of Dermatopathology, 33, 790–5.
Rosen, C.L., DePalma, L., and Morita, A. (2000). Primary angiitis of the cen- Saravanan, M. and Turnbull, I.M. (2009). Brain:  non-infective and
tral nervous system as a first presentation in Hodgkin’s disease: a case non-neoplastic manifestations of HIV. British Journal of Radiology, 82,
report and review of the literature. Neurosurgery, 46, 1504–10. 956–65.
Rosen, Y., Moon, S., Huang, C.T., Gourin, A., and Lyons, H.A. (1977). Sartwell, P.E. (1976). Racial differences in sarcoidosis. Annals of the New York
Granulomatous pulmonary angiitis in sarcoidosis. Archives of Pathology Academy of Sciences, 278, 368.
and Laboratory Medicine, 101, 170–4. Sato, Y., Notohara, K., Kojima, M., Takata, K., Masaki, Y., and Yoshino, T.
Rowley, A.H., Wolinsky, S.M., Relman, D.A., Sambol, S.P., Sullivan, J., Terai, (2010). IgG4-related disease:  historical overview and pathology of
M., and Shulman, S.T. (1994). Search for highly conserved viral and hematological disorders. Pathology International, 60, 247–58.
bacterial nucleic acid sequences corresponding to an etiologic agent of Sato, Y., Ohshima, K., Ichimura, K., Sato, M., Yamadori, I., Tanaka, T.,
Kawasaki disease. Pediatric Research, 36, 567–71. Takata, K., Morito, T., Kondo, E., and Yoshino, T. (2008). Ocular
Rybicki, B.A., Major, M., Popovich, J. Jr, et al. (1997). Racial differences in adnexal IgG4-related disease has uniform clinicopathology. Pathology
sarcoidosis incidence: a 5-year study in a health maintenance organiza- International, 58, 465–70.
tion. American Journal of Epidemiology, 145, 234–41. Schaff, Z., Heine, U., and Dalton, A.J. (1972). Ultramorphological and ultra-
Sackier, J.M., Kelly, S.B., Clarke, D., Rees, A.J., and Wood, B. (1991). Small cytochemical studies of tubuloreticular structures in lymphoid cells.
bowel haemorrhage due to cytomegalovirus vasculitis. Gut, 32, 1419–20. Cancer Research, 32, 2696.
Saeki, T., Imai, N., Ito, T., Yamazaki, H., and Nishi, S. (2009). Membranous Scheinfeld, N. (2011). Commentary on Degos disease A  C5b-9/
nephropathy associated with IgG4-related systemic disease and without interferon-α-mediated endotheliopathy syndrome by Magro
autoimmune pancreatitis. Clinical Nephrology, 71, 173–8. et al: A reconsideration of Degos disease as hematologic or endothelial
Saeki, T., Nishi, S., Ito, T., Yamazaki, H., Miyamura, S., Emura, I., Imai, N., genetic disease. Dermatology Online Journal, 17, 6.
Ueno, M., Saito, A., and Gejyo, F. (2007). Renal lesions in IgG4-related Schuknecht, H.F. and Nadol, J.B. (1994). Temporal bone pathology in a case
systemic disease. Internal Medicine, 46, 1365–71. of Cogan’s syndrome. Laryngoscope, 104, 1135–42.
Sah, R.P. and Chari, S.T. (2011). Serologic issues in IgG4-related systemic dis- Schwartz, H.R., McDuffie, F.C., Black, L.F., Schroeter, A.L., and Conn, D.L.
ease and autoimmune pancreatitis. Current Opinion in Rheumatology, (1982). Hypocomplementemic urticarial vasculitis: association with chronic
23, 108–13. obstructive pulmonary disease. Mayo Clinic Proceedings, 57, 231–8.
Sahni, S.K. and Rydkina, E. (2009). Host-cell interactions with pathogenic Scotto, G., Cibelli, D.C., Saracino, A., Prato, R., Palumbo, E., Fazio, V.,
Rickettsia species. Future Microbiology, 4, 323–39. Scarabaggio, T., Monno, L., and Angarano, G. (2006). Cryoglobulinemia
Saidha, S., Sotirchos, E.S., and Eckstein, C. (2012). Etiology of sarcoido- in subjects with HCV infection alone, HIV infection and HCV/HIV
sis: does infection play a role? Yale Journal of Biology and Medicine, coinfection. Journal of Infection, 52, 294–9.
85, 133–41. Shah, P., Luqmani, R.A., Murray, P.I., Honan, W.P., Corridan, P.G.J., and
Saigal, K., Valencia, I.C., Cohen, J., and Kerdel, F.A. (2003). Emery, P. (1994). Posterior scleritis: an unusual manifestation of Cogan’s
Hypocomplementemic urticarial vasculitis with angioedema, a rare pres- syndrome. British Journal of Rheumatology, 33, 774–5.
entation of systemic lupus erythematosus: rapid response to rituximab. Shapiro, M.R. (2010). Rickettsia 364D:  a newly recognized cause of
Journal of the American Academy of Dermatology, 49 (5 Suppl.), S283–5. eschar-associated illness in California. Clinical Infectious Diseases,
Sakalli, H., Baskin, E., Bayrakçi, U.S., Melek, E., Cengiz, N., and Ozdemir, 50, 541–8.
B.H. (2010). Parvovirus B19-induced multisystemic vasculitis and acute Sharifi, R., Sinclair, J.C., Gilmour, K.C., Arkwright, P.D., Kinnon, C., Thrasher,
endocapillary proliferative glomerulonephritis in a child. Renal Failure, A.J., and Gaspar, H.B. (2004). SAP mediates specific cytotoxic T-cell
32, 506–9. functions in X-linked lymphoproliferative disease. Blood, 103, 3821–7.
CHAPTER 41 miscellaneous forms of vasculitis 595

Sheehy, N., Sheehan, K, Brett, F., Kay, E., Grogan, L., and Delanty, N. (2003). Stephansson, E.A., Niemi, K.M., Jouhikainen, T., Vaarala, O., and Palosuo, T.
Hodgkins disease presenting as granulomatous angiitis of the central (1991). Lupus anticoagulant and the skin. A longterm follow-up study
nervous system. Journal of Neurology, 250, 112–3. of SLE patients with special reference to histopathological findings. Acta
Shi, R.J., Simpson-Haidaris, P.J., Lerner, N.B., Marder, V.J., Silverman, D.J., Dermato-Venereologica, 71, 416–22.
and Sporn, L.A. (1998). Transcriptional regulation of endothelial cell Stone, J.H., Khosroshahi, A., Deshpande, V., and Stone, J.R. (2010).
tissue factor expression during Rickettsia rickettsii infection: involve- IgG4-related systemic disease accounts for a significant proportion of
ment of the transcription factor NF-κB. Infection and Immunity, 66, thoracic lymphoplasmacytic aortitis cases. Arthritis Care and Research
1070–5. (Hoboken), 62, 316–22.
Shigemitsu, H., Nagai, S., and Sharma, O.P. (2007). Pulmonary hyperten- Stone, J.H., Zen, Y., and Deshpande, V. (2012). IgG4-related disease. New
sion and granulomatous vasculitis in sarcoidosis. Current Opinion in England Journal of Medicine, 366, 539–51.
Pulmonary Medicine, 13, 434–8. Stone, J.R. (2011). Aortitis, periaortitis, and retroperitoneal fibrosis, as
Shimizu, S., Nakamura, Y., Togawa, Y., Kamada, N., Kambe, N., and Matsue, manifestations of IgG4-related systemic disease. Current Opinion in
H. (2008). Erythema elevatum diutinum with primary Sjogren syn- Rheumatology, 23, 88–94.
drome associated with IgA antineutrophil cytoplasmic antibody. British Strickland, D.K. and Ware, R.E. (1995). Urticarial vasculitis: an autoimmune
Journal of Dermatology, 159, 733–5. disorder following therapy for Hodgkin’s disease. Medical and Pediatric
Silver, B., Nagel, M.A., Mahalingam, R., Cohrs, R., Schmid, D.S., and Gilden, Oncology, 25, 208–12.
D. (2012). Varicella zoster virus vasculopathy: A treatable form of rap- Strole, W.E. Jr, Clark, W.H. Jr, and Isselbacher, K.J. (1967). Progressive arterial
idly progressive multi-infarct dementia after 2years’ duration. Journal of occlusive disease (Kohlmeier-Degos): A frequently fatal cutaneo-systemic
the Neurological Sciences, 323, 245–7. disorder. New England Journal of Medicine, 276, 195–201.
Simon, A., Drewe, E., van der Meer, J.W., Powell, R.J., Kelley, R.I., Stalenhoef, Sturgess, A.S. and Littlejohn, G.O. (1988). Jaccoud’s arthritis and panvasculi-
A.F., and Drenth, J.P. (2004). Simvastatin treatment for inflammatory tis in the hypocomplementemic urticarial vasculitis syndrome. Journal
attacks of the hyperimmunoglobulinemia D and periodic fever syn- of Rheumatology, 15, 858–61.
drome. Clinical Pharmacology and Therapeutics, 75, 476–83. Su, W.P., Schroeter, A.L., Lee, D.A., Hsu, T., and Muller, S.A. (1985). Clinical
Simsek, I., Pay, S., Pekel, A., Dinc, A., Musabak, U., Erdem, H., and Sengul, A. and histologic findings in Degos’ syndrome (malignant atrophic papu-
(2007). Serum proinflammatory cytokines directing T helper 1 polari- losis). Cutis, 35, 131–8.
zation in patients with familial Mediterranean fever. Rheumatology Suarez, J., Miguelez, M., and Villalba, R. (1998). Nodular erythema elevatum
International, 27, 807–11. diutinum in an HIV-1 infected woman: response to dapsone and antiret-
Sivakumar, M. and Chee, S.P. (1998). A case series of ocular disease as the pri- roviral therapy. British Journal of Dermatology, 138, 717–8.
mary manifestation in sarcoidosis. Annals of the Academy of Medicine Subbiah, P., Wijdicks, E., Muenter, M., Carter, J., and Connolly, S. (1996).
Singapore, 27, 560–6. Skin lesion with a fatal neurologic outcome (Degos’ disease). Neurology,
Slaviero, F., Annes, R.D., Frighetto, L., Schirmer, L.M., Vanzin, J.R., Fröhlich, 46, 636–40.
A.C., Ferraz, J., and Azambuja, N.D. Jr (2009). Köhlmeier-Degos disease Sumegi, J., Huang, D., Lanyi, A., et al. (2000). Correlation of mutations of the
(malignant atrophic papulosis) and neurologic involvement. Arquivos SH2D1A gene and epstein-barr virus infection with clinical phenotype
de Neuro-psiquiatria, 67, 692–4. and outcome in X-linked lymphoproliferative disease. Blood, 96, 3118–25.
Slavov, S.N. (2011). Human parvovirus B19:  general considerations and Sweiss, N., Patterson, K., Sawaqed, R., Jabbar, U., Korsten, P., Hogarth, K.,
impact on patients with sickle-cell disease and thalassemia and on blood Wollman, R., Garcia, J.G., Niewold, T.B., and Baughman, R.P. (2010).
transfusions. FEMS Immunology and Medical Microbiology, 62, 247–62. Rheumatologic manifestations of sarcoidosis. Seminars in Respiratory
Solans-Laque, R., Bosch-Gil, J.A., Perez-Bocanegra, C., Selva-O’Callaghan, and Critical Care Medicine, 31, 463–73.
A., Simeon-Aznar, C.P., and Vilardell-Tarres, M. (2008). Paraneoplastic Takada, K., Aksentijevich, I., Mahadevan, V., Dean, J.A., Kelley, R.I., and
vasculitis in patients with solid tumors: report of 15 cases. Journal of Kastner, D.L. (2003). Favorable preliminary experience with etanercept
Rheumatology, 35, 294–304. in two patients with the hyperimmunoglobulinemia D and periodic
Soldo-Juresa, D., Radman, M., Pejsa, V., and Bozikov, V. (2010). Reactive fever syndrome. Arthritis and Rheumatism, 48, 2645–51.
macrophage activation syndrome in a patient with parvovirus B19 Takemura, T., Matsui, Y., Oritsu, M., Akiyama, O., Hiraga, Y., Omichi, M.,
infection, lymphocytic lichenoid vasculitis, urticaria and angioedema. Hirasawa, M., Saiki, S., Tamura, S., and Mochizuki, I. (1991). Pulmonary
Annals of Saudi Medicine, 30, 88–9. vascular involvement in sarcoidosis: granulomatous angiitis and micro-
Soni, B.P., Wiiliford, P.M., and White, W.L. (1998). Erythematous nodules in angiopathy in transbronchial lung biopsies. Virchows Archives in
a patient infected with the human immunodeficiency virus. Erythema Pathology Anatomy and Histopathology, 418, 361–8.
elevatum diutinum (EED). Archives of Dermatology, 134, 232–6. Takemura, T., Shishiba, T., Akiyama, O., Oritsu, M., Matsui, Y., and Eishi,
Soravia-Dunand, V.A., Loo, V.G., and Salit, I.E. (1999). Aortitis due to Y. (1997). Vascular involvement in cutaneous sarcoidosis. Pathology
Salmonella: report of 10 cases and comprehensive review of the litera- International, 47, 84–9.
ture. Clinical Infectious Diseases, 29, 862–8. Talaat, K.R., Rothman, J.A., Cohen, J.I., et al. (2009). Lymphocytic vasculitis
Soter, N.A., Murphy, G.F., and Mihm, M.C. Jr (1982). Lymphocytes and involving the central nervous system occurs in patients with X-linked
necrosis of the cutaneous microvasculature in malignant atrophic papu- lymphoproliferative disease in the absence of Epstein-Barr virus infec-
losis: A refined light microscopy study. Journal of American Academy of tion. Pediatric Blood Cancer, 13, 229–36.
Dermatology, 7, 620–30. Tanaka, S., Komori, K., Okadome, K., Sugimachi, K., and Mori, R. (1994).
Sotrel, A., Lacson, A.G., and Huff, K.R. (1983). Childhood Kohlmeier-Degos Detection of active cytomegalovirus infection in inflammatory aortic
disease with atypical skin lesions. Neurology, 33, 1146–51. aneurysms with RNA polymerase chain. Journal of Vascular Surgery,
Stankovic, K., Miailhes, P., Bessis, D., Ferry, T., Broussolle, C., and Sève, P. 20, 235–43.
(2007). Kawasaki-like syndromes in HIV-infected adults. Journal of Taniwaki, S., Kataoka, M., Tanaka, H., Mizuno, Y., and Hirose, M. (1997).
Infection, 55, 488–494. Multiple ulcers of the ileum due to Cytomegalovirus infection in a
Stashower, M.E., Rennie, T.A., Turiansky, G.W., and Gilliland, W.R. (1999). patient who showed no evidence of an immunocompromised state.
Ovarian cancer presenting as leukocytoclastic vasculitis. Journal of Journal of Gastroenterology, 32, 548–52.
American Academy of Dermatology, 40, 287–9. Tasanen, K., Raudasoja, R., Kallioinen, M., and Ranki, A. (1997). Erythema
St Clair, E.W. and McCallum, R.M. (1999). Cogan’s syndrome. Current elevatum diutinum in association with coeliac disease. British Journal of
Opinion in Rheumatology, 11, 47–52. Dermatology, 136, 624–7.
596 SECTION 5 vasculitic diseases and syndromes and related disorders

Tatsis, E., Reinhold-Keller, E., Steindorf, K., Feller, A.C., and Gross, W.L. van der Ven, A., van Oostenbrugge, R., Kubat, B., and Cohen Tervaert, J.W.
(1999). Wegener’s granulomatosis associated with renal cell carcinoma. (2002). Cerebral vasculitis after initiation antiretroviral therapy. AIDS,
Arthritis and Rheumatism, 42, 751–6. 16, 2362–4.
Tatum, E.T., Sun, P.C., and Cohn, D.L. (1989). Cytomegalovirus vasculitis van Son, J.A., O’Marcaigh, A.S., Edwards, W.D., Julsrud, P.R., and Danielson,
and colon perforation in a patient with the acquired immunodeficiency G.K. (1995). Successful resection of thoracic aortic aneurysms in
syndrome. Pathology, 21, 235–8. Wiskott-Aldrich syndrome. Annals of Thoracic Surgery, 60, 685–7.
Tauber, E., Wojnarowski, C., Horcher, E., Dekan, G., and Frischer, T. (1999). Vella, J.P., O’Callaghan, J., Hickey, D., and Walshe, J.J. (1997). Renal artery ste-
Necrotizing sarcoid granulomatosis in a 14-yr-old female. European nosis complicating Cogan’s syndrome. Clinical Nephrology, 47, 407–8.
Respiratory Journal, 13, 703–5. Veraldi, S., Mancuso, R., Rizzitelli, G., Gianotti, R., and Ferrante, P. (1999).
Tedeschi, A., Barate, C., Minola, E., and Morra, E. (2007). Cryoglobulinemia. Henoch-Schonlein syndrome associated with human Parvovirus B19
Blood Reviews, 21, 183–200. primary infection. European Journal of Dermatology, 9, 232–3.
Tekin, M., Yalcinkaya, F., Tumer, N., Akar, N., Misirlioglu, M., and Cakar, Vincent, D., Dubas, F., Hauw, J.J., Godeau, P., Lhermitte, F., Buge, A., and
N. (2000). Clinical, laboratory and molecular characteristics of chil- Castaigne, P. (1986). Nerve and muscle microvasculitis in periph-
dren with Familial Mediterranean Fever-associated vasculitis. Acta eral neuropathy:  a remote effect of cancer? Journal of Neurology,
Paediatrica, 89, 177–82. Neurosurgery, and Psychiatry, 49, 1007–10.
Thomas, H.G. (1992). Case report:  clinical and radiological features of Vital, A., Lagueny, A., Ferrer, X., Louiset, P., Canron, M.H., and Vital, C.
Cogan’s syndrome—non-syphilitic interstitial keratitis, audiovestibular (2008). Sarcoid neuropathy: clinico-pathological study of 4 new cases
symptoms and systemic manifestations. Clinical Radiology, 45, 418–21. and review of the literature. Clinical Neuropathology, 27, 96–105.
Ting, A.C., Cheng, S.W., Ho, P., Poon, J.T., and Tsu, J.H. (2005). Surgical treat- Walker, D.H. (1989). Rocky Mountain spotted fever:  a disease in need of
ment of infected aneurysms and pseudoaneurysms of the thoracic and microbiological convern. Clinical Microbiology Reviews, 2, 227.
abdominal aorta. American Journal of Surgery, 189, 150–4. Walker, D.H. (1990). The role of host factors in the severity of spotted fever
Tong, D., Manolios, N., Howe, G., and Spencer, D. (2012). New onset and typhus rickettsioses. Annals of the New York Academy of Sciences,
sarcoid-like granulomatosis developing during anti-TNF therapy:  an 590, 10–9.
under-recognised complication. Internal Medicine Journal, 42, 89–94. Walker, D.H., Gay, R.M., and Valdes-Dapena, M. (1981). The occurrence
Torok, L., Husz, S., Korom, I., Horvath, K., and Forizs, A. (1993). Systemic of eschars in Rocky Mountain spotted fever. Journal of the American
lupus erythematosus with pigmented skin. Cutis, 51, 433–6. Academy of Dermatology, 4, 571–6.
Touma, Z., Nawwar, R., Hadi, U., Hourani, M., and Arayssi, T. (2007). Walker, D.H. and Ismail, N. (2008). Emerging and re-emerging rickettsi-
The use of TNF-alpha blockers in Cogan’s syndrome. Rheumatology oses: endothelial cell infection and early disease events. Nature Reviews
International, 27, 995–6. Microbiology, 6, 375–86.
Trapani, S., Ermini, M., and Falcini, F. (1999). Human parvovirus B19 infec- Walker, D.H. and Raoult, D. (2010). Rickettsia rickettsii and other spotted
tion:  its relationship with systemic lupus erythematosus. Seminars in fever group rickettsiae (Rocky Mountain spotted fever and other spot-
Arthritis and Rheumatism, 28, 319–25. ted fevers). In Principles and practice of infectious diseases, 6th edn.
Trendelenburg, M., Courvoisier, S., Spath, P.J., Moll, S., Mihatsch, M., Itin, P., (ed. G.L. Mandell, J.E. Bennett, and R. Dolin), pp. 2499–505. Churchill
and Schifferli, J.A. (1999). Hypocomplementemic urticarial vasculitis or Livingstone, Philadelphia, PA, USA.
systemic lupus erythematosus? American Journal of Kidney Diseases, Wang J., Parisier, S.C., Weiss, M.H., Chute, P.M., Hellman, S.A., and Sauris,
34, 745–51. E. (2010). Cochlear implantation in patients with autoimmune inner
Tseng, J.F., Cambria, R.P., Aretz, H.T., and Brewster, B.C. (1999). ear disease including cogan syndrome:  a comparison with age- and
Thoracoabdominal aortic aneurysm in Cogan’s syndrome. Journal of sex-matched controls. Laryngoscope, 120, 2478–83.
Vascular Surgery, 30, 565–8. Wang, J.H., Liu, Y.C., Yen, M.Y., Chen, Y.S., Wann, S.R., and Cheng, D.L.
Tufan, G. and Demir, S. (2010). Uncommon clinical pattern of FMF: pro- (1996). Mycotic aneurysm due to non-typhi salmonella:  report of 16
tracted febrile myalgia syndrome. Rheumatology International, 30, cases. Clinical Infectious Diseases, 23, 743–7.
1089–90. Watanabe, T., Mochizuki, M., and Yamaguchi, K. (1977). HTLV-1 uveitis
Tunca, M., Akar, S., Onen, F., Ozdogan, H., Kasapcopur, O., Yalcinkaya, F., (HU). Leukemia, 11 (Suppl. 3), 582–4.
Tutar, E., Ozen, S., Topaloglu, R., Yilmaz, E., Arici, M., Bakkaloglu, A., Webster, E., Corman, L.C., and Braylan, R.C. (1986). Syndrome of tempo-
Besbas, N., Akpolat, T., Dinc, A., Erken, E.; Turkish FMF Study Group ral arteritis with perivascular infiltration by malignant cells in a patient
(2005). Familial Mediterranean fever (FMF) in Turkey:  results of a with follicular small cleaved cell lymphoma. Journal of Rheumatology,
nationwide multicenter study. Medicine (Baltimore), 84, 1–11. 13, 1163–6.
Tzanck, A., Civatte, A., and Sidi, E. (1948). Presentation de mou- Weiler, V., Redtenbacher, S., Bancher, C., Fischer, M.B., and Smolen, J.S.
lage: Ulerytheme porcelain en gouttes. Bulletin de la Societe Francaise (2000). Concurrence of sarcoidosis and aortitis: case report and review
de Dermatologie et de Syphiligraphie, 55, 10–2. of the literature. Annals of the Rheumatic Diseases, 59, 850–3.
Umehara, H., Okazaki, K., Masaki, Y., Kawano, M., Yamamoto, M., Saeki, T., Weiss, D.J., Greenfield, J.W. Jr, O’Rourke, K.S., and McCune, W.J. (1993).
Matsui, S., Sumida, T., Mimori, T., Tanaka, Y., Tsubota, K., Yoshino, T., Systemic cytomegalovirus infection mimicking an exacerbation of
Kawa, S., Suzuki, R., Takegami, T., Tomosugi, N., Kurose, N., Ishigaki, Y., Wegener’s granulomatosis. Journal of Rheumatology, 20, 155–7.
Azumi, A., Kojima, M., Nakamura, S., and Inoue D. (2012). A novel clin- Weissen-Plenz, G., Sezer, O., Vahlhaus, C., Robenek, H., Hoffmeier, A.,
ical entity, IgG4-related disease (IgG4RD): general concept and details. Tjan, T.D., Scheld, H.H., and Sindermann, J.R. (2010). Aortic dis-
Modern Rheumatology, 22, 1–14. section associated with cogans’s syndrome: deleterious loss of vascu-
Uzman, B.G., Saito, H., and Kasac, M. (1971). Tubular arrays in the endo- lar structural integrity is associated with GM-CSF overstimulation
plasmic reticulum in human tumor cells. Laboratory Investigation, in macrophages and smooth muscle cells. Journal of Cardiothoracic
24, 492–8. Surgery, 5, 66.
van der Hilst, J.C., Drenth, J.P, Bodar, E.J., Bijzet, J., van der Meer, J.W., and Wilkinson, S.M., English, J.S., Smith, N.P., Wilson-Jones, E., and Winkelmann,
Simon, A. (2005). Serum amyloid A serum concentrations and genotype R.K. (1992). Erythema elevatum diutinum: a clinicopathological study.
do not explain low incidence of amyloidosis in Hyper-IgD syndrome. Clinical and Experimental Dermatology, 17, 87–93.
Amyloid, 12, 115–9. Wilson, D., McCluggage, W.G., and Wright, G.D. (2002). Urticarial vascu-
van der Hilst, J.C. and Frenkel, J. (2010). Hyperimmunoglobulin D syndrome litis: a paraneoplastic presentation of B-cell non-Hodgkin’s lymphoma.
in childhood. Current Rheumatology Reports, 12, 101–7. Rheumatology (Oxford), 41, 476–7.
CHAPTER 41 miscellaneous forms of vasculitis 597

Wisnieski, J.J. (2000). Urticarial vasculitis. Current Opinion in Rheumatology, of CMV after solid organ transplantation. Netherlands Journal of
12, 24–31. Medicine, 63, 408–12.
Wisnieski, J.J., Baer, A.N., Christensen, J., Cupps, T.R., Flagg, D.N., Jones, Zeiss, C.R., Burch, F.X., Marder, R.J., Furey, N.L., Schmid, F.R., and Gewurz,
J.V., Katzenstein, P.L., McFadden, E.R., McMillen, J.J., Pick, M.A., H. (1980). A hypocomplementemic vasculitic urticarial syndrome.
et  al. (1995). Hypocomplementemic urticarial vasculitis syndrome. Report of four new cases and definition of the disease. American Journal
Clinical and serologic findings in 18 patients. Medicine (Baltimore), of Medicine, 68, 867–75.
74, 24–41. Zen, Y., Inoue, D., Kitao, A., Onodera, M., Abo, H., Miyayama, S., Gabata,
Wisnieski, J.J. and Jones, S.M. (1992a). Comparison of autoantibodies to the T., Matsui, O., and Nakanuma, Y. (2009a). IgG4-related lung and pleu-
collagen-like region of C1q in hypocomplementemic urticarial vasculitis ral disease: a clinicopathologic study of 21 cases. American Journal of
syndrome and systemic lupus erythematosus. Journal of Immunology, Surgical Pathology, 33, 1886–93.
148, 1396–403. Zen, Y., Onodera, M., Inoue, D., Kitao, A., Matsui, O., Nohara, T., Namiki, M.,
Wisnieski, J.J. and Jones, S.M. (1992b). IgG autoantibody to the collagen-like Kasashima, S., Kawashima, A., Matsumoto, Y., Katayanagi, K., Murata,
region of C1q in hypocomplementemic urticarial vasculitis syndrome, T., Ishizawa, S., Hosaka, N., Kuriki, K., and Nakanuma, Y. (2009b).
systemic lupus erythematosus, and 6 other musculoskeletal or rheu- Retroperitoneal fibrosis: a clinicopathologic study with respect to immu-
matic diseases. Journal of Rheumatology, 19, 884–8. noglobulin G4. American Journal of Surgical Pathology, 33, 1833–9.
Woods, M.E. and Olano, J.P. (2008). Host defenses to Rickettsia rickettsii Zhang, J., Shehabeldin, A., da Cruz, L.A., Butler, J., Somani, A.K., McGavin,
infection contribute to increased microvascular permeability in human M., Kozieradzki, I., dos Santos, A.O., Nagy, A., Grinstein, S., Penninger,
cerebral endothelial cells. Journal of Clinical Immunology, 28, 174–85. J.M., and Siminovitch, K.A. (1999). Antigen receptor-induced activa-
Zakrzewska, K., Corcioli, F., Carlsen, K.M., Giuggioli, D., Fanci, R., Rinieri, tion and cytoskeletal rearrangement are impaired in Wiskott-Aldrich
A., Ferri, C., and Azzi, A. (2009). Human parvovirus B19 (B19V) infec- syndrome protein-deficient lymphocytes. Journal of Experimental
tion in systemic sclerosis patients. Intervirology, 52, 279–82. Medicine, 190, 1329–42.
Zandberg, M., de Maar, E.F., Hofker, H.S., Homan van der Heide, J.J., Rosati, Zurada, J.M., Ward, K.M., and Grossman, M.E. (2006). Henoch-Schonlein
S., and van Son, W.J. (2005). Initial cytomegalovirus prophylaxis with prefurpura associated with malignancy in adults. Journal of the
ganciclovir: no guarantee for prevention of late serious manifestations American Academy of Dermatology, 55 (5 Suppl.), S65–70.
CHAPTER 42

Experimental therapies
for vasculitis
Sebastian Unizony and John H. Stone

Primary systemic vasculitis was not recognized as a clinical entity Large-vessel vasculitis
until 1866, when Adolf Kussmaul and Rudolf Maier described
the case of a 27-year-old journeyman tailor who developed fever, Giant cell arteritis (GCA) and Takayasu’s arteritis (TAK) are the
pain in his extremities and abdomen, and oliguria (Kussmaul most common forms of large-vessel vasculitis (LVV). Although
and Maier 1866). On autopsy, they noted nodular inflamma- clear demographic differences exist between these conditions, both
tory lesions of the medium- and small-sized arteries that today diseases are characterized by the occurrence of a granulomatous
would be recognized as polyarteritis nodosa, the archetype for inflammatory process that affects the aorta and its main branches
all forms of primary systemic vasculitis. In their famous descrip- (Kerr et al. 1994; Salvarani et al. 2002). Increased knowledge of the
tion of this disorder, they wrote that he was ‘one of those patients pathogenesis of LVV has lead to the identification of new therapeu-
for whom one can already give the prognosis before the diagno- tic targets, prompting the initiation of clinical trials.
sis . . . . The first impression was one of a lost soul whose . . . days
were numbered . . . ’. Giant cell arteritis
The prognosis for vasculitis is not nearly so grim in the mod- The aetiology of the immune-mediated response against the arte-
ern era, as the use of corticosteroids (CS) and cytotoxic agents has rial wall in GCA remains unknown, but the mechanism of dis-
dramatically improved the immediate survival of these patients. In ease has been partially clarified (Weyand et al. 2011; Wagner et al.
contrast, decreased mortality has made therapy-related toxicity an 1994). In untreated patients, an expanded repertoire of autoreactive
evident problem. Ironically, treatments themselves frequently lead CD4-positive T lymphocytes, including interferon (IFN)-γ produc-
to the most serious consequences in terms of long-term morbidity ing T helper (Th) 1 cells and interleukin (IL)-17-secreting Th17
and quality of life. cells, orchestrates the formation of granulomatous vascular inflam-
In the last 10  years, modifications to the original mation (Deng et al. 2010). Moreover, the number of FOXP3 regula-
cyclophosphamide-based protocols (i.e. CYCAZAREM trial) tory T cells (Treg), which normally serve to limit an immunological
(Jayne et  al. 2003)  and the emergence of new treatment strate- response, appears to be decreased (Terrier et al. 2012).
gies for ANCA associated vasculitis (i.e. rituximab) (Stone et al. No clearly defined alternative to long-term CS treatment cur-
2010)  have maintained high rates of disease control, with the rently exists for GCA (Mukhtyar et al. 2009). Glucocorticoids are
advantage of decreasing the exposure to glucocorticoids and highly effectively at controlling systemic inflammation and pre-
alkylating agents in an attempt to minimize long-term compli- venting acute damage (i.e. vision loss), but generally fail to cure
cations. Unfortunately, clearly effective glucocorticoid-sparing the disease or to induce long-term remissions (Mazlumzadeh
options have been elusive for disorders such as giant cell arteritis et al. 2006). More than half of the patients relapse upon weaning
and Takayasu’s arteritis, and these patients still require long CS treatment and the majority develop CS-induced toxicity (i.e. bone
courses that invariably lead to undesirable side-effects. Similarly, fractures, cataracts, diabetes, and many others) (Salvarani et  al.
the treatment of other forms of medium and small-sized vessel 2002; Proven et al. 2003). Other immunosuppressive medications,
vasculitis (i.e. cryoglobulinaemic vasculitis, eosinophilic granulo- including methotrexate (MTX), and tumour necrosis factor alpha
matosis with polyangiitis, polyarteritis nodosa, etc.) continues to (TNF-α) antagonists, have generally led to disappointing results
be suboptimal for reasons of both efficacy and safety. (Hoffman et al. 2002; Hoffman et al. 2007; Jover et al. 2001; Spiera
Recent advances in the understanding of the immunopathogen- et al. 2001; Martinez-Taboada et al. 2008).
esis of vasculitides have opened novel treatment avenues. However,
new therapies will need rigorous testing in controlled studies before Anti-IL-6 receptor therapy (tocilizumab)
they can be regarded as efficacious in this spectrum of diseases. IL-6 is a proinflammatory cytokine with a wide variety of bio-
This chapter will describe some of the experimental strategies that logical functions depending on its target cell (Naka et  al. 2002).
may one day become part of a new standard of care for this group Under physiological conditions, IL-6 triggers the synthesis of acute
of immune-mediated disorders. phase proteins, promotes the transition from acute to chronic
600 SECTION 5 vasculitic diseases and syndromes and related disorders

inflammation, and facilitates the development of specific immunity 0–6 mg). Of note, active LVV was observed pathologically in the
(Jones et al. 2005). IL-6 modulates the activation, proliferation, dif- autopsy of an 81-year-old woman who died from an unrelated
ferentiation, and function of different T-cell subpopulations (i.e. cause. This patient had been in clinical and serological remission
cytotoxic CD8 cells, Th17 cells, and Treg cells) (Miossec et al. 2009; for 4 months while on tocilizumab and low-dose prednisone
Sakaguchi et al. 2010), stimulates the terminal differentiation of B Based on these reports, an adequately powered randomized con-
cells, enhances the survival of plasmocytes, and induces cells of the trolled trial designed to rigorously test the efficacy and safety of
monocyte, endothelial, and stromal lineages to acquire an ‘proin- TCZ for GCA (GiACTA) has been initiated in the United States and
flammatory’ phenotype. Europe (Unizony et al. 2013).
In GCA patients, IL-6 is up-regulated within inflamed arteries
Co-stimulation blockade (abatacept)
(Weyand et al. 1994) and its concentration is elevated in periph-
A population of dendritic cells (DCs) that reside at the
eral circulation (Garcia-Martinez et al. 2010; Dasgupta and Panayi
adventitia-media border of normal large arteries may play a role in
1990). Serum IL-6 levels mirror disease activity and decline with
the early stages of GCA (Weyand et al. 2005). After the recognition
adequate CS treatment (Weyand et  al. 2000; Roche et  al. 1993).
of still undefined ‘danger signals’, these antigen-presenting cells
As suggested by research in other immune-mediated conditions,
may be responsible for recruiting lymphocytes into the arterial wall
IL-6 blockade could ameliorate GCA-related vascular inflamma-
(Ma-Krupa et al. 2002; Krupa et al. 2002; Han et al. 2008). Once
tion via multiple possible mechanisms that include: (1) inhibition
in the vascular tissue and properly stimulated by DCs, autoreac-
of the upstream differentiation of autoreactive lymphocytes (prob-
tive T cells would undergo clonal expansion, initiating the arteritic
ably Th17 and Th1) (Fujimoto et  al. 2008; Fujimoto et  al. 2008;
process.
Haruta et al. 2011); (2) generation of Treg cells (Korn et al. 2008);
A pathogenic role for DCs in GCA was suggested by the dem-
and (3)  deamplification of the downstream innate inflammatory
onstration that depletion of this cell type ameliorated vasculitis in
response (Emilie et al. 1994).
human artery–SCID chimeras (Ma-Krupa et al. 2002). Upon anti-
A limited number of GCA patients have been treated with tocili-
gen capture, immature DCs upregulate co-stimulatory molecules
zumab (TCZ), a humanized monoclonal anti-IL-6 receptor (IL-6R)
(i.e. CD80/86) and become an important source of cytokines and
antibody (Salvarani et  al. 2012b; Seitz et  al. 2011; Sciascia et  al.
chemokines. This maturation process endows DCs with the pro-
2011; Beyer et al. 2011; Vinit et al. 2012; Unizony et al. 2012). The
ficiency to interact with CD4 T cells and give specific ‘shape’ and
great majority of these patients had relapsing / refractory disease.
magnitude to an immunological response (Th1, Th2, Th17, etc.).
So far, TCZ (mainly 8 mg/kg every 4 weeks) has been tolerated
On this basis, a randomized, controlled trial of co-stimulatory
without any major safety concern besides expected side-effects (e.g.
blockade with abatacept is currently ongoing for patients with LVV.
cytopenias, mild transaminase elevation, infection), and prelimi-
nary results are encouraging. Takayasu’s arteritis
Seitz et al. (2011) reported rapid and complete clinical improve-
Early TAK lesions mainly consist of adventitial infiltration by T
ment in five patients treated with TCZ for 7–8 months. Two sub-
cells, natural killer (NK) cells, and macrophages. Granulomas con-
jects, treatment naïve, received biological monotherapy. For the
taining giant cells are subsequently found in the tunica media of
other three cases (two previously failing MTX), TCZ was added
elastic arteries. Late-stage (‘burned out’) damage, on the other hand,
to a background of glucocorticoids. Patients achieved and main-
demonstrates extensive fibrosis and intimal hyperplasia. In contrast
tained disease remission while on anti-IL-6R therapy and were able
to GCA, a pathogenic role for B cells has been proposed in TAK not
to taper their prednisone dose from a mean of 19 mg/day (range
only by the identification of polyclonal hypergammaglobulinaemia
10–40 mg) at the first TCZ administration to 3 mg/day (range 0–5
and circulating antiendothelial antibodies (Park et al. 2006; Wang
mg) at 12 weeks. Evidence of active LVV, by MRI, resolved after
et al. 2011), but also the demonstration of an increased number of
3 months of treatment in three individuals. Beyer et al. (2011) also
plasmablasts in peripheral blood (Hoyer et al. 2012) and the obser-
reported good clinical and radiographic response in three patients
vation of B cells infiltrating the tunica adventitia of patient’s aortic
who received TCZ for 6  months, given their refractory disease
samples (Inder et al. 2000).
despite the use of prednisone (mean dose 30 mg/day), azathioprine
As in GCA, CS are the mainstay for the treatment of TAK
(AZA), mycophenolate mofetil (MMF), and MTX. Once on IL-6R
(Mukhtyar et  al. 2009). Unfortunately, relapses, CS dependence,
blockade, all subjects were able to reduce their CS to 7.5 mg/day
and subclinical radiographic progression are seen in more than
or less without experiencing disease exacerbations. Positron emis-
two-thirds of the patients, and further immunosuppression is
sion tomography (PET) / computed tomography (CT) scans, that
often required. Conventional agents (i.e. MTX, AZA, MMF, CYC)
originally showed increased 18-fluorodeoxyglucose (FDG) uptake
have shown limited CS-sparing potential in uncontrolled series
at baseline in two cases, normalized after 24 weeks of treatment.
(Hoffman et al. 1994; Maksimowicz-McKinnon et al. 2007; Goel
Our group published the largest open-label experience to date
et al. 2010). In contrast, retrospective and open-label studies sug-
using TCZ for LVV (Unizony et al. 2012). Within a cohort of ten
gest that TNF-α, IL-6, and B-cell directed therapies might have a
cases, seven patients with GCA whose disease had been highly
role in the treatment of this disease (Hoffman et al. 2004; Tanaka
refractory to glucocorticoids and multiple immunosuppressive
et al. 2006; Molloy et al. 2008; Mekinian et al. 2012; Comarmond
agents (AZA, MTX, cyclophosphamide (CYC), and TNF-α inhibi-
et al. 2012).
tors), rapidly entered and maintain remission while receiving TCZ
for a mean period of time of 8 months (range 6–12). Given their B-cell depletion (rituximab)
excellent clinical response, all patients were able to significantly B-cells not only represent a source of antibodies, but also modu-
reduce their prednisone from an average daily dose of 23 mg at late effector, memory, and regulatory T-cell responses through
baseline (range 8–60 mg) to 2.2 mg at the end of follow-up (range antibody-independent mechanisms (Lund and Randall 2012;
CHAPTER 42 experimental therapies for vasculitis 601

Shlomchik et al. 2001). Some of these mechanisms involve anti- Small-vessel vasculitis
gen presentation (Griffin et  al. 2011)  and cytokine secretion
(Barr et  al. 2012). Furthermore, subpopulations of B cells with Cryoglobulinaemic vasculitis
intrinsic regulatory capacity have been characterized (Mauri and Cryoglobulins are immunoglobulins (Ig) that precipitate in vitro
Bosma 2012). at temperatures below 37°C and dissolve upon rewarming. The
Rituximab (RTX) is a chimeric IgG1 antibody that upon bind- presence of cryoglobulins in circulation (cryoglobulinaemia) can
ing to CD20 depletes circulating naïve and memory B cells for be asymptomatic or lead to a small-vessel vasculitis, often target-
6–12 months via FcγR-mediated antibody-dependent cell cytotox- ing the skin, kidneys, and peripheral nerves (Ramos-Casals et al.
icity and complement-dependent cytotoxicity. Galarza et al. (2008) 2012). Based on its clonality, cryoglobulins are classified in three
described good clinical response to RTX in one of two patients with types (Brouet et al. 1974). Type I or simple cryoglobulinaemia is
TAK refractory to MTX and TNF-α inhibition. Hoyer et al. (2012) defined by monoclonal Ig (IgM or IgG), which may self-aggregate
reported three patients with refractory TAK, despite prednisone, and cause cold-induced vascular occlusions. In contrast, the mixed
mycophenolic acid, ciclosporin, and adalimumab, who responded cryoglobulinaemias are mainly composed by either monoclonal
to B-cell depletion. Of note, anti-CD20 therapy normalized the (type II) or polyclonal (type III) IgM that form aggregates with
number of plasmablasts, which subsequently increased during polyclonal Ig or other molecules (i.e. viral proteins) and precipitate
relapse in two subjects that were successfully re-treated. within the microvasculature, leading to immune complex-mediated
Anti-interleukin (IL)-6 therapy (tocilizumab) inflammation.
Cryoglobulins are produced by one or more expanded B-cell
As in GCA, IL-6 is up-regulated in TAK (Seko et  al. 1996), and
clones. Type I cryoglobulinaemia suggest the presence of a lym-
serum levels correlate with disease activity (Park et al. 2006). During
phoproliferative disease or plasma cell dyscrasia, whereas mixed
the early stages of disease, IL-6 might be important in stimulating T
cryoglobulinaemia tends to arise in the context of persistent
cells and recruiting monocytes to the sites of arterial inflammation.
immune stimulation by chronic infection (i.e. HCV, HIV), auto-
Later on, IL-6 could be involved in promoting vascular angiogen-
immune disease (i.e. Sjögren’s syndrome), or under no obvious
esis and fibrosis.
circumstances in a condition termed idiopathic or essential mixed
Nishimoto et  al. (2008) reported the successful use of TCZ in
cryoglobulinaemia (Trejo et al. 2001).
a patient with refractory TAK for the first time. Since then, nine
additional cases of TAK treated with this agent have been pub- Regulatory T-cell enhancement for HCV-related
lished (Unizony et al. 2012; Salvarani et al. 2012a; Salvarani et al. mixed cryoglobulinaemia
2012b; Seitz et al. 2011; Bredemeier et al. 2012). For these ten cases, Forty to 60% of patients with chronic HCV infection have
IL-6R therapy was utilized for a mean period of 11 months (range circulating cryoglobulins but only 5–10% of them develop
4–41 months). Two cases were treatment naïve and received TCZ cryoglobulinaemia-related symptoms (Cacoub et al. 1999). Given
monotherapy, and eight subjects were refractory to concomitant its high prevalence, however, HCV accounts for 70–90% of the
prednisone (mean dose 23 mg/day; range 5–40 mg/day) and other cases of cryoglobulinaemic vasculitis. Standard of care for this
immunosuppressants (MTX, AZA, MMF, CYC, ciclosporin, inf- entity is discussed in detail in Chapter 40. Here we comment on
liximab, and adalimumab). All subjects achieved disease control, recently explored therapies that deserve some attention.
and those on glucocorticoids were able to either discontinue or Patients with symptomatic HCV-related mixed cryoglobulinae-
significantly taper prednisone after 3–6  months of TCZ therapy. mia have a quantitative deficit of functional Tregs in circulation
Serial imaging in nine patients (MRA n=2; CT n=2; PET/CT n=5) (Boyer et al. 2004). In a pilot study, ten patients with HCV-related
showed improvement of vasculitic features in eight cases. One mixed cryoglobulinaemia intolerant or refractory to conventional
subject relapsed while on TCZ after 8  months of treatment, and treatments received four cycles of low-dose IL-2 therapy within
a second subject flared within 3  months of discontinuing IL-6R 9 weeks (1.5–3 million IU daily SC for 5 days). IL-2 significantly
blockade. Controlled clinical trials that include longitudinal arte- increased the number of CD25high—FOXP3+ Treg cells from a
rial imaging to evaluate the possibility of subclinical vascular dis- mean baseline of 3.6% to 11.8% of circulating CD4 cells with-
ease progression are required at this juncture. out worsening the viral load or precipitating a vasculitic flare.
Newer antimetabolites (leflunomide) Clinical symptoms improved in eight patients and cryoglobulin
levels decreased in seven cases. Transcriptome analysis of periph-
Immunity is dependent on rapidly dividing immune cells that
eral blood mononuclear cells (PBMC) revealed that IL-2 attenu-
require continual synthesis of DNA. Lefunomide, an agent widely
ated inflammatory and oxidative stress gene signatures (Saadoun
used in rheumatoid arthritis, has immunomodulatory effects by
et al. 2011).
inhibiting pyrimidine synthesis.
In a prospective study from Brazil (de Souza et al. 2012), favour- Conventional approach to treatment of non-HCV
able clinical response to leflunomide was seen in 80% of sub- cryoglobulinaemic vasculitis
jects within a group of 15 TAK patients whose disease had been Besides treatment for the underlying disease (i.e. antivirals for
refractory to CS, MTX, AZA, CYC, MMF, and TNF-α antago- HCV, chemotherapy for lymphoma, etc.), when cryoglobulinaemia
nists. Disease activity scores, CRP levels, and CS use significantly causes significant morbidity (i.e. glomerulonephritis), traditional
declined after a median treatment period of 9 months. Of note, at therapeutics are oriented to: (1) decrease the inflammatory con-
the end of follow-up, patients still required a mean daily prednisone sequences of cryoglobulin deposition within the microcirculation
dose of 13.9 mg, and two cases had silent radiographic progression (i.e. glucocorticoids); (2) reduce the cryoglobulin production (i.e.
despite being classified in remission by clinical examination and immunosuppression or B-cell depletion); or (3) remove the path-
laboratory analysis. ogenic Ig (i.e. plasma exchange). Despite this approach a sizable
602 SECTION 5 vasculitic diseases and syndromes and related disorders

subset of patients experiences continued disease activity and there- to one published case of RA-related disease successfully treated
fore poor outcomes (Ramos-Casals et al. 2012). with TCZ (Cohen et al. 2012).
A retrospective analysis of 33 cases of non-HCV mixed cry- Plasma-cell-targeted therapies such as lenalidomide, thalido-
oglobulinaemia (Foessel et  al. 2011)  showed poor response mide, or bortezomib have been rarely used in patients with cry-
to conventional immunosuppression and partial response to oglobulinaemic vasculitis. These subjects, who mainly had type
B-cell-depletion therapy. There was transient improvement with I cryoglobulinaemia refractory to conventional immunosuppres-
CS, but all patients relapsed upon steroid tapering. Further sion (i.e. prednisone, CYC, AZA, and RTX), had favourable clinical
immunosuppression was required in 21 cases (CYC n = 13; aza- response (Foessel et al. 2011; Calabrese et al. 2011; Lin et al. 2010;
thioprine n = 2; mycophenolate n = 5; MTX n = 1; chlorambucil Spizzo et al. 2010; Talamo et al. 2008; Sampson et al. 2006; Cem Ar
n = 2). Except for one patient with rheumatoid arthritis treated et al. 2005; Rodriguez-Paez et al. 2009).
with MTX, complete remission was never achieved. Seven Given the biological rational and the paucity of clinical experi-
patients, including four subjects with idiopathic disease, received ence, IL-6 signalling blockade, proteasome inhibitors, and other
RTX. B-cell depletion induced sustained remission in only two B-cell/ plasmocyte directed therapies (e.g. anti-BLyS agents, Btk
patients. The rest either had partial response or withdrawn ther- inhibitors) need further exploration for the treatment of cryoglo-
apy due to adverse events. Data from the French AutoImmune bulinaemic vasculitis.
Registry (AIR) on 23 cases of non-viral mixed cryoglobulinaemia
treated with RTX showed that among eight patients with essential ANCA-associated vasculitis
cryoglobulinaemia treated with RTX, four had complete clinical Antineutrophil cytoplasmic antibody (ANCA)-associated vascu-
response, one had partial response, and three failed therapy sec- litis (AAV) comprises granulomatosis with polyangiitis (GPA or
ondary to severe infection. Complete immunological response Wegener’s granulomatosis), microscopic polyangiitis (MPA), and
defined as negativization of the serum cryoglobulins and/or nor- eosinophilic granulomatosis with polyangiitis (EGPA or Churg–
malization of C4 levels occurred in only two patients. Most of Strauss syndrome). These related autoimmune diseases, which
the responders relapsed after a median time of 13.5 months and affect the respiratory tract and kidneys among other organs, are
required RTX re-dosing. characterized by the occurrence of pauci-immune necrotizing
inflammation of the microcirculation, and the presence of antibod-
Novel B-cell and plasmocyte-targeted therapies for non-HCV ies directed against neutrophil and macrophage granule compo-
cryoglobulinaemia nents: namely, myeloperoxidase (MPO-ANCA) and proteinase-3
Treatment options beyond classic immunosuppression and B-cell (PR3-ANCA).
depletion have been insufficiently explored in cryoglobulinaemia.
Long-lived autoreactive plasma cells may contribute to the perpet- GPA and MPA
uation of autoimmunity, and possibly represent a target for ther- A complex network of innate and adaptive immunity cells and
apy. Unlike B-lymphocytes, plasmocytes do not have proliferative molecules are involved in the pathogenesis of GPA and MPA. CD4
potential and lack the expression of CD20. Therefore, these cells cells, B lymphocytes, neutrophils, monocyte/ macrophages, and
are not significantly affected by alkylating agents or RTX (Hoyer endothelial cells are key cellular players. ANCAs, complement, and
et al. 2004). soluble mediators such as BLyS (B-lymphocyte stimulator), TNF-α,
IL-6 is not only important for the adaptive cellular and innate and IL-17 are only a few of the humoral pathogenic factors (Jennette
immunity, but also participates in the regulation of the B-cell/ plas- et al. 2006; Holden et al. 2011; Krumbholz et al. 2005; Ludviksson
mocyte compartment (Shapiro-Shelef et al. 2005; Eto et al. 2011; et al. 1998; Nassonov et al. 1997; Noronha et al. 1993; Falk et al.
Jego et al. 2001; Cassese et al. 2003). Serum IL-6 levels are increased 1990; Stone et al. 2001; Little et al. 2006; Nogueira et al. 2010).
in antibody-driven conditions such as neuromyelitis optica and In the last decade, animal models of AAV have been generated
systemic lupus erythematosus (SLE), and, of note, in patients with and, although limitations exist when trying to extrapolate experi-
HCV-related mixed cryoglobulinaemia (Antonelli et  al. 2009). mental findings to human disease (i.e. animal AAV phenotype is
Initially identified as B-cell stimulatory factor 2 (BSF-2), IL-6 often less severe), several lessons can be derived.
is known to promote late B-cell differentiation (Muraguchi et al.
1988). Upon binding to its receptor, IL-6 signals through STAT-3 ANCA and neutrophil-oriented therapies
(signal transducer and activator of transcription-3) to stimulate the Cumulative evidence suggests that tissue injury in AAV is secondary
synthesis of BLIMP-1 (B-lymphocyte-induced maturation protein to ANCA-mediated neutrophil activation in the microcirculation
1), an essential transcriptional regulator required for plasma-cell (Van Timmeren et al. 2009). Neonatal pulmonary-renal syndrome
function. Furthermore, once homed in the bone marrow, plasmo- has been reported in association with transplacental passage of
cytes induce stromal cells to produce IL-6, which in turn acts as a anti-MPO antibodies (Bansal and Tobin 2004). In patients with
paracrine survival factor for long-lived plasma cells (Minges Wols ANCA-associated glomerulonephritis, the number of activated
et al. 2002). intraglomerular neutrophils correlates with the severity of renal
In humans, Roll et al. reported that the frequency of peripheral injury. In vitro, ANCAs induce cytokine-primed neutrophils to not
pre- and postswitch memory B cells was significantly decreased only degranulate and produce oxygen radicals, but also adhere to
after treatment with TCZ in a group of patients with RA (Roll et al. and damage bystander endothelial cells (Savage et al. 2002; Little
2011). In individuals with active SLE, increased numbers of circu- et al. 2005). In vivo, passive transfer of anti-MPO antibodies trig-
lating plasmablasts (Odendahl et al. 2000; Illei et al. 2010; Jacobi gers pauci-immune crescentic glomerulonephritis in immunocom-
et al. 2010) declined after anti-IL-6R therapy (Illei et al. 2010). In petent and lymphocyte immunodeficient mice (Rag2−/−). Of note,
mixed cryoglobulinaemia, however, the experience is only limited kidney insult in anti-MPO transfer models is completely prevented
CHAPTER 42 experimental therapies for vasculitis 603

by neutrophil depletion (Xiao et al. 2005) or by the artificial elimi- Small inhibitory molecules
nation of MPO from neutrophils when chimeric mice containing Janus kinase (JAK) inhibitors have been approved for the treatment
MPO−/− haematopoietic cells are engineered. Finally, anti-PR3 of rheumatoid arthritis and compounds that modulate other intra-
antibodies have also been found to be pathogenic in humanized cellular signalling pathways are in preclinical and clinical develop-
mice (NOD-scid-IL2Rγ−/−) (Little et al. 2012). ment for different autoimmune disorders
ANCA neutrophil activation is a Fc gamma receptor (Zerbini and Lomonte 2012; Bonilla-Hernan et al. 2011).
(FcγR)-dependent process (Colman et  al. 2007). Pretreating No human data currently exist on the use of small molecules
anti-MPO IgG with endoglycosidase S or genetic ablation of FcγR for patients with AAV. However, pharmacological inhibition of
ameliorates disease in experimental conditions (van Timmeren PI3Kγ (AS605250) and p38 MAPK (AR-447) has shown to abro-
et  al. 2010; Nolan et  al. 2008). Downstream to FcγR, intracel- gate ANCA-induced neutrophil oxygen burst and degranulation
lular signal transduction pathways involve different proteins in vitro, and reduce the glomerular influx of leucocytes (Schreiber
including diacylglycerol kinase (DAGK) (Williams et  al. 2007), et al. 2010) and crescent formation in vivo (van der Veen et al.
phosphoinositol-3-kinase (PI3K) (Kettritz et al. 2002), the tyrosine 2011). Further exploration of these and other targets (i.e. Syk,
kinase Syk (Hewins et al. 2004), and p38 mitogen-activated pro- ERK1/2, etc.) may lead to the discovery of new therapeutic agents
tein kinase (P38MAPK) (Polzer et al. 2008). Once stimulated by for AAV.
ANCA, neutrophils become a source of proinflammatory cytokines
(e.g. IL-6 and IL-23) (Hoshino et al. 2008), which in turn regulate Complement inhibition
upstream lymphocyte differentiation and therefore integrate the Because subjects with GPA and MPA are ‘normocomplementae-
innate and adaptive immune systems (Cua et al. 2010). In paral- mic’ and end-organ damage is characterized by the absence (or
lel, ANCA-activated neutrophils release neutrophil extracellular minimal presence) of complement deposition, the participation
traps (NETs) that contain chromatin, PR3, MPO, and the protein of the complement cascade in the pathogenesis of AAV was not
LL37. NETs fuel the immune system with autoantigens, and pro- suspected until recently. In mice, complement depletion prevents
vide TLR9-dependent interferogenic stimulation (i.e. MPO-DNA kidney damage induced by anti-MPO challenge (Xiao et al. 2007;
complexes) to B cells and plasmacytoid dendritic cells (pDC) Schreiber et al. 2009). Furthermore, knock out phenotypes for fac-
(Kessenbrock et al. 2009). tor B, factor C5, and C5a receptor (C5aR) are protective, and phar-
Based on the ANCA-neutrophil interactions, therapeutic strat- macological inhibition of C5 cleavage (Huugen et al. 2007) or C5aR
egies for AAV may include removal of the offending autoanti- signalling attenuates the disease (Schreiber et al. 2009).
bodies (plasma exchange), modulation of the neutrophil FcγR Human healthy donor neutrophils stimulated with MPO and
signalling pathways, suppression of NET formation, and anticy- PR3 ANCAs activate the alternative complement pathway on
tokine therapy. their surface, possibly through the release of properdin or reactive
oxygen species (ROS) (Schreiber et al. 2009; Camous et al. 2011).
Plasma exchange The C5a generated by this mechanism not only contributes to the
The pathogenic role of ANCAs in animal models has led to the amplification of the inflammatory process by targeting other cells
investigation of plasma exchange (PLEX) in human disease. (i.e. endothelium, monocytes/ macrophages), but also chemoat-
Current PLEX techniques involve blood filtration or centrifugation tracks and furthers primes other neutrophils that up-regulate
to separate the plasma from other elements and then replace the their membrane expression of MPO and PR3, therefore creating a
removed volume with a substitute, such as albumin. Besides remov- vicious cycle (Schreiber et al. 2009; Hao et al. 2012).
ing ANCAs, PLEX could theoretically exert beneficial effects by On this basis, RCTs with C5aR antagonists and the anti-C5
extracting cytokines, chemokines, and activated complement and monoclonal antibody eculizumab are currently ongoing. On the
coagulation factors. other hand, since C5aR also signals though PI3K and the MAPK
In a RCT of AAV (Szpirt et  al. 2011), 32 patients with GPA family member ERK1/2, small inhibitory molecules have the
were followed for 5  years after receiving treatment with PLEX potential to ameliorate AAV not only through FcγR function
and immunosuppression (n  =  16) or immunosuppression alone modulation, but also by decreasing the inflammatory effects of the
(n = 16). The study found that PLEX was associated with preserva- complement anaphylatoxins (see Section Experimental ANCA and
tion of the renal function (renal survival), which was most marked Neutrophil-Oriented Therapies).
at 1 month and in patients presenting with creatinine levels above
2.9 mg/dl. Nonetheless, no differences in relapse rate or mortality T-cell-directed therapies
were observed. The adaptive immune system is fundamental for the genesis and
In the MEPEX trial (Jayne et al. 2007), 137 AAV patients with perpetuation of autoimmunity, but also provides effector path-
severe renal failure (creatinine >5.7 mg/dl) received oral CS and ways of tissue damage. Changes in the number and activation sta-
CYC, and were randomized to either seven PLEX sessions or three tus of circulating T-cell populations have been described in AAV
daily infusions of 1000 mg of methylprednisolone. Although the (Abdulahad et al. 2011; Popa et al. 1999; Berden et al. 2009). A sub-
progression to end-stage renal disease (ESRD) was reduced by 24% set of effector memory T cells (TEM) with NK like features (e.g.
at 12 months in the PLEX group, there was no mortality benefit and granzyme induced cytotoxicity) is expanded in peripheral circu-
the initial advantage of PLEX for renal survival was not appreciated lation of GPA patients in remission, and decreases during peri-
on long-term follow-up (Casian and Jayne 2011). ods of activity, probably due to migration into the target organs
The ongoing PEXIVAS study, designed to enrol 500 patients (Abdulahad et al. 2006). Quantitative and qualitative defects in the
with eGFR below 50 ml/min and/or pulmonary haemorrhage, will Treg compartment, and up-regulation of the Th17 axis have been
hopefully clarify the effects of PLEX on ESRD and mortality. described in GPA and MPA (Nogueira et al. 2010; Abdulahad et al.
604 SECTION 5 vasculitic diseases and syndromes and related disorders

2006). Of note, IL-17 neutralization via genetic ablation or mono- of remission in AAV. Clear indications for RTX maintenance still
clonal antibody blockade has demonstrated therapeutic effects in remain to be determined and long-term RTX safety in patients with
animal models (Gan et al. 2010; Summers et al. 2011). AAV requires further investigation. Mechanistic studies from the
Based on these findings, cytokine immunomodulatory therapy RAVE trial suggest that reconstitution of the circulating B-cell pool
directed to suppress the Th17 response (i.e. IL-17, IL-23, IL-6, and detectable ANCA titres may define the patients with highest
IL-21) or increase the number or function of Treg cells are options likelihood of relapse. Therefore, risk stratification may be the strat-
to explore in the future. egy that will allow physicians to rationalize the use of B-cell deple-
tion therapies for maintenance of remission in AAV.
B-cell and plasmocyte-directed therapies
Long-lived plasma cell depletion
B-cell depletion (RTX for long-term maintenance therapy)
Long-lived plasmocytes may develop early on in the course of
Two randomized controlled trials have provided the most con-
autoimmunity and could contribute to the perpetuation of disease.
clusive data about the pathogenic role of B cells in AAV (Jones
Unlike rapidly dividing lymphocytes, plasma cells are resistant to
et al. 2010; Stone et al. 2010). In these studies, RTX demonstrated
alkylating agents, antimetabolites, and anti-CD20 B cell depletion
equivalent efficacy compared to CYC for remission induction and
treatments (i.e. RTX).
maintenance with a similar short-term safety profile and lower per-
Bortezomib targets plasmablasts and mature plasma cells by
centage of leucopenia. Although a single course of RTX is becom-
inhibiting the 26S proteasome and inducing cell death through the
ing standard of care for relapsed and newly diagnosed GPA/MPA
activation of the terminal unfolded protein response. In animal
patients, no recommendations are yet available concerning the
models, early bortezomib treatment reduces the titres of ANCA
need for maintenance. In RITUXVAS (Jones et al. 2010), 15% of the
and abrogates glomerulonephritis by decreasing the number of
patients in the RTX group and 10% in the CYC group had a relapse
total and MPO-specific splenic and bone marrow plasmocytes
by 12 months of follow-up. Long-term results from the RAVE trial
without affecting T or B cells. Further clarification of the role of
(Stone et  al. 2010)  showed that one course of RTX was as effec-
long-live plasma cells in AAV is required before investigating this
tive for maintenance of remission as standard therapy. At 12 and
therapeutic area in humans with GPA or MPA.
18 months, 42% and 36% of the patients in the RTX arm versus 38%
and 31% in the CYC arm remained in remission off glucocorticoids
(Specks et al. 2013). Tumour necrosis factor (TNF) α monoclonal antibodies
In a retrospective study (Rhee et al. 2010), 20 patients in com- Despite preclinical data supporting the notion that TNF-α is an
plete or partial remission continued their background immuno- important mediator in the pathogenesis of GPA (Ludviksson et al.
suppression and received RTX every 4 months. Cytotoxic therapy 1998; Nassonov et al. 1997; Noronha et al. 1993; Falk et al. 1990;
was tapered off and prednisone was discontinued in 70% and 45% Stone et al. 2001; Little et al. 2006), the only randomized trial of
of the cases, respectively, by 24 months. RTX infusions were gen- TNF-α inhibition with etanercept (p75 TNF-α fusion protein),
erally well tolerated and side-effects were infrequent. All patients demonstrated no efficacy for remission maintenance in a cohort of
achieved disease control and only 3 (15%) had a flare. Two sub- 180 patients (Wegener's Granulomatosis Etanercept Trial (WGET)
jects developed late-onset neutropenia (LON) without infectious Research Group). In contrast, etanercept may increase the risk
complication. of solid malignancy when used in combination with cyclophos-
Among 65 patients with refractory AAV despite conventional phamide (Stone et  al. 2006). Unlike etanercept, the monoclonal
therapy analysed by Jones et al. (2009), none of 15 who received anti-TNF-α antibodies infliximab and adalimumab neutralize both
pre-emptive RTX infusions relapsed, in contrast to 57% of those soluble and membrane-bound TNF-α, providing complete block-
not prescribed RTX maintenance therapy (median follow-up ade of the TNF pathway and possible therapeutic advantage.
11.5 months). Sixty-two per cent of the patients taking oral immu- In 2002, Lamprecht et  al. (2002) and Bertolucci et  al. (2002)
nosuppression (n=60) were able to withdrawn these agents within reported good preliminary results with infliximab (3–5 mg/kg) in
1 year. Side-effects included 16 serious infections and two deaths. 13 patients with AAV refractory to conventional therapy (glucocor-
Of note, two patients developed short-lived LON without clinical ticoids, cyclophosphamide, AZA, MMF, and MTX). In 2004, Booth
implications. et al. conducted an open-label study of infliximab (5 mg/kg) added
Roubaud-Baudron et  al. (2012) have published the outcomes to standard therapy in 32 subjects with relapsing/ refractory AAV.
of 28 AAV patients treated with preventive RTX infusions for a Twenty-four patients (88%) achieved disease remission, but dur-
mean period of 38 months. Subjects received a median of four RTX ing a mean follow-up period of 16.8 months, one out of every five
maintenance doses with regimens that included 375 mg biannually patients flared (on infliximab maintenance), and approximately one
(n=13), 1 g biannually (n=4), 1 g annually (n=3), and others (n=8). out of every five subjects developed a severe infection (i.e. pneumo-
Eighty per cent of the patients received concomitant glucocorti- nia, urosepsis, Nocardia endophthalmitis, etc.) (Booth et al. 2004).
coids (median dose 5 mg/day), and 50% oral immunosuppression In 2010, Laurino et al. (2010) completed an open-label study of
(i.e. AZA, MMF, LEF, MTX). At last evaluation, two patients (7%) subcutaneous adalimumab in 14 subjects with newly diagnosed
had had pulmonary relapses, 17 (57%) were in complete remission, (n  =  13) or relapsed disease. Patients were treated with a taper
nine (32%) persisted with vasculitic activity, and one had died from course of prednisone, six to ten pulses of CYC (15 mg/kg), and
H1N1 flu infection. adalimumab 40 mg every 2 weeks for 3 months. At 3–6 months,
Continuous RTX monotherapy to prevent disease exacerbation subjects continued prednisone (0.1 mg/kg daily) and started AZA
have not been evaluated in controlled settings and the heterogeneity (2 mg/kg) or MMF (0.5–1 g twice daily). Eleven subjects (78.5%)
of treatments used in the studies outlined in this section precludes met the primary outcome of remission induction by week 14, but
definite conclusions about the precise role of RTX for maintenance 36% relapsed during a mean follow-up period of 40 months. Two
CHAPTER 42 experimental therapies for vasculitis 605

patients had severe infections, one of which resulted in death, and a 2009), this approach has been sporadically used in refractory EGPA
second patient died from an unknown cause at 42 months. with mixed results (Termeer et al. 2001; Mercie et al. 2000; Lesens
Thirty-three subjects with active AAV enrolled in a prospective et al. 2002; Reissig et al. 2003; Simon et al. 2003).
study received conventional therapy (glucocorticoids and CYC) In 1998, Tatsis et  al. (1998) showed that IFN-α administered
with (n=16) or without (n=17) additional infliximab at 0, 2, 6, and by subcutaneous (s.c.) injection three times a week, induced and
10 weeks. Adjuvant tumour necrosis factor antagonism did not maintained remission in four cases of EGPA not responding to
influence remission rates, adverse events, damage index scores, conventional therapy (i.e. CS and CYC). Of note, during IFN-α
relapse rates, or levels of biomarker (Morgan et al. 2011). treatment, blood eosinophil counts decreased in a dose-dependent
These results suggest that monoclonal anti-TNF-α inhibitors manner and paralleled clinical improvement and CS tapering in all
have no role beyond traditional immunosuppression in newly diag- patients. A phase II study that included seven subjects with EGPA
nosed patients, but no firm conclusions can be formulated regard- refractory to CS and CYC or MTX, showed that IFN-α (3 million
ing cases of refractory disease. Safety concerns and the availability IU three times weekly s.c.) induced and maintain disease remis-
of more effective treatment options have decreased the enthusiasm sion for at least 6 months, allowing the taper of prednisone from a
to perform controlled studies to appropriately evaluate the efficacy mean dose of 20 mg to 7 mg daily. In two cases, residual asthmatic
of these agents in refractory AAV. manifestations persisted (Metzler et al. 2008). The same research
group subsequently evaluated the safety and efficacy of this strategy
Eosinophilic granulomatosis with polyangiitis for long-term disease control in 13 patients followed for a mean
(churg–strauss syndrome, allergic period of 64 months, and found that in nine cases, IFN-α had to
granulomatous angiitis) be discontinued due to either lack of efficacy (n = 5), side-effects
Eosinophilic granulomatosis with polyangiitis (EGPA) is a disor- (n = 2), or both (n = 2).
der characterized by asthma, blood eosinophilia, and involvement IL-5 inhibition (mepolizumab)
of various organs (i.e. respiratory tract, heart, skin, gastrointes- IL-5 is the most potent inducer of maturation and activation of
tinal system, peripheral nerves, kidneys, etc.) with necrotizing eosinophils. Mepolizumab is a fully humanized anti-IL5 monoclo-
small-vessel vasculitis, eosinophil-rich infiltrates, and eosinophilic nal (IgG1) antibody that binds with high affinity to free IL-5 and pre-
granulomatosis. ANCAs are detected in less than half of EGPA vents cytokine–receptor coupling (Smith et al. 2011). Mepolizumab
patients. Standard of care recommends CS monotherapy in the decreases the number and function of eosinophils, and has shown
absence of poor prognostic factors and the addition of cyclophos- efficacy in controlled trials of eosinophil-driven conditions such
phamide for severe cases (i.e. crescentic glomerulonephritis, coro- as Fip-1-like 1-platelet derived growth factor receptor α fusion
nary vasculitis). However, limited therapeutic options exist for an gene (FIP1L1-PDGFRA) negative HES (Rothenberg et  al. 2008),
important group of patients located between the ends of this spec- prednisone-dependent asthma with sputum eosinophilia (Nair
trum, who commonly fail trials of ‘traditional’ CS-sparing agents et al. 2009), and eosinophilic oesophagitis (Straumann et al. 2010).
such as MTX, AZA, and MMF. In a study of seven patients with CS-dependent EGPA, monthly
B-cell-depletion therapy (rituximab) infusions of mepolizumab (750 mg) allowed patients to taper pred-
Granulomatous inflammation, angiitis, pauci-immune glomerulone- nisone from a mean dose of 12.9 mg/day to 4.6 mg daily by 12
phritis, and circulating ANCA, are characteristics that group EGPA weeks, regardless of the baseline IL-5 level. During anti-IL-5 ther-
with other forms of AAV. Based on these features, B-cell depletion apy, eosinophil counts decreased and subjects remained clinically
has been tried and demonstrated encouraging preliminary results stable except for two asthma exacerbations. Upon drug discontinu-
in small uncontrolled series of patients with ‘ANCA positive’ and ation after 4 months of treatment, 18 relapses occurred, mostly in
‘ANCA-negative’ disease and various disease manifestations (Vaglio the setting of the eosinophil counts returning to pretreatment levels
et al. 2012; Cartin-Ceba et al. 2011; Pepper et al. 2008; Koukoulaki (~3.4%) (Kim et al. 2010). In a subsequent study, ten patients with
et al. 2006). Confirmatory studies are needed at this time. active EGPA despite prednisone 12.5 mg daily were treated with
monthly mepolizumab (750 mg) infusions for 9 months. Eight of
Eosinophil-targeted therapies the subjects achieved the primary outcome of remission and CS
Prominent blood and tissue eosinophilia are hallmarks of EGPA dosage of less than 7.5 mg/day at 32 weeks. Patients were able to
(Hellmich et al. 2003). IL-5 is a cardinal regulator of the eosino- decrease their prednisone intake from a median dose of 19 mg/day
phil lineage biology and selectively regulates eosinophil matura- at baseline to 4 mg/day at the end of follow-up (Moosig et al. 2011).
tion, mobilization, activation, and survival. The IL-5 pathway is After mepolizumab was discontinued, seven relapses occurred. No
over expressed in patients with EGPA (Schonermarck et al. 2000; major safety concerns arose from these pilot studies.
Tsukadaira et al. 1999), and the serum levels of this cytokine cor-
relate with disease activity (Hellmich et  al. 2005). Experimental
therapies that directly or indirectly impact the IL-5 axis have been Conclusion
tried for refractory/ relapsing patients. Recent advances in understanding the immunopathogenesis of sys-
Interferon-α temic vasculitides have created a scenario where the change from
In vitro, IFN-α decreases the production of Th2-related cytokines non-specific immunosuppression to therapies targeting specific
by CD4 cells (including IL-5), and inhibits the release of cytotoxic pathogenic pathways seems more possible. Throughout the devel-
mediators by eosinophils. (Krishnaswamy et  al. 1996; Aldebert opment of these novel approaches, initial enthusiasm is frequently
et al. 1996). Because IFN-α therapy has CS-sparing effects in cer- supplanted by more sober interpretations, and, therefore, these
tain types of hypereosinophilic syndromes (HES) (Ogbogu et al. early reports should be viewed warily. Unfortunately, our current
606 SECTION 5 vasculitic diseases and syndromes and related disorders

knowledge is far from comprehensive and not only further research Cacoub, P., Poynard, T., Ghillani, P., Charlotte, F., Olivi, M., Piette, J.C.,
will need to be done to clear uncertainties, close the gaps, and sug- and Opolon, P. (1999). Extrahepatic manifestations of chronic hepa-
gest new targets, but of greatest importance, experimental thera- titis C. MULTIVIRC Group. Multidepartment Virus C. Arthritis and
Rheumatism, 42, 2204–12.
peutic interventions will require the scrutiny of controlled trials to
Calabrese, C., Faiman, B., Martin, D., Reu, F., and Calabrese, L.H. (2011).
rigorously evaluate their efficacy and safety before becoming part of Type 1 cryoglobulinemia: response to thalidomide and lenalidomide.
the armamentum against vasculitis. Journal of Clinical Rheumatology, 17, 145–7.
Camous, L., Roumenina, L., Bigot, S., Brachemi, S., Fremeaux-Bacchi, V.,
References Lesavre, P., and Halbwachs-Mecarelli, L. (2011). Complement alterna-
Abdulahad, W.H., Lamprecht, P., and Kallenberg, C.G. (2011). T-helper cells tive pathway acts as a positive feedback amplification of neutrophil acti-
as new players in ANCA-associated vasculitides. Arthritis Research and vation. Blood, 117, 1340–9.
Therapy, 13, 236. Cartin-Ceba, R., Keogh, K.A., Specks, U., Sethi, S., and Fervenza, F.C. (2011).
Abdulahad, W.H., Stegeman, C.A., Limburg, P.C., and Kallenberg, C.G. Rituximab for the treatment of Churg-Strauss syndrome with renal
(2008). Skewed distribution of Th17 lymphocytes in patients with involvement. Nephrology Dialysis Transplantation, 26, 2865–71.
Wegener’s granulomatosis in remission. Arthritis and Rheumatism, 58, Casian, A. and Jayne, D. (2011). Plasma exchange in the treatment of
2196–205. Wegener’s granulomatosis, microscopic polyangiitis, Churg-Strauss syn-
Abdulahad, W.H., van der Geld, Y.M., Stegeman, C.A., and Kallenberg, drome and renal limited vasculitis. Current Opinion in Rheumatology,
C.G. (2006). Persistent expansion of CD4+ effector memory T cells in 23, 12–7.
Wegener’s granulomatosis. Kidney International, 70, 938–47. Cassese, G., Arce, S., Hauser, A.E., Lehnert, K., Moewes, B., Mostarac, M.,
Aldebert, D., Lamkhioued, B., Desaint, C., Gounni, A.S., Goldman, M., Muehlinghaus, G., Szyska, M., Radbruch, A., and Manz, R.A. (2003).
Capron, A., Prin, L., and Capron, M. (1996). Eosinophils express a func- Plasma cell survival is mediated by synergistic effects of cytokines and
tional receptor for interferon alpha: inhibitory role of interferon alpha adhesion-dependent signals. Journal of Immunology, 171, 1684–90.
on the release of mediators. Blood, 87, 2354–60. Cem Ar, M., Soysal, T., Hatemi, G., Salihoglu, A., Yazici, H., and Ulku, B.
Antonelli, A., Ferri, C., Ferrari, S.M., Ghiri, E., Goglia, F., Pampana, A., (2005). Successful management of cryoglobulinemia-induced leukocy-
Bruschi, F., and Fallahi, P. (2009). Serum levels of proinflammatory toclastic vasculitis with thalidomide in a patient with multiple myeloma.
cytokines interleukin-1beta, interleukin-6, and tumor necrosis fac- Annals of Hematology, 84, 609–13.
tor alpha in mixed cryoglobulinemia. Arthritis and Rheumatism, 60, Cohen, C., Mekinian, A., Saidenberg-Kermanac, H.N., Stirnemann, J.,
3841–7. Fenaux, P., Gherardi, R., and Fain, O. (2012). Efficacy of tocilizumab
Bansal, P.J. and Tobin, M.C. (2004). Neonatal microscopic polyangiitis sec- in rituximab-refractory cryoglobulinemia vasculitis. Annals of the
ondary to transfer of maternal myeloperoxidase-antineutrophil cytoplas- Rheumatic Diseases, 71, 628–9.
mic antibody resulting in neonatal pulmonary hemorrhage and renal Colman, R., Hussain, A., Goodall, M., Young, S.P., Pankhurst, T., Lu, X.,
involvement. Annals of Allergy, Asthma and Immunology, 93, 398–401. Jefferis, R., Savage, C.O., and Williams, J.M. (2007). Chimeric antibodies
Barr, T.A., Shen, P., Brown, S., Lampropoulou, V., Roch, T., Lawrie, S., Fan, to proteinase 3 of IgG1 and IgG3 subclasses induce different magnitudes
B., O’Connor, R.A., Anderton, S.M., Bar-Or, A., et  al. (2012). B cell of functional responses in neutrophils. Annals of the Rheumatic Diseases,
depletion therapy ameliorates autoimmune disease through ablation of 66, 676–82.
IL-6-producing B cells. Journal of Experimental Medicine, 209, 1001–10. Comarmond, C., Plaisier, E., Dahan, K., Mirault, T., Emmerich, J., Amoura,
Bartolucci, P., Ramanoelina, J., Cohen, P., Mahr, A., Godmer, P., Le Hello, C., Z., Cacoub, P., and Saadoun, D. (2012). Anti TNF-alpha in refrac-
and Guillevin, L. (2002). Efficacy of the anti-TNF-alpha antibody inflixi- tory Takayasu’s arteritis:  Cases series and review of the literature.
mab against refractory systemic vasculitides: an open pilot study on 10 Autoimmunity Reviews, 11, 678–84.
patients. Rheumatology (Oxford), 41, 1126–32. Cua, D.J. and Tato, C.M. (2010). Innate IL-17-producing cells: the sentinels of
Berden, A.E., Kallenberg, C.G., Savage, C.O., Yard, B.A., Abdulahad, W.H., the immune system. Nature Reviews Immunology, 10, 479–89.
de Heer, E., Bruijn, J.A., and Bajema, I.M. (2009). Cellular immunity in Dasgupta, B. and Panayi, G.S. (1990). Interleukin-6 in serum of patients
Wegener’s granulomatosis: characterizing T lymphocytes. Arthritis and with polymyalgia rheumatica and giant cell arteritis. British Journal of
Rheumatism, 60, 1578–87. Rheumatology, 29, 456–8.
Beyer, C., Axmann, R., Sahinbegovic, E., Distler, J.H., Manger, B., Schett, G., Deng, J., Younge, B.R., Olshen, R.A., Goronzy, J.J., and Weyand, C.M. (2010).
and Zwerina, J. (2011). Anti-interleukin 6 receptor therapy as rescue treat- Th17 and Th1 T-cell responses in giant cell arteritis. Circulation, 121,
ment for giant cell arteritis. Annals of the Rheumatic Diseases, 70, 1874–5. 906–15.
Bonilla-Hernan, M.G., Miranda-Carus, M.E., and Martin-Mola, E. (2011). de Souza, A.W., da Silva, M.D., Machado, L.S., Oliveira, A.C., Pinheiro,
New drugs beyond biologics in rheumatoid arthritis: the kinase inhibi- F.A., and Sato, E.I. (2012). Short-term effect of leflunomide in patients
tors. Rheumatology (Oxford), 50, 1542–50. with Takayasu arteritis: an observational study. Scandinavian Journal of
Booth, A., Harper, L., Hammad, T., Bacon, P., Griffith, M., Levy, J., Savage, C., Rheumatology, 41, 227–30.
Pusey, C., and Jayne, D. (2004). Prospective study of TNFalpha blockade Emilie, D., Liozon, E., Crevon, M.C., Lavignac, C., Portier, A., Liozon, F., and
with infliximab in anti-neutrophil cytoplasmic antibody-associated sys- Galanaud, P. (1994). Production of interleukin 6 by granulomas of giant
temic vasculitis. Journal of the American Society of Nephrology, 15, 717–21. cell arteritis. Human Immunology, 39, 17–24.
Boyer, O., Saadoun, D., Abriol, J., Dodille, M., Piette, J.C., Cacoub, P., and Eto, D., Lao, C., DiToro, D., Barnett, B., Escobar, T.C., Kageyama, R., Yusuf, I.,
Klatzmann, D. (2004). CD4+CD25+ regulatory T-cell deficiency in and Crotty, S. (2011). IL-21 and IL-6 are critical for different aspects of B
patients with hepatitis C-mixed cryoglobulinemia vasculitis. Blood, 103, cell immunity and redundantly induce optimal follicular helper CD4 T
3428–30. cell (Tfh) differentiation. PLoS One, 6, e17739.
Bredemeier, M.R.C., Barbosa, M.V., and Pitrez, E.H. (2012). One-year clini- Falk, R.J., Terrell, R.S., Charles, L.A., and Jennette, J.C. (1990). Anti-neutrophil
cal and radiological evolution of a patient with refractory Takayasu’s cytoplasmic autoantibodies induce neutrophils to degranulate and pro-
arteritis under treatment with tocilizumab. Clinical and Experimental duce oxygen radicals in vitro. Proceedings of the National Academy of
Rheumatology, 30, S98–S100. Sciences of the U S A, 87, 4115–9.
Brouet, J.C., Clauvel, J.P., Danon, F., Klein, M., and Seligmann, M. (1974). Foessel, L., Besancenot, J.F., Blaison, G., Magy-Bertrand, N., Jaussaud,
Biologic and clinical significance of cryoglobulins. A report of 86 cases. R., Etienne, Y., Maurier, F., Audia, S., and Martin, T. (2011). Clinical
American Journal of Medicine, 57, 775–88. spectrum, treatment, and outcome of patients with type II mixed
CHAPTER 42 experimental therapies for vasculitis 607

cryoglobulinemia without evidence of hepatitis C infection. Journal of Holden, N.J., Williams, J.M., Morgan, M.D., Challa, A., Gordon, J., Pepper,
Rheumatology, 38, 716–22. R.J., Salama, A.D., Harper, L., and Savage, C.O. (2011). ANCA-stimulated
Fujimoto, M., Serada, S., Mihara, M., Uchiyama, Y., Yoshida, H., Koike, N., neutrophils release BLyS and promote B cell survival: a clinically rel-
Ohsugi, Y., Nishikawa, T., Ripley, B., Kimura, A., et al. (2008). Interleukin-6 evant cellular process. Annals of the Rheumatic Diseases, 70, 2229–33.
blockade suppresses autoimmune arthritis in mice by the inhibition of Hoshino, A., Nagao, T., Nagi-Miura, N., Ohno, N., Yasuhara, M., Yamamoto,
inflammatory Th17 responses. Arthritis and Rheumatism, 58, 3710–9. K., Nakayama, T., and Suzuki, K. (2008). MPO-ANCA induces IL-17
Galarza, C., Valencia, D., Tobon, G.J., Zurita, L., Mantilla, R.D., production by activated neutrophils in vitro via classical complement
Pineda-Tamayo, R., Rojas-Villarraga, A., Rueda, J.C., and Anaya, J.M. pathway-dependent manner. Journal of Autoimmunity, 31, 79–89.
(2008). Should rituximab be considered as the first-choice treatment for Hoyer, B.F., Moser, K., Hauser, A.E., Peddinghaus, A., Voigt, C., Eilat, D.,
severe autoimmune rheumatic diseases? Clinical Reviews in Allergy and Radbruch, A., Hiepe, F., and Manz, R.A. (2004). Short-lived plasmablasts
Immunology, 34, 124–8. and long-lived plasma cells contribute to chronic humoral autoimmun-
Gan, P.Y., Steinmetz, O.M., Tan, D.S., O’Sullivan, K.M., Ooi, J.D., Iwakura, ity in NZB/W mice. Journal of Experimental Medicine, 199, 1577–84.
Y., Kitching, A.R., and Holdsworth, S.R. (2010). Th17 cells promote Hoyer, B.F., Mumtaz, I.M., Loddenkemper, K., Bruns, A., Sengler, C.,
autoimmune anti-myeloperoxidase glomerulonephritis. Journal of the Hermann, K.G., Maza, S., Keitzer, R., Burmester, G.R., Buttgereit, F.,
American Society of Nephrology, 21, 925–31. et al. (2012). Takayasu arteritis is characterised by disturbances of B cell
Garcia-Martinez, A., Hernandez-Rodriguez, J., Espigol-Frigole, G., homeostasis and responds to B cell depletion therapy with rituximab.
Prieto-Gonzalez, S., Butjosa, M., Segarra, M., Lozano, E., and Cid, M.C. Annals of the Rheumatic Diseases, 71, 75–9.
(2010). Clinical relevance of persistently elevated circulating cytokines Huugen, D., van Esch, A., Xiao, H., Peutz-Kootstra, C.J., Buurman, W.A.,
(tumor necrosis factor alpha and interleukin-6) in the long-term fol- Tervaert, J.W., Jennette, J.C., and Heeringa, P. (2007). Inhibition
lowup of patients with giant cell arteritis. Arthritis Care and Research of complement factor C5 protects against anti-myeloperoxidase
(Hoboken), 62, 835–41. antibody-mediated glomerulonephritis in mice. Kidney International,
Goel, R., Danda, D., Mathew, J., and Edwin, N. (2010). Mycophenolate 71, 646–54.
mofetil in Takayasu’s arteritis. Clinical Rheumatology, 29, 329–32. Illei, G.G., Shirota, Y., Yarboro, C.H., Daruwalla, J., Tackey, E., Takada, K.,
Griffin, D.O. and Rothstein, T.L. (2011). A small CD11b(+) human B1 cell Fleisher, T., Balow, J.E., and Lipsky, P.E. (2010). Tocilizumab in systemic
subpopulation stimulates T cells and is expanded in lupus. Journal of lupus erythematosus: data on safety, preliminary efficacy, and impact on
Experimental Medicine, 208, 2591–8. circulating plasma cells from an open-label phase I dosage-escalation
Han, J.W., Shimada, K., Ma-Krupa, W., Johnson, T.L., Nerem, R.M., Goronzy, study. Arthritis and Rheumatism, 62, 542–52.
J.J., and Weyand, C.M. (2008). Vessel wall-embedded dendritic cells Inder, S.J., Bobryshev, Y.V., Cherian, S.M., Wang, A.Y., Lord, R.S., Masuda,
induce T-cell autoreactivity and initiate vascular inflammation. K., and Yutani, C. (2000). Immunophenotypic analysis of the aortic
Circulation Research, 102, 546–53. wall in Takayasu’s arteritis: involvement of lymphocytes, dendritic cells
Hao, J., Meng, L.Q., Xu, P.C., Chen, M., and Zhao, M.H. (2012). p38MAPK, and granulocytes in immuno-inflammatory reactions. Cardiovascular
ERK and PI3K Signaling Pathways Are Involved in C5a-Primed Surgery, 8, 141–8.
Neutrophils for ANCA-Mediated Activation. PLoS One, 7, e38317. Jacobi, A.M., Mei, H., Hoyer, B.F., Mumtaz, I.M., Thiele, K., Radbruch,
Haruta, H., Ohguro, N., Fujimoto, M., Hohki, S., Terabe, F., Serada, S., A., Burmester, G.R., Hiepe, F., and Dorner, T. (2010). HLA-DRhigh/
Nomura, S., Nishida, K., Kishimoto, T., and Naka, T. (2011). Blockade CD27high plasmablasts indicate active disease in patients with
of interleukin-6 signaling suppresses not only th17 but also interpho- systemic lupus erythematosus. Annals of the Rheumatic Diseases,
toreceptor retinoid binding protein-specific Th1 by promoting regula- 69, 305–8.
tory T cells in experimental autoimmune uveoretinitis. Investigative Jayne, D.R., Gaskin, G., Rasmussen, N., Abramowicz, D., Ferrario, F.,
Ophthalmology and Visual Science, 52, 3264–71. Guillevin, L., Mirapeix, E., Savage, C.O., Sinico, R.A., Stegeman, C.A.,
Hellmich, B., Csernok, E., and Gross, W.L. (2005). Proinflammatory et  al. (2007). Randomized trial of plasma exchange or high-dosage
cytokines and autoimmunity in Churg-Strauss syndrome. Annals of the methylprednisolone as adjunctive therapy for severe renal vasculitis.
New York Academy of Sciences, 1051, 121–31. Journal of the American Society of Nephrology, 18, 2180–8.
Hellmich, B., Ehlers, S., Csernok, E., and Gross, W.L. (2003). Update on the Jayne, D., Rasmussen, N., Andrassy, K., et al. (2003). A randomized trial of
pathogenesis of Churg-Strauss syndrome. Clinical and Experimental maintenance therapy for vasculitis associated with antineutrophil cyto-
Rheumatology, 21 (6 Suppl. 32), S69–77. plasmic autoantibodies. New England Journal of Medicine, 349, 36–44.
Hewins, P., Williams, J.M., Wakelam, M.J., and Savage, C.O. (2004). Jego, G., Bataille, R., and Pellat-Deceunynck, C. (2001). Interleukin-6 is
Activation of Syk in neutrophils by antineutrophil cytoplasm antibod- a growth factor for nonmalignant human plasmablasts. Blood, 97,
ies occurs via Fcgamma receptors and CD18. Journal of the American 1817–22.
Society of Nephrology, 15, 796–808. Jennette, J.C., Xiao, H., and Falk, R.J. (2006). Pathogenesis of vascular
Hoffman, G.S., Cid, M.C., Hellmann, D.B., Guillevin, L., Stone, J.H., inflammation by anti-neutrophil cytoplasmic antibodies. Journal of the
Schousboe, J., Cohen, P., Calabrese, L.H., Dickler, H., Merkel, P.A., et al. American Society of Nephrology, 17, 1235–42.
(2002). A multicenter, randomized, double-blind, placebo-controlled Jones, R.B., Ferraro, A.J., Chaudhry, A.N., Brogan, P., Salama, A.D.,
trial of adjuvant methotrexate treatment for giant cell arteritis. Arthritis Smith, K.G., Savage, C.O., and Jayne, D.R. (2009). A multicenter sur-
and Rheumatism, 46, 1309–18. vey of rituximab therapy for refractory antineutrophil cytoplasmic
Hoffman, G.S., Cid, M.C., Rendt-Zagar, K.E., Merkel, P.A., Weyand, C.M., antibody-associated vasculitis. Arthritis and Rheumatism, 60, 2156–68.
Stone, J.H., Salvarani, C., Xu, W., Visvanathan, S., and Rahman, M.U. Jones, R.B., Tervaert, J.W., Hauser, T., Luqmani, R., Morgan, M.D., Peh, C.A.,
(2007). Infliximab for maintenance of glucocorticosteroid-induced Savage, C.O., Segelmark, M., Tesar, V., van Paassen, P., et  al. (2010).
remission of giant cell arteritis: a randomized trial. Annals of Internal Rituximab versus cyclophosphamide in ANCA-associated renal vasculi-
Medicine, 146, 621–30. tis. New England Journal of Medicine, 363, 211–20.
Hoffman, G.S., Leavitt, R.Y., Kerr, G.S., Rottem, M., Sneller, M.C., and Fauci, Jones, S.A. (2005). Directing transition from innate to acquired immu-
A.S. (1994). Treatment of glucocorticoid-resistant or relapsing Takayasu nity: defining a role for IL-6. Journal of Immunology, 175, 3463–8.
arteritis with methotrexate. Arthritis and Rheumatism, 37, 578–82. Jover, J.A., Hernandez-Garcia, C., Morado, I.C., Vargas, E., Banares, A., and
Hoffman, G.S., Merkel, P.A., Brasington, R.D., Lenschow, D.J., and Liang, P. Fernandez-Gutierrez, B. (2001). Combined treatment of giant-cell arte-
(2004). Anti-tumor necrosis factor therapy in patients with difficult to ritis with methotrexate and prednisone. a randomized, double-blind,
treat Takayasu arteritis. Arthritis and Rheumatism, 50, 2296–304. placebo-controlled trial. Annals of Internal Medicine, 134, 106–14.
608 SECTION 5 vasculitic diseases and syndromes and related disorders

Kerr, G.S., Hallahan, C.W., Giordano, J., Leavitt, R.Y., Fauci, A.S., Rottem, Ludviksson, B.R., Sneller, M.C., Chua, K.S., Talar-Williams, C., Langford,
M., and Hoffman, G.S. (1994). Takayasu arteritis. Annals of Internal C.A., Ehrhardt, R.O., Fauci, A.S., and Strober, W. (1998). Active
Medicine, 120, 919–29. Wegener’s granulomatosis is associated with HLA-DR+ CD4+ T cells
Kessenbrock, K., Krumbholz, M., Schonermarck, U., Back, W., Gross, W.L., exhibiting an unbalanced Th1-type T cell cytokine pattern: reversal with
Werb, Z., Grone, H.J., Brinkmann, V., and Jenne, D.E. (2009). Netting neu- IL-10. Journal of Immunology, 160, 3602–9.
trophils in autoimmune small-vessel vasculitis. Nature Medicine, 15, 623–5. Lund, F.E. and Randall, T.D. (2010). Effector and regulatory B cells: modula-
Kettritz, R., Choi, M., Butt, W., Rane, M., Rolle, S., Luft, F.C., and Klein, J.B. tors of CD4(+) T cell immunity. Nature Review Immunology, 10, 236–47.
(2002). Phosphatidylinositol 3-kinase controls antineutrophil cytoplas- Ma-Krupa, W., Jeon, M.S., Spoerl, S., Tedder, T.F., Goronzy, J.J., and Weyand,
mic antibodies-induced respiratory burst in human neutrophils. Journal C.M. (2004). Activation of arterial wall dendritic cells and breakdown
of the American Society of Nephrology, 13, 1740–9. of self-tolerance in giant cell arteritis. Journal of Experimental Medicine,
Kim, S., Marigowda, G., Oren, E., Israel, E., and Wechsler, M.E. (2010). 199, 173–83.
Mepolizumab as a steroid-sparing treatment option in patients with Maksimowicz-McKinnon, K., Clark, T.M., and Hoffman, G.S. (2007).
Churg-Strauss syndrome. Journal of Allergy and Clinical Immunology, Limitations of therapy and a guarded prognosis in an American cohort
125, 1336–43. of Takayasu arteritis patients. Arthritis and Rheumatism, 56, 1000–9.
Korn, T., Mitsdoerffer, M., Croxford, A.L., Awasthi, A., Dardalhon, V.A., Martinez-Taboada, V.M., Rodriguez-Valverde, V., Carreno, L., Lopez-Longo,
Galileos, G., Vollmar, P., Stritesky, G.L., Kaplan, M.H., Waisman, A., et al. J., Figueroa, M., Belzunegui, J., Mola, E.M., and Bonilla, G. (2008). A
(2008). IL-6 controls Th17 immunity in vivo by inhibiting the conver- double-blind placebo controlled trial of etanercept in patients with giant
sion of conventional T cells into Foxp3+ regulatory T cells. Proceedings cell arteritis and corticosteroid side effects. Annals of the Rheumatic
of the National Academy of Sciences of the U S A, 105, 18460–5. Diseases, 67, 625–30.
Koukoulaki, M., Smith, K.G., and Jayne, D.R. (2006). Rituximab in Matteson EL. (1999). A history of early investigation in polyarteritis nodosa.
Churg-Strauss syndrome. Annals of the Rheumatic Diseases, 65, 557–9. Arthritis Care and Research, 12, 294-302.
Krishnaswamy, G., Smith, J.K., Srikanth, S., Chi, D.S., Kalbfleisch, J.H., and Mauri, C. and Bosma, A. (2012). Immune regulatory function of B cells.
Huang, S.K. (1996). Lymphoblastoid interferon-alpha inhibits T cell Annual Review of Immunology, 30, 221–41.
proliferation and expression of eosinophil-activating cytokines. Journal Mazlumzadeh, M., Hunder, G.G., Easley, K.A., Calamia, K.T., Matteson, E.L.,
of Interferon and Cytokine Research, 16, 819–27. Griffing, W.L., Younge, B.R, Weyand, C.M., and Goronzy, J.J. (2006).
Krumbholz, M., Specks, U., Wick, M., Kalled, S.L., Jenne, D., and Meinl, E. Treatment of giant cell arteritis using induction therapy with high-dose
(2005). BAFF is elevated in serum of patients with Wegener’s granulo- glucocorticoids: a double-blind, placebo-controlled, randomized pro-
matosis. Journal of Autoimmunity, 25, 298–302. spective clinical trial. Arthritis and Rheumatism, 54, 3310–8.
Krupa, W.M., Dewan, M., Jeon, M.S., Kurtin, P.J., Younge, B.R., Goronzy, Mekinian, A., Neel, A., Sibilia, J., Cohen, P., Connault, J., Lambert, M.,
J.J., and Weyand, C.M. (2002). Trapping of misdirected dendritic cells Federici, L., Berthier, S., Fiessinger, J.N., Godeau, B., et al. (2012). Efficacy
in the granulomatous lesions of giant cell arteritis. American Journal of and tolerance of infliximab in refractory Takayasu arteritis: French mul-
Pathology, 161, 1815–23. ticentre study. Rheumatology (Oxford), 51, 882–6.
Kussmaul, A. and Maier, R. (1866). Ueber eine bisher nicht beschriebene Mercie, P., Viallard, J.F., Faure, I., Trimoulet, P., Vital, A., Lifermann, F., Leng,
eigenthümliche Arterienerkrankung (Periarteritis nodosa), die mit B., and Pellegrin, J.L. (2000). Hepatitis C virus infection with and with-
Morbus Brightii und rapid fortschreitender allgemeiner Muskellähmung out cryoglobulinemia as a case of Churg-Strauss syndrome. Journal of
einhergeht. Deutsches Archiv für Klinische Medizin, 1, 484–518. Rheumatology, 27, 814–7.
Lamprecht, P., Voswinkel, J., Lilienthal, T., Nolle, B., Heller, M., Gross, W.L., Metzler, C., Schnabel, A., Gross, W.L., and Hellmich, B. (2008). A phase II
and Gause, A. (2002). Effectiveness of TNF-alpha blockade with inflixi- study of interferon-alpha for the treatment of refractory Churg-Strauss
mab in refractory Wegener’s granulomatosis. Rheumatology (Oxford), syndrome. Clinical and Experimental Rheumatology, 26 (3 Suppl. 49),
41, 1303–7. S35–40.
Laurino, S., Chaudhry, A., Booth, A., Conte, G., and Jayne, D. (2010). Minges Wols, H.A., Underhill, G.H., Kansas, G.S., and Witte, P.L. (2002). The
Prospective study of TNFalpha blockade with adalimumab in role of bone marrow-derived stromal cells in the maintenance of plasma
ANCA-associated systemic vasculitis with renal involvement. cell longevity. Journal of Immunology, 169, 4213–21.
Nephrology Dialysis Transplantation, 25, 3307–14. Miossec, P., Korn, T., and Kuchroo, V.K. (2009). Interleukin-17 and type 17
Lesens, O., Hansmann, Y., Nerson, J., Pasquali, J., Gasser, B., Wihlm, J., and helper T cells. New England Journal of Medicine, 361, 888–98.
Christmann, D. (2002). Severe Churg-Strauss syndrome with mediasti- Molloy, E.S., Langford, C.A., Clark, T.M., Gota, C.E., and Hoffman, G.S.
nal lymphadenopathy treated with interferon therapy. European Journal (2008). Anti-tumour necrosis factor therapy in patients with refrac-
of Internal Medicine, 13, 458. tory Takayasu arteritis: long-term follow-up. Annals of the Rheumatic
Lin, R.J., Curran, J.J., Zimmerman, T.M., Song, J., Niewold, T.B., and Sweiss, Diseases, 67, 1567–9.
N.J. (2010). Lenalidomide for the treatment of cryoglobulinemia and Moosig, F., Gross, W.L., Herrmann, K., Bremer, J.P., and Hellmich, B. (2011).
undifferentiated spondyloarthropathy in a patient with multiple mye- Targeting interleukin-5 in refractory and relapsing Churg-Strauss syn-
loma. Journal of Clinical Rheumatology, 16, 90–1. drome. Annals of Internal Medicine, 155, 341–43.
Little, M.A., Al-Ani, B., Ren, S., Al-Nuaimi, H., Leite, M. Jr, Alpers, C.E., Morgan, M.D., Drayson, M.T., Savage, C.O., and Harper, L. (2011). Addition
Savage, C.O., and Duffield, J.S. (2012). Anti-proteinase 3 anti-neutrophil of infliximab to standard therapy for ANCA-associated vasculitis.
cytoplasm autoantibodies recapitulate systemic vasculitis in mice with a Nephron Clinical Practice, 117, c89–97.
humanized immune system. PLoS One, 7, e28626. Mukhtyar, C., Guillevin, L., Cid, M.C., Dasgupta, B., de Groot, K., Gross,
Little, M.A., Bhangal, G., Smyth, C.L., Nakada, M.T., Cook, H.T., Nourshargh, W., Hauser, T., Hellmich, B., Jayne, D., Kallenberg, C.G., et al. (2009).
S., and Pusey, C.D. (2006). Therapeutic effect of anti-TNF-alpha EULAR recommendations for the management of large vessel vasculitis.
antibodies in an experimental model of anti-neutrophil cytoplasm Annals of the Rheumatic Diseases, 68, 318–23.
antibody-associated systemic vasculitis. Journal of the American Society Muraguchi, A., Hirano, T., Tang, B., Matsuda, T., Horii, Y., Nakajima, K.,
of Nephrology, 17, 160–9. and Kishimoto, T. (1988). The essential role of B cell stimulatory fac-
Little, M.A., Smyth, C.L., Yadav, R., Ambrose, L., Cook, H.T., Nourshargh, S., tor 2 (BSF-2/IL-6) for the terminal differentiation of B cells. Journal of
and Pusey, C.D. (2005). Antineutrophil cytoplasm antibodies directed Experimental Medicine, 167, 332–44.
against myeloperoxidase augment leukocyte-microvascular interactions Nair, P., Pizzichini, M.M., Kjarsgaard, M., Inman, M.D., Efthimiadis, A.,
in vivo. Blood, 106, 2050–8. Pizzichini, E., Hargreave, F.E., and O’Byrne, P.M. (2009). Mepolizumab
CHAPTER 42 experimental therapies for vasculitis 609

for prednisone-dependent asthma with sputum eosinophilia. New Roche, N.E., Fulbright, J.W., Wagner, A.D., Hunder, G.G., Goronzy, J.J., and
England Journal of Medicine, 360, 985–93. Weyand, C.M. (1993). Correlation of interleukin-6 production and dis-
Naka, T., Nishimoto, N., and Kishimoto, T. (2002). The paradigm of IL-6: from ease activity in polymyalgia rheumatica and giant cell arteritis. Arthritis
basic science to medicine. Arthritis Research, 4 (Suppl. 3), S233–242. and Rheumatism, 36, 1286–94.
Nassonov, E.L., Samsonov, M.Y., Tilz, G.P., Beketova, T.V., Semenkova, E.N., Rodriguez-Paez, A.C., Seetharaman, M., and Brent, L.H. (2009).
Baranov, A., Wachter, H., and Fuchs, D. (1997). Serum concentrations Cryoglobulin crystal arthropathy in a patient with multiple myeloma.
of neopterin, soluble interleukin 2 receptor, and soluble tumor necrosis Journal of Clinical Rheumatology, 15, 238–40.
factor receptor in Wegener’s granulomatosis. Journal of Rheumatology, Roll, P., Muhammad, K., Schumann, M., Kleinert, S., Einsele, H., Dorner, T.,
24, 666–70. and Tony, H.P. (2011). In vivo effects of the anti-interleukin-6 recep-
Nishimoto, N., Nakahara, H., Yoshio-Hoshino, N., and Mima, T. (2008). tor inhibitor tocilizumab on the B cell compartment. Arthritis and
Successful treatment of a patient with Takayasu arteritis using a human- Rheumatism, 63, 1255–64.
ized anti-interleukin-6 receptor antibody. Arthritis and Rheumatism, 58, Rothenberg, M.E., Klion, A.D., Roufosse, F.E., Kahn, J.E., Weller, P.F., Simon,
1197–200. H.U., Schwartz, L.B., Rosenwasser, L.J., Ring, J., Griffin, E.F., et  al.
Nogueira, E., Hamour, S., Sawant, D., Henderson, S., Mansfield, N., Chavele, (2008). Treatment of patients with the hypereosinophilic syndrome with
K.M., Pusey, C.D., and Salama, A.D. (2010). Serum IL-17 and IL-23 mepolizumab. New England Journal of Medicine, 358, 1215–28.
levels and autoantigen-specific Th17 cells are elevated in patients with Roubaud-Baudron, C., Pagnoux, C., Meaux-Ruault, N., Grasland, A., Zoulim,
ANCA-associated vasculitis. Nephrology Dialysis Transplantation, 25, A., Leg, J., Prud’homme, A., Bienvenu, B., de Menthon, M., Camps, S.,
2209–17. et al. (2012). Rituximab maintenance therapy for granulomatosis with
Nolan, S.L., Kalia, N., Nash, G.B., Kamel, D., Heeringa, P., and Savage, C.O. polyangiitis and microscopic polyangiitis. Journal of Rheumatology, 39,
(2008). Mechanisms of ANCA-mediated leukocyte-endothelial cell 125–30.
interactions in vivo. Journal of the American Society of Nephrology, 19, Saadoun, D., Rosenzwajg, M., Joly, F., Six, A., Carrat, F., Thibault, V., Sene,
973–84. D., Cacoub, P., and Klatzmann, D. (2011). Regulatory T-cell responses to
Noronha, I.L., Kruger, C., Andrassy, K., Ritz, E., and Waldherr, R. (1993). In low-dose interleukin-2 in HCV-induced vasculitis. New England Journal
situ production of TNF-alpha, IL-1 beta and IL-2R in ANCA-positive of Medicine, 365, 2067–77.
glomerulonephritis. Kidney International, 43, 682–92. Sakaguchi, S., Miyara, M., Costantino, C.M., and Hafler, D.A. (2010).
Odendahl, M., Jacobi, A., Hansen, A., Feist, E., Hiepe, F., Burmester, G.R., FOXP3+ regulatory T cells in the human immune system. Nature
Lipsky, P.E., Radbruch, A., and Dorner, T. (2000). Disturbed peripheral Review Immunology, 10, 490–500.
B lymphocyte homeostasis in systemic lupus erythematosus. Journal of Salvarani, C., Cantini, F., Boiardi, L., and Hunder, G.G. (2002). Polymyalgia
Immunology, 165, 5970–9. rheumatica and giant-cell arteritis. New England Journal of Medicine,
Ogbogu, P.U., Bochner, B.S., Butterfield, J.H., Gleich, G.J., Huss-Marp, J., 347, 261–71.
Kahn, J.E., Leiferman, K.M., Nutman, T.B., Pfab, F., Ring, J., et al. (2009). Salvarani, C., Magnani, L., Catanoso, M., et al. (2012a). Rescue treatment
Hypereosinophilic syndrome:  a multicenter, retrospective analysis of with tocilizumab for Takayasu arteritis resistant to TNF-alpha blockers.
clinical characteristics and response to therapy. Journal of Allergy and Clinical and Experimental Rheumatology, 30 (1 Suppl. 70), S90–3.
Clinical Immunology, 124, 1319–25. Salvarani, C., Magnani, L., Catanoso, M., Pipitone, N., Versari, A., Dardani,
Park, M.C., Lee, S.W., Park, Y.B., and Lee, S.K. (2006). Serum cytokine pro- L., Pulsatelli, L., Meliconi, R., and Boiardi, L. (2012b). Tocilizumab: a
files and their correlations with disease activity in Takayasu’s arteritis. novel therapy for patients with large-vessel vasculitis. Rheumatology
Rheumatology (Oxford), 45, 545–8. (Oxford), 51, 151–6.
Park, M.C., Park, Y.B., Jung, S.Y., Lee, K.H., and Lee, S.K. (2006). Sampson, A. and Callen, J.P. (2006). The cutting edge:  Thalidomide for
Anti-endothelial cell antibodies and antiphospholipid antibodies in type 1 cryoglobulinemic vasculopathy. Archives of Dermatology,
Takayasu’s arteritis: correlations of their titers and isotype distributions 142, 972–4.
with disease activity. Clinical and Experimental Rheumatology, 24 (2 Savage, C.O., Harper, L., and Holland, M. (2002). New findings in pathogen-
Suppl. 41), S10–16. esis of antineutrophil cytoplasm antibody-associated vasculitis. Current
Pepper, R.J., Fabre, M.A., Pavesio, C., Gaskin, G., Jones, R.B., Jayne, D., Pusey, Opinion in Rheumatology, 14, 15–22.
C.D., and Salama, A.D. (2008). Rituximab is effective in the treatment of Schonermarck, U., Csernok, E., Trabandt, A., Hansen, H., and Gross, W.L.
refractory Churg-Strauss syndrome and is associated with diminished (2000). Circulating cytokines and soluble CD23, CD26 and CD30 in
T-cell interleukin-5 production. Rheumatology (Oxford), 47, 1104–5. ANCA-associated vasculitides. Clinical and Experimental Rheumatology,
Polzer, K., Soleiman, A., Baum, W., Axmann, R., Distler, J., Redlich, K., Kilian, 18, 457–63.
A., Kronke, G., Schett, G., and Zwerina, J. (2008). Selective p38MAPK Schreiber, A., Rolle, S., Peripelittchenko, L., Rademann, J., Schneider, W.,
isoform expression and activation in antineutrophil cytoplasmatic Luft, F.C., and Kettritz, R. (2010). Phosphoinositol 3-kinase-gamma
antibody-associated crescentic glomerulonephritis: role of p38MAPKal- mediates antineutrophil cytoplasmic autoantibody-induced glomerulo-
pha. Annals of the Rheumatic Diseases, 67, 602–8. nephritis. Kidney International, 77, 118–28.
Popa, E.R., Stegeman, C.A., Bos, N.A., Kallenberg, C.G., and Tervaert, J.W. Schreiber, A., Xiao, H., Jennette, J.C., Schneider, W., Luft, F.C., and Kettritz, R.
(1999). Differential B- and T-cell activation in Wegener’s granulomato- (2009). C5a receptor mediates neutrophil activation and ANCA-induced
sis. Journal of Allergy and Clinical Immunology, 103, 885–94. glomerulonephritis. Journal of the American Society of Nephrology, 20,
Proven, A., Gabriel, S.E., Orces, C., O’Fallon, W.M., and Hunder, G.G. (2003). 289–98.
Glucocorticoid therapy in giant cell arteritis: duration and adverse out- Sciascia, S., Rossi, D., and Roccatello, D. (2011). Interleukin 6 blockade as
comes. Arthritis and Rheumatism, 49, 703–8. steroid-sparing treatment for 2 patients with giant cell arteritis. Journal
Ramos-Casals, M., Stone, J.H., Cid, M.C., and Bosch, X. (2012). The cryoglo- of Rheumatology, 38, 2080–1.
bulinaemias. Lancet, 379, 348–60. Seitz, M., Reichenbach, S., Bonel, H.M., Adler, S., Wermelinger, F., and
Reissig, A., Forster, M., Mock, B., Schilder, C., and Kroegel, C. (2003). Villiger, P.M. (2011). Rapid induction of remission in large vessel vascu-
(Interferon-alpha treatment of the Churg-Strauss syndrome). Deutsche litis by IL-6 blockade. A case series. Swiss Medical Weekly, 141, w13156.
Medizinische Wochenschrift, 128, 1475–8. Seko, Y., Sato, O., Takagi, A., Tada, Y., Matsuo, H., Yagita, H., Okumura, K.,
Rhee, E.P., Laliberte, K.A., and Niles, J.L. (2010). Rituximab as maintenance and Yazaki, Y. (1996). Restricted usage of T-cell receptor Valpha-Vbeta
therapy for anti-neutrophil cytoplasmic antibody-associated vasculitis. genes in infiltrating cells in aortic tissue of patients with Takayasu’s arte-
Clinical Journal of the American Society of Nephrology, 5, 1394–400.  ritis. Circulation, 93, 1788–90.
610 SECTION 5 vasculitic diseases and syndromes and related disorders

Serada, S., Fujimoto, M., Mihara, M., Koike, N., Ohsugi, Y., Nomura, S., Terrier, B., Geri, G., Chaara, W., Allenbach, Y., Rosenzwajg, M.,
Yoshida, H., Nishikawa, T., Terabe, F., Ohkawara, T., et al. (2008). IL-6 Costedoat-Chalumeau, N., Fouret, P., Musset, L., Benveniste, O., Six, A.,
blockade inhibits the induction of myelin antigen-specific Th17 cells and et al. (2012). IL-21 modulates Th1 and Th17 responses in giant cell arte-
Th1 cells in experimental autoimmune encephalomyelitis. Proceedings of ritis. Arthritis and Rheumatism, 64, 2001–11.
the National Academy of Sciences of the U S A, 105, 9041–6. Trejo, O., Ramos-Casals, M., Garcia-Carrasco, M., Yague, J., Jimenez,
Shapiro-Shelef, M. and Calame, K. (2005). Regulation of plasma-cell devel- S., de la Red, G., Cervera, R., Font, J., and Ingelmo, M. (2001).
opment. Nature Review Immunology, 5, 230–42. Cryoglobulinemia: study of etiologic factors and clinical and immuno-
Shlomchik, M.J., Craft, J.E., and Mamula, M.J. (2001). From T to B and back logic features in 443 patients from a single center. Medicine (Baltimore),
again: positive feedback in systemic autoimmune disease. Nature Review 80, 252–62.
Immunology, 1, 147–53. Tsukadaira, A., Okubo, Y., Kitano, K., Horie, S., Momose, T., Takashi, S.,
Simon, H.U., Seelbach, H., Ehmann, R., and Schmitz, M. (2003). Clinical and Suzuki, J., Isobe, M., and Sekiguchi, M. (1999). Eosinophil active
immunological effects of low-dose IFN-alpha treatment in patients with cytokines and surface analysis of eosinophils in Churg-Strauss syn-
corticosteroid-resistant asthma. Allergy, 58, 1250–5. drome. Allergy and Asthma Proceedings, 20, 39–44.
Smith, D.A., Minthorn, E.A., and Beerahee, M. (2011). Pharmacokinetics Unizony, S., Arias-Urdaneta, L., Miloslavsky, E., Arvikar, S., Khosroshahi,
and pharmacodynamics of mepolizumab, an anti-interleukin-5 mono- A., Keroack, B., and Stone, J. (2012). Tocilizumab for the treatment
clonal antibody. Clinical Pharmacokinetics, 50, 215–27. of large-vessel vasculitis (giant cell arteritis, takayasu arteritis) and
Specks U, Merkel PA, Seo P, Spiera R, Langford CA, Hoffman GS, Kallenberg polymyalgia rheumatica. Arthritis Care and Research (Hoboken), 64,
CG, St Clair EW, Fessler BJ, Ding L, et al. Efficacy of remission-induction 1720–9.
regimens for ANCA-associated vasculitis. N Engl J Med. 2013 Aug Unizony, S.H., Dasgupta, B., Fisheleva, E., Rowell, L., Schett, G., Spiera, R.,
369: 417-27. Stone JH; RAVE-ITN Research Group. Zwerina, J., Harari, O., and Stone, J.H. (2013). Design of the tocilizumab
Spiera, R.F., Mitnick, H.J., Kupersmith, M., Richmond, M., Spiera, H., Peterson, in giant cell arteritis trial. International Journal of Rheumatology, 2013,
M.G., and Paget, S.A. (2001). A prospective, double-blind, randomized, 912562.
placebo controlled trial of methotrexate in the treatment of giant cell arte- Vaglio, A., Moosig, F., and Zwerina, J. (2012). Churg-Strauss syn-
ritis (GCA). Clinical and Experimental Rheumatology, 19, 495–501. drome: update on pathophysiology and treatment. Current Opinion in
Spizzo, G., Mitterer, M., and Gunsilius, E. (2010). Bortezomib for the Rheumatology, 24, 24–30.
treatment of refractory Type-1 cryoglobulinaemia. British Journal of van der Veen, B.S., Chen, M., Muller, R., van Timmeren, M.M., Petersen,
Haematology, 150, 235–7. A.H., Lee, P.A., Satchell, S.C., Mathieson, P.W., Saleem, M.A., Stegeman,
Stone, J.H., Holbrook, J.T., Marriott, M.A., Tibbs, A.K., Sejismundo, L.P., C.A., et al. (2011). Effects of p38 mitogen-activated protein kinase inhi-
Min, Y.I., Specks, U., Merkel, P.A., Spiera, R., Davis, J.C., et al. (2006). bition on anti-neutrophil cytoplasmic autoantibody pathogenicity in
Solid malignancies among patients in the Wegener’s Granulomatosis vitro and in vivo. Annals of the Rheumatic Diseases, 70, 356–65.
Etanercept Trial. Arthritis and Rheumatism, 54, 1608–18. Van Timmeren, M.M., Chen, M., and Heeringa, P. (2009). Review arti-
Stone, J.H., Merkel, P.A., Spiera, R., Seo, P., Langford, C.A., Hoffman, G.S., cle: Pathogenic role of complement activation in anti-neutrophil cyto-
Kallenberg, C.G., St Clair, E.W., Turkiewicz, A., Tchao, N.K., et  al. plasmic auto-antibody-associated vasculitis. Nephrology (Carlton),
(2010). Rituximab versus cyclophosphamide for ANCA-associated vas- 14, 16–25.
culitis. New England Journal of Medicine, 363, 221–32. van Timmeren, M.M., van der Veen, B.S., Stegeman, C.A., Petersen, A.H.,
Stone, J.H., Uhlfelder, M.L., Hellmann, D.B., Crook, S., Bedocs, N.M., and Hellmark, T., Collin, M., and Heeringa, P. (2010). IgG glycan hydrolysis
Hoffman, G.S. (2001). Etanercept combined with conventional treat- attenuates ANCA-mediated glomerulonephritis. Journal of the American
ment in Wegener’s granulomatosis: a six-month open-label trial to eval- Society of Nephrology, 21, 1103–14.
uate safety. Arthritis and Rheumatism, 44, 1149–54. Vinit, J., Bielefeld, P., Muller, G., and Besancenot, J.F. (2012). Efficacy of tocili-
Straumann, A., Conus, S., Grzonka, P., Kita, H., Kephart, G., Bussmann, zumab in refractory giant cell arteritis. Joint Bone Spine, 79, 317–8.
C., Beglinger, C., Smith, D.A., Patel, J., Byrne, M., et  al. (2010). Wagner, A.D., Goronzy, J.J., and Weyand, C.M. (1994). Functional profile
Anti-interleukin-5 antibody treatment (mepolizumab) in active eosino- of tissue-infiltrating and circulating CD68+ cells in giant cell arteri-
philic oesophagitis:  a randomised, placebo-controlled, double-blind tis. Evidence for two components of the disease. Journal of Clinical
trial. Gut, 59, 21–30. Investigation, 94, 1134–40.
Summers, S.A., Steinmetz, O.M., Gan, P.Y., Ooi, J.D., Odobasic, D., Kitching, Wang, H., Ma, J., Wu, Q., Luo, X., Chen, Z., and Kou, L. (2011). Circulating
A.R., and Holdsworth, S.R. (2011). Toll-like receptor 2 induces Th17 mye- B lymphocytes producing autoantibodies to endothelial cells play a role
loperoxidase autoimmunity while Toll-like receptor 9 drives Th1 autoim- in the pathogenesis of Takayasu arteritis. Journal of Vascular Surgery,
munity in murine vasculitis. Arthritis and Rheumatism, 63, 1124–35. 53, 174–80.
Szpirt, W.M., Heaf, J.G., and Petersen, J. (2011). Plasma exchange for induc- Wegener's Granulomatosis Etanercept Trial (WGET) Research Group
tion and cyclosporine A  for maintenance of remission in Wegener’s (2005). Etanercept plus standard therapy for Wegener’s granulomatosis.
granulomatosis—a clinical randomized controlled trial. Nephrology New England Journal of Medicine, 352, 351–61.
Dialysis Transplantation, 26, 206–13. Weyand, C.M., Fulbright, J.W., Hunder, G.G., Evans, J.M., and Goronzy,
Talamo, G., Claxton, D., Tricot, G., Fink, L., and Zangari, M. (2008). Response J.J. (2000). Treatment of giant cell arteritis: interleukin-6 as a biologic
to bortezomib in refractory type I cryoglobulinemia. American Journal marker of disease activity. Arthritis and Rheumatism, 43, 1041–8.
of Hematology, 83, 883–4. Weyand, C.M., Hicok, K.C., Hunder, G.G., and Goronzy, J.J. (1994). Tissue
Tanaka, F., Kawakami, A., Iwanaga, N., Tamai, M., Izumi, Y., Aratake, K., cytokine patterns in patients with polymyalgia rheumatica and giant cell
Arima, K., Kamachi, M., Nakamura, H., and Huang, M., et al. (2006). arteritis. Annals of Internal Medicine, 121, 484–91.
Infliximab is effective for Takayasu arteritis refractory to glucocorticoid Weyand, C.M., Ma-Krupa, W., Pryshchep, O., Groschel, S., Bernardino, R.,
and methotrexate. Internal Medicine, 45, 313–6. and Goronzy, J.J. (2005). Vascular dendritic cells in giant cell arteritis.
Tatsis, E., Schnabel, A., and Gross, W.L. (1998). Interferon-alpha treatment Annals of the New York Academy of Sciences, 1062,195–208.
of four patients with the Churg-Strauss syndrome. Annals of Internal Weyand, C.M., Younge, B.R., and Goronzy, J.J. (2011). IFN-gamma and
Medicine, 129, 370–4. IL-17: the two faces of T-cell pathology in giant cell arteritis. Current
Termeer, C.C., Simon, J.C., and Schopf, E. (2001). Low-dose interferon Opinion in Rheumatology, 23, 43–9.
alfa-2b for the treatment of Churg-Strauss syndrome with prominent Williams, J.M., Pettitt, T.R., Powell, W., Grove, J., Savage, C.O., and Wakelam,
skin involvement. Archives of Dermatology, 137, 136–8. M.J. (2007). Antineutrophil cytoplasm antibody-stimulated neutrophil
CHAPTER 42 experimental therapies for vasculitis 611

adhesion depends on diacylglycerol kinase-catalyzed phosphatidic acid Xiao, H., Schreiber, A., Heeringa, P., Falk, R.J., and Jennette, J.C. (2007).
formation. Journal of the American Society of Nephrology, 18, 1112–20. Alternative complement pathway in the pathogenesis of disease medi-
Xiao, H., Heeringa, P., Liu, Z., Huugen, D., Hu, P., Maeda, N., Falk, R.J., and ated by anti-neutrophil cytoplasmic autoantibodies. American Journal
Jennette, J.C. (2005). The role of neutrophils in the induction of glo- of Pathology, 170, 52–64.
merulonephritis by anti-myeloperoxidase antibodies. American Journal Zerbini, C.A. and Lomonte, A.B. (2012). Tofacitinib for the treatment of
of Pathology, 167, 39–45. rheumatoid arthritis. Expert Review of Clinical Immunology, 8, 319–31.
SECTION 6

Mimickers of vasculitis
CHAPTER 43

Antiphospholipid syndrome
David P. D’Cruz, Munther A. Khamashta,
and Graham R.V. Hughes

Introduction Moore and Mohr recognized that biologically false-positive sero-


logical tests for syphilis could occur, especially in lupus (Moore
The antiphospholipid (Hughes’) syndrome (APS) is one of the com- and Mohr 1952). Soon afterwards, two reports (Beaumont 1954;
monest causes of acquired thrombosis and pregnancy morbidity. Nilsson et al. 1975) linked a circulating anticoagulant with recur-
It is characterized by a vasculopathy that involves the vessel wall, rent foetal deaths, spontaneous abortions, and intrauterine growth
vascular endothelium, and procoagulant factors in the blood asso- restriction. Bowie et al. (1963) reported the paradoxical occurrence
ciated with the presence of antiphospholipid antibodies (aPL). The of thrombotic lesions in patients with a circulating anticoagu-
clinical presentation of APS is often confused for vasculitis, espe- lant. In 1972, the term ‘lupus’ anticoagulant (LA) was introduced
cially when seen in the context of systemic lupus erythematosus by Feinstein and Rapaport and in 1982, at the Heberden Round,
(SLE), and patients may be treated with immunosuppression rather Graham Hughes presented a patient with primary APS (PAPS)
than anticoagulation. It is now clear that this is a non-inflammatory (Feinstein and Rapaport 1972). In 1983, the first solid-phase radio-
vasculopathy. Nevertheless, it can sometimes be difficult to distin- immunoassay for the detection of anticardiolipin antibodies (aCL)
guish lesions seen in APS from those due to vasculitides such as was described (Harris et al. 1983). A few years later, Gharavi and
polyarteritis nodosa. For example, Asherson et al. (1992) reported colleagues developed an enzyme-linked immunosorbent assay
two patients with APS and skin lesions resembling vasculitis but (ELISA) while working in Hughes’ laboratory (Gharavi et  al.
who had microthrombosis at biopsy. Acalculous cholecystitis, an 1987). Between 1983 and 1990, numerous clinical associations
uncommon feature of vasculitis, has also been reported in APS were described, including livedo reticularis, thrombosis in mul-
(Dessailloud et al. 1998) and the angiographic appearance of APS tiple sites, pulmonary hypertension, myelopathy, chorea, bowel
may also rarely mimic vasculitis (Provenzale et al. 1998). Indeed, infarction, dementia, and pregnancy-related complications. More
it is being increasingly recognized that APS and systemic vascu- recently, these observations have been extended to include stenotic
litides can coexist, leading to diagnostic confusion and increased lesions of the renal and coeliac arteries, fractures, demyelinating
morbidity (Norden et al. 1995; Rocca et al. 1994; Rees et al. 2006). syndromes, and involvement of the eyes and ears. In 1990, three
Elsewhere in this book, the association between ANCA vasculitis groups (Takao Koike, Monica Galli, Steven Krilis, and their col-
and venous thromboembolism has been described—on the whole leagues) reported that in sera from patients with thrombosis, aCL
these patients are negative for aPL but this diagnosis should be kept bound to a plasma cofactor β2-glycoprotein 1 (β2GPI), a natu-
in mind. rally occurring anticoagulant (McNeil et al. 1990; Galli et al. 1990;
The main clinical features of this syndrome are arterial and Matsuura et al. 1990). It is now clear that β2GPI is one of the main
venous thrombosis, thrombocytopenia, and recurrent pregnancy antigens recognized by aPL. Several animal models were described
loss.. However, the spectrum of clinical manifestations seen in APS during the 1990s and the link between accelerated atheroma and
continues to expand and virtually every branch of medicine has aPL has been elucidated, particularly by Vaarala (1997). In recogni-
been influenced by this disorder. tion of Graham Hughes’s descriptions and major contributions to
this field, the eponym ‘Hughes’ syndrome’ was introduced in 1994.

APS: a brief history Antiphospholipid


The first test that detected an antibody against a phospholipid
dates back to 1906 when Wasserman developed a serological test
antibodies: methods of detection
for syphilis (Wasserman et  al. 1906). This involved a reaction aPL are a heterogeneous group of immunoglobulins directed at
between a lipid tissue antigen and an autoantibody in syphilitic protein antigens binding anionic phospholipids. Anticardiolipin
sera. Subsequently, it was determined that an alcoholic extract antibodies (aCL) may be detected routinely with phospholipid
of beef heart, the antigen in this test, contains a phospholipid, and β2GPI-dependent solid phase immunoassays (ELISAs).
termed cardiolipin by Mary Pangborn (1942). In 1952, Conley and Prolongation of phospholipid-dependent coagulation tests with
Hartmann described the association between a circulating antico- correction testing using a phospholipid source, such as washed
agulant and SLE (Conley and Hartmann 1952). In the same year, platelets, detects the lupus anticoagulant (LA). The assays for aCL
616 SECTION 6 mimickers of vasculitis

involve coating plastic plates with pure cardiolipin dissolved in picograms of antibody in serum. In contrast, larger amounts of anti-
ethanol, then blocking non-specific binding by incubation with body are required to prolong coagulation assays. A positive result in
foetal calf or adult bovine serum, and incubation with diluted an LA assay would suggest a higher titre of antibody than a positive
patient test sera. Bound antibody is identified using radioactive or result in an aCL assay, and the greater predictive value may be due to
enzyme-labelled antihuman antibody. the lower sensitivity of these assays (Tincani et al. 1998). Thus LA may
β2GPI is a 50-kD plasma protein that is a member of the com- be more specific but less sensitive than aCL testing for APS, reinforc-
plement control protein super family of molecules, possessing five ing the need to request both assays when suspecting this syndrome.
characteristic domains which contain the epitopes for aCL bind-
ing (Reid et al. 1986). β2GPI circulates in a ring form and, follow-
ing oxidation, this structure opens out into the ‘fishook’ structure
Demographics
that binds to anionic phospholipids on the cell surface in con- APS has, perhaps misleadingly, been recognized largely as a dis-
junction with molecules such as annexin A2 (Ioannou et al. 2012; ease of young women due to the association with SLE and preg-
Giannakopoulos et al. 2007). In vitro, β2GPI binds to anionic phos- nancy loss. The age of first thrombosis in APS has been shown to be
pholipids, DNA, and heparin, and has anticoagulant properties. predominately between 30 and 45 years (Piette and Cacoub 1998);
aCL found in infections such as HIV disease, unlike however, the syndrome has been described in children and the
autoimmune-associated aCL, do not bind β2GPI. This may explain elderly. There are problems with reporting bias, as young patients
the lack of thrombosis with infections. The presence of anti-β2GPI with thrombosis and pregnancy loss are more likely to be inves-
antibodies may provide a higher positive predictive value for the tigated, hence skewing the results. In fact, aPL are being increas-
development of thrombotic events compared to aCL and so-called ingly recognized in a diverse number of conditions, and in older
triple positivity may confer a significantly higher predictive value subjects. Among the latter, the prevalence may be as high as 7–12%
for the risk of thrombosis than single positivity (Pengo et al. 2011). (Fields et al. 1989; Schved et al. 1994) but the clinical significance
The term ‘lupus anticoagulant’ describes a laboratory phenom- is unclear. Racial differences have been noted, with IgA aCL more
enon in which plasma autoantibodies impair the function of ani- common in Afro-Caribbeans (Molina et al. 1997).
onic phospholipid in a variety of phospholipid-dependent in vitro There have been several large studies of the prevalence of aPL in SLE
coagulation tests. These include the activated partial thromboplas- patients. Perhaps the largest is the Euro-Lupus study, which found a
tin time, the Russell viper venom clotting time, the kaolin clotting prevalence of 24% IgG aCL, 13% IgM aCL, and 15% LA in a cohort
time, and the prothrombin time determined using dilute thrombo- of 1000 patients with SLE (Cervera et al. 1993). The prevalence of aPL
plastin. The main problems come from the laboratory techniques and definite APS may increase with longer follow-up, further preg-
that are necessary to detect the LA activity. These involve coagu- nancies, and repeat testing for aPS. Thus Perez-Vazquez et al. (1993)
lation procedures that are subject to a variety of conditions. It is showed that the prevalence of APS increased from 10% to 23% after
recommended that the presence of an LA be confirmed by a plate- 15–18 years in a large cohort of SLE patients. A further study of 1000
let neutralization test. To rule out coagulation factor deficiencies, APS patients has detailed the clinical features of the disorder (Cervera
the test should be performed with a mixture of patient and control et al. 2002). The Hopkins Lupus Cohort Study longitudinally studied
plasmas (Pengo et al. 2009). aPL and showed that SLE patients positive for LA had a 50% chance of
LA are not only directed against anionic phospholipids but a venous thrombotic event within 10 years of diagnosis (Somers et al.
against β2GPI and prothrombin (de Laat et  al. 2004). LA may 2002). In studies of patients with established APS, the risk of recurrent
also be directed at oxidized low-density lipoprotein and other thrombotic events is substantially increased, especially if tight antico-
phospholipid-binding proteins such as protein C, protein S, and agulation is not maintained (Khamashta et al. 1995).
heparan sulphate.
Concordance rates for the presence of aCL and LA in a given
patient have been estimated at between 60% and 70%. Some patients Definition and classification of APS
can be positive for only LA or aCL, but not both, and samples with An international consensus statement on classification criteria for
both LA and aCL have been separated into fractions with only one definite APS was published after a workshop in 1998 (the so-called
reactivity each, suggesting that the antibodies are related but clearly Sapporo criteria) (Wilson et al. 1999), which have been updated
differ in their binding to phospholipid (McNeil et al. 1989). For this (Sydney criteria) (Miyakis et al. 2006) (Table 43.1). Classification
reason, both tests should be requested when APS is suspected and criteria are used to facilitate research studies and are not a substi-
it is recommended that anti-β2GPI antibodies should also be tested tute for diagnostic criteria. When considering a diagnosis of APS,
to aid risk prediction. it is critically important for the clinician to exclude other possible
Other aPL have been identified, including antibodies to phos- causes of thrombosis. There are, however, very few causes of arterial
phatidylserine, phosphatidylinositol, phosphatidic acid, phos- and venous events in combination with pregnancy morbidity that
phatidylcholine, phosphatidylethanolamine, sphingomyelin, or a can occur in the same patient other than the APS.
mixture of these compounds. Other novel antigens that are under- Piette has urged clinicians to be aware of suspicious symptoms.
going clinical testing include antiprothrombin antibodies and anti- Fever and weight loss are unusual in APS and suggest infection
bodies to phosphatidylserine/ prothrombin complexes (Atsumi or malignancy. Splenomegaly is not a feature of APS unless com-
et al. 2000; Bertolaccini et al. 2005; Bertolaccini et al. 2013). At pre- plicated by other conditions. Thrombocytosis or leukocytosis are
sent, their detection adds little clinical information and is restricted unusual (Piette 1998). HIV infection can be a great mimicker, remi-
to research centres. niscent of syphilis in the centuries before effective treatment was
Several studies have shown that LA has a greater positive predictive available. Thrombosis associated with aPL in patients with HIV
value for thrombosis than aCL. ELISAs can be very sensitive, detecting disease has occurred but is rare.
CHAPTER 43 antiphospholipid syndrome 617

Table 43.1 Classification criteria for antiphospholipid syndrome Table 43.2 Indications for testing for antiphospholipid syndrome

Clinical criteria Venous/ arterial thrombosis before the age of 45 years


1. Vascular thrombosis Thrombosis after trivial provocation
One or more clinical episodes of arterial, venous, or small-vessel Association of arterial and venous thrombosis
thrombosis, in any tissue or organ. Thrombosis must be confirmed by
objective validated criteria, i.e. unequivocal findings of appropriate imaging Association of thrombosis and foetal loss
studies or histopathology. For histopathological confirmation, thrombosis Recurrent thrombotic events
should be present without significant evidence of inflammation in the
vessel wall. Family history

2. Pregnancy morbidity Thrombosis in an unusual site: retinal veins, portal, cerebral veins,


(a) One or more unexplained deaths of a morphologically normal renal veins, axillary veins
foetus at or beyond the 10th week of gestation, with normal foetal Coumarin-induced skin necrosis
morphology documented by ultrasound or by direct examination of
the foetus, or Systemic lupus erythematosus
(b) One or more premature births of a morphologically normal neonate Recurrent superficial thrombophlebitis
at or before the 34th week of gestation because of (i) eclampsia or
Recurrent miscarriage
severe pre-eclampsia defined according to standard definitions or (ii)
recognized features of severe placental insufficiency, or Foetal loss, especially stillbirth
(c) Three or more unexplained consecutive spontaneous abortions before Severe early pre-eclampsia
the 10th week of gestation, with maternal anatomic or hormonal
abnormalities and paternal and maternal chromosomal causes
excluded.
In studies of populations of patients who have more than one type of thrombophilias (Alarcón-Segovia et  al. 1996; Ames et  al. 1998;
pregnancy morbidity, investigators are strongly encouraged to stratify groups Brenner et al. 1996; Peddi and Kant 1995; Schutt et al. 1998).
of subjects according to (a), (b), or (c) above. The commonest site of venous thrombosis is the large veins of
Laboratory criteria the lower limb which may be associated with pulmonary emboli.
1. Lupus anticoagulant present in plasma on two or more occasions at least However, superficial thrombophlebitis, upper limb thrombo-
12 weeks apart, detected according to the guidelines of the International sis, portal vein thrombosis, Budd–Chiari syndrome, and cerebral
Society on Thrombosis and Hemostasis. venous thrombosis have all been reported.
2. Anticardiolipin antibody of IgG and/or IgM isotype in serum or plasma, Arterial thromboses, especially strokes in young patients, are a
present in medium or high titre (i.e. >40 GPL or MPL or >99th percentile), hallmark of this disorder. For example, Nencini et al. (1992) found
on two or more occasions, at least 12 weeks apart, measured by a standard 18% of young stroke patients (mean age 38 years) were positive for
enzyme linked immunosorbent assay. aPL (LA and aCL) whereas the Antiphospholipid Antibodies in
3. Anti-β2-glycoprotein 1 antibody of IgG and/or IgM isotype in serum or Stroke Study Group (APASS 1993) found 9.7% of first stroke patients
plasma (in titre >99th percentile), present on two or more occasions at had a positive aCL. Acute occlusion of peripheral arteries is unusual,
least 12 weeks apart, measured by a standardized ELISA, according to but premature peripheral vascular disease may be a feature of APS.
recommended procedures. In myocardial infarction, the prevalence of aCL is between 5% and
Definite APS is considered to be present if at least one of the clinical and one of the 15% (Vaarala 1998). Although this does not necessarily imply cau-
laboratory criteria are met (Wilson et al. 1999; Miyakis et al. 2006). sation, the high prevalence of both aPL and myocardial ischaemia
in SLE, combined with increasing evidence for a link between aPL
and atherosclerosis, is suggestive. Urbanus et al. (2009) highlighted
the risk of stroke and myocardial infarction in young women with
Clinical features of APS lupus anticoagulant, especially if there are other cardiovascular risk
factors such as smoking and oral contraceptive pill use.
Thrombosis Patients with persistent aPL may not experience thrombosis,
Arterial and venous thrombosis are major features of the syndrome. though, over time, patients with medium to high titre aPL, espe-
aPL should be included in the work up of unexplained thrombosis, cially if they are ‘triple positive’ have an increased risk of thrombosis
particularly in unusual sites or in young patients. Indications for (Shah et al. 1998; Pengo et al. 2011). Many women have recurrent
testing for aPL are shown in Table 43.2. Thrombosis may be spon- pregnancy morbidity as the only manifestation of the disease and
taneous or it may occur in association with other recognized risk these women are at increased risk of future thrombotic events. In a
factors. These risk factors can be divided into primary (hereditary large observational study the annual rates of deep vein thrombosis,
thrombophilias) and secondary (acquired) causes. The primary pulmonary embolism, superficial vein thrombosis, and cerebrovas-
causes include protein C, S or antithrombin deficiency, Factor V cular events were significantly higher in aPL-positive women than
Leiden mutations, and homocystinuria, etc. The acquired causes are in the control groups despite low-dose aspirin primary prophylaxis
immobility, trauma, surgery, use of oral contraceptives, pregnancy, (Gris et al. 2012). The various studies that have addressed the risk of
smoking, malignancy, diabetes, nephrotic syndrome, and vasculitis. thrombosis are summarized in Table 43.3. A major focus of research
There is some anecdotal evidence that thrombosis due to aPL may is identification of risk factors for thrombosis so that patients who
be more severe when occurring in association with the hereditary require more aggressive treatment can be identified. Although the
618 SECTION 6 mimickers of vasculitis

Table 43.3 Selected studies on thrombotic risk in APS

Study Design Patients aPL type Risk of thrombosis Comments


Rosove and Brewer Retrospective Primary (51) and aCL, LA 53% recurrence rate, with mean 91% followed initial pattern
1992 secondary APS (14) follow-up 5.2 years of thrombosis
Course after first Highest INR coincident with
thrombosis thrombosis: 2.6

Bongard et al. 1992 Retrospective VTE (107) and suspected aCL No association
VTE (186)
Khamashta et al. 1995 Retrospective SLE (85), PAPS (66), LA, aCL 69% recurrence rate for 20% had bleeding
10 years follow-up thrombosis complications, all had INR >3
Significantly reduced rate of
recurrent events in INR >3 group
Horbach et al. 1996 Retrospective, SLE (175), PAPS (23) IgG, IgM aCL, LA Odds ratio VTE: LA: 6.55*, IgM Higher titres of aCL in the
multivariate analysis Controls (blood donors) aCL>20 MPL units: 3.9* thrombotic SLE groups
Arterial: LA: 9.77* No additional information
aCL: NS from β2-GPI

Ginsburg et al. 1992 Nested case control American physicians aCL aCL>33GPL units
Relative risk:
VTE: 5.3*
CVA: 1.35 NS
Nencini et al. 1992 Case controlled Young strokes LA, aCL 18% young strokes positive for Recurrence rate for stroke
(15–44 years), aPL, 2% healthy controls higher
healthy volunteers in the aPL-positive group
APASS 1993 Case controlled Stroke (248) vs. non- IgM, IgG, Odds ratio: 2.33* Strength of association
stroke hospitalized (257) IgA aCL equivalent to hypertension
Ghirardello et al. 1994 Cross-sectional SLE (107) IgG aCL, LA Thrombosis associated with LA,
less extent aCL
Ginsberg et al. 1995 Cross-sectional VTE vs. no VTE IgG aCL, LA Odds ratio VTE LA: 9.4*
prospective cohort aCL: 0.7
aCL >50GPL: 1.9 NS
Vaarala et al. 1995 Nested case control Men with LDL cholesterol IgG aCL Cardiac end points (infarction, Risk independent of other
Helsinki Heart Study >5.2 mmol/l death) risk factors
Highest quartile of aCL: aCL levels higher in smokers
Odds ratio: 2.0*
Abu Shakra Prospective cohort SLE (390) LA, aCL LA: thrombosis, Odds ratio:7.96* No correlation with recurrent
et al. 1995 aCL: Coombs, thrombocytopenia fetal loss
No association with thrombosis
Simioni et al. 1996 Cross-sectional, VTE (59) vs. no VTE (117) LA LA: Odds ratio for DVT: 10.7* Prevalence of LA in idopathic
case control VTE: 10.7%
Finazzi et al. 1996 Prospective cohort PAPS (165) IgG aCL, LA IgG >40 GPL units, relative risk With previous thrombotic
Italian APS Registry Secondary APS:SLE (69) for thrombosis: 3.66* LA: NS history the relative risk rose
to 4.9
Schulman et al. 1998 Prospective cohort VTE (897 first episode) IgG aCL Risk ratio for recurrent VTE: 2.1* No recurrence in group with
6 months INR 2.0–2.85 (follow-up
post-thrombosis 4 years)
Of 20 recurrences in the aCL
positive group, 14/20 had
negative
aCL at time of thrombosis
(continued)
CHAPTER 43 antiphospholipid syndrome 619

Table 43.3 (Continued)

Study Design Patients aPL type Risk of thrombosis Comments


Kearon et al. 1999 Prospective ‘Idiopathic VTE’ LA, aCL Risk ratio for recurrence LA:
6.8* aCL: 2.3
Wahl et al. 1997 Meta-analysis SLE LA, aCL Odds ratio for VTE: LA: 6.32*,
aCL: 2.17*
Wahl et al. 1997 Meta-analysis aPL-positive patients non- LA, aCL Odds ratio for VTE: Odds ratio increased to3.21
autoimmune disease or LA: 11.1* for high titre aCL
previous thrombosis aCL: 1.64
Cervera et al. 2002 Prospective cohort aPL-positive patients LA, aCL Primary APS:53.1% Largest series to date
1000 patients APS with SLE: 36.2%
Urbanus et al. 2009 Controlled aPL-positive women with LA, aCL Hazard ratio for stroke: Large population based
observational stroke or myocardial aβ2GP1 201 case–control study (RATIO)
study infarction

Gris et al. 2012 Controlled aPL-positive women with LA, aCL Hazard ratio for VTE: Large prospective study of
observational pregnancy morbidity aβ2GP1 1.85 thrombosis risk following
study pregnancy loss in aPL +ve
women

* Significant result; NS, not significant; VTE, venous thromboembolism; CVA cerebrovascular accident; INR international normalized ratio.

data cited above suggests that 50% of patients with high titre aPL or secondary to intravascular obstruction or vessel wall disease.
may have a thrombotic event over 10 years, it is not possible to pre- Pathological livedo reticularis is usually extensive, occurring on the
dict with any certainty which individual patients will go on to have limbs, especially the forearms and knees, trunk, and buttocks, and
an event. These studies also show that low-dose aspirin alone is not has a broken pattern: so called livedo racemosa. The list of patho-
sufficient as primary prophylaxis against thrombosis. A prospec- logical causes includes cardiac failure, oxalosis, thrombocythae-
tive study of low-dose aspirin versus low-intensity anticoagulation mia, cryoglobulins, cold agglutinins, and arteritis, including PAN.
with warfarin (target INR 1.5) failed to recruit sufficient patients to The changes are initially reversible if the underlying cause can be
address the benefit of low-dose warfarin. At this stage, the best pre- corrected, but after a time the vessels become permanently dilated
dictor appears to be previous thrombotic history and persistently and anticoagulation has no effect on its extent or severity. Livedo
positive LA, and high titres of aCL and anti-β2GPI antibodies. reticularis is a marker for poor prognosis and more severe disease
in APS and as such is a powerful physical sign when this disorder is
Seronegative APS suspected (Toubi et al. 2005). Livedo reticularis may also be a useful
Over the last few years it has become apparent that there are many sign in patients with seronegative APS (Sangle et al. 2005a).
patients with the typical clinical features of APS, including preg- Livedo can be seen with nodules (cutaneous polyarteritis nodosa),
nancy losses, thrombotic events, and livedo reticularis, but who and with a segmental hyalinizing vasculitis, often with ulceration of
remain persistently negative for conventional testing for aPL: the the lower limbs, called livedoid vasculitis. This is also referred to as
so-called ‘seronegative APS’ (Hughes and Khamashta 2003; segmental hyalinizing vasculitis, or livedo reticularis with summer
Sangle et  al. 2005a). A  tiny minority of these patients may have
anti-β2-glycoprotein 1 and/or antiprothrombin antibodies, but it
may be that these patients have other explanations for their clinical
features (Bertolaccini et al. 1998). Rodriguez-Garcia et al. (2012)
showed no significant differences in the frequency of thrombotic
events or obstetric morbidity in patients with the so-called seron-
egative APS versus patients with seropositive APS.
Cutaneous disease
The most striking cutaneous lesion is that of livedo reticularis
(Figure 43.1). ‘This is a cyanotic, mottled discolouration of the skin
with a characteristic network pattern, which is accentuated by cold’
(Gibbs et al. 2005). The mottling of livedo reticularis may corre-
spond to watershed areas where the blood supply (larger arterioles)
is relatively diminished; hence the discoloration is due to dilata-
tion and stagnation of blood within capillaries and minute small
vessels. Livedo may be physiological (cutis marmorata), primary, Fig. 43.1 Livedo reticularis in a patient with systemic lupus erythematosus.
620 SECTION 6 mimickers of vasculitis

(tPA) highlights that thrombosis may be a key factor in this condi-


tion (Klein and Pittelkow 1992).
Other reported cutaneous associations include anetoderma,
ulcers, Degos’ disease, splinter haemorrhages, superficial throm-
bophlebitis, distal cutaneous ischaemia, and Sneddon’s syndrome.
However, closer examination of Sneddon’s syndrome (strokes and
livedo reticularis) reveals that many of these patients in fact have
APS with the finding of aCL and antiprothrombin antibodies
(Kalashnikova et al. 1990; Kalashnikova et al. 1999).

Neurological
There is a very broad spectrum of CNS involvement in APS, as
shown in Table 43.4. Stroke is a hallmark of APS. The age of onset
in APS, though, is several decades earlier than in the typical stroke
population (Levine et al. 1995) and the ischaemic events may occur
in any territory (Coull et al. 1992).
In SLE patients, MRI abnormalities are common in both
aPL-positive and negative patients. Sailer et al. (1997) found that
abnormalities greater than 8 mm in diameter were more likely to
be aPL-related than inflammatory (Figures 43.4 and 43.5). In many
patients, these clinical and radiological signs may be mistaken for
multiple sclerosis. Radiologically, it is almost impossible to dis-
tinguish large high-signal lesions on brain MRI due to APS from
Fig. 43.2 Livedo and leg ulceration in a patient with livedoid vasculitis. those due to multiple sclerosis (Cuadrado et al. 2000). Anecdotally,
some patients who apparently have multiple sclerosis but who are
positive for aPL have benefited from anticoagulation, though this is
often a difficult clinical decision (Ferreira et al. 2005).
ulceration (Figure 43.2). The latter condition is a chronic disease
Many patients complain of other problems such as migraine, poor
with ulcers, which tend to heal with hyperpigmentation and atro-
memory, and cognitive impairment. Brain MRI is often helpful in
phie blanche (Figure 43.3), affecting the feet and lower legs. The
absence of a sufficient perivascular infiltrate is against vasculitis
as a primary cause. It is not usually associated with aPL; however,
Acland et  al. (1999) reported four patients in whom aPL were Table 43.4 Spectrum of CNS involvement in antiphospholipid
syndrome
found. Successful treatment of resistant livedoid vasculitis in six
patients with non-healing ulcers by tissue plasminogen activator
Cerebrovascular ischaemia
Stroke
Transient ischaemic attack
Cerebral venous sinus thrombosis
Ocular ischaemia
Dementia
Acute ischaemic encephalopathy
Atypical migraine
Seizures
Chorea
Transverse myelopathy
Guillain–Barré syndrome
Diabetic peripheral neuropathy
Sensorineural hearing loss
sudden onset
progressive
Transient global amnesia
Fig. 43.3 Livedo vasculitis. Characteristic lesions on the medial lower leg showing
stellate shaped areas of necrosis and stellate white scars (atrophie blanche) Psychiatric disorders
surrounded by macular purpura and secondary haemosiderin pigmentation. Orthostatic hypotension
(From Sams and Sams (2002) Cutaneous manifestations. In Vasculitis, Ball, G.V. and
Bridges, S.L. Jr, eds. Oxford University Press.) (Source: Brey and Escalante 1998.)
CHAPTER 43 antiphospholipid syndrome 621

More severe associations include dementia (Gomez-Puerta et al.


2005b), psychosis, myelopathy, multiple sclerosis-like syndrome,
chorea, sensorineural hearing loss, cerebral artery or vein occlu-
sion, and retinal lesions. Although the pathological lesion is likely
to be thrombotic (small or large-vessel ischaemia), or embolic,
there is some evidence for direct neurotoxicity (Furie et al. 1994).
Two controversial studies have examined the association
between abnormal cognitive function and aPL. Hanly et al. (1999)
prospectively studied cognitive function in SLE patients and found
that psychomotor speed was reduced in patients who had persis-
tently positive IgG aCL. Menon et al. (1999) found similar results
in 45 SLE patients. Those patients with persistently elevated IgG
aCL over a 2 to 3-year period performed poorer in tasks requir-
ing speed of attention and concentration. No association was found
with anti-DNA antibody or C3 levels.
The prevalence of aPL in migraine is debated. A case–control study
failed to find an association in patients less than 60  years old for
migraine with or without focal neurological deficits compared with
controls (Tietjen et al. 1998). Cavestro et al. (2011) found that 12% of
migraineurs had one or more positive aPL compared to 3% of healthy
controls, supporting an association between migraine and positive aPL.

Fig. 43.4 MRI of brain showing lesions indistinguishable radiologically from Pregnancy


multiple sclerosis. APS is frequently diagnosed following investigation for recur-
rent miscarriage, pregnancy morbidity being one of the major
manifestations of the syndrome. In pregnancies that do not end in
these patients, revealing high-signal lesions. Electroencephalography miscarriage or foetal loss, there is a high incidence of early-onset
(EEG) is also a useful test in these patients. Lampropoulos et  al. pre-eclampsia, intrauterine growth restriction, placental abruption,
(2005) showed that EEG abnormalities of bitemporal slow activity, and premature delivery (Lima et al. 1996). Since the classification
suggesting cerebrovascular insufficiency, are common and correlate criteria for APS have been amended (Wilson et al. 1999; Miyakis
with the presence of aPL, even when the MRI is normal. et al. 2006), a patient with adverse pregnancy outcome may now be
labelled as APS without a history of foetal loss. The prevalence of
aPL in the general obstetric population is low (<2%), so universal
screening is not warranted (Harris and Spinnato 1991); however,
AH
any woman with a history of three or more first trimester miscar-
riages should be tested for these antibodies. Rai et al. (1995) have
reported that 15% of women with a history of three or more con-
secutive miscarriages have persistently positive aPL results. Testing
for aPL other than LA and aCL in women with recurrent miscar-
riage is of no clinical value (Branch et al. 1997) although anti-β2GPI
antibodies remain highly relevant (Simchen et al. 2011). The risk
of pregnancy loss is directly related to antibody titre:  the higher
the titre the greater the risk of adverse obstetric outcome (Simchen
et al. 2011) although the presence of aPL does not preclude success-
ful pregnancy. Once the diagnosis of APS is made and confirmed,
serial aPL determinations are not useful. Previous poor obstetric
history remains the most important predictor of future risk (Lima
et al. 1996; Danza et al. 2012).
The mechanism of pregnancy loss in APS remains uncertain. It
seems likely that progressive thrombosis of the microvasculature of
the placenta, which leads to infarction (Figure 43.6) may cause pla-
cental insufficiency, which in turn leads to foetal growth restriction
and foetal death in some patients. However, not all placentas exam-
ined have shown areas of thrombosis or infarction and it is, there-
fore, likely that other mechanisms are operative (Lockshin 1998).
A variety of hypotheses have been proposed to explain how aPL
might cause pregnancy loss, including: (1) inhibition of placental
prostaglandins and thromboxane; (2) inhibition of placental gon-
Fig. 43.5 Large-vessel disease with a parieto-occipital infarct. adotrophin; (3) competition with annexin V; (4) displacement of
622 SECTION 6 mimickers of vasculitis

1989; Straaton et al. 1989) and the association of aPL and cardiac


valve disease in SLE has been confirmed in a meta-analysis (Zuily
et al. 2011). In the absence of SLE, Doppler echocardiogram has
revealed a prevalence of valvular lesions in patients with APS of
between 32% and 38% (Hojnik et al. 1996). In non-APS patients
referred for valve replacement, aPL were found in 19 of 89 (21%)
compared with 9% of controls (Bouillanne et al. 1996). The mitral
valve is affected more commonly than the aortic valve and the
lesions may be vegetations or valvular thickening (Khamashta
et al. 1990). Functionally the valve may become regurgitant, but
stenosis is rarely encountered. The haemodynamic significance
is usually minor, although valve replacement has been required
in some patients. The vegetations may be a source of thrombo-
embolism. Infective endocarditis is uncommon, but a condition
of pseudoendocarditis has been described in both SLE and pri-
Fig. 43.6 Placental infarcts.
mary APS, and is characterized by fever, murmurs, valve vegeta-
tions, splinter haemorrhages, increased aPL, and negative blood
cultures.
annexin V; and (5) inhibition of trophoblast proliferation (reviewed It is likely that thrombosis on a damaged valve and healing with
in Meroni et al. 2012). fibrosis are key players in the valve deformity. Whether aPL are
Landmark animal studies have suggested that complement, espe- instrumental in initial damage of the valves remains to be eluci-
cially C5a, may have an important role in the pathogenesis of foetal dated, but deposits of immunoglobulin (including aCL) and com-
loss (Girardi et al. 2004). Heparin may block the effects of comple- plement in diseased valves have been documented, suggesting a
ment and thus prevent foetal loss by an anti-inflammatory rather role in the development of valvular pathology. APS patients with
than antithrombotic mechanism. It is not clear if these mechanisms valve disease undergoing valve replacement surgery are at high
pertain to the human situation and a study from Italy did not sup- risk of complications with a mortality rate of 12.5% and significant
port this (Reggia et al. 2012). postoperative complications (Erdozain et al. 2012).
Many investigators have found a statistically higher rate of aPL Diastolic dysfunction has also been documented by a number
in women with infertility, though not all agree. Proof that aPL of groups (Hasnie et al. 1995). Coudray et al. (1995) studied 18
are associated with a specific type of infertility or influence preg- patients with echocardiogram and found impairment of myocar-
nancy outcome with assisted reproductive technology is lacking dial relaxation and filling dynamics of the left ventricle in PAPS
(Buckingham and Chamley 2009). compared with controls. They concluded that the abnormalities
may be due to subclinical myocardial damage, possibly second-
Pulmonary ary to microvascular thrombosis. This is supported by the obser-
The commonest manifestation is pulmonary emboli and thrombo- vation (Kaplan et al. 1992) that widespread cardiac dysfunction
embolic pulmonary hypertension. Gertner and Lie (1993) noted due to multiple arteriolar thrombi can occur in the absence of
that other manifestations may be seen in primary APS, including coronary artery disease. Cardiac MRI has supported this obser-
a syndrome resembling adult respiratory distress syndrome, alveo- vation with a relatively high prevalence of asymptomatic myo-
lar haemorrhage, microvascular pulmonary thrombosis, transient/ cardial ischaemic disease in patients with antiphospholipid
migratory infiltrates on chest radiography, and (pathologically) syndrome (Sacré 2010).
capillaritis. Pulmonary arterial hypertension is also associated with aPL both
as a consequence of thromboembolic disease and the ‘plexogenic’
Haematological type of pulmonary hypertension (Asherson and Cervera 2007).
Thrombocytopenia has been found in up to 30% of primary APS,
and is seen more frequently in SLE with aPL. Despite thrombocyto- Musculoskeletal
penia, some patients are still at risk of thrombosis. Coombs-positive Avascular necrosis (AVN) of bone (also referred to as osteonecro-
haemolytic anaemia may also occur. sis) may be associated with aPL. Tektonidou et al. (2003) found MRI
scanning to be a sensitive test with 20% of primary APS patients
Cardiac showing evidence of AVN. A previous group of 800 SLE patients
Several cardiac lesions apart from coronary artery disease have were retrospectively reviewed and 37 were found to have devel-
been reported in association with aPL (Tenedios et  al. 2006). oped AVN (4.6%); 27 (73%) were positive for aPL (Asherson et al.
Libman–Sacks endocarditis was first described in 1924 in patients 1993). Given the aPL prevalence figures in SLE, the expected rate
with sterile lesions of the valvular and mural endocardium would have been 30–40%, suggesting that aPL may be a risk fac-
(Libman and Sacks 1924). The association between Libman– tor for AVN. However, Alarcón-Segovia et al. (1989) and Houssiau
Sacks endocarditis and aPL was first noted in 1985 (D’Alton et al. et al. (1998) failed to find any association between aPL and AVN in
1985) in a young woman with transient ischaemic attacks, valvu- SLE patients. Although glucocorticoid treatment may influence the
lar disease, and LA. Several groups have highlighted a probable incidence of AVN in the SLE population, it has also been reported
role for aPLs in the pathogenesis of valvular lesions in patients in primary APS patients who are glucocorticoid-naïve (Asherson
with SLE (Chartash et al. 1989; Galve et al. 1989; Khamashta et al. et al. 1993; Nagasawa et al. 1989).
CHAPTER 43 antiphospholipid syndrome 623

Spontaneous metatarsal fractures are another manifestation obsolescence of glomeruli may result, culminating in renal impair-
linked to aPL that have been described (Sangle et al. 2004). These ment or failure (Amigo et al. 1992). Renal impairment may also
patients presented with fractures that occurred either spontane- result from cortical renal ischaemia due to occlusion of small renal
ously or after trivial trauma and many had normal bone mineral vessels, which may give rise to foci of cortical necrosis. This may
density and were not taking glucocorticoids. How aPL cause these be multiple in the catastrophic APS. The risks of renal biopsy in
fractures remains unknown but speculatively it is possible that patients with APS or aPL may be increased due to bleeding com-
thrombosis of the blood supply to the bones may be an explanation. plications and these patients need special precautions (Jordan
et al. 2013).
Endocrine It is not uncommon for patients with aPL to come to renal trans-
Addison’s disease due to adrenal thrombosis/ haemorrhage is a rec- plantation. This may be due to associated lupus nephritis or to the
ognized clinical association of aPL. Anterior pituitary failure has primary effect of aPL on the kidney. Loss of renal allograft may
been reported in a patient with APS (Pandolfi et al. 1997). occur due to a thrombotic vascular lesion or to a recurrence of
thrombotic microangiopathy (Vaidya et al. 1998). D’Cruz (2005)
Renal has reviewed the broad spectrum of renal complications of APS.
Almost any part of the renal vasculature can be affected in the APS.
Since the early descriptions of the disease, labile hypertension has Gastrointestinal
been noted (Hughes 1983). Amigo et al. (1992) found renal involve- Intestinal ischaemia and perforations due to thrombosis are rare
ment in up to 25% of patients with primary APS. There are now but well described, especially in the context of the catastrophic APS.
several well-documented cases of renal artery thrombosis or steno- There have been descriptions of mesenteric angina in aPL-positive
sis occurring in primary or secondary APS (Asherson et al. 1991; patients, some of whom had abdominal bruits. Investigation of
Ostuni et al. 1990) (Figure 43.7). The exact aetiology is unclear, as these patients disclosed celiac (Figure 43.8) and mesenteric artery
there are case reports of thrombosis and improvement with antico- stenoses, further supporting the idea that a medium to large-vessel
agulation, while other lesions are probably atherosclerotic in nature. vasculopathy is a feature of the APS (Sangle et al. 2006).
Godfrey et al. (2000) documented five cases of renal artery steno-
sis in APS patients, three of which developed in patients already on Catastrophic APS
anticoagulation. A case–control study has confirmed these findings The term catastrophic APS was first introduced by Asherson in
showing a significantly increased prevalence of renal artery steno- 1992 to describe an accelerated form that results in multiorgan fail-
sis in 26% of patients with aPL compared to 8% and 3% in control ure (Figure 43.9)(Asherson 1992). The majority of the patients to
groups with hypertension or who were renal organ donors, respec- date have suffered with primary APS and precipitating factors have
tively (Sangle at al. 2003). Furthermore, anticoagulation appeared to
improve outcome in these patients (Sangle et al. 2005b). Renal corti-
cal infarcts may occur due to occlusion of renal arteries secondary to
thrombosis, stenosis, or embolism. Renal vein thrombosis has also
been documented (Gluek et al. 1985; Morgan and Feneley 1994).
Tektonidou et al. (2004) found a strong association between APS
nephropathy, arterial thrombosis, and livedo reticularis.
Thrombotic microangiopathy occurs in primary or second-
ary APS (Bhandari et al. 1998). The microangiopathy can involve
both the vascular tree and the glomerular tufts, and ischaemic

Fig. 43.7 Right renal artery stenosis in APS. Fig. 43.8 Celiac artery stenosis in APS.
624 SECTION 6 mimickers of vasculitis

(a) (b) of an abnormal ABI was significantly higher than in healthy con-
trols. Medina et al. (2003) showed that patients with primary APS
had significantly increased carotid artery intima–media thickness
(IMT) compared to controls. These patients were also more likely
to have reduced lumen diameter. Patients with increased IMT were
more likely to have had arterial events such as strokes. The descrip-
tion of renal and celiac artery lesions further supports this idea,
although the precise mechanisms remain elusive. Post-mortem
histology in patients with SLE and aPL suggests that vascular
intimal fibrous hyperplasia is a common finding (Sipek-Dolnicar
et al. 2002).

Differences between primary and


secondary APS
Vianna et al. (1994) found that primary APS and APS secondary
to SLE had similar clinical features, but heart valve disease, auto-
immune haemolytic anaemia, lymphopenia, neutropenia, and low
C4 levels were more common in patients with SLE. Anti-dsDNA
Fig. 43.9 (a and b) Catastrophic APS in a patient with severe skin necrosis and antibodies or antibodies to extractable nuclear antigens (Sm and
gangrene from widespread thrombosis. RNP) were not found in PAPS and their presence should suggest
a secondary cause. The distinction between PAPS and APS due to
SLE can sometimes be difficult. Features such as thrombocytope-
been infections, operations, anticoagulant withdrawal, and drugs. nia, anaemia, renal, and CNS disease may be seen in both condi-
Classification criteria for the catastrophic APS have been validated tions. Piette et al. (1993) have been strong advocates of exclusion
(Cervera et al. 2005). criteria for primary APS (Table  43.5). There does not appear to
Asherson (1998) initially reviewed 50 patients with catastrophic be any difference in rates of arterial or venous thrombosis, or foe-
APS documented in the literature and this has recently been tal loss (Finazzi et al. 1996; Krnic-Barrie et al. 1997; Vianna et al.
extended to 250 patients using registry data (Bucciarelli et  al. 1994). Shah et al. (1998) found IgM aCL more often in SLE than
2006; Cervera and Espinosa 2012). Fatal outcome was seen in 44%, PAPS (22 of 42 patients, vs. one of ten patients) but no difference in
with cerebral disease (mainly stroke), cerebral haemorrhage, and thrombotic rates.
encephalopathy being the most common cause of death, followed The number of cases reported in the literature of patients with
by cardiac disease and infection. The most significant prognostic primary APS evolving into SLE is small. Silver et  al. (1994) and
factor was the presence of SLE. Renal involvement, hypertension, Mujic et al. (1995) have reported the evolution in small numbers
and pulmonary disease (ARDS in 50%, pulmonary emboli, pulmo- (seven of 71 and three of 80, respectively) but Asherson et al. (1989)
nary oedema, and infrequently intra-alveolar haemorrhage) were and Vianna et  al. (1994) did not find any such progression. The
also common. short period of follow-up may have been responsible for the latter
result (5 and 2 years respectively), as several patients have devel-
Atherosclerosis oped the syndrome after 10 years. The presence of high titre ANA
The aetiology of atherosclerosis is multifactorial with traditional (>1:320) and lymphopenia may be predictive of subsequent SLE
risk factors including hyperlipidaemia, hypertension, smok-
ing, diabetes, and family history. In recent years, the role of the
immune system has begun to be appreciated. The role of aPL is Table 43.5 Proposed exclusion criteria for primary APS
also being unravelled. It is well recognized that SLE patients have
accelerated atherosclerosis and the risk is increased in the setting Malar or discoid rash
of aPL. It is also known that aPL can bind and activate endothelial
Oral, pharyngeal, or nasal ulceration
cells, increase monocyte adherence in vitro, and bind to oxidized
LDL, perhaps resulting in increased uptake into vessel walls via Fc Frank arthritis
receptors (Del Papa et al. 1997). Hasunuma et al. (1997) showed Pleurisy/ pericarditis
that macrophages take up labelled oxidized LDL at an increased
Persistent proteinuria >0.5 g/day, due to biopsy-proven immune complex
rate in the presence of aPL. Further evidence supporting a role for
aPL in atherosclerosis comes from mouse models where immuni- Inflammatory/ immune complex-mediated glomerulonephritis
zation of LDL receptor-deficient mice with human β2GPI led to Lymphopenia (<1000 cells/μl)
acceleration of early atherosclerosis (George et al. 1998).
Antibodies to dsDNA (crithidia or radioimmunoassay), or ENA
Studies using the ankle-brachial pressure index (ABI) have shown
that an abnormal ABI (<1.0) occurs in 19% of APS patients with a ANA >1:320
history of previous thrombotic events (Baron et al. 2005), and in Treatment with drugs known to produce aPL
APS patients with a previous history of pregnancy morbidity but
Follow-up >5 years from the initial clinical manifestation.
without thrombotic events, an abnormal ABI was found in 23% of
patients (Christodoulou et al. 2006). In both studies the prevalence (Piette et al. 1993)
CHAPTER 43 antiphospholipid syndrome 625

(Seisdedos et al. 1997). A larger study of 128 patients confirmed cultured endothelial cells and normal monocytes (Amengual et al.
the low rate of progression from primary APS to SLE; during the 1998; Cuadrado et  al. 1997; Reverter et  al. 1996; Tripplett 1995;
follow-up and after a median disease duration of 8 years, 11 (8%) Lopez-Pedrera et al. 2006). Tissue factor is not normally expressed
patients developed SLE, and six (5%) developed lupus-like dis- by cells in contact with blood, but with inflammatory stimuli, tissue
ease. A positive Coombs test increased the risk of developing SLE factor expression on monocytes and endothelial cells can be induced,
(Gomez-Puerta et al. 2005a). which may then initiate normal and pathological coagulation.
β2GPI can adhere to endothelial cells, offering suitable epitopes
Pathophysiology for circulating aPL, especially anti-β2GPI, antibodies. Functional
changes then occur to induce cell activation. This has been demon-
Immunogenetics strated in vitro by up-regulation of adhesion molecule expression
A number of studies have examined HLA antigens in patients with and by production of proinflammatory cytokines rendering the
primary and secondary APS. Most studies have involved small endothelium prothrombotic (Del Papa et al. 1997; Simantov et al.
numbers of patients. In primary APS, an association has been 1995). Pierangeli et al. (1999) have demonstrated both in vitro and
noted with HLA-DR53, -DR4, -DQ7, and -DR7. In disease associ- in vivo endothelial activation when affinity-purified aPL was pre-
ated with SLE and aCL, HLA-DR7, -DR4, -DR53, or no associa- sented to human umbilical vein endothelial cells, and in a mouse
tion emerge (Granados et al. 1997; Schutt et al. 1998). Examination model of the microcirculation.
of alleles encoding complement components has found a variable Analysis of monoclonal aPL has shown that arginine residues
association. There is geographical variation, with HLA-DR7 more are critical in determining the binding of human monoclonal
common in Latins (Sebastiani et al. 1996). antiphospholipid antibodies to clinically relevant antigens (Giles
The gene encoding β2GPI (apolipoprotein H, or apoH) is located et al. 2006).
on chromosome 17 (Steinkasserer et al. 1992). A number of alleles, Finally, a mechanism of action of LA through platelet activation
designated APOH*1, APOH*2, APOH*3, and APOH*4, have been has been postulated and demonstrated in primary APS by detec-
identified and allele frequency varies among populations. The tion of activated platelets by flow cytometry (Emmi et al. 1997) as
binding site of β2GPI to anionic phospholipids resides in the fifth well as under flow conditions (Font et al. 2002).
domain. Genetic mutations in the fifth domain of β2GPI can affect It would seem likely that a two-hit hypothesis is necessary for
aPL binding and the production of β2GPI-dependent aPL (Kamboh thrombosis to occur. The prothrombotic state is present, but a sec-
and Mehdi 1998). ond event may be necessary to trigger thrombosis. Various animal
Gharavi et  al. (1999) demonstrated that lysine-rich peptides models exist but no model has been able to mimic all features of
from cytomegalovirus (CMV), adenovirus, and Bacillus subti- APS. In particular, venous thrombosis has been rarely produced
lis, which have sequence homology for the phospholipid bind- without non-physiological stimuli. The pathogenesis of APS has
ing region of β2GPI, can induce production of high levels of aPL been reviewed by Pierangeli et  al. (2006), Giannakopoulos et  al.
and anti-β2GPI antibodies when bound to bovine serum albumin (2007 and 2013), and Ioannou (2012).
in Freund’s adjuvant. This may be a mechanism of production of A large number of conditions have been reported in association
infection-associated aPL, but the latter are not usually associated with aPL (Table 43.6). Thrombosis is unusual in most of these con-
with thrombosis in humans, so this model may not explain the pro- ditions except for SLE. In a prospective study, Merkel et al. (1996)
duction of autoimmune aPL.

Mechanism of pathogenicity Table 43.6 Conditions associated with aPL


The exact mechanism by which aPL leads to thrombosis is
unknown. Mechanisms may be divided into those where antibod- Autoimmune disorders
ies interfere with haemostatic reactions of physiological anticoagu- SLE/ Sjögren syndrome
lants or those that affect cell mediated events such as endothelial Systemic vasculitis
cells, monocytes, and platelets. The heterogeneity of the antibodies
and the multiple proposed mechanisms may explain the clinical Malignancy
features, in particular why some individuals experience only preg- Infection
nancy loss and others venous or arterial thrombosis. Syphilis
At a simplistic level, the mechanism of thrombosis may be Lyme disease
through binding of natural anticoagulants such as β2GPI, protein Human immunodeficiency virus
C, protein S, or thrombomodulin, the primary activator of protein
Hepatitis C
C (Oosting et al. 1993). However, under physiological conditions,
Cytomegalovirus
β2GPI is a weak anticoagulant and individuals who lack β2GPI do
not appear to suffer with thrombosis (Bancsi et al. 1992). Inhibition Mycoplasma
of the function of protein C has been documented and remains Drugs
one of the hypotheses for the mechanism of thrombosis. Galli et al. Chlorpromazine
(1998) have shown activated protein C resistance in patients with Quinine/ quinidine
aPL; the effect was β2GPI dependent, but antiprothrombin antibod- Hydralazine
ies were not associated with the phenotype.
Procainamide
Various studies have shown that the sera or IgGs from patients with
Phenytoin
LA or aCL can induce tissue factor (a cell glycoprotein) expression in
626 SECTION 6 mimickers of vasculitis

found 16% of rheumatoid arthritis and SLE patients had either IgG triple positive, should be managed. There is no doubt that some of
or IgM aCL, compared with 4% of blood donors. Patients with scle- these patients will have a future event, but predicting those at high
roderma, myositis, undifferentiated connective tissue disease, and risk has proven difficult.
ANCA-associated vasculitis did not differ from the control blood Many physicians recommend low-dose aspirin for management
donors in the prevalence of aPL. The manifestations of APS are very of those patients without previous thrombosis, particularly if titres
rarely seen in infection or drug-associated aCL, although occa- of aPL are high; however, there is little objective evidence to sup-
sional case reports of the development of thrombosis in patients port this. Gris et al. (2012) showed that thrombosis risk is increased
with AIDS and CMV infection have been described (Soweid et al. despite low-dose aspirin in women with aPL and previous preg-
1995). Procainamide has been shown to produce β2GPI-dependent nancy morbidity. A small prospective randomized controlled trial
antibodies that are potentially pathogenic (Merrill et al. 1997). of low-dose aspirin in persistently aPL-positive individuals also
showed no benefit (Erkan et  al. 2007). In SLE patients, there is
evidence supporting use of hydroxychloroquine, as it was protec-
APS and systemic vasculitis tive against future thrombosis in the Hopkins Lupus Cohort (Petri
APS may complicate systemic vasculitis (Rees et al. 2006). aPL were 1997). It has also been used as a prophylactic agent to prevent DVT
found in 17% of 144 patients; nine patients had definite APS and a in patients undergoing orthopaedic surgery.
further four had clinical and serological features of APS but did not Traveller’s thrombosis has become topical and, in the absence
meet all the classification criteria. APS has been described in a patient of evidence, it seems reasonable to recommend simple meas-
with granulomatosis with polyangiitis (Shovman et  al. 2013)  and ures such as support stockings, hydration, and exercises to APS
previous studies showed that aPL may occur in the systemic vascu- patients undertaking long journeys by any transport mode. In very
litides. Bleil et al. (1991) did not find significantly elevated levels in high-risk aPL-positive patients, some clinicians recommend a sin-
patients with granulomatosis with polyangiitis (Wegener’s) granulo- gle injection of heparin prior to commencing the journey, though
matosis compared with controls, but there were significant associa- this is clearly anecdotal. Other high-risk periods include the post-
tions with four patients with polyarteritis nodosa and 24 patients operative period, especially after abdominal, gynaecological, and
with polymyalgia rheumatica/ giant cell arteritis (GCA). Consistent orthopaedic surgery where heparin prophylaxis is important.
with these findings, other authors have found significant associa-
tions with GCA (Duhaut et al. 1998; McHugh et al. 1990; Watts et al. Thrombotic events
1990). Duhaut et al. (1998), in multivariate analyses, did not find Arterial and venous events are treated in similar fashion, that is
any association with thrombosis. They postulated that aCL may be initially with unfractionated heparin or low molecular weight hep-
produced as a consequence of endothelial damage and not have a arins, followed by oral anticoagulation with agents such as warfarin.
relationship to thrombosis. Thrombosis is a feature of Behçet’s syn- When using unfractionated heparin, it is important to examine the
drome, however aPL have not been consistently found (Aydintug baseline activated partial thromboplastin time (APTT) level, which
et  al. 1993). Case reports have also described aPL in Takayasu’s may indicate the presence of LA. If there is concern that the APTT
arteritis (Yokoi et al. 1996) and classical PAN (Dasgupta et al. 1997; will not accurately reflect the degree of anticoagulation, other tests
Norden et al. 1995). There is an increasing literature describing the can be employed such as a factor Xa inhibition test. Alternatively,
occurrence of aPL and APS in Henoch-Schönlein purpura (IgA low molecular weight heparins such as tinzaparin or enoxaparin can
related disease) (Liu and Zhang 2012). be used. Similar problems may occur rarely with warfarin and the
It is not clear if the aPL in these conditions are pathogenic and International Normalized Ratio (INR) level, but can be avoided by
contribute to the clinical features, although it is postulated that a using chromogenic factor X levels and prothrombin-proconvertin
more severe clinical course may occur if both vasculitis and aPL are times (Bartholomew and Kottke-Marchant 1998).
present at the same time (Norden et al. 1995). Given the description The current recommendation is that anticoagulation should be
of a high rate of venous thromboses in Wegener’s granulomatosis lifelong, given the risk of recurrent events; however, lifelong antico-
(Merkel et al. 2005) discussed in Chapter 31, this finding may be agulation is not without hazard, with major bleeding occurring in
highly relevant. up to 20–40% in some long-term studies of non-APS patients (Fihn
et al. 1993; Landefeld and Goldman 1989). It is tempting not to rec-
Treatment ommend lifelong anticoagulation if the initial venous thrombosis is
minor, particularly if there has been a clear-cut precipitating factor.
General measures In this situation, co-morbid factors as well as titres of aPL should
Given the increased thrombotic risk of these patients and the possi- be considered and discussed with the patient. No such uncertainty
ble association with atherosclerosis, vascular risk factors should be should exist with arterial events, particularly cerebral, where life-
addressed. Lifestyle measures and management of risk factors, such long anticoagulation is usually indicated.
as obesity, smoking, hypertension, diabetes, and hyperlipidaemia, The optimal intensity of anticoagulation has been debated.
are important. The combined oral contraceptive pill and hormone Khamashta et al. (1995) and Rosove and Brewer (1992) found no
replacement therapy should be avoided. thrombosis in patients who maintained the INR greater than 3 and
2.9 respectively, and Schulman et al. (1998) found no recurrence in
Asymptomatic patients a group of patients with venous thromboembolic events who main-
Increasing numbers of asymptomatic or recurrent miscarriage tained their INR in the range of 2.0–2.85. In those patients that
patients are being detected due to improved awareness of the con- fail high-intensity warfarin, it has been our practice to add aspi-
dition. A major dilemma is how these patients, who have not had a rin although little evidence, other than anecdotal, exists to support
thrombotic event but remain persistently aPL positive, especially if or discourage this approach. Randomized, controlled trials have
CHAPTER 43 antiphospholipid syndrome 627

suggested that lower INR levels may be sufficient in APS patients feature of normal pregnancy and that low molecular weight hepa-
with only venous thrombotic events (reviewed in Lim et al. 2006; rin does not exacerbate this loss.
Ruiz-Irastorza et al. 2011). For patients who continue to have pregnancy losses despite hepa-
Treatment of catastrophic APS has involved glucocorticoids, rin and low-dose aspirin treatment, intravenous immunoglobulin
cyclophosphamide, plasmapheresis, rituximab, intravenous gam- (IVIg) may be an option. IVIg is expensive and definitive proof of
maglobulin, anticoagulation, and, in the occasional patient, throm- its efficacy is needed before endorsing its use as a first-line therapy
bolytic therapy. More than one modality has been tried in most (Gordon and Kilby 1998).
patients. Although no clear approach has emerged, one study of Most authorities agree that one of the main reasons for the
mortality suggests that anticoagulation, glucocorticoids, and improving outcome of APS pregnancies is closer obstetric surveil-
plasma exchange may be useful treatments (Bucciarelli et al. 2006; lance (Mascola and Repke 1997). Viable APS pregnancies have a
Cervera and Espinosa 2012). high incidence of obstetric and foetal complications, including
Thrombocytopenia is common in APS. Patients remain at risk of intrauterine growth restriction, prematurity and pre-eclampsia;
thrombosis despite this and warfarin should be continued unless hence, close monitoring including uterine artery Doppler scans
the platelet count falls below 50 × 109/l. Anticoagulation may be and timely delivery may improve foetal outcome in these women.
continued even when the platelet count is below 50  × 109/l, but
the intensity of anticoagulation may need to be reduced. In patients Conclusions
with severe thrombocytopenia, glucocorticoids and intravenous
immunoglobulins are usually effective. Other agents such as dana- The identification of APS and its pathophysiology have significantly
zol, dapsone, and even aspirin have been tried. There is some debate advanced in the last 25 years, touching nearly all branches of medi-
as to the effectiveness of splenectomy in APS patients. Hakim et al. cine. In the field of systemic vasculitis, it is becoming clear that APS
(1998) and Galindo et al. (1999) have shown that splenectomy may patients may also suffer the consequences of thrombosis, contribut-
allow successful long-term outcomes. ing to a poor prognosis. It would therefore be prudent to consider
A frequent problem in the management of female patients is screening newly diagnosed systemic vasculitis patients for aPL.
menorrhagia, particularly when INRs are maintained at high levels.
In this situation, the ‘Mirena’ coil, an intrauterine coil with a silas-
References
tic capsule containing levonorgestrel, has been found to be helpful Abu-Shakra, M., Gladman, D., Urowitz, M., and Farewell, V. (1995).
(Pisoni et al. 2006). Anticardiolipin antibodies in systemic lupus erythematosus:  Clinical
and Laboratory correlations. American Journal of Medicine, 99, 624–8.
A number of novel oral anticoagulants have been licensed for
Acland, K., Darvay, A., Wakelin, S., and Russell Jones, R. (1999). Livedoid
use in postoperative thromboprophylaxis and atrial fibrillation, vasculitis:  a manifestation of the antiphospholipid syndrome? British
including drugs such as rivaroxaban, dabigatran, and apixiban. Journal of Dermatology, 140, 131–5.
These drugs do not need therapeutic INR monitoring and are set to Alarcón-Segovia, D., Deleze, M., Oria, C., et  al. (1989). Antiphospholipid
revolutionize anticoagulation strategies in a number of thrombotic antibodies and the antiphospholipid syndrome in systemic lupus ery-
disorders. It is not clear if these new agents are as effective as warfa- thematosus: A prospective study of 500 consecutive patients. Medicine,
rin especially in complicated thrombotic APS with recurrent events 68, 353–65.
and clinical trials are on-going. Alarcón-Segovia, D., Ruiz-Arguelles, G., Garces-Eisele, J., et  al. (1996).
Inherited activated protein C resistance in a patient with familial pri-
Pregnancy mary antiphospholipid syndrome. Journal of Rheumatology, 23, 2162–5.
Amengual, O., Atsumi, T., Khamashta, M., and Hughes, G. (1998). The role of
The management of pregnancy in women with APS remains con- the tissue factor pathway in the hypercoaguable state in patients with the
troversial. Antithrombotic treatment is preferred to glucocorti- antiphospholipid syndrome. Thrombosis and Haemostasis, 79, 276–81.
coids (which were once widely recommended). The current choice Ames, P., Tommasino, C., D’Andrea, G., Iannaccone, L., Brancaccio, V., and
is aspirin or heparin, or both, and this has been reviewed in detail Margaglione, M. (1998). Thrombophilic genotypes in subjects with
by Derksen et al. (2004). This area remains controversial, especially idiopathic antiphospholpid antibodies—prevalence and significance.
since Farquharson et al. (2002) showed in a randomized, controlled Thrombosis and Haemostasis, 79, 46–9.
trial that high pregnancy success rates were achieved with low-dose Amigo, M., Garcia-Torres, R., Robles, M., Bochiccio, T., and Reyes, P. (1992).
Renal involvement in primary antiphospholipid syndrome. Journal of
aspirin and no advantage was gained by the addition of low molec-
Rheumatology, 19, 1181–5.
ular weight heparin. Women with APS and previous thromboem-
Antiphospholipid Antibodies in Stroke Study Group (APASS) (1993). Clinical,
bolism are at extremely high risk in pregnancy and the puerperium, radiological, and pathological aspects of cerebrovascular disease associ-
and should be given antenatal thromboprophylaxis with subcuta- ated with antiphospholipid antibodies. Stroke, 24 (Suppl. 1), 120–3.
neous heparin. Asherson, R.A. (1992). The catastrophic antiphospholipid syndrome. Journal
Warfarin exposure between 6 and 12 weeks of gestation can be of Rheumatology, 19, 508–12.
associated with embryopathy, which is characterized by stippled Asherson, R.A. (1998). The catastrophic antiphospholipid syndrome.
epiphyses and nasal hypoplasia. Therefore, the change from war- A review of the clinical features, possible pathogenesis and treatment.
farin to heparin should be achieved prior to 6 weeks’ gestation. Lupus, 7 (Suppl. 2), S55–62.
Heparin does not cross the placenta and is not known to cause Asherson, R.A. and Cervera, R. (2007). Pulmonary hypertension, antiphos-
pholipid antibodies, and syndromes. Clinical Reviews in Allergy and
any adverse foetal effects. Baglin et al. (2006) published guidelines
Immunology, 32, 153–8.
for the use of heparin, including the use of low molecular weight Asherson, R., Jacobelli, S., Rosenberg, H., Mckee, P., and Hughes G. (1992).
heparin in pregnancy. The risk of osteoporosis in the mother Skin nodules and macules resembling vasculitis in the antiphospholipid
from long-term heparin use has probably been overestimated; a syndrome: a report of two cases. Clinical and Experimental Dermatology,
controlled study by Carlin et al. (2004) showed that bone loss is a 17, 266–9.
628 SECTION 6 mimickers of vasculitis

Asherson, R., Khamashta, M., Ordi Ros, J., et  al. (1989). The primary Brenner, B., Vulfsons, S., Lanir, N., and Nahir, M. (1996). Coexistence of
antiphospholipid syndrome:  Major clinical and serological features. familial antiphospholipid syndrome and factor V Leiden:  Impact on
Medicine, 68, 366–74. thrombotic diathesis. British Journal of Haematology, 94, 166–7.
Asherson, R., Liote, F., Page, B., et al. (1993). Avascular necrosis of bone and Brey, R. and Escalante, A. (1998). Neurological manifestations of antiphos-
antiphospholpid antibodies in systemic lupus erythematosus. Journal of pholipid antibody syndrome. Lupus, 7 (Suppl. 2), S67–74.
Rheumatology, 20, 284–8. Bucciarelli, S., Espinosa, G., Cervera, R., Erkan, D., Gomez-Puerta, J.A.,
Asherson, R., Noble, G., and Hughes, G. (1991). Hypertension, renal artery Ramos-Casals, M., Font, J., Asherson, R.A.; European Forum on
stenosis and the ‘primary’ antiphospholipid syndrome. Journal of Antiphospholipid Antibodies (2006). Mortality in the catastrophic
Rheumatology, 18, 1413–5. antiphospholipid syndrome: causes of death and prognostic factors in a
Atsumi, T., Ieko, M., Bertolaccini, M.L., Ichikawa, K., Tsutsumi, A., Matsuura, series of 250 patients. Arthritis and Rheumatism, 54, 2568–76.
E., and Koike, T. (2000). Association of autoantibodies against the Buckingham, K.L., Chamley, L.W. (2009). A critical assessment of the role of
phosphatidylserine-prothrombin complex with manifestations of the antiphospholipid antibodies in infertility. J Reprod Immunol, 80, 132–45.
antiphospholipid syndrome and with the presence of lupus anticoagu- Carlin, A.J., Farquharson, R.G., Quenby, S.M., Topping, J., and Fraser, W.D.
lant. Arthritis and Rheumatism, 43, 1982–93. (2004). Prospective observational study of bone mineral density dur-
Aydintug, A., Tokgoz, G., D’Cruz, D., et al. (1993). Antibodies to endothe- ing pregnancy:  low molecular weight heparin versus control. Human
lial cells in patients with Behcet’s Disease. Clinical Immunology and Reproduction, 19, 1211–4.
Immunopathology, 67, 157–62. Cavestro, C., Micca, G., Molinari, F., Bazzan, M., Di Pietrantonj, C., Aloi,
Baglin, T., Barrowcliffe, T.W., Cohen, A., Greaves, M.; British Committee for R., Pedemonte, E., Iannini, R., Frigeri, M.C., and Roccatello, D. (2011).
Standards in Haematology (2006). Guidelines on the use and monitor- Migraineurs show a high prevalence of  antiphospholipid  antibodies.
ing of heparin. British Journal of Haematology, 133, 19–34. Journal of Thrombosis and Haemostasis, 9, 1350–4. 
Bancsi, L., van der Linden, I., and Bertina, R. (1992). Beta 2-glyco-protein Cervera, R. and Espinosa, G. (2012). Update on the catastrophic antiphos-
I deficiency and the risk of thrombosis. Thrombosis and Haemostasis, pholipid syndrome and the ‘CAPS Registry’. Seminars in Thrombosis and
67, 649–53. Hemostasis, 38, 333–8.
Baron, M.A., Khamashta, M.A., Hughes, G.R., and D’Cruz, D.P. (2005). Cervera, R., Font, J., Gomez-Puerta, J.A., Espinosa, G., Cucho, M., Bucciarelli, S.,
Prevalence of an abnormal ankle-brachial index in patients with primary Ramos-Casals, M., Ingelmo, M., Piette, J.C., Shoenfeld, Y., Asherson, R.A.;
antiphospholipid syndrome: preliminary data. Annals of the Rheumatic Catastrophic Antiphospholipid Syndrome Registry Project Group (2005).
Diseases, 64, 144–6. Validation of the preliminary criteria for the classification of catastrophic
Bartholomew, J. and Kottke-Marchant, K. (1998). Monitoring anticoagu- antiphospholipid syndrome. Annals of the Rheumatic Diseases, 64, 1205–9.
lant therapy in patients with the lupus anticoagulant. Journal of Clinical Cervera, R., Khamashta, M.A., Font, J., Sebastiani, G.D., Gil, A., Lavilla, P.,
Rheumatology, 4, 307–11. Domenech, I., Aydintug, A.O., Jedryka-Goral, A., de Ramon, E., et  al.
Beaumont, J. (1954). Syndrome hemorrhagique acquis de a un anticoagulant. (1993). Systemic lupus erythematosus: clinical and immunologic patterns
Sang, 25, 1–5. of disease expression in a cohort of 1,000 patients. The European Working
Bertolaccini, M.L., Atsumi, T., Koike, T., Hughes, G.R., and Khamashta, M.A. Party on Systemic Lupus Erythematosus. Medicine (Baltimore), 72, 113–24.
(2005). Antiprothrombin antibodies detected in two different assay sys- Cervera, R., Piette, J.C., Font, J., Khamashta, M.A., Shoenfeld, Y., Camps,
tems. Prevalence and clinical significance in systemic lupus erythemato- M.T., et al.; Euro-Phospholipid Project Group (2002). Antiphospholipid
sus. Thrombosis and Haemostasis, 93, 289–97. syndrome: clinical and immunologic manifestations and patterns of dis-
Bertolaccini, M.L., Roch, B., Amengual, O., Atsumi, T., Khamashta, M.A., ease expression in a cohort of 1,000 patients. Arthritis and Rheumatism,
and Hughes, G.R. (1998). Multiple antiphospholipid tests do not 46, 1019–27.
increase the diagnostic yield in antiphospholipid syndrome. British Chartash, E., Lans, D., Paget, S., Qamar, T., and Lockshin, M. (1989). Aortic
Journal of Rheumatology, 37, 1229–32. insufficiency and mitral regurgitation in patients with systemic lupus
Bertolaccini, M.L., Sciascia, S., Murru, V., et  al. (2013). Prevalence erythematosus and the antiphospholipid syndrome. American Journal
of antibodies to prothrombin in solid phase (aPT) and to of Medicine, 86, 407–12.
phosphatidylserine-prothrombin complex (aPS/PT) in patients with and Christodoulou, C., Zain, M., Bertolaccini, M.L., Sangle, S., Khamashta, M.A.,
without lupus anticoagulant. Thrombosis and Haemostasis, 109, 207–13. Hughes, G.R., and D’Cruz, D.P. (2006). Prevalence of an abnormal
Bhandari, S., Harnden, P., Brownjohn, A., and Turney, J. (1998). Association ankle-brachial index in patients with antiphospholipid syndrome with
of anticardiolipin antibodies with intraglomeruli thrombi and renal dys- pregnancy loss but without thrombosis: a controlled study. Annals of the
function in lupus nephritis. Quarterly Journal of Medicine, 91, 401–9. Rheumatic Diseases, 65, 683–4.
Bleil, L., Manger, B., Winkler, T., et al. (1991). The role of antineutrophil cyto- Conley, C.L. and Hartmann, R.C. (1952). A hemorrhagic disorder caused by
plasmic antibodies, anticardiolipin antibodies, von Willebrand factor circulating anticoagulant in patients with disseminated lupus erythema-
antigen, and fibronectin for the diagnosis of systemic vasculitis. Journal tosus. Journal of Clinical Investigation, 31, 621–2.
of Rheumatology, 18, 1199–206. Coudray, N., de Zuttere, D., Bletry, O., et al. (1995). M Mode and doppler
Bongard, O., Reber, G., Bounameaux, H., and de Moerloose, P. (1992). echocardiographic assessment of left ventricular diastolic function in
Anticardiolipin in acute venous thromboembolism. Thrombosis and primary antiphospholipid syndrome. British Heart Journal, 74, 531–5.
Haemostasis, 67, 724. Coull, B., Levine, S., and Brey, R. (1992). The role of antiphospholipid anti-
Bouillanne, O., Millaire, A., de Groote, P., et al. (1996). Prevalence and clini- bodies in stroke. Neurology Clinics, 10, 125–43.
cal significance of antiphospholipid antibodies in heart valve disease: a Cuadrado, M.J., Khamashta, M.A., Ballesteros, A., Godfrey, T., Simon, M.J., and
case control study. American Heart Journal, 132, 790–5. Hughes, G.R. (2000). Can neurologic manifestations of Hughes (antiphos-
Bowie, E., Thompson, J., Pascuzzi, C., and Owen, C. (1963). Thrombosis pholipid) syndrome be distinguished from multiple sclerosis? Analysis of
in SLE despite circulating anticoagulants. Journal of Laboratory and 27 patients and review of the literature. Medicine (Baltimore), 79, 57–68.
Clinical Medicine, 62, 416–30. Cuadrado, M.J., Lopez-Pedrera, C., Khamashta, M., et al. (1997). Thrombosis
Branch, D., Silver, R., Pierangeli, S., Van Leeuwen, I., and Harris, E. (1997). in primary antiphospholipid syndrome: a pivotal role for monocyte tis-
Antiphospholipid antibodies other than lupus anticoagulant and anti- sue factor expression. Arthritis and Rheumatism, 40, 834–41.
cardiolipin antibodies in women with recurrent pregnancy loss, fertile D’Alton, J., Preston, D., Bormanis, J., Green, M., and Kraag, G. (1985).
controls and antiphospholipid syndrome. Obstetrics and Gynecology, 89, Multiple transient ischaemic attacks, lupus anticoagulant and verrucous
549–55. endocarditis. Stroke, 16, 512–4.
CHAPTER 43 antiphospholipid syndrome 629

Danza, A., Ruiz-Irastorza, G., and Khamashta, M. (2012). Antiphospohlipid Galindo, M., Khamashta, M., and Hughes, G. (1999). Splenectomy for
syndrome in obstetrics. Best Practice and Research. Clinical Obstetrics refractory thrombocytopenia in the antiphospholipid syndrome.
and Gynaecology, 26, 65–76. Rheumatology, 38, 848–53.
Dasgupta, B., Almond, M., and Tanqueray, A. (1997). Polyarteritis nodosa Galli, M., Comfurius, P., Maassen, C., et al. (1990). Anticardiolipin antibod-
and the antiphospholipid syndrome. British Journal of Rheumatology, ies directed not to cardiolipin but to a plasma protein cofactor. Lancet,
36, 1210–12 335, 1544–7.
D’Cruz, D.P. (2005). Renal manifestations of the antiphospholipid syndrome. Galli, M., Comfurius, P., Barbui, T., Zwaal, R., and Bevers, E. (1992).
Lupus, 14, 45–8. Anticoagulant activity of beta 2GPI is potentiated by a distinct group
de Laat, H.B., Derksen, R.H., Urbanus, R.T., Roest, M., and de Groot, P.G. (2004). of anticardiolipin antibodies. Thrombosis and Haemostasis, 68, 297–300.
Beta2-glycoprotein I-dependent lupus anticoagulant highly correlates with Galli, M., Ruggeri, L., and Barbui, T. (1998). Differential effects of anti-beta
thrombosis in the antiphospholipid syndrome. Blood, 104, 3598–602. 2GP1 and antiprothrombin antibodies on the anticoagulant activity of
Del Papa, N., Guidali, L., Sala, A., et al. (1997). Endothelial cells as target activated protein C. Blood, 91, 1999–2004.
for antiphospholpid antibodies: human polyclonal and monoclonal anti- Galve, E., Ordi, J., Candell-Riera, J., Permanyer-Miralda, G., Vilardell, M.,
beta 2-glycoprotein 1 antibodies react in vitro with endothelial cells and Soler-Soler, J. (1989). Valvular heart disease in systemic lupus ery-
through adherent beta 2-glycoprotein 1 and induce endothelial activa- thematosus. New England Journal of Medicine, 320, 740–1.
tion. Arthritis and Rheumatism, 40, 551–61. George, J., Afek, A., Gilburd, B., et al. (1998). Induction of early atheroscle-
Derksen, R.H., Khamashta, M.A., and Branch, D.W. (2004). Management of rosis in LDL receptor deficient mice by immunization with beta 2 glyco-
the obstetric antiphospholipid syndrome. Arthritis and Rheumatism, 50, protein 1. Circulation, 98, 1108–15.
1028–39. Gertner, E. and Lie, J. (1993). Pulmonary capillaritis, alveolar haemorrhage,
Dessailloud, R., Papo, T., Vaneecloo, S., Gamblin, C., Vanhille, P., and Piette, and recurrent microvascular thrombosis in primary antiphospholipid
J. (1998). Acalculous ischaemic gallbladder necrosis in the catastrophic syndrome. Journal of Rheumatology, 20, 1224–8.
antiphospholipid syndrome. Arthritis and Rheumatism, 41, 1318–20. Gharavi, A.E., Harris, E.N., Asherson, R.A., and Hughes, G.R.V. (1987).
Duhaut, P., Berruyer, M., Pinede, L., et al. (1998). Anticardiolipin antibod- Anticardiolipin antibody: isotype distribution and phospholipid speci-
ies and giant cell arteritis: a prospective, multicenter case-control study. ficity. Annals of the Rheumatic Diseases, 46, 1–6.
Arthritis and Rheumatism, 41, 701–9. Gharavi, E., Chaimovich, H., Cucurull, E., et  al. (1999). Induction of
Emmi, L., Bergamini, C., Spinelli, et al. (1997). Possible pathogenic role of antiphospholipid antibodies by immunisation with synthetic viral and
activated platelets in the primary antiphospholipid syndrome involving bacterial peptides. Lupus, 8, 449–55.
the central nervous system. Annals of the New York Academy of Science, Ghirardello, A., Doria, A., Ruffati, A., et al. (1994). Antiphospholipid anti-
823, 188–200. bodies (aPL) in systemic lupus erythematosus. Are they specific tools
Erdozain, J.G., Ruiz-Irastorza, G., Segura, M.I., Amigo, M.C., Espinosa, G., for the diagnosis of aPL syndrome. Annals of the Rheumatic Diseases,
Pomar, J.L., Perez-Valero, I., Ateka-Barrutia, O., and Khamashta, M.A. 53, 140–2.
(2012). Cardiac  valve replacement in patients with antiphospholipid Giannakopoulos, B. and Krilis, S.A. (2013). The pathogenesis of the antiphos-
syndrome. Arthritis Care and Research (Hoboken), 64, 1256–60. pholipid syndrome. New England Journal of Medicine, 368, 1033–44.
Erkan, D., Harrison, M.J., Levy, R., Peterson, M., Petri, M., Sammaritano, Giannakopoulos, B., Passam, F., Rahgozar, S., and Krilis, S.A. (2007). Current
L., Unalp-Arida, A., Vilela, V., Yazici, Y., and Lockshin, M.D. (2007). concepts on the pathogenesis of the antiphospholipid syndrome. Blood,
Aspirin for primary thrombosis prevention in the antiphospholipid syn- 109, 422–30.
drome: a randomized, double-blind, placebo-controlled trial in asymp- Gibbs, M.B., English, J.C. 3rd, Zirwas, M.J. (2005). Livedo reticularis: an
tomatic antiphospholipid antibody-positive individuals. Arthritis and update. J Am Acad Dermatol, 52, 1009–19.
Rheumatism, 56, 2382–91. Giles, I., Lambrianides, N., Pattni, N., Faulkes, D., Latchman, D., Chen,
Farquharson, R.G., Quenby, S., and Greaves, M. (2002). Antiphospholipid P., Pierangeli, S., Isenberg, D., and Rahman, A. (2006). Arginine resi-
syndrome in pregnancy:  a randomized, controlled trial of treatment. dues are important in determining the binding of human monoclonal
Obstetrics and Gynecology, 100, 408–13. antiphospholipid antibodies to clinically relevant antigens. Journal of
Feinstein, D. and Rapaport, S. (1972). Acquired inhibitors of blood coagula- Immunology, 177, 1729–36.
tion. Progress in Haemostasis and Thrombosis, 1, 75–95. Ginsberg, J., Wells, P., Brill-Edwards, P., et al. (1995). Antiphospholipid anti-
Ferreira, S., D’Cruz, D.P., and Hughes, G.R. (2005). Multiple sclerosis, neu- bodies and venous thromboembolism. Blood, 86, 3685–91.
ropsychiatric lupus and antiphospholipid syndrome: where do we stand? Ginsburg, K., Liang, M., Newcomer, L., et al. (1992). Anticardiolipin antibod-
Rheumatology (Oxford), 44, 434–42. ies and the risk for ischaemic stroke and venous thrombosis. Annals of
Fields, R., Toubbeh, H., Searles R., and Bankhurst, A. (1989). The prevalence Internal Medicine, 117, 997–1002.
of anticardiolipin antibodies in a healthy elderly population and its asso- Girardi, G., Redecha, P., and Salmon, J.E. (2004). Heparin prevents antiphos-
ciation with antinuclear antibodies. Journal of Rheumatology, 16, 623–5. pholipid antibody-induced fetal loss by inhibiting complement activa-
Fihn, S., McDonell, M., Martin, D., et al. (1993). Risk factors for complica- tion. Nature Medicine, 10, 1222–6.
tions of chronic anticoagulation: A multicentre study. Annals of Internal Gluek, H., Kant, K., Weiss, M., Pollak, V., Miller, M., and Coots, M. (1985).
Medicine, 118, 511–20. Thrombosis in systemic lupus erythematosus. Relation to the pres-
Finazzi, G., Brancaccio, V., Moia, M., et al. (1996). Natural history and risk ence of circulating anticoagulants. Archives of Internal Medicine, 145,
factors for thrombosis in 360 patients with antiphopsholipid antibod- 1389–95.
ies. A four year prospective study from the Italian Registry. American Godfrey, T., Abbs, I., Khamashta, M., and Hughes, G. (2000). Antiphospholipid
Journal of Medicine, 100, 530–6. syndrome and renal artery stenosis. Quarterly Journal of Medicine,
Font, J., Espinosa, G., Tassies, D., Pino, M., Khamashta, M.A., Gallart, T., 93, 127–9.
Cervera, R., Escolar, G., Hughes, G.R., Ingelmo, M., Ordinas, A., and Gomez-Puerta, J.A., Martin, H., Amigo, M.C., Aguirre, M.A., Camps, M.T.,
Reverter, J.C. (2002). Effects of beta2-glycoprotein I  and monoclonal Cuadrado, M.J., Hughes, G.R., and Khamashta, M.A. (2005a). Long-term
anticardiolipin antibodies in platelet interaction with subendothelium follow-up in 128 patients with primary antiphospholipid syndrome: do
under flow conditions. Arthritis and Rheumatism, 46, 3283–9. they develop lupus? Medicine (Baltimore), 84, 225–30.
Furie, R., Ishikawa, T., Dhawan, V., and Eidelberg, D. (1994). Alternating Gomez-Puerta, J.A., Cervera, R., Calvo, L.M., Gomez-Anson, B., Espinosa,
hemichorea in primary antiphospholipid syndrome: evidence for con- G., Claver, G., Bucciarelli, S., Bove, A., Ramos-Casals, M., Ingelmo, M.,
tralateral striatal metabolism. Neurology, 44, 2197–9. and Font, J. (2005b). Dementia associated with the antiphospholipid
630 SECTION 6 mimickers of vasculitis

syndrome:  clinical and radiological characteristics of 30 patients. Kamboh, M. and Mehdi, H. (1998). Genetics of apolipoprotein H (2-glycoprotein
Rheumatology (Oxford), 44, 95–9. 1) and anionic phospholipid binding. Lupus, 7 (Suppl. 2), S10–13.
Gordon, C. and Kilby, M.D. (1998). Use of intravenous immunoglobulin Kaplan, S., Chartash, E., Pizzarello, R., and Furies, R. (1992). Cardiac mani-
therapy in pregnancy in systemic lupus erythematosus and antiphos- festations of the antiphospholipid syndrome. American Heart Journal,
pholipid syndrome. Lupus, 7, 429–33. 124, 1331–8.
Granados, J., Vargas-Alarcon, G., Drenkard, C., et al. (1997). Relationship of Kearon, C., Gent, M., Hirsh, J., et al. (1999). A comparison of three months
anticardiolpin antibodies and antiphospholipid syndrome to HLA DR7 of anticoagulation with extended anticoagulation for a first episode of
in Mexican patients with SLE. Lupus, 6, 57–62. idiopathic venous thromboembolism. New England Journal of Medicine,
Gris, J.C., Bouvier, S., Molinari, N., Galanaud, J.P., Cochery-Nouvellon, E., 340, 901–7.
Mercier, E., Fabbro-Peray, P., Balducchi, J.P., Marès, P., Quéré, I., and Khamashta, M., Cervera, R., Asherson, R., et al. (1990). Association of anti-
Dauzat, M. (2012). Comparative incidence of a first thrombotic event bodies against phospholipids with heart valve disease in systemic lupus
in purely obstetric antiphospholipid syndrome with pregnancy loss: the erythematosus. Lancet, 335, 1541–2
NOH-APS observational study. Blood,119, 2624–32. Khamashta, M., Cuadrado, M., Mujic, F., et al. (1995). The management of
Hakim, A., Machin, S., and Isenberg, D. (1998). Autoimmune thrombocy- thrombosis in the antiphospholipid-antibody syndrome. New England
topenia in primary antiphospholipid syndrome and systemic lupus Journal of Medicine, 332, 993–7.
erythematosus: The response to splenectomy. Seminars in Arthritis and Khamashta, M., Gil, A., Asherson, R., Vazquez, J., and Hughes, G. (1989).
Rheumatism, 28, 20–5. Antiphospholipid antibodies, valvular heart disease, and systemic lupus
Hanly, J., Hong, C., Smith, S., and Fisk, J. (1999). A prospective analysis of erythematosus (Letter). American Journal of Medicine, 86, 633–4.
cognitive function and anticardiolipin antibodies in systemic lupus ery- Klein, K. and Pittelkow, M. (1992). Tissue plasminogen activator for treat-
thematosus. Arthritis and Rheumatism, 42, 728–34. ment of livedoid vasculitis. Mayo Clinic Proceedings, 67, 923–33.
Harris, E., Gharavi, A., Boey, M., et  al. (1983). Anticardiolipin antibod- Krnic-Barrie, S., O’Connor, C., Looney, S., Pierangeli, S., and Harris, N.
ies: detection by radioimmunoassay and association with thrombosis in (1997). A retrospective review of 61 patients with antiphospholipid syn-
SLE. Lancet, 2, 1211–14. drome. Analysis of factors influencing recurrent thrombosis. Archives of
Harris, E. and Spinnato, J. (1991). Should anticardiolipin tests be performed Internal Medicine, 157, 2101–8.
in otherwise healthy pregnant women? American Journal of Obstetrics Lampropoulos, C.E., Koutroumanidis, M., Reynolds, P.P., Manidakis, I.,
and Gynecology, 165, 1272–7. Hughes, G.R., and D’Cruz, D.P. (2005). Electroencephalography in the
Hasnie, A., Stoddard, M., Gleason, C., et al. (1995). Diastolic dysfunction is a assessment of neuropsychiatric manifestations in antiphospholipid syn-
feature of the antiphospholipid syndrome. American Heart Journal, 129, drome and systemic lupus erythematosus. Arthritis and Rheumatism,
1009–13. 52, 841–6.
Hasunuma, Y., Matsuura, E., Makita, Z., Katahira, T., Nishi, S., and Koike T. Landefeld, C.S. and Goldman, L. (1989). Major bleeding in outpatients
(1997). Involvement of beta 2-glycoprotein 1 and anticardiolipin anti- treated with warfarin: Incidence and prediction by factors known at the
bodies in oxidatively modified lowdensity lipoprotein uptake by mac- start of outpatient therapy. American Journal of Medicine, 87, 144–52.
rophages. Clinical and Experimental Immunology, 107, 569–73. Levine, S., Brey, R., Sawaya, K., et  al. (1995). Recurrent stroke and
Hojnik, M., George, J., Ziporen, L., and Shoenfeld, Y. (1996). Heart valve thrombo-occlusive events in the antiphospholipid syndrome. Annals of
involvement (Libman-Sacks endocarditis) in the antiphospholipid syn- Neurology, 38, 119–24.
drome. Circulation, 93, 1579–87. Libman, E. and Sacks, B. (1924). A hitherto undescribed form of valvular and
Horbach, D., Oort, E., Donders, R., Derksen, R., and de Groot, P. (1996). mural endocarditis. Archives of Internal Medicine, 33, 701–37.
Lupus anticoagulant is the strongest risk factor for both venous and Lie J.T. (1996). Vasculopathy of the antiphospholipid syndromes revisited:
arterial thrombosis in patients with systemic lupus erythematosus. thrombosis is the culprit and vasculitis the consort. Lupus, 5, 368–71.
Thrombosis and Haemostasis, 76, 916–24. Lim, W., Crowther, M.A., and Eikelboom, J.W. (2006). Management of
Houssiau, F.A., N’Zeusseu Toukap, A., Depresseux, G., Maldague, B.E., antiphospholipid antibody syndrome: a systematic review. Journal of the
Malghem, J., Devogelaer, J.P., and Vande Berg, B.C. (1998). Magnetic American Medical Association, 295, 1050–7.
resonance imaging-detected avascular osteonecrosis in systemic lupus Lima, F., Khamashta, M.A., Buchanan, N.M., Kerslake, S., Hunt, B.J., and Hughes,
erythematosus:  lack of correlation with antiphospholipid antibodies. G.R. (1996). A study of sixty pregnancies in patients with the antiphospho-
British Journal of Rheumatology, 37, 448–53. lipid syndrome. Clinical and Experimental Rheumatology, 14, 131–6.
Hughes, G., Harris, E., and Gharavi, A. (1987). The anticardiolipin syndrome. Liu, A. and Zhang, H. (2012). Detection of  antiphospholipid  antibody in
Journal of Rheumatology, 13, 486–9. children with Henoch-Schönlein purpura and central nervous system
Hughes, G.R. and Khamashta, M.A. (2003). Seronegative antiphospholipid involvement. Pediatric Neurology, 47, 167–70.
syndrome. Annals of the Rheumatic Diseases, 62, 1127. Lockshin, M. (1998). Pregnancy loss and antiphospholipid antibodies. Lupus,
Hughes, G.R.V. (1983). Thrombosis, abortion, cerebral disease and the lupus 7 (Suppl. 2), S86–9.
anticoagulant. British Medical Journal, 287, 1088–9. Lopez-Pedrera, C., Buendia, P., Cuadrado, M.J., Siendones, E., Aguirre, M.A.,
Ioannou, Y. (2012). The Michael Mason Prize:  Pathogenic antiphospho- Barbarroja, N., Montiel-Duarte, C., Torres, A., Khamashta, M., and
lipid antibodies, stressed out antigens and the deployment of decoys. Velasco, F. (2006). Antiphospholipid antibodies from patients with the
Rheumatology (Oxford), 51, 32–6. antiphospholipid syndrome induce monocyte tissue factor expression
Jordan, N.P., Chaib, A., Sangle, S., Tungekar, F., Sabharwal, T., Abbs, I., through the simultaneous activation of NF-kappaB/Rel proteins via the
Khamashta, M.A.,  and D'Cruz, D. (2013). Association of thrombotic p38 mitogen-activated protein kinase pathway, and of the MEK-1/ERK
microangiopathy and intimal hyperplasia with bleeding post renal pathway. Arthritis and Rheumatism, 54, 301–11.
biopsy in antiphospholipid antibody positive patients. Arthritis Care and Mascola, M.A. and Repke, J.T. (1997). Obstetric management of the high-risk
Research (Hoboken), in press. lupus pregnancy. Rheumatic Disease Clinics of North America, 23, 119–32.
Kalashnikova, L.A., Korczyn, A.D., Shavit, S., Rebrova, O., Reshetnyak, T., Matsuura, E., Igarasha, T., Fujimoto, M., Ichikawa, K., and Koike, T. (1990).
and Chapman, J. (1999). Antibodies to prothrombin in patients with Anticardiolipin cofactor(s) and differential diagnosis of autoimmune
Sneddon’s syndrome. Neurology, 53, 223–5. disease. Lancet, 336, 117–8.
Kalashnikova, L.A., Nasonov, E.L., Kushekbaeva, A.E., and Gracheva, L.A. McHugh, N., James, I., and Plant, G. (1990). Anticardilopin and antineu-
(1990). Anticardiolipin antibodies in Sneddon’s syndrome. Neurology, trophil antibodies in giant cell arteritis. Journal of Rheumatology, 17,
40, 464–7. 916–22.
CHAPTER 43 antiphospholipid syndrome 631

McNeil, H.P., Chesterman, C.N., and Krilis, S.A. (1989). Anticardiolipin and Pandolfi, C., Gianni, A., Fregoni, V., Nalli, G., and Faggi, L. (1997).
lupus anticoagulants comprise separate antibody subgroups with differ- Hypopituitarism and antiphospholipid syndrome. Minerva
ent phospholipid binding characteristics. British Journal of Haematology, Endocrinologica, 22, 103–5.
73, 506–12. Pangborn, M. (1942). Isolation and purification of a serologically active phos-
McNeil, H., Simpson, R., Chesterman, C., and Krilis, S. (1990). pholipid from beef heart. Journal of Biological Chemistry, 143, 247–56.
Antiphospholipid antibodies are directed against a complexed antigen Peddi, V. and Kant, K. (1995). Catastrophic secondary antiphospholipid
that includes a lipid binding inhibitor of coagulation: beta-2-glycoprotein syndrome with concomitant antithrombin III deficiency. Journal of the
1. Proceedings of the National Academy of Science USA, 87, 4120–4. American Society of Nephrology, 5, 1882–7.
Medina, G., Casaos, D., Jara, L.J., Vera-Lastra, O., Fuentes, M., Barile, L., Pengo, V., Ruffatti, A., Legnani, C., et al. (2011). Incidence of a first throm-
and Salas, M. (2003). Increased carotid artery intima-media thickness boembolic event in asymptomatic carriers of high-risk antiphospho-
may be associated with stroke in primary antiphospholipid syndrome. lipid antibody profile:  a multicenter prospective study. Blood, 118,
Annals of the Rheumatic Diseases, 62, 607–10. 4714–8.
Menon, S., Jameson-Shortall, E., Newman, S., Hall-Craggs, R., Chinn, R., Pengo, V., Tripodi, A., Reber, G., Rand, J.H., Ortel, T.L., Galli, M., De
and Isenberg, D. (1999). A longitudinal study of anticardiolipin anti- Groot, P.G.; Subcommittee on Lupus Anticoagulant/Antiphospholipid
body levels and cognitive functioning in sytemic lupus erythematosus. Antibody of the Scientific and Standardisation Committee of the
Arthritis and Rheumatism, 42, 735–41. International Society on Thrombosis and Haemostasis. (2009). Update
Merkel, P., Chang, Y., Pierangeli, S., Convery, K., Harris, N., and Polisson, R. of the guidelines for lupus anticoagulant detection. Subcommittee on
(1996). The prevalence and clinical association of anticardiolipin anti- Lupus Anticoagulant/Antiphospholipid Antibody of the Scientific and
bodies in a large inception cohort of patients with connective tissue dis- Standardisation Committee of the International Society on Thrombosis
eases. American Journal of Medicine, 101, 576–83. and Haemostasis. Journal of Thrombosis and Haemostasis, 7, 1737–40.
Merkel, P.A., Lo, G.H., Holbrook, J.T., et al. (2005). Brief communication: high Pérez-Vázquez, M.E., Villa, A.R., Drenkard, C., Cabiedes, J., and
incidence of venous thrombotic events among patients with Wegener Alarcón-Segovia, D. (1993). Influence of disease duration, continued
granulomatosis:  the Wegener’s Clinical Occurrence of Thrombosis followup and further antiphospholipid testing on the frequency and
(WeCLOT) Study. Annals of International Medicine, 142, 620–6. classification category of antiphospholipid syndrome in a cohort of
Meroni, P.L., Raschi, E., Grossi, C., Pregnolato, F., Trespidi, L., Acaia, B., and patients with systemic lupus erythematosus. Journal of Rheumatology,
Borghi, M.O. (2012). Obstetric and vascular APS: same autoantibodies 20, 437–42.
but different diseases? Lupus, 21, 708–10. Petri, M. (1997). Pathogenesis and treatment of APS. Medical Clinics of North
Merrill, J., Shen, C., Gugnani, M., Lahita, R., and Mongey, A. (1997). High America, 81, 151–61.
prevalence of antiphospholipid antibodies in patients taking procaina- Pierangeli, S., Colden-Stanfield, M., Liu, X., Barker, J., Anderson, G., and
mide. Journal of Rheumatology, 24, 1083–8. Harris, N. (1999). Antiphospholipid antibodies from antiphospho-
Miyakis, S., Lockshin, M.D., Atsumi, T., Branch, D.W., Brey, R.L., Cervera, R., lipid syndrome patients activate endothelial cell in vitro and in vivo.
et al. (2006). International consensus statement on an update of the clas- Circulation, 99, 1997–2002.
sification criteria for definite antiphospholipid syndrome (APS). Journal Pierangeli, S.S., Chen, P.P., and Gonzalez, E.B. (2006). Antiphospholipid anti-
of Thrombosis and Haemostasis, 4, 295–306. bodies and the antiphospholipid syndrome: an update on treatment and
Molina, J.F., Gutierrez-Urena, S., Molina, J., et al. (1997). Variability of anti- pathogenic mechanisms. Current Opinion in Hematology, 13, 366–75.
cardiolipin antibody isotype distribution in 3 geographic populations of Piette, J. (1998). Criteria for the antiphosholipid syndrome. Lupus, 7
patients with systemic lupus erythematosus. Journal of Rheumatology, (Suppl. 2), S149–57.
24, 291–6. Piette, J. and Cacoub, P. (1998). Antiphospholipid syndrome in the
Moore, J. and Mohr, C. (1952). Biologically false positive serological tests elderly: Caution. Circulation, 97, 2195–6.
for syphilis. Type, incidence and cause. Journal of the American Medical Piette, J., Weschler, B., Frances, C., Papo, T., and Godeau, P. (1993).
Association, 150, 467–73. Exclusion criteria for primary antiphospholipid syndrome. Journal of
Morgan, R. and Feneley, C. (1994). Renal vein thrombosis caused by primary Rheumatology, 20, 1802–4.
antiphospholipid syndrome. British Journal of Urology, 74, 807–8. Pisoni, C.N., Cuadrado, M.J., Khamashta, M.A., and Hunt, B.J. (2006).
Mujic, F., Cuadrado, M., Lloyd, M., et al. (1995). Primary antiphospholipid Treatment of menorrhagia associated with oral anticoagulation: efficacy
syndrome evolving into SLE. Journal of Rheumatology, 22, 1589–92. and safety of the levonorgestrel releasing intrauterine device (Mirena
Nagasawa, K, Ishii, T., Mayumi, T., et al. (1989). Avascular necrosis of bone in coil). Lupus, 15, 877–80.
systemic lupus erythematosus: Possible role of haemostatic abnormali- Provenzale, J., Barboriak, D., Allen, N., and Ortel, T. (1998). Antiphospholipid
ties. Annals of the Rheumatic Diseases, 48, 672–6. antibodies: findings at arteriography. American Journal of Neuroradiology,
Nencini, P., Baruffi, M., Abbate, R., Massai, G., Amaducci, L., and Inzitari, 19, 611–6.
D. (1992). Lupus anticoagulant and anticardiolipin antibodies in young Rai, R.S., Regan, L., Clifford, K., et al. (1995). Antiphospholipid antibodies and
adults with cerebral ischaemia. Stroke, 23, 189–93. beta 2-glycoprotein I in 500 women with recurrent miscarriage: results
Nilsson, I., Astedt, B., Hedner, U., and Berezin, D. (1975). Intrauterine of a comprehensive screening approach. Human Reproduction, 10,
death and circulating ‘antithromboplastin’. Acta Medica Scandinavica, 2001–5.
197, 153–9. Rees, J.D., Lanca, S., Marques, P.V., Gomez-Puerta, J.A., Moco, R., Oliveri, C.,
Norden, D., Ostrov, B., Shafritz, A., and Von Feldt, J. (1995). Vasculitis Khamashta, M.A., Hughes, G.R., and D’Cruz, D.P. (2006). Prevalence of
associated with antiphospholipid syndrome. Seminars in Arthritis and the antiphospholipid syndrome in primary systemic vasculitis. Annals of
Rheumatism, 24, 273–81. the Rheumatic Diseases, 65, 109–11.
Oosting, J., Derkson, R., Bobbink, I., Hackeng, T., Bouma, B., and de Groot, P. Reggia, R., Ziglioli, T., Andreoli, L., Bellisai, F., Iuliano, A., Gerosa, M.,
(1993). Antiphopsholipid antibodies directed against a combination of Ramoni, V., Tani, C., Brucato, A., Galeazzi, M., Mosca, M., Caporali,
phospholipids with prothrombin, protein C, or protein S: an explanation R., Meroni, P.L., and Tincani, A. (2012). Primary anti-phospholipid syn-
for their pathogenic mechanism? Blood, 81, 2618–25. drome: any role for serum complement levels in predicting pregnancy
Ostuni, P., Lazzarin, P., Pengo, V., Ruffati, A., Schiavon, F., and Gambari, P. complications? Rheumatology (Oxford), 51, 2186–90.
(1990). Renal artery thrombosis and hypertension in a 13-year-old girl Reid, K., Bentley, D., and Campbell, R. (1986). Complement system proteins
with antiphospholipid syndrome. Annals of the Rheumatic Diseases, which interact with C3b or C4b: A super-family of structurally related
49, 184–7. proteins. Immunology Today, 7, 230–4.
632 SECTION 6 mimickers of vasculitis

Reverter, J., Tassies, D., Font, J., et al. (1996). Hypercoaguable state in patients Shah, N., Khamashta, M., Atsumi, T., and Hughes, G. (1998). Outcome
with antiphospholipid syndrome is related to high induced tissue fac- of patients with anticardiolpin antibodies:  a 10  year follow up of 52
tor expression on monocytes and to low free protein S. Arteriosclerosis, patients. Lupus, 7, 3–6.
Thrombosis and Vascular Biology, 16, 1319–26. Shovman, O., Langevitz, P., Gilburd, B., and Shoenfeld, Y. (2013). Coincidence
Rocca, P., Siegel, L., and Cupps, T. (1994). The concomitant expression of vas- of granulomatosis and polyangiitis with atypical clinical manifestation
culitis and coagulopathy: Synergy for marked tissue ischaemia. Journal and antiphospholipid syndrome. Lupus, 22, 320–3.
of Rheumatology, 21, 556–60. Silver, R., Draper, M., Scoff, J., Lyon, J., Reading, J., and Branch, D. (1994).
Rodriguez-Garcia, J.L., Bertolaccini, M.L., Cuadrado, M.J., Sanna, G., Clinical consequences of antiphospholipid antibodies. An historic
Ateka-Barrutia, O., and  Khamashta, M.A. (2012). Clinical manifesta- cohort study. Obstetrics and Gynaecology, 83, 372–7.
tions of antiphospholipid syndrome (APS) with and without antiphos- Simantov, R., LaSala, J., Lo, S., et al. (1995). Activation of cultured vascu-
pholipid antibodies (the so-called ‘seronegative APS’). Annals of the lar endothelial cells by antiphospholipid antibodies. Journal of Clinical
Rheumatic Diseases, 71, 242–4. Investigation, 96, 2211–19.
Rosove, M. and Brewer, P. (1992). Antiphospholipid thrombosis:  Clinical Simchen, M.J., Dulitzki, M., Rofe, G., Shani, H., Langevitz, P., Schiff, E., and
course after the first thrombotic event in 70 patients. Annals of Internal Pauzner, R. (2011). High positive antibody titers and adverse  preg-
Medicine, 117, 303–8. nancy  outcome in women with antiphospholipid syndrome. Acta
Ruiz-Irastorza, G., Cuadrado, M.J., Ruiz-Arruza, I., et  al. (2011). Obstetricia et Gynecologica Scandinavica, 90, 1428–33.
Evidence-based recommendations for the prevention and long-term Simioni, P., Prandoni, P., Zanon, B., et al. (1996). Deep venous thrombosis
management of thrombosis in antiphospholipid antibody-positive and lupus anticoagulant. Thrombosis and Haemostasis, 76, 187–9.
patients:  report of a task force at the 13th International Congress on Sipek-Dolnicar, A., Hojnik, M., Bozic, B., et al. (2002). Clinical presentations
antiphospholipid antibodies. Lupus, 20, 206–18. and vascular histopathology in autopsied patients with systemic lupus
Sacré, K., Brihaye, B., Hyafil, F., Serfaty, J.M., Escoubet, B., Zennaro, M.C., erythematosus an anticardiolipin antibodies. Clinical and Experimental
Lidove, O., Laissy, J.P., and Papo, T. (2010). Asymptomatic myocardial Rheumatology, 20, 335–42.
ischemic disease in antiphospholipid syndrome: a controlled cardiacmag- Somers, E., Magder, L.S., and Petri, M. (2002). Antiphospholipid anti-
netic resonance imaging study. Arthritis and Rheumatism, 62, 2093–100.  bodies and incidence of venous thrombosis in a cohort of patients
Sailer, M., Burchert, W., Smid, H., et al. (1997). Positron emission tomog- with systemic lupus erythematosus. Journal of Rheumatology, 29,
raphy and magnetic resonance imaging for cerebral involvement in 2531–6.
patients with SLE. Journal of Neurology, 244, 186–93. Soweid, A., Hajjar, R., Hewan-Lowe, K., and Gonzalez, E. (1995). Skin
Sangle, S.R., D’Cruz, D.P., Jan, W., Karim, M.Y., Khamashta, M.A., Abbs, necrosis indicating antiphospholipid syndrome in patients with AIDS.
I.C., and Hughes, G.R. (2003). Renal artery stenosis in the antiphospho- Southern Medical Journal, 88, 786–8.
lipid (Hughes) syndrome and hypertension. Annals of the Rheumatic Straaton, K., Chatham, W., Smith, S., and Koopman, W. (1989). Valvular
Diseases, 62, 999–1002. heart disease in systemic lupus erythematosus. New England Journal of
Sangle, S., D’Cruz, D.P., Khamashta, M.A., and Hughes, G.R. (2004). Medicine, 320, 740.
Antiphospholipid antibodies, systemic lupus erythematosus, and Steinkasserer, A., Cockburn, D., Black, D., Boyd, Y., Solomon, E., and Sim, R.
non-traumatic metatarsal fractures. Annals of the Rheumatic Diseases, (1992). Assignment of apolipoprotein H to human chromosome 17q23
63, 1241–3. qter; determination of the major expression site. Cytogenetics and Cell
Sangle, S., D’Cruz, D.P., and Hughes, G.R. (2005a). Livedo reticularis and Genetics, 60, 31–3.
pregnancy morbidity in patients negative for antiphospholipid antibod- Tektonidou, M.G., Malagari, K., Vlachoyiannopoulos, P.G., Kelekis, D.A.,
ies. Annals of the Rheumatic Diseases, 64, 147–8. and Moutsopoulos, H.M. (2003). Asymptomatic avascular necrosis in
Sangle, S.R., D’Cruz, D.P., Abbs, I.C., Khamashta, M.A., and Hughes, G.R. patients with primary antiphospholipid syndrome in the absence of
(2005b). Renal artery stenosis in hypertensive patients with antiphos- corticosteroid use: a prospective study by magnetic resonance imaging.
pholipid (Hughes) syndrome:  outcome following anticoagulation. Arthritis and Rheumatism, 48, 732–6.
Rheumatology (Oxford), 44, 372–7. Tektonidou, M.G., Sotsiou, F., Nakopoulou, L., Vlachoyiannopoulos, P.G.,
Sangle, S.R., Jan, W., Lau, I.S., Bennett, A.N., Hughes, G.R., and D’Cruz, and Moutsopoulos, H.M. (2004). Antiphospholipid syndrome nephrop-
D.P. (2006). Coeliac artery stenosis and antiphospholipid (Hughes) athy in patients with systemic lupus erythematosus and antiphos-
syndrome/antiphospholipid anti-bodies. Clinical and Experimental pholipid antibodies:  prevalence, clinical associations, and long-term
Rheumatology, 24, 349. outcome. Arthritis and Rheumatism, 50, 2569–79.
Schulman, S., Svenungsson, E., Granqvist, S., and the Duration of Tenedios, F., Erkan, D., and Lockshin, M.D. (2006). Cardiac manifestations
Anticoagulation Study Group (1998). Anticardiolipin antibodies predict in the antiphospholipid syndrome. Rheumatic Disease Clinics of North
early recurrence of thromboembolism and death among patients with America, 32, 491–507.
venous thromboembolism following anticoagulation therapy. American Tietjen, G., Day, M., Norris, L., et al. (1998). Role of anticardiolipin antibod-
Journal of Medicine, 104, 332–8. ies in young persons with migraine and transient focal neurological
Schutt, M., Kluter, H., Hagedorn-Greiwe, M., Fehm, H., and Wiedemann, events. Neurology, 50, 1433–40.
G. (1998). Familial co-existence of primary antiphospholipid syndrome Tincani, A., Balestrieri, G., Spatola, L., Cinquini, M., Meroni, P., and Roubey,
and Factor V Leiden. Lupus, 7, 176–82. R. (1998). Anticardiolipin and anti-beta 2glycoprotein 1 immuno-
Schved, J., Dupuy-Fons, C., Biron, C., Quere, I., and Janbon, C. (1994). A assays in the diagnosis of antiphospholipid syndrome. Clinical and
prospective epidemiological study on the occurrence of antiphos- Experimental Rheumatology, 16, 396–402.
pholipid antibody:  The Montpellier Antiphospholipid (MAP) Study. Toubi, E., Krause, I., Fraser, A., Lev, S., Stojanovich, L., Rovensky, J., Blank,
Haemostasis, 24, 175–82. M., and Shoenfeld, Y. (2005). Livedo reticularis is a marker for predict-
Sebastiani, G., Galeazzi, M., Morozzi, G., and Marcolongo, R. (1996). The ing multi-system thrombosis in antiphospholipid syndrome. Clinical
immunogenetics of the antiphospholipid syndrome, anticardiolipin anti- and Experimental Rheumatology, 23, 499–504.
bodies, and lupus anticoagulant. Seminars in Arthritis and Rheumatism, Tripplett, D. (1995). Protean clinical presentation of antiphospholipid
25, 414–20. -protein antibodies. Thrombosis and Haemostasis, 74, 329–37.
Seisdedos, L., Munos-Rodriguez, F., Cervera, R., Font, J., and Ingelmo, M. Urbanus, R.T., Siegerink, B., Roest, M., et al. (2009). Antiphospholipid anti-
(1997). Primary antiphospholipid syndrome evolving into SLE. Lupus, bodies and risk of myocardial infarction and ischaemic stroke in young
6, 285–6.
CHAPTER 43 antiphospholipid syndrome 633

women in the RATIO study: a case-control study. Lancet Neurology, 8, with antiphospholipid antibodies without underlying autoimmune dis-
998–1005. ease or previous thrombosis. Lupus, 7, 15–22.
Vaarala, O. (1997). Atherosclerosis in SLE and Hughes’ syndrome. Lupus, 6, Wasserman, A., Neisser, A., and Bruck, C. (1906). Eine Serodiagnostiche
489–90. reaktion bei Syphilis. Deutsche Medizinische Wochenschrift,
Vaarala, O. (1998). Antiphospholipid antibodies and myocardial infarction. 32, 745–6.
Lupus, 7 (Suppl. 2), S132–4. Watts, M., Greaves, M., Clearkin, L., et al. (1990). Antiphospholipid antibod-
Vaarala, O., Männttäri, M., Manninen, V., et al. (1995). Anti-cardiolipin anti- ies and ischaemic optic neuropathy. Lancet, 335, 613–4.
bodies and risk of myocardial infarction in a prospective cohort of mid- Wilson, W., Gharavi, A., Koike, T., et al. (1999). International consensus state-
dle aged men. Circulation, 91, 23–7. ment on preliminary classification for definite antiphospholipid syn-
Vaidya, S., Wang, C., Gugliuzza, C., and Fish, J. (1998). Relative risk of drome. Report of an International Workshop. Arthritis and Rheumatism,
post-transplant renal thrombosis in patients with antiphospholpid anti- 42, 1309–11.
bodies. Clinical Transplantation, 12, 439–44. Yokoi, K., Hosoi, E., Akaike, M., Shigekiyo, T., and Saito, S. (1996). Takayasu’s
Vianna, J., Khamashta, M., Ordi-Ros, J., et al. (1994). Comparison of the pri- arteritis associated with antiphospholipid antibodies. Report of two
mary and secondary antiphospholipid syndrome: A European multicen- cases. Angiology, 47, 315–9.
tre study of 114 patients. American Journal of Medicine, 96, 3–9. Zuily, S., Regnault, V., Selton-Suty, C., Eschwège, V., Bruntz, J.F., Bode-Dotto,
Wahl, D., Guillemin, F., de Maistre, B., Perret, C., Lecompte, T., and Thibaut, G. E., De Maistre, E., Dotto, P., Perret-Guillaume, C., Lecompte, T., and
(1997). Risk for venous thrombosis related to antiphospholipid antibod- Wahl, D. (2011). Increased risk for heart valve disease associated with
ies in systemic lupus erythematosus—A meta analysis. Lupus, 6, 467–73. antiphospholipid antibodies in patients with systemic lupus erythe-
Guillemin, F., de Maistre, B., Perret-Guillaume, C., Lecompte, T., and Thibaut, matosus: meta-analysis of echocardiographic studies. Circulation, 124,
G. (1998). Meta-analysis of the risk of venous thrombosis in individuals 215–24.
CHAPTER 44

Imitators of vasculitis
Sharon A. Chung and Kenneth E. Sack

Introduction latency of 20  years (Lie 1992a). Associated illnesses and the site
of the biopsy may also affect the histopathological picture. Thus,
To most physicians, ‘vasculitis’ connotes an immune-mediated dis- acute arterial hypertension may cause fibrinoid changes in vessel
ease. Yet, strictly speaking, the term simply means inflammation walls, usually without cellular infiltration. In areas where stasis
of the vessel wall, regardless of the cause. Confusion arises when occurs, such as the lower leg, a mild degree of perivascular cel-
non-inflammatory vascular damage produces the same signs and lular infiltration and vascular hypertrophy are the rule (Ryan and
symptoms as those resulting from true vasculitis (Table 44.1). For Wilkinson 1986).
these reasons, defining imitators of vasculitis becomes somewhat
arbitrary. In this chapter, we describe conditions of both vascular
and non-vascular aetiology that foster the mistaken diagnosis of
Table 44.1 Non-inflammatory causes of vascular damage
immune-mediated vasculitis and promote inappropriate use of
immunosuppressive agents.
Occlusive processes Atheroembolic disease

Mechanisms of vascular damage Thrombotic disorders

The initial cause of vascular injury is often obscure because dif- Antiphospholipid antibody syndrome
ferent stimuli can produce identical responses in the vessel wall. Thrombotic thrombocytopenic purpura
Non-immunological causes of vascular injury include infection Sickle cell anaemia
(Blanco et  al. 1999; Golden et  al. 1994; Hogarth et  al. 1999; Lie
1996; Pandey and LeRoy 1998; Somer and Finegold 1995; Walker Thromboembolism
and Mattern 1980), neoplasia (Fredericks et al. 1991b; Kao et al. Abnormal proteins
1992; Lie 1992b; Sheibani et al. 1986; Sienknecht 1995; Wick et al. Cryoglobulins
1986), ischaemia, congenital or inherited abnormalities (Imahori
et  al. 1969), and various forms of external (Martens et  al. 1996; Cryofibrinogens
Sijpkens et al. 1997; Nance et al. 1997; Olivero 1997; Pineda et al. Paraproteins
1985; Shim 1998) and internal (Ryan and Wilkinson 1986) injury Neoplasia Cardiac myxoma
to the vessel wall.
Other neoplasms

Pitfalls of angiography External injury Exposure to cold


Radiation exposure
Irregularities in vessel walls, as well as segmental vascular occlu-
sions and dilatations, are the commonly recognized angiographic Internal injury Hypertension, arterial dissection
manifestations of vasculitis (see Chapter 17). These same findings, Biochemical abnormalities
however, occur in a host of other conditions (Table 44.2).
Infection (See Table 44.3)

Pitfalls of histology Congenital or inherited


abnormalities
Pseudoxanthoma elasticum

Histopathological examination of vascular tissue does not always


Ehlers–Danlos syndrome
yield definitive information. A  vessel damaged by any mecha-
nism may accumulate immune complexes or fibrin; inflamma- Neurofibromatosis
tion in neighbouring tissues occasionally triggers an inflammatory Fibromuscular dysplasia
response in the vessel wall (Ryan and Wilkinson 1986). Furthermore,
Miscellaneous conditions Drug effects
the microscopic findings are a function of the type of injury and
the time between the original insult and the obtaining of tissue. Moyamoya disease
Anoxia, for example, can damage vascular smooth muscle within Others (see Table 44.4)
2 hours (Ryan and Wilkinson 1986). Conversely, radiation injury
Non-vascular
may cause atherosclerotic changes and periarterial fibrosis after a
636 SECTION 6 mimickers of vasculitis

Table 44.2 Angiographic imitators of vasculitis 1995; Darsee 1979; Young et  al. 1986), abdominal pain with or
without gastrointestinal bleeding (Darsee 1979; Hendel et al. 1989;
Imitator Authors Moolenaar and Lamers 1996), angina pectoris (Cappiello et  al.
1989), myocardial infarction (Darsee 1979), transient ischaemic
Amyloidosis Salvarani et al. 1994
attacks (Cappiello et al. 1989; Stanton and Nickeleit 1996; Young
Atheromatous emboli Cappiello et al. 1989 et  al. 1986), and other stroke syndromes (Cappiello et  al. 1989;
Atrial myxoma New et al. 1970; Thomas 1981 Sijpkens et al. 1997; Winter 1957; Young et al. 1986).
Laboratory abnormalities commonly seen in atheroembolic dis-
Cold exposure Jacob et al. 1986
ease include hypocomplementaemia, because crystalline choles-
Drug abuse Rumbaugh et al. 1971a terol and lipids from atheromata can fix complement. This process
Ehlers–Danlos syndrome Imahori et al. 1969 also results in the production of C5a, a potent chemotactic factor
for eosinophils and neutrophils (Hammerschmidt et al. 1981; Cosio
Fibromuscular dysplasia Meyers et al. 1974
et  al. 1985). Thus, eosinophilia and eosinophiluria are also fre-
Hypertension Garner et al. 1990 quently seen (Lye et al. 1993; Cosio et al. 1985; Cappiello et al. 1989;
Infection Marks and Kuskov 1995 Kasinath and Lewis 1987; Fine et al. 1987). Other laboratory abnor-
malities that accompany atheroembolic disease are similar to those
Migraine Masuzawa et al. 1983; Serdaru et al. 1984
seen in immune-mediated conditions:  markers of inflammation
Moyamoya Provost et al. 1991; Ueki et al. 1994 (anaemia, leukocytosis, elevated erythrocyte sedimentation rate);
Neurofibromatosis Finley and Dabbs 1988; Tomsick et al. 1976 azotaemia; thrombocytopenia; hyperamylasaemia; positive antinu-
clear antibody or rheumatoid factor; proteinuria; and granular or
Neoplasia Fredericks 1991b; Leeds and Rosenblatt 1972
hyaline urinary casts (Cappiello et al. 1989; Goldman et al. 1980;
Pseudoxanthoma elasticum Travers et al. 1979 Young et al. 1986; Fine et al. 1987; Stanton and Nickeleit 1996).
Radiation exposure McCready et al. 1983 While biopsy of the skin lesion (Falanga et al. 1986) or kidney
Thrombotic Orbison 1952
(Gupta et al. 1993; Smith et al. 1981) can yield the diagnosis in the
thrombocytopenic purpura appropriate setting, biopsy of other organs such as muscle or bone
marrow can also aid in diagnosis (Anderson 1965; Pierce et  al.
Trauma Savader et al. 1988; Suwanwela and 1978). Usual methods for preparing histological specimens dis-
Suwanwela 1972
solve cholesterol, leaving characteristic clefts (Figure 44.1). Of note,
an acute inflammatory response resulting in intimal hyperplasia
and panarteritis with foreign body giant cells can develop after the
Representative syndromes deposition of atheromatous material in the arterial lumen (Leeds
and Rosenblatt 1972; Cross 1991; Retan and Miller 1966). In cases
Occlusive processes where necrotizing angiitis is the dominant pathological finding, the
Atheroembolic disease cholesterol clefts may go unnoticed (Sijpkens et al. 1997; Anderson
Atheroembolic disease results from the occlusion of distal arteries 1965; Anderson and Richards 1968).
by atheromatous debris, including fibrin, platelets, cholesterol crys- Prognosis for recovery from atheroembolic disease is poor
tals, and calcium fragments (Kennedy et al. 1989; Eliot et al. 1964). (Dahlberg et  al. 1989; Gupta et  al. 1993; Lye et  al. 1993). The
This process is suggested by the clinical triad of a precipitating mortality is 70–90% in some series (Moolenaar and Lamers
event, acute or subacute renal failure, and skin lesions. Precipitating 1996; Hendel et  al. 1989; Falanga et  al. 1986; Fine et  al. 1987),
events are generally vascular procedures such as cardiac catheteri- but 20–30% of patients can have partial renal recovery even after
zation and anticoagulation (Scolari and Ravani 2010; Fukumoto some period of dialysis support (Scolari and Ravani 2010; Smith
et al. 2003). Anticoagulation may predispose to atheroemboli by
preventing the formation of a ‘protective’ thrombus over an ather-
omatous plaque (Nevelsteen et al. 1992; Moldveen-Geronimus and
Merriam 1967; Hyman et al. 1987). When there is no obvious trig-
gering event, other signs and symptoms of atherosclerotic disease
or an aortic aneurysm are usually present (Stanton and Nickeleit
1996; Kalter et al. 1985; Smith et al. 1981; Fine et al. 1987).
The most frequent, visible manifestation of atheroembolic dis-
ease is purple discoloration of the toes, occasionally followed by
ulceration and gangrene. Peripheral pulses are usually normal,
and the patient often has livedo reticularis symmetrically affecting
the lower body (Anderson and Richards 1968; Falanga et al. 1986;
Kalter et al. 1985). Unexplained acute or progressive renal failure
(Bradley 1995; Gupta et al. 1993; Lye et al. 1993; Smith et al. 1981;
Spring et al. 1998; Thadhani et al. 1995) or hypertension (Bradley
1995; Dahlberg et al. 1989) should also suggest atheroembolic dis-
ease. Additional clinical conditions caused by atheroemboli include
acute pancreatitis (Orvar and Johlin 1994), amaurosis (Bradley Fig. 44.1 Atheromatous embolus lodged in an afferent renal arteriole.
CHAPTER 44 imitators of vasculitis 637

et  al. 1981). Survival probably relates to the cause and extent of material ordinarily comes from the heart (in situ thrombi, marantic
the disease (Smith et al. 1981). Thus, a patient with limited emboli valvular lesions) or a large artery (Halasz and Strauss 1998), but
from a discrete source, such as an aneurysm or localized plaque, venous thrombi occasionally embolize paradoxically through a pat-
may do well upon removal of the offending lesion (Darsee 1979). ent foramen ovale (Karl 1975). Rarely, the emboli consist of air, fat,
Glucocorticoids can temporarily alleviate some of the disease or neoplastic tissue (Lee and Hodes 1967).
manifestations but do not affect the outcome (Hendel et al. 1989). In addition to vasculitis, embolic disease is one of the many
Use of low-molecular-weight dextran, antiplatelet drugs, or vaso- causes of ‘blue toe syndrome’ (the sudden onset of discrete painful,
dilators does not alter the course substantially (Hendel et al. 1989; blue or purple discolorations on the foot or toes). Other conditions
Kalter et al. 1985; Cappiello et al. 1989). Agents of potential ben- associated with this phenomenon include de novo thrombi, various
efit include pentoxifylline (Carr et al. 1994), lipid-lowering drugs infections, cyanotic heart disease, hyperviscosity, calciphylaxis, and
(Woolfson and Lachmann 1998; Kawakami et al. 1990), and drugs phaeochromocytoma (Abdelmalek and Spittell 1995; Federman
with the ability to stabilize atherosclerotic plaques (Kullo et  al. et al. 1994; Nevelsteen et al. 1992; O’Keeffe et al. 1992).
1998). If anticoagulants precipitate the syndrome, the patient may
Abnormal proteins
improve after discontinuing them (Bruns et al. 1978).
Materials other than atheromatous or thrombotic debris can
occlude vessels and simulate vasculitis. Temperature may play an
Thrombotic disorders important role in this process—cryoglobulins and cryofibrinogens
Thrombosis in multiple vessels can masquerade as vasculitis, and precipitate in cold temperature and occlude blood vessels (Korst
when inflammation and thrombosis occur together in a vessel, and Kratochvil 1955; Waxman and Dove 1969), causing cutane-
determining which came first can be difficult. The antiphospholipid ous ulcerations and acral purpura (Sack 1993; Stoane et al. 1966;
antibody (APL) syndrome is the most common hypercoagulable Schwarz et  al. 1972). Cryofibrinogens occur most commonly in
state that simulates vasculitis (see Chapter 43). Of the many skin patients with neoplasia or diabetes mellitus (Kirsner et  al. 1993;
manifestations associated with APL syndrome, livedo reticula- Smith and Arkin 1972). The characteristic histopathological feature
ris, often accompanied by acrocyanosis, is the most characteristic is eosinophilic thrombi in dermal vessels associated with minimal
(Alarcon-Segovia et al. 1997; Ingram et al. 1987; Naldi et al. 1993; signs of inflammation (Beightler et al. 1991; Burruss et al. 1994).
Sammaritano et al. 1990; Hughes 1993; Alegre et al. 1989; Bowles Rarely, whole immunoglobulins or light chains can form occlusive
1990; Smith et al. 1990; Stephens 1991), and is associated with arte- crystals at cool temperatures giving rise to polyarthralgia, palpable
rial events (Frances et al. 2005). Other cutaneous manifestations purpura, necrotic cutaneous ulcers, nerve palsies, or combinations
include small, non-blanching, erythematous, or cyanotic areas on thereof (Grossman et al. 1972; Dotten et al. 1976; Stone et al. 1989).
the hands and feet, as well as haemorrhages, ulcers, and gangrene Non-cryoprecipitable paraproteins, such as those occurring in
(Hughes 1993; Alegre et  al. 1989; Bowles 1990; Stephens 1991; multiple myeloma, may cause an occlusive vasculopathy (Dornan
Grob et al. 1991; Frances et al. 2005). Histopathological examina- et  al. 1985). A  similar vasculopathy also appears in a condition
tion of the skin occasionally shows inflammation of small vessels termed POEMS (polyneuropathy, organomegaly, endocrinopathy,
(Naldi et al. 1993; Goldberger et al. 1992), but thrombosis is proba- M protein, and skin changes) (Lesprit 1996; Bardwick et al. 1980;
bly the primary event (Dessailloud et al. 1998; Fessler 1997; Ingram Viard et al. 1988; Manning et al. 1992; Soubrier et al. 1994). The
et al. 1987; Bowles 1990; Smith et al. 1990; Stephens 1991; Lie 1994; cardinal feature of POEMS is severe, progressive sensorimotor
Gertner and Lie 1994). The antibodies in this syndrome occasion- polyneuropathy. Other common findings are plasma cell dyscrasia,
ally induce widespread thrombosis, causing ischaemia in single in association with osteosclerotic bone lesions, production of an
(Dessailloud et al. 1998; Gertner 1999; Marie et al. 1997; Sneddon M protein, hepatosplenomegaly, lymphadenopathy, thickening and
1965) or multiple (Asherson et al. 1998) organs. In these instances, hyperpigmentation of the skin, and endocrine dysfunction (diabe-
angiograms may demonstrate occlusions or narrowing of involved tes mellitus, hypothyroidism, adrenal insufficiency, amenorrhea,
vessels (Roberts et al. 1994). gynaecomastia, or impotence).
A pentad of microangiopathic haemolytic anaemia, thrombo-
cytopenic purpura, neurological abnormalities, renal dysfunction, Neoplasia
and fever is characteristic of thrombotic thrombocytopenic pur-
Cardiac myxoma
pura (Fox et al. 1986; Jain et al. 1994). Histopathological examina-
tion of tissues such as gingiva or kidney shows hyaline thrombi, Myxomas are the most common type of cardiac neoplasms. These
microaneurysm formation, and endothelial cell proliferation in tumours usually arise as a single, pedunculated, friable mass rang-
small arteries and arterioles (Orbison 1952; Fox et  al. 1986; Jain ing from a few millimetres to greater than 10 cm in diameter (Wold
et al. 1994). Similar changes occur when injury to vascular endothe- and Lie 1980; Markel et al. 1987). Cardiac myxomas typically occur
lium induces disseminated intravascular coagulation (Gilbert and in the left atrium (Markel et al. 1987; Tazelaar et al. 1992), most often
Scalzi 1993). attached to the septum near the fossa ovalis (Reynen 1995; Markel
Sickle cell anaemia occasionally gives rise to vascular narrowing et al. 1987; Greenwood 1968). Occasionally, these tumours appear
(Liebeskind et al. 1973; Calabrese et al. 1992). Thrombocythaemia in the right atrium, and about 5% involve the ventricles (Markel
can cause digital gangrene and livedo reticularis without demon- et  al. 1987). Cardiac myxomas usually affect patients between
strable arterial disease (Singh and Wetherley-Mein 1977). the ages of 30 and 60  years, and are more common in women.
Approximately 10% of cardiac myxomas are familial (Siltanen et al.
Thromboembolism 1976; Liebler et al. 1976), and can be seen in hereditary conditions
Thromboembolism, by occluding multiple arterial beds, can create such as the Carney complex. This autosomal dominant condition,
the clinical picture of vasculitis (Sprabery et al. 1994). The embolic linked to genetic mutations in the PRKAR1α gene, is characterized
638 SECTION 6 mimickers of vasculitis

by recurrent familial cardiac (and other organ) myxomas, spotty echocardiogram shows not only its size, shape, and location, but
skin pigmentation, and endocrine hyperactivity and neoplasms also estimates its mobility (Markel et al. 1987; Alam and Sun 1991).
(Casey et al. 2000; Shetty Roy et al. 2011; Carney et al. 1985). Computed tomography (CT) and magnetic resonance imaging
Myxomas characteristically cause systemic, obstructive, and (MRI) can detect myxomas 0.5 cm or greater in diameter (Reynen
embolic symptoms (Reynen 1995; Greenwood 1968; Markel et al. 1995). The angiographic findings of myxomatous emboli (vascular
1987; Nasser et al. 1972; St. John Sutton et al. 1980). Some patients irregularities, dilatations, or aneurysms) (Thomas 1981; New et al.
have fever, weight loss, arthralgias, myalgias, or Raynaud’s phenom- 1970; Markel et al. 1987; Boussen et al. 1991; Stoane et al. 1966;
enon (Greenwood 1968; Reynen 1995; Thomas 1981; Wold and Lie Schwarz et al. 1972; Leonhardt and Kullenberg 1977) can also lead
1980; Markel et al. 1987; Nasser et al. 1972; St. John Sutton et al. one to incorrectly diagnose a systemic vasculitis.
1980; Fitzpatrick et al. 1986; Kaminsky et al. 1979; Boussen et al. Microscopic examination of myxoma tissue shows a paucicellu-
1991). In addition, a skin rash may appear that ranges from ery- lar collection of polygonal and stellate cells in an amorphous muco-
thematous (or livedoid) macules or papules, to frank ulcerations or polysaccharide matrix with variable vascularity (Markel et al. 1987;
telangiectasias (Reynen 1995; Navarro et al. 1995; Gravallese et al. Tazelaar et al. 1992). The cells contain few mitotic figures, but the
1995; Byrd et al. 1980; Bridges and Hector 1989). Clubbing may fact that myxomas sometimes recur locally or in peripheral sites
also occur (Reynen 1995; Greenwood 1968). Obstruction of the confirms their neoplastic nature (New et al. 1970; Price et al. 1970;
mitral valve can cause shortness of breath, which may improve on Read et  al. 1974). On immunohistochemical and ultrastructural
recumbency, fatigue, weakness, or syncope, which can occur upon study, myxoma cells may show endothelial, epithelial, smooth mus-
recumbency (Reynen 1995; Markel et al. 1987; Nasser et al. 1972; cle, and fibroblastic features, suggesting a multipotential, mesen-
St. John Sutton et al. 1980; Harvey 1968; Selzer et al. 1972; Peters chymal cell origin (Reynen 1995; Landon et al. 1986; Goldman et al.
et  al. 1974). New-onset congestive heart failure, chest pain, and 1987). Embolic fragments of myxoma are sometimes detectable in
episodic pulmonary oedema are the most common cardiac-related tissue specimens (Greenwood 1968; Markel et  al. 1987; Boussen
symptoms (Bulkley and Hutchins 1979). et al. 1991), but occasionally one sees only ‘vasculitis’ (Leonhardt
One-quarter to one-half of patients with atrial myxoma will have and Kullenberg 1977).
one or more embolic events (Greenwood 1968; Reynen 1995; Wold Improved methods of detection and better surgical techniques
and Lie 1980; Bulkley and Hutchins 1979; St. John Sutton et  al. have increased survival substantially (Reynen 1995; Greenwood
1980), half of which affect the central nervous system (Schmidley 1968; Markel et al. 1987; Bulkley and Hutchins 1979; Nasser et al.
1993; Markel et al. 1987) causing focal abnormalities (New et al. 1972; St. John Sutton et al. 1980; Peters et al. 1974). Because the
1970; Stoane et al. 1966; Schwarz et al. 1972). Peripheral emboli tumour is occasionally multicentric, direct visualization of all four
may cause mononeuropathy multiplex, simulating vasculitis (Byrd cardiac chambers is important (Markel et al. 1987). Full thickness
et al. 1980). Right-sided myxomatous emboli often produce symp- excision of the tumour at the base of its pedicle is usually cura-
toms and signs of pulmonary thromboembolism, leading in some tive, but recurrences do occur. Most recurrences are local and occur
instances to pulmonary hypertension (Wold and Lie 1980; Markel within the first or second postoperative year. In some cases, how-
et al. 1987). ever, the interval is longer (Desousa et al. 1978; Markel et al. 1987;
While the cardiac examination may be normal, some patients St. John Sutton et al. 1980; Markel et al. 1986). Myxomas may also
have an accentuated first heart sound, an increased pulmonic com- recur at extracardiac sites (Desousa et al. 1978; Markel et al. 1986),
ponent of the second heart sound, and a diastolic rumble suggest- presumably from slow growth of embolic tissue.
ing mitral stenosis (Greenwood 1968; Markel et al. 1987; St. John
Sutton et al. 1980; Harvey 1968; Selzer et al. 1972). A unique, but Other neoplasms
infrequent finding is a low-frequency tumour plop heard 0.08 to Both haematological and solid organ malignancies can be
0.15 seconds after the second heart sound; it is mistaken at times associated with a paraneoplastic vasculitis (Fain et  al. 2007;
for an S3 or opening snap (Greenwood 1968; Markel et al. 1987; Solans-Laque et  al. 2008). Neoplasms can injure blood vessels
St. John Sutton et al. 1980; Harvey 1968). Rarely, a to-and-fro car- by inducing immune-mediated inflammation (Desousa et  al.
diac rub with a crunching quality may appear (Greenwood 1968). 1978; Markel et al. 1986), occluding the vessel via embolization
Right-sided myxomas can produce murmurs of tricuspid stenosis of tumour particles (Mertz and Conn 1992; Greenwood 1968;
or regurgitation, jugular venous distension, hepatomegaly, oedema, Markel et al. 1987; Boussen et al. 1991; Primka et al. 1993), induc-
or ascites (Markel et al. 1987). Ventricular myxomas can cause signs ing a hypercoagulable state (Mertz and Conn 1992; O’Keeffe et al.
of ventricular outflow obstruction (Markel et al. 1987; Wold and 1992; Nachman and Silverstein 1993; Fengler et al. 1990), produc-
Lie 1980). ing an abnormal protein (Mertz and Conn 1992; Stone et al. 1989;
The common findings of anaemia, leukocytosis, thrombocy- Dornan et al. 1985; Kois et al. 1991), or by directly invading the
topenia, hypocomplementaemia, elevated ESR, acute phase reac- vessel wall (Kanno 1987; Gabriel et al. 1986; Webster et al. 1986;
tants, and serum autoantibodies (Gravallese 1995; Reynen 1995; Thomas et al. 1994; Leeds and Rosenblatt 1972; Leeds et al. 1971).
Greenwood 1968; Wold and Lie 1980; St. John Sutton et al. 1980; Furthermore, tumours sometimes invade the microscopic nerves
Byrd et  al. 1980; Selzer et  al. 1972; Savige et  al. 1988)  probably supplying the vessel (O’Connor 1884) and in some instances the
result from the immune-stimulating properties of myxomas, such tumour involves major nerves per se, creating the picture of mon-
as the production of IL-6. For unclear reasons, polycythaemia may oneuritis multiplex (Jones and Edgar 1995), thereby simulating
accompany right atrial myxomas (Greenwood 1968; Wold and Lie vasculitis. Phaeochromocytoma, perhaps by releasing catecho-
1980; Markel et al. 1987; St. John Sutton et al. 1980). lamines, can generate ischaemic lesions and angiographic find-
Echocardiography confirms a mass in virtually 100% of the ings typical of vasculitis (Armstrong and Hayes 1961; McColl and
cases (Markel et al. 1987; Rajpal et al. 1979). A two-dimensional Fraser 1995).
CHAPTER 44 imitators of vasculitis 639

Angiotropic lymphomas and the related condition, lymphoma- ulcerations are slow to heal and may leave scarring or postinflam-
toid granulomatosis, can also imitate vasculitis (Glass et al. 1993; matory pigmentation (Herman et al. 1981). Middle-aged women
Kleinschmidt-DeMasters et al. 1992; Walker et al. 1994; Roux et al. seem especially vulnerable (Herman et al. 1981; Jacob et al. 1986),
1995; Thomas et al. 1994; Wu et al. 2005). Malignant angioendo- perhaps because of hyper-reactivity of their arterial circulation to
theliomatosis, once considered a neoplasm of vascular endothelial cold, or because of diminished temperature in the skin overlying
cell origin, is now recognized as a B-cell lymphoma (Fredericks a relatively thick layer of subcutaneous fat (Jacob et al. 1986; Page
et  al. 1991b; DiGiuseppe 1994; Sienknecht et  al. 1995; Sheibani and Shear 1988). Other possible predisposing factors include sys-
et al. 1986; Kao et al. 1992; Wick et al. 1986) with a predilection for temic illness, nutritional deficiencies, neuromuscular dysfunction,
vessels of the central nervous system (Lie 1992b; Kamesaki et al. and genetic traits (Millard and Rowell 1978; Herman et al. 1981;
1990; Detsky et al. 2006; Pless et al. 2010) and skin (Asada et al. Thomas 1964; Kelly and Dowling 1985).
2007). Primary vascular sarcomas may obstruct vessels or embolize Histopathological examination of cutaneous lesions demon-
distally (Mason et al. 1982); occasionally they induce aneurysms strates non-specific inflammation and oedema in the papillary der-
(O’Donnell and O’Connell 1993). mis along with perivascular mononuclear cell infiltration around
dermal arterioles (Herman et al. 1981; Page and Shear 1988; Wall
External injury and Smith 1981). Proliferation of the vessel intima (Page and
Trauma Shear 1988), deposition of fibrin, and a true lymphocytic vasculitis
The ‘hypothenar hammer syndrome’ exemplifies how external (Herman et al. 1981) are occasional findings. Angiography of an
trauma can affect a blood vessel (Savader et al. 1988; Pineda et al. affected extremity may show vascular occlusions and aneurysms
1985; Vayssairat et al. 1987; Wernick and Smith 1989). The typical (Jacob et al. 1986).
history is one of hitting or pushing a hard surface with the hypoth- Protection from cold is the sine qua non of treating pernio (Jacob
enar aspect of the hand. This circumstance conjures up the pic- et al. 1986; Page and Shear 1988). Additionally, a weight loss pro-
ture of a blacksmith—the patient’s hand is the ‘hammer’, the hook gramme could reduce the insulating layer of subcutaneous fat and
of the hamate bone is the ‘anvil’, and the unprotected superficial thereby raise skin temperature (Page and Shear 1988). Use of calcium
palmar branch of the ulnar artery is the ‘horseshoe’. Intermittent channel blockers is sometimes successful (Dowd et al. 1986) and
lancinating pain ensues, followed by a dull ache over the hypoth- glucocorticoid creams can decrease itching (Ganor 1983).
enar eminence. The subsequent ischaemic symptoms often lead Polyvinyl chloride
to a diagnosis of Raynaud’s phenomenon, but careful questioning
Exposure to polyvinyl chloride tubing during haemodialysis can
reveals that neither a triphasic colour change nor involvement of
cause a necrotizing dermatitis. Histopathological examination
the thumb occurs (Pineda et  al. 1985). An angiogram will show
shows thrombosis of vessels with fibrin deposits in the walls of
an irregularity, an aneurysm, or an occlusion of the ulnar artery,
arterioles and inflammatory infiltrates without leukocytoclasis.
sometimes in association with occlusion of the more distal arter-
Precipitates of IgG and complement occur in small vessels, but
ies (Sack 1993; Savader et al. 1988; Pineda et al. 1985; Vayssairat
serum levels of immunoglobulin and complement are normal.
et al. 1987) (see Figure 17.17). Histopathological examination often
When polyurethane is substituted for polyvinyl chloride, the rash
demonstrates thrombosis on the intimal surface and fibrosis in the
does not occur (Bommer et al. 1979).
media of the affected vessels (Vayssairat et al. 1987). Trauma to the
radial artery can cause a ‘thenar hammer syndrome’, with digital Radiation
cyanosis and splinter haemorrhages (White and Parke 2005). External radiation can injure vascular endothelial cells within
The vascular narrowing seen on cerebral angiograms after hours (Fonkalsrud et al. 1977; Page and Shear 1988). There may be
severe head trauma could result from pathogenic mechanisms subsequent thickening and irregularity of the intima as well as focal
similar to those of the hypothenar hammer syndrome. In cases of fibrosis and necrosis of the media. Exposure to 3600 to 6800 rad
head trauma, however, cerebral vasospasm consequent to intrac- (McCready et al. 1983) can cause rupture or occlusion of an artery
ranial haemorrhage may be an additional factor (Suwanwela and within several weeks or after many years (Lie 1992a; McCready
Suwanwela 1972). Trauma to the carotid artery can mimic cerebral et al. 1983) (Figure 44.2). Mediastinal radiation can cause rapidly
vasculitis by causing dissection of the vessel wall (Nance et al. 1997; progressive coronary artery stenosis even in the absence of risk fac-
Lanczik et  al. 2003)  or formation of a thrombus that embolizes tors for cardiovascular disease, with a latency of onset between 3
distally (Thomas and Lowitt 1995). Likewise, if atlantoaxial sub- and 29 years (Bramkamp et al. 2007).
luxation causes injury to the vertebral arteries, multiple cerebellar
infarctions may ensue (Shim et al. 1998).
Internal injury
Exposure to cold Hypertension can be considered a form of internal vascular ‘trauma’.
Prolonged exposure to non-freezing cold, particularly when the Acute elevations of blood pressure may produce angiographic find-
humidity is high, can cause the condition known as pernio, or chil- ings of vasoconstriction and dilatation of cerebral vessels (Garner
blains (Herman et al. 1981; Jacob et al. 1986; Page and Shear 1988). In et al. 1990). Segmental narrowing of these vessels may also occur in
the acute, self-limited form of this disorder, pruritic or painful pur- the postpartum (Call et al. 1988; Bogousslavsky et al. 1989) as well
plish swellings appear on the extremities (or rarely the face) about as before, during, or after episodes of migraine (Masuzawa et al.
24 hours after the exposure. Tender blue nodules, and occasionally 1983; Serdaru et al. 1984; Call et al. 1988; Bogousslavsky et al. 1989).
ulcerations, follow and persist for 10–14 days (Herman et al. 1981). Dissections of extracranial and intracranial arteries, and occasion-
Some patients acquire a chronic form of pernio in which the char- ally of visceral arteries, may induce long foci of arterial narrow-
acteristic lesions recur during winter months. In such cases, the ing on angiogram. This finding can prompt an erroneous diagnosis
640 SECTION 6 mimickers of vasculitis

specific for this process, include segmental narrowing and dilata-


tion (‘string of beads’) of the arteries. Changes are usually bilateral
and diffuse. A key feature of the syndrome is that the vasoconstric-
tion is transient—resolution of the vascular narrowing can occur in
as early as a few days. Brain biopsy is not recommended for diagno-
sis (Ducros 2012; Calabrese et al. 2007).
No randomized clinical trials have identified treatments for
RCVS. Treatments are generally supportive since most episodes
are self-limited. Recommendations include avoiding potential trig-
gers and ceasing the use of vasoactive drugs. Medications targeting
vasospasm are frequently considered, and include calcium channel
blockers such as nimodipine and verapamil. Immunosuppression
with glucocorticoids may worsen the clinical course and is not rec-
ommended (Ducros 2012; Calabrese et al. 2007).

Biochemical abnormalities
Amyloid
Amyloid angiopathy can appear as an isolated phenomenon in
the central nervous system (Silbert et al. 1995; Okazaki et al. 1989;
Gilbert and Vinters 1983; Mandybur 1986) or as part of systemic
amyloidosis with an associated paraprotein (Breathnach and Wells
Fig. 44.2 Radiation-induced vasculopathy—total occlusion of the middle 1980; Jennette et  al. 1982). Its clinical features include progres-
cerebral artery and marked narrowing of the anterior cerebral artery in a young girl sive dementia, multiple cerebral infarctions, and signs suggesting
3 years status post radiation therapy. temporal arteritis (Silbert et al. 1995; Okazaki et al. 1989; Gilbert
and Vinters 1983; Mandybur 1986; Salvarani et al. 1994; Rao and
Allen 1993; Churchill et al. 2003). Moreover, when intracerebral
of arteritis (Caplan and Louis 1996).Obstruction of vasa vasorum haemorrhage accompanies dementia, amyloid cerebrovascular
by any mechanism can produce ischaemic necrosis of an arterial disease is a leading diagnostic consideration (Gilbert and Vinters
wall. Mycotic aneurysms may form by such a process (Olmsted and 1983). Amyloid can also affect peripheral vessels and produce
McGee 1977). ischaemic organ damage or purpura (Breathnach and Wells 1980;
Jennette et al. 1982; Auethavekiat et al. 2004). Unexplained renal
Reversible cerebral vasoconstriction syndrome failure or haematuria, with or without proteinuria, sometimes her-
Reversible cerebral vasoconstriction syndrome (RCVS) is a tran- alds amyloidosis (Friman and Pettersson 1996). Angiography may
sient, non-inflammatory, non-atherosclerotic constriction of the show vascular narrowing or occlusion (Auethavekiat et al. 2004).
cerebral arteries previously known by several names, including Histological evaluation of affected vessels demonstrates infiltration
‘Call–Fleming syndrome’ or ‘benign angiopathy of the CNS’ (see with amyloid, accompanied at times by obliterative intimal changes,
Chapter  36). This acute and typically self-limited process more aneurysm formation, perivascular or transmural inflammatory
commonly affects women between the ages of 40 and 50 years. The infiltrates, and fibrinoid necrosis (Okazaki et al. 1989; Mandybur
main symptom of RCVS is the ‘thunderclap headache’—a severe 1986; Breathnach and Wells 1980; Jennette et al. 1982).
headache that reaches peak intensity within seconds, similar to the
headache observed in subarachnoid haemorrhage. This headache α1-antitrypsin deficiency
can be occipital or diffuse, lasting from a few minutes to several α1-antitrypsin, the most abundant proteinase inhibitor in human
days, and be associated with nausea, vomiting, and photosensitiv- plasma, plays a major role in protecting tissues (including blood
ity. Many patients report a trigger such as stressful situations, vas- vessels) from endogenous toxins (Cox 1994). A deficiency in this
soactive drugs, and physical exertion. Neurological deficits such inhibitor reportedly increases the incidence of fibromuscular dys-
as visual defects, hemiplegia, dysarthria, and ataxia can occur due plasia (Schievink et al. 1994b), intracranial aneurysm (Schievink
to cerebral ischaemia from the constricted artery. Seizures have et al. 1994a), spontaneous dissection of cervical or peripheral arter-
been reported in up to 17% of cases, but epilepsy does generally ies (Cattan et al. 1994), panniculitis (Schievink et al. 1994a), and
not develop. Ischaemic and haemorrhagic stroke as well as cerebral vasculitis (Mahr et al. 2010). Some of these associations, however,
haemorrhage causing death has been reported (Calabrese et  al. remain unproven.
2007; Ducros 2012; Singhal et al. 2009).
Physical examination is usually unremarkable in the absence of Calciphylaxis
neurological deficits. Laboratory measures, such as blood counts, Calciphylaxis is a small-vessel vasculopathy that causes tissue
liver and kidney function tests, inflammatory markers, and analy- ischaemia and is most commonly seen in patients with renal fail-
sis of cerebrospinal fluid are usually normal. The results of initial ure. This arteriopathy typically occurs in patients with a high cal-
neuroimaging using CT or MRI are normal in 55–80% of patients. cium phosphate product (Roe et al. 1994; Conn et al. 1973; Gipstein
Abnormal findings include cerebral infarcts, intraparenchymal et al. 1976), and elevated parathyroid levels are frequently noted.
haemorrhage, reversible brain oedema, and posterior reversible However, calciphylaxis has developed in patients without an abnor-
encephalopathy syndrome. Angiographic findings, which are not mal calcium phosphate product or renal failure (Jacobs-Kosmin
CHAPTER 44 imitators of vasculitis 641

and Dehoratius 2007). Diabetes, female gender, and obesity may (a)
also be risk factors (Fine and Zacharias 2002; Mazhar et al. 2001).
The most common manifestations tend to involve the skin and
extremities. Affected patients typically develop a painful livedo
reticularis-like rash that becomes plaque-like, nodular, or bullous,
and may progress to necrosis with ulceration. Lesions can develop
initially in the distal extremities (e.g. fingers or toes) or more
proximally on the thighs. Other organ involvement includes skel-
etal muscle and intestinal tissue (Jacobs-Kosmin and Dehoratius
2007), but infarction of major organs is rare (Adrogue et al. 1981).
Upon histological evaluation, pathological specimens characteristi-
cally show intravascular deposits in arterioles and venules. Lesions
can progress to develop intimal proliferation and endovascular
fibrosis, which corresponds to end organ damage. Affected vessels
tend to have little inflammation in or surrounding the vessel walls
(Jacobs-Kosmin and Dehoratius 2007). (b)
Unfortunately, treatment of advanced disease is rarely successful,
and mortality remains high. Normalization of the calcium phos-
phate product through dietary changes, phosphate binders, and
low calcium dialysis baths is recommended. Parathryoidectomy is
also often recommended (Blumberg and Weidmann 1977; Angelis
et  al. 1997), but a survival benefit has not been clearly shown
(Jacobs-Kosmin and Dehoratius 2007). Other treatment options
such as intravenous sodium thiosulphate (Jacobs-Kosmin and
Dehoratius 2007), bisphosphonate infusion, cinacalcet, and hyper-
baric oxygen (Vassa et al. 1994) may provide benefit.

Fabry’s disease (angiokeratoma corporis diffusum)


Fabry’s disease is sphingolipid storage disease caused by a defi-
ciency in the lysosomal enzyme α-galactosidase A.  Lack of this
enzyme results in widespread deposition of neutral glycosphin-
golipids, primarily globotriaosylceramide, and galactosylceramide
(Anonymous 1990; Meschia et al. 2005; Zarate and Hopkin 2008), Fig. 44.3 Angiokeratomas in the mouth (a) and on the penis (b) of a young man
leading to cellular dysfunction. Accumulation of these molecules with Fabry’s disease.
in the vascular endothelium is thought to cause poor perfusion of
affected organs and irreversible organ dysfunction. This disease is
inherited in an X-linked recessive fashion, but it is unique in that it burning paresthesias, the neuropathy may intensify with exertion
may cause symptoms in female carriers (Serdaru et al. 1984; Gupta or fever, and be accompanied by hypohidrosis (Desnick et al. 1989;
et al. 2005; Rolfs et al. 2005), although symptom onset tends to be Serdaru et al. 1984). Abnormalities of thermal sensation, especially
at a later age (Zarate and Hopkin 2008). cold sensitivity, are common, even in otherwise asymptomatic car-
In adult men, virtually any organ system can be involved. As riers (Morgan et  al. 1990). A  characteristic finding on slit-lamp
a consequence, the clinical findings vary considerably. The char- examination of the cornea is a whorl-like lesion similar to that
acteristic skin manifestations are angiokeratomas (Figure  44.3), occasionally seen after use of phenothiazines, chloroquine, or indo-
red-purple papules that blanch on pressure if not thrombosed. These methacin, which can be seen in asymptomatic carriers (Meschia
lesions typically appear in a ‘bathing suit’ distribution (genitalia, et al. 2005). Additional ocular findings are cataracts, corneal opaci-
buttocks, and lower abdomen) during childhood or adolescence. ties, and aneurysmal dilatations of conjunctival and retinal vessels
Occasionally, they occur on the elbows, thighs, fingers, lips, and (Klein 1986). Other manifestations of this disorder include: cere-
mucous membranes (Sack 1993; Serdaru et al. 1984; Anonymous brovascular occlusions (Serdaru et al. 1984); obstructive airway dis-
1990). The major complications of Fabry’s disease are kidney and ease (Rosenberg et al. 1980); non-destructive arthropathy (Sheth
heart disease. Renal involvement is often discovered via abnormal and Bernhard 1979); ischaemic necrosis of bone (Ross et al. 1993);
laboratory studies, which can include proteinuria (a poor prognos- bowel dysfunction (Rowe et al. 1974); vertigo; sensorineural hear-
tic sign) and an inability to concentrate urine. Renal disease gener- ing loss; and lymphadenopathy.
ally progresses to renal failure without therapy (Zarate and Hopkin Diagnosis in affected men can be made by detecting deficient
2008). The classic cardiac manifestation is hypertrophic cardiomy- or absent α-galactosidase A activity in plasma or gene sequencing;
opathy causing mild diastolic dysfunction, which can progress to affected women have low to normal levels of α-galactosidase activ-
systolic dysfunction. Patients are also at risk for cardiac arrhyth- ity and require genetic diagnosis (Meschia et al. 2005). Serum levels
mias (Nakao et al. 1995; Becker et al. 1975; Fisher et al. 1992; Zarate of globotriaosylceramide help confirm the diagnosis (Serdaru et al.
and Hopkin 2008). A painful autonomic neuropathy is one of the 1984), and can be used to monitor treatment response if elevated at
earliest and most debilitating manifestations. Often presenting as diagnosis (Zarate and Hopkin 2008). On biopsy, an angiokeratoma
642 SECTION 6 mimickers of vasculitis

typically shows ectatic cutaneous capillaries protruding into a Finding an increased 24-hour urinary total oxalate excretion
hyperkeratotic epidermis. Similar lesions, however, may appear in establishes the diagnosis of hyperoxaluria; biopsy of affected tis-
other inherited lysosomal disorders (Rodriguez-Serna et al. 1996; sues reveals the vascular oxalate deposits (Kuiper 1996; Baethge
Kanzaki et al. 1993). et al. 1988). With the onset of renal failure, serum oxalate levels
Treatment is largely symptomatic. Enzyme replacement ther- are difficult to interpret (Baethge et al. 1988). Consequently, dis-
apy has been to shown to delay the development of clinical events tinguishing primary from secondary forms of oxalosis may require
(Banikazemi et al. 2007; Mehta et al. 2009) and has been avail- specific enzyme assays. Although oxalate is readily dialysable, hae-
able since 2003 in the United States. While clinical effect has modialysis and peritoneal dialysis cannot keep up with its synthe-
been shown in studies, the benefit for an individual patient is sis. Thus, most effective approaches for preserving renal function
difficult to document. Damage present at the start of treatment enhance oxalate solubility in the urine (phosphate, magnesium
is often irreversible, and enzyme replacement therapy does not oxide, or citrate supplementation) or decrease its production (high
resolve all symptoms. The cost of such therapy, however, is pro- doses of pyridoxine) (MacConnell and Ferro 1995; Milliner et al.
hibitive (Desnick et  al. 2003; Schiffmann et  al. 2001; Eng et  al. 1994). Combined liver and renal transplantation offer the pos-
2001; Meschia et  al. 2005). Prior to the use of enzyme therapy, sibility of reversing the underlying metabolic defect (Bastani and
most patients succumbed to renal failure by age 50 if not started Nahass 1999).
on dialysis (Anonymous 1990). The long-term effect of enzyme Infection
therapy on this outcome is not yet known. Renal transplantation,
Direct infection of vessel walls, as well as infection-initiated immu-
although occasionally of temporary benefit (Mosnier et al. 1991),
nological or toxic processes, can compromise vascular integrity and
does not appear to supply enough of the missing enzyme to stop
simulate vasculitis (Sundy and Haynes 1995; Somer and Finegold
progression of the disease.
1995; Lie 1996; Mandel and Calabrese 1998). An array of organ-
Homocystinuria isms of various types may be the culprits (Table 44.3). The clinical
Homocysteine can injure vascular endothelium, potentiate the oxi- manifestations range from non-specific systemic complaints and a
dation of low-density lipoprotein cholesterol, and promote throm- variety of skin lesions to infarction of major organs. In addition,
bosis (Stein and McBride 1998; Welch and Loscalzo 1998; Clarke changes of ‘vasculitis’ on cerebral angiograms may actually stem
et  al. 1991; Robinson et  al. 1994). Patients who are homozygous from:  bacterial endocarditis (Leeds and Goldberg 1971); viral
for deficiency of cystathionine β-synthase have hyperhomocyst- infections, including HIV and members of the herpes family (Lie
einaemia (and homocystinuria) and are thereby susceptible to 1996; Victor and Green 1976; Goldstein et al. 2010; Mehta et al.
premature atherosclerosis and thromboembolism (Robinson et al. 2011; Neuteboom et  al. 2011; Saraya et  al. 2006); mycobacterial
1994). Heterozygote patients (approximately 1–2% of the popula- infection (Clarke et al. 2005); spirochetal infection (Kakumani and
tion) have milder elevations of homocysteine levels but are also Hajj-Ali 2009); basilar meningitis secondary to bacteria, mycobac-
predisposed to occlusive vascular disease (Graham et  al. 1997; teria, fungi, or spirochetes (Leeds et al. 1971; Greitz 1964; Lehrer
Welch and Loscalzo 1998; Clarke et  al. 1991; Boers et  al. 1985; 1966; Thomas and Hopkins 1972; Vatz et al. 1974; Tjia et al. 1985;
Selhub et al. 1995). Potential treatments include pyridoxine, folate, Veenendaal-Hilbers et al. 1988; Wheat et al. 1990; Williams et al.
and vitamin B12 supplementation, as well as a diet low in methio- 1992), or brain abscess. An example of Salmonella aortitis is shown
nine (Meschia et al. 2005). When low serum levels of folate accom- in Figure 17.12.
pany hyperhomocysteinaemia, supplementation of this vitamin Some infections cause cutaneous papules, nodules, and ulcers
can decrease serum homocysteine levels and could conceivably by non-vascular mechanisms. An example is the granulomatous
ameliorate the associated vasculopathy (Stein and McBride 1998; panniculitis (erythema induratum) induced by Mycobacterium
Selhub et al. 1995). Patients with homocystinuria and a history of tuberculosis (Baselga et  al. 1997; Rademaker et  al. 1989; Ollert
ischaemic stroke should be placed on antiplatelet therapy (Meschia et al. 1993). Infections with organisms such as Strongyloides ster-
et al. 2005). coralis (Wachter et al. 1984; Reiman et al. 2002; Mora et al. 2006),
trichinella (Frayha 1981), acanthamoeba (Murakawa et  al. 1995;
Hyperoxaluria
Chandrasekar et al. 1997; Slater et al. 1994), HIV (Lie 1996; Lipton
Hyperoxaluria is an autosomal recessive disorder that causes
and Ma 1996; Mandel and Calabrese 1998; Marks and Kuskov 1995;
accumulation of oxalic acid, an end-product of glycine metabo-
Simpson and Tagliati 1994), Pythium insidiosum (Prasertwitayakij
lism (Baethge et al. 1988). Patients with this condition typically
et al. 2003), and rabies virus can also produce symptoms and signs
have a history of nephrocalcinosis, often beginning before age 5
identical to those of vasculitis. We have observed a patient with
(Kuiper 1996; Baethge et al. 1988; Blackburn et al. 1975). Renal
myalgias, scalp tenderness, jaw pain, and a markedly elevated ESR
failure may eventually ensue, after which calcium oxalate deposits
in whom the initial diagnosis of ‘giant cell arteritis’ changed to
in extrarenal tissues such as heart, skin, bone, joints, blood ves-
orbital cellulitis when orbital swelling, proptosis, and gaze paralysis
sels, and eyes. Oxalate crystals may also accumulate in the media
subsequently appeared.
of the vessel wall or physically occlude small vessels (Baethge
et al. 1988; Blackburn et al. 1975). In addition, calcium oxalate Congenital and inherited abnormalities
activates complement and thus may trigger neutrophil-mediated Genetic disorders that affect the large vessels can mimic the pri-
injury of endothelial cells (Boogaerts et al. 1983). Thus, vascu- mary large vessel vasculitides. These disorders include Marfan’s
lar complications are common (Kuiper 1996; Baethge et al. 1988; syndrome, an autosomal dominant disease caused by mutations
Blackburn et  al. 1975), consisting of acrocyanosis, Raynaud’s in the fibrillin-1 gene. In addition to the classic manifestations
phenomenon, livedo reticularis, decreased pulses, and peripheral of above average height and subluxation of the ocular lens (ecto-
gangrene. pia lentis), Marfan’s is also known to cause ascending thoracic
CHAPTER 44 imitators of vasculitis 643

Table 44.3 Infectious organisms, diseases, or syndromes associated intimal and adventitia of blood vessels, resulting in arterial sten-
with vasculitis oses or aneurysms (Tomsick et al. 1976; DiPrete et al. 1990; Finley
and Dabbs 1988; Jones et al. 1998; Chan et al. 1998; Meyers et al.
Organism, diseases, or syndrome Reference(s) 1974)  (see Figure 17.18). These abnormalities most commonly
involve the renal arteries (DiPrete et al. 1990; Finley and Dabbs
Bacterial
1988), but other major vessels may be affected (Tomsick et al. 1976;
Neisseria gonorrhea Mastrolonardo et al. 1994
DiPrete et al. 1990). In addition to the characteristic findings of
Neisseria meningitides Speranza and Javaheri 2008 small yellow to orange skin papules and ocular angoid streaks,
Burkholderia pseudomallei Torrens et al. 1999 pseudoxanthoma elasticum can cause premature atherosclerosis
(melioidosis) due abnormal calcification of the internal elastic lamina (Nishida
Fungal Benedict et al. 1992 et al. 1990; Lebwohl et al. 1993; Travers et al. 1979).
Histoplasmosis Stone et al. 1998 Fibromuscular dysplasia is a non-inflammatory,
non-atherosclerotic vascular disease that can affect the intima,
Aspergillus Seton et al. 2008
media, or adventita of small and medium sized vessels. The dam-
Fusarium species Nelson et al. 1994
age can cause stenosis, dilatation, or aneursymal formations, which
Sporotrichosis Byrd et al. 1998 can be misdiagnosed as vasculitis The renal arteries are affected in
Cryptococcus Aberfeld and Gladstone 1967 60–75% of cases, and fibromuscular dysplasia is a well-recognized
Mycobacterial cause of renovascular hypertension. However, any artery can be
Mycobacterium tuberculosis Baselga et al. 1997; Blanco et al. 1999; involved (Slovut and Olin 2004; Jones et al. 1998; Rybka and Novick
Clarke et al. 2005 1983; Stokes et al. 1996). Involvement of the carotid and vertebral
Mycobacterium leprae Camps-Garcia et al. 2011 arteries can result in stroke and subarachnoid haemorrhage. The
Viral
cause of this process is unknown, but a genetic component is sus-
pected since this disease is more common in first-degree relatives
HIV/AIDS Gruber et al. 1997; Marks and
of affected individuals. Treatment is comprised of antihypertensive
Kuskov 1995; Joshi et al. 1987
therapy for individuals with renovascular hypertension and revas-
Varicella-zoster virus Erhard et al. 1995; Gray et al. 1994
cularization either through percutaneous angioplasty or surgical
Epstein–Barr virus Murakami et al. 1998 methods (Slovut and Olin 2004; Pontes Tde et al. 2012). Segmental
Rubella Esterly and Oppenheimer 1967 arterial mediolysis may be a form of fibromuscular dysplasia (Slavin
Parvovirus B19 Finkel et al. 1994 et al. 1995; Chan et al. 1998) (see Figure 17.19). Grange syndrome
Spirochetal/Rickettsial is a newly recognized hereditary disorder characterized by the ste-
Neurosyphilis Peters et al. 1993 nosis or occlusion of multiple arteries, including renal, abdominal,
and cerebral arteries. This arterial occlusive disease appears simi-
Borreliosis/ Lyme disease May and Jabbari 1990; Fontana et al.
1996;
lar to fibromuscular dysplasia on angiography, and can also cause
hypertension. This syndrome is associated with congenital heart
Rickettsia Wenzel et al. 1986
defects, brachysyndactyly, bone fragility, and learning disabilities
Parasitic (Grange et al. 1998).
Chagas’ disease Oddo et al. 1992
(Trypanosomiasis americana)
Miscellaneous conditions
Cysticercosis Salgado et al. 1997 Drug effects
Strongyloides Wachter et al. 1984; Reiman et al. 2002 Ergot derivatives (Kapoor 1976; Henry et  al. 1984)  and sym-
pathomimetic drugs (Fallis and Fisher 1985; Citron et  al. 1970;
Visceral larva migrans Kraus et al. 1995
Matick et  al. 1983; Lake et  al. 1990; Sloan et  al. 1991; Pearlson
Toxoplasmosis Huang and Chou 1988
et al. 1993; Tapia and Schumacher 1993; Fredericks et  al. 1991a;
Kaye and Fainstat 1987; Greene 2005), particularly amphetamines
and ephedrine (Calabrese and Mallek 1988), can induce vascular
abnormalities. The cause of these abnormalities is probably mul-
aneurysms and dissection (Judge and Dietz 2005). Ehlers–Danlos tifactorial. Vasospasm plays a role, as evidenced by the transient
syndrome type IV is also an autosomal dominant disorder, caused segmental arterial constrictions visible on angiograms soon after
by mutations in the type III procollagen gene, associated with exposure to the offending agent (Rumbaugh et al. 1971a; Kapoor
arterial aneurysm, dissection, and rupture (even in the absence 1976; Henry et  al. 1984). In some instances, histopathological
of aneurysms). Patients with this type do not generally exhibit studies show true vasculitic lesions (Citron et al. 1970; Tapia and
joint hypermobility (Imahori et  al. 1969). Loeys–Dietz syn- Schumacher 1993), and angiograms occasionally show more per-
drome is an autosomal dominant disorder associated with aortic manent changes, for example aneurysms (Citron et al. 1970; Matick
and arterial aneurysms with tortuosity, as well as bifid uvula or et al. 1983). Thus, vasoconstriction may be the initial event, fol-
cleft palate and orbital hypertelorism. Mutations in the either of lowed by ischaemia with resultant inflammation of the vessel wall
the two transforming growth factor β receptors have been iden- (Fredericks et al. 1991a; Rumbaugh et al. 1971b; Lake et al. 1990;
tified as the cause of the syndrome (Van Hemelrijk et al. 2010). Karch and Billingham 1988). Some drugs, on the other hand, can
Neurofibromatosis, a hamartomatous abnormality of neural crest stimulate the release of toxic mediators or induce a coagulopathy
tissue resulting from mutations in the ABCC6 gene, can affect the (Rumbaugh et al. 1971a). Concomitant infection or emboli of drug
644 SECTION 6 mimickers of vasculitis

contaminants occasionally play a role (Rumbaugh et  al. 1971a; Gulati and Donato 2012). Neutropenia and hypocomplementae-
Citron et al. 1970) (see Figure 17.13). mia are common (Graf et al. 2011; Milman and Smith 2011; Bose
Alpha-interferon, a protein with antiviral and antitumour activ- and Calabrese 2012; Poon et al. 2011). ANCA antibodies have been
ity, occasionally induces Raynaud’s phenomenon or a thrombotic detected in all patients and frequently exhibit high-titre perinu-
angiopathy (Schapira et  al. 2002; Zuber et  al. 2002). Likewise, clear staining on indirect immunofluorescence testing. However,
recombinant human erythropoietin can induce a thrombotic vas- the ANCA patterns detected on immunofluorescence testing are
culopathy (Gibson et al. 2005). often discordant with the specific antigenic targets seen on ELISA
Tumour necrosis factor (TNF) inhibitors can induce vasculitic testing. ANCAs found in patients with levamisole-induced vas-
skin ulcerations, scleritis, mononeuritis, leukocytoclastic vascu- culopathy typically recognize other neutrophil antigens besides
litis, and proliferative lupus nephritis. These lesions resolve after myeloperoxidase and proteinase 3, such as elastase, cathepsin G,
withdrawal of the agent and treatment with glucocorticoids (Jarrett or lactoferrin. Other autoantibodies are also frequently observed,
et al. 2003; Mor et al. 2005). including ANAs, anti-dsDNA autoantibodies, anticardiolipin anti-
Female reproductive hormones may exert a toxic effect on the bodies, and lupus anticoagulants (Milman and Smith 2011; Graf
vasculature. Intimal proliferation reportedly occurs in the ves- et al. 2011). Levamisole can be detected in the urine of affected
sel walls of women who are pregnant, postpartum, or taking oral patients using liquid or gas chromatography in tandem with mass
contraceptives (Irey and Norris 1973). Such changes might also be spectrometry.
responsible for the ‘transient cerebrovascular disease of pregnancy’ The optimal treatment of levamisole-induced vasculopathy is
(Brick 1988). unclear. Cessation of cocaine use may stop the progression of the
While propylthiouracil is well recognized to cause a drug-induced skin disease; the cutaneous lesions become chronic and progressive
antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis with continued exposure to cocaine (and presumably to levami-
that can resolve with cessation of the medication (Aloush et al. 2006; sole) (Milman and Smith 2011). Supportive care, immunosuppres-
Chen et al. 2012), this syndrome can also be observed with other sion with prednisone and rituximab, as well as anticoagulation with
medications such as proton pump inhibitors (Jacobs-Kosmin et al. warfarin has been used to treat the disease manifestations (Poon
2006). Polyarteritis nodosa-like vasculitis has also been observed et al. 2011; Gulati and Donato 2012).
with use of minocycline (Kermani et al. 2012).
Moyamoya disease
Cocaine and levamisole-associated vasculopathy Moyamoya disease is an occlusive vasculopathy primarily affect-
Long-term intranasal cocaine use can cause perforation of the nasal ing the arteries of the circle of Willis (Ueki et al. 1994; Suzuki and
septum, and potentially extensive destruction of the nose, sinuses, Takaku 1969; Takeuchi and Shimizu 1957). Although first recog-
and palate. These cocaine-induced midline destructive lesions nized in Japan (Takeuchi and Shimizu 1957), it affects virtually all
can be particularly difficult to distinguish from granulomatosis races (Ueki et al. 1994). The onset of disease has two peaks, one
with polyangiitis (Wegener’s) since biopsy of the affected tissue in the first and the other in the fourth to fifth decade of life (Ueki
can reveal necrotizing inflammation, and patients frequently pro- et al. 1994; Tzeng et al. 2005). Ischaemic attacks characterize the
duce ANCA (Friedman and Wolfsthal 2005; Rowshani et al. 2004; childhood form, whereas cerebral haemorrhage predominates in
Trimarchi et al. 2001). Perinuclear staining is the most common pat- adults (Scott and Smith 2009). In the early stages, angiograms
tern seen on indirect immunofluorescence testing, but cytoplasmic show stenoses of both carotid arteries at their suprasellar posi-
staining has also been observed. A high proportion of the ANCA tions. As these arteries progressively narrow, a characteristic
from patients with these lesions target human neutrophil elastase, network of moyamoya (‘hazy puff of smoke’) vessels appears
although a small proportion does target proteinase-3 (Wiesner (Figure 44.4). These increase in prominence as major trunks of the
et al. 2004; Peikert et al. 2008). Antihuman neutrophil elastase is anterior circle of Willis become occluded. With involvement of
found uncommonly in autoimmune diseases, so its presence sug- all the components (including the posterior cerebral arteries), the
gests cocaine-induced midline destructive lesions. Cocaine use has moyamoya vessels diminish in size and may completely disappear
also been associated with crescentic glomuleronephritis and ulcer- as collaterals develop from the extracranial circulation (Suzuki
ated skin lesions. and Takaku 1969). Conventional dye-based angiography as well
Levamisole, an antihelminthic agent approved for use in vet- as magnetic resonance imaging are the most common techniques
erinary medicine, has been identified in over 70% of the cocaine used to identify the vascular and parenchymal changes seen in
seized by the United States Drug Enforcement Administration (as this disease and can be used to confirm the diagnosis (Scott and
of 2010) and in cocaine samples worldwide (Gulati and Donato Smith 2009). Histopathological examination of stenotic vessels
2012). Previously used to treat children with nephrotic syndrome, shows fibrotic thickening without arteriosclerotic or inflamma-
this drug was subsequently removed from human use in 1999 due tory changes. Dilated vessels have attenuation of the media and
to its toxic effects including agranulocytosis and cutaneous ulcers fragmentation of the elastic lamina (Yamashita et al. 1983; Scott
from vasculopathy (Bose and Calabrese 2012). and Smith 2009).
Levamisole-induced vasculopathy is characterized most promi- Multiple factors may contribute to the pathogenesis of moy-
nently by retiform purpura and cutaneous necrosis of the extremi- amoya disease, including Asian heritage and previous radio-
ties, face, and ears. While the predominant histological pattern is therapy to the head or neck (Hoshimaru et al. 1991; Yamamoto
small-vessel thrombotic vasculopathy, leukocytoclastic vasculitis et  al. 1998; Masuda et  al. 1993; Tzeng et  al. 2005; Scott and
also occurs (Bose and Calabrese 2012; Graf et  al. 2011). Other Smith 2009). Case reports describe familial occurrences, an
reported manifestations include arthritis and arthralagia, as well increased incidence in Down’s syndrome (Ueki et al. 1994) and
as pauci-immune glomerulonephritis (Milman and Smith 2011; sickle cell disease, and antibodies to Ro (SS-A) and La (SS-B)
CHAPTER 44 imitators of vasculitis 645

(Provost et al. 1991). This disease typically progresses and has a


high mortality rate. Treatments are directed at improving blood
flow to the affected tissue. Medical therapy such as antiplatelet
therapy, anticoagulation, and calcium channel blockers do not
tend to halt the disease (Scott and Smith 2009). Surgical proce-
dures including both direct and indirect revascularization are
becoming accepted as the primary treatment method given their
documented success (Kinugasa et al. 1993; Ueki et al. 1994; Scott
and Smith 2009).
A variety of other miscellaneous conditions may imitate vascu-
litis (Table 44.4).
IgG4-related disease
IgG4-related disease is a recently identified systemic fibroinflam-
matory condition that can affect virtually every organ system,
including the aorta and its branches. While the epidemiologi-
cal characteristics of this disease are poorly defined, it is strik-
ing that the majority of affected individuals are men (62–83%)
over 50 years of age. This is in contrast to the female predomi-
nance of autoimmune diseases that mimic IgG4-related disease,
such as Sjögren’s syndrome and primary biliary cirrhosis (Stone
et al. 2012).
Fig. 44.4 Moyamoya—total occlusion of the middle cerebral artery and While the most recognized manifestations of IgG4-related dis-
marked narrowing of the anterior cerebral artery; prominent collateral ease are autoimmune sclerosing pancreatitis and submandibular/
moyamoya vessels. parotid gland enlargement, approximately 20% of patients have

Table 44.4 Miscellaneous imitators of vasculitis

Imitator Clinical features Vasculitis simulated


Angiolymphoid hyperplasia with eosinophilia Lymphadenopathy, erythematous papules Polyarteritis nodosa
Anorexia nervosa Livedo reticularis Connective tissue disease
Atrophie blanche Multifocal cutaneous infarctions of lower extremities Cutaneous vasculitis
Brain abscess Temporal headache Giant cell arteritis
Cerebral autosomal dominant arteriopathy with subcortical Focal cerebral ischaemia and progressive dementia CNS vasculitis
infarcts and leukoencephalopathy (CADASIL)
Cerebral segmental vasoconstriction (reversible) Headache, focal CNS ischaemia CNS vasculitis
Ear cerumen Temporal headache Giant cell arteritis
Glaucoma Temporal headache Giant cell arteritis
Gluten sensitivity Ataxia CNS vasculitis
Heparin-induced thrombotic thrombocytopenia Skin necrosis, cerebral haemorrhage, GI bleeding, adrenal Systemic vasculitis
haemorrhage
Inflammatory bowel disease CNS white matter lesions noted on MRI (without clinical CNS vasculitis
findings)
Mitochondrial myopathy, encephalopathy, lactic acidosis, and Headache, CNS ischemia CNS vasculitis
stroke-like episodes (MELAS)
Non-bacterial thrombotic (marantic) endocarditis Multiple organ infarctions Polyarteritis nodosa
Orbital infection Temporal headache CNS vasculitis
Sickle cell anaemia Headache, CNS ischaemia, segmental narrowing of cerebral CNS vasculitis
vessels
Small infarctions of the cochlear, retinal, and encephalic tissue Focal CNS ischaemia, visual and auditory disturbances CNS vasculitis
(SICRET syndrome)
Sphenoid pneumoceles Episodic blindness Giant cell arteritis
646 SECTION 6 mimickers of vasculitis

aortic involvement. In fact, approximately 10–50% of all inflam- Table 44.5 Non-vascular cutaneous conditions that may simulate
matory aortitis cases may be IgG4-related. This disease can pre- vasculitis
sent with frank aortitis, thoracic and abdominal aortic aneurysms,
periaortitis and periarteritis, and aortic dissection. However, many Familial leg ulcers
cases are found incidentally upon radiological imaging, because Infections
most patients do not have constitutional or vascular symptoms.
Insect bites (brown recluse spider)
In addition, many patients with IgG4-related disease experience
allergy symptoms such as asthma, eczema, rhinorrhoea, and can Malignant neoplasm
have modest peripheral eosinophilia. Other sinonasal symp- Neutrophilic dermatosis (Sweet’s syndrome)
toms include chronic sinusitis and nasal obstruction. While rare,
Panniculitis (erythema nodosum)
destructive bony lesions can occur. These allergic and sinus symp-
toms can lead to an erroneous diagnosis of eosinophilic granu- Polymorphic eruption of pregnancy
lomatosis with polyangiitis (previously known as Churg–Strauss Pyoderma gangrenosum
syndrome) or granulomatosis with polyangiitis (Wegener’s gran-
Sarcoidosis
ulomatosis). Many patients present with single-organ involve-
ment, but multiorgan disease can develop over time. On exam, Scurvy
the most prominent finding is parotid or submandibular gland
swelling, if those organs are involved (Stone et al. 2012; Zen and
Nakanuma 2010).
Diagnosis is made on radiological, serological, and histo-
References
pathological features. IgG4-related aortitis causes arterial wall Abdelmalek, M.F. and Spittell, P.C. (1995). 79-year-old woman with blue toes.
Mayo Clinic Proceedings, 70, 292–5.
thickening with homogeneous enhancement at the late phase of
Aberfeld, D. and Gladstone, J. (1967). Cryptococcal meningoencephalitis
contrast-enhanced computed tomography. Imaging of the sinuses
presenting with hemiplegia of sudden onset. Journal of the American
can show nasal polyps as well as mucosal thickening. The major- Medical Association, 202, 90–1.
ity of patients with IgG4-related disease have elevated IgG4 levels, Adrogue, H.J., Frazier, M.R., Zeluff, B., and Suki, W.N. (1981). Systemic calci-
but 30% of cases have normal levels. For these cases, an elevated phylaxis revisited. American Journal of Nephrology, 1, 177–83.
ratio of serum IgG4 to the total IgG level may be diagnostic (Stone Alam, M. and Sun, I. (1991). Transesophageal echocardiographic evalu-
et al. 2012). It is important to note that the histological findings ation of left atrial mass lesions. Journal of the American Society of
are similar regardless of the organ involved. Features highly Echocardiography, 4, 323–30.
suggestive of IgG4-related disease include a dense lymphoplas- Alarcon-Segovia, D., Mestanza, M., Cabiedes, J., and Cabral, A.R. (1997). The
macytic infiltrate, focal areas of storifrom fibrosis, and oblitera- antiphospholipid/cofactor syndromes. II. A variant in patients with sys-
temic lupus erythematosus with antibodies to beta 2-glycoprotein I but
tive phlebitis. Increased eosinophils can also be observed. IgG4
no antibodies detectable in standard antiphospholipid assays. Journal of
immunostaining is essential for the pathological diagnosis of this Rheumatology, 24, 1545–51.
disease. The number of IgG4+ plasma cells/hpf suggestive of this Alegre, V.A., Gastineau, D.A., and Winkelmann, R.K. (1989). Skin lesions
disease varies by tissue; however, an IgG4+/IgG+ plasma cell ratio associated with circulating lupus anticoagulant. British Journal of
of >40% confirms diagnosis of IgG4-related disease (Deshpande Dermatology, 120, 419–29.
et al. 2012). Aloush, V., Litinsky, I., Caspi, D., and Elkayam, O. (2006). Propylthiouracil-
Optimal treatment has not been well-defined, but use of immu- induced autoimmune syndromes:  two distinct clinical presentations
nosuppression is warranted when vital organs are threatened, with different course and management. Seminars in Arthritis and
Rheumatism, 36, 4–9.
and glucocorticoids are considered first-line therapy. Disease
Anderson, W.R. (1965). Necrotizing angiitis associated with embolization of
flares are common upon glucocorticoid tapering and azatho-
cholesterol. Case report, with emphasis on the use of the muscle biopsy
prine, methotrexate, and mycophenolate mofetil have been used as a diagnostic aid. American Journal of Clinical Pathology, 43, 65–71.
as glucocorticoid-sparing agents. Rituximab may be effective for Anderson, W.R. and Richards, A.M. (1968). Evaluation of lower extrem-
patients with refractory disease. Treatment is likely to be more suc- ity muscle biopsies in the diagnosis of atheroembolism. Archives of
cessful when initiated early, prior to the development of extensive Pathology, 86, 535–41.
fibrosis (Stone et al. 2012). Angelis, M., Wong, L.L., Myers, S.A., and Wong, L.M. (1997). Calciphylaxis in
patients on hemodialysis: a prevalence study. Surgery, 122, 1083–9; 1089–90.
Non-vascular diseases Anonymous (1990). Anderson-Fabry disease. Lancet, 336, 24–5.
Non-vascular diseases of almost any organ can simulate vasculitis Armstrong, F.S. and Hayes, G.J. (1961). Segmental cerebral arterial constric-
(Rosenstein and Kramer 1994). For example, occult subacute thy- tion associated with pheochromocytoma. Report of a case with arterio-
roiditis can masquerade as giant cell arteritis, and antineutrophil grams. Journal of Neurosurgery, 18, 843–6.
cytoplasmic antibody can accompany inflammatory bowel disease Asada, N., Odawara, J., Kimura, S., et al. (2007). Use of random skin biopsy
for diagnosis of intravascular large B-cell lymphoma. Mayo Clinic
(Stebbing et al. 1999) or infectious pulmonary processes (Byrd et al.
Proceedings, 82, 1525–7.
1998). Keep in mind also that ‘imitators’ may accompany true vas-
Asherson, R.A., Cervera, R., Piette, J.C., et al. (1998). Catastrophic antiphos-
culitis. Thus, pulmonary nodules or cavitary lesions in a patient pholipid syndrome. Clinical and laboratory features of 50 patients.
with granulomatosis with polyangiitis (Wegener’s) or polyarteri- Medicine (Baltimore), 77, 195–207.
tis can just as easily result from infection as from vasculitis. And Auethavekiat, P., Murali, N.S., and Manek, N.J. (2004). Clinical
finally, non-vascular cutaneous conditions, such as those shown in images: Primary systemic amyloidosis masquerading as necrotizing vas-
Table 44.5, can simulate vasculitis. culitis. Arthritis and Rheumatism, 50, 3400.
CHAPTER 44 imitators of vasculitis 647

Baethge, B.A., Sanusi, I.D., Landreneau, M.D., Rohr, M.S., and Mcdonald, Brick, J.F. (1988). Vanishing cerebrovascular disease of pregnancy. Neurology,
J.C. (1988). Livedo reticularis and peripheral gangrene associated with 38, 804–6.
primary hyperoxaluria. Arthritis and Rheumatism, 31, 1199–203. Bridges, B.F. and Hector, D.A. (1989). Possible association of cutaneous
Banikazemi, M., Bultas, J., Waldek, S., et  al. (2007). Agalsidase-beta ther- telangiectasia with cardiac myxoma. American Journal of Medicine,
apy for advanced Fabry disease: a randomized trial. Annals of Internal 87, 483–5.
Medicine, 146, 77–86. Bruns, F.J., Segel, D.P., and Adler, S. (1978). Control of cholesterol emboliza-
Bardwick, P.A., Zvaifler, N.J., Gill, G.N., Newman, D., Greenway, G.D., and tion by discontinuation of anticoagulant therapy. American Journal of
Resnick, D.L. (1980). Plasma cell dyscrasia with polyneuropathy, orga- the Medical Sciences, 275, 105–8.
nomegaly, endocrinopathy, M protein, and skin changes: the POEMS Bulkley, B.H. and Hutchins, G.M. (1979). Atrial myxomas: a fifty year review.
syndrome. Report on two cases and a review of the literature. Medicine, American Heart Journal, 97, 639–43.
59, 311–22. Burruss, J.B., Fabrae, V.C., and Callen, J.P. (1994). Painful acral purpuric nod-
Baselga, E., Margall, N., Barnadas, M.A., Coll, P., and Demoragas, J.M. ules. Arthritis and Rheumatism, 37, 1812–5.
(1997). Detection of Mycobacterium tuberculosis in lobular granuloma- Byrd, R., Hourany, J., Cooper, C., and Roy, T.M. (1998). False-positive anti-
tous panniculitis (erythema induratum-nodular vasculitis). Archives of neutrophil cytoplasmic antibodies in a patient with cavitary pulmonary
Dermatology, 133, 457–62. sporotrichosis. American Journal of Medicine, 104, 101–3.
Bastani, B. and Nahass, G. (1999). Images in clinical medicine. Type I pri- Byrd, W.E., Matthews, O.P., and Hunt, R.E. (1980). Left atrial myxoma pre-
mary hyperoxaluria. New England Journal of Medicine, 341, 1979. senting as a systemic vasculitis. Arthritis and Rheumatism, 23, 240–3.
Becker, A.E., Schoorl, R., Balk, A.G., and Van Der Heide, R.M. (1975). Calabrese, L.H., Dodick, D.W., Schwedt, T.J., and Singhal, A.B. (2007).
Cardiac manifestations of Fabry’s disease. Report of a case with mitral Narrative review:  reversible cerebral vasoconstriction syndromes.
insufficiency and electrocardiographic evidence of myocardial infarc- Annals of Internal Medicine, 146, 34–44.
tion. American Journal of Cardiology, 36, 829–35. Calabrese, L.H., Furlan, A.J., Gragg, L.A., and Ropos, T.J. (1992). Primary
Beightler, E., Diven, D.G., Sanchez, R.L., and Solomon, A.R. (1991). angiitis of the central nervous system: diagnostic criteria and clinical
Thrombotic vasculopathy associated with cryofibrinogenemia. Journal approach. Cleveland Clinic Journal of Medicine, 59, 293–306.
of the American Academy of Dermatology, 24, 342–5. Calabrese, L.H. and Mallek, J.A. (1988). Primary angiitis of the central nerv-
Benedict, L., Kusne, S., Torre-Cisneros, J., and Hunt, S.J. (1992). Primary ous system. Report of 8 new cases, review of the literature, and proposal
cutaneous fungal infection after solid-organ transplantation: report of for diagnostic criteria. Medicine, 67, 20–39.
five cases and review. Clinical Infectious Diseases, 15, 17–21. Call, G.K., Fleming, M.C., Sealfon, S., Levine, H., Kistler, J.P., and Fisher,
Blackburn, W.E., Mcroberts, J.W., Bhathena, D., Vazquez, M., and Luke, R.G. C.M. (1988). Reversible cerebral segmental vasoconstriction. Stroke, 19,
(1975). Severe vascular complications in oxalosis after bilateral nephrec- 1159–70.
tomy. Annals of Internal Medicine, 82, 44–6. Camps-Garcia, T., Pedro, I.P., Gomez, I.M., Narankiewicz, D., Ayala-Gutierrez,
Blanco, F., Blas, M.S., and Gonzalez, M.F. (1999). Histopathologic features M., and Sanz-Trelles, A. (2011). Clinical images:  cutaneous necrotiz-
of cerebral vasculitis associated with Mycobacterium tuberculosis. ing vasculitis in a patient with lepromatous leprosy. Arthritis and
Arthritis and Rheumatism, 42, 383. Rheumatism, 63, 3639.
Blumberg, A. and Weidmann, P. (1977). Successful treatment of ischaemic Caplan, D. and Louis, D. (1996). Case records of the Massacusetts General
ulceration of the skin in azotaemic hyperparathyroidism with parathy- Hospital. New England Journal of Medicine, 335, 952–9.
roidectomy. British Medical Journal, 1, 552–3. Cappiello, R.A., Espinoza, L.R., Adelman, H., Aguilar, J., Vasey, F.B., and
Boers, G.H., Smals, A.G., Trijbels, F.J., et al. (1985). Heterozygosity for homo- Germain, B.F. (1989). Cholesterol embolism:  a pseudovasculitic syn-
cystinuria in premature peripheral and cerebral occlusive arterial dis- drome. Seminars in Arthritis and Rheumatism, 18, 240–6.
ease. New England Journal of Medicine, 313, 709–15. Carney, J.A., Gordon, H., Carpenter, P.C., Shenoy, B.V., and Go, V.L. (1985).
Bogousslavsky, J., Despland, P.A., Regli, F., and Dubuis, P.Y. (1989). The complex of myxomas, spotty pigmentation, and endocrine overac-
Postpartum cerebral angiopathy: reversible vasoconstriction assessed by tivity. Medicine, 64, 270–83.
transcranial Doppler ultrasounds. European Neurology, 29, 102–5. Carr, M.E. Jr, Sanders, K., and Todd, W.M. (1994). Pain relief and clinical
Bommer, J., Ritz, E., and Andrassy, K. (1979). Necrotizing dermatitis result- improvement temporally related to the use of pentoxifylline in a patient
ing from hemodialysis with polyvinylchloride tubing. Annals of Internal with documented cholesterol emboli—a case report. Angiology, 45, 65–9.
Medicine, 91, 869–70. Casey, M., Vaughan, C.J., He, J., et al. (2000). Mutations in the protein kinase
Boogaerts, M.A., Hammerschmidt, D.E., Roelant, C., Verwilghen, R.L., A  R1alpha regulatory subunit cause familial cardiac myxomas and
and Jacob, H.S. (1983). Mechanisms of vascular damage in gout and Carney complex. Journal of Clinical Investigation, 106, R31–8.
oxalosis:  crystal induced, granulocyte mediated, endothelial injury. Cattan, S., Mariette, X., Labrousse, F., and Brouet, J.C. (1994). Iliac artery dis-
Thrombosis and Haemostasis, 50, 576–80. section in alpha 1-antitrypsin deficiency. Lancet, 343, 1371–2.
Bose, N. and Calabrese, L. (2012). Recognizing rheumatologic aspects of Chan, R., Goodman, T.A., Aretz, T.H., and Lie, J.T. (1998). Segmental medio-
cocaine abuse. Journal of Musculoskeletal Medicine, 29, 34–40. lytic arteriopathy of the splenic and hepatic arteries mimicking systemic
Boussen, K., Moalla, M., Blondeau, P., Ben Ayed, H., and Lie, J.T. (1991). necrotizing vasculitis. Arthritis and Rheumatism, 41, 935–8.
Embolization of cardiac myxomas masquerading as polyarteritis Chandrasekar, P., Nandi, P.S., Fairfax, M.R., and Crane, L.R. (1997). Cutaneous
nodosa. Journal of Rheumatology, 18, 283–5. infections due to Acanthamoeba in patients with acquired immunodefi-
Bowles, C.A. (1990). Vasculopathy associated with the antiphospholipid ciency syndrome. Archives of Internal Medicine, 157, 569–72.
antibody syndrome. Rheumatic Diseases Clinics of North America, 16, Chen, Y.X., Zhang, W., Chen, X.N., et al. (2012). Propylthiouracil-induced
471–90. antineutrophil cytoplasmic antibody (ANCA)-associated renal vascu-
Bradley, M. (1995). Images in clinical medicine. Spontaneous atheroembo- litis versus primary ANCA-associated renal vasculitis:  a comparative
lism. New England Journal of Medicine, 332, 998. study. Journal of Rheumatology, 39, 558–63.
Bramkamp, M., Rock, T., Schneemann, M., Binggeli, C., and Kucher, N. Churchill, C.H., Abril, A., Krishna, M., Callman, M.L., and Ginsburg, W.W.
(2007). Radiotherapy and the heart. Lancet, 369, 1762. (2003). Jaw claudication in primary amyloidosis: unusual presentation
Breathnach, S.M. and Wells, G.C. (1980). Amyloid vascular disease: cord-like of a rare disease. Journal of Rheumatology, 30, 2283–6.
thickening of mucocutaneous arteries, intermittent claudication and Citron, B.P., Halpern, M., McCarron, M., et  al. (1970). Necrotizing angii-
angina in a case with underlying myelomatosis. British Journal of tis associated with drug abuse. New England Journal of Medicine, 283,
Dermatology, 102, 591–5. 1003–11.
648 SECTION 6 mimickers of vasculitis

Clarke, P., Glick, S., and Reilly, B.M. (2005). Clinical problem-solving. On the Esterly, J. and Oppenheimer, E. (1967). Vascular lesions in infants with con-
threshold—a diagnosis of exclusion. New England Journal of Medicine, genital rubella. Circulation, 36, 544–50.
352, 919–24. Fain, O., Hamidou, M., Cacoub, P., et al. (2007). Vasculitides associated with
Clarke, R., Daly, L., Robinson, K., et al. (1991). Hyperhomocysteinemia: an malignancies: analysis of sixty patients. Arthritis and Rheumatism, 57,
independent risk factor for vascular disease. New England Journal of 1473–80.
Medicine, 324, 1149–55. Falanga, V., Fine, M.J., and Kapoor, W.N. (1986). The cutaneous manifesta-
Conn, J., Jr, Krumlovsky, F.A., Del Greco, F., and Simon, N.M. (1973). tions of cholesterol crystal embolization. Archives of Dermatology, 122,
Calciphylaxis:  etiology of progressive vascular calcification and gan- 1194–8.
grene? Annals of Surgery, 177, 206–10. Fallis, R. and Fisher, M. (1985). Cerebral vasculitis and hemmorrhage associ-
Cosio, F.G., Zager, R.A., and Sharma, H.M. (1985). Atheroembolic renal dis- ated with phenylpropanolamine. Neurology, 35, 405–7.
ease causes hypocomplementaemia. Lancet, 2, 118–21. Federman, D.G., Valdivia, M., and Kirsner, R.S. (1994). Syphilis presenting
Cox, D.W. (1994). Alpha 1-antitrypsin: a guardian of vascular tissue. Mayo as the ‘blue toe syndrome’. Archives of Internal Medicine, 154, 1029–31.
Clinic Proceedings, 69, 1123–4. Fengler, S.A., Berenberg, J.L., and Lee, Y.T. (1990). Disseminated coagulopa-
Cross, S.S. (1991). How common is cholesterol embolism? Journal of Clinical thies and advanced malignancies. American Surgeon, 56, 335–8.
Pathology, 44, 859–61. Fessler, B.J. (1997). Thrombotic syndromes and autoimmune diseases.
Dahlberg, P.J., Frecentese, D.F., and Cogbill, T.H. (1989). Cholesterol embo- Rheumatic Diseases Clinics of North America, 23, 461–79.
lism:  experience with 22 histologically proven cases. Surgery, 105, Fine, A. and Zacharias, J. (2002). Calciphylaxis is usually non-ulcerating: risk
737–46. factors, outcome and therapy. Kidney International, 61, 2210–7.
Darsee, J.R. (1979). Cholesterol embolism: the great masquerader. Southern Fine, M.J., Kapoor, W., and Falanga, V. (1987). Cholesterol crystal embo-
Medical Journal, 72, 174–80. lization: a review of 221 cases in the English literature. Angiology, 38,
Deshpande, V., Zen, Y., Chan, J.K., et al. (2012). Consensus statement on the 769–84.
pathology of IgG4-related disease. Modern Pathology, 25, 1181–92. Finkel, T.H., Teoreok, T.J., Ferguson, P.J., et al. (1994). Chronic parvovirus
Desnick, R.J., Astrin, K.H., and Bishop, D.F. (1989). Fabry disease: molecular B19 infection and systemic necrotising vasculitis: opportunistic infec-
genetics of the inherited nephropathy. Advances in Nephrology from the tion or aetiological agent? Lancet, 343, 1255–8.
Necker Hospital, 18, 113–27. Finley, J.L. and Dabbs, D.J. (1988). Renal vascular smooth muscle prolifera-
Desnick, R.J., Brady, R., Barranger, J., et  al. (2003). Fabry disease, an tion in neurofibromatosis. Human Pathology, 19, 107–10.
under-recognized multisystemic disorder: expert recommendations for Fisher, E.A., Desnick, R.J., Gordon, R.E., Eng, C.M., Griepp, R., and Goldman,
diagnosis, management, and enzyme replacement therapy. Annals of M.E. (1992). Fabry disease: an unusual cause of severe coronary disease
Internal Medicine, 138, 338–46. in a young man. Annals of Internal Medicine, 117, 221–3.
Desousa, A.L., Muller, J., Campbell, R., Batnitzky, S., and Rankin, L. (1978). Atrial Fitzpatrick, A.P., Lanham, J.G., and Doyle, D.V. (1986). Cardiac tumours
myxoma: a review of the neurological complications, metastases, and recur- simulating collagen vascular disease. British Heart Journal, 55, 592–5.
rences. Journal of Neurology, Neurosurgery and Psychiatry, 41, 1119–24. Fonkalsrud, E.W., Sanchez, M., Zerubavel, R., and Mahoney, A. (1977).
Dessailloud, R., Papo, T., Vaneecloo, S., Gamblin, C., Vanhille, P., and Piette, Serial changes in arterial structure following radiation therapy. Surgery,
J.C. (1998). Acalculous ischemic gallbladder necrosis in the catastrophic Gynecology and Obstetrics, 145, 395–400.
antiphospholipid syndrome. Arthritis and Rheumatism, 41, 1318–20. Fontana, P., Gabutti, L., Piffaretti, J.C., and Marone, C. (1996). Antibiotic
Detsky, M.E., Chiu, L., Shandling, M.R., Sproule, M.E., and Ursell, M.R. treatment for giant cell arteritis? Lancet, 348, 1630.
(2006). Clinical problem-solving. Heading down the wrong path. New Fox, D.A., Faix, J.D., Coblyn, J., Fraser, P., Smith, B., and Weinblatt, M.E.
England Journal of Medicine, 355, 67–74. (1986). Thrombotic thrombocytopenic purpura and systemic lupus ery-
DiGiuseppe, J., Nelson, W.G., Seifter, E.J., Boitnott, J.K., and Mann, R.B. thematosus. Annals of the Rheumatic Diseases, 45, 319–22.
(1994). Intravascular lymphomatosis:  a clinicopatgologic study of 10 Frances, C., Niang, S., Laffitte, E., Pelletier, F., Costedoat, N., and Piette,
casesand assessment of response to chemotherapy. Journal of Clinical J.C. (2005). Dermatologic manifestations of the antiphospholipid syn-
Oncology, 12, 2573–9. drome: two hundred consecutive cases. Arthritis and Rheumatism, 52,
Diprete, D.A., Abuelo, J.G., Abuelo, D.N., and Cronan, J.J. (1990). Acute renal 1785–93.
insufficiency due to renal infarctions in a patient with neurofibromato- Frayha, R.A. (1981). Trichinosis-related polyarteritis nodosa. American
sis. American Journal of Kidney Diseases, 15, 357–60. Journal of Medicine, 71, 307–12.
Dornan, T.L., Blundell, J.W., Morgan, A.G., Burden, R.P., Reeves, W.G., and Fredericks, R., Lefkowitz, D.S., Challa, V.R., and Troost, B.T. (1991a).
Cotton, R.E. (1985). Widespread crystallisation of paraprotein in myelo- Cerebral Vasculitis associated with cocaine abuse. Stroke, 22, 1437–9.
matosis. Quarterly Journal of Medicine, 57, 659–67. Fredericks, R., Walker, F.O., Elster, A., and Challa, V. (1991b). Angiotropic
Dotten, D.A., Pruzanski, W., Olin, J., and Brown, T.C. (1976). intravascular large-cell lymphoma (malignant angioendotheliomato-
Cryocrystalglobulinemia. Canadian Medical Association Journal, 114, sis): report of a case and review of the literature. Surgical Neurology, 35,
909–12. 218–23.
Dowd, P.M., Rustin, M.H., and Lanigan, S. (1986). Nifedipine in the treat- Friedman, D.R. and Wolfsthal, S.D. (2005). Cocaine-induced pseudovasculi-
ment of chilblains. British Medical Journal Clinical Research Edition, tis. Mayo Clinic Proceedings, 80, 671–3.
293, 923–4. Friman, C. and Pettersson, T. (1996). Amyloidosis. Current Opinion in
Ducros, A. (2012). Reversible cerebral vasoconstriction syndrome. Lancet Rheumatology, 8, 62–71.
Neurology, 11, 906–17. Fukumoto, Y., Tsutsui, H., Tsuchihashi, M., Masumoto, A., and Takeshita, A.
Eliot, R.S., Kanjuh, V.I., and Edwards, J.E. (1964). Atheromatous embolism. (2003). The incidence and risk factors of cholesterol embolization syn-
Circulation, 30, 611–8. drome, a complication of cardiac catheterization: a prospective study.
Eng, C.M., Guffon, N., Wilcox, W.R., et  al. (2001). Safety and efficacy of Journal of the American College of Cardiology, 42, 211–6.
recombinant human alpha-galactosidase A—replacement therapy in Gabriel, S.E., Conn, D.L., Phyliky, R.L., Pittelkow, M.R., and Scott, R.E.
Fabry’s disease. New England Journal of Medicine, 345, 9–16. (1986). Vasculitis in hairy cell leukemia: review of literature and con-
Erhard, H., Runger, T.M., Kreienkamp, M., Muller, J., Muller-Hermelink, sideration of possible pathogenic mechanisms. Journal of Rheumatology,
H.K., et al. (1995). Atypical varicella-zoster virus infection in an immu- 13, 1167–72.
nocompromised patient: result of a virus-induced vasculitis. Journal of Ganor, S. (1983). Corticosteroid therapy for pernio. Journal of the American
the American Academy of Dermatology, 32, 908–11. Academy of Dermatology, 8, 136.
CHAPTER 44 imitators of vasculitis 649

Garner, B.F., Burns, P., Bunning, R.D., and Laureno, R. (1990). Acute blood antiphospholipid antibody syndrome. Journal of the American Academy
pressure elevation can mimic arteriographic appearance of cerebral of Dermatology, 24, 53–8.
vasculitis—a postpartum case with relative hypertension. Journal of Grossman, J., Abraham, G.N., Leddy, J.P., and Condemi, J.L. (1972).
Rheumatology, 17, 93–7. Crystalglobulinemia. Annals of Internal Medicine, 77, 395–400.
Gertner, E. (1999). Diffuse alveolar hemorrhage in the antiphospholipid syn- Gruber, B., Schranz, J.A., Fuhrer, J., and Kane, P.B. (1997). Isolated pulmo-
drome:  spectrum of disease and treatment. Journal of Rheumatology, nary microangiitis mimicking pneumonia in a patient infected with
26, 805–7. human immunodeficiency virus. Journal of Rheumatology, 24, 759–62.
Gertner, E. and Lie, J.T. (1994). Systemic therapy with fibrinolytic agents and Gulati, S. and Donato, A.A. (2012). Lupus anticoagulant and ANCA asso-
heparin for recalcitrant nonhealing cutaneous ulcer in the antiphospho- ciated thrombotic vasculopathy due to cocaine contaminated with lev-
lipid syndrome. Journal of Rheumatology, 21, 2159–61. amisole: a case report and review of the literature. Journal of Thrombosis
Gibson, A., Gardner, J., and O’Donnell, J. (2005). Erythropoietin and painful Thrombolysis, 34, 7–10.
leg ulcers: thrombosis or vasculitis? Arthritis and Rheumatism, 53, 792. Gupta, B.K., Spinowitz, B.S., Charytan, C., and Wahl, S.J. (1993). Cholesterol
Gilbert, J.A. Jr and Scalzi, R.P. (1993). Disseminated intravascular coagula- crystal embolization-associated renal failure after therapy with recom-
tion. Emergency Medicine Clinics of North America, 11, 465–80. binant tissue-type plasminogen activator. American Journal of Kidney
Gilbert, J.J. and Vinters, H.V. (1983). Cerebral amyloid angiopathy: incidence Diseases, 21, 659–62.
and complications in the aging brain. I. Cerebral hemorrhage. Stroke, Gupta, S., Ries, M., Kotsopoulos, S., and Schiffmann, R. (2005). The rela-
14, 915–23. tionship of vascular glycolipid storage to clinical manifestations of Fabry
Gipstein, R.M., Coburn, J.W., Adams, D.A., et  al. (1976). Calciphylaxis in disease: a cross-sectional study of a large cohort of clinically affected het-
man. A  syndrome of tissue necrosis and vascular calcification in 11 erozygous women. Medicine (Baltimore), 84, 261–8.
patients with chronic renal failure. Archives of Internal Medicine, 136, Halasz, C.L. and Strauss, E.B. (1998). Images in clinical medicine. Unilateral
1273–80. livedo reticularis. New England Journal of Medicine, 338, 1127.
Glass, J., Hochberg, F.H., and Miller, D.C. (1993). Intravascular lymphoma- Hammerschmidt, D.E., Greenberg, C.S., Yamada, O., Craddock, P.R., and
tosis. Cancer, 71, 3156–64. Jacob, H.S. (1981). Cholesterol and atheroma lipids activate comple-
Goldberger, E., Elder, R.C., Schwartz, R.A., and Phillips, P.E. (1992). Vasculitis ment and stimulate granulocytes. A possible mechanism for amplifica-
in the antiphospholipid syndrome. A cause of ischemia responding to tion of ischemic injury in atherosclerotic states. Journal of Laboratory
corticosteroids. Arthritis and Rheumatism, 35, 569–72. and Clinical Medicine, 98, 68–77.
Golden, M.P., Hammer, S.M., Wanke, C.A., and Albrecht, M.A. (1994). Harvey, W.P. (1968). Clinical aspects of cardiac tumors. American Journal of
Cytomegalovirus vasculitis. Case reports and review of the literature. Cardiology, 21, 328–43.
Medicine, 73, 246–55. Hendel, R.C., Cuenoud, H.F., Giansiracusa, D.F., and Alpert, J.S. (1989).
Goldman, B.I., Frydman, C., Harpaz, N., Ryan, S.F., and Loiterman, D. Multiple cholesterol emboli syndrome. Bowel infarction after retrograde
(1987). Glandular cardiac myxomas. Histologic, immunohistochemi- angiography. Archives of Internal Medicine, 149, 2371–4.
cal, and ultrastructural evidence of epithelial differentiation. Cancer, 59, Henry, P.Y., Larre, P., Aupy, M., Lafforgue, J.L., and Orgogozo, J.M. (1984).
1767–75. Reversible cerebral arteriopathy associated with the administration of
Goldman, M., Goldman, A., Dereume, J.P., Heenen, M., Appelboom, T., and ergot derivatives. Cephalalgia, 4, 171–8.
Bellens, R. (1980). Aortic atheromatosis presenting as a cutaneous vas- Herman, E.W., Kezis, J.S., and Silvers, D.N. (1981). A distinctive variant of
culitis with antinuclear antibody. Arthritis and Rheumatism, 23, 1407–8. pernio. Clinical and histopathologic study of nine cases. Archives of
Goldstein, D.A., Timpone, J., and Cupps, T.R. (2010). HIV-associated intrac- Dermatology, 117, 26–8.
ranial aneurysmal vasculopathy in adults. Journal of Rheumatology, 37, Hogarth, M.B., Qureshi, T., Lloyd, J., and Rees, R.G. (1999). A blistering rash
226–33. and swollen knees. Lancet, 353, 978.
Graf, J., Lynch, K., Yeh, C.L., et al. (2011). Purpura, cutaneous necrosis, and anti- Hoshimaru, M., Takahashi, J.A., Kikuchi, H., Nagata, I., and Hatanaka, M.
neutrophil cytoplasmic antibodies associated with levamisole-adulterated (1991). Possible roles of basic fibroblast growth factor in the pathogen-
cocaine. Arthritis and Rheumatism, 63, 3998–4001. esis of moyamoya disease: an immunohistochemical study. Journal of
Graham, I.M., Daly, L.E., Refsum, H.M., et al. (1997). Plasma homocysteine Neurosurgery, 75, 267–70.
as a risk factor for vascular disease. The European Concerted Action Huang, T. and Chou, S. (1988). Occlusive hypertrophic arteritis as the cause
Project. JAMA, 277, 1775–81. of discrete necrosis in CNS toxoplasmosis in the acquired immunodefi-
Grange, D.K., Balfour, I.C., Chen, S.C., and Wood, E.G. (1998). Familial syn- ciency syndrome. Human Pathology, 19, 1210–4.
drome of progressive arterial occlusive disease consistent with fibromus- Hughes, G.R. (1993). The antiphospholipid syndrome: ten years on. Lancet,
cular dysplasia, hypertension, congenital cardiac defects, bone fragility, 342, 341–4.
brachysyndactyly, and learning disabilities. American Journal of Medical Hyman, B.T., Landas, S.K., Ashman, R.F., Schelper, R.L., and Robinson, R.A.
Genetics, 75, 469–80. (1987). Warfarin-related purple toes syndrome and cholesterol micro-
Gravallese, E., Waksmonski, C., Winters, G.L., and Simms, R.W. (1995). embolization. American Journal of Medicine, 82, 1233–7.
Fever, arthralgias, skin lesions, and ischemic digits in a 59-year old man. Imahori, S., Bannerman, R.M., Graf, C.J., and Brennan, J.C. (1969).
Clinicopathologic conference. Arthritis and Rheumatism, 38, 1161–8. Ehlers-Danlos syndrome with multiple arterial lesions. American
Gray, F., Belec, L., Lescs, M.C., Chretien, F., Ciardi, A., Hassine, D., et  al. Journal of Medicine, 47, 967–77.
(1994). Varicell-zoster virus infection of the central nervous system in Ingram, S.B., Goodnight, S.H. Jr, and Bennett, R.M. (1987). An unusual
the acquired immune deficiency syndrome. Brain, 117, 987–99. syndrome of a devastating noninflammatory vasculopathy associ-
Greene, R.R. (2005). Clinical images:  Afrin-induced central nervous sys- ated with anticardiolipin antibodies: report of two cases. Arthritis and
tem vasospasm and thunderclap headache. Arthritis and Rheumatism, Rheumatism, 30, 1167–72.
52, 3314. Irey, N.S. and Norris, H.J. (1973). Intimal vascular lesions associated with
Greenwood, W.F. (1968). Profile of atrial myxoma. American Journal of female reproductive steroids. Archives of Pathology, 96, 227–34.
Cardiology, 21, 367–75. Jacob, J.R., Weisman, M.H., Rosenblatt, S.I., and Bookstein, J.J. (1986).
Greitz, T. (1964). Angiography in tuberculous meningitis. Acta Radiologica, Chronic pernio. A historical perspective of cold-induced vascular dis-
2, 369–78. ease. Archives of Internal Medicine, 146, 1589–92.
Grob, J.J., San Marco, M., Aillaud, M.F., et al. (1991). Unfading acral micro- Jacobs-Kosmin, D. and Dehoratius, R.J. (2007). Calciphylaxis: an important
livedo. A  discrete marker of thrombotic skin disease associated with imitator of cutaneous vasculitis. Arthritis and Rheumatism, 57, 533–7.
650 SECTION 6 mimickers of vasculitis

Jacobs-Kosmin, D., Derk, C.T., and Sandorfi, N. (2006). Pantoprazole and use: a single-center case series. Seminars in Arthritis and Rheumatism,
perinuclear antineutrophil cytoplasmic antibody-associated vasculitis. 42, 213–21.
Journal of Rheumatology, 33, 629–32. Kinugasa, K., Mandai, S., Kamata, I., Sugiu, K., and Ohmoto, T. (1993).
Jain, R., Chartash, E., Susin, M., and Furie, R. (1994). Systemic lupus ery- Surgical treatment of moyamoya disease:  operative technique for
thematosus complicated by thrombotic microangiopathy. Seminars in encephalo-duro-arterio-myo-synangiosis, its follow-up, clinical results,
Arthritis and Rheumatism, 24, 173–82. and angiograms. Neurosurgery, 32, 527–31.
Jarrett, S.J., Cunnane, G., Conaghan, P.G., et al. (2003). Anti-tumor necro- Kirsner, R.S., Eaglstein, W.H., Katz, M.H., Kerdel, F.A., and Falanga, V.
sis factor-alpha therapy-induced vasculitis:  case series. Journal of (1993). Stanozolol causes rapid pain relief and healing of cutaneous
Rheumatology, 30, 2287–91. ulcers caused by cryofibrinogenemia. Journal of the American Academy
Jennette, J.C., Sheps, D.S., and Mcneill, D.D. (1982). Exclusively vascular of Dermatology, 28, 71–4.
systemic amyloidosis with visceral ischemia. Archives of Pathology and Klein, P. (1986). Ocular manifestations of Fabry’s disease. Journal of the
Laboratory Medicine, 106, 323–7. American Optometric Association, 57, 672–4.
Jones, H. and Edgar, M. (1995). Case records of the Massachusetts General Kleinschmidt-Demasters, B., Filley, C.M., and Bitter, M.A. (1992). Central
Hospital. New England Journal of Medicine, 332, 730–7. nervous system angiocentric, angiodestructive T-cell lymphoma (lym-
Jones, H.J., Staud, R., and Williams, R.C. Jr (1998). Rupture of a hepatic artery phomatoid granulomatosis). Surgical Neurology, 37, 130–7.
aneurysm and renal infarction: 2 complications of fibromuscular dyspla- Kois, J.M., Sexton, F.M., and Lookingbill, D.P. (1991). Cutaneous manifesta-
sia that mimic vasculitis. Journal of Rheumatology, 25, 2015–8. tions of multiple myeloma. Archives of Dermatology, 127, 69–74.
Joshi, V., Pawel, B., Connor, E., Sharer, L., Oleske, J.M., et  al. (1987). Korst, D.R. and Kratochvil, C.H. (1955). ‘Cryofibrinogen’ formation in a case
Arteriopathy in children with acquired immune deficiency syndrome. of lung neoplasm associated with thrombophlebitis migrans. Blood, 10,
Pediatric Pathology, 7, 261–75. 945–53.
Judge, D.P. and Dietz, H.C. (2005). Marfan’s syndrome. Lancet, 366, 1965–76. Kraus, A., Valencia, X., Cabral, A.R., and De La Vega, G. (1995). Visceral
Kakumani, P.L. and Hajj-Ali, R.A. (2009). A forgotten cause of central nerv- larva migrans mimicking rheumatic diseases. Journal of Rheumatology,
ous system vasculitis. Journal of Rheumatology, 36, 655. 22, 497–500.
Kalter, D.C., Rudolph, A., and Mcgavran, M. (1985). Livedo reticularis due Kuiper, J.J. (1996). Initial manifestation of primary hyperoxaluria type I in
to multiple cholesterol emboli. Journal of the American Academy of adults—recognition, diagnosis, and management. Western Journal of
Dermatology, 13, 235–42. Medicine, 164, 42–53.
Kamesaki, H., Matsui, Y., Ohno, Y., et al. (1990). Angiotropic lymphoma with Kullo, I.J., Edwards, W.D., and Schwartz, R.S. (1998). Vulnerable
histologic features of neoplastic angioendotheliomatosis presenting with plaque:  pathobiology and clinical implications. Annals of Internal
predominant respiratory and hematologic manifestations. Report of a Medicine, 129, 1050–60.
case and review of the literature. American Journal of Clinical Pathology, Lake, C.R., Gallant, S., Masson, E., and Miller, P. (1990). Adverse drug
94, 768–72. effects attributed to phenylpropanolamine: a review of 142 case reports.
Kaminsky, M.E., Ehlers, K.H., Engle, M.A., Klein, A.A., Levin, A.R., and American Journal of Medicine, 89, 195–208.
Subramanian, V.A. (1979). Atrial myxoma mimicking a collagen disor- Lanczik, O., Szabo, K., Gass, A., and Hennerici, M.G. (2003). Tinnitus after
der. Chest, 75, 93–5. cycling. Lancet, 362, 292.
Kanno, J., Takemura, T., and Kasaga, T. (1987). Malignant endothelioma of Landon, G., Ordaoanez, N.G., and Guarda, L.A. (1986). Cardiac myxo-
the aorta. Virchows Archiv A, 412, 183–8. mas. An immunohistochemical study using endothelial, histiocytic,
Kanzaki, T., Yokota, M., Irie, F., Hirabayashi, Y., Wang, A.M., and Desnick, R.J. and smooth-muscle cell markers. Archives of Pathology and Laboratory
(1993). Angiokeratoma corporis diffusum with glycopeptiduria due to defi- Medicine, 110, 116–20.
cient lysosomal alpha-N-acetylgalactosaminidase activity. Clinical, mor- Lebwohl, M., Halperin, J., and Phelps, R.G. (1993). Brief report: occult pseu-
phologic, and biochemical studies. Archives of Dermatology, 129, 460–5. doxanthoma elasticum in patients with premature cardiovascular dis-
Kao, N.L., Broy, S., and Tillawi, I. (1992). Malignant angioendotheliomatosis ease. New England Journal of Medicine, 329, 1237–9.
mimicking systemic necrotizing vasculitis. Journal of Rheumatology, 19, Lee, K.F. and Hodes, P.J. (1967). Intracranial ischemic lesions. Radiologic
1133–5. Clinics of North America, 5, 363–93.
Kapoor, O.P. (1976). Iatrogenic ergot vasospastic angiitis. A  case report. Leeds, N.E. and Goldberg, H.I. (1971). Angiographic manifestations in cer-
Vascular Surgery, 10, 58–60. ebral inflammatory disease. Radiology, 98, 595–604.
Karch, S.B. and Billingham, M.E. (1988). The pathology and etiology of Leeds, N.E. and Rosenblatt, R. (1972). Arterial wall irregularities in intracranial
cocaine-induced heart disease. Archives of Pathology and Laboratory neoplasms. The shaggy vessel brought into focus. Radiology, 103, 121–4.
Medicine, 112, 225–30. Leeds, N.E., Rosenblatt, R., and Zimmerman, H.M. (1971). Focal angio-
Karl, M. (1975). Clinicopathologic conference. American Journal of Medicine, graphic changes of cerebral lymphoma with pathologic correlation.
59, 837–43. A report of two cases. Radiology, 99, 595–9.
Kasinath, B.S. and Lewis, E.J. (1987). Eosinophilia as a clue to the diagnosis of Lehrer, H. (1966). The angiographic triad in tuberculous meningitis. A radio-
atheroembolic renal disease. Archives of Internal Medicine, 147, 1384–5. graphic and clinicopathologic correlation. Radiology, 87, 829–35.
Kawakami, Y., Hirose, K., Watanabe, Y., et al. (1990). Management of mul- Leonhardt, E.T. and Kullenberg, K.P. (1977). Bilateral atrial myxomas with
tiple cholesterol embolization syndrome—a case report. Angiology, 41, multiple arterial aneurysms—a syndrome mimicking polyarteritis
248–52. nodosa. American Journal of Medicine, 62, 792–4.
Kaye, B.R. and Fainstat, M. (1987). Cerebral vasculitis associated with Lesprit, P., Authier, F.J., Gherardi, R., Belec, L., Paris, D., et al. (1996). Acute
cocaine abuse. JAMA, 258, 2104–6. arterial obliteration: a new feature of the POEMS syndrome? Medicine,
Kelly, J.W. and Dowling, J.P. (1985). Pernio. A  possible association with 75, 226–32.
chronic myelomonocytic leukemia. Archives of Dermatology, 121, Lie, J. (1996). Vasculitis associated with infectious agents. Current Opinion in
1048–52. Rheumatology, 8, 26–9.
Kennedy, A., Cumberland, D., and Gaines, P. (1989). The pathology of cho- Lie, J.T. (1992a). Vasculitis simulators and vasculitis look-alikes. Current
lesterol embolism arising as a complication of intra-aortic catheteriza- Opinion in Rheumatology, 4, 47–55.
tion. Histopathology, 15, 515–21. Lie, J.T. (1992b). Malignant angioendotheliomatosis intravascular lympho-
Kermani, T.A., Ham, E.K., Camilleri, M.J., and Warrington, K.J. (2012). matosis clinically simulating primary angiitis of the central nervous sys-
Polyarteritis nodosa-like vasculitis in association with minocycline tem. Arthritis and Rheumatism, 35, 831–4.
CHAPTER 44 imitators of vasculitis 651

Lie, J.T. (1994). Vasculitis in the antiphospholipid syndrome: culprit or con- Mehta, B., Mueller, W., and Efthimiou, F. (2011). CNS vasculitis:  A  pro-
sort? Journal of Rheumatology, 21, 397–9. tean manifestation of HIV/AIDS. Journal of Musculoskeletal Medicine,
Liebeskind, A., Cohen, S., Anderson, R., Schechter, M.M., and Zingesser, 28, 462–4.
L.H. (1973). Unusual segmental cerebrovascular changes. Radiology, Mertz, L.E. and Conn, D.L. (1992). Vasculitis associated with malignancy.
106, 119–22. Current Opinion in Rheumatology, 4, 39–46.
Liebler, G.A., Magovern, G.J., Park, S.B., Cushing, W.J., Begg, F.R., and Joyner, Meschia, J.F., Brott, T.G., and Brown, R.D. Jr (2005). Genetics of cerebrovas-
C.R. (1976). Familial myxomas in four siblings. Journal of Thoracic and cular disorders. Mayo Clinic Proceedings, 80, 122–32.
Cardiovascular Surgery, 71, 605–8. Meyers, D.S., Grim, C.E., and Keitzer, W.F. (1974). Fibromuscular dysplasia
Lipton, S. and Ma, M. (1996). Case records of the Massachusetts General of the renal artery with medial dissection. A case simulating polyarteritis
Hospital. New England Journal of Medicine, 335, 1587–95. nodosa. American Journal of Medicine, 56, 412–6.
Lye, W.C., Cheah, J.S., and Sinniah, R. (1993). Renal cholesterol embolic Millard, L.G. and Rowell, N.R. (1978). Chilblain lupus erythematosus
disease. Case report and review of the literature. American Journal of Hutchinson. A  clinical and laboratory study of 17 patients. British
Nephrology, 13, 489–93. Journal of Dermatology, 98, 497–506.
MacConnell, T. and Ferro, A. (1995). A sore eye and meningitis. Lancet, Milliner, D.S., Eickholt, J.T., Bergstralh, E.J., Wilson, D.M., and Smith, L.H.
346, 1269. (1994). Results of long-term treatment with orthophosphate and pyri-
Mahr, A.D., Edberg, J.C., Stone, J.H., et  al. (2010). Alpha(1)-antitrypsin doxine in patients with primary hyperoxaluria. New England Journal of
deficiency-related alleles Z and S and the risk of Wegener’s granuloma- Medicine, 331, 1553–8.
tosis. Arthritis and Rheumatism, 62, 3760–7. Milman, N. and Smith, C.D. (2011). Cutaneous vasculopathy associated with
Mandel, B. and Calabrese, L. (1998). Infections and systemic vasculitis. cocaine use. Arthritis Care and Research (Hoboken), 63, 1195–202.
Current Opinion in Rheumatology, 10, 51–7. Moldveen-Geronimus, M. and Merriam, J.C. Jr (1967). Cholesterol embo-
Mandybur, T.I. (1986). Cerebral amyloid angiopathy:  the vascular pathol- lization. From pathological curiosity to clinical entity. Circulation, 35,
ogy and complications. Journal of Neuropathology and Experimental 946–53.
Neurology, 45, 79–90. Moolenaar, W. and Lamers, C.B. (1996). Cholesterol crystal embolisation to
Manning, W.J., Goldberger, A.L., Drews, R.E., et al. (1992). POEMS syndrome the alimentary tract. Gut, 38, 196–200.
with myocardial infarction:  observations concerning pathogenesis and Mor, A., Bingham, C., 3rd, Barisoni, L., Lydon, E., and Belmont, H.M. (2005).
review of the literature. Seminars in Arthritis and Rheumatism, 22, 151–61. Proliferative lupus nephritis and leukocytoclastic vasculitis during treat-
Marie, I., Levesque, H., Heron, F., Cailleux, N., Borg, J.Y., and Courtois, H. ment with etanercept. Journal of Rheumatology, 32, 740–3.
(1997). Acute adrenal failure secondary to bilateral infarction of the Mora, C.S., Segami, M.I., and Hidalgo, J.A. (2006). Strongyloides stercoralis
adrenal glands as the first manifestation of primary antiphospholipid hyperinfection in systemic lupus erythematosus and the antiphospho-
antibody syndrome. Annals of the Rheumatic Diseases, 56, 567–8. lipid syndrome. Seminars in Arthritis and Rheumatism, 36, 135–43.
Markel, M.L., Armstrong, W.F., Waller, B.F., and Mahomed, Y. (1986). Left Morgan, S.H., Rudge, P., Smith, S.J., et al. (1990). The neurological compli-
atrial myxoma with multicentric recurrence and evidence of metastases. cations of Anderson-Fabry disease (alpha-galactosidase A deficiency)—
American Heart Journal, 111, 409–13. investigation of symptomatic and presymptomatic patients. Quarterly
Markel, M.L., Waller, B.F., and Armstrong, W.F. (1987). Cardiac myxoma. Journal of Medicine, 75, 491–507.
A review. Medicine, 66, 114–25. Mosnier, J.F., Degott, C., Bedrossian, J., et al. (1991). Recurrence of Fabry’s
Marks, C. and Kuskov, S. (1995). Pattern of arterial aneurysms in acquired disease in a renal allograft eleven years after successful renal transplanta-
immunodeficiency disease. World Journal of Surgery, 19, 127–32. tion. Transplantation, 51, 759–62.
Martens, P., Levine, J.A., and Hunder, G.G. (1996). Splinter hemmorhages Murakami, K., Oshawa, M., Hu, S.X., Kanno, H., and Aozasa, K., et al. (1998).
following arterial puncture. Arthritis and Rheumatism, 39, 169–70. Large-vessel vasculitis associated with chronic active Epstein-Barr virus
Mason, M.S., Wheeler, J.R., Gregory, R.T., and Gayle, R.G. (1982). Primary infection. Arthritis and Rheumatism, 41, 369–73.
tumors of the aorta:  report of a case and review of the literature. Murakawa, G., McCalmont, T., Altman, J., Telang, G.H., Hoffman, M.D.,
Oncology, 39, 167–72. et  al. (1995). Disseminated acanthamebiasis in patients with AIDS.
Mastrolonardo, M., Loconsole, F., Conte, A., and Rantuccio, F. (1994). Archives of Dermatology, 131, 1291–6.
Cutaneous vasculitis as the sole manifestation of disseminated gonococ- Nachman, R.L. and Silverstein, R. (1993). Hypercoagulable states. Annals of
cal infection: case report. Genitourinary Medicine, 70, 130–1. Internal Medicine, 119, 819–27.
Masuda, J., Ogata, J., and Yutani, C. (1993). Smooth muscle cell proliferation Nakao, S., Takenaka, T., Maeda, M., et  al. (1995). An atypical variant of
and localization of macrophages and T cells in the occlusive intracranial Fabry’s disease in men with left ventricular hypertrophy. New England
major arteries in moyamoya disease. Stroke, 24, 1960–7. Journal of Medicine, 333, 288–93.
Masuzawa, T., Shinoda, S., Furuse, M., Nakahara, N., Abe, F., and Sato, F. Naldi, L., Locati, F., Marchesi, L., et  al. (1993). Cutaneous manifestations
(1983). Cerebral angiographic changes on serial examination of a patient associated with antiphospholipid antibodies in patients with suspected
with migraine. Neuroradiology, 24, 277–81. primary antiphospholipid syndrome: a case-control study. Annals of the
Matick, H., Anderson, D., and Brumlik, J. (1983). Cerebral vasculitis associ- Rheumatic Diseases, 52, 219–22.
ated with oral amphetamine overdose. Archives of Neurology, 40, 253–4. Nance, J., Abbott, K., Morris, L., and Couper, R. (1997). An unfortunate con-
May, E.F. and Jabbari, B. (1990). Stroke in neuroborreliosis. Stroke, 21, sequence of being tickled. Lancet, 349, 1142.
1232–5. Nasser, W.K., Davis, R.H., Dillon, J.C., et al. (1972). Atrial myxoma. I. Clinical
Mazhar, A.R., Johnson, R.J., Gillen, D., et al. (2001). Risk factors and mor- and pathologic features in nine cases. American Heart Journal, 83,
tality associated with calciphylaxis in end-stage renal disease. Kidney 694–704.
International, 60, 324–32. Navarro, P.H., Bravo, F.P., and Beltran, G.G. (1995). Atrial myxoma with
McColl, G.J. and Fraser, K. (1995). Pheochromocytoma and pseudovasculi- livedoid macules as its sole cutaneous manifestation. Journal of the
tis. Journal of Rheumatology, 22, 1442–3. American Academy of Dermatology, 32, 881–3.
McCready, R.A., Hyde, G.L., Bivins, B.A., Mattingly, S.S., and Griffen, W.O., Nelson, P., Dignani, M.C., and Anaissie, E.J. (1994). Taxonomy, biology, and
Jr (1983). Radiation-induced arterial injuries. Surgery, 93, 306–12. clinical aspects of Fusarium species. Clinical Microbiology Reviews, 7,
Mehta, A., Beck, M., Elliott, P., et al. (2009). Enzyme replacement therapy 479–504.
with agalsidase alfa in patients with Fabry’s disease: an analysis of regis- Neuteboom, R.F., Lequin, M.H., De Wit, M.C., and Catsman-Berrevoets,
try data. Lancet, 374, 1986–96. C.E. (2011). Lumbar puncture in paediatric stroke. Lancet, 378, 848.
652 SECTION 6 mimickers of vasculitis

Nevelsteen, A., Kutten, M., Lacroix, H., and Suy, R. (1992). Oral anticoagu- woman with paresthesias and ataxia. New England Journal of Medicine,
lant therapy:  a precipitating factor in the pathogenesis of cholesterol 362, 1129–38.
embolization? Acta Chirurgica Belgica, 92, 33–6. Pontes Tde, C., Rufino, G.P., Gurgel, M.G., Medeiros, A.C., and Freire, E.A.
New, P.F., Price, D.L., and Carter, B. (1970). Cerebral angiography in cardiac (2012). Fibromuscular dysplasia:  a differential diagnosis of vasculitis.
myxoma. Correlation of angiographic and histopathological findings. Revista Brasileira de Reumatologia, 52, 70–4.
Radiology, 96, 335–45. Poon, S.H., Baliog, C.R., Jr, Sams, R.N., Robinson-Bostom, L., Telang, G.H.,
Nishida, H., Endo, M., Koyanagi, H., Ichihara, T., Takao, A., and Maruyama, and Reginato, A.M. (2011). Syndrome of cocaine-levamisole-induced
M. (1990). Coronary artery bypass in a 15-year-old girl with pseudoxan- cutaneous vasculitis and immune-mediated leukopenia. Seminars in
thoma elasticum. Annals of Thoracic Surgery, 49, 483–5. Arthritis and Rheumatism, 41, 434–44.
O’Connor, B. (1884). Symmetrical gangrene. British Medical Journal, 1, 460. Prasertwitayakij, N., Louthrenoo, W., Kasitanon, N., Thamprasert, K., and
Oddo, D., Casanova, M., Acuna, G., Ballesteros, J., and Morales, B. (1992). Vanittanakom, N. (2003). Human pythiosis, a rare cause of arteritis: case
Acute Chagas’ disease (Trypanosomiasis americana) in acquired report and literature review. Seminars in Arthritis and Rheumatism, 33,
immune deficiency syndrome: report of two cases. Human Pathology, 204–14.
23, 41–4. Price, D.L., Harris, J.L., New, P.F., and Cantu, R.C. (1970). Cardiac myxoma.
O’Donnell, T.J. and O’Connell, J.X. (1993). Case records of the Massachusetts A clinicopathologic and angiographic study. Archives of Neurology, 23,
General Hospital. Weekly clinicopathological exercises. Case 26-1993. 558–67.
A 73-year-old man with an enlarging inguinal mass 10 years after treat- Primka, E.J.D., King, C., and O’Keefe, E.J. (1993). Malignant melanoma
ment for prostate and colon cancers. New England Journal of Medicine, of unknown origin presenting as a systemic vasculitis. Archives of
329, 43–8. Dermatology, 129, 1205–7.
Okazaki, H., Reagan, T.J., and Campbell, R.J. (1989). Clinicopathologic stud- Provost, T.T., Moses, H., Morris, E.L., et al. (1991). Cerebral vasculopathy
ies of primary cerebral amyloid angiopathy. Mayo Clinic Proceedings, associated with collateralization resembling moya moya phenomenon
54, 22–31. and with anti-Ro/SS-A and anti-La/SS-B antibodies. Arthritis and
O’Keeffe, S.T., Woods, B.O., Breslin, D.J., and Tsapatsaris, N.P. (1992). Blue Rheumatism, 34, 1052–5.
toe syndrome. Causes and management. Archives of Internal Medicine, Rademaker, M., Lowe, D.G., and Munro, D.D. (1989). Erythema indura-
152, 2197–202. tum Bazin’s disease. Journal of the American Academy of Dermatology,
Olivero, J.J. (1997). Case in point. Hospital Practice, 32, 30. 21, 740–5.
Ollert, M.W., Thomas, P., Korting, H.C., Schraut, W., and Braun-Falco, O. Rajpal, R.S., Leibsohn, J.A., Liekweg, W.G., et al. (1979). Infected left atrial
(1993). Erythema induratum of Bazin. Evidence of T-lymphocyte hyper- myxoma with bacteremia simulating infective endocarditis. Archives of
responsiveness to purified protein derivative of tuberculin: report of two Internal Medicine, 139, 1176–8.
cases and treatment. Archives of Dermatology, 129, 469–73. Rao, J.K. and Allen, N.B. (1993). Primary systemic amyloidosis masquer-
Olmsted, W.W. and McGee, T.P. (1977). The pathogenesis of peripheral aneu- ading as giant cell arteritis. Case report and review of the literature.
rysms of the central nervous system: a subject review from the AFIP. Arthritis and Rheumatism, 36, 422–5.
Radiology, 123, 661–6. Read, R.C., White, H.J., Murphy, M.L., Williams, D., Sun, C.N., and Flanagan,
Orbison, J.L. (1952). Morphology of thrombocytopenic purpura with dem- W.H. (1974). The malignant potentiality of left atrial myxoma. Journal of
onstration of aneurysms. American Journal of Pathology, 28, 129–43. Thoracic and Cardiovascular Surgery, 68, 857–68.
Orvar, K. and Johlin, F.C. (1994). Atheromatous embolization resulting in Reiman, S., Fisher, R., Dodds, C., Trinh, C., Laucirica, R., and Whigham,
acute pancreatitis after cardiac catheterization and angiographic studies. C.J. (2002). Mesenteric arteriographic findings in a patient with strongy-
Archives of Internal Medicine, 154, 1755–61. loides stercoralis hyperinfection. Journal of Vascular and Interventional
Page, E.H. and Shear, N.H. (1988). Temperature-dependent skin disorders. Radiology, 13, 635–8.
Journal of the American Academy of Dermatology, 18, 1003–19. Retan, J.W. and Miller, R.E. (1966). Microembolic complications of athero-
Pandey, J. and Leroy, E. (1998). Human cytomegalovirus and the vasculopa- sclerosis. Literature review and report of a patient. Archives of Internal
thies of autoimmune diseases (especially scleroderma), allograft rejec- Medicine, 118, 534–45.
tion, and coronary restenosis. Arthritis and Rheumatism, 41, 10–5. Reynen, K. (1995). Cardiac myxomas. New England Journal of Medicine, 333,
Pearlson, G.D., Jeffery, P.J., Harris, G.J., Ross, C.A., Fischman, M.W., and 1610–7.
Camargo, E.E. (1993). Correlation of acute cocaine-induced changes Roberts, W.N., De Meo, J.H., and Breitbach, S.A. (1994). Well-imaged large
in local cerebral blood flow with subjective effects. American Journal of vessel vasculitis attributed to anticardiolipin antibody. Arthritis and
Psychiatry, 150, 495–7. Rheumatism, 37, 1254–7.
Peikert, T., Finkielman, J.D., Hummel, A.M., et al. (2008). Functional char- Robinson, K., Mayer, E., and Jacobsen, D.W. (1994). Homocysteine and coro-
acterization of antineutrophil cytoplasmic antibodies in patients with nary artery disease. Cleveland Clinic Journal of Medicine, 61, 438–50.
cocaine-induced midline destructive lesions. Arthritis and Rheumatism, Rodriguez-Serna, M., Botella-Estrada, R., Chabas, A., et  al. (1996).
58, 1546–51. Angiokeratoma corporis diffusum associated with beta-mannosidase
Peters, M., Gottschalk, D., Boit, R., Pohle, H.D., and Ruf, B. (1993). deficiency. Archives of Dermatology, 132, 1219–22.
Meningovascular neurosyphilis in human immunodeficiency virus Roe, S.M., Graham, L.D., Brock, W.B., and Barker, D.E. (1994).
infection as a differential diagnosis of focal CNS lesions: a clinicopatho- Calciphylaxis: early recognition and management. American Journal of
logical study. Journal of Infection, 27, 57–62. Surgery, 60, 81–6.
Peters, M.N., Hall, R.J., Cooley, D.A., Leachman, R.D., and Garcia, E. (1974). Rolfs, A., Bottcher, T., Zschiesche, M., et al. (2005). Prevalence of Fabry dis-
The clinical syndrome of atrial myxoma. JAMA, 230, 695–701. ease in patients with cryptogenic stroke:  a prospective study. Lancet,
Pierce, J.R. Jr, Wren, M.V., and Cousar, J.B. Jr (1978). Cholesterol embo- 366, 1794–6.
lism: diagnosis antemortem by bone marrow biopsy. Annals of Internal Rosenberg, D.M., Ferrans, V.J., Fulmer, J.D., et  al. (1980). Chronic air-
Medicine, 89, 937–8. flow obstruction in Fabry’s disease. American Journal of Medicine, 68,
Pineda, C.J., Weisman, M.H., Bookstein, J.J., and Saltzstein, S.L. (1985). 898–905.
Hypothenar hammer syndrome. Form of reversible Raynaud’s phenom- Rosenstein, E.D. and Kramer, N. (1994). Occult subacute thyroiditis mimick-
enon. American Journal of Medicine, 79, 561–70. ing classic giant cell arteritis. Arthritis and Rheumatism, 37, 1618–20.
Pless, M.L., Chen, Y.B., Copen, W.A., and Frosch, M.P. (2010). Case records Ross, G., Kuwamura, F., and Goral, A. (1993). Association of Fabry’s disease
of the Massachusetts General Hospital. Case 9-2010. A  37-year-old with femoral head avascular necrosis. Orthopedics, 16, 471–3.
CHAPTER 44 imitators of vasculitis 653

Roux, S., Grossin, M., De Brandt, M., Palazzo, E., Vachon, F., et al. (1995). Serdaru, M., Chiras, J., Cujas, M., and L’Hermitte, F. (1984). Isolated benign
Angiotropic large cell lymphoma with mononeuritis multiplex mim- cerebral vasculitis or migrainous vasospasm? Journal of Neurology,
icking systemic vasculitis. Journal of Neurology, Neurosurgery and Neurosurgery and Psychiatry, 47, 73–6.
Psychiatry, 58, 363–6. Seton, M., Pless, M., Fishman, J.A., Caruso, P.A., and Hedley-Whyte, E.T.
Rowe, J.W., Gilliam, J.I., and Warthin, T.A. (1974). Intestinal manifestations (2008). Case records of the Massachusetts General Hospital. Case
of Fabry’s disease. Annals of Internal Medicine, 81, 628–31. 18-2008. A 68-year-old man with headache and visual changes after liver
Rowshani, A.T., Schot, L.J., and Ten Berge, I.J. (2004). c-ANCA as a serologi- transplantation. New England Journal of Medicine, 358, 2619–28.
cal pitfall. Lancet, 363, 782. Sheibani, K., Battifora, H., Winberg, C.D., et al. (1986). Further evidence that
Rumbaugh, C.L., Bergeron, R.T., Fang, H.C., and McCormick, R. (1971a). ‘malignant angioendotheliomatosis’ is an angiotropic large-cell lym-
Cerebral angiographic changes in the drug abuse patient. Radiology, phoma. New England Journal of Medicine, 314, 943–8.
101, 335–44. Sheth, K.J. and Bernhard, G.C. (1979). The arthropathy of Fabry disease.
Rumbaugh, C.L., Bergeron, R.T., Scanlan, R.L., et al. (1971b). Cerebral vas- Arthritis and Rheumatism, 22, 781–3.
cular changes secondary to amphetamine abuse in the experimental ani- Shetty Roy, A.N., Radin, M., Sarabi, D., and Shaoulian, E. (2011). Familial
mal. Radiology, 101, 345–51. recurrent atrial myxoma:  Carney’s complex. Clinical Cardiology,
Ryan, T.J. and Wilkinson, D.S. (1986). Cutaneous vascultis; ‘angiitis’. In 34, 83–6.
Textbook of dermatology, 4th ed. (ed. A. Rook, D.S. Wilkinson, F.J.G. Shim, S., Yoo, D.H., Lee, J.K., Koh, H.K., Lee, S.R., et al. (1998). Multiple cere-
Ebling, R.H. Champion, and J.L. Burton), pp. 1121–85. Blackwell bellar infarction due to vertebral artery obstruction and bulbar symptoms
Scientific. associated with verticle subluxation and atlanto-occipital subluxation in
Rybka, S.J. and Novick, A.C. (1983). Concomitant carotid, mesenteric and ankylosing spondylitis. Journal of Rheumatology, 25, 2464–8.
renal artery stenosis due to primary intimal fibroplasia. Journal of Sienknecht, C., Whetsell, W.O., and Pollock, P. (1995). Intravascular malig-
Urology, 129, 798–800. nant lymphoma (‘malignant angioendotheliomatosis’). mimicking pri-
Sack, K.E. (1993). When vasculitis is not vasculitis. Hospital Practice Office mary angiitis of the central nervous system. Journal of Rheumatology,
Edition, 28, 94, 97–100, 103. 22, 1769–70.
Salgado, P., Rojas, R., and Sotelo, J. (1997). Cysticercosis. Clinical classifi- Sijpkens, Y., Westendorp, R., Van Kemenade, F., Van Duinen, S., and
cation based on imaging studies. Archives of Internal Medicine, 157, Breedveld, F. (1997). Vasculitis due to cholesterol embolism. American
1991–7. Journal of Medicine, 102, 302–3.
Salvarani, C., Gabriel, S.E., Gertz, M.A., Bjornsson, J., Li, C.Y., and Hunder, Silbert, P., Bartleson, J.D., Miller, G.M., Parisi, J.E., Goldman, M.S., et al.
G.G. (1994). Primary systemic amyloidosis presenting as giant cell arte- (1995). Cortical petechial hemorrhage, leukoencephalopathy, and suba-
ritis and polymyalgia rheumatica. Arthritis and Rheumatism, 37, 1621–6. cute dementia associated with seizures due to cerebral amyloid angiopa-
Sammaritano, L.R., Gharavi, A.E., and Lockshin, M.D. (1990). thy. Mayo Clinic Proceedings, 70, 477–80.
Antiphospholipid antibody syndrome:  immunologic and clinical Siltanen, P., Tuuteri, L., Norio, R., Tala, P., Ahrenberg, P., and Halonen, P.I.
aspects. Seminars in Arthritis and Rheumatism, 20, 81–96. (1976). Atrial myxoma in a family. American Journal of Cardiology,
Saraya, T., Shimura, C., Wada, H., Aoshima, M., and Goto, H. (2006). 38, 252–6.
Evidence for vascular spread of varicella zoster-associated vasculopathy. Simpson, D.M. and Tagliati, M. (1994). Neurologic manifestations of HIV
Annals of Internal Medicine, 144, 535–7. infection. Annals of Internal Medicine, 121, 769–85.
Savader, S.J., Savader, B.L., and Drewry, G.R. (1988). Hypothenar hammer Singh, A.K. and Wetherley-Mein, G. (1977). Microvascular occlusive lesions
syndrome with embolic occlusion of digital arteries. Clinical Radiology, in primary thrombocythaemia. British Journal of Haematology, 36,
39, 324–5. 553–64.
Savige, J.A., Yeung, S.P., Davies, D.J., Ebeling, P., and Hunt, D.H. (1988). Singhal, A.B., Kimberly, W.T., Schaefer, P.W., and Hedley-Whyte, E.T. (2009).
Anti-neutrophil cytoplasmic antibodies associated with atrial myxoma. Case records of the Massachusetts General Hospital. Case 8-2009.
American Journal of Medicine, 85, 755–6. A 36-year-old woman with headache, hypertension, and seizure 2 weeks
Schapira, D., Nahir, A.M., and Hadad, N. (2002). Interferon-induced post partum. New England Journal of Medicine, 360, 1126–37.
Raynaud’s syndrome. Seminars in Arthritis and Rheumatism, 32, 157–62. Slater, C.A., Sickel, J.Z., Visvesvara, G.S., Pabico, R.C., and Gaspari, A.A.
Schievink, W.I., Prakash, U.B., Piepgras, D.G., and Mokri, B. (1994a). Alpha (1994). Brief report: successful treatment of disseminated acanthamoeba
1-antitrypsin deficiency in intracranial aneurysms and cervical artery infection in an immunocompromised patient. New England Journal of
dissection. Lancet, 343, 452–53. Medicine, 331, 85–7.
Schievink, W.I., Bjeornsson, J., Parisi, J.E., and Prakash, U.B. (1994b). Arterial Slavin, R., Saeki, K., Bhagavan, B., and Maas, A.E. (1995). Segmental arterial
fibromuscular dysplasia associated with severe alpha 1-antitrypsin defi- mediolysis: a precursor to fibromuscular dysplasia? Modern Pathology,
ciency. Mayo Clinic Proceedings, 69, 1040–3. 8, 287–94.
Schiffmann, R., Kopp, J.B., Austin, H.A. 3rd, et al. (2001). Enzyme replace- Sloan, M.A., Kittner, S.J., Rigamonti, D., and Price, T.R. (1991). Occurrence
ment therapy in Fabry disease:  a randomized controlled trial. JAMA, of stroke associated with use/abuse of drugs. Neurology, 41, 1358–64.
285, 2743–9. Slovut, D.P. and Olin, J.W. (2004). Fibromuscular dysplasia. New England
Schmidley, J.W. (1993). Neurological presentations of atrial myxoma. Heart Journal of Medicine, 350, 1862–71.
Disease and Stroke, 2, 483–6. Smith, K.J., Skelton, H.G.D., James, W.D., et  al. (1990). Cutaneous histo-
Schwarz, G.A., Schwartzman, R.J., and Joyner, C.R. (1972). Atrial myxoma. pathologic findings in ‘antiphospholipid syndrome’. Correlation with
Cause of embolic stroke. Neurology, 22, 1112–21. disease, including human immunodeficiency virus disease. Archives of
Scolari, F. and Ravani, P. (2010). Atheroembolic renal disease. Lancet, 375, Dermatology, 126, 1176–83.
1650–60. Smith, M.C., Ghose, M.K., and Henry, A.R. (1981). The clinical spectrum
Scott, R.M. and Smith, E.R. (2009). Moyamoya disease and moyamoya syn- of renal cholesterol embolization. American Journal of Medicine, 71,
drome. New England Journal of Medicine, 360, 1226–37. 174–80.
Selhub, J., Jacques, P.F., Bostom, A.G., et  al. (1995). Association between Smith, S.B. and Arkin, C. (1972). Cryofibrinogenemia:  incidence, clinical
plasma homocysteine concentrations and extracranial carotid-artery correlations, and a review of the literature. American Journal of Clinical
stenosis. New England Journal of Medicine, 332, 286–91. Pathology, 58, 524–30.
Selzer, A., Sakai, F.J., and Popper, R.W. (1972). Protean clinical manifestations Sneddon, I.B. (1965). Cerebro-Vascular Lesions and Livedo Reticularis.
of primary tumors of the heart. American Journal of Medicine, 52, 9–18. British Journal of Dermatology, 77, 180–5.
654 SECTION 6 mimickers of vasculitis

Solans-Laque, R., Bosch-Gil, J.A., Perez-Bocanegra, C., Selva-O’Callaghan, Thomas, P. and Lowitt, N.R. (1995). A traumatic experience. New England
A., Simeon-Aznar, C.P., and Vilardell-Tarres, M. (2008). Paraneoplastic Journal of Medicine, 333, 307–10.
vasculitis in patients with solid tumors:  report of 15 cases Journal of Thomas, R., Vuitch, F., and Lakhanpal, S. (1994). Angiocentric T cell lym-
Rheumatology, 35, 294–304. phoma masquerading as cutaneous vasculitis. Journal of Rheumatology,
Somer, T. and Finegold, S. (1995). Vasculitides associated with infections, 21, 760–2.
immunization, and antimicrobial drugs. Clinical Infectious Diseases, 20, Thomas, V.H. and Hopkins, I.J. (1972). Arteriographic demonstration of vas-
1010–36. cular lesions in the study of neurologic deficit in advanced Haemophilus
Soubrier, M.J., Dubost, J.J., and Sauvezie, B.J. (1994). POEMS syndrome: a influenzae meningitis. Developmental Medicine and Child Neurology,
study of 25 cases and a review of the literature. French Study Group on 14, 783–7.
POEMS Syndrome. American Journal of Medicine, 97, 543–53. Tjia, T.L., Yeow, Y.K., and Tan, C.B. (1985). Cryptococcal meningitis. Journal
Speranza, G. and Javaheri, S. (2008). Flu-like illness—and a rash. Lancet, of Neurology, Neurosurgery and Psychiatry, 48, 853–8.
372, 688. Tomsick, T.A., Lukin, R.R., Chambers, A.A., and Benton, C. (1976).
Sprabery, A.T., Newman, K., and Lohr, K.M. (1994). Aortic mural thrombus Neurofibromatosis and intracranial arterial occlusive disease.
presenting as pseudovasculitis. Chest, 106, 282–3. Neuroradiology, 11, 229–34.
Spring, M.W., Hartley, B., Scoble, J.E., and Viberti, G.C. (1998). A man with Torrens, J.K., McWhinney, P.H., and Tompkins, D.S. (1999). A deadly thorn: a
diabetes and unexplained renal failure. Lancet, 352, 956. case of imported melioidosis. Lancet, 353, 1016.
Stanton, R.C. and Nickeleit, V. (1996). Case records of the Massachusetts Travers, R.L., Allison, D.J., Brettle, R.P., and Hughes, G.R. (1979). Polyarteritis
General Hospital. New England Journal of Medicine, 334, 973–9. nodosa:  a clinical and angiographic analysis of 17 cases. Seminars in
Stebbing, J., Askin, F., Fishman, E., and Stone, J. (1999). Pulmonary mani- Arthritis and Rheumatism, 8, 184–99.
festations of ulcerative colitis mimicking Wegener’s granulomatosis. Trimarchi, M., Gregorini, G., Facchetti, F., et  al. (2001). Cocaine-induced
Journal of Rheumatology, 26, 1617–21. midline destructive lesions: clinical, radiographic, histopathologic, and
Stein, J.H. and McBride, P.E. (1998). Hyperhomocysteinemia and atheroscle- serologic features and their differentiation from Wegener granulomato-
rotic vascular disease: pathophysiology, screening, and treatment. off. sis. Medicine (Baltimore), 80, 391–404.
Archives of Internal Medicine, 158, 1301–6. Tzeng, D.Z., Fein, J., Boe, N., and Chan, A. (2005). A pregnant woman with
Stephens, C.J. (1991). The antiphospholipid syndrome. Clinical correlations, headaches, seizures, and hypertension. Lancet, 365, 2150.
cutaneous features, mechanism of thrombosis and treatment of patients Ueki, K., Meyer, F.B., and Mellinger, J.F. (1994). Moyamoya disease: the disor-
with the lupus anticoagulant and anticardiolipin antibodies. British der and surgical treatment. Mayo Clinic Proceedings, 69, 749–57.
Journal of Dermatology, 125, 199–210. Van Hemelrijk, C., Renard, M., and Loeys, B. (2010). The Loeys-Dietz syn-
St. John Sutton, M.G., Mercier, L.A., Giuliani, E.R., and Lie, J.T. (1980). Atrial drome: an update for the clinician. Current Opinion in Cardiology, 25,
myxomas:  a review of clinical experience in 40 patients. Mayo Clinic 546–51.
Proceedings, 55, 371–6. Vassa, N., Twardowski, Z.J., and Campbell, J. (1994). Hyperbaric oxygen
Stoane, L., Allen, J.H. Jr, and Collins, H.A. (1966). Radiologic observations therapy in calciphylaxis-induced skin necrosis in a peritoneal dialysis
in cerebral embolization from left heart myxomas. Radiology, 87, 262–6. patient. American Journal of Kidney Diseases, 23, 878–81.
Stokes, J.B., Bonsib, S.M., and McBride, J.W. (1996). Diffuse intimal fibromus- Vatz, K.A., Scheibel, R.L., Keiffer, S.A., and Ansari, K.A. (1974). Neurosyphilis
cular dysplasia with multiorgan failure. Archives of Internal Medicine, and diffuse cerebral angiopathy: a case report. Neurology, 24, 472–6.
156, 2611–4. Vayssairat, M., Debure, C., Cormier, J.M., Bruneval, P., Laurian, C., and
Stone, G.C., Wall, B.A., Oppliger, I.R., et  al. (1989). A vasculopathy with Juillet, Y. (1987). Hypothenar hammer syndrome: seventeen cases with
deposition of lambda light chain crystals. Annals of Internal Medicine, long-term follow-up. Journal of Vascular Surgery, 5, 838–43.
110, 275–8. Veenendaal-Hilbers, J.A., Perquin, W.V., Hoogland, P.H., and Doornbos, L.
Stone, J., Pomper, M.G., and Hellmann, D.B. (1998). Histoplasmosis mimick- (1988). Basal meningovasculitis and occlusion of the basilar artery in
ing vasculitis of the central nervous system. Journal of Rheumatology, two cases of Borrelia burgdorferi infection. Neurology, 38, 1317–9.
25, 1644–8. Viard, J.P., Lesavre, P., Boitard, C., et al. (1988). POEMS syndrome present-
Stone, J.H., Zen, Y., and Deshpande, V. (2012). IgG4-related disease. New ing as systemic sclerosis. Clinical and pathologic study of a case with
England Journal of Medicine, 366, 539–51. microangiopathic glomerular lesions. American Journal of Medicine,
Sundy, J. and Haynes, B. (1995). Pathogenic mechanisms of vessel damage in 84, 524–8.
vasculitis syndromes. Rheumatic Disease Clinics of North America, 21, Victor, D.I. and Green, W.R. (1976). Temporal artery biopsy in herpes zoster
861–81. ophthalmicus with delayed arteritis. American Journal of Ophthalmology,
Suwanwela, C. and Suwanwela, N. (1972). Intracranial arterial narrowing and 82, 628–30.
spasm in acute head injury. Journal of Neurosurgery, 36, 314–23. Wachter, R.M., Burke, A.M., and MacGregor, R.R. (1984). Strongyloides ster-
Suzuki, J. and Takaku, A. (1969). Cerebrovascular ‘moyamoya’ disease. coralis hyperinfection masquerading as cerebral vasculitis. Archives of
Disease showing abnormal net-like vessels in base of brain. Archives of Neurology, 41, 1213–6.
Neurology, 20, 288–99. Walker, D.H. and Mattern, W.D. (1980). Rickettsial vasculitis. American
Takeuchi, K. and Shimizu, K. (1957). Hypogenesis of bilateral internal carotid Heart Journal, 100, 896–906.
arteries. No To Shinkei, 9, 37–43. Walker, U., Herbst, E.W., Ansorge, O., and Peter, H.H. (1994). Intravascular
Tapia, J. and Schumacher, J. (1993). Case records of the Massachusetts lymphoma simulating vasculitis. Rheumatology International, 14, 131–3.
General Hospital. New England Journal of Medicine, 329, 117–24. Wall, L.M. and Smith, N.P. (1981). Perniosis:  a histopathological review.
Tazelaar, H.D., Locke, T.J., and Mcgregor, C.G. (1992). Pathology of surgically Clinical and Experimental Dermatology, 6, 263–71.
excised primary cardiac tumors. Mayo Clinic Proceedings, 67, 957–65. Waxman, S. and Dove, J.T. (1969). Cryofibrinogenemia aggravated during
Thadhani, R.I., Camargo, C.A. Jr, Xavier, R.J., Fang, L.S., and Bazari, H. hypothermia. New England Journal of Medicine, 281, 1291–2.
(1995). Atheroembolic renal failure after invasive procedures. Natural Webster, E., Corman, L.C., and Braylan, R.C. (1986). Syndrome of tempo-
history based on 52 histologically proven cases. Medicine (Baltimore), ral arteritis with perivascular infiltration by malignant cells in a patient
74, 350–8. with follicular small cleaved cell lymphoma. Journal of Rheumatology,
Thomas, E.W.P. (1964). Chapping and chilblains. Practioner, 193, 775–60. 13, 1163–6.
Thomas, M.H. (1981). Myxoma masquerading as polyarteritis nodosa. Welch, G.N. and Loscalzo, J. (1998). Homocysteine and atherothrombosis.
Journal of Rheumatology, 8, 133–7. New England Journal of Medicine, 338, 1042–50.
CHAPTER 44 imitators of vasculitis 655

Wenzel, R.P., Hayden, F.G., Greoschel, D.H., et al. (1986). Acute febrile cere- Wold, L.E. and Lie, J.T. (1980). Cardiac myxomas: a clinicopathologic profile.
brovasculitis: a syndrome of unknown, perhaps rickettsial, cause. Annals American Journal of Pathology, 101, 219–40.
of Internal Medicine, 104, 606–15. Woolfson, R.G. and Lachmann, H. (1998). Improvement in renal cholesterol
Wernick, R. and Smith, D.L. (1989). Bilateral hypothenar hammer syn- emboli syndrome after simvastatin. Lancet, 351, 1331–2.
drome: an unusual and preventable cause of digital ischemia. American Wu, S.M., Min, Y., Ostrzega, N., Clements, P.J., and Wong, A.L. (2005).
Journal of Emergency Medicine, 7, 302–6. Lymphomatoid granulomatosis: a rare mimicker of vasculitis. Journal of
Wheat, L.J., Batteiger, B.E., and Sathapatayavongs, B. (1990). Histoplasma Rheumatology, 32, 2242–5.
capsulatum infections of the central nervous system. A clinical review. Yamamoto, M., Aoyagi, M., Fukai, N., Matsushima, Y., and Yamamoto, K.
Medicine, 69, 244–60. (1998). Differences in cellular responses to mitogens in arterial smooth
White, W.B. and Parke, A.L. (2005). Acute pain in the tip of the index finger. muscle cells derived from patients with moyamoya disease. Stroke, 29,
American Journal of Medicine, 118, 1223–4. 1188–93.
Wick, M.R., Mills, S.E., Scheithauer, B.W., Cooper, P.H., Davitz, M.A., and Yamashita, M., Oka, K., and Tanaka, K. (1983). Histopathology of the brain
Parkinson, K. (1986). Reassessment of malignant ‘angioendotheliomato- vascular network in moyamoya disease. Stroke, 14, 50–8.
sis’. Evidence in favor of its reclassification as ‘intravascular lymphoma- Young, D.K., Burton, M.F., and Herman, J.H. (1986). Multiple cholesterol
tosis’. American Journal of Surgical Pathology, 10, 112–23. emboli syndrome simulating systemic necrotizing vasculitis. Journal of
Wiesner, O., Russell, K.A., Lee, A.S., et al. (2004). Antineutrophil cytoplas- Rheumatology, 13, 423–6.
mic antibodies reacting with human neutrophil elastase as a diagnostic Zarate, Y.A. and Hopkin, R.J. (2008). Fabry’s disease. Lancet, 372,
marker for cocaine-induced midline destructive lesions but not autoim- 1427–35.
mune vasculitis. Arthritis and Rheumatism, 50, 2954–65. Zen, Y. and Nakanuma, Y. (2010). IgG4-related disease: a cross-sectional
Williams, P.L., Johnson, R., Pappagianis, D., et  al. (1992). Vasculitic and study of 114 cases. American Journal of Surgical Pathology, 34,
encephalitic complications associated with Coccidioides immitis infec- 1812–9.
tion of the central nervous system in humans: report of 10 cases and Zuber, J., Martinez, F., Droz, D., Oksenhendler, E., and Legendre, C. (2002).
review. Clinical Infectious Diseases, 14, 673–82. Alpha-interferon-associated thrombotic microangiopathy:  a clinico-
Winter, W.J. (1957). Atheromatous emboli; a cause of cerebral infarction. pathologic study of 8 patients and review of the literature. Medicine
AMA Archives of Pathology, 64, 137–142. (Baltimore), 81, 321–31.
Index

Note: page numbers in italics refer to figures and tables.

A cerebral amyloid angiopathy ( amyloid β-related polyarteritis nodosa 125, 183, 184, 233, 235,
abatacept, giant cell arteritis 600 angiitis) 499 258, 259
abdominal pain anaphylatoxin inactivator 55 pulmonary artery 147, 151
cryoglobulinaemic vasculitis 556 anaphylatoxins 54–5, 72 Takayasu’s arteritis 228, 229, 254
differential diagnosis in children 538 ANCA-associated vasculitis (AAV) 4, 7, 62 angiogenesis 43
IgA vasculitis 285, 530, 538–9 animal models abnormalities in vasculitis 48
polyarteritis nodosa 339 MPO-ANCA vasculitis 96–8, 97 defects in scleroderma 213
systemic lupus erythematosus 448 PR3-ANCA vasculitis 98 inhibitors 45, 46–7
thromboangiitis obliterans 508 SCG/Kj mouse strain 93–4 mediators 45–6
abnormal proteins 637 childhood disease 289 angiogenesis therapy, thromboangiitis obliterans
acne-like lesions, Behçet’s syndrome 470–1, 474 eosinophilic granulomatosis with polyangiitis 513–14
juvenile disease 493 291 angiographically defined PACNS 499
acute haemorrhagic oedema of infancy granulomatosis with polyangiitis 289–90 angiography 279
(Finkelstein’s disease) 125, 538 microscopic polyangiitis 290–1 Behçet’s syndrome 237–8
Acute Physiology and Chronic Health Evaluation clinical features characteristic patterns 227
(APACHE) score 343 cardiopulmonary disease 144, 148 CNS vasculitis 503
adalimumab ocular manifestations 135–6 connective tissue disease-associated vasculitis
in Behçet’s syndrome 480 renal involvement 198–200 239–40
in giant cell arteritis 315 EMEA classification algorithm 402 CT angiography 248
in granulomatosis with polyangiitis 604–5 environmental and genetic factors 66 digital ischaemia 216–17
Addison’s disease, antiphospholipid syndrome 623 epidemiology giant cell arteritis 232–3, 234
adipose tissue, role in endothelial regeneration 43 childhood disease 19 granulomatosis with polyangiitis 409–10
age-related incidence of vasculitis 7 eosinophilic granulomatosis with polyangiitis infection-related vasculitis 238–9
agranulocytosis, cyclophosphamide-induced 428 17–18 Kawasaki’s disease 233, 378
AIDS see human immunodeficiency virus granulomatosis with polyangiitis 13–16 microscopic polyangiitis 355
airway lesions 143–4 microscopic polyangiitis 16–17 midaortic syndrome 242
allergy, origins of term 29 malignancy-associated 584 mimics of vasculitis 240–1, 636
alpha-1-antitrypsin (α1-AT) see SERPINA1 management, experimental therapies 602–5 MR angiography 249
alpha-1-antitrypsin deficiency 80–1, 640 pathogenesis 72 polyarteritis nodosa 233, 235, 258, 342
alpha2-adrenergic receptors genetic factors 80–1 renal involvement 338
control of blood vessel calibre 212 role of complement 57 tibial artery involvement 339
inhibition of 220 role of infections 82 Raynaud’s phenomenon 216–17, 243
alpha-galactosidase A deficiency, Fabry’s disease pathophysiology of glomerulonephritis 200–1 reversible cerebral vasoconstriction syndromes
641–2 renal biopsy 201–2 (RCVS) 504
alpha smooth muscle actin (α-SMA), role in IgA see also eosinophilic granulomatosis with Takayasu’s arteritis 228–30, 251, 253, 254, 322–3
vasculitis 534 polyangiitis; granulomatosis with thromboangiitis obliterans 236–7, 509, 510
alternative complement activation pathway 53–4 polyangiitis; microscopic polyangiitis angioimmunolymphoproliferative lesions (AIL),
alveolar haemorrhage anchor ELISA 64 CNS vasculitis 500
eosinophilic granulomatosis with polyangiitis 436 aneurysm formation 105, 107 angiokeratoma corporis diffusum (Fabry’s disease)
microscopic polyangiitis 354 Behçet’s syndrome 115, 237–8, 261, 472, 473 641–2
American College of Rheumatology (ACR) bypass grafts 231 angiopoeitin-1/Tie2 system 45
classification of vasculitic syndromes 3, 7 giant cell arteritis 255, 256, 315–16 inhibition of 46
GPA 401 infection-related vasculitis 238 angiostatin 47
polyarteritis nodosa 331 aortitis 570 angiotensin converting enzyme (ACE) inhibitors,
criteria for cutaneous leukocytoclastic vasculitis inflammatory aneurysms 262, 263 in Raynaud’s phenomenon 219
517, 518 Kawasaki’s disease 233, 260, 287, 374–5, animal models 98
amphetamines, vascular effects 643 376, 377 complement-induced endothelial damage 56–7
amyloidosis 640 angiography 378 giant cell arteritis 308
Behçet’s syndrome 476 mycotic aneurysms 115 induced
658 index

animal models (Cont.) antinuclear antibodies (ANA) 277 juvenile disease 493
models of ANCA-associated vasculitis 76, antioxidant therapy, in Raynaud’s phenomenon 220 management 480
96–8, 97, 201, 352, 393–4 antiphospholipid antibodies, cutaneous cryoglobulinaemic vasculitis 555
models of anti-glomerular basement polyarteritis nodosa 365, 368 cutaneous polyarteritis nodosa 364
membrane disease 95 antiphospholipid syndrome (Hughes’ syndrome) hyperimmunoglobulinaemia D syndrome 582
models of immune complex vasculitis 94–5 117, 637 hypocomplementaemic urticarial vasculitis 580
models of infection-induced vasculitis 95–6 associated conditions 625–6 IgA vasculitis 285, 531
spontaneous clinical features polyarteritis nodosa 335
in inbred mouse strains 93 Addison’s disease 623 thromboangiitis obliterans 509
interleukin-1 receptor antagonist deficient atherosclerosis 624 Arthus reaction 29, 30, 32–3, 56
mice 94 cardiac manifestations 622 aspirin
McH5/lpr mouse strain 94 catastrophic APS 623–4 in antiphospholipid syndrome 619, 626
MRL/MpJ-Faslpr mouse strain 93, 94 cutaneous lesions 619–20 in Degos’ disease 579
NZB/NZW F1 and NZW/BXSB F1 mice 94 gastrointestinal involvement 623 in giant cell arteritis 315
SCG/Kj mouse strain 93–4 musculoskeletal manifestations 622–3 in Kawasaki’s disease 288, 378, 379
Takayasu’s arteritis 320–1 neurological manifestations 501, 620–1 in Raynaud’s phenomenon 219, 220
anterior ischaemic optic neuropathy (AION), giant pregnancy morbidity 621–2 in Takayasu’s arteritis 293
cell arteritis 311 pulmonary manifestations 448–9, 622 in thromboangiitis obliterans 512
anterior uveitis, seronegative renal involvement 623 assessment of disease 299–300
spondyloarthropathies 454 thrombocytopaenia 622 asthma 143
anti-aquaporin 4 antibodies 452 thrombosis 617, 619 eosinophilic granulomatosis with polyangiitis
antibiotic therapy thrombotic risk 618–19 150, 435, 439
Behçet’s syndrome 480–1 definition and classification 616–17 asthma drugs, association with EGPA 79
cutaneous leukocytoclastic vasculitis 524 demographics 616 asymmetrical polyneuropathy 161, 166
antibody-dependent cellular cytotoxicity differences between primary and secondary asymmetric dimethylarginine (ADMA), role in
(ADCC) 78 disease 624–5 vascular injury 42–3
antibody reactions, type II hypersensitivity 32 historical background 615 asymptomatic vasculitic neuropathy 166–7
anti-C1q antibodies 448 imaging 260 atheroembolic disease 636–7
anticardiolipin antibodies 615 indications for testing 617 atherosclerosis 41
methods of detection 615–16 methods of antibody detection 615–16 antiphospholipid syndrome 624
thromboangiitis obliterans 509 pathophysiology 625–6 Behçet’s syndrome 474
anticoagulation 626–7 seronegative disease 619 atopy 30–2
in acute digital ischaemia 217–18 treatment 626–7 atypical ANCA 61–2
in antiphospholipid syndrome 619, 626 anti-SSA/Ro antibodies 447 auditory symptoms, Cogan’s syndrome 576–7
in Behçet’s syndrome 482 antithymocyte globulin (ATG), granulomatosis autoamputation 219, 220, 221
in cutaneous polyarteritis nodosa 369 with polyangiitis 420 autoantibodies 32, 61
in Degos’ disease 579 antithyroid drugs see also antiendothelial cell antibodies;
in Kawasaki’s disease 379 association with vasculitis 79–80 antineutrophil cytoplasmic antibodies
anti-dsDNA antibodies 447 see also propylthiouracil Autoantigen Complementarity, Theory of 74
antiendothelial cell antibodies (AECAs) 47–8, 61 antiviral therapy autonomic neuropathy, Sjögren’s syndrome 451
pathogenic role 77–8, 352–3 HBV-related PAN 346 avascular necrosis of bone, antiphospholipid
in polyarteritis nodosa 334 see also ribavirin syndrome 622
in systemic lupus erythematosus 447 aortic dissection, polyarteritis nodosa 338 axonal degeneration 159, 160
in Takayasu’s arteritis 320 aortic aneurysm, giant cell arteritis 315–16 azathioprine
anti-glomerular basement membrane disease aortitis 149, 151 adverse effects 427
(Goodpasture’s syndrome) 146 antiphospholipid syndrome 260 in Behçet’s syndrome 480, 481, 482
animal models 95, 201 bacterial 82 in childhood primary angiitis of the central
ocular manifestations 136 Cogan’s syndrome 577 nervous system 294
antineutrophil cytoplasmic antibodies (ANCA) 61 in connective tissue disease 444 in Cogan’s syndrome 577
antigenic specificity 74 rheumatoid vasculitis 445 in cutaneous polyarteritis nodosa 368
determination of 63–4 giant cell arteritis 255, 256, 308, 311 in cutaneous leukocytoclastic vasculitis 523, 524
diagnostic and prognostic value 64–5, 74–6 imaging 313, 314 in eosinophilic granulomatosis with polyangiitis
monitoring levels during follow-up 65–6 IgG4-related disease 455, 582, 645–6 438
disease evaluation 301 infectious 238, 570 in giant cell arteritis 315
in drug-induced vasculitis 79–80 relapsing polychondritis 453 in granulomatosis with polyangiitis 290,
elastase-ANCA and hLAMP-2-ANCA 65 Takayasu’s arteritis 251, 252, 253, 321 422–3, 427
in eosinophilic granulomatosis with aphthous ulcers maintenance of remission 420
polyangiitis 435 Behçet’s syndrome 468–70, 469 in IgA vasculitis 286, 539
in granulomatosis with polyangiitis 392–4, juvenile disease 492–3 in IgG4-related disease 583
406–7 hyperimmunoglobulinaemia D syndrome 582 in microscopic polyangiitis 291, 357
in IgA vasculitis 533, 538 apolipoprotein H (APOH) genes 625 in polyarteritis nodosa 292, 345
laboratory tests 277 apoptosis in sarcoidosis 576
indications for 66 AECA-induced 78 in Takayasu’s arteritis 324
indirect immunofluorescence 62, 63 role in ANCA-associated vasculitis 77 in vasculitic neuropathy 169, 172
in microscopic polyangiitis 351–2, 355 role of PR3-ANCA 393 IgA nephritis 539
pathogenic role 66, 73–4, 75–7, 75, 200–1, 202 appendix
effects on neutrophils 77 IgA vasculitis 186 B
interaction of activated neutrophils with microscopic polyangiitis 186 bacterial endocarditis 569–70
endothelial cells 76–7 polyarteritis nodosa 183, 339, 346 bacterial superantigens (SAg) 82
in polyarteritis nodosa 333, 342 arrhythmias 148 bactericidal/permeability-increasing protein
in systemic lupus erythematosus 447 polyarteritis nodosa 338 (BPI)-ANCA 63, 75
targets in small-vessel vasculitides 62–3 arteriosclerosis obliterans 511–12 barium enema, signs of bowel ischaemia 180
terminology 61–2 arthralgia/arthritis barrier dysfunction, granulomatosis with
see also ANCA-associated vasculitis Behçet’s syndrome 474 polyangiitis 390–1
index 659

basophils, role in type I hypersensitivity 31 bladder, polyarteritis nodosa 340 antiphospholipid syndrome 622
B-cell clonal expansion, cryoglobulin production bladder cancer risk, cyclophosphamide 420–1 Behçet’s syndrome 473–4
551–2 prophylaxis 425, 427–8 management 482
B-cell depletion Blau’s syndrome 321 clinical evaluation 149–50
in cryoglobulinaemic vasculitis 602 blood pressure cryoglobulinaemic vasculitis 556
in GPA and MPA 604 Takayasu’s arteritis 322 eosinophilic granulomatosis with polyangiitis 437
in Takayasu’s arteritis 600–1 thromboangiitis obliterans 509 granulomatosis with polyangiitis 409
see also rituximab see also hypertension imaging 259, 260
B-cells blood vessel calibre, physiological control 212 Kawasaki’s disease 376–7
role in eosinophilic granulomatosis with ‘blue toe syndrome’ 637 microscopic polyangiitis 354, 355
polyangiitis 434 bone polyarteritis nodosa 338, 340
role in granulomatosis with polyangiitis 395 avascular necrosis 622 rheumatoid vasculitis 445
Behçet’s syndrome (BS) polyarteritis nodosa 340 scope
clinical features 479, 492 bone marrow cell transplantation, thromboangiitis cardiomyopathy and heart failure 148
amyloidosis and renal involvement 476 obliterans 513–14 conduction abnormalities 148
atherosclerosis 474 bortezomib coronary artery disease 148
cardiac manifestations 473–4 in cryoglobulinaemic vasculitis 561, 602 pericarditis 148
cardiopulmonary disease 144, 147, 148, in GPA and MPA 604 valvular heart disease 148
149, 151 in IgG4-related disease 583 seronegative spondyloarthropathies 454
central nervous system disease 474–5 bosentan Sjögren’s syndrome 452
cutaneous lesions 470–1 in Raynaud’s phenomenon 219 systemic lupus erythematosus 449
frequencies 469 in thromboangiitis obliterans 513 cardiac myxoma 637–8
gastrointestinal involvement 186–7, 475–6 botulinum toxin, in Raynaud’s phenomenon 219 cardiomyopathy 148
genital ulcers 470 bowel ischaemia in EGPA 150
major vessel involvement 472–3 clinical features 180 cardiovascular risk factors, giant cell arteritis 10
musculoskeletal manifestations 474 imaging 180–1, 182, 183, 185 Carney complex 637–8
ocular manifestations 131, 137, 471–2 pathophysiology 179–80 catastrophic APS 623–4
oral ulcers 468–70, 469 see also gastrointestinal manifestations treatment 627
pathergy 471, 478 brain biopsy 503–4 CC chemokines
urological problems 476 brain lesions angiostatic 47
diagnosis 479 Behçet’s syndrome 261 role in angiogenesis 46
diagnostic criteria 469 childhood primary angiitis of the central role in cryoglobulinaemic vasculitis 553
epidemiology 467–8 nervous system 293–4 role in giant cell arteritis 309
historical background 467 see also central nervous system involvement CD4 T cells, role in giant cell arteritis 310
imaging 261 bronchoalveolar lavage (BAL) CD44 44
angiography 237–8 in eosinophilic granulomatosis with polyangiitis CD59 55
PET 270 438 CD95/CD95 ligand system, role in EGPA 434
laboratory investigations 478–9 in granulomatosis with polyangiitis 408 CD99 44
management bronchoscopy 150 CD226 386
arthritis 481 in diffuse alveolar haemorrhage 146 celastrol 46
central nervous system disease 482 in granulomatosis with polyangiitis 408 cell adhesion molecules (CAMs) 41, 42, 43, 44
gastrointestinal disease 482 brucellosis, association with cryoglobulinaemic adhesion mechanisms 45
major vessel disease 481–2 vasculitis 550 effect of AECA 78
mucocutaneous lesions 480–1 bruits 276, 311 role in angiogenesis 46
ocular manifestations 481 Takayasu’s arteritis 324 role in vasculitis 48
pathogenesis 476–8 Budd–Chiari syndrome 187 SLE 447
pathological features 107, 114–15, 187, 470 Behçet’s syndrome 473 cell-mediated hypersensitivity reactions 30
prognosis 480 Buerger’s disease see thromboangiitis obliterans cell-mediated immunity, pathogenic role 83
see also juvenile Behçet’s syndrome bypass grafts animal models 96
benign angiopathy of the central nervous system in Kawasaki’s disease 379 central nervous system involvement 497
(BACNS) 501 in Takayasu’s arteritis 325 antiphospholipid syndrome 620–1
benign atrophic papulosis (localized Degos’ in thromboangiitis obliterans 513 Behçet’s syndrome 261, 474–5
disease) 578 juvenile disease 493
β2GPI 615, 616, 625 C management 482
biomarkers 300 C1 inhibitor (C1 INH) 55 childhood primary angiitis of the central
granulomatosis with polyangiitis 407 C3a 72 nervous system 293–4
biopsy 278–9 functions 54–5 cryoglobulinaemic vasculitis 556
of brain 503–4 C3b, functions 54 Degos’ disease 578
of cardiopulmonary lesions 149–50 C5a 72 diagnostic studies 502–4
of cutaneous lesions 123, 365 functions 54–5 eosinophilic granulomatosis with polyangiitis 437
in cutaneous leukocytoclastic vasculitis 522–3 role in endothelial injury 55–6 IgA vasculitis 533
in granulomatosis with polyangiitis 405 C5b-9 (membrano-lytic attack complex), role in polyarteritis nodosa 336
of muscle 162, 163 Degos’ disease 579 primary angiitis of the central nervous system
of nerves 160, 161–2, 163 cadherins 43 497–9
selection of site 167 calcification, Takayasu’s arteritis 253–4, 255 rheumatoid vasculitis 445
in polyarteritis nodosa 334–5 calciphylaxis 640–1 secondary forms 499–501
in rheumatoid vasculitis 446 calcium-channel blockers, in Raynaud’s Sjögren’s syndrome 451–2
selection of sites 116–17 phenomenon 218, 220 treatment and prognosis 504
see also renal biopsy; temporal artery biopsy cANCA 61, 74 varicella zoster virus infection 573
Birmingham Vasculitis Activity Score (BVAS) 300, Candida albicans infection, animal model of cerebral amyloid angiopathy (CAA, amyloid
301, 411, 418 vasculitis 95 β-related angiitis) 499
in childhood disease 283–4 cannabis arteritis 507 cerebrospinal fluid (CSF)
in polyarteritis nodosa 343 capture ELISA 64 Behçet’s syndrome 475
prognostic value 303 cardiac manifestations 150–2 CNS vasculitis 503
660 index

Chapel Hill Consensus Conference (CHCC) small-vessel vasculitis 517 cryoglobulinaemic vasculitis 553
Nomenclature of Vasculitides 4–5, 7, Takayasu’s arteritis 323 glomerulonephritis 201
332, 401 claudication systemic lupus erythematosus 447
microscopic polyangiitis 351 giant cell arteritis 311, 316 role in pregnancy morbidity, antiphospholipid
chest radiography 278 thromboangiitis obliterans 508 syndrome 622
eosinophilic granulomatosis with polyangiitis clinical features computed tomography (CT) 247–9, 250, 278, 280
436, 438 antiphospholipid syndrome 617, 619–24 Behçet’s syndrome 261
granulomatosis with polyangiitis 407, 408 Behçet’s syndrome 468–76, 479 bowel ischaemia 181, 182, 183
Takayasu’s arteritis 323 juvenile 492–3, 494 cardiopulmonary evaluation 149
Chikungunya virus infection, association with cardiopulmonary disease 143–52 chronic periaortitis 262, 263
cryoglobulinaemic vasculitis 550 cryoglobulinaemic vasculitis 553–7 CT angiography 248
childhood vasculitis cutaneous manifestations 121–8 eosinophilic granulomatosis with polyangiitis
ANCA-associated vasculitis 289 cutaneous leukocytoclastic vasculitis 519–20, 521 262–4, 436
eosinophilic granulomatosis with digital ischaemia and Raynaud’s phenomenon giant cell arteritis 255, 256, 313, 314
polyangiitis 291 209–21 granulomatosis with polyangiitis 262–4, 407–8
granulomatosis with polyangiitis 289–90 eosinophilic granulomatosis with polyangiitis IgA vasculitis 531
microscopic polyangiitis 290–1 435–7 Kawasaki’s disease 260
Blau’s syndrome 321 gastrointestinal manifestations 179–90 microscopic polyangiitis 259
classification 283 giant cell arteritis 310–11 pulmonary fibrosis 354
Cogan’s syndrome 294 granulomatosis with polyangiitis 404–5 polyarteritis nodosa 258, 259
disease evaluation 283–4, 303 IgA vasculitis 529–33 splenic infarct 339
epidemiology 18–19 Kawasaki’s disease 376–7 SLE and APS 260–1
ages of onset 284 microscopic polyangiitis 353–5 Takayasu’s arteritis 251, 254, 320, 323
features suggestive of vasculitis 284 neuropathy 157–74 see also cross-sectional imaging
IgA vasculitis 284–6, 527 ocular manifestations 131–8 conduction abnormalities 148
classification criteria 536–7 polyarteritis nodosa 335–42 conjunctival lesions 131–2
clinical course and outcome 540–1 polymyalgia rheumatica 311–12 granulomatosis with polyangiitis 135
clinical features 529–33 renal manifestations 197–204 Kawasaki’s disease 287
diagnosis 536, 536–7 rheumatoid vasculitis 445 connective tissue diseases
differences from adult disease 537–8 skin lesions 444 angiography 239–40
differential diagnosis 538 Sjögren’s syndrome 451–3 association with cryoglobulinaemic vasculitis
epidemiology 527–8 systemic lupus erythematosus 447–50 549, 550–1
genetic factors 528 Takayasu’s arteritis 321–2 clinical features
histology 535–6 thromboangiitis obliterans 508–9 cardiopulmonary disease 152
precipitating factors 528–9 clinical trial design, granulomatosis with characteristics of associated vasculitis 444
treatment 538–40 polyangiitis 417–19 CNS vasculitis 501
juvenile Behçet’s syndrome 491–4 clopidogrel digital ischaemia, treatment 217
Kawasaki’s disease 286–9, 373–80 in cutaneous polyarteritis nodosa 369 gastrointestinal vasculitis 188–9
polyarteritis nodosa 291–2, 341–2 in Kawasaki’s disease 379 Raynaud’s phenomenon 211
cutaneous 292, 369 clusterin 55 and Degos’ disease 578–9
primary angiitis of the central nervous system cocaine-associated vasculitis 387, 522, 644 differentiation from IgA vasculitis 538
293–4 ANCA 65, 75 IgG4-related disease 455
Sneddon’s syndrome 294 Cogan’s syndrome 294, 576–7 inflammatory myopathies 454–5
Susac’s syndrome 294 clinical features, ocular manifestations 137 pathological features of vasculitis 113–14
Takayasu’s arteritis 292–3, 321 imaging 262 relapsing polychondritis 114, 453–4
chilblains (pernio) 639 colchicine seronegative spondyloarthropathies 454
choroidal vasculitis, polyarteritis nodosa 135 in Behçet’s syndrome 480, 481 see also rheumatoid vasculitis; systemic lupus
Churg–Strauss granulomas 127, 436 juvenile disease 494 erythematosus; systemic sclerosis
Churg–Strauss syndrome see eosinophilic in cryoglobulinaemic vasculitis 561 Coombs, R.R.A., classification of hypersensitivity
granulomatosis with polyangiitis in cutaneous polyarteritis nodosa 368 reactions 30
ciclosporin in cutaneous leukocytoclastic vasculitis 524 corkscrew collaterals, thromboangiitis obliterans
in Behçet’s syndrome 481 in IgA vasculitis 286 236, 509, 510
in Cogan’s syndrome 577 in polyarteritis nodosa 346 coronary artery disease 148, 151
in cutaneous leukocytoclastic vasculitis 523 cold exposure eosinophilic granulomatosis with polyangiitis
in IgA nephritis 539–40 pernio 639 437
in IgA vasculitis 286 see also cryoglobulinaemic vasculitis Kawasaki’s disease 184, 233, 260, 287, 373–5,
in Kawasaki’s disease 378 collateral vessels, thromboangiitis obliterans 236, 374, 376
in Takayasu’s arteritis 324 509, 510 angiography 378
classical complement activation pathway 53 complement deficiency 80 management 378–9
classification of vasculitic syndromes 3–5, 7–8, 332 IgA vasculitis 528, 534 prognosis 379–80
ANCA-associated vasculitis 402 immune complex disease 33, 73 microscopic polyangiitis 355
antiphospholipid syndrome 616–17 see also hypocomplementaemic urticarial polyarteritis nodosa 233, 338
childhood disease 283 vasculitis systemic lupus erythematosus 449
cryoglobulinaemic vasculitis 548, 559 complement-induced endothelial damage 55–6 corticosteroids
cutaneous leukocytoclastic vasculitis 517, 521 animal models 56–7 adverse effects 315, 426
eosinophilic granulomatosis with polyangiitis complement inhibition, GPA and MPA 603 in Behçet’s syndrome 480, 481, 482
433–4 complement levels 277 juvenile disease 494
giant cell arteritis 312 complement system 53 in childhood primary angiitis of the central
granulomatosis with polyangiitis 401–2, 417–18 activation pathways 53–4 nervous system 294
IgA vasculitis 536–7 biologically active activation products 54–5 in CNS vasculitis 504
microscopic polyangiitis 351 interaction with endothelial cells 43 in Cogan’s syndrome 577
polyarteritis nodosa 331, 333 regulation 55 in cryoglobulinaemic vasculitis 561
primary angiitis of the central nervous system role in clinical vasculitis 57, 72–3 in cutaneous polyarteritis nodosa 368
497–8 animal models 95 in cutaneous leukocytoclastic vasculitis 523, 524
index 661

in digital ischaemia 221 historical background 547 historical background 363


in eosinophilic granulomatosis with polyangiitis pathogenesis 551–3 laboratory investigations 363, 365
291, 438 association with HCV 81–2, 173 management 346, 368–9
in giant cell arteritis 313–15 pathological features 114, 552, 554, 555 pathogenesis 368
in granulomatosis with polyangiitis 289–90, prognosis 558–9 prognosis 369
419–20, 426 treatment 559–61 CXC chemokines
in IgA vasculitis 186, 286, 538–9 experimental therapies 601–2 angiostatic 47
in IgG4-related disease 455, 583 therapeutic approaches 562 role in angiogenesis 45–6
in Kawasaki’s disease 184, 288, 378, 379 cryoglobulins 547, 548 role in cryoglobulinaemic vasculitis 553
in malignancy-associated vasculitis 585 biochemical characteristics 552–3 CYCAZAREM trial 421, 422–3
in microscopic polyangiitis 291, 356, 357 detection of 557 cyclophosphamide
in polyarteritis nodosa 198, 292, 344–5 production of 551–2 adverse effects 420–1, 426, 428
in relapsing polychondritis 453 CTLA-4 386 malignancy risk 387, 413, 425, 427–8
in rheumatoid vasculitis 446 cutaneous lesions 121 in Behçet’s syndrome 482
in sarcoidosis 576 acute haemorrhagic oedema of infancy 125 in childhood primary angiitis of the central
in seronegative spondyloarthropathies 454 antiphospholipid syndrome 619–20, 637 nervous system 294
in Sjögren’s syndrome 451, 452 atheroembolic disease 636 in CNS vasculitis 504
in systemic lupus erythematosus 448, 449, 450 Behçet’s syndrome 468–71, 470 in Cogan’s syndrome 577
in systemic sclerosis 453 juvenile disease 493 in cryoglobulinaemic vasculitis 560, 561
in Takayasu’s arteritis 293, 324 management 480–1 in cutaneous polyarteritis nodosa 368
in thromboangiitis obliterans 512 biopsy and direct immunofluorescence in cutaneous leukocytoclastic vasculitis 523
in vasculitic neuropathy 169, 171–2 123, 365 in diffuse alveolar haemorrhage 146–7
co-trimoxazole, Pneumocystis jiroveci common morphologies 121–3 in digital ischaemia 221
prophylaxis 425 cryoglobulinaemic vasculitis 125, 553–4 in eosinophilic granulomatosis with polyangiitis
CPVL 478 cutaneous leukocytoclastic vasculitis 123–4, 291, 438
CR1 55, 73 519–20, 521 in granulomatosis with polyangiitis 289, 290,
cranial nerve involvement Degos’ disease 578 420–1, 422, 426
polyarteritis nodosa 336 dermatomyositis 454 remission induction 419
rheumatoid vasculitis 445 eosinophilic granulomatosis with polyangiitis in IgA nephritis 539
Sjögren’s syndrome 451 127, 436–7 in Kawasaki’s disease 378
C-reactive protein (CRP) 48, 277 erythema elevatum diutinum 124, 581 in malignancy-associated vasculitis 585
CNS vasculitis 503 giant cell arteritis 127 in microscopic polyangiitis 291, 356–7
giant cell arteritis 312 granulomatosis with polyangiitis 126–7 in polyarteritis nodosa 184, 198, 292, 345
crescentic glomerulonephritis 108, 199 HIV infection 573–4 in rheumatoid vasculitis 446
animal models 93, 96, 97 hyperimmunoglobulinaemia D syndrome 582 in sarcoidosis 576
see also glomerulonephritis IgA vasculitis 124, 285, 529–30 in Sjögren’s syndrome 452
Creutzfeldt–Jakob disease 173–4 histology 534–5 in systemic lupus erythematosus 448, 449, 450
Crohn’s disease Kawasaki’s disease 128, 287 in Takayasu’s arteritis 293, 324
differentiation from Behçet’s syndrome 476 malignancy-associated vasculitis 583 in vasculitic neuropathy 169–70, 171–2
see also inflammatory bowel disease microscopic polyangiitis 126, 354 CYCLOPS trial 421
cross-sectional imaging 247, 249–50 polyarteritis nodosa 125, 336–7 cytolytic/cytotoxic hypersensitivity reactions 30
antiphospholipid syndrome 260 relapsing polychondritis 453 cytomegalovirus (CMV)
Behçet’s syndrome 261 rheumatoid vasculitis 444, 445 association with vasculitis 82, 572–3
chronic periaortitis 262, 263 sarcoidosis 576 GPA 386, 394
Cogan’s syndrome 262 seronegative spondyloarthropathies 454 in HIV infection 574
computed tomography 247–9, 248 Sjögren’s syndrome 451
eosinophilic granulomatosis with polyangiitis systemic lupus erythematosus 447–8 D
262–4 Takayasu’s arteritis 127, 322 damage assessment 299–300, 301–2
giant cell arteritis 255–8 urticarial vasculitis 124–5 dapsone
granulomatosis with polyangiitis 262–4 cutaneous leukocytoclastic vasculitis (cutaneous in cutaneous polyarteritis nodosa 368
Kawasaki’s disease 260 small-vessel vasculitis) 3–4, 517 in cutaneous leukocytoclastic vasculitis 524
magnetic resonance imaging 249 aetiology 518 in erythema elevatum diutinum 581
microscopic polyangiitis 258–60 classification 517 in IgA vasculitis 286
polyarteritis nodosa 258–60 special subtypes 521 in polyarteritis nodosa 346
rheumatoid arthritis 260 clinical features 123–4, 519–20, 521 in systemic lupus erythematosus 448
systemic lupus erythematosus 260–1 gastrointestinal manifestations 189–90 deaths
Takayasu’s arteritis 250–5 ocular manifestations 137–8 Behçet’s syndrome 480
thromboangiitis obliterans 261–2 differential diagnosis 520–1 microscopic polyangiitis 355
cryocrit 557–8 epidemiology 518 polyarteritis nodosa 342–3
cryofibrinogens 637 evaluation 522 decay-accelerating factor (DAF, CD55) 55
cryoglobulinaemia, paraneoplastic 584 pathogenesis 72, 518–19 deep vein thrombosis (DVT)
cryoglobulinaemic vasculitis pathological features 107, 522–3 antiphospholipid syndrome 617
causes 548–51, 549 systemic involvement 522 Behçet’s syndrome 472
classification criteria 548, 559 treatment 523–4 see also venous thromboembolic disease
clinical features 553–7, 558 cutaneous polyarteritis nodosa (cPAN) Degos’ disease 577–8
cardiopulmonary disease 152 126, 340–1 clinical features 578
cutaneous lesions 125 childhood disease 292, 369 histology 578
gastrointestinal vasculitis 190 clinical features 363–5 pathogenesis 579
ocular manifestations 136 diagnostic approach 367 pathological features 578–9
renal involvement 203, 204 diagnostic criteria 367 treatment 579–80
definition 547 differential diagnosis 365–7 delayed-type hypersensitivity (DTH) 34–5
diagnosis 557–8 epidemiology 363 dendritic cells, role in giant cell arteritis 309, 600
differentiation from IgA vasculitis 538 disease associations and exposures 367–8 Dengue virus infection, association with
epidemiology 547–8 histopathology 365, 366, 367 cryoglobulinaemic vasculitis 550
662 index

deoxyspergualin,in granulomatosis with IgA vasculitis 18, 529, 537 cardiopulmonary disease 143, 144, 150
polyangiitis microscopic polyangiitis 17 cutaneous lesions 127
maintenance of remission 420 pathogenesis 78–80 gastrointestinal vasculitis 185
refractory disease 420, 424 pathological features 114 ocular manifestations 131–2, 136
dermatomyositis (DM) 454–5 duplex ultrasound, in bowel ischaemia 180–1, 182 renal involvement 199–200
gastrointestinal vasculitis 189 diagnosis, ANCA 65
diabetes mellitus, vasculitic neuropathy 173 E differential diagnosis 436, 437–8
diagnosis 275 echocardiography 149 drug associations 435
biopsy 278–9 Kawasaki’s disease 260, 288, 377 epidemiology 17–18, 435
confirmatory tests 278 Takayasu’s arteritis 323 historical background 433
initial approach 275–7 EC-selective adhesion molecule (ESAM) 44 imaging 262, 436, 438
laboratory investigations 277 ectopic lymphoid tissue, granulomatosis with laboratory investigations 438
vascular imaging 279–80 polyangiitis 392 malignancy-associated 584
see also under individual conditions eculizumab, Degos’ disease 579, 580 pathogenesis 72, 434–5
diagnostic criteria 3 Egami score, Kawasaki’s disease 288 association with asthma drugs 79
diffuse alveolar haemorrhage (DAH) 145–7, 146 Ehlers–Danlos syndrome 643 granuloma formation 83
differential diagnosis 147 elastase-ANCA 63, 65 pathological features 107, 110–11, 112, 433, 434
rheumatoid vasculitis 445 elastic lamina disruption, medium-vessel prognosis 439
systemic lupus erythematosus 448–9 vasculitis 105 treatment 438–9
digital gangrene, treatment 219, 220 electroencephalography (EEG), antiphospholipid experimental therapies 605
digital infarction 210, 216 syndrome 621 eosinophils, role in EGPA 434, 435
digital ischaemia 209, 216 electromyography (EMG) 162, 167–8 eosinophil-targeted therapies 605
clinical evaluation Elschnig’s spots 135 ephedrine, vascular effects 643
colour changes 214–15 embolization therapy 227–8 epidemiology 7–8
differential diagnosis 213–14 endarteritis, infectious 570 ANCA-associated vasculitis
physical examination 215 endoglin (CD105) 44 eosinophilic granulomatosis with polyangiitis
cryoglobulinaemic vasculitis 557 endomyocardial biopsy 150 17–18, 435
historical background 209–10 endostatin 47 granulomatosis with polyangiitis 13–16, 289,
investigations 215–16, 217 endothelial cells (ECs) 41–2 385–7, 402–4
angiography 216–17 influence on local blood flow 212 microscopic polyangiitis 16–17, 290, 351
polyarteritis nodosa 338 interaction with ANCA-activated neutrophils antiphospholipid syndrome 616
thromboangiitis obliterans 508 76–7, 200–1, 202 Behçet’s syndrome 467–8
treatment 220–1 leukocyte adhesion 44–5 childhood vasculitis 18–19
guidelines 217–18 release of von Willebrand factor 77 cutaneous leukocytoclastic vasculitis 518
see also Raynaud’s phenomenon retraction and contraction 42 IgA vasculitis 18, 19, 284, 527–8
digital pitting 210 endothelial dysfunction large-vessel vasculitis
digital pulp atrophy 216 AECA-induced 78 giant cell arteritis 8–11, 307–8
digital ulceration, treatment 219 associated factors and mechanisms 41 Takayasu’s arteritis 11, 292, 319
dipyridamole clinical assessment 42, 43–4 medium-vessel vasculitis (MVV)
Degos’ disease 579 complement-induced 55–6 Kawasaki’s disease 11–12, 286, 373
IgA nephritis 539 animal models 56–7 polyarteritis nodosa 12–13, 333, 363
direct ELISA 64 mechanisms 42–3 Raynaud’s phenomenon 211–12
direct immunofluorescence (DIF) 123 in vasculitis 47–8 rheumatoid vasculitis 443
cutaneous polyarteritis nodosa 365 Behçet’s syndrome 477 Sjögren’s syndrome 451
cutaneous leukocytoclastic vasculitis 517, Raynaud’s phenomenon 213 systemic lupus erythematosus 446
522, 523 thromboangiitis obliterans 508 thromboangiitis obliterans 507
disease activity assessment 299 endothelial morphology, changes in vasculitis 42 vasculitic neuropathy 163–4
childhood disease 283–4, 303 endothelial progenitor cells (EPCs) 47 vasculitis associated with specific conditions
giant cell arteritis 315 dysfunction 48 19–20
granulomatosis with polyangiitis 411–12, endothelial regeneration 43 episcleritis 132, 133, 138
418–19 endothelin-1 (ET-1) granulomatosis with polyangiitis 126
imaging 301 inhibition of 46 epoprostenol
laboratory investigations 301 role in vascular injury 43 in Raynaud’s phenomenon 220
uses in vasculitis 300–1 endovascular procedures, thromboangiitis in thromboangiitis obliterans 513
Disease Extent Index 418 obliterans 513 Epstein–Barr virus (EBV) infection 574–5
granulomatosis with polyangiitis 300, 411, 412 ENT assessment, granulomatosis with polyangiitis association with cryoglobulinaemic vasculitis
disease states 303 407 550
dissecting aneurysms ENTAS 411 erythema elevatum diutinum (EED) 124, 138, 521,
giant cell arteritis 256 enterocolic lymphocytic phlebitis (ELP) 190 581
polyarteritis nodosa 338 environmental factors erythema nodosum 367
Doppler ultrasound ANCA-associated vasculitis 66 in Behçet’s syndrome 470
giant cell arteritis 256 eosinophilic granulomatosis with polyangiitis 17 juvenile disease 493
Takayasu’s arteritis 251, 253 giant cell arteritis 10–11 in Takayasu’s arteritis 322
thromboangiitis obliterans 262 granulomatosis with polyangiitis 16, 386–7 erythrocyte sedimentation rate (ESR) 277
drug abuse vasculitis 238, 239 IgA vasculitis 18 CNS vasculitis 503
drug effects, as mimic of vasculitis 643–4 Kawasaki’s disease 12 giant cell arteritis 312
drug-induced vasculitis 148, 387, 410, 522 microscopic polyangiitis 17 E-selectin 42, 44
associated drugs 79 enzyme-linked immunosorbent assays (ELISA) 63–4 etanercept
cutaneous polyarteritis nodosa 368 eosinophil activation, role in IgA vasculitis 534 in Behçet’s syndrome 480
cutaneous leukocytoclastic vasculitis 518 eosinophilic granulomatosis with polyangiitis in granulomatosis with polyangiitis 424, 604
diagnosis 79 (EGPA, Churg–Strauss syndrome) ethnic differences
ANCA 65 childhood disease 291 Behçet’s syndrome 467–8
eosinophilic granulomatosis with polyangiitis classification 433–4 giant cell arteritis 10
17, 435 clinical features 435–7 granulomatosis with polyangiitis 14–15
index 663

IgA vasculitis 18 γδT cells, role in Takayasu’s arteritis 320 polyarteritis nodosa 13
Kawasaki’s disease 12 gastrointestinal involvement 180 Raynaud’s phenomenon 211
microscopic polyangiitis 17 antiphospholipid syndrome 623 Takayasu’s arteritis 319
polyarteritis nodosa 13 Behçet’s syndrome 186–7, 475–6 geographic necrosis, granulomatosis with
Takayasu’s arteritis 11 juvenile disease 493 polyangiitis 108, 109, 387, 388, 391–2
Euro-Lupus study 616 management 482 giant cell arteritis (GCA) 307
European Medicines Agency (EMEA), connective tissue disease 188–9 ACR classification 312
classification of AAV 402 Sjögren’s syndrome 452–3 aetiology 308
surrogate markers 403 systemic lupus erythematosus 261, 448 association with antiphospholipid antibodies
European Vasculitis Study Group (EUVAS), cryoglobulinaemic vasculitis 190, 556 626
categorization of AAV disease stages 402 Degos’ disease 578 clinical features 310–11
evidence, categorization of 422 differential diagnosis and management 181 association with polymyalgia rheumatica 8
experimental therapies eosinophilic granulomatosis with polyangiitis cardiopulmonary disease 148, 149, 151
cryoglobulinaemic vasculitis 601–2 185, 437 cutaneous lesions 127
giant cell arteritis 599–600 giant cell arteritis 182 gastrointestinal vasculitis 182
Takayasu’s arteritis 600–1 granulomatosis with polyangiitis 184–5, 409 ocular manifestations 134
eye IgA vasculitis 186, 530–1 renal involvement 197
vascular systems 131 histology 535 differentiation from Takyasu’s arteritis 4
see also ocular manifestations treatment 538–9 disease evaluation 300, 301
eyelid lesions, granulomatosis with polyangiitis 127 imaging 180–1, 182, 183, 185 epidemiology 19, 307–8
Kawasaki’s disease 184 cardiovascular risk factors 10
F leukocytoclastic vasculitis 189–90 environmental factors 10–11
Fabry’s disease (angiokeratoma corporis diffusum) lymphoproliferative disease 190 ethnic differences 10
641–2 microscopic polyangiitis 185–6, 354 genetic factors 10
factor H, complement regulation 55 polyarteritis nodosa 182–4, 259–60, 338–40 geographical factors 8, 10
factor I, complement regulation 55 management 344 incidence rates 8, 9
familial disease regional (localized) 181 time trends 8
Behçet’s syndrome 477 Takayasu’s arteritis 181, 183 imaging 255–8, 313, 314
juvenile disease 493 thromboangiitis obliterans 187–8, 508 angiography 232–3, 234
IgA vasculitis 284, 528 gastrointestinal system CT angiography 248
familial Mediterranean fever (FMF) 528, 581 blood supply 179 PET 268–9
Fasciola hepatica infection 82 microbiota 179 laboratory investigations 312
fasudil 220 pathophysiology of bowel ischaemia 179–80 malignancy-associated 584
FCGR3B 386 Gell, P.C.G., classification of hypersensitivity management 313–15
Fc receptors 201 reactions 30 experimental therapies 599–600
polymorphisms 80 gender differences in incidence pathogenesis 309–10
role in ANCA-associated vasculitis 603 giant cell arteritis 8 pathological features 101–4, 103, 104, 308
role in hypersensitivity reactions 32, 33 Kawasaki’s disease 11–12, 373 prognosis 315–16
fever, Kawasaki’s disease 287 Raynaud’s phenomenon 211 temporal artery biopsy 312–13
fever of unknown origin, FDG-PET 267 Takayasu’s arteritis 11 giant cells 103, 104
fibrinoid necrosis thromboangiitis obliterans 507 differential diagnosis 101
cutaneous leucocytoclastic vasculitis 107 gene expression, prognostic value 303 granulomatosis with polyangiitis 109, 110
cutaneous polyarteritis nodosa 365, 366, 367 genetic factors 80–1 Takayasu’s arteritis 102
giant cell arteritis 103 ANCA-associated vasculitis 66 gingival lesions, granulomatosis with polyangiitis
granulomatosis with polyangiitis 109, 110 antiphospholipid syndrome 625 127
medium-vessel vasculitis 104, 105 atopy 30 glomerulonephritis
microscopic polyangiitis 106 Behçet’s syndrome 476, 477–8 ANCA-associated 198–200
mimics of vasculitis 117 juvenile disease 493–4 pathophysiology 200–1
fibrinolytic therapy, Behçet’s syndrome 482 complement component deficiencies 73 Behçet’s syndrome 476
fibroblast growth factor (FGF), role in giant cell cryoglobulinaemic vasculitis 549 cryoglobulinaemic vasculitis 203, 204, 555
arteritis 310 eosinophilic granulomatosis with polyangiitis 434 eosinophilic granulomatosis with polyangiitis
fibromuscular dysplasia 643 familial Mediterranean fever 581 437
angiography 241 giant cell arteritis 10, 308 histological classification 391
ficolins 53 granulomatosis with polyangiitis 15–16, 289, IgA vasculitis 203, 532
fingolimod (FTY720) 46 385–6 histology 535–6
Finkelstein’s disease see acute haemorrhagic IgA nephropathy 73 prognosis 540–1
oedema of infancy IgA vasculitis 284, 528 treatment 539–40
fisetin 46 Kawasaki’s disease 12, 286, 375 inflammatory myopathies 454–5
Five Factor Score 302–3, 355–6, 412 microscopic polyangiitis 353 microscopic polyangiitis 355
eosinophilic granulomatosis with polyangiitis rheumatoid vasculitis 444 necrotizing crescentic GN 388–9, 391
438, 439 sarcoidosis 575 Sjögren’s syndrome 452
polyarteritis nodosa 344 Takayasu’s arteritis 11, 319–21 systemic lupus erythematosus 449
fludarabine, cryoglobulinaemic vasculitis 561 thromboangiitis obliterans 507 glucocorticoids see corticosteroids
fluoro-18-deoxyglucose (FDG) 267 genital ulcers, Behçet’s syndrome 470 Goodpasture’s syndrome see anti-glomerular
see also positron emission tomography (PET) juvenile disease 493 basement membrane disease
management 480–1 granulolocyte-colony stimulating factor, cutaneous
G genitourinary involvement, IgA vasculitis 533 polyarteritis nodosa 368
galactose-deficient IgA1 284 geographical variation granuloma formation 83, 127
gallbladder Behçet’s syndrome 467–8 differential diagnosis 101
eosinophilic granulomatosis with polyangiitis 185 eosinophilic granulomatosis with polyangiitis 18 granulomatosis with polyangiitis (GPA, Wegener’s
IgA vasculitis 186 giant cell arteritis 11–12 granulomatosis) 13, 127
microscopic polyangiitis 186 granulomatosis with polyangiitis 14, 402–3 association with antiphospholipid antibodies
polyarteritis nodosa 183, 339, 346 Kawasaki’s disease 11–12, 11–12, 373 626
systemic lupus erythematosus 448 microscopic polyangiitis 15, 16–17 childhood disease 289–90
664 index

granulomatosis with polyangiitis (GPA, Wegener’s granulomatous angiitis of the CNS (GACNS) human T-lymphotropic virus type 1 (HTLV-I)
granulomatosis) (Cont.) 498–9 association with vasculitis 82, 573
classification 401–2 diagnostic studies 502–4 neuropathy 174
in clinical trials 417–18 granulomatous arteritis see giant cell arteritis hydralazine, association with microscopic
clinical features 290, 404–5 growth factors polyangiitis 17
cardiopulmonary disease 143, 143–4, role in angiogenesis 45 hydroxychloroquine, in antiphospholipid
145, 150 role in giant cell arteritis 310 syndrome 626
cutaneous lesions 126–7 hydroxyzine, in cutaneous leukocytoclastic
digital infarction 210 H vasculitis 523, 524
gastrointestinal vasculitis 184–5 haematological malignancy, association with Hygiene Hypothesis 31
ocular manifestations 132, 133, 135–6 vasculitis 83, 583–4 hyperacute rejection 56
renal involvement 198–9 haemoptysis hypercoagulability syndromes 512
complications, treatment and prevention 425, association with pulmonary capillaritis 108 hyperimmunoglobulinaemia D syndrome (HIDS)
426–7 microscopic polyangiitis 354 581–2
diagnosis hairy cell leukaemia, association with vasculitis 83 hyperoxaluria 642
ANCA 64–5, 406–7 Halicephalobus deletrix infection, association with hypersensitivity reactions
clinical examination 405 vasculitis 83 classification 30
differential diagnosis 410–11 halo sign 280, 313 historical background 29–30
disease course and prognosis 413 headache type I 30–2
disease evaluation 300, 411–12 Behçet’s syndrome 474–5 type II 32
biomarkers of disease activity 407 giant cell arteritis 310 type III 32–3
epidemiology 385, 402–4 ‘healed arteritis’, pathological features 308 type IV 33–5
association with malignancy 387 hearing loss, Cogan’s syndrome 576–7 hypersensitivity vasculitis
childhood disease 19 heart failure 148 ACR criteria 517, 518
environmental factors 16, 386–7 microscopic polyangiitis 355 differentiation from IgA vasculitis 536, 537
ethnic factors 14–15 polyarteritis nodosa 338 see also cutaneous leukocytoclastic vasculitis
genetic factors 15–16, 385–6 heat-shock proteins, role in Behçet’s syndrome 477 hypertension 149
geographical factors 14 Helicobacter pylori, association with IgA vasculitis 529 cryoglobulinaemic vasculitis 555
time trends 14, 15 Henoch–Schönlein purpura see IgA vasculitis as mimic of vasculitis 639
historical background 401 heparin, in acute digital ischaemia 217–18 polyarteritis nodosa 337
imaging 262, 407–9 hepatitis A infection, association with Takayasu’s arteritis 292–3, 322
CT 250 cryoglobulinaemic vasculitis 550 hyperviscosity syndrome, cryoglobulinaemic
PET 270, 271 hepatitis B infection, association with vasculitis 557
investigations cryoglobulinaemic vasculitis 550 hypervolaemia induction, thromboangiitis
laboratory investigations 405–6 hepatitis B-related polyarteritis nodosa 13, 81, 333, obliterans 513
renal biopsy 201–2 334, 342 hypocomplementaemic urticarial vasculitis (HUV)
malignancy-associated 584 clinical features 340, 341 448, 521, 580–1
pathological features 108–9, 110, 405 management 345–6 ocular manifestations 136
diagnostic criteria 387 hepatitis C infection 81–2 hypopyon, Behçet’s syndrome 471, 472
glomerulonephritis, histological cryoglobulinaemic vasculitis 547–50 hypothenar hammer syndrome 240, 639
classification 391 pathogenesis 551–2, 553 hypoxia-inducible factors (HIFs)
GPA score 390 prognosis 558–9 inhibition of 46
necrotizing crescentic glomerulonephritis treatment 559–61, 601 role in angiogenesis 45
(NCGN) 388–9, 391 vasculitic neuropathy 173
necrotizing granulomatous inflammation herpes virus infection 82, 334 I
387–8, 389 role in Takayasu’s arteritis 320 IgA vasculitis (Henoch–Schönlein purpura)
vasculitis 388, 390 Heymann nephritis 32 associated conditions 537
pathophysiology 72 histamine trap test 523 association with antiphospholipid antibodies 626
barrier dysfunction 390–1 histology see pathological features classification criteria 536–7
cell-mediated immunity 83 history-taking 275–6 clinical course and outcome 540–1
ectopic lymphoid tissue 392 HLA associations clinical features 284–5, 533
genetic factors 80, 81 antiphospholipid syndrome 625 cardiopulmonary disease 151
granuloma formation 83 Behçet’s syndrome 476, 477, 477–8 cutaneous lesions 124, 529–30
neutrophilic microabscesses and geographic juvenile disease 493–4 gastrointestinal manifestations 186, 530–1
necrosis 391–2 cryoglobulinaemic vasculitis 549 joint manifestations 531
role of ANCA 392–4 eosinophilic granulomatosis with polyangiitis 434 ocular manifestations 136
role of B lymphocytes 395 giant cell arteritis 10, 308 renal involvement 73, 202–3, 531–3
role of T lymphocytes 394–5 granulomatosis with polyangiitis 15, 289, 385–6 definition 527
therapeutic trial design 417–19 IgA vasculitis 284, 528 diagnosis 285–6, 536
treatment 417, 419, 426–7 microscopic polyangiitis 353 differences between adult and childhood disease
azathioprine 422–3 rheumatoid vasculitis 444 537–8
of complications 425, 426–7 Takayasu’s arteritis 11, 319 differential diagnosis 538
corticosteroids 419–20 thromboangiitis obliterans 507 epidemiology 18, 19, 284, 527–8
cyclophosphamide 420–1, 422 Hodgkin’s lymphoma, CNS vasculitis 500 familial Mediterranean fever 581
experimental therapies 602–5 homocystinuria 642 historical background 527
leflunomide 423 Hopkins Lupus Cohort Study 616 laboratory investigations 533–4
maintenance of remission 420 Hughes–Stovin syndrome 115, 472 malignancy-associated 584
methotrexate 422 Hughes’ syndrome see antiphospholipid syndrome pathological features 114, 534–6
mycophenolate mofetil 423 human immunodeficiency virus (HIV) pathophysiology 73, 284, 534
non-medical therapies 424–5 anticardiolipin antibodies 616 genetic factors 284, 528
refractory disease 420, 424 association with vasculitis 82, 573–4 precipitating factors 528–9
remission induction 419 CNS vasculitis 500 prognosis 286
rituximab 421–2 cryoglobulinaemic vasculitis 550 treatment 286, 538–40
trimethoprim/sulfamethoxazole 423–4 vasculitic neuropathy 157–8, 172–3 IgE, type I hypersensitivity reactions 31–2
index 665

IgG4-related disease 30, 455, 582–3, 645–6 infliximab K


IL-6 inhibitors 600 in Behçet’s syndrome 480, 481 karyorrhexis 107
in cryoglobulinaemic vasculitis 602 in giant cell arteritis 315 Kawasaki-like syndrome, HIV infection 573
in Takayasu’s arteritis 324–5, 601 in granulomatosis with polyangiitis 604, 605 Kawasaki’s disease (KD, mucocutaneous lymph
see also tocilizumab refractory disease 420, 424 node syndrome)
iloprost in Kawasaki’s disease 288, 378, 379 animal models 95–6
in Raynaud’s phenomenon 218–19 mechanism of action 46 cardiac evaluation 377
in thromboangiitis obliterans 513 integrins 43, 44 clinical features 287, 376–7
imaging 225, 278, 279–80 role in angiogenesis 46 cardiopulmonary disease 144, 148, 151
antiphospholipid syndrome 620–1 intercellular adhesion molecules (ICAMs) 42, 44 cutaneous lesions 128
cardiopulmonary evaluation 149 role in vasculitis 48 gastrointestinal vasculitis 184
childhood primary angiitis of the central interferon alpha ocular manifestations 135
nervous system 294 in Behçet’s syndrome 481 renal involvement 198
CNS vasculitis 503 in cryoglobulinaemic vasculitis 559–60 diagnosis 287, 375–6, 377
Cogan’s syndrome 577 in cutaneous leukocytoclastic vasculitis 523 epidemiology 11–12, 286, 373
disease evaluation 301 in eosinophilic granulomatosis with polyangiitis historical background 373
eosinophilic granulomatosis with polyangiitis 438, 605 imaging 260
436, 438 role in polyarteritis nodosa 334 angiography 233
giant cell arteritis 313, 314 vascular effects 644 management 288, 378–9
granulomatosis with polyangiitis 289, 407–9 interferon gamma (IFN-γ) follow-up 288
IgA vasculitis 531 role in generalised Shwartzman reaction 29–30 percutaneous endovsacular interventions 380
infectious aortitis 570 role in Takayasu’s arteritis 320 pathogenesis 286–7, 375
interventional radiology techniques 227–8 role in Th1 reactions 34–5 bacterial superantigens 82
Kawasaki’s disease 288, 377 interleukin-1 receptor antagonist deficient mice 94 Epstein–Barr virus infection 574
microscopic polyangiitis 354, 355 interleukin genes, association with Takayasu’s genetic factors 286, 375
polyarteritis nodosa 342 arteritis 11 pathological features 104–5, 106, 373–5
renal involvement 338 interleukins prognosis 288–9, 379–80
positron emission tomography 267–71 IL-2 therapy, cryoglobulinaemic vasculitis 560, relationship to polyarteritis nodosa 375
Takayasu’s arteritis 322–4 601 keratoconjunctivitis sicca (KCS) 132, 138
thromboangiitis obliterans 509, 510 IL-4, role in type I hypersensitivity 31 KIR2DS2 444
see also angiography; computed tomography; IL-5-targeted therapies 605 Koch, R. 29
cross-sectional imaging; magnetic IL-6 Kohlmeier–Degos arteritis (malignant atrophic
resonance imaging; ultrasonography role in cryoglobulinaemic vasculitis 602 papulosis) 173
immune complex-associated vasculitis 4, 7, 332 role in giant cell arteritis 599–600
animal models 94–5 role in Takayasu’s arteritis 601 L
clinical features, ocular manifestations 136 IL-12 laboratory investigations 277
pathogenesis 71–3 role in generalised Shwartzman reaction 29–30 atheroembolic disease 636
see also cutaneous leukocytoclastic vasculitis; role in Th1 reactions 34–5 Behçet’s syndrome 478–9
IgA vasculitis IL-17, role in Kawasaki’s disease 373 childhood primary angiitis of the central
immune complexes 32–3 role in angiogenesis 45 nervous system 293
role in Behçet’s syndrome 477 role in Behçet’s syndrome 478 CNS vasculitis 502–3
role in vasculitic neuropathy 161 role in EGPA 434 cryoglobulinaemic vasculitis 557–8
immunodeficiency-associated vasculitis 575 role in giant cell arteritis 310 cutaneous polyarteritis nodosa 363, 365
Indian Takayasu Arteritis Score 300 role in granulomatosis with polyangiitis 394 cutaneous leukocytoclastic vasculitis 522
infection-related vasculitis 148, 569 role in polyarteritis nodosa 334 disease evaluation 301
angiography 238–9 role in Takayasu’s arteritis 320, 324–5 eosinophilic granulomatosis with polyangiitis 438
antiphospholipid syndrome 625 interstitial lung disease, rheumatoid vasculitis 445 giant cell arteritis 312
associated pathogens 570 interventional radiology techniques 227–8 granulomatosis with polyangiitis 289, 405–6
bacterial endocarditis 569–70 Kawasaki’s disease 379, 380 IgA vasculitis 286, 533–4
cryoglobulinaemic vasculitis 548–50 midaortic syndrome 242 Kawasaki’s disease 288, 376
cutaneous polyarteritis nodosa 368 Takayasu’s arteritis 231–2, 325 microscopic polyangiitis 290, 355
cutaneous leukocytoclastic vasculitis 518 intravenous immunoglobulin (IVIg) polyarteritis nodosa 291, 342
giant cell arteritis 308 in cutaneous polyarteritis nodosa 368–9 Takayasu’s arteritis 293, 322
granulomatosis with polyangiitis 386 in granulomatosis with polyangiitis, refractory thromboangiitis obliterans 509
IgA vasculitis 528–9, 537 disease 424 Lactobacillus casei infection, animal model of
infectious endarteritis and aortitis 570 in IgA nephritis 540 vasculitis 95
mimics of vasculitis 642, 643 in Kawasaki’s disease 184, 288, 378, 379 large-vessel giant cell arteritis (LV-GCA) 311
pathogenesis 81–3 in microscopic polyangiitis 358 large-vessel vasculitis (LVV) 4, 7, 332
animal models 95–6 in vasculitic neuropathy 170 in Behçet’s syndrome 472–3
pathological features 115 intussusception, IgA vasculitis 285, 530, 538 juvenile disease 493
polyarteritis nodosa 333, 334, 342 ischaemia/reperfusion injury, animal models 56–7 management 481–2
rickettsial vasculitis 571 ischaemic optic neuropathy (ION) 134 clinical features, ocular manifestations 134
septic phlebitis 570–1 isolated vasculitis 115–16 epidemiology
Takayasu’s arteritis 320 giant cell arteritis 8–11
viral-induced disease 571–5 J Takayasu’s arteritis 11
see also hepatitis B infection; hepatitis C janus kinase (JAK) inhibitors 603 pathological features 101–4
infection; tuberculosis jaw claudication, giant cell arteritis 310–11 see also giant cell arteritis; Takayasu’s arteritis
inflammatory bowel disease Jenner, E. 29 lectin complement activation pathway 53
ANCA 75 junctional cell adhesion molecules (JAMs) 44 leflunomide
association with cutaneous polyarteritis juvenile Behçet’s syndrome (JBS) 491–2 adverse effects 427
nodosa 368 characteristic features 493 in granulomatosis with polyangiitis 420, 423, 427
association with Takayasu’s arteritis 321, 322 clinical features 492–3, 494 in Takayasu’s arteritis 601
inflammatory myopathies 454–5 immunogenetics 493–4 leishmaniasis, association with cryoglobulinaemic
characteristics of vasculitis 444 management 494 vasculitis 550
666 index

Lemierre’s syndrome 570–1 granulomatosis with polyangiitis 262–4, 407, 408 microaneurysms, polyarteritis nodosa 233, 338,
lenalinomide, in cryoglobulinaemic vasculitis Kawasaki’s disease 260 339–40, 342
561, 602 microscopic polyangiitis 259 microbiome 179
LEPR 386 polyarteritis nodosa 259, 259 microscopic polyangiitis (MPA) 12–13
leprosy 174 muscle involvement 335, 336 childhood disease 290–1
leukocyte–endothelial adhesion 44–5 systemic lupus erythematosus 260 classification criteria 351
leukocyte-mediated vascular injury 42 Takayasu’s arteritis 251, 252, 253 clinical features 353–5
leukocytoclastic vasculitis thromboangiitis obliterans 261–2 cardiopulmonary disease 144, 145, 150
differentiation from IgA vasculitis 536, 538 see also cross-sectional imaging cutaneous lesions 126
gastrointestinal manifestations 189–90 MAINRITSAN trial 421–2 gastrointestinal vasculitis 185–6
malignancy-associated 583 malignancy-associated vasculitis 83, 583, 638–9 ocular manifestations 136
seronegative spondyloarthropathies 454 cryoglobulinaemic vasculitis 549, 551 renal involvement 199
systemic lupus erythematosus 447 cutaneous leukocytoclastic vasculitis 518 diagnosis
see also cutaneous leukocytoclastic vasculitis haematological malignancies 583–4 ANCA 65
leukotriene inhibitors, association with IgA vasculitis 537 differentiation from PAN 331, 333
eosinophilic granulomatosis with paraneoplastic vasculitic neuropathy 172 imaging 258–60
polyangiitis 17 polyarteritis nodosa 334 laboratory investigations 355
leukotriene receptor antagonists (LTRAs), role in solid tumours 584 epidemiology 16–17, 351
EGPA 79, 150, 435 treatment 585 geographical factors 15
leukotrienes, role in type I hypersensitivity 31 malignancy risk, cyclophosphamide 387, 413, time trends 15
levamisole-induced vasculitis 522, 644 420–1 malignancy-associated 584
livedoid vasculitis 619–20 prophylaxis 425 management 355–8
livedo racemosa 364 malignant atrophic papulosis (Kohlmeier–Degos experimental therapies 602–5
livedo reticularis 121 arteritis) 173, 578 outcome and prognosis 355
antiphospholipid syndrome 619 mannan-binding lectin (MBL) 53 pathogenesis 351–3
microscopic polyangiitis 126 Marfan’s syndrome 642–3 pathological features 105, 106, 108, 111, 113
polyarteritis nodosa 126, 337, 364 mass lesion presentation, PACNS 499 midaortic syndrome 241, 242
liver disease mast cells migraine, and antiphospholipid syndrome 621
association with cryoglobulinaemic vasculitis 551 role in Arthus reaction 33 mimics of vasculitis 117, 276, 645
Behçet’s syndrome 187 role in type I hypersensitivity 31 abnormal proteins 637
cryoglobulinaemic vasculitis 555 matrix metalloproteins (MMPs), role in angiography 240–1, 636
eosinophilic granulomatosis with polyangiitis vasculitis 48 atheroembolic disease 636–7
185 giant cell arteritis 310 biochemical abnormalities
imaging 259 McH5/lpr mouse strain 94 alpha-1-antitrypsin deficiency 640
polyarteritis nodosa 184, 339 medial laminar necrosis 308 amyloidosis 640
systemic lupus erythematosus 448 medium-vessel vasculitis (MVV) 4, 7, 332 calciphylaxis 640–1
Loeys–Dietz syndrome 643 clinical features, ocular manifestations 135 Fabry’s disease 641–2
low-antigen-content (LAC) diet, epidemiology homocystinuria 642
cryoglobulinaemic vasculitis 561 Kawasaki’s disease 11–12 hyperoxaluria 642
L-selectin 44 polyarteritis nodosa 12–13 cold exposure 639
lung biopsy 150, 279 pathological features 104–6 congenital and inherited disorders 642–3
lung nodules and cavities 145 see also Kawasaki’s disease; polyarteritis nodosa drug effects 643–4
lupus anticoagulant 615 MEFV gene 581 histological imitators 635
methods of detection 615–16 membrane-attack complex (MAC) 54, 55 IgG4-related disease 645–6
lupus choroidopathy 138 IgA vasculitis 534 infections 642, 643
lupus mesenteric vasculitis (LMV), imaging 261 role in Degos’ disease 579 internal injury 639–40
lupus retinopathy 138 role in endothelial injury 55, 56 moyamoya disease 644–5
Lyme disease 174 membrane-cofactor protein (MCP, CD46) 55 neoplasia 638–9
lymphadenopathy, Kawasaki’s disease 287 membranous nephritis 32 cardiac myxoma 637–8
lymphocytic PACNS 499 meningitis, granulomatosis with polyangiitis 409 non-inflammatory causes of vascular damage 635
lymphomatoid granulomatosis (LG) 574–5 menorrhagia, antiphospholipid syndrome 627 non-vascular disease 646
lymphoproliferative disease MEPEX trial 603 of PACNS 498, 500, 501–2
CNS vasculitis 500 mepolizumab, in eosinophilic granulomatosis with polyvinyl chloride exposure 639
gastrointestinal vasculitis 190 polyangiitis 438–9, 605 radiation exposure 639
lysosome-associated membrane protein 2 mesenteric vasculitis see gastrointestinal reversible cerebral vasoconstriction syndromes 640
(LAMP-2)-ANCA 63, 65, 74 manifestations thromboembolism 637
pathogenic role 66, 74, 394 mesna, bladder cancer prophylaxis 425, 427–8 thrombotic disorders 637
methotrexate trauma 639
M adverse effects 426–7 see also antiphospholipid syndrome
macrophages, pathogenic role 83 in Cogan’s syndrome 577 minocycline
role in giant cell arteritis 309–10 in cutaneous leukocytoclastic vasculitis 523, 524 association with polyarteritis nodosa 368, 644
macular purpura 121, 122 in giant cell arteritis 315 in Behçet’s syndrome 480
MAGIC syndrome 479 in granulomatosis with polyangiitis 290, 422, miscarriage, antiphospholipid syndrome 621–2
magnetic resonance imaging (MRI) 249, 250, 278, 426–7 mizoribine
279–80 maintenance of remission 420 in cutaneous polyarteritis nodosa 368
angiography 181 remission induction 419 in sarcoidosis 576
antiphospholipid syndrome 260, 620–1 in microscopic polyangiitis 291, 357 monoclonal cryoglobulins 552–3
Behçet’s syndrome 261 in polyarteritis nodosa 345, 368 see also cryoglobulins
cardiac 149 in rheumatoid vasculitis 446 mononeuropathy 165
chronic periaortitis 262, 263 in sarcoidosis 576 polyarteritis nodosa 336
CNS vasculitis 503 in Takayasu’s arteritis 293, 324 mononeuropathy multiplex 161, 165
eosinophilic granulomatosis with polyangiitis in vasculitic neuropathy 172 associated diseases 166
262–4 2-methoxyestradiol (2-ME), angiogenesis microscopic polyangiitis 354
giant cell arteritis 256, 257, 313 inhibition 47 polyarteritis nodosa 336
index 667

mortality rates, granulomatosis with see also glomerulonephritis nitroglycerin, topical (MQX-503), in Raynaud’s
polyangiitis 413 necrotizing scleritis 132 phenomenon 219
moyamoya disease 644–5 nephritis-associated plasmin receptor 529 nodules
MRL/MpJ-Faslpr mouse strain 93 nerve biopsy 160, 161–2, 279 pulmonary 145
genetic studies 94 diagnostic sensitivity 163 subcutaneous 122, 125, 126, 127, 337, 363–4, 447
necrotizing glomerulonephritis 94 in polyarteritis nodosa 335 nomenclature of vasculitides 4–5, 8
mucin accumulation, role in Degos’ disease 579 selection of site 167 see also classification of vasculitic syndromes
multiorgan failure, cryoglobulinaemic vasculitis 556 nerve conduction studies 167–8 non-Hodgkin’s lymphoma
muscle biopsy 162, 279 NETosis 389, 392 association with cryoglobulinaemic vasculitis
diagnostic sensitivity 163 role of ANCA 393 551, 556–7
in polyarteritis nodosa 335 neurofibromatosis 643 CNS vasculitis 500
musculoskeletal manifestations, antiphospholipid tubular stenosis 241 non-steroidal anti-inflammatory drugs (NSAIDs)
syndrome 622–3 neurological manifestations in cryoglobulinaemic vasculitis 561
myalgia antiphospholipid syndrome 620–1 in cutaneous polyarteritis nodosa 368
polyarteritis nodosa 335, 336, 364–5 associated diseases 173–4 in cutaneous leukocytoclastic vasculitis 523
see also polymyalgia rheumatica diabetes mellitus 173 in thromboangiitis obliterans 512
mycobacterial infections HCV infection 173 non-systemic vasculitic neuropathy (NSVN)
as mimic of vasculitis 642 HIV infection 172–3 170–2
role in cutaneous polyarteritis nodosa 368 Behçet’s syndrome 474–5 NORAM trial 422
see also tuberculosis juvenile disease 493 NOS2A polymorphism 528
mycophenolate mofetil cryoglobulinaemic vasculitis 555 NOTCH pathway, role in giant cell arteritis 310
adverse effects 427 diagnosis 169 NZB/NZW F1 and NZW/BXSB F1 mice 94
in Behçet’s syndrome 480 electrophysiological findings 167–8
in childhood primary angiitis of the central laboratory investigations 168 O
nervous system 294 muscle biopsy 162 occupational exposure
in cutaneous leukocytoclastic vasculitis 523, 524 nerve biopsy 161–2 role in granulomatosis with polyangiitis 16
in giant cell arteritis 315 sensitivity of nerve and muscle biopsy 163 role in microscopic polyangiitis 17
in granulomatosis with polyangiitis 423, 427 differences between SNV and NSNV 171 ocular manifestations 131
maintenance of remission 420 eosinophilic granulomatosis with Behçet’s syndrome 471–2
refractory disease 420, 424 polyangiitis 437 juvenile disease 493
in IgA nephritis 540 giant cell arteritis 311 management 481
in IgG4-related disease 583 granulomatosis with polyangiitis 409 Cogan’s syndrome 576–7
in microscopic polyangiitis 291, 357 IgA vasculitis 533 large-vessel vasculitis 134
in systemic lupus erythematosus 448 microscopic polyangiitis 354 giant cell arteritis 311
in Takayasu’s arteritis 293 non-systemic vasculitic neuropathy 170–2 medium-vessel vasculitis 135
mycotic aneurysms 115 paraneoplastic 172 Kawasaki’s disease 287
myelodysplastic syndrome, associated vasculitis pathogenesis 160–1 polyarteritis nodosa 340
583, 584 pathological features 159 relapsing polychondritis 453
myeloperoxidase (MPO)-ANCA 62–3, 72–3, 74 ‘active vasculitis’ 158 rheumatoid vasculitis 445
animal models 96–8, 97 axonal degeneration 160 seronegative spondyloarthropathies 454
diagnostic and prognostic value 64–5 criteria of definite and probable vasculitis 158 single-organ vasculitis 137–8
drug-induced vasculitis 79–80, 387 healed vasculitic lesions 158–60 small-vessel vasculitis 135–6
eosinophilic granulomatosis with ‘inactive vasculitis’ 158 granulomatosis with polyangiitis 126–7, 409
polyangiitis 435 patterns of degeneration 160 microscopic polyangiitis 355
glomerulonephritis 199, 200–1 perivascular inflammatory infiltration 157–8 in systemic disease 138
granulomatosis with polyangiitis 406 pattern of involvement 161 systemic lupus erythematosus 450–1
laboratory tests 277 polyarteritis nodosa 335–6, 365 tissues subject to systemic vasculitis 131–4
microscopic polyangiitis 351–2, 355 prognosis 170 variable-vessel vasculitis 137
pathogenic role 66, 75–7 relapsing polychondritis 453 oesophagus
myelosuppression, cyclophosphamide-induced 428 rheumatoid vasculitis 445 Behçet’s syndrome 186
myocardial infarction sarcoidosis 576 polyarteritis nodosa 339
antiphospholipid syndrome 617 Sjögren’s syndrome 451–2 oestrogen, role in Raynaud’s phenomenon 212–13
association with cryoglobulinaemic vasculitis 551 in systemic necrotizing vasculitis omalizumab, association with eosinophilic
Kawasaki’s disease 376–7 associated diseases 164 granulomatosis with polyangiitis 17
myocarditis clinical features 164–7 opportunistic infections, in HIV infection 574
Kawasaki’s disease 375, 377 distribution of neuropathy patterns 165 oral lesions
systemic lupus erythematosus 449 incidence and risk factors 163–4 Behçet’s syndrome 468–70, 469
myopathy treatment 169–70 juvenile disease 492–3
Behçet’s syndrome 474 vulnerability of peripheral nerves 157 management 480–1
cryoglobulinaemic vasculitis 555 neuropsychiatric SLE (NPSLE) 449–50 granulomatosis with polyangiitis 127
protracted febrile myalgia 581 neutropenia, cyclophosphamide-induced 428 hyperimmunoglobulinaemia D syndrome 582
Sjögren’s syndrome 452 neutrophilic microabscesses, granulomatosis with Kawasaki’s disease 287
myxoma, cardiac 637–8 polyangiitis 387, 388, 391–2 orbital lesions 133–4
neutrophil-oriented therapies, ANCA-associated granulomatosis with polyangiitis 126–7, 135,
N vasculitis 602–3 408, 409
nailfold capillaries, examination technique 214, 215 neutrophils polyarteritis nodosa 135
nasal biopsy 279 ANCA-induced effects 77, 200–1, 202, 392–3 orchitis, polyarteritis nodosa 340
nasopharyngeal lesions, granulomatosis with role in Behçet’s syndrome 477 osteoporosis prophylaxis 425
polyangiitis 127 role in immune complex disease 33 outcome parameters, granulomatosis with
necrosis role in vascular injury 42, 76–7 polyangiitis 418
cutaneous 122 NFKB1L1 polymorphism 320 ovarian thrombosis, septic 571
see also fibrinoid necrosis nitric oxide (NO) ovary, polyarteritis nodosa 340
necrotizing crescentic glomerulonephritis (NCGN) role in Raynaud’s phenomenon 213 oxygen, hyperbaric, cutaneous polyarteritis
64, 65, 388–9, 391 role in vascular injury 42 nodosa 368
668 index

P infectious vasculitis 115 phosphodiesterase inhibitors, in Raynaud’s


p38 MAPK inhibition 603 isolated vasculitis 115–16 phenomenon 219, 220
paclitaxel, angiogenesis inhibition 46 large-vessel vasculitis 101–4 physical examination 276–7
paediatric vasculitis activity score (PVAS) 303 giant cell arteritis 308 granulomatosis with polyangiitis 405
pain, neuropathic 170 Takayasu’s arteritis 292, 321 PI3Kγ inhibition 603
pain relief medium-vessel vasculitis 104–6, 183 placenta, in antiphospholipid syndrome 621–2
cryoglobulinaemic vasculitis 561 Kawasaki’s disease 287–8, 373–5 plasma cell-targeted therapies
thromboangiitis obliterans 513 polyarteritis nodosa 334–5, 365, 366, 367 cryoglobulinaemic vasculitis 602
palpable purpura 121, 122, 124 mimics of vasculitis 635 GPA and MPA 604
acute haemorrhagic oedema of infancy 125 sarcoidosis 576 plasma exchange (PLEX)
cryoglobulinaemic vasculitis 125, 553–4 SLE 189 in cryoglobulinaemic vasculitis 560–1
cutaneous polyarteritis nodosa 126 small-vessel vasculitis 106–8 in granulomatosis with polyangiitis 424–5, 603
cutaneous leukocytoclastic vasculitis 519 connective tissue diseases 113–14 remission induction 419
differential diagnosis 538 cryoglobulinaemia 114 in IgA vasculitis 540
eosinophilic granulomatosis with polyangiitis cutaneous 522–3 in Kawasaki’s disease 378
127 drug-induced vasculitis 114 in microscopic polyangiitis 358
granulomatosis with polyangiitis 126 eosinophilic granulomatosis with polyangiitis in polyarteritis nodosa 345
in HIV infection 573–4 110–11, 112, 433, 434 in vasculitic neuropathy 170
IgA vasculitis 124, 285, 529, 530 granulomatosis with polyangiitis 108–10, platelet-activating factor (PAF), role in type I
microscopic polyangiitis 126 387–90, 391, 405 hypersensitivity 31
polyarteritis nodosa 125, 337 IgA vasculitis 114, 203, 285–6, 534–6 platelet activation, role in Raynaud’s phenomenon
pANCA 61–2, 74 microscopic polyangiitis 355 213
see also antineutrophil cytoplasmic antibodies thromboangiitis obliterans 115, 116, 188, 233, platelet-derived growth factor (PDGF), role in
pancreatitis 510–11 giant cell arteritis 310
eosinophilic granulomatosis with polyangiitis vasculitic neuropathy 157–60 platelet-endothelial adhesion molecule 1 (PECAM-
185 pathophysiology 1, CD31) 44
IgA vasculitis 186 antiphospholipid syndrome 625–6 pleural disease 148
microscopic polyangiitis 186 giant cell arteritis 308 eosinophilic granulomatosis with polyangiitis
polyarteritis nodosa 339 granulomatosis with polyangiitis 390 436
papulopustular lesions, Behçet’s syndrome 470–1, barrier dysfunction 390–1 pneumatosis cystoide intestinalis 189
474 ectopic lymphoid tissue 392 Pneumocystis jiroveci pneumonia (PJP) 574
paraneoplastic vasculitic neuropathy 172 neutrophilic microabscesses and geographic prophylaxis 315, 344, 425
paraneoplastic vasculitis see malignancy-associated necrosis 391–2 POEMS (polyneuropathy, organomegaly,
vasculitis role of ANCA 392–4 endocrinopathy, M protein, and skin
parasitic diseases, association with vasculitis 82–3 role of B lymphocytes 395 changes) 637
parenchymal lung disease 144–5 role of T lymphocytes 394–5 polyarteritis nodosa (PAN)
parvovirus B19-associated vasculitis 334, 550, 572 IgA vasculitis 534 aetiology and precipitating factors 334
passive antibody transfer model, MPO-ANCA 96 pauci-immune glomerulonephritis 199, 200 association with antiphospholipid antibodies
pathergy phenomenon, Behçet’s syndrome 471, 478 pegylated interferon, cryoglobulinaemic vasculitis 626
pathogenesis 71 559–60 association with hairy cell leukaemia 83
ANCA 66, 73–7 penis, polyarteritis nodosa 340 CHCC definition 4
anti-endothelial cell antibodies 77–8 pentoxyfilline, cutaneous polyarteritis nodosa 368 childhood disease 291–2, 341–2
Behçet’s syndrome 476–8 percutaneous endovsacular interventions 227–8 classification criteria 331, 333
cell-mediated immunity and granuloma Kawasaki’s disease 379, 380 clinical features 335
formation 83 midaortic syndrome 242 aortic dissection and peripheral vascular
cryoglobulinaemic vasculitis 551–3 Takayasu’s arteritis 231–2, 325 manifestations 338, 339
cutaneous polyarteritis nodosa 368 periaortitis arterial hypertension 337
cutaneous leukocytoclastic vasculitis 518–19 in connective tissue disease 444 bone manifestations 340
drug-induced vasculitis 78–80 in IgG4-related disease 455 cardiac manifestations 338
eosinophilic granulomatosis with polyangiitis imaging 262, 263 cardiopulmonary disease 144, 148, 150
434–5 PET 269, 270 cutaneous lesions 125–6, 336–7
genetic factors 80–1 pericarditis 148 gastrointestinal manifestations 182–4, 338–40
giant cell arteritis 309–10 eosinophilic granulomatosis with polyangiitis HBV-associated disease 340, 341
IgA vasculitis 284 437 localised forms 340–1
immune complexes and complement 71–3 Kawasaki’s disease 377 main organ/system involvements 335
infectious agents 81–3 polyarteritis nodosa 338 neurological manifestations 335–6
Kawasaki’s disease 286–7, 375 systemic lupus erythematosus 449 ocular manifestations 135, 340
microscopic polyangiitis 351–3 peripheral nerves, vulnerability to vasculitic orchitis 340
polyarteritis nodosa 333–4 neuropathy 157 pulmonary manifestations 340
Raynaud’s phenomenon 212–13 peripheral neuropathy renal involvement 198, 337, 338
rheumatoid vasculitis 443–5 polyarteritis nodosa 335–6, 365 ureteral and urogenital manifestations 340
systemic lupus erythematosus 447 relapsing polychondritis 453 diagnostic criteria 333
Takayasu’s arteritis 319–21 rheumatoid vasculitis 445 epidemiology 12–13, 333
thromboangiitis obliterans 507–8 Sjögren’s syndrome 451 familial Mediterranean fever 581
tumour cell-mediated damage 83 systemic lupus erythematosus 450 historical background 4
vasculitic neuropathy 160–1 peripheral ulcerative keratitis (PUK) 132 imaging 258–60
pathological features 101 granulomatosis with polyangiitis 135–6 angiography 233, 235, 338, 339, 342
Behçet’s syndrome 114–15, 187, 470 microscopic polyangiitis 136 laboratory investigations 342
CNS vasculitis 503–4 polyarteritis nodosa 135 malignancy-associated 584
cryoglobulinaemic vasculitis 552, 554, 555 pernio (chilblains) 639 management 344–6
cutaneous lesions 123 peroneal nerve biopsy 161–2 outcome and prognosis 342–4
Degos’ disease 578–9 PEXIVAS trial 603 pathogenesis 333–4
glomerulonephritis 199–200 phaeochromocytoma 638 association with HBV 13, 81
IgG4-related disease 582 phlebitis, septic 570–1 association with HCV 82
index 669

pathological features 104–5, 183, 334–5 glomerulonephritis 200–1 rapidly progressive glomerulonephritis (RPGN)
relationship to Kawasaki’s disease 375 granulomatosis with polyangiitis 406 198–9
see also cutaneous polyarteritis nodosa neutrophil activation and degranulation 392–3 Raynaud’s phenomenon 209, 210
polymyalgia rheumatica (PMR) laboratory tests 277 clinical evaluation
association with giant cell arteritis 8 microscopic polyangiitis 351–2, 355 colour changes 214–15
classification criteria 311 pathogenic role 66, 75–7 differentiation of primary from secondary
clinical features 311–12 animal models 98 disease 214–15
PET 269 proteinase-activated receptors (PAR), role in type I differential diagnosis 213–14
polymyositis (PM) 454–5 hypersensitivity 31 physical examination 215
polyvinyl chloride exposure 639 proteinuria cryoglobulinaemic vasculitis 555
popliteal vascular entrapment syndrome 512 IgA vasculitis 532, 533 epidemiology 211–12
Portier, P. 29 see also renal involvement historical background 209–10
portovenous system, septic thrombophlebitis 571 protracted febrile myalgia (PFM) 581 investigations 215–16, 217
positron emission tomography (PET) 267, 280 PRTN3 15, 386 angiography 216–17, 241, 243
Behçet’s syndrome 270 P-selectin 44, 56 nomenclature 210–11
chronic periaortitis 269, 270 pseudo-obstruction, intestinal, Kawasaki’s disease pathogenesis 212–13
giant cell arteritis 268–9, 313, 314 184 polyarteritis nodosa 338
medium-vessel vasculitis 270, 271 PTPN22 SNP 81, 385, 386 systemic lupus erythematosus 239
polymyalgia rheumatica 268–9 pulmonary arterial hypertension 147 thromboangiitis obliterans, angiography 236
small-vessel vasculitis 270, 271 pulmonary artery lesions 147, 151 treatment 217–20
Takayasu’s arteritis 269, 322, 323–4 Behçet’s syndrome 237–8, 261, 472, 473 reactive oxygen intermediates (ROI), role in
postanginal sepsis (Lemierre’s syndrome) 570–1 management 482 vascular injury 42, 77
postocclusive reactive hyperaemia (PORH) 43–4 Takayasu’s arteritis 254–5, 321, 322, 323 recombinant human granulocyte-colony
post-partum angiopathy 501 pulmonary assessment, granulomatosis with stimulating factor (rhG-CSF) 428
prazosin, in Raynaud’s phenomenon 218 polyangiitis 407–8, 409 refractory disease, granulomatosis with
prednisone see corticosteroids pulmonary capillaritis 108 polyangiitis 420, 424
pregnancy cryoglobulinaemic vasculitis 114 regulatory T cells (Treg)
ANCA-associated vasculitis 75–6 differential diagnosis 147 changes in granulomatosis with polyangiitis 395
antiphospholipid syndrome 621–2 granulomatosis with polyangiitis 109 role in cryoglobulinaemic vasculitis 601
management 627 systemic lupus erythematosus 114 relapses
Takayasu’s arteritis 325 pulmonary fibrosis, microscopic polyangiitis 354 ANCA titres 65–6
primary angiitis of the central nervous system pulmonary function testing 149 IgA vasculitis 540
(PACNS) eosinophilic granulomatosis with polyangiitis 438 microscopic polyangiitis 355
association with varicella zoster infection 500 pulmonary manifestations 144, 150–2 polyarteritis nodosa 342
classification and diagnostic criteria 497–8 antiphospholipid syndrome 622 relapsing polychondritis (RP) 453–4
clinical subsets 498–500 clinical evaluation 149–50, 438 characteristics of vasculitis 444
diagnostic studies 502–4 cryoglobulinaemic vasculitis 556 pathological features 114
differentiation fromRCVS 502 eosinophilic granulomatosis with polyangiitis remission, definition 418
malignancy-associated 584 435–6 remission induction, granulomatosis with
mimics 498, 500, 501–2 hypocomplementaemic urticarial vasculitis 580 polyangiitis 419
treatment and prognosis 504 IgA vasculitis 533 remission maintenance, granulomatosis with
see also reversible cerebral vasoconstriction histology 536 polyangiitis 420
syndromes imaging 259, 260 renal artery stenosis 197–8
primary Raynaud’s phenomenon inflammatory myopathies 454 renal assessment, granulomatosis with polyangiitis
clinical evaluation 214–15 microscopic polyangiitis 354, 355 408–9
nomenclature 211 polyarteritis nodosa 338, 340 renal biopsy 150, 201–2, 279
pathophysiology 212–13 rheumatoid vasculitis 445 cryoglobulinaemic vasculitis 204, 555
see also Raynaud’s phenomenon sarcoidosis 576 granulomatosis with polyangiitis 199
procalcitonin levels, granulomatosis with scope IgA vasculitis 203, 535–6
polyangiitis 407 asthma and airways disease 143–4 microscopic polyangiitis 355
prognosis diffuse alveolar haemorrhage 145–7 renal involvement
Behçet’s syndrome 480 drug toxicity 148 ANCA-associated vasculitis
CNS vasculitis 504 infections 148 comparison of PR3-ANCA and MPO-ANCA
cryoglobulinaemic vasculitis 558–9 lung nodules and cavities 145 associated disease 200
cutaneous polyarteritis nodosa 369 parenchymal lung disease 144–5 eosinophilic granulomatosis with polyangiitis
eosinophilic granulomatosis with polyangiitis pleural disease 148 199–200, 437
439 pulmonary arterial hypertension 147 granulomatosis with polyangiitis 198–9,
giant cell arteritis 315–16 pulmonary artery lesions 147 388–9, 391
granulomatosis with polyangiitis 413 upper respiratory tract disease 143 idiopathic necrotizing and crescentic
IgA vasculitis 540–1 seronegative spondyloarthropathies 454 glomerulonephritis 200
Kawasaki’s disease 379–80 Sjögren’s syndrome 452 microscopic polyangiitis 199, 354
long-term outcomes 303, 304 systemic lupus erythematosus 448 pathophysiology 200–1
microscopic polyangiitis 355–6 pulmonary–renal syndrome 146 antiphospholipid syndrome 623
polyarteritis nodosa 342–4, 344 pulse, loss of 276 atheroembolic disease 636
predictive tools 302–3 Takayasu’s arteritis 322 Behçet’s syndrome 476
thromboangiitis obliterans 512 pulse cyclophosphamide 421 cryoglobulinaemic vasculitis 203, 555
propylthiouracil purine nucleoside phosphorylase (PNP) deficiency giant cell arteritis 197
association with microscopic polyangiitis 17 575 IgA vasculitis 202–3, 285, 531–3
association with vasculitis 79–80, 644 pylephlebitis 571 prognosis 540–1
prostaglandin D2, role in type I hypersensitivity 31 pyoderma gangrenosum, Takayasu’s arteritis 322 treatment 539–40
protease inhibitors 46 imaging 259
proteinase 3 (PR3)-ANCA 62, 74 R inflammatory myopathies 454–5
diagnostic and prognostic value 64–5 rabies 642 Kawasaki’s disease 198
drug-induced vasculitis 387 radiation exposure 639, 640 polyarteritis nodosa 198, 337, 338
670 index

renal involvement (Cont.) scalp necrosis, giant cell arteritis 127 pathological features 106–8
relapsing polychondritis 453 SCG/Kj mouse strain 93–4 Behçet’s disease 114–15
seronegative spondyloarthropathies 454 Schick, B. 29 connective tissue diseases 113–14
Sjögren’s syndrome 452 scleritis 132 cryoglobulinaemia 114
systemic lupus erythematosus 448, 449 granulomatosis with polyangiitis 126 drug-induced vasculitis 114
Takayasu’s arteritis 197–8 polyarteritis nodosa 135 eosinophilic granulomatosis with polyangiitis
renal transplantation rheumatoid arthritis 138 110–11, 112
antiphospholipid syndrome 623 systemic lupus erythematosus 138 granulomatosis with polyangiitis 108–9, 110
association with cryoglobulinaemic vasculitis 551 scleromalacia perforans 132 IgA vasculitis 114
granulomatosis with polyangiitis 425 scopolin 46 see also ANCA-associated vasculitis;
IgA vasculitis 286, 540 scrotal swelling, IgA vasculitis 124 antiglomerular basement membrane
renin–angiotensin system gene polymorphism, seasonality disease; cryoglobulinaemic vasculitis;
role in IgA vasculitis 528 giant cell arteritis 11 eosinophilic granulomatosis with
retiform purpura 121–2 granulomatosis with polyangiitis 16 polyangiitis; granulomatosis with
giant cell arteritis 127 Kawasaki’s disease 12 polyangiitis; hypocomplementaemic
retinal vasculitis 133 secondary Raynaud’s phenomenon urticarial vasculitis; IgA vasculitis;
Behçet’s syndrome 137, 471–2 clinical evaluation 214–15 microscopic polyangiitis
management 481 nomenclature 211 smoking
granulomatosis with polyangiitis 136 pathophysiology 213 and giant cell arteritis 10
polyarteritis nodosa 135 see also Raynaud’s phenomenon and oral ulcers 469
systemic lupus erythematosus 450 selectins 43, 44 and Raynaud’s phenomenon 218
retinocochleocerebral vasculopathy 501 role in angiogenesis 46 and thromboangiitis obliterans 507, 512
retroperitoneal fibrosis sensory neuropathy 164, 166 Sneddon’s syndrome 294, 620
in connective tissue disease 444 Sjögren’s syndrome 451 soluble CD30 levels, granulomatosis with
in IgG4-related disease 455 seronegative spondyloarthropathies (SNSA) 454 polyangiitis 407
reversible cerebral vasoconstriction syndromes characteristics of vasculitis 444 soluble IL-2 receptor levels, granulomatosis with
(RCVS) 497, 501–2, 640 serotonin reuptake inhibitors, in Raynaud’s polyangiitis 407
angiography 504 phenomenon 219 spinal cord vasculitis 499
rheumatoid factor 277 SERPINA1 (α1-AT) 15, 16, 386 spleen
rheumatoid vasculitis 444 deficiency of 80–1, 640 Behçet’s syndrome 187
clinical features 445 serum sickness 29, 32, 33 polyarteritis nodosa 339
gastrointestinal vasculitis 188 animal models 95 systemic lupus erythematosus 448
ocular manifestations 138 pathogenesis 71–2 standing-wave pattern, thromboangiitis obliterans
skin lesions 444 Shwartzman reaction 29–30 237
diagnosis and treatment 446 sickle cell anaemia 637 Staphylococcus aureus
epidemiology 15, 19–20, 443 sildenafil, in Raynaud’s phenomenon 219, 220 antigenic similarity to cPR3 74
imaging 260 silica exposure association with granulomatosis with
angiography 240 role in granulomatosis with polyangiitis 387 polyangiitis 386, 391
pathogenesis 443–5 role in microscopic polyangiitis 17 statins, in Raynaud’s phenomenon 219–20
pathological features 113 single-organ vasculitis 4, 332 stem cell therapy, thromboangiitis obliterans 513
RhoA/Rho kinase inhibition 220 clinical features, ocular manifestations 137–8 stenosis
RhoA/Rho kinase pathway 212 see also cutaneous leucocytoclastic vasculitis; giant cell arteritis 234
ribavirin, cryoglobulinaemic vasculitis 559–60 primary angiitis of the central nervous neurofibromatosis 241
Richet, C. 29 system Takayasu’s arteritis 229–30
rickettsial vasculitis 571 sinuses percutaneous endovsacular interventions
rituximab biopsy of 279 231–2
adverse effects 426 eosinophilic granulomatosis with polyangiitis stents 227
in ANCA-associated vasculitis 76 435–6 drug-eluting 228
eosinophilic granulomatosis with polyangiitis Sjögren’s syndrome strawberry tongue, Kawasaki’s disease 376
291, 438 association with cryoglobulinaemic vasculitis streptococcal infection
granulomatosis with polyangiitis 419, 420, 551 association with polyarteritis nodosa 342
421–2, 424, 426, 604 characteristics of vasculitis 444 role in IgA vasculitis 529
microscopic polyangiitis 357–8, 604 clinical features ‘string of beads’ appearance, fibromuscular
in Cogan’s syndrome 577 cardiopulmonary disease 452 dysplasia 241
in cryoglobulinaemic vasculitis 560, 561, 602 cutaneous lesions 451 stroke, antiphospholipid syndrome 620
in cutaneous leukocytoclastic vasculitis 524 gastrointestinal vasculitis 452–3 subcutaneous nodules 122
in diffuse alveolar haemorrhage 146–7 myopathy 452 polyarteritis nodosa 125, 126, 337, 363–4
in digital ischaemia 221 neurological manifestations 451–2, 501 systemic lupus erythematosus 447
in IgG4-related disease 583 renal involvement 452 Takayasu’s arteritis 127
in Sjögren’s syndrome 451, 452 epidemiology 451 subglottic stenosis, granulomatosis with
in systemic lupus erythematosus 448 pathological features 114 polyangiitis 408, 425
in Takayasu’s arteritis 293, 600–1 skin biopsy 279 sulfapyridine, cutaneous polyarteritis nodosa 368
in vasculitic neuropathy 169–70 cutaneous leukocytoclastic vasculitis 522–3 superantigens, bacterial 82
RITUXVAS trial 421, 604 polyarteritis nodosa 335, 365 superficial fibular nerve biopsy 161–2
Rocky Mountain spotted fever 571 skin lesions see cutaneous lesions sural nerve biopsy 161, 162
small-vessel vasculitis (SVV) 4, 7, 332 surrogate markers 3
S ANCA-associated vasculitis, epidemiology Susac’s syndrome 294, 501
saddle nose deformity, granulomatosis with 13–18 Sweet’s syndrome 471
polyangiitis 126 ANCA targets 62–3 symmetrical polyneuropathy 161, 166
salmonella arteritis 238, 570 classification 517 sympathectomy, thromboangiitis obliterans 513
Sano score, Kawasaki’s disease 288 cutaneous, pathogenesis 72 sympathetic nervous system, control of blood
sarcoidosis 575–6 immune complex-associated, epidemiology vessel calibre 212
neuropathy 174 18, 19 syphilis 82
ocular manifestations 138 ocular manifestations 135–6 aortitis 238
index 671

systemic lupus erythematosus (SLE) role in granulomatosis with polyangiitis 394–5 in systemic lupus erythematosus 448
animal models 93–4 role in microscopic polyangiitis 353 in Takayasu’s arteritis 601
association with cryoglobulinaemic vasculitis 551 role in polyarteritis nodosa 334 topical therapies, mucocutaneous ulcers 480
clinical features 444 role in rheumatoid vasculitis 444 tracheal stenosis 144
cardiac and peripheral vascular role in vascular injury 42, 83 transforming growth factor (TGF)-β, role in
manifestations 449 role in vasculitic neuropathy 161 Kawasaki’s disease 373, 375
cutaneous lesions 367, 447–8 temporal arteritis see giant cell arteritis transplacental transfer, ANCA 75–6
epidemiology 446 temporal artery biopsy 103–4, 134, 278–9, 312–13 transverse myelitis
gastrointestinal manifestations 188–9, 448 pathological features 308 Sjögren’s syndrome 452
neurological manifestations 449–50, 501, in polyarteritis nodosa 335 systemic lupus erythematosus 450
620, 621 sensitivity 267 trauma, as mimic of vasculitis 639
ocular manifestations 138, 450–1 temporal artery imaging 134 treatment see under individual disorders and drugs
pathogenesis 447 ultrasonography 256 trimethoprim/sulfamethoxazole, in granulomatosis
pulmonary manifestations 448–9 Th1 response 34 with polyangiitis 423–4
Raynaud’s phenomenon 217 in Behçet’s syndrome 476 maintenance of remission 420
renal involvement 449 granulomatosis with polyangiitis 394–5 remission induction 419
and hypocomplementaemic urticarial vasculitis role in giant cell arteritis 310 tuberculin reaction 34
580–1 Th17 response tuberculosis
imaging 260–1 in Behçet’s syndrome 476 ANCA 75
angiography 239 granulomatosis with polyangiitis 394–5 role in Takayasu’s arteritis 320
pathological features 113, 114, 189 role in giant cell arteritis 310 tubular stenosis, neurofibromatosis 241
see also antiphospholipid syndrome thalidomide tumour necrosis factor (TNF-α)
systemic rheumatoid vasculitis see rheumatoid in Behçet’s syndrome 480 role in endothelial dysfunction 43, 45, 48
vasculitis in cryoglobulinaemic vasculitis 561, 602 role in generalised Shwartzman reaction 29–30
systemic sclerosis (SSc) 453 in cutaneous leukocytoclastic vasculitis 523 role in IgA vasculitis 534
angiography 240 in systemic lupus erythematosus 448 role in polyarteritis nodosa 334
characteristics of vasculitis 444 thromboangiitis obliterans (TAO, Buerger’s disease) role in Takayasu’s arteritis 320, 324–5
pathological features of vasculitis 114 clinical features 508–9 role in type I hypersensitivity 31
physical examination 215 digital infarction 210 see also TNF inhibitors
Raynaud’s phenomenon gastrointestinal vasculitis 187–8 type I hypersensitivity 30–2
pathophysiology 213 diagnostic criteria 511–12 type II hypersensitivity 32
treatment 218–20 epidemiology 507 type III hypersensitivity 32–3
historical background 507 type IV hypersensitivity 33–5
T imaging 261–2 tyrosine phosphorylation, role in Raynaud’s
tacrolimus angiography 217, 236–7 phenomenon 212
Behçet’s syndrome 481 investigations 509–10
Cogan’s syndrome 577 outcome and prognosis 512 U
tadalafil, in Raynaud’s phenomenon 219 pathogenesis 507–8 UBAC2 478
Takayasu’s arteritis (TAK) pathological features 115, 116, 188, 233, 510–11 UBASH3B 478
aetiology and pathogenesis 319–21 treatment ulcers 122–3, 124
childhood disease 292–3 angiogenesis therapy 513–14 Behçet’s syndrome 468–70, 475–6
classification 323 conservative 512–13 juvenile disease 492–3
clinical features 321–2 surgery 513 management 480–1
cardiopulmonary disease 144, 147, 148, 149, thrombocytopaenia, antiphospholipid syndrome conjunctival 131
151 622, 627 cryoglobulinaemic vasculitis 554
cutaneous lesions 127 thromboembolism 637 cutaneous polyarteritis nodosa 126, 337, 364
gastrointestinal vasculitis 181, 183 thrombolytic therapy 227 cutaneous leukocytoclastic vasculitis 520
ocular manifestations 134 thrombomodulin 43 digital 219
renal involvement 197–8 thrombophlebitis, thromboangiitis obliterans 508 see also digital ischaemia; Raynaud’s
diagnosis 324 thrombotic disorders 637 phenomenon
differentiation from giant cell arteritis 4 antiphospholipid syndrome 617, 619 gastrointestinal 180, 183, 184, 185, 186–7, 475–6
laboratory investigations 322 pathophysiology 625–6 livedoid vasculitis 620
disease evaluation 300 studies on thrombotic risk 618–19 palate 407
epidemiology 11, 19, 319 see also venous thromboembolic disease (VTE) ulinastatin, Kawasaki’s disease 378
historical background 319 thrombotic thrombocytopaenic purpura 637 ultrasonography 247, 280
imaging 250–5, 320, 322–4 time trends giant cell arteritis 256, 258, 313
angiography 228–30 giant cell arteritis 8 IgA vasculitis 530–1
PET 269 granulomatosis with polyangiitis 14, 15 Takayasu’s arteritis 251, 252, 253, 323
malignancy-associated 584 microscopic polyangiitis 15, 16 thromboangiitis obliterans 262, 509
management 324–5 systemic rheumatoid vasculitis 15, 19–20 see also echocardiography
experimental therapies 600–1 TNF inhibitors ultraviolet radiation (UVR) exposure
interventional radiographic treatment 230–2 in Behçet’s syndrome 481 and giant cell arteritis 10
pathogenesis, role of AECA 77–8 in Cogan’s syndrome 577 and granulomatosis with polyangiitis 16
pathological features 101–2, 321 in granulomatosis with polyangiitis 604–5 upper respiratory tract disease 143
prognosis 325 in Kawasaki’s disease 378, 379 granulomatosis with polyangiitis 404
tamoxifen in microscopic polyangiitis 358, 604–5 systemic lupus erythematosus 449
cutaneous polyarteritis nodosa 368 in Takayasu’s arteritis 324–5 ureters
IgG4-related disease 455 vascular effects 644 microscopic polyangiitis 354
T-cell-directed therapies, GPA and MPA 603–4 see also adalimumab; etanercept; infliximab polyarteritis nodosa 340
T-cells tocilizumab urokinase pulse therapy, IgA nephritis 539
γδT cells, role in Takayasu’s arteritis 320 in Behçet’s syndrome 482 urological problems, Behçet’s syndrome 476
role in Behçet’s syndrome 476–7 in cryoglobulinaemic vasculitis 602 urticarial papules 121
role in EGPA 434 in giant cell arteritis 315, 600 urticarial vasculitis 124–5, 521, 580–1
role in giant cell arteritis 310 mechanism of action 46 systemic lupus erythematosus 447–8
672 index

uterus, polyarteritis nodosa 340 vascular leakage, mechanisms 42 visual impairment


uveal tract 131 vascular regeneration 43 Behçet’s syndrome 472
uveitis 133 vascular smooth muscle, defects in scleroderma see also ocular manifestations
Behçet’s disease 137 213 vitamin D, and giant cell arteritis 10
seronegative spondyloarthropathies 454 vasculitis damage index (VDI) 302, 411–12 vitamin D supplementation, Sjögren’s syndrome
prognostic value 303 451
V vasculogenesis 47 vitronectin 55
valvular heart disease 148, 151 abnormalities in vasculitis 48 von Pirquet, C. P. 29
antiphospholipid syndrome 622 vasoconstriction, reversible cerebral von Willebrand factor (vWF), release by
Behçet’s syndrome 473 vasoconstriction syndromes (RCVS) 501–2 endothelial cells 77
Kawasaki’s disease 377 vasodilatation, mechanisms 42
relapsing polychondritis 453 venography 249, 260, 261 W
seronegative spondyloarthropathies 454 venous thromboembolic disease (VTE) 147 Wegener’s granulomatosis see granulomatosis with
systemic lupus erythematosus 449 antiphospholipid syndrome 617, 619 polyangiitis
variable-vessel vasculitis 4, 7, 332 studies on thrombotic risk 618–19 WEGENT trial 422, 423
ocular manifestations 137 Behçet’s syndrome 261, 472–3 WGET (Wegener’s Granulomatosis Etanercept
see also Behçet’s syndrome; Cogan’s syndrome management 481–2 Trial) 424
varicella zoster virus (VZV) systemic lupus erythematosus 260 Wiskott–Aldrich syndrome (WAS) 575
association with vasculitis 82, 573 very late antigen 4 (VLA-4) 44 wound care
CNS vasculitis 500 vessel size, use in diagnosis and classification 4 cutaneous leukocytoclastic vasculitis 524
in HIV infection 574 vestibular symptoms, Cogan’s syndrome 576–7 thromboangiitis obliterans 512
vascular adhesion protein 1 (VAP-1) 44 viral-induced vasculitis 82, 571–2
vascular cell adhesion molecule 1 (VCAM-1) 44 cytomegalovirus 82, 386, 394, 572–3 X
role in vasculitis 48 Epstein–Barr virus 550, 574–5
vascular endothelial growth factor (VEGF) human T-lymphotropic virus type 1 82, xenotransplantation, hyperacute rejection 56
inhibition of 46 174, 573 X-linked lymphoproliferative disorder
role in angiogenesis 45 parvovirus B19 572 (XLP) 575
role in giant cell arteritis 310 varicella zoster virus 573
role in IgA vasculitis 528 see also hepatitis B infection; hepatitis C Z
role in vasculitis 48 infection; herpes virus infection; human
therapeutic use, thromboangiitis obliterans 513 immunodeficiency virus Z score, coronary arteries 376, 379

You might also like