Download as pdf or txt
Download as pdf or txt
You are on page 1of 2511

Harper’s

Textbook of 
Pediatric Dermatology
Harper’s
Textbook of
Pediatric
Dermatology
IN TWO VOLUMES
FOURTH EDITION

EDITED BY

Peter Hoeger
Veronica Kinsler
Albert Yan

EDITORIAL ADVISORS

John Harper
Arnold Oranje

ASSOCIATE EDITORS

Christine Bodemer
Margarita Larralde
David Luk
Vibhu Mendiratta
Diana Purvis
This edition first published 2020 © 2020 John Wiley & Sons Ltd

Edition History
Blackwell Publishing Ltd (1e, 2000 2e, 2006; 3e, 2011)

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, electronic, mechanical, photocopying, recording or
otherwise, except as permitted by law. Advice on how to obtain permission to reuse material
from this title is available at http://www.wiley.com/go/permissions.

The right of Peter Hoeger, Veronica Kinsler, Albert Yan, John Harper and Arnold Oranje to be
identified as the authors of the editorial material in this work has been asserted in accordance
with law.

Registered Offices
John Wiley & Sons, Inc., 111 River Street, Hoboken, NJ 07030, USA
John Wiley & Sons Ltd, The Atrium, Southern Gate, Chichester, West Sussex, PO19 8SQ, UK

Editorial Office
9600 Garsington Road, Oxford, OX4 2DQ, UK

For details of our global editorial offices, customer services, and more information about Wiley
products visit us at www.wiley.com.

Wiley also publishes its books in a variety of electronic formats and by print‐on‐demand. Some
content that appears in standard print versions of this book may not be available in other formats.

Limit of Liability/Disclaimer of Warranty


The contents of this work are intended to further general scientific research, understanding, and
discussion only and are not intended and should not be relied upon as recommending or
promoting scientific method, diagnosis, or treatment by physicians for any particular patient. In
view of ongoing research, equipment modifications, changes in governmental regulations, and
the constant flow of information relating to the use of medicines, equipment, and devices, the
reader is urged to review and evaluate the information provided in the package insert or
instructions for each medicine, equipment, or device for, among other things, any changes in the
instructions or indication of usage and for added warnings and precautions. While the publisher
and authors have used their best efforts in preparing this work, they make no representations or
warranties with respect to the accuracy or completeness of the contents of this work and
specifically disclaim all warranties, including without limitation any implied warranties of
merchantability or fitness for a particular purpose. No warranty may be created or extended by
sales representatives, written sales materials or promotional statements for this work. The fact
that an organization, website, or product is referred to in this work as a citation and/or potential
source of further information does not mean that the publisher and authors endorse the
information or services the organization, website, or product may provide or recommendations it
may make. This work is sold with the understanding that the publisher is not engaged in
rendering professional services. The advice and strategies contained herein may not be suitable
for your situation. You should consult with a specialist where appropriate. Further, readers
should be aware that websites listed in this work may have changed or disappeared between
when this work was written and when it is read. Neither the publisher nor authors shall be liable
for any loss of profit or any other commercial damages, including but not limited to special,
incidental, consequential, or other damages.

Library of Congress Cataloging‐in‐Publication Data


Names: Hoeger, Peter H., editor. | Kinsler, Veronica, editor. | Yan, Albert C., editor.
Title: Harper’s textbook of pediatric dermatology / edited by Peter Hoeger, Veronica Kinsler,
Albert Yan ; editorial advisors, John Harper, Arnold Oranje ; associate editors, Christine
Bodemer, Margarita Larralde, David Luk, Vibhu Mendiratta, Diana Purvis.
Other titles: Textbook of pediatric dermatology
Description: Fourth edition. | Hoboken, NJ : Wiley-Blackwell, 2020. | Includes bibliographical
references and index.
Identifiers: LCCN 2019031032 (print) | ISBN 9781119142195 (hardback) | ISBN 9781119142805
(adobe pdf) | ISBN 9781119142737 (epub)
Subjects: MESH: Skin Diseases | Child | Infant
Classification: LCC RJ511 (print) | LCC RJ511 (ebook) | NLM WS 265 | DDC 618.92/5–dc23
LC record available at https://lccn.loc.gov/2019031032
LC ebook record available at https://lccn.loc.gov/2019031033

Cover image: CP Photo Art/Getty Images


Cover design by Wiley

Set in 9.5/11.5 pt Palatino by SPi Global, Pondicherry, India

10 9 8 7 6 5 4 3 2 1
 v

Contents

List of Contributors, xi 10 Differential Diagnosis of Neonatal Erythroderma, 121


Hagen Ott & Peter H. Hoeger
Preface to the Fourth Edition, xxiv
11 Vesiculopustular, Bullous and Erosive Diseases
Dedication to Arnold P. Oranje, xxv of the Neonate, 134
Caroline Mahon & Anna E. Martinez
Acknowledgements, xxvi
12 Iatrogenic Disorders of the Newborn, 154
List of Abbreviations, xxvii Elia F. Maalouf  & Wilson Lopez

Section 3 Atopic Dermatitis and Related


VOLUME 1 Disorders

Section 1 Development, Structure and 13 Epidemiology of Atopic Dermatitis, 167


Physiology of the Skin Carsten Flohr, Jonathan I. Silverberg, Joy Wan &
Sinéad M. Langan
1 Embryogenesis of the Skin, 1
14 Genetics and Aetiology of Atopic Dermatitis, 184
Lara Wine Lee & Karen A. Holbrook
Elke Rodriguez & Stephan Weidinger
2 Molecular Genetics in Paediatric Dermatology, 36
15 Clinical Features and Diagnostic Criteria of Atopic
Anna C. Thomas & Veronica A. Kinsler
Dermatitis, 193
3 Cutaneous Microbiome, 46 Sinéad M. Langan & Hywel C. Williams
Carrie C. Coughlin & William H. McCoy IV
16 Severity Scoring and Quality of Life Assessment
4 Physiology of Neonatal Skin, 56 in Atopic Dermatitis, 212
Peter H. Hoeger Christian Apfelbacher, Cecilia A.C. (Sanna) Prinsen,
Daniel Heinl & Hywel C. Williams

17 Atopic Dermatitis and Related Disorders:


Section 2 Skin Disorders of the Neonate Special Types of Presentation, 228
and Young Infant Nawaf Almutairi

5 Neonatal Skin Care, 63 18 Atopic Dermatitis: Complications, 245


Peter H. Hoeger Kevin B. Yarbrough & Eric L. Simpson

6 Transient Skin Disorders in the Neonate 19 Management of Atopic Dermatitis, 253


and Young Infant, 72 Lea Solman & Mary Glover
Margarita Larralde & Maria Eugenia Abad

7 Congenital and Acquired Infections in the Neonate, 84


Scott H. James, Nico G. Hartwig, David W. Kimberlin &
Section 4 Other Types of Dermatitis
Peter H. Hoeger
20 Napkin Dermatitis, 265
8 Transplacentally Acquired Dermatoses, 93 Arnold P. Oranje, Ernesto Bonifazi, Paul J. Honig &
Paula Carolina Luna Albert C. Yan

9 Developmental Anomalies, 101 21 Adolescent Seborrhoeic Dermatitis, 279


Marion Wobser & Henning Hamm Roselyn Kellen & Nanette Silverberg
vi Contents

22 Irritant Contact Dermatitis, 287 39 Pitted Keratolysis, Erythrasma and Erysipeloid, 456


David Luk Zhe Xu, Yuanyuan Xiao, Ying Xiu & Lin Ma

23 Allergic Contact Dermatitis, 300 40 Lyme Borreliosis, 463


Sharon E. Jacob, Hannah Hill & Alina Goldenberg Susan O’Connell

24 Hypereosinophilic Disorders, 316 41 Bartonella Infections, 475


Eirini E. Merika & Nerys Roberts Sonia Kamath & Minnelly Luu

25 Juvenile Plantar Dermatosis, 335 42 Mycobacterial Skin Infections, 485


John C. Browning & Margaret Brown G. Sethuraman, Tanvi Dev & V. Ramesh

26 Perioral Dermatitis, 338 43 Rickettsial Disease, 503


Marius Rademaker Arun C. Inamadar & Aparna Palit

44 Endemic Treponematoses: Yaws, Pinta and Endemic


Syphilis, 515
Section 5 Psoriasis Herman Jan H. Engelkens

45 Tropical Ulcer, 523


27 Psoriasis: Epidemiology, 343 Vibhu Mendiratta & Soumya Agarwal
Matthias Augustin & Marc Alexander Radtke

28 Psoriasis: Aetiology and Pathogenesis, 350


Jonathan Barker Section 8 Fungal Skin Infections
29 Psoriasis: Clinical Features and Comorbidities, 354
Derek H. Chu & Kelly M. Cordoro 46 Superficial Fungal Infections, 527
Peter Mayser & Yvonne Gräser
30 Psoriasis: Classification, Scores and Diagnosis, 362
Nirav Patel & Megha Tollefson 47 Deep Fungal Infections, 560
María Teresa García‐Romero
31 Psoriasis: Management, 368
Marieke M.B. Seyger

Section 9 Viral Skin Infections and Opportunistic


Infections
Section 6 Other Papulosquamous Disorders
48 Molluscum Contagiosum, 579
32 Pityriasis Rubra Pilaris, 377 Joachim J. Bugert, Ali Alikhan & Tor Shwayder
Liat Samuelov & Eli Sprecher
49 Human Papillomavirus Infection, 588
33 Lichen Planus, 390 Yun Tong, Stephen K. Tyring & Zsuzsanna Z. Szalai
Vibhu Mendiratta & Sarita Sanke
50 Herpes Simplex Virus Infections, 598
34 Lichen Nitidus, 403 Manuraj Singh, Helen M. Goodyear & Judith Breuer
Jasem M. Alshaiji
51 Varicella Zoster Virus Infections, 612
35 Lichen Striatus, 408 Manuraj Singh & Judith Breuer
Franck Boralevi & Alain Taïeb
52 Poxvirus Infections, 624
36 Pityriasis Rosea, 416 Susan Lewis‐Jones & Jane C. Sterling
Antonio A.T. Chuh & Vijay Zawar
53 HIV and HTLV‐1 Infection, 649
Neil S. Prose, Ncoza C. Dlova, Rosalia A. Ballona &
Coleen K. Cunningham
Section 7 Bacterial Skin Infections 54 Viral Exanthems, 660
Jusleen Ahluwalia, Pamela Gangar & Sheila Fallon
37 Pyodermas and Bacterial Toxin‐mediated Friedlander
Syndromes, 423
James R. Treat, Christian R. Millett, 55 Eruptive Hypomelanosis, 681
Warren R. Heymann & Steven M. Manders Vijay Zawar & Antonio A.T. Chuh

38 Cutaneous Manifestations of Gram‐negative 56 Cutaneous Infections in Immunocompromised


Infections, 434 Children, 684
Saul N. Faust, Diane Gbesemete & Robert S. Heyderman Miriam Weinstein, Hagen Ott & Peter H. Hoeger
Contents vii

Section 10 Parasitic Skin Infestations Section 14  Blistering Disorders


and Sting Reactions
73 Differential Diagnosis of Vesiculobullous Lesions, 859
57 Leishmaniasis, 693 Sharleen F. Hill & Dédée F. Murrell
Bernardo Gontijo & Carolina Talhari
74 Autoimmune Bullous Diseases, 868
58 Helminthic Infections, 702 Nina van Beek & Enno Schmidt
Héctor Cáceres‐Ríos & Felipe Velasquez
75 Childhood Dermatitis Herpetiformis, 898
59 Scabies and Pseudoscabies, 711 Carmen Liy Wong & Irene Lara‐Corrales
Wingfield E. Rehmus & Julie S. Prendiville
76 Epidermolysis Bullosa and Kindler Syndrome, 907
60 Pediculosis and Cimicosis, 723 Jemima E. Mellerio, Anna E. Martinez & Christina Has
Sandipan Dhar & Sahana M. Srinivas

61 Noxious and Venomous Creatures, 733


Kam Lun Ellis Hon, Theresa Ngan Ho Leung & Section 15 Photodermatoses, Photoprotection
Ting Fan Leung and Environmental Skin Disorders
62 Aquatic Dermatoses, 746
77 The Idiopathic Photodermatoses and Skin Testing, 943
Sarah Hill
Erhard Hölzle & Robert Dawe

78 The Porphyrias, 957


Section 11 Urticaria, Erythemas and Drug Jorge Frank
Reactions
79 Photoprotection, 969
Lachlan Warren & Genevieve Casey
63 Urticaria, 751
Bettina Wedi 80 Skin Reactions to Plants, Cold, Heat
and Chemicals, 983
64 Annular Erythemas, 764
Tuyet A. Nguyen, Christopher Lovell &
Kimberly A. Horii
Andrew C. Krakowski
65 Gianotti–Crosti Syndrome, 771
Carlo M. Gelmetti

66 Erythema Multiforme, Stevens–Johnson Syndrome Section 16  Granulomatous Diseases


and Toxic Epidermal Necrolysis, 777
Benjamin S. Daniel, Lizbeth Ruth Wheeler & 81 Sarcoidosis, 995
Dédée F. Murrell Lisa M. Arkin, Julie L. Cantatore‐Francis & Julie V. Schaffer

67 Hypersensitivity Reactions to Drugs, 785 82 Granuloma Annulare, 1006


Mohannad Abu‐Hilal & Neil Shear Annalisa Patrizi & Iria Neri

83 Orofacial Granulomatosis, 1017


Lisa Weibel & Martin Theiler
Section 12  Acne and Acneiform Disorders

68 Acne, 803
Marissa J. Perman, Bodo C. Melnik & Anne W. Lucky Section 17  Neutrophilic Dermatoses
69 Childhood Rosacea, 821
84 Sweet Syndrome, 1023
Clio Dessinioti & Andreas Katsambas
Peter von den Driesch
70 Hidradenitis Suppurativa, 825
85 Pyoderma Gangrenosum, 1027
Peter Theut Riis & Gregor B.E. Jemec
Karolina Gholam

Section 13  Nutritional Disorders


Section 18  Lymphocytic Disorders
71 Skin Manifestations of Nutritional Disorders, 831
Carola Durán McKinster & Luz Orozco‐Covarrubias 86 Pityriasis Lichenoides, 1035
Christine T. Lauren & Maria C. Garzon
72 Skin Manifestations of Paediatric Metabolic
Syndrome, 841 87 Jessner Lymphocytic Infiltrate of the Skin, 1040
Gregor Holzer & Beatrix Volc‐Platzer R.M. Ross Hearn
viii Contents

88 Primary Cutaneous Lymphoma, 1044 102 Panniculitis in Children, 1207


Rebecca Levy & Elena Pope Christine Bodemer

89 Childhood Leukaemias and Lymphomas, 1063 103 Lipodystrophies, 1221


Keith Morley & Jennifer Huang Robert K. Semple

Section 19  Histiocytic Disorders Section 23 Mosaic Disorders, Naevi and


Hamartomas
90 Langerhans Cell Histiocytosis, 1071
Sylvie Fraitag & Jean Donadieu 104 An Introduction to Mosaicism, 1229
Veronica A. Kinsler
91 Juvenile Xanthogranuloma and Other
Non‐Langerhans Cell Histiocytoses, 1078 105 Melanocytic Naevi, 1237
Gudrun Ratzinger & Bernhard W.H. Zelger Veronica A. Kinsler

106 Epidermal Naevi, 1260


Leopold M. Groesser & Christian Hafner
Section 20  Mastocytosis 107 Other Naevi and Hamartomas, 1276
Jonathan A. Dyer
92 Paediatric Mastocytosis, 1097
Laura Polivka & Christine Bodemer 108 Proteus Syndrome and Other Localized Overgrowth
Disorders, 1283
Veronica A. Kinsler

Section 21  Disorders of Connective Tissue 109 Mosaic Disorders of Pigmentation, 1296
Veronica A. Kinsler
93 Ehlers–Danlos Syndromes, 1111
Nigel P. Burrows
Section 24  Nonvascular Skin Tumours
94 Pseudoxanthoma Elasticum and Cutis Laxa, 1125
Sean D. Reynolds & Lionel Bercovitch
110 Differential Diagnosis of Skin Nodules and Cysts, 1313
95 Buschke–Ollendorff Syndrome, Marfan Syndrome Susanne Abraham & Peter H. Hoeger
and Osteogenesis Imperfecta, 1139
111 Adnexal Disorders, 1325
Marc Lacour
Andrew Wang & Robert Sidbury
96 Anetodermas and Atrophodermas, 1151
112 Calcification and Ossification in the Skin, 1338
Marc Lacour
Amanda T. Moon, Albert C. Yan & Eulalia T. Baselga
97 Hyalinoses, Stiff Skin Syndrome and Restrictive
113 Angiolymphoid Hyperplasia with Eosinophilia, 1350
Dermopathy, 1164
Jasem M. Alshaiji
David G. Paige
114 Fibromatoses, 1356
98 Striae in Children and Adolescents, 1172
Jenna L. Streicher, Moise L. Levy & Albert C. Yan
Marcelo Ruvertoni
115 Carcinomas of the Skin, 1370
99 Morphoea (Localized Scleroderma), 1175
Karen Agnew
Despina Eleftheriou & Lindsay Shaw
116 Childhood Melanoma, 1377
100 Systemic Sclerosis in Childhood, 1183
Birgitta Schmidt & Elena B. Hawryluk
Christopher P. Denton & Carol M. Black
117 Other Malignant Skin Tumours, 1382
Index, i1 Andrea Bettina Cervini, Marcela Bocian,
María Marta Bujan & Paola Stefano

Section 25  Vascular Tumours and Malformations


VO L U M E 2
118 Vascular Malformations, 1399
Section 22  Disorders of Fat Tissue Laurence M. Boon & Miikka Vikkula

101 Lipoma and Lipomatosis, 1195 119 Infantile Haemangiomas, 1425


Siriwan Wananukul & Susheera Chatproedprai Anna L. Bruckner, Ilona J. Frieden & Julie Powell
Contents ix

120 Other Vascular Tumours, 1440 135 Focal Dermal Hypoplasia, 1706
Ann M. Kulungowski, Taizo A. Nakano & Bret L. Bostwick, Ignatia B. Van den Veyver &
Anna L. Bruckner V. Reid Sutton

121 Disorders of Lymphatics, 1452 136 Incontinentia Pigmenti, 1718


Arin K. Greene & Jeremy A. Goss Elizabeth A. Jones & Dian Donnai

137 Premature Ageing Syndromes, 1725


Helga V. Toriello & Caleb P. Bupp
Section 26  Disorders of Pigmentation

122 Inherited and Acquired Hyperpigmentation, 1463


Leslie Castelo‐Soccio & Alexis Weymann Perlmutter
Section 29 Genetic Diseases Predisposing
to Malignancy
123 Vitiligo, 1476
Julien Seneschal, Juliette Mazereeuw‐Hautier & 138 Xeroderma Pigmentosum and Related Diseases, 1743
Alain Taïeb Steffen Schubert & Steffen Emmert

124 Albinism, 1486 139 Gorlin (Naevoid Basal Cell Carcinoma) Syndrome, 1769
Fanny Morice‐Picard & Alain Taïeb Kai Ren Ong & Peter A. Farndon

125 Disorders of Hypopigmentation, 1492 140 Rothmund–Thomson Syndrome, Bloom Syndrome,


M.W. Bekkenk & A. Wolkerstorfer Dyskeratosis Congenita, Fanconi Anaemia
and Poikiloderma with Neutropenia, 1786
126 Dyschromatosis, 1499 Lisa L. Wang & Moise L. Levy
Liat Samuelov & Eli Sprecher
141 Other Genetic Disorders Predisposing to Malignancy, 1802
Julie V. Schaffer

Section 27 Disorders of Keratin and


Keratinization Section 30 Neurofibromatosis, RASopathies and
Hamartoma-Overgrowth Syndromes
127 Review of Keratin Disorders, 1515
Maurice A.M. van Steensel & Peter M. Steijlen 142 The Neurofibromatoses, 1823
Amy Theos, Kevin P. Boyd & Bruce R. Korf 
128 Mendelian Disorders of Cornification (MEDOC):
The Keratodermas, 1524 143 Tuberous Sclerosis Complex, 1837
Edel A. O’Toole Francis J. DiMario Jr

129 Mendelian Disorders of Cornification 144 Other RASopathies, 1857


(MEDOC): The Ichthyoses, 1549 Fanny Morice‐Picard
Angela Hernández, Robert Gruber & Vinzenz Oji

130 Keratosis Pilaris and Darier Disease, 1599


Flora B. de Waard‐van der Spek & Arnold P. Oranje
Section 31  Vasculitic and Rheumatic Syndromes

131 The Erythrokeratodermas, 1608 145 Cutaneous Vasculitis, 1865


Juliette Mazereeuw‐Hautier, S. Leclerc‐Mercier & Joyce C. Chang & Pamela F. Weiss
E. Bourrat
146 Purpura Fulminans, 1891
132 Netherton Syndrome, 1613 Michael Levin, Brian Eley & Saul N. Faust
Wei‐Li Di & John Harper
147 Kawasaki Disease, 1906
133 Porokeratosis, 1623 Wynnis L. Tom & Jane C. Burns
Leslie Castelo‐Soccio
148 Polyarteritis Nodosa, Granulomatosis with Polyangiitis
and Microscopic Polyangiitis, 1918
Paul A. Brogan
Section 28 Focal or Generalized Hypoplasia 149 Juvenile Idiopathic Arthritis, Systemic Lupus
and Premature Ageing Erythematosus and Juvenile Dermatomyositis, 1933
Elena Moraitis & Despina Eleftheriou
134 Ectodermal Dysplasias, 1629
Cathal O’Connor, Yuka Asai & Alan D. Irvine 150 Behçet Disease and Relapsing Polychondritis, 1952
Sibel Ersoy‐Evans, Ayşen Karaduman & Seza Özen
x Contents

151 Erythromelalgia, 1961 Section 37 Psychological Aspects of Skin


Nedaa Skeik Disease in Children

165 Assessing and Scoring Life Quality, 2241


Section 32 Cutaneous Manifestations of Andrew Y. Finlay
Systemic Disease
166 Coping with the Burden of Disease, 2255
152 Metabolic Disorders and the Skin, 1965 Sarah L. Chamlin
Fatma Al Jasmi, Hassan Galadari, Peter T. Clayton & 167 Physiological Habits, Self‐Mutilation and Factitious
Emma J. Footitt Disorders, 2262
153 Cystic Fibrosis, 1988 Arnold P. Oranje, Jeroen Novak & Robert A.C. Bilo
Roderic J. Phillips

154 Cutaneous Manifestations of Endocrine Disease, 1993


Devika Icecreamwala & Tor A. Shwayder Section 38  Principles of Treatment in Children

155 Autoinflammatory Diseases and Amyloidosis, 2010 168 Topical Therapy, 2275


Antonio Torrelo, Sergio Hernández‐Ostiz & Teri A. Kahn Johannes Wohlrab
156 Immunodeficiency Syndromes, 2028 169 Systemic Therapy in Paediatric Dermatology, 2282
Julie V. Schaffer, Melanie Makhija & Amy S. Paller Blanca Rosa Del Pozzo‐Magana & Irene Lara‐Corrales
157 Graft‐Versus‐Host Disease, 2067 170 New Genetic Approaches to Treating Diseases
John Harper & Paul Veys of the Skin, 2301
Stephen Hart & Amy Walker

171 Surgical Therapy, 2310


Section 33  The Oral Cavity Julianne A. Mann & Jane S. Bellet

158 The Oral Mucosa and Tongue, 2079 172 Laser Therapy, 2319
Jane Luker & Crispian Scully Samira Batul Syed, Maria Gnarra & Sean Lanigan

173 Sedation and Anaesthesia, 2330


Brenda M. Simpson, Yuin‐Chew Chan &
Lawrence F. Eichenfield
Section 34  Hair, Scalp and Nail Disorders

159 Hair Disorders, 2103


Elise A. Olsen & Matilde Iorizzo Section 39 Diagnostic Procedures in
Dermatology
160 Alopecia Areata, 2139
Kerstin Foitzik‐Lau
174 Approach to the Paediatric Patient, 2341
161 Nail Disorders, 2147 Diana Purvis
Antonella Tosti & Bianca Maria Piraccini
175 Dermoscopy of Melanocytic Lesions in the Paediatric
Population, 2357
Maria L. Marino, Jennifer L. DeFazio, Ralph P. Braun &
Section 35  Anogenital Disease in Children Ashfaq A. Marghoob

176 The Role of Histopathology and Molecular Techniques


162 Genital Disease in Children, 2159
in Paediatric Dermatology, 2378
Gayle O. Fischer
Lori Prok & Adnan Mir
163 Sexually Transmitted Diseases in Children
and Adolescents, 2195
Arnold P. Oranje, Robert A.C. Bilo & Nico G. Hartwig
Section 40 Nursing Care of Cutaneous Disorders
in Children

Section 36 Cutaneous Signs of Child 177 Nursing Care of the Skin in Children, 2393
Maltreatment and Sexual Abuse Bisola Laguda, Hilary Kennedy, Jackie Denyer,
Heulwen Wyatt, Jean Robinson & Karen Pett
164 Maltreatment, Physical and Sexual Abuse, 2219
Bernhard Herrmann Index, i1
xi

List of Contributors

Maria Eugenia Abad, MD Ali Alikhan Rosalia A. Ballona


Dermatology Department University of Cincinnati Division of Dermatology
Hospital Alemán Department of Dermatology Instituto del Salud del Niño
Pediatric Dermatology Department Cincinnati, OH, USA Lima, Peru
Hospital Ramos Mejía
Buenos Aires, Argentina Nawaf Almutairi, MD Jonathan Barker, MD, FRCP,
Professor FRCPath
Susanne Abraham, MD Department of Medicine Professor of Dermatology
Department of Dermatology Faculty of Medicine St John’s Institute of Dermatology (King’s
Medical Faculty Carl‐Gustav‐Carus Kuwait University College)
Technical University of Dresden Kuwait Guy’s Hospital
Dresden, Germany London, UK
Jasem M. Alshaiji, MD
Mohannad Abu‐Hilal, MD Head of Dermatology Department Eulalia T. Baselga, MD
Assistant Professor Head of Pediatric Dermatology Unit Pediatric Dermatology Unit
Division of Dermatology Amiri Hospital Hospital de la Santa Creu I Sant Pau
Department of Medicine Kuwait Universitat Autònoma de Barcelona
McMaster University Spain
Hamilton, ON, Canada Christian Apfelbacher, PhD
Medical Sociology M.W. Bekkenk, MD, PhD
Soumya Agarwal, MD Institute of Epidemiology and Preventive Dermatologist
Senior Resident Medicine Netherlands Institute for Pigment Disorders
Department of Dermatology University of Regensburg Amsterdam University Medical Centers
Lady Hardinge Medical College and Regensburg, Germany Amsterdam, The Netherlands
Associated Hospitals
New Delhi, India Lisa M. Arkin, MD Jane S. Bellet, MD
Department of Dermatology Associate Professor of Dermatology and
Karen Agnew, MBChB, FRACP, University of Wisconsin School of Medicine Pediatrics
FNZDS and Public Health Duke University Medical Center
Consultant Dermatologist Madison, WI, USA Durham, NC, USA
Starship Children’s and Auckland City
Hospitals Yuka Asai, MSc, PhD, MD Lionel Bercovitch, MD
Auckland, New Zealand
Assistant Professor Professor of Dermatology
Division of Dermatology Warren Alpert Medical School of Brown
Jusleen Ahluwalia, MD Queen’s University University
Resident Physician Kingston, ON, Canada Director of Pediatric Dermatology
Department of Pediatric and Adolescent Hasbro Children’s Hospital
Dermatology Matthias Augustin, MD Providence, RI, USA
Rady Children’s Hospital Medical Director
Professor
San Diego, CA, USA PXE International, Inc.
Institute for Health Services Research in
Washington, DC, USA
Dermatology and Nursing (IVDP)
Fatma Al Jasmi, MBBS, FRCPC, University Medical Center Hamburg‐
FCCMG Eppendorf (UKE) Robert A.C. Bilo
Associate Professor Hamburg, Germany Department of Forensic Medicine
College of Medicine and Health Science Section on Forensic Pediatrics
United Arab Emirates University Netherlands Forensic Institute
Al Ain, United Arab Emirates The Hague, The Netherlands
xii  List of Contributors

Carol M. Black, DBE, MD, FRCP, Ralph P. Braun, MD Jane C. Burns, MD


MACP, FMedSci Dermatology Clinic Professor of Pediatrics
Principal of Newnham College Cambridge, University Hospital Zürich Director, Kawasaki Disease Research Center
Expert Adviser on Health and Work to NHS Zürich, Switzerland University of California, San Diego
England and Public Health England, Chair of Rady Children’s Hospital
Think Ahead, Chair of the British Library Judith Breuer, MBBS, MD, La Jolla, CA, USA
Centre for Rheumatology FRCPath
Royal Free Hospital and UCL Division of
Professor of Virology Nigel P. Burrows, MBBS, MD,
Medicine FRCP
UCL
London, UK
Honorary Consultant Virologist Consultant Dermatologist and Associated
Great Ormond Street Hospital Lecturer
Christine Bodemer, MD, PhD UCL Division of Infection and Immunity Department of Dermatology
Professor of Dermatology London, UK Addenbrooke’s Hospital
Department of Dermatology Cambridge University Hospitals NHS
Imagine Institute Paul A. Brogan, MBChB, Foundation Trust
Necker‐Enfants Malades Hospital Cambridge, UK
Paris, France
FRCPCH, PhD
Professor of Vasculitis and Honorary
Consultant Paediatric Rheumatologist Héctor Cáceres‐Ríos, MD
Marcela Bocian, MD Section Head: Infection and Inflammation and Consultant in Pediatric Dermatology
Assistant Physician Rheumatology Department of Pediatric Dermatology
Dermatology Department Co‐Director of Education (Clinical Academics) Instituto de Salud del Niño
Hospital de Pediatría ‘Prof. Dr. Juan P. Garrahan’ UCL Institute of Child Health Lima, Peru
Buenos Aires, Argentina Great Ormond Street Hospital NHS
Foundation Trust Julie L. Cantatore‐Francis, MD
Ernesto Bonifazi, MD London, UK
Dermatology Physicians of Connecticut
Professor of Dermatology Shelton, CT, USA
Dermatologia Pediatrica Association Margaret Brown, MD
Bari, Italy Division of Dermatology Genevieve Casey
The University of Texas Health Science Center
Specialist Registrar
Laurence M. Boon, MD, PhD at San Antonio
Department of Dermatology
San Antonio, TX, USA
Coordinator of the Center for Vascular Women’s & Children’s Hospital
Anomalies Adelaide, SA, Australia
Division of Plastic Surgery John C. Browning, MD, FAAD,
Cliniques Universitaires Saint Luc and FAAP Leslie Castelo‐Soccio, MD, PhD
Human Molecular Genetics Assistant Professor Professor of Pediatrics and Dermatology
de Duve Institute Baylor College of Medicine Department of Pediatrics
University of Louvain Chief of Dermatology Section of Pediatric Dermatology
Brussels, Belgium Children’s Hospital of San Antonio University of Pennsylvania Perelman School
San Antonio, TX, USA of Medicine and Children’s Hospital of
Franck Boralevi, MD, PhD Philadelphia
Pediatric Dermatology Unit Anna L. Bruckner, MD, MSCS Philadelphia, PA, USA
Hôpital Pellegrin‐Enfants Associate Professor of Dermatology and
Bordeaux, France Pediatrics Andrea Bettina Cervini, MD
University of Colorado School of Medicine Dermatologist, Pediatric Dermatologist
Bret L. Bostwick, MD Section Head Head of Dermatology Department
Assistant Professor Division of Dermatology Hospital de Pediatría ‘Prof. Dr. Juan P.
Department of Molecular and Human Children’s Hospital Colorado Garrahan’
Genetics Aurora, CO, USA Buenos Aires, Argentina
Baylor College of Medicine and Texas
Children’s Hospital Joachim J. Bugert, MD, PhD Sarah L. Chamlin, MD
Houston, TX, USA Lab Group Leader Professor of Pediatrics and Dermatology
Institut für Mikrobiologie der Bundeswehr The Ann and Robert H. Lurie Children’s
E. Bourrat, MD München, Germany Hospital of Chicago and Northwestern
Reference Center for Inherited Skin Disease University
Dermatology Department María Marta Bujan, MD Feinberg School of Medicine
CHU Saint Louis Assistant Physician Chicago, IL, USA
Paris, France Dermatology Department
Hospital de Pediatría ‘Prof. Dr. Juan P. Garrahan’ Yuin‐Chew Chan
Kevin P. Boyd, MD Buenos Aires, Argentina Dermatologist
Clinical Assistant Professor Dermatology Associates
University of Alabama at Birmingham Caleb P. Bupp, MD Gleneagles Medical Centre
Birmingham, AL, USA Medical Geneticist Singapore
Spectrum Health Medical Group
Grand Rapids, MI, USA
List of Contributors  xiii

Joyce C. Chang, MD Robert Dawe, MBCh, MD, FRCPE Francis J. DiMario Jr, MD
Instructor Photodermatology Unit Professor of Pediatrics and Neurology
Division of Rheumatology Department of Dermatology University of Connecticut School of Medicine
The Children’s Hospital of Philadelphia Ninewells Hospital and Medical School Farmington, CT, USA
Philadelphia, PA, USA Dundee, UK Associate Chair for Academic Affairs,
Department of Pediatrics,
Susheera Chatproedprai, MD Jennifer L. DeFazio, MD Director, Neurogenetic‐Tuberous Sclerosis Clinic
Division of Pediatric Neurology
Associate Professor of Paediatrics Department of Dermatology
Connecticut Children’s Medical Center
Head of Division of Paediatric Dermatology Memorial Sloan‐Kettering Cancer Center
Hartford, CT, USA
Department of Paediatrics New York, NY, USA
Faculty of Medicine
Chulalongkorn University, Blanca Rosa Del Pozzo‐Magana Ncoza C. Dlova
Bangkok, Thailand London Health Sciences Center and Western Department of Dermatology
University University of Kwazulu‐Natal
Derek H. Chu, MD London, ON, Canada Durban, South Africa

Clinical Assistant Professor of Dermatology


and Pediatrics Christopher P. Denton, PhD, Jean Donadieu, MD, PhD
Stanford University School of Medicine FRCP Service d’Hémato‐Oncologie Pédiatrique
Palo Alto, CA, USA Registre des Histiocytoses
Professor of Experimental Rheumatology
Centre de Référence des Histiocytoses
Centre for Rheumatology
Antonio A.T. Chuh, MD Hopital Trousseau
Royal Free Hospital and UCL Division of
Paris, France
Department of Family Medicine Medicine
and Primary Care London, UK
The University of Hong Kong and Queen Dian Donnai, CBE, FMedSci,
Mary Hospital Jackie Denyer FRCP, FRCOG
Pokfulam, Hong Kong Clinical Nurse Specialist in Paediatric Professor of Medical Genetics
JC School of Public Health and Primary Care Dermatology Manchester Centre for Genomic Medicine
The Chinese University of Hong Kong and the Great Ormond Street Hospital St Mary’s Hospital
Prince of Wales Hospital London, UK Manchester University NHS Foundation Trust
Shatin, Hong Kong Manchester, UK
Division of Evolution and Genomic Sciences
Clio Dessinioti
Peter T. Clayton, BA, MBBS, Department of Dermatology
Faculty of Biology Medicine and Health
MSc, MRCP University of Manchester
Andreas Syggros Hospital
Manchester, UK
Professor University of Athens
Institute of Child Health Greece
University College London with Great Carola Durán McKinster, MD
Ormond Street Hospital for Children Tanvi Dev, MD Paediatric Dermatologist and Professor of
NHS Trust Pediatric Dermatology
Senior Resident (Fellow)
London, UK Universidad Nacional Autonoma de México
Department of Dermatology
Head of the Department of Pediatric
All India Institute of Medical Sciences
Kelly M. Cordoro, MD Dermatology
New Delhi, India
National Institute of Paediatrics of Mexico
Associate Professor of Dermatology and
Mexico City, Mexico
Pediatrics Flora B. de Waard‐van der
University of California San Francisco Spek, MD, PhD
San Francisco, CA, USA Jonathan A. Dyer, MD
Paediatric Dermatologist
Associate Professor of Dermatology and
Department of Dermatology
Carrie C. Coughlin, MD Child Health
Franciscus Gasthuis and Vlietland
Departments of Dermatology and Child Health
Assistant Professor Rotterdam/Schiedam, The Netherlands
University of Missouri
Division of Dermatology
Columbia, MO, USA
Department of Medicine and Department of Sandipan Dhar, MBBS, MD,
Pediatrics
DNB, FRCP(Edin) Lawrence F. Eichenfield, MD
Washington University School of Medicine
Professor and Head
St Louis, MO, USA Professor of Clinical Dermatology
Department of Pediatric Dermatology
Pediatric and Adolescent Dermatology
Institute of Child Health
Coleen K. Cunningham Rady Children’s Hospital
Kolkata, West Bengal, India
San Diego University of California
Department of Pediatrics and Dermatology
San Diego School of Medicine
Duke University Medical Center Wei‐Li Di, MBBS, PhD San Diego, CA, USA
Durham, NC, USA
Associate Professor in Skin Biology
Infection, Immunity and Inflammation
Benjamin S. Daniel Programme
Department of Dermatology Immunobiology Section
St George Hospital and University of New Institute of Child Health
South Wales University College London
Sydney, NSW, Australia London, UK
xiv  List of Contributors

Despina Eleftheriou, MBBS, Gayle O. Fischer, MBBS, FACD, Pamela Gangar, MD


PhD, MRCPCH MD Resident Physician
Associate Professor in Paediatric Rheumatology Associate Professor in Dermatology University of Arizona Department of Pediatrics
Infection, Inflammation and Rheumatology The Northern Clinical School Tucson, AZ, USA
Section The University of Sydney
UCL Institute of Child Health Sydney, NSW, Australia María Teresa García‐Romero,
Paediatric Rheumatology Department, Great MD, MPH
Ormond Street Hospital for Children NHS Carsten Flohr, MD, PhD Attending Physician
Foundation Trust
Professor of Dermatology Department of Dermatology
Arthritis Research UK Centre for Adolescent
Unit for Population‐Based Dermatology National Institute for Pediatrics
Rheumatology
Research Member of the National System of Researchers
London, UK
St John’s Institute of Dermatology Mexico City, Mexico
Guy’s and St Thomas’ NHS Foundation Trust
Brian Eley, BSc (Hons) (Med and King’s College Maria C. Garzon, MD
Biochem), MBChB (Cape Town), London, UK
Columbia University Medical Center
FCP (SA) New York, NY, USA
Professor of Paediatric Infectious Diseases Kerstin Foitzik‐Lau, MD
University of Cape Town Physician
South Africa
Diane Gbesemete, BM,
Skin and Vein Clinic Winterhude
Hamburg, Germany
MRCPCH, PGDipID
Clinical Research Fellow
Steffen Emmert, MD
NIHR Southampton Clinical Research Facility
Professor of Dermatology Emma J. Footitt, MB, BS, BSc, University of Southampton and University
Director PhD Hospital Southampton NHS Foundation Trust
Clinic for Dermatology and Venereology
Institute of Child Health Southampton, UK
University Medical Center Rostock
University College London with Great Ormond
Rostock, Germany
Street Hospital for Children NHS Trust Carlo M. Gelmetti
London, UK
Professor of Dermatology and Venereology
Herman Jan H. Engelkens, MD,
Department of Pathophysiology and
PhD Sylvie Fraitag, MD Transplantation
Department of Dermatology and Venereology Dermatopathologie Pédiatrique Università degli Studi di Milano
Ikazia Hospital Service d’Anatomo‐Pathologie Head
Rotterdam, The Netherlands Hôpital Necker‐Enfants Malades Unit of Pediatric Dermatology
Paris, France Fondazione IRCCS Ca’ Granda ‘Ospedale
Sibel Ersoy‐Evans, MD Maggiore Policlinico’
Professor of Dermatology Jorge Frank, MD Milan, Italy
Hacettepe University School of Medicine Professor of Dermatology
Department of Dermatology Department of Dermatology, Venereology Karolina Gholam, MBSS, MSc,
Ankara, Turkey and Allergology FRCPCH, SCEderm
University Medical Center Göttingen Consultant Paediatric Dermatologist
Peter A. Farndon, MSc, MD, Göttingen, Germany Great Ormond Street Hospital
FRCP London, UK
Professor of Clinical Genetics (Retired) llona J. Frieden, MD
University of Birmingham Professor of Dermatology and Pediatrics Mary Glover, MA, FRCP, FRCPCH
Birmingham, UK Division of Pediatric Dermatology Consultant Paediatric Dermatologist
San Francisco School of Medicine Great Ormond Street Hospital for Children
Saul N. Faust, FRCPCH, PhD University of California NHS Foundation Trust
San Francisco, CA, USA London, UK
Professor of Paediatric Immunology and
Infectious Diseases and Director of the NIHR
Southampton Clinical Research Facility Sheila Fallon Friedlander, MD Maria Gnarra, MD, PhD
University of Southampton and University Professor of Dermatology and Pediatrics Research Fellow
Hospital Southampton NHS Foundation Trust Department of Pediatric and Adolescent Paediatric Dermatology
Southampton, UK Dermatology Great Ormond Street Hospital for Children
Rady Children’s Hospital NHS Trust
Andrew Y. Finlay, CBE, FRCP San Diego, CA, USA London, UK
(Lond. and Glasg.)
Professor of Dermatology Hassan Galadari, MD Alina Goldenberg, MD
Division of Infection and Immunity Associate Professor Resident in‐training
Cardiff University School of Medicine College of Medicine and Health Science Department of Dermatology
Cardiff, UK United Arab Emirates University University of California
Al Ain, United Arab Emirates San Diego, CA, USA
List of Contributors  xv

Bernardo Gontijo, MD, PhD John Harper, MBBS, MD, FRCP, Bernhard Herrmann, MD
Professor of Dermatology FRCPCH Consultant
Federal University of Minas Gerais Honorary Professor of Paediatric Child Protection Center
Medical School Dermatology Pediatric and Adolescent Gynecology
Belo Horizonte, MG, Brazil Great Ormond Street Hospital for Children Department of Pediatrics
NHS Trust Klinikum Kassel
Helen M. Goodyear, MB, ChB, London, UK Kassel, Germany

FRCP, FRCPCH, MD, MMEd, MA


Health Education England (West Midlands) Stephen Hart, PhD Robert S. Heyderman, PhD,
Associate Postgraduate Dean Professor in Molecular Genetics FRCP, DTM & H
Heart of England NHS Foundation Trust Experimental and Personalised Medicine Professor of Infectious Diseases
Birmingham, UK UCL Great Ormond Street Institute of University College London
Child Health London, UK
Jeremy A. Goss, MD London, UK

Research Fellow Warren R. Heymann, MD


Department of Plastic and Oral Surgery Nico G. Hartwig, MD, PhD Head, Division of Dermatology,
Vascular Anomalies Center Department of Paediatrics Clinical Professor of Dermatology
Boston Children’s Hospital Franciscus Gasthuis & Vlietland University of Pennsylvania School of
Harvard Medical School Rotterdam, The Netherlands Medicine
Boston, MA, USA Professor of Medicine and Paediatrics
Christina Has, MD University of Medicine and Dentistry of
Yvonne Gräser, PhD New Jersey
Consultant Dermatologist and Professor
Robert Wood Johnson Medical School
Professor of Molecular Mycology Molecular Dermatology
Camden, NJ, USA
The National Reference Laboratory for Medical Center
Dermatophytes University of Freiburg
Universitätsmedizin – Charité Freiburg, Germany Hannah Hill, MD
Institute of Microbiology and Hygiene Resident in‐training
Berlin, Germany Elena B. Hawryluk, MD, PhD Department of Dermatology
Mayo Clinic
Department of Dermatology
Arin K. Greene, MD, MMSc Scottsdale, AZ, USA
Massachusetts General Hospital
Professor of Surgery Harvard Medical School;
Department of Plastic and Oral Surgery Dermatology Program Sarah Hill, MBChB, FRACP
Vascular Anomalies Center Division of Allergy and Immunology Paediatric and General Dermatologist
Boston Children’s Hospital Department of Medicine Department of Dermatology
Harvard Medical School Boston Children’s Hospital Waikato Hospital
Boston, MA, USA Harvard Medical School Hamilton, New Zealand
Boston, MA, USA
Leopold M. Groesser, Dr med. Sharleen F. Hill, BM BSc MRCP
Department of Dermatology R.M. Ross Hearn Dermatology Clinical Research Fellow
University of Regensburg Department of Dermatology and St George Hospital
Regensburg, Germany Photobiology Conjoint Associate Lecturer
Ninewells Hospital and Medical School University of New South Wales
Robert Gruber, MD Dundee, UK Sydney, NSW, Australia

Department of Dermatology and Division of


Human Genetics Daniel Heinl, MD Peter H. Hoeger, MD
Medical University of Innsbruck Medical Sociology Professor of Paediatrics and Dermatology
Innsbruck, Austria Institute of Epidemiology and Preventive (University of Hamburg)
Medicine Head, Departments of Paediatrics and
Christian Hafner, Dr med. University of Regensburg Dermatology
Regensburg, Germany Catholic Children’s Hospital, Wilhelmstift
Professor of Dermatology
Hamburg, Germany
Department of Dermatology
University of Regensburg Angela Hernández, MD
Regensburg, Germany Department of Dermatology Karen A. Holbrook, MD
Hospital Infantil del Niño Jesús (Retired)
Henning Hamm, MD Madrid, Spain Department of Physiology and Cell Biology
Ohio State University
Professor of Dermatology
Sergio Hernández‐Ostiz, MD Columbus, OH, USA
Department of Dermatology, Venereology and
Allergology Department of Dermatology
University Hospital Würzburg Hospital Infantil del Niño Jesús
Würzburg, Germany Madrid, Spain
xvi  List of Contributors

Gregor Holzer, MD Arun C. Inamadar, MD, FRCP Ayşen Karaduman, MD


Department of Dermatology Professor and Head
Professor of Dermatology
Donauspital SMZ Ost Department of Dermatology,
Hacettepe University School of Medicine
Vienna, Austria Venereology & Leprosy
Department of Dermatology
Sri B.M.Patil Medical College
Ankara, Turkey
Hospital & Research Centre
Erhard Hölzle, MD BLDE University
Professor of Dermatology Vijayapur, Karnataka, India Andreas Katsambas
Director Professor of Dermatology
Department of Dermatology and Allergology
University Hospital
Matilde Iorizzo, MD Department of Dermatology
Andreas Syggros Hospital
Oldenburg, Germany Private Dermatology Practice
University of Athens
Bellinzona and Lugano
Greece
Switzerland
Kam Lun Ellis Hon, MBBS,
MD, FAAP, FCCM, FHKCPaed, Roselyn Kellen, MD
Alan D. Irvine, MD, FRCPI, FRCP
FHKAM(Paed) Resident Physician
Professor
Honorary Professor Icahn School of Medicine at Mount Sinai
Paediatric Dermatology
Department of Paediatrics, The Chinese New York, NY, USA
Trinity College Dublin and Our Lady’s
University of Hong Kong
Children’s Hospital
Consultant
The Hong Kong Children’s Hospital
Dublin, Ireland Hilary Kennedy
Hong Kong Clinical Nurse Specialist in Paediatric
Sharon E. Jacob, MD Dermatology
Professor Great Ormond Street Hospital
Paul J. Honig, MD London, UK
Department of Dermatology
Division of Dermatology
Loma Linda University
Denver Children’s Hospital
Denver, CO, USA
Loma Linda, CA, USA David W. Kimberlin, MD
Department of Pediatrics Department of Pediatrics
Perelman School of Medicine at the University Scott H. James, MD Division of Infectious Diseases
of Pennsylvania Department of Paediatrics University of Alabama
Philadelphia, PA, USA Division of Infectious Diseases Birmingham, AL, USA
University of Alabama
Kimberly A. Horii, MD Birmingham, AL, USA Veronica A. Kinsler, MA, MB,
Associate Professor of Pediatrics BChir, FRCPCH, PhD
Division of Dermatology Gregor B.E. Jemec, MD, DMSc Professor of Paediatric Dermatology and
University of Missouri‐Kansas City School Department of Dermatology Dermatogenetics
of Medicine Roskilde Hospital Paediatric Dermatology Department
Children’s Mercy‐Kansas City Roskilde, Denmark Great Ormond Street Hospital for Children
Kansas City, MO, USA NHS Foundation Trust
Genetics and Genomic Medicine
Elizabeth A. Jones, MA MB,
UCL Great Ormond Street Institute of
Jennifer Huang, MD BChir, FRCP, PhD Child Health
Assistant Professor of Dermatology Consultant Clinical Geneticist London, UK
Dermatology Program Manchester Centre for Genomic Medicine
Boston Children’s Hospital St Mary’s Hospital
Boston, MA, USA Manchester University NHS Foundation Trust
Bruce R. Korf, MD, PhD
Manchester, UK Professor and Chairman of Department
of Genetics
Devika Icecreamwala, MD Division of Evolution and Genomic Sciences
Faculty of Biology Medicine and Health University of Alabama at Birmingham
Pediatric Dermatology Fellow
University of Manchester Birmingham, AL, USA
Department of Dermatology
Henry Ford Health System Manchester, UK
Detroit, MI, USA Andrew C. Krakowski, MD
Teri A. Kahn, MD, MPH Chief
Department of Dermatology
Ying Liu, MD, PhD Associate Professor
Department of Dermatology St Luke’s University Health Network
Associate Chief Physician
University of Maryland Easton, PA, USA
Department of Dermatology
Beijing Children’s Hospital Baltimore, MD, USA
Capital Medical University Ann M. Kulungowski, MD
National Center for Children’s Health Sonia Kamath, MD Assistant Professor of Surgery and Pediatrics
Beijing, China Resident Physician University of Colorado School of Medicine
Department of Dermatology Surgical Director
Keck School of Medicine of University of Vascular Anomalies Center
Southern California Children’s Hospital Colorado
Los Angeles, CA, USA Aurora, CO, USA
List of Contributors  xvii

Marc Lacour, MD Ting Fan Leung, MBChB(CUHK), Pediatrics and Pediatric Dermatology
MD(CUHK), MRCP(UK), FRCPCH, Cincinnati Children’s Hospital
Paediatrician
Cincinnati, OH, USA
Pediatric Dermatology Clinic FAAAAI, FHKCPaed,
Carouge, Switzerland FHKAM(Paediatrics)
David Luk, FHKAM(Paed),
Chairman and Professor
Bisola Laguda Department of Paediatrics FHKCPaed, FRCPCH
Consultant The Chinese University of Hong Kong Consultant Paediatrician
Paediatric Dermatology Hong Kong United Christian Hospital, Hong Kong
Chelsea and Westminster Hospital Hong Kong Children’s Hospital
Honorary Clinical Assistant Professor
London, UK Michael Levin, FRCPCH, PhD The Chinese University of Hong Kong
Professor of Paediatrics and International The University of Hong Kong
Sinéad M. Langan, MD, PhD Child Health President
Associate Professor of Epidemiology Imperial College London Hong Kong Paediatric and Adolescent
Faculty of Epidemiology and Population Health London, UK Dermatology Society
London School of Hygiene and Tropical
Medicine Moise L. Levy, MD Jane Luker, BDS, PhD, FDSRCS
London, UK
Professor Eng @ Edin DDR, RCR
Department of Pediatrics
Consultant Dental Surgeon
Sean Lanigan, MD, FRCP, DCH Texas Children’s Hospital
Bristol Dental Hospital
Regional Medical Director Baylor College of Medicine
University Hospitals Bristol NHS Foundation
sk:n Limited Houston, TX, USA
Trust
Birmingham, UK Department of Pediatrics
Bristol, UK
Dell Medical School/University of Texas and
Dell Children’s Medical Center
Irene Lara‐Corrales, MD Paula Carolina Luna, MD
Austin, TX, USA
Associate Professor of Pediatrics Dermatology Department
Pediatric Dermatology Fellow Hospital Alemán
Section of Dermatology Rebecca Levy, MD, FRCPC
Buenos Aires, Argentina
Division of Paediatric Medicine Clinical Fellow
Hospital for Sick Children Pediatric Dermatology
University of Toronto University of Toronto Minnelly Luu, MD
Toronto, ON, Canada The Hospital for Sick Children Assistant Professor of Clinical Dermatology
Toronto, ON, Canada Department of Dermatology
Keck School of Medicine of University of
Margarita Larralde, PhD, MD
Susan Lewis‐Jones, FRCP, Southern California
Head Los Angeles, CA, USA
Dermatology Department FRCPCH Division of Pediatric Dermatology
Hospital Alemán Honorary Consultant Dermatologist Children’s Hospital Los Angeles
Head Ninewells Hospital & Medical School Los Angeles, CA, USA
Pediatric Dermatology Department Dundee, UK
Hospital Ramos Mejía
Buenos Aires, Argentina Lin Ma, MD, PhD
Carmen Liy Wong Professor, Director
Pediatric Dermatology Fellow Department of Dermatology
Christine T. Lauren, MD Section of Dermatology Beijing Children’s Hospital
Assistant Professor of Dermatology and Division of Paediatric Medicine Capital Medical University
Pediatrics Hospital for Sick Children National Center for Children’s Health
Columbia University Medical Center Toronto, ON, Canada Beijing, China
New York, NY, USA
Wilson Lopez, MBBS, MD, MRCP, Elia F. Maalouf, MBChB, MRCP,
S. Leclerc‐Mercier, MD FRCPCH, DCH, MSc FRCPCH, MD
Reference Center for Rare and Inherited Skin Consultant Neonatologist Consultant in General Paediatrics and
Diseases (MAGEC) Neonatal Unit Neonatal Medicine
Departments of Dermatology and Pathology Barking, Havering and Redbridge University Neonatal Unit
CHU Necker‐Enfants Malades Hospitals NHS Trust Homerton University Hospital NHS
Paris, France UK Foundation Trust
London, UK
Theresa Ngan Ho Leung, Christopher Lovell
MBBS, FRCPCH, FHKCPaed, Consultant Dermatologist Caroline Mahon, MD
FHKAM(Paed) Kinghorn Dermatology Unit Consultant Paediatric Dermatologist
Clinical Associate Professor Royal United Hospital Department of Dermatology
Department of Paediatrics and Adolescent Bath, UK Bristol Royal Infirmary
Medicine University Hospitals Bristol NHS Foundation Trust
The University of Hong Kong Anne W. Lucky, MD Bristol, UK
Hong Kong
Adjunct Professor of Pediatrics and Dermatology
Divisions of General and Community
xviii  List of Contributors

Melanie Makhija, MD, MSc Jemima E. Mellerio, MD, FRDP Dermatology


Reference Center for Rare Skin Diseases
Assistant Professor of Pediatrics Consultant Dermatologist and Honorary
Hôpital Saint André
Department of Pediatrics Professor
Bordeaux, France
Northwestern University Paediatric Dermatology Department
Feinberg School of Medicine, Great Ormond Street Hospital for Children
Chicago, IL, USA NHS Trust Keith Morley, MD
and St John’s Institute of Dermatology Paediatric Dermatology Fellow
Steven M. Manders, MD Guy’s and St Thomas’ NHS Foundation Trust Dermatology Program
London, UK Boston Children’s Hospital
Professor of Medicine and Paediatrics
Boston, MA, USA
Division of Dermatology
University of Medicine and Dentistry of New Bodo C. Melnik, MD
Jersey Adjunct Professor of Dermatology Dédée F. Murrell,
Robert Wood Johnson Medical School Department of Dermatology, Environmental MA(Cambridge), BMBCh (Oxford),
Camden, NJ, USA Medicine and Health Theory FAAD(USA), MD (UNSW), FACD,
University of Osnabrück
FRCP(Edin)
Julianne A. Mann, MD Osnabrück, Germany
Head
Assistant Professor of Dermatology Department of Dermatology
Dartmouth‐Hitchcock Medical Center Vibhu Mendiratta, MD St George Hospital
Lebanon, NH, USA Director and Professor Professor of Dermatology
Department of Dermatology University of New South Wales
Lady Hardinge Medical College and Sydney, NSW, Australia
Ashfaq A. Marghoob, MD
associated hospitals
Department of Dermatology
Memorial Sloan‐Kettering Cancer Center
New Delhi, India Taizo A. Nakano, MD
New York, NY, USA Assistant Professor of Pediatrics
Eirini E. Merika, MBBS, iBSc, University of Colorado School of Medicine

Maria L. Marino, MD MRCP Derm Medical Director


Consultant Paediatric Dermatologist Vascular Anomalies Center
Department of Dermatology Children’s Hospital Colorado
Chelsea and Westminster Hospital
Memorial Sloan‐Kettering Cancer Center Aurora, CO, USA
London, UK
New York, NY, USA

Christian R. Millett, MD Iria Neri


Anna E. Martinez, FRCPCH Professor of Dermatology
Forefront Dermatology
Consultant Paediatric Dermatologist Department of Specialized, Diagnostic and
Vienna, VA, USA
Paediatric Dermatology Department Experimental Medicine
Great Ormond Street Hospital for Children Division of Dermatology
NHS Trust Adnan Mir, MD, PhD University of Bologna
London, UK Assistant Professor of Dermatology Bologna, Italy
University of Texas Southwestern Medical
Peter Mayser, MD Center and Children’s Medical Center Dallas
Tuyet A. Nguyen, MD
Dallas, TX, USA
Professor of Dermatology Kaiser Permanente Dermatology
Clinic of Dermatology, Allergology and Los Angeles, CA, USA
Venereology Amanda T. Moon, MD
Justus Liebig University (UKGM) Departments of Pediatrics and Dermatology
Giessen, Germany Jeroen Novak
Perelman School of Medicine at the University
of Pennsylvania GGZ Momentum
Breda, The Netherlands
Juliette Mazereeuw‐Hautier, Philadelphia, PA, USA
MD, PhD Section of Dermatology
Children’s Hospital of Philadelphia Susan O’Connell, MD
Professor of Dermatology
Philadelphia, PA, USA Formerly Lyme Borreliosis Unit
Reference Center for Rare Skin Diseases
Department of Dermatology Health Protection Agency Microbiology
CHU Larrey Elena Moraitis, MBBS, PhD Laboratory
Toulouse, France Consultant in Paediatric Rheumatology Southampton University Hospitals NHS Trust
Infection, Inflammation and Rheumatology Southampton, UK

William H. McCoy IV, MD, PhD Section


UCL Institute of Child Health Cathal O’Connor, MD
Resident Physician
London, UK Paediatric Dermatology
Division of Dermatology
Paediatric Rheumatology Department Trinity College Dublin and Our Lady’s
Department of Medicine
Great Ormond Street Hospital for Children Children’s Hospital
Washington University School of Medicine
NHS Foundation Trust Dublin, Ireland
St Louis, MO, USA
London, UK

Vinzenz Oji, MD
Fanny Morice‐Picard, MD, PhD Department of Dermatology
Department of Dermatology and Paediatric University Hospital Münster
Münster, Germany
List of Contributors  xix

Elise A. Olsen, MD Aparna Palit, MD Laura Polivka, MD, PhD


Professor of Dermatology and Medicine Professor Department of Dermatology
Director, Cutaneous Lymphoma Research and Department of Dermatology Imagine Institute
Treatment Center Venereology and Leprosy Necker‐Enfants Malades Hospital
Director, Hair Disorders Research and Sri B.M.Patil Medical College Paris, France
Treatment Center Hospital & Research Centre
Director, Dermatopharmacology Study Center BLDE University Elena Pope, MSc, FRCPC
Departments of Dermatology and Medicine Vijayapur, Karnataka, India
Professor of Paediatrics
Duke University Medical Center
University of Toronto
Durham, NC, USA Amy S. Paller, MD, MSc Fellowship Director and Section Head
Walter J. Hamlin Professor and Chair of Paediatric Dermatology
Kai Ren Ong, MD, MRCP Dermatology, Professor of Pediatrics The Hospital for Sick Children
Consultant Clinical Geneticist Departments of Pediatrics and Dermatology Toronto, ON, Canada
West Midlands Regional Clinical Genetics Service Northwestern University
Birmingham Women’s Hospital Feinberg School of Medicine Julie Powell, MD, FRCPC
Birmingham, UK Chicago, IL, USA
Director
Pediatric Dermatology
Arnold P. Oranje, MD, PhD Nirav Patel, MD Professor of Dermatology (Pediatrics)
(Deceased) Departments of Dermatology and Pediatrics Division of Dermatology
Professor of Pediatric Dermatology Mayo Clinic Department of Pediatrics
Kinderhuid.nl, Rotterdam, The Netherlands Rochester, MN, USA CHU Sainte‐Justine
Hair Clinic, Breda, The Netherlands University of Montreal
Dermicis Skin Clinic, Alkmaar, The Netherlands Annalisa Patrizi, MD Montreal, QC, Canada

Professor
Luz Orozco‐Covarrubias, MD Head of Dermatology Julie S. Prendiville, MBBCH,
Paediatric Dermatologist and Associated Department of Specialized, Diagnostic and DCH, BAO, FRCPC
Professor of Pediatric Dermatology Experimental Medicine, Chief
Universidad Nacional Autonoma de México Division of Dermatology Pediatric Dermatology
Attending Physician University of Bologna Sidra Medicine
Department of Pediatric Dermatology Bologna, Italy Doha, Qatar
National Institute of Paediatrics of Mexico
Mexico City, Mexico Marissa J. Perman, MD Cecilia A.C. (Sanna) Prinsen,
Assistant Professor of Pediatrics and PhD
Edel A. O’Toole, MB, PhD, FRCP, Dermatology VU University Medical Center
FRCPI Children’s Hospital of Philadelphia and Department of Epidemiology and Biostatistics
Professor of Molecular Dermatology and The University of Pennsylvania Amsterdam Public Health Research Institute
Honorary Consultant Dermatologist Philadelphia, PA, USA Amsterdam, The Netherlands
Department of Dermatology
Royal London Hospital Karen Pett Lori Prok, MD
Barts Health NHS Trust and Centre for Cell
Clinical Nurse Specialist in Paediatric Associate Professor of Dermatology and
Biology and Cutaneous Research
Dermatology Pathology
Barts and the London School of Medicine
West Hertfordshire Hospitals NHS Trust University of Colorado Denver and Children’s
and Dentistry
St Albans, UK Hospital Colorado
London, UK
Denver, CO, USA
Roderic J. Phillips, BSc(Hons),
Hagen Ott, MD
MBBS, PhD, FRACP, AMAM, CIRF Neil S. Prose, MD
Head of the Division of Pediatric Dermatology
Associate Professor Professor
and Allergology
Paediatric Dermatologist Department of Pediatrics and Dermatology
Epidermolysis Bullosa Centre Hannover
Royal Children’s Hospital Duke University Medical Center
Children’s Hospital AUF DER BULT
Honorary Research Fellow Research Professor of Global Health, Duke
Hannover, Germany
Murdoch Children’s Research Institute Global Health Institute
Adjunct Professor Co‐Director, Duke Health Humanities Lab
Seza Özen, MD Paediatrics Durham, NC, USA
Professor of Pediatrics Monash University
Hacettepe University School of Medicine Melbourne, VIC, Australia
Department of Pediatric Rheumatology
Diana Purvis, MB ChB, MRCPCH,
Ankara, Turkey FRACP
Bianca Maria Piraccini, MD, PhD Paediatric Dermatologist
Dermatology Starship Children’s Hospital
David G. Paige, MBBS, MA, Department of Experimental, Diagnostic and Honorary Senior Lecturer
FRCP Specialty Medicine Department of Paediatrics
Consultant Dermatologist University of Bologna University of Auckland
Department of Dermatology, Bologna, Italy Auckland, New Zealand
Bart’s and The London NHS Trust
London, UK
xx  List of Contributors

Marius Rademaker, BM, Marcelo Ruvertoni, MD Julien Seneschal, MD, PhD


FRCP(Edin), FRACP, DM Paediatric Dermatologist and Paediatrician Professor
Clinical Director British Hospital Department of Dermatology and Paediatric
Dermatology Department Montevideo, Uruguay Dermatology
Waikato District Health Board Reference Center for Rare Skin Diseases
Hon Associate Professor Liat Samuelov, MD Hôpital Saint André
Waikato Clinical Campus Bordeaux, France
Vice Chair
Faculty of Medical and Health Sciences
Department of Dermatology
The University of Auckland
Tel Aviv Sourasky Medical Center G. Sethuraman, MD
Hamilton, New Zealand
Tel Aviv, Israel Professor of Dermatology
Department of Dermatology
Marc Alexander Radtke, MD Sarita Sanke, MD All India Institute of Medical Sciences
Professor New Delhi, India
Dermatology and STD
Institute for Health Services Research in
Lady Hardinge Medical College and
Dermatology and Nursing (IVDP)
associated hospitals Marieke M.B. Seyger, MD, PhD
University Medical Center Hamburg‐
New Delhi, India Associate Professor of Dermatology
Eppendorf (UKE)
Department of Dermatology
Hamburg, Germany
Radboud University Medical Center
Julie V. Schaffer, MD Nijmegen, The Netherlands
V. Ramesh, MD Associate Professor of Pediatrics
Division of Pediatric and Adolescent
Professor of Dermatology
Dermatology Lindsay Shaw, MBBS, MRCPCH
Department of Dermatology
Hackensack University Medical Center Consultant in Paediatric Rheumatology
Vardhman Mahavir Medical College &
Hackensack, NJ, USA Paediatric Dermatology
Safdarjung Hospital
Great Ormond Street Hospital for Children
New Delhi, India
NHS Foundation Trust
Birgitta Schmidt, MD London and Bristol Children’s Hospital
Gudrun Ratzinger, MD Department of Pathology Bristol UK
Boston Children’s Hospital
Professor of Dermatology
Harvard Medical School
Department of Dermatology, Venereology and
Boston, MA, USA Neil Shear, MD, FRCPC, FACP
Allergology
Professor of Medicine and Pharmacology
Medical University Innsbruck
Division of Dermatology
Austria Enno Schmidt, MD, PhD Sunnybrook Health Sciences Center and
Professor of Dermatology University of Toronto
Wingfield E. Rehmus, MD Lübeck Institute of Experimental Toronto, ON, Canada
Dermatology (LIED)
Clinical Assistant Professor
Lübeck, Germany
Department of Pediatrics Tor A. Shwayder, MD
University of British Columbia and British
Director
Columbia’s Children’s Hospital Steffen Schubert Pediatric Dermatology
Vancouver, BC, Canada Department of Dermatology, Venereology Department of Dermatology
and Allergology Henry Ford Hospital
Sean D. Reynolds, MB BCh, BAO University Medical Center Göttingen Detroit, MI, USA
Göttingen, Germany
Department of Dermatology
Warren Alpert Medical School of Brown Brenda M. Simpson
University Crispian Scully, CBE, DSc, DChD, Dermatologist
Providence, RI, USA DMed (HC), Dhc (multi), MD, PhD, El Paso Dermatology Center
PhD (HC), FMedSci, MDS, MRCS, El Paso, TX, USA
Nerys Roberts, MD, FRCP, BSc, FDSRCS, FDSRCPS, FFDRCSI,
MRCPCH, BSc FDSRCSEd, FRCPath, FHEA Robert Sidbury, MD, MPH
Consultant Paediatric Dermatologist (Deceased) Professor
Chelsea & Westminster Hospital Emeritus Professor of Oral Medicine at UCL Department of Pediatrics
London, UK Bristol Dental Hospital Chief
University Hospitals Bristol NHS Division of Dermatology
Jean Robinson Foundation Trust Seattle Children’s Hospital
Bristol, UK University of Washington School of Medicine
Clinical Nurse Specialist in Paediatric
University College London Seattle, WA, USA
Dermatology
London, UK
Royal London Hospital
London, UK Jonathan I. Silverberg, MD,
Robert K. Semple, PhD, FRCP PhD
Elke Rodriguez, PhD Professor of Translational Molecular Medicine
Associate Professor of Dermatology
Centre for Cardiovascular Sciences, Queens
Senior Researcher Northwestern University Feinberg School
Medical Research Institute
Department of Dermatology, Allergology and of Medicine
University of Edinburgh
Venereology Chicago, IL, USA
Edinburgh, UK
University Hospital Schleswig‐Holstein
Campus Kiel
Kiel, Germany
List of Contributors  xxi

Nanette Silverberg, MD Peter M. Steijlen, MD, PhD Amy Theos, MD


Clinical Professor of Dermatology Professor of Dermatology and Chair Associate Professor of Department of
Icahn School of Medicine at Mount Sinai Department of Dermatology Dermatology
Chief Maastricht University Medical Center University of Alabama at Birmingham
Pediatric Dermatology Maastricht, The Netherlands Birmingham, AL, USA
Mount Sinai Health System
Director Jane C. Sterling, MB, BChir, MA, Peter Theut Riis, MD
Pediatric and Adolescent Dermatology
Department of Dermatology
FRCP, PhD Department of Dermatology
Consultant Dermatologist Roskilde Hospital
New York, NY, USA
Department of Dermatology Roskilde, Denmark
Cambridge University Hospitals NHS
Eric L. Simpson, MD Foundation Trust Anna C. Thomas, BSc, PhD
Professor of Dermatology Addenbrooke’s Hospital
Post‐doctoral Research Associate
School of Medicine Cambridge, UK
Genetics and Genomic Medicine
Department of Dermatology
UCL Great Ormond Street Institute of Child
Oregon Health and Science University
Portland, OR, USA
Jenna L. Streicher, MD Health
Clinical Assistant Professor London, UK
Department of Pediatrics, Dermatology Section
Manuraj Singh, MBBS, MRCP, Children’s Hospital of Philadelphia Megha Tollefson, MD
PhD, DipRCPath (Dermpath) Departments of Pediatrics and Dermatology
Departments of Dermatology and Pediatrics
Consultant Dermatologist and Perelman School of Medicine at the University
Mayo Clinic
Dermatopathologist of Pennsylvania
Rochester, MN, USA
St George’s University Hospitals Philadelphia, PA, USA
London, UK
Wynnis L. Tom, MD
V. Reid Sutton, MD
Associate Clinical Professor of Dermatology
Nedaa Skeik, MD, FACP, FSVM, RPVI Professor
and Pediatrics
Associate Professor of Medicine Department of Molecular and Human
University of California, San Diego
Section Head, Vascular Medicine Department Genetics
Rady Children’s Hospital
Medical Director, Thrombophilia & Baylor College of Medicine and Texas
San Diego, CA, USA
Anticoagulation Clinic Children’s Hospital
Medical Director, Hyperbaric Medicine Houston, TX, USA
Medical Director, Vascular Laboratories Yun Tong, MD
Minneapolis Heart Institute at Abbott Clinical Research Fellow
Samira Batul Syed, MBBS, DCH,
Northwestern Hospital – part of Allina Health Department of Dermatology, University of
Minneapolis, MN, USA
DCCH, RCPEd, RCGP, FCM, BTEC California San Diego
Adv LASER, DPD San Diego, CA, USA
Associate Specialist in Paediatrics
Lea Solman, MD, FRCPCH Dermatology
Consultant Paediatric Dermatologist Helga V. Toriello, PhD
Great Ormond Street Hospital for Children
Department of Paediatric Dermatology NHS Trust Professor
Great Ormond Street Hospital for Children London, UK Department of Pediatrics/Human
NHS Trust Development
London, UK Michigan State University College of Human
Zsuzsanna Z. Szalai, MD Medicine
Professor and Head Grand Rapids, MI, USA
Eli Sprecher, MD, PhD Department of Pediatric Dermatology
Professor and Chair Heim Pál Children’s Hospital
Department of Dermatology and Deputy Antonio Torrelo, MD
Budapest, Hungary
Director General for Patient Safety Head
Tel Aviv Sourasky Medical Center Department of Dermatology
Frederick Reiss Chair of Dermatology Alain Taïeb, MD, PhD Hospital Infantil del Niño Jesús
Sackler Faculty of Medicine Professor of Dermatology Madrid, Spain
Tel Aviv University Department of Dermatology and Paediatric
Tel Aviv, Israel Dermatology, Reference Center for Rare Skin
Antonella Tosti, MD
Diseases
Hôpital Saint André Department of Dermatology and Cutaneous
Sahana M. Srinivas, DNB, DVD, Bordeaux, France Surgery
FRGUHS (Paediatric Dermatology) Miller Medical School University of Miami
Consultant Paediatric Dermatologist Miami, FL, USA
Department of Pediatric Dermatology
Carolina Talhari, MD, PhD
Adjunct Professor of Dermatology
Indira Gandhi Institute of Child Health James R. Treat, MD
Bangalore, Karnataka, India Amazon State University
Manaus, AM, Brazil Associate Professor of Clinical Pediatrics and
Dermatology
Paola Stefano, MD Fellowship Director, Pediatric Dermatology
Martin Theiler, MD Education Director, Pediatric Dermatology
Assistant Physician
Dermatology Department Paediatric Dermatology Department Children’s Hospital of Philadelphia
Hospital de Pediatría ‘Prof. Dr. Juan P. Garrahan’ University Children’s Hospital Zurich Dermatology Section
Buenos Aires, Argentina Switzerland Perelman School of Medicine at the University
of Pennsylvania
Philadelphia, PA, USA
xxii  List of Contributors

Stephen K. Tyring, MD, PhD Peter von den Driesch, MD Miriam Weinstein, BSc, BScN,
Clinical Professor Professor of Dermatology MD, FRCPC (Paediatrics) FRCPC
Department of Dermatology Head (Dermatology)
University of Texas Health Science Center Center for Dermatology Department of Paediatrics
at Houston Klinikum Stuttgart Hospital for Sick Children
Houston, TX, USA Stuttgart, Germany Toronto, ON, Canada

Nina van Beek, MD Amy Walker, MRes, BSc Pamela F. Weiss, MD, MSCE
Department of Dermatology Experimental and Personalised Medicine Associate Professor of Pediatrics and
University of Lübeck UCL Great Ormond Street Institute of Epidemiology
Lübeck, Germany Child Health Divison of Rheumatology
London, UK Children’s Hospital of Philadelphia
Ignatia B. Van den Veyver, MD Center for Clinical Epidemiology and
Professor Lachlan Warren Biostatistics
Departments of Obstetrics and Gynecology Consultant Dermatologist University of Pennsylvania
and Molecular and Human Genetics Department of Dermatology Philadelphia, PA, USA
Director of Clinical Prenatal Genetics Women’s & Children’s Hospital
BCM and Texas Children’s Hospital Pavilion Adelaide, SA, Australia Alexis Weymann Perlmutter,
for Women MD
Investigator Joy Wan, MD, MSCE Resident
Jan and Dan Duncan Neurological Research
Postdoctoral Fellow of Dermatology Department of Dermatology
Institute at Texas Children’s Hospital
Department of Biostatistics and Epidemiology Geisinger Medical Center
Baylor College of Medicine
University of Pennsylvania Perelman School PA, USA
Houston, TX, USA
of Medicine
Philadelphia, PA, USA Lizbeth Ruth Wheeler
Maurice A.M. van Steensel,
Department of Dermatology
MD, PhD Siriwan Wananukul, MD St George Hospital and University of New
Professor of Dermatology and Skin Biology South Wales
Professor of Paediatrics
Lee Kong Chian School of Medicine Sydney, NSW, Australia
Head of Department of Paediatrics
Singapore
Faculty of Medicine
Research Director
Skin Research Institute of Singapore
Chulalongkorn University Hywel C. Williams, MD, PhD
Bangkok, Thailand
Singapore Director of the NIHR Health Technology
Assessment Programme
Andrew Wang, MD Co‐Director of the Centre of Evidence Based
Felipe Velasquez, MD Dermatology
Brookline Dermatology Associates
Consultant in Pediatric Dermatology University of Nottingham
West Roxbury
Department of Pediatric Dermatology Nottingham, UK
MA, USA
Instituto de Salud del Niño
Lima, Peru
Lisa L. Wang, MD Lara Wine Lee, MD, PhD
Associate Professor Departments of Dermatology and Pediatrics
Paul Veys Medical University of South Carolina
Department of Pediatrics
Director Charleston, SC, USA
Texas Children’s Hospital
Bone Marrow Transplantation Unit
Baylor College of Medicine
Great Ormond Street Hospital for Children
NHS Trust
Houston, TX, USA Marion Wobser, MD
London, UK Department of Dermatology, Venereology and
Bettina Wedi, MD, PhD Allergology
Professor of Dermatology University Hospital Würzburg
Miikka Vikkula, MD, PhD Würzburg, Germany
Department of Dermatology and Allergology
Head of Laboratory of Human Molecular
Hannover Medical School
Genetics
de Duve Institute
Hannover, Germany Johannes Wohlrab, MD
University of Louvain Professor of Dermatology
Brussels, Belgium Lisa Weibel, MD Department of Dermatology and Venereology
Paediatric Dermatology Department and Institute of Applied Dermatopharmacy
University Children’s Hospital Zurich Martin‐Luther‐University Halle‐Wittenberg
Beatrix Volc‐Platzer, MD Halle (Saale), Germany
Switzerland
Professor of Dermatology
Department of Dermatology
Donauspital SMZ Ost Stephan Weidinger, MD A. Wolkerstorfer, MD, PhD
Vienna, Austria Professor of Dermatology Netherlands Institute for Pigment Disorders
Deputy Head Amsterdam University Medical Centers
Department of Dermatology, Allergology and Amsterdam, The Netherlands
Venereology
University Hospital Schleswig‐Holstein
Campus Kiel
Kiel, Germany
List of Contributors  xxiii

Heulwen Wyatt Albert C. Yan, MD, FAAP, FAAD Bernhard W.H. Zelger, MD,
Clinical Nurse Specialist in Paediatric Section of Dermatology MSc
Dermatology Children’s Hospital of Philadelphia Professor of Dermatology
Dermatology Unit Philadelphia, PA, USA Department of Dermatology, Venereology and
St Woolos Hospital, Departments of Pediatrics and Dermatology Allergology
Newport, UK Perelman School of Medicine at the University Medical University Innsbruck
of Pennsylvania Austria
Yuanyuan Xiao, MD Philadelphia, PA, USA

Associate Chief Physician


Department of Dermatology Kevin B. Yarbrough, MD
Beijing Children’s Hospital Staff Physician
Capital Medical University Department of Pediatric Dermatology
National Center for Children’s Health Phoenix Children’s Hospital
Beijing, China Phoenix, AZ, USA

Zhe Xu, MD, PhD Vijay Zawar, MD


Chief Physician Skin Diseases Centre
Department of Dermatology Nashik, India
Beijing Children’s Hospital Professor
Capital Medical University Department of Dermatology
National Center for Children’s Health MVP’s Dr Vasantrao Pawar Medical College
Beijing, China and Research Centre
Nashik, Maharashtra, India
xxiv 

Preface to the Fourth Edition

It is with much delight and pride that we present the The fourth edition appears at a time when digital instant
fourth edition of this textbook. The new edition continues access to information, retrievable anywhere and at anytime,
to provide state‐of‐the‐art information on all aspects of is the norm. Nevertheless, the value of having a physical
skin disease in children. Existing content has been textbook to read and study remains an integral part of clini-
refreshed and fully updated to reflect emerging thinking cal practice and academia; but, in order to keep up to date,
and to incorporate the latest in research and clinical the book will also be accessible online on an interactive plat-
data  –  especially at the genetic level. The three Editors, form via laptops and smartphones. The book’s virtue is, in
two Editorial Advisors and five Associate Editors from our opinion, its comprehensiveness and in‐depth informa-
across the world bring a truly global perspective to the tion written by international experts on each subject.
work. Some 31 countries are represented by 313 We hope that this fourth edition will be as warmly
contributors, of whom 192 are new contributors, who received as its three predecessors and will contribute to
have extensively updated or completely rewritten the 177 the improved care of children with skin diseases.
chapters. The book represents a definitive reference text PH
for dermatologists, paediatricians, clinician scientists, VK
research workers and all other individuals involved in the AY
care of children with skin disease. JIH
 xxv

Dedication

In memory of Arnold P. Oranje, 1948–2016

This 4th edition of the textbook is dedicated to my dear friend and colleague Arnold Oranje. Arnold’s untimely death
shocked us all and is a great loss to the world of paediatric dermatology. He was one of the three original editors of this
book, with Neil Prose and myself, and was working on this edition at the time of his death.
On a personal note, my collaboration with Arnold was unique: his enthusiasm, passion for the subject, expertise and exuber-
ant laughter made him a joy to work with. I hope that Arnold would be immensely proud of this new edition.
John Harper

Taïeb A, Stalder J‐F, de Waard‐van der Spek F, Harper J. In Memoriam: Arnold P. Oranje. Pediatr Dermatol 2017; 34: 231–4.
xxvi 

Acknowledgements

We wish to thank the following people: the Associate However, the biggest thank you has to be to our patients
Editors and chapter authors for their invaluable contri- who provide the inspiration and motivation for our work
bution to the book; the editorial staff and production on a daily basis.
staff at Wiley‐Blackwell, freelance project editor, Alison PH
Nick, project manager, Nik Prowse, and the copy-editors VK
for their tireless efforts; the families who gave permis- AY
sion for the photographs of their children to be used in JIH
this book and our own families for their understanding
and support.
 xxvii

List of Abbreviations

AA alopecia areata AMMoL acute myelomonocytic leukaemia


AA amyloid A AMN acquired melanocytic naevi
AAD American Academy of Dermatology AMoL acute monocytic leukaemia
AAV adeno‐associated virus AMP antimicrobial peptide
AAV antineutrophil cytoplasmic antibody (ANCA)‐ AMPK adenosine monophosphate‐activated protein kinase
associated vasculitides AN acanthosis nigricans
ABC ATP‐binding cassette ANA antinuclear antibody
ACA acrodermatitis chronica atrophicans ANCA antineutrophilic cytoplasmic antibody
ACA anticentromere antibodies AP actinic prurigo
ACC aplasia cutis congenita AP adaptor protein complex
ACD allergic contact dermatitis AP anteroposterior
ACD amyloidosis cutis dyschromica APC antigen‐presenting cell
ACE angiotensin‐converting enzyme APECED autoimmune polyendocrinopathy–candidiasis–
aCGH array‐comparative genomic hybridization ectodermal dystrophy (syndrome)
ACR American College of Rheumatology APL acquired partial lipoatrophy
ACS Aicardi–Goutiéres syndrome APL atrichia with papular lesions
ACTH adrenocorticotropic hormone APP atrophoderma of Pasini and Pierini
AD atopic dermatitis APS antiphospholipid antibody syndrome
AD autosomal dominant APSS acral peeling skin syndrome
ADA adenosine deaminase AR androgen receptor
ADCL autosomal dominant cutis laxa AR autosomal recessive
ADCL anergic diffuse cutaneous leishmaniasis ARC arthrogryposis–renal dysfunction–cholestasis
ADHD attention deficit hyperactivity disorder (syndrome)
ADR adverse drug reaction ARCI autosomal recessive congenital ichthyosis
ADM atypical dermal melanocytosis ARCL autosomal recessive cutis laxa
ADULT acro‐dermato‐ungual‐lacrimal‐tooth ARD adult Refsum disease
ADWH autosomal‐dominant woolly hair AR‐HIES autosomal recessive hyperimmunoglobulin E
AEC ankyloblepharon, ectodermal defects and cleft lip/ syndrome
palate ARKID autosomal recessive keratoderma ichthyosis and
AEDS atopic eczema/dermatitis syndrome deafness (syndrome)
AEI annular epidermolytic ichthyosis ARMS amplification‐refractory mutation system
AFB acid‐fast bacilli ASD autism spectrum disorders
AGA androgenetic alopecia ASM aggressive systemic mastocytosis
AGEP acute generalized exanthematous pustulosis ASO antistreptolysin O
aGVHD acute GVHD ASPS alveolar soft part sarcoma
AGL acquired generalized lipoatrophy ASST autologous serum skin test
AGM acrylate gelling material AST aspartate transaminase
AHA American Heart Association AST aspartate aminotransferase
AHA antihistone antibody AT ataxia telangiectasia
AHEI acute haemorrhagic oedema of infancy ATG anti‐thymocyte globulin
AHO Albright hereditary osteodystrophy ATGL adipose triglyceride lipase
AHT abusive head trauma ATP adenosine triphosphate
AID autoinflammatory disease ATPase adenosine triphosphatase
AIDS acquired immune deficiency syndrome ATS arterial tortuosity syndrome
AIP acute intermittent porphyria ATT antitubercular therapy
ALHE angiolymphoid hyperplasia with eosinophilia AUC area under the concentration‐time curve
ALL acute lymphoblastic leukaemia AUG acute ulcerative gingivitis
ALN actinic lichen nitidus AV arteriovenous
ALP actinic lichen planus AVF arteriovenous fistula
ALT alanine transaminase AVM arteriovenous malformations
ALU aphthous‐like ulceration AV atrophoderma vermiculata
AML acute myeloid leukaemia AZA azathioprine
xxviii  List of Abbreviations

BAL bronchoalveolar lavage CFU colony‐forming unit


BB β‐blockers CGD chronic granulomatous disease
BCC basal cell carcinoma CGH comparative genomic hybridization
BCG bacillus Calmette–Guérin CGL congenital generalized lipodystrophy
BCIE bullous congenital ichthyosiform erythroderma CGRP calcitonin gene‐related product
BD Behçet disease CGS contiguous gene syndrome
BDCS Bazex–Dupré–Christol syndrome cGVHD chronic graft versus host disease
BDD blistering distal dactylitis CH congenital haemangiomas
BDNG British Dermatological Nursing Group CHAND curly hair, ankyloblepheron, nail dysplasia
BER base excision repair (syndrome)
BGS Baller–Gerold syndrome CHCC Chapel Hill Consensus Conference
BHDS Birt–Hogg–Dubé syndrome CHH Conradi–Hünermann–Happle syndrome
BHPR British Health Professionals in Rheumatology CHH cartilage hair hypoplasia
BMD bone mineral density CHIKV Chikungunya virus
BMI body mass index CHILD congenital hemidysplasia with ichthyosiform
BMP bone morphogenetic protein erythroderma and limb defect (syndrome)
BMT bone marrow transplantation CHIME colobomas, congenital heart disease, early‐onset
BO branchio‐otic ichthyosiform dermatosis, mental retardation and ear
BOR branchio‐oto‐renal abnormalities
BOS Buschke–Ollendorff syndrome CHP cytophagic histiocytic panniculitis
BP blood pressure CHRPE congenital hypertrophy of the retinal pigment
BP bullous pemphigoid epithelium
BRBN blue rubber bleb naevus CHS Chédiak–Higashi syndrome
BRRS Bannayan–Riley–Ruvalcaba syndrome CI congenital ichthyoses
BS Bloom syndrome CIAD Childhood Impact of Atopic Dermatitis
BSA body surface area CID combined immunodeficiency
BSCL Berardinelli–Seip congenital lipodystrophy CIE congenital ichthyosiform erythroderma
BSI bathing suit ichthyosis CIL‐F congenital infiltrating lipomatosis of the face
BSLE benign summer light eruption CL cutaneous leishmaniasis
BSR British Society for Rheumatology CLM cutaneous larva migrans
BSS Brooke–Spiegler syndrome CLOVE congenital lipomatous overgrowth, vascular
BTK Bruton tyrosine kinase malformations and epidermal naevi (syndrome)
BV bacterial vaginitis CLQI Children’s Life Quality Index
BWS Beckwith–Wiedemann syndrome CLSM confocal laser scanning microscopy
CA condyloma acuminata CM capillary malformation
CAD chronic actinic dermatitis CM cutaneous mastocytosis
CADIS Childhood Atopic Dermatitis Impact Scale CM‐AVM capillary malformation‐arteriovenous malformation
C‐ALCL cutaneous anaplastic large cell lymphoma CMC chronic mucocutaneous candidiasis
CALM café‐au‐lait macules CML chronic myeloid leukaemia
CAM cell adhesion molecule CMN congenital melanocytic naevi
CA‐MRSA community‐associated meticillin‐resistant CMN congenital mesoblastic nephroma
Streptococcus aureus CMO carotenoid 15,15′‐mono‐oxygenase
CAN child abuse and neglect CMRDS constitutional mismatch repair deficiency syndrome
c‐ANCA cytoplasmic antineutrophil cytoplasmic antibody CMTC cutis marmorata telangiectatica congenita
CANDLE chronic atypical neutrophilic dermatosis with CMV cytomegalovirus
lipodystrophy and elevated temperature CNS central nervous system
CAPS cryopyrin‐associated autoinflammatory syndromes CNTP connective tissue naevus of the proteoglycan type
CAT cutaneovisceral angiomatosis with thrombocyopenia CNV copy number variations
CAVB complete atrioventricular block CoA co‐enzyme A
CAVM capillary–arteriolovenular malformations C1‐INH C1‐esterase inhibitor
CBCL cutaneous B‐cell lymphoma CoNS coagulase‐negative Staphylococcus
CBS cystathionine β‐synthase COFS cerebro‐oculo‐facio‐skeletal
CCA congenital contractural arachnodactyly CPAP continuous positive airway pressure
CCC congenital cutaneous candidiasis CRP C‐reactive protein
CCTN cerebriform connective tissue naevi CRIE congenital reticular ichthyosiform erythroderma
CD coeliac disease CRISPR clustered regularly interspaced palindromic repeats
CDLQI Children’s Dermatology Life Quality Index CPD cyclobutanepyrimidine dimers
CDP chondrodysplasia punctata CS Cockayne syndrome
CDPX2 X‐linked dominant chondrodysplasia punctata CS Costello syndrome
CDS Chanarin–Dorfman syndrome CSA child sexual abuse
CE cell envelope CSD cat scratch disease
CEA carcinoembryonic antigen CSF cerebrospinal fluid
CEDNIK cerebral dysgenesis, neuropathy, ichthyosis, CSMH congenital smooth muscle hamartoma
keratoderma (syndrome) CST Christ–Siemens–Touraine
CEP congenital erythropoietic porphyria CT Chlamydia trachomatis
CEVD congenital erosive and vesicular dermatosis (with CT computed tomography
reticulated supple scarring) CTCL cutaneous T‐cell lymphoma
CFCS cardiofaciocutaneous syndrome CTGF connective tissue growth factor
CFTR cystic fibrosis transmembrane conductance regulator CTN connective tissue naevi
List of Abbreviations  xxix

CTL cytotoxic T‐lymphocyte ECM1 extracellular matrix protein 1


CTLA‐4 cytotoxic T‐lymphocyte antigen‐4 ECP eosinophil cationic protein
CVG cutis verticis gyrata ECP extracorporeal photopheresis
CVID common variable immunodeficiency ED ectodermal dysplasia
CVS chorionic villus sampling EDA ectodysplasin‐A
CVS congenital varicella syndrome EDP erythema dyschromicum perstans
CVST cerebral venous sinus thrombosis EDS Ehlers–Danlos syndrome
Cx connexin EDTA ethylenediaminetetraacetic acid
DADA2 deficiency of adenosine deaminase type 2 EDV epidermodysplasia verruciformis
DASI Dyshidrotic Eczema Area and Severity Index EEC ectrodactyly, ectodermal dysplasia and cleft lip/
DBPCDC double‐blind placebo‐controlled drug challenge palate
DBS DeBarsy syndrome EED erythema elevatum diutinum
DC dyskeratosis congenita EEG electroencephalogram
DCM diffuse cutaneous mastocytosis EF eosinophilic fasciitis
dcSSc diffuse cutaneous systemic sclerosis EGA estimated gestational age
DD Darier disease EGF epidermal growth factor
DDD Dowling–Degos disease EGFR epidermal growth factor receptor
DDEB dominant dystrophic epidermolysis bullosa EGPA eosinophilic granulomatosis with polyangiitis
DE dermoepidermal EGW external genital wart
DEB dystrophic epidermolysis bullosa EH eczema herpeticum
DEJ dermoepidermal junction EH epithelioid haemangioma
DEPPK diffuse epidermolytic palmoplantar keratoderma EHK epidermolytic hyperkeratosis
DFA direct fluorescent antibody EI epidermolytic ichthyosis
DFI Dermatitis Family Impact EIA enzyme immunoassay
DFSP dermatofibrosarcoma protuberans EIB erythema induratum of Bazin
DG1 DGI disseminated gonoccocal infection EKA erythrokeratoderma with ataxia
DGS DiGeorge syndrome EKC erythrokeratoderma en cocardes
DH dermatitis herpetiformis EKV erythrokeratoderma variabilis
DHEA dihydroepiandrosterone EKVP erythrokeratoderma variabilis progressiva
DHEAS dihydroepiandrosterone sulphate ELISA enzyme‐linked immunosorbent assay
DI dentinogenensis imperfecta EM erythema migrans
DIF direct immunofluorescence EM erythema multiforme
DIHS/AHS drug‐induced or anticonvulsant hypersensitivity EMG electromyogram
syndrome EMLA eutectic mixture of local anaesthetics
DIRA deficiency of the IL‐1 receptor antagonist EMS electromagnetic spectrum
DKS dyskeratosis congenita EN epidermal naevus
DLE discoid lupus erythematosus EN erythema nodosum
DMARD disease modifying antirheumatic drugs EN‐D epidermal naevus – Darier type
DMEG dysplastic megalencephaly ENDA European Network for Drug Allergy
DMSA dimercaptosuccinic acid ENS epidermal naevus syndrome
DMSO dimethyl sulphoxide ENT ear, nose and throat
DNEPPK diffuse nonepidermolytic palmoplantar keratoderma EORTC European Research and Treatment of Cancer
DOC disorders of cornification EOS early‐onset sarcoidosis
DOPA dihydroxyphenylalanine EP eccrine poroma
DP dyschromatosis ptychotropica EPDS erosive pustular dermatosis of the scalp
DPR dermatopathia pigmentosa reticularis EPI eosinophilic pustulosis of infancy
DRESS drug rash with eosinophilia and systemic symptoms EPF eosinophilic pustular folliculitis
DSAP disseminated superficial actinic porokeratosis EPP erythropoietic protoporphyria
DSH dyschromatosis symmetrica hereditaria ER endoplasmic reticulum
DSP disseminated superficial porokeratosis ERA enthesitis‐related arthritis
DTE desmoplastic trichoepithelioma ES epithelioid sarcoma
DUH dyschromatosis universalis hereditaria ESPD European Society of Pediatric Dermatology
DVA developmental venous anomaly ESR erythrocyte sedimentation rate
DVT deep vein thrombosis ESRD end‐stage renal disease
EAACI European Academy of Allergy and Clinical ET exfoliative toxin
Immunology ETN erythema toxicum neonatorum
EASI Eczema Area and Severity Index EULAR European League Against Rheumatism
EB epidermolysis bullosa EV eczema vaccinatum
EBA epidermolysis bullosa acquisita EV enteroviruses
EBP emopamil binding protein EV epidermodysplasia verruciformis
EBV Epstein–Barr virus EV‐HPV epidermodysplasia verruciformis‐associated human
EBS epidermolysis bullosa simplex papillomavirus
EBS‐DM epidermolysis bullosa simplex Dowling–Meara FADH fatty alcohol dehydrogenase
EBS‐MP epidermolysis bullosa simplex with mottled FAE fumaric acid ester
pigmentation FALDH fatty aldehyde dehydrogenase
ECCL encephalocraniocutaneous lipomatosis FAP familial adenomatous polyposis
ECF eosinophil chemotactic factor FAO fibroadipose hyperplasia or overgrowth
ECG electrocardiogram FATP fatty acid transport protein
ECHO enteric cytopathic human orphan FC familial cylindromatosis
xxx  List of Abbreviations

FDA US Food and Drug Administration HCC harlequin colour change


FDE fixed drug eruptions HCG human chorionic gonadotropin
FDEIA food‐dependent exercise‐induced urticaria/ HCP hereditary coproporphyria
anaphylaxis HCT haemopoietic cell transplantation
FDH focal dermal hypoplasia HDL high‐density lipoprotein
FFD Fox–Fordyce disease HDN haematodermic neoplasm
FFDD focal facial dermal dysplasias HE hypereosinophilia
FFM focus‐floating microscopy HED hypohidrotic ectodermal dysplasia
FFP fresh frozen plasma HEP hepatoerythropoietic porphyria
FFPE formalin‐fixed paraffin‐embedded HES hypereosinophilic syndrome
FGFR fibroblast growth factor receptor HFMD hand, foot and mouth disease
FH familial hypercholesterolaemia HFS hyaline fibromatosis syndrome
FHL familial haemophagocytic lymphohistiocytosis HGA homogentisic acid
FII fabricated or induced illness HGPS Hutchinson–Gilford progeria syndrome
FISH fluorescence in situ hybridization HHD Hailey–Hailey disease
FITC fluorescein isothiocyanate HHML hemihyperplasia–multiple lipomatosis syndrome
5‐FU 5‐fluorouracil HHT hereditary haemorrhagic telangiectasia
FMF familial Mediterranean fever HHV human herpesvirus
FML familial multiple lipomatosis HI haemangioma of infancy
FNAC fine needle aspiration cytology HI harlequin ichthyosis
FOP fibrodysplasia ossificans progressiva HI hypomelanosis of Ito
FPHH familial progressive hyperpigmentation and HID hystrix‐like ichthyosis with deafness (syndrome)
hypopigmentation HIES hyperimmunoglobulin E syndrome
FPLD familial partial lipodystrophies HIF hypoxia inducible factor
FSH follicle‐stimulating hormone HIMS hyperimmunoglobulin M syndrome
FTC familial tumoural calcinosis HIP helix initiation peptide
FTG full‐thickness skin graft HIV human immunodeficiency virus
FTI farnesyltransferase inhibitors HJMD hypotrichosis with juvenile macular dystrophy
FVC forced vital capacity HLA human leucocyte antigen
GA granuloma annulare HLH haemophagocytic lymphohistiocytosis
GABA γ‐aminobutyric acid HLRCC hereditary leiomyomatosis and renal cell cancer
GACI generalized arterial calcification of infancy HMG high mobility group
GAS Group A Streptococcus HMS Haim–Munk syndrome
GBFDE generalized bullous fixed drug eruption HOME Harmonising Outcome Measures for Eczema
GCDFP gross cystic disease fluid protein HOPP hypotrichosis–osteolysis–periodontitis–palmoplantar
GCS Giannott—Crosti syndrome keratoderma
GD Gaucher disease HP hydroxylysylpyridinoline
GFR glomerular filtration rate HPC haemangiopericytoma
GGR global genome repair HPS Hermansky–Pudlak syndrome
GGT gamma‐glutamyl transpeptidase HPETE hydroperoxyeicosatetraenoic acid
GH growth hormone HPV human papillomavirus
GI gastrointestinal HR H1‐receptor
GLA generalized lymphatic anomaly HRCT high resolution computed tomography
GM‐CSF granulocyte macrophage colony‐stimulating factor HRM high‐resolution melt PCR
GMS Gomori’s methenamine silver HS hidradenitis suppurativa
GNCST granular nerve cell sheath tumour HSCT haematopoietic stem cell transplantation
GO geroderma osteodysplastica HSD holocarboxylase synthetase deficiency
GOSH Great Ormond Street Hospital for Children HSP heat shock proteins
GPA granulomatosis with polyangiitis HSP Henoch–Schönlein purpura
GPP generalized pustular psoriasis HSV herpes simplex virus
GPS Griscelli–Prunieras syndrome HTLV human T‐lymphotropic virus
GS Griscelli syndrome HTP helix termination peptide
GSD Gorham–Stout disease HUV hypocomplementemic urticarial vasculitis
GSE gluten‐sensitive enteropathy HUVS hypocomplementemic urticarial vasculitis syndrome
G6PD glucose‐6‐phosphate dehydrogenase HV hydroa vacciniforme
GSS granulomatous slack skin IA infantile acropustulosis
GVHD graft‐versus‐host disease IBD inflammatory bowel disease
GVHR graft‐versus‐host reaction IBIDS ichthyosis, brittle hair, intellectual impairment,
GVL graft‐versus‐leukaemia decreased fertility and short stature
GVM glomuvenous malformation IBS ichthyosis bullosa of Siemens
GWAS genome‐wide association study ICAM intracellular adhesion molecule
HAART highly active antiretroviral therapy ICD irritant contact dermatitis
HAE hereditary angioedema IC1/IC2 imprinting centre 1/2
HA‐MRSA hospital‐associated meticillin‐resistant Streptococcus ID infective dermatitis
aureus IDQoL Infants’ Dermatitis Quality of Life Index
H&E haematoxylin and eosin IEM inborn errors of metabolism
HBD human β‐defensin IF immunofluorescence
HBT hereditary benign telangiectasia IF infantile fibrosarcoma
HBV hepatitis B virus IF intermediate filament
List of Abbreviations  xxxi

IFA indirect fluorescence assay KS Kindler syndrome


IFAP ichthyosis follicularis, alopecia and photophobia KTS Klippel–Trenaunay syndrome
IFM immunofluorescence mapping KWE keratolytic winter erythema
IFN interferon LABD linear immunoglobulin IgA bullous dermatosis
Ig immunoglobulin LAD leucocyte adhesion deficiency
IgA1 IgA subtype 1 LAD linear IgA dermatosis
Ig ε RI high‐affinity IgE receptor LAH localized autosomal recessive hypotrichosis
IGF insulin‐like growth factor LAM linear atrophoderma of Moulin
IGFBP insulin‐like growth factor binding protein LAM lymphangiomyomatosis
IgM immunoglobulin M LAS loose anagen syndrome
IGRA interferon‐γ release assay LB Lyme borreliosis
IH infantile haemangiomas LCD liquor carbonis detergens
IHCM ichthyosis hystrix of Curth–Macklin LCH Langerhans cell histiocytosis
IHS ichthyosis hypotrichosis syndrome LCR locus control region
IHSC ichthyosis–hypotrichosis–sclerosing cholangitis lcSSC limited cutaneous systemic sclerosis
(syndrome) LED light‐emitting diode
IIF indirect immunofluorescence LIC localized intravascular coagulopathy
IL interleukin LD linkage disequilibrium
ILAR International League of Associations for LD lymphoedema‐distichiasis
Rheumatology LDF laser Doppler flowmetry
ILC ichthyosis linearis circumflexia LDH lactate dehydrogenase
ILVEN inflammatory linear verrucous epidermal naevus LDL low‐density lipoprotein
IMF immunofluorescence LDS Loeys–Dietz syndrome
IM infectious mononucleosis LE lupus erythematosus
IM intramuscular LEC lymphatic endothelial cells
IP incontinentia pigmenti LEKTI lymphoepithelial Kazal‐type inhibitor
iPCS inducible pluripotential stem cells LET lidocaine/epinephrine/tetracaine
IPEX immune dysregulation, polyendocrinopathy, LFA‐3 lymphocyte function‐associated antigen‐3
enteropathy, X‐linked LH luteinizing hormone
IPP infantile perineal protrusion LI lamellar ichthyosis
IPS ichthyosis prematurity syndrome LJ Lowenstein–Jensen (medium)
IR insulin resistance LM lymphatic malformation
IRAK‐4 IL‐1 receptor associated kinase‐4 LMP last menstrual period
IRIS immune reconstitution inflammatory syndrome LMP1 latent membrane protein 1
IS infantile spasms LMS Lenz–Majewski syndrome
ISAAC International Study of Asthma and Allergies in LMS limb–mammary syndrome
Childhood LMX liposomal lidocaine
ISD infantile seborrhoeic dermatitis LN lichen nitidus
ISH infantile systemic hyalinosis LOH loss of heterozygosity
ISM indolent systemic mastocytosis LOSSI localized scleroderma severity index
ISSVA International Society for the Study of Vascular LOX lipoxygenase
Anomalies LP lichen planus
ISU idiopathic solar urticaria LP lupus panniculitis
IV ichthyosis vulgaris LP lysylpyridinoline
IVF in vitro fertilization LPL lipoprotein lipase deficiency
IVIG intravenous immunoglobulin LPP lichen planopilaris
JAK janus kinase LPS lipopolysaccharide
JDM juvenile dermatomyositis LS lichen sclerosus
JEB junctional epidermolysis bullosa LS lichen scrofulosorum
JHF juvenile hyaline fibromatosis LS lichen striatus
JIA juvenile idiopathic arthritis LSC lichen simplex chronicus
JLI Jessner lymphocytic infiltrate LSc localized scleroderma
JPD juvenile plantar dermatosis LT‐β lymphotoxin‐β
JSPD Japanese Society for Pediatric Dermatology LTT lymphocyte transformation test
JSLE juvenile systemic lupus erythematosus LV lentiviral vector
JSSc juvenile‐onset systemic sclerosis LV livedoid vasculopathy
JXG juvenile xanthogranuloma LV lupus vulgaris
KD Kawasaki disease LyP lymphomatoid papulosis
KEN keratinocytic epidermal naevus MAC membrane attack complex
KFSD keratosis follicularis spinulosa decalvans MACS macrocephaly‐alopecia‐cutis laxa‐scoliosis
KHE kaposiform haemangioendothelioma MACS magnetic‐activated cell sorting
KID keratitis, ichthyosis and deafness (syndrome) MAD mandibuloacral dysplasia
KIF keratin intermediate filaments MAIC M. avium‐intracellulare complex
KLA Kaposiform lymphangiomatosis MALDI‐TOF matrix‐assisted laser desorption/ionization
KLICK keratosis linearis‐ichthyosis congenita‐keratoderma time‐of‐flight
KLK kallikrein MALT mucosa‐associated lymphoid tissue
KMP Kasabach–Merritt phenomenon MAPK mitogen‐activated protein kinase
KPI keratinopathic ichthyoses MAS macrophage activation syndrome
KS Kaposi sarcoma MAS McCune–Albright syndrome
xxxii  List of Abbreviations

MBL mannose‐binding lectin NBCC naevoid basal cell carcinoma


MBP major basic protein NBD nucleotide‐binding domain
MBTPS2 membrane‐bound transcription factor protease, site 2 NBS Nijmegen breakage syndrome
MC mast cells NBT nitroblue tetrazolium
MC molluscum contagiosum NB‐UVB narrow‐band UVB
MCAS mast cell activation symptoms NC naevus comedonicus
MC&S microscopy, culture and (antibiotic) sensitivity NCAM neural cell adhesion molecule
MCL mast cell leukaemia NCIE nonbullous congenital ichthyosiform erythroderma
MCL mucocutaneous leishmaniasis NCP neonatal cephalic pustulosis
M‐CM megalencephaly‐capillary malformation NCV nerve conduction velocity
MCS mast cell lymphoma ND naevus depigmentosus
MCV molluscum contagiosum virus Nd:YAG neodymium:yttrium aluminium garnet
MDM minor determinant mixture NEP neonatal eosinophilic pustulosis
MDP mandibular hypoplasia, deafness and progeria NEPPK nonepidermolytic palmoplantar keratoderma
(syndrome) NER nucleotide excision repair
MDR multidrug‐resistant NET neutrophil extracellular trap
MED minimal erythema dose NF necrotizing fasciitis
MEDNIK mental retardation, enteropathy, deafness, peripheral NFJ Naegeli‐Franceschetti‐Jadassohn (syndrome)
neuropathy, ichthyosis, keratoderma NF‐κB nuclear factor κB
MeDOC mendelian disorders of cornification NF1 neurofibromatosis type 1
MEN‐1 multiple endocrine neoplasia type‐1 NGCO non‐gestational ovarian choriocarcinoma
MetS metabolic syndrome NGF nerve growth factor
MF mycosis fungoides NGFR nerve growth factor receptor
MFS Marfan syndrome NGS next‐generation sequencing
MFT multiple familial trichoepitheliomas NHL non‐Hodgkin lymphoma
mHA minor histocompatibility antigen NICE National Institute for Health and Care Excellence (UK)
MHC major histocompatibility complex NICH noninvoluting congenital haemangioma
MIDAS microphthalmia, dermal aplasia and sclerocornea NICU neonatal intensive care unit
(syndrome) NIH National Institutes of Health (USA)
MKD mevalonate kinase deficiency NISCH neonatal ichthyosis‐sclerosing cholangitis
MLPA multiplex ligation‐dependent probe amplification (syndrome)
MLT multifocal lymphangioendotheliomatosis with NK natural killer (cell)
thrombocyopenia NL necrobiosis lipoidica
MMA methylmalonic acidaemia NL neonatal lupus
MMF mycophenolate mofetil NLCH non‐Langerhans cell histiocytosis
MMP matrix metalloproteinases NLCS naevus lipomatosus cutaneous superficialis
MMP mucous membrane pemphigoid NLD necrobiosis lipoidica
MMPH multifocal micronodular pneumocyte hyperplasia NLP nail lichen planus
MMR mismatch repair NLS naevus lipomatosus superficialis
MODY maturity‐onset diabetes of the young NLS Neu‐Laxova syndrome
MPA microscopic polyangiitis NLSD neutral lipid storage disease
MPCM maculopapular cutaneous mastocytosis NLSDI neutral lipid storage disease with ichthyosis
MPDE maculopapular drug eruptions or exanthems NMF natural moisturizing factor
MPE maculopapular exanthems NOMID neonatal onset multisystemic inflammatory disorder
MPNST malignant peripheral nerve sheath tumour N2O nitrous oxide
MPO myeloperoxidase NPSA National Patient Safety Agency (UK)
MPS mucopolysaccharidoses NP‐SLE neuropsychiatric systemic lupus erythematosus
MRA magnetic resonance angiography NPY neuropeptide Y
MRI magnetic resonance imaging NS naevus sebaceus
MRSA meticillin‐resistant Staphylococcus aureus NS Netherton syndrome
MRSS modified Rodnan skin score NS Noonan syndrome
MSD multiple sulphatase deficiency NSML Noonan syndrome with multiple lentigines
MSF Milton sterilizing fluid NSAIDs nonsteroidal anti‐inflammatory drugs
MSH melanocyte‐stimulating hormone NSIP nonspecific interstitial pneumonia
MSL multiple symmetric lipomatosis NSV nonsegmental vitiligo
MSS Mulvihill–Smith syndrome NTM nontuberculous mycobacteria
MSUD maple syrup urine disease NUV normocomplementemic urticarial vasculitis
MTC Mycobacterium tuberculosis complex OA ocular albinism
mtDNA mitochondrial DNA OC osteoma cutis
mTOR mammalian target of rapamycin OCA oculocutaneous albinism
MTS Muir–Torre syndrome OCCS oculocerebrocutaneous syndrome
MTX methotrexate OES oculoectodermal syndrome
MUGA multiple uptake gated acquisition angiography OFC oral food challenge
MUHH Marie–Unna hereditary hypotrichosis OFG orofacial granulomatosis
MVP mitral valve prolapse OI osteogenesis imperfecta
MWS Muckle–Wells syndrome OL‐EDA‐ID osteopetrosis, lymphoedema, anhidrotic ectodermal
NA naevus anaemicus dysplasia and immune deficiency
NAC N‐acetylcysteine OMIM Online Mendelian Inheritance in Man
NADPH nicotinamide adenine dinucleotide phosphate OODD odonto‐onycho‐dermal dysplasia
List of Abbreviations  xxxiii

OPG optic pathway glioma PKDL post‐kala‐azar dermal leishmaniasis


OPK osteopoikilosis PKS Pallister–Killian syndrome
ORF open reading frame PKU phenylketonuria
OS Omenn syndrome pI isoelectric point
OSD occult spinal dysraphism PI3 proteinase inhibitor 3
OTU operational taxonomic unit PIP proximal interphalangeal
O/W oil in water PL pityriasis lichenoides
PA pityriasis alba PLC pityriasis lichenoides chronica
PA phytanic acid PLCA primary localized cutaneous amyloidosis
PABA para‐aminobenzoic acid PLE polymorphous light eruption
PAF platelet‐activating factor PLEVA pityriasis lichenoides et varioliformis acuta
PAH phenylalanine hydroxylase PLS Papillon–Lefèvre syndrome
PAHX phytanoyl CoA hydroxylase PM porokeratosis of Mibelli
PAI‐1 plasminogen activator inhibitor 1 PML progressive multifocal leucoencephalopathy
PAN polyarteritis nodosa PMLE polymorphous light eruption
p‐ANCA perinuclear antineutrophilic cytoplasmic antibody PMN polymorphonuclear cell
PAP peak arterial pressure PN poikiloderma with neutropenia
PAPA pyogenic arthritis, pyoderma gangrenosum and acne PN prurigo nodularis
PAR2 protease‐activated receptor 2 PNET peripheral neuroectodermal tumour
PAS periodic acid–Schiff PNF plexiform neurofibroma
PASI Psoriasis Area and Severity Index POC point of care
PC pachyonychia congenita P1cp procollagen type 1 carboxy‐terminal peptide
PCFCL primary cutaneous follicle‐centre lymphoma PNT papulonecrotic tuberculid
PCFH precalcaneal congenital fibrolipomatous hamartoma POEM Patient‐Oriented Eczema Measure
PCL primary cutaneous lymphoma POEMS polyneuropathy, organomegaly, endocrinopathy,
PCLBCL primary cutaneous diffuse large B‐cell lymphoma M protein, skin changes (syndrome)
PCMZL primary cutaneous marginal zone B‐cell lymphoma POH progressive osseous heteroplasia
PCNA proliferating cell nuclear antigen PP prurigo pigmentosa
PCOS polycystic ovarian syndrome PPAR peroxisome proliferator‐activated receptor
PCR polymerase chain reaction PPGSS papular‐purpuric gloves‐and‐socks syndrome
PCT porphyria cutanea tarda PPi inorganic pyrophosphate
PCT primary care trust PPK palmoplantar keratoderma
PDGF platelet‐derived growth factor PPK phacomatosis pigmentokeratotica
PDL pulsed‐dye laser PPKN palmoplantar keratoderma Nagashima
PDT photodynamic therapy PPL penicilloyl‐polylysine
PE pulmonary embolism PPP palmoplantar pustulosis
PEG polyethylene glycol PPV phakomatosis pigmentovascularis
PEH palmoplantar eccrine hidradenitis PPPD porokeratosis palmaris et plantaris disseminata
PELVIS perianal haemangioma, external genitalia PPT positive patch test
malformations, lipomyelomeningocoele, vesicorenal PPTR positive patch test reaction
abnormalities, imperforate anus and skin tags PR pagetoid reticulosis
PEP postexposure prophylaxis PRES Paediatric Rheumatology European Society
PEPD paroxysmal extreme pain disorder PRINTO Paediatric Rheumatology International Trials
PEN porokeratotic eccrine naevus Organization
PENS papular epidermal naevus with skyline basal cell layer PRIS propofol‐related infusion syndrome
PEODDN porokeratotic eccrine ostial and dermal duct naevus PRNT plaque‐reduction neutralization test
PET positron emission tomography PRO patient‐reported outcome
PF pemphigus foliaceus PROM patient‐reported outcome measure
PFT pulmonary function tests PROS PIK3CA‐related overgrowth spectrum disorders
PG pyoderma gangrenosum PRP pityriasis rubra pilaris
PG pyogenic granuloma PRR pathogen recognition receptors
PGA Physician Global Assessment PR3 proteinase 3
PGD preimplantation genetic diagnosis PS Proteus syndrome
PGP 9.5 protein gene product 9.5 PSEK progressive symmetric erythrokeratoderma
PGRP peptidoglycan recognition protein PSH premature sebaceous hyperplasia
PH palmoplantar hidradenitis PSS peeling skin syndromes
PH pulmonary hypertension PT prothrombin time
PHA phytohaemagglutinin PTC premature termination codon
PHACES posterior fossa brain malformations, large or PTEN phosphatase and TENsin homologue
complex haemangiomas of the face, arterial PTH parathyroid hormone
anomalies, cardiac anomalies and eye abnormalities PTHrP parathyroid‐hormone‐related peptide
PHP pseudo‐hypoparathyroidism PTT partial thromboplastin time
PHTS PTEN hamartoma tumour syndrome PUVA psoralens plus UVA
PhyH phytanoyl‐CoA2‐hydroxylase PV pemphigus vulgaris
PICH partially involuting congenital haemangioma PV psoriasis vulgaris
PID pelvic inflammatory disease PVL Panton–Valentine leukocidin
PID primary immunodeficiency PWS port wine stain
PILA papillary intralymphatic angioendothelioma PXE pseudoxanthoma elasticum
PJS Peutz–Jeghers syndrome QoL quality of life
xxxiv  List of Abbreviations

qPCR quantitative polymerase chain reaction SFT solitary fibrous tumour


qRT‐PCR quantitative real‐time PCR SGC Shprintzen–Goldberg craniosynostosis
QUADAS Quality Assessment of Diagnostic Accuracy tool SHBG sex hormone‐binding globulin
RAK reticulate acropigmentation of Kitamura SHCB self‐healing collodion baby
RAMBA retinoic acid metabolism blocking agent SHH Sonic Hedgehog
RAS recurrent aphthous stomatitis SHFM split hand–split foot syndrome
RAST radio‐allergosorbent test SHP Schönlein–Henoch purpura
RBP retinol‐binding protein SIB self‐injurious behaviour
RCC renal cell carcinoma SICI self‐improving congenital ichthyosis
RCDP rhizomelic chondrodysplasia punctata SID sudden infant death (syndrome)
RCT randomized controlled trial sIgE drug‐specific IgE antibodies
RD restrictive dermopathy siRNA small interfering RNA
RDD Rosai–Dorfman disease SIRS systemic inflammatory response syndrome
RDEB recessive dystrophic epidermolysis bullosa 6‐4PP pyrimidine‐6,4‐pyrimidone photoproducts
RICH rapidly involuting congenital haemangioma SJIA systemic juvenile idiopathic arthritis
RF rheumatoid factor SJS Stevens–Johnson syndrome
RF rib fracture SLADP Latin American Society for Pediatric Dermatology
RFC replication factor C S‐LAM spontaneous lymphangiomyomatosis
RFLP restriction fragment length polymorphism SLC27 solute carrier family 27
RH retinal haemorrhage SLE systemic lupus erythematosus
RMH rhabdomyomatous mesenchymal hamartoma SLICC Systemic Lupus International Collaborating Clinics
RMS rhabdomyosarcoma SLN speckled lentiginous naevus
RNP ribonucleoprotein SLOS Smith–Lemli–Opitz syndrome
ROAT repeated open application test SLPI secretory leucocyte protease inhibitor
ROS reactive oxygen species SLS Sjögren–Larsson syndrome
RP relapsing polychondritis SLS sodium lauryl sulphate
RPA replication protein A SM systemic mastocytosis
RPE recurrent toxin‐mediated perineal erythema SM‐AHN systemic mastocytosis with an associated
RRP recurrent respiratory papillomatosis haematological neoplasm
RSC respiratory syncytial virus SMO smoothened
RT‐PCR reverse transcription polymerase chain reaction SNA spherical nucleic acid
RTS Rothmund–Thomson syndrome SNP single nucleotide polymorphism
RTX rituximab SNV single nucleotide variant
RV retroviral vector SP syringocystadenoma papilliferum
RXLI recessive X‐linked ichthyosis SPD Society for Pediatric Dermatology
SA Streptococcus aureus SPECT single‐photon emission computed tomography
SAA serum amyloid A SPF sun protection factor
SAM severe dermatitis, multiple allergies and metabolic SPINK serine protease inhibitor Kazal type
wasting SPRR small proline‐rich proteins
SAPHO synovitis, acne, pustulosis, hyperostosis and osteitis SPTCL subcutaneous panniculitis‐like T‐cell lymphoma
SASSAD six‐area, six‐sign atopic dermatitis (score) SR systemic retinoids
SC stratum corneum SS Sézary syndrome
SC subcutaneous SS Sjögren syndrome
SCALP sebaceous naevus, central nervous system SS Sweet syndrome
abnormalities, aplasia cutis, limbal dermoid and SSc systemic sclerosis
pigmented naevus (syndrome) SSG split‐thickness skin graft
SCAP syringocystadenoma papillifera SSKI saturated solution of potassium iodide
SCC squamous cell carcinoma SSLR serum sickness‐like reaction
SCCE stratum corneum chymotryptic enzyme SSM smouldering systemic mastocytosis
SCE sister chromatid exchange SSP Schöpf–Schulz–Passarge
SCF stem cell factor SSPE subacute sclerosing panencephalitis
SCFN subcutaneous fat necrosis SSRI selective serotonin reuptake inhibitors
SCH spindle cell haemangioma SSS stiff skin syndrome
SCID severe combined immunodeficiency SSSS staphylococcal scalded skin syndrome
SCLE subacute cutaneous lupus erythematosus SSTI skin and soft tissue infection
SCORAD SCORing Atopic Dermatitis STD sexually transmitted disease
SCT stem cell transplantation STI sexually transmitted infection
SCTE stratum corneum tryptic enzyme STS sodium thiosulfate
SD seborrhoeic dermatitis STS steroid sulphatase
SDA Sabouraud dextrose agar STS soft tissue sarcoma
SDH subdural haematoma STSS streptococcal toxic shock syndrome
SEGA subependymal giant cell astrocytoma SWS Sturge–Weber syndrome
SegPD segmental pigmentary disorder TA tufted angioma
SEI superficial epidermolytic ichthyosis TA teichoic acid
SEN scalp–ear–nipple (syndrome) TAC tetracaine/adrenaline/cocaine
SEN subependymal nodules TBE tickborne encephalitis
SERCA2 sarco/endoplasmic reticulum ATPase type 2 TBSA total body surface area
SFD scrofuloderma TCC triple‐combination cream
List of Abbreviations  xxxv

TCI topical calcineurin inhibitors US/LS upper to lower segment


TCR T‐cell receptor UTR untranslated region
TCS topical corticosteroids UV ultraviolet
TDO trichodento‐osseous UVA ultraviolet A
TEN toxic epidermal necrolysis UVB ultraviolet B
TEWL transepidermal water loss UV‐DDB UV‐damaged DNA‐binding
TFIIH transcription factor IIH UVSS UV‐sensitive syndrome
TG transglutaminase UVR ultraviolet radiation
TG triacylglycerol VACTERL vertebral anomalies, anal atresia, congenital cardiac
TGF transforming growth factor anomalies, tracheo‐oesophageal fistula and/or
TG1 transglutaminase‐1 oesophageal atresia, renal anomalies, radial dysplasia
TIMP tissue inhibitor of metalloproteinase and other limb defects
TJ tight junction VDLR Venereal Disease Research Laboratory
TLR toll‐like receptor VEGF vascular endothelial growth factor
TMD transmembrane domains VEGFR3 vascular endothelial growth factor receptor 3
TMEP telangiectasia macularis eruptiva perstans VIG vaccinia immune globulin
TMP‐SMX trimethoprim‐sulfamethoxazole VIT venom immunotherapy
TND twenty‐nail dystrophy VL visceral leishmaniasis
TNF tumour necrosis factor VL visible light
TNFR tumour necrosis factor receptor VLBW very low birthweight
TNPM transient neonatal pustular melanosis VLCFA very‐long‐chain fatty acid
tPA tissue plasminogen activator VLDL very low‐density lipoproteins
TPM transient pustular melanosis VM venous malformation
TPMT thiopurine methyltransferase VMCM cutaneomucosal venous malformation
TPN total parenteral nutrition VS Vohwinkel syndrome
TRAPS TNF receptor superfamily 1A‐associated periodic VZIG varicella zoster immunoglobulin
fever syndrome VZV varicella zoster virus
TREC T‐cell receptor excision circle WAO World Allergy Organization
TRPS trichorhinophalangeal syndrome WAS Wiskott–Aldrich syndrome
TRT thermal relaxation time WGS whole‐genome shotgun
TS tuberous sclerosis WHO World Health Organization
TSC tuberous sclerosis complex W/O water in oil
TSC‐IS TSC‐associated infantile spasms WPWS Wolff–Parkinson–White syndrome
TSH thyroid‐stimulating hormone WRS Wiedemann–Rautenstrauch syndrome
TSS toxic shock syndrome WS Werner syndrome
TST tuberculosis skin test XD X‐linked dominant
TTD trichothiodystrophy XLA X‐linked agammaglobulinaemia
TU tropical ulcer XLI X‐linked ichthyosis
TV Trichomonas vaginalis XLMR X‐linked mental retardation
UD unrelated donor XP xeroderma pigmentosum
uE3 unconjugated oestriol XPV xeroderma pigmentosum variant
UNT unilateral naevoid telangiectasia XR X‐linked recessive
UP urticaria pigmentosa YNS yellow nail syndrome
UPD uniparental disomy ZIKV Zika virus
URDS Urban–Rifkin–Davis syndrome ZNS Zunich neuroectodermal syndrome
US ultrasound
1

C HA PTER   1

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Embryogenesis of the Skin
Lara Wine Lee1 & Karen A. Holbrook2
 Departments of Dermatology and Pediatrics, Medical University of South Carolina, Charleston, SC, USA
1

 Formerly Department of Physiology and Cell Biology, Ohio State University, Columbus, OH, USA
2

Introduction, 1 Embryonic–fetal transition, 10 Conclusion, 35


Time‐scale of skin development, 2 Fetal skin, 16
Embryonic skin, 3 Unique features of developing human skin, 20

Abstract gene regulation; apoptosis; differentiation (structural, biochemical


and functional); and certain longstanding basic ­p henomena
of development such as induction, pattern ­formation and dif-
The skin is a large and complex organ. While skin development be-
ferentiation. Rigorous understanding of embryogenesis ­allows
gins during early embryogenesis, full development is not ­complete
­definition of critical periods when the skin may be more vulner-
until well into the postnatal years. Studies of skin development
able to developmental errors. Further understanding of these
can shed light on a number of basic problems in c­ ontemporary bi-
critical processes advances the study of developmental d ­ isorders
ology: epithelial–mesenchymal interactions that establish organs
of the skin with the promise of improving therapeutic options for
(in skin, these tissue interactions occur in follicle, sweat gland and
these disorders.
nail formation); cell–cell interactions through soluble mediators;

­Introduction in a continuum of events that modifies the skin. It is


The skin is an ideal organ in which to study development c­ haracterized by morphogenetic processes, activation of
because it is readily accessible for observation, sampling new genes and gain of function. In contrast, ageing may
and evaluation. As an interface, it straddles the internal, involve morpholytic processes in which genes are turned
systemic world of the individual and the external environ­ off, resulting in a loss of function. Consideration of this
ment and is modified by both. The skin itself is a compli­ continuum, and the genetic and environmental interac­
cated and complex organ, with the normal structure and tions that come into play throughout life, provides a con­
function of each ‘part’ highly dependent upon what ceptual framework for discussing the place and role of the
­happens in other parts of the skin. In other words, one events in skin morphogenesis.
cannot understand, for example, changes that occur in the Understanding the stages and events of normal human
epidermis without understanding the nature of the der­ skin development is also important from a biomedical
mis since the dermis has major influences on the activities perspective. Skin embryogenesis allows the definition of
and functions of the epidermis. This is the case for each critical periods when the skin may be more vulnerable to
region or structure of the skin. developmental errors. It provides an opportunity to study
Development offers an opportunity to study skin the evolution of skin function, establishing a background
structure and function under more controlled conditions for understanding the natural history of expression of
because the environment of the developing skin is rea­ genetic skin disease in its earliest form. Moreover, advances
sonably constant (controlled light, temperature, pressure, in gene therapy may provide intervention rooted in
etc.). It is therefore possible to investigate how the prop­ understanding normal morphological processes.
erties of the different regions and structures of the skin The unique morphological properties of developing
are coordinately established, presumably under the human skin have always intrigued investigators.
directions of a genetic programme. Specific aspects of the skin that are found only in the
Some of the structures of the skin may be fully formed fetus, such as the periderm, and specific events that
early in the fetal period whereas other structures or result in the formation of complex structures, such as
regions are not complete until well into the postnatal follicle or sweat gland, were often described for specific
years. Full establishment of adult functions of the skin ages only (reviewed in [1–4]). Expansion of studies to
always requires an extended period of development characterize the complete ontogeny of the tissue, region
beyond the stages in utero. Development is the first period or structure then began to include data derived from

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
2 Section 1  Development, Structure and Physiology of the Skin

­ iochemical or immunohistocytochemical assays for the


b fetal period. Histogenesis of all skin regions is initiated
STRUCTURE AND PHYSIOLOGY

expression of specific molecules that were known to in the embryo, and differentiation of some of those tis­
SECTION 1: DEVELOPMENT,

correlate with the state of differentiation or with a sues begins to occur in the first trimester [2]. The boundary
­specific property such as barrier function adhesion. between the first and second trimesters, at 3 months of
Culturing and grafting human embryonic and fetal skin age, is based only on fetal age and not on any specific
(reviewed in [5–8]) and skin‐derived cells [9,10] and changes in structure, composition or function of any
evaluation of skin from fetuses affected with genoder­ region of the skin.
matoses (reviewed in [11–13]), or under conditions of The second trimester includes many important
growth retardation, have also provided insight into events in skin development. Morphogenesis of new
human skin development. Our understanding of skin structures is initiated and there is terminal differentia­
development continues to increase as we apply tion of others. During the third trimester, all parts of
more modern tools of biology to study the skin at all the skin are assembled and the functions of each of
stages of life. them are unfolding. The end of this period is not the
final state of the skin, as there is significant reorganiza­
­References tion of certain units of the skin (e.g. the vasculature),
1 Holbrook KA. Structure and function of the developing human skin. additions to the skin in volume (e.g. the dermal matrix)
In: Goldsmith LA (ed.) Physiology, Biochemistry and Molecular
and functional maturation of many structures of the
Biology of the Skin, 2nd edn. Oxford: Oxford University Press,
1991:63–110. skin (e.g. nerves, sweat glands and stratum corneum)
2 Holbrook KA. Structural and biochemical organogenesis of skin and after birth [3–7].
cutaneous appendages in the fetus and neonate. In: Polin RA, Fox Other important times that should be recognized in
WW (eds) Neonatal and Fetal Medicine Physiology and
Pathophysiology. New York: Grune & Stratton, 1992:527–51.
skin development are the ages at which diagnostic proce­
3 Holbrook KA, Wolff K. The structure and development of skin. In: dures are performed for the purpose of evaluating the
Fitzpatrick TB, Eisen AZ, Wolff K et  al. (eds) Dermatology in condition of a fetus at risk for a genetic skin disease
General Medicine, 6th edn. New York: McGraw‐Hill, 1993:97–144. (reviewed in [8]). Fetal deoxyribonucleic acid (DNA)
4 Holbrook KA, Sybert VP. Basic science. In: Schachner L, Hansen R
(eds) Pediatric Dermatology, 2nd edn. New York: Churchill can be extracted from chorionic villi sampled around
Livingstone, 1995. 10 weeks EGA and amniotic fluid cells can be obtained at
5 Holbrook KA, Minami SA. Hair follicle morphogenesis in the human: around 14–16 weeks EGA. Fetal skin can be sampled as
characterization of events in vivo and in vitro. NY Acad Sci 1991;
642:167–96.
early as 16 weeks EGA, however this technique is largely
6 Zeltinger J, Holbrook KA. A model system for long term, serum‐free, obsolete and has been replaced by more advanced diag­
suspension organ culture of human fetal tissues: experiments using nostic and genetic methods. Newer technology allows
digits and skin from multiple body regions. Cell Tissue Res 1997; isolation of fetal DNA from maternal plasma as early as
290:51–60.
7 Lane AT, Scott GA, Day KH. Development of human fetal skin trans­ 9 weeks EGA. The cell‐free DNA testing is currently not
planted to the nude mouse. J Invest Dermatol 1989;93:787–91. widely used for microdeletion syndromes but offers
8 Gilhar A, Gershoni‐Baruch R, Margolis A et al. Dopa reaction of fetal promising noninvasive first‐trimester testing in the
melanocytes before and after skin transplantation onto nude mice.
future. In addition, preimplantation genetic diagnosis has
Br J Dermatol 1995;133:884–9.
9 Oliver AM. The cytokeratin expression of cultured human foetal been used successfully for diagnosis of severe skin d ­ isease
keratinocytes. Br J Dermatol 1990;123:707–16. (reviewed in [8]).
10 Scott G, Ewing J, Ryan D et  al. Stem cell factor regulates human
­melanocyte–matrix interactions. Pigment Cell Res 1994;7:44–51.
11 Holbrook KA, Smith LT, Elias S. Prenatal diagnosis of genetic skin ­References
disease using fetal skin biopsy samples. Arch Dermatol 1993; 1 Holbrook KA, Odland GF. The fine structure of developing human
129:1437–54. epidermis: light, scanning and transmission electron microscopy of
12 Sybert VP, Holbrook KA, Levy M. Prenatal diagnosis of severe the periderm. J Invest Dermatol 1975;65:16–38.
­dermatologic diseases. Adv Dermatol 1992;7:179–209. 2 Holbrook KA, Dale BA, Smith LT et al. Markers of adult skin expressed
13 Sybert VP, Holbrook KA. Antenatal pathology of the skin. In:
in the skin of the first trimester fetus. Curr Prob Dermatol
Claireaux AE, Reed GB (eds) Diseases of the Fetus and Newborn: 1987;16:94–108.
Pathology, Radiology and Genetics. New York: Cockburn, Chapman 3 Holbrook KA. Structure and function of the developing human skin.
& Hall, 1995:755–68. In: Goldsmith LA (ed.) Physiology, Biochemistry and Molecular
Biology of the Skin, 2nd edn. Oxford: Oxford University Press,
1991:63–110.
­Time‐scale of skin development 4 Holbrook KA. Structural and biochemical organogenesis of skin
and cutaneous appendages in the fetus and neonate. In: Polin RA,
There are several schemes for categorizing stages of Fox WW (eds) Neonatal and Fetal Medicine Physiology and
Pathophysiology. New York: Grune & Stratton, 1992:527–51.
skin development (Fig. 1.1) [1]. Development is defined 5 Holbrook KA, Wolff K. The structure and development of skin. In:
by estimated gestational age (EGA) starting at the time Fitzpatrick TB, Eisen AZ, Wolff K et al (eds) Dermatology in General
of fertilization, which differs from calculations based on Medicine, 6th edn. New York: McGraw‐Hill, 1993:97–144.
the last menstrual period (LMP). Fertilization occurs on 6 Holbrook KA, Sybert VP. Basic science. In: Schachner L, Hansen R
(eds) Pediatric Dermatology, 2nd edn. New York: Churchill
average 2 weeks after the LMP. Human development is Livingstone, 1995.
separated into the embryonic period, before the onset of 7 Holbrook KA. A histologic comparison of infant and adult skin. In:
bone marrow function, which corresponds to fertiliza­ Boisits E, Maibach HI (eds) Neonatal Skin: Structure and Function.
New York: Marcel Dekker, 1982:3–31.
tion to 2 months EGA, and the fetal period from 8 Luu M, Cantatore‐Francis L, Glick S. Prenatal diagnosis of
2  months EGA until birth. The first trimester includes genodermatoses: current scope and future capabilities. Int J Derm
the entire embryonic period and the first stages of the 2010;49:353–61.
Chapter 1  Embryogenesis of the Skin 3

*Organogenesis

STRUCTURE AND PHYSIOLOGY


Histogenesis

SECTION 1: DEVELOPMENT,
Maturation

Amniocentesis: Fetal skin


Chorionic villus biochemical, biopsy:
sampling: DNA chromosomal morphological
analysis analysis analysis

1 2 3 4 5 6 7 8 9
Months

First trimester Second trimester Third trimester

Embryonic Fetal

Fig. 1.1  Time‐scale diagram identifying specific Embryonic-fetal Follicular Interfollicular


stages of skin development and identifying the transition keratinization epidermal
ages at which prenatal diagnosis can be performed keratinization
using each of the various methods currently
employed. Source: Adapted from Polin RA, Fox *Dashed lines suggest that the starting/ending point
WW. Fetal and Neonatal Physiology, 2nd edn. Vol. for these events is uncertain
1. Philadelphia: W.B. Saunders, 1998:730.

­Embryonic skin
The primitive ectoderm of the developing blastocyst is
established at 1 week EGA, and by 20–50 days EGA the
development of major organs and organ systems of the
human embryo is initiated. The integumentary system
exhibits characteristics of the skin at 30 days EGA. The
epidermis, dermoepidermal junction (DEJ) and dermis
are well delineated and the tissue is innervated and vas­
cularized (Fig.  1.2). The boundary between the dermis
and subcutaneous tissue is not clearly defined in all body
sites, but in some regions these two zones are distinct
from one another on the basis of a greater density of cells
and matrix in the dermis compared with the hypodermis.
The skin is closely associated with the underlying devel­
oping striated muscle or cartilage on the appendages. (a)
There is no morphological evidence that epidermal
appendages have begun to form.
In most regions of the embryo, the epidermis is a sim­
ple, flat, two‐layered epithelium consisting of basal and
periderm cells (Figs 1.2 and 1.3). The periderm is a dis­
tinct embryonic layer that is eventually shed. Both types
of cells are mostly filled with glycogen, a molecule that
is characteristic of the cytoplasm of developing and
regenerating tissues, where it most likely serves as a
(b)
source of energy [1] (Fig.  1.3). Microvilli project from
the peridermal surface into the amniotic fluid (Figs 1.3b Fig. 1.2  (a) Tissue of the body wall of a 36‐day EGA human embryo and
and 1.4). The nucleus is centrally located in periderm (b) the skin from a 45‐day EGA human embryo. Note the two‐layered epidermis,
dermis and subcutaneous tissue and the more linear orientation of dermal cells
and basal cells, and the cytoplasmic organelles are
in contrast to the pleomorphic shapes of the subcutaneous mesenchyme. In
sparse and distributed either around the nucleus or at (b) note the periderm and basal cells of the epidermis, the closely associated
the periphery of the cell (Fig. 1.3b). Both layers contain fibroblastic cells in the dermis proximal to the epidermis and a nerve–vascular
distinct keratin intermediate filament proteins (Fig. 1.5) plane separating the dermis from the subcutaneous tissue (×200).
4 Section 1  Development, Structure and Physiology of the Skin

Fig. 1.3  Transmission electron micrographs of the embryonic epidermis. In


STRUCTURE AND PHYSIOLOGY

(a) note the glycogen (G)‐filled basal (B) and periderm (P) layer cells.
SECTION 1: DEVELOPMENT,

Desmosomes are evident between basal cells and between basal cells and
periderm cells. The DEJ is flat and shows few sites of increased density,
suggesting sites of desmosome formation. In (b) one periderm cell and
P portions of two basal cells are shown. Note the nature and disposition of
cytoplasmic organelles within both cell types, the keratin filaments
associated with desmosomes (arrow) and the microvilli extending from the
periderm surface (a, ×11 525; b, ×25 000).

(a)

Fig. 1.4  Scanning electron micrograph of the surface of 55‐day EGA


embryonic skin from the surface of the developing foot. The layer of cells
shown is the periderm. Note the microvilli and the variable size and shape
of the cells (×1000).

(50 kDa), that are characteristic of adult basal layer


keratinocytes [2,3] and additional keratin polypep­
tides, K19 (40 kDa) and K8 (52 kDa), that are specific to
embryonic/fetal basal cells and periderm cells [2,3]. At
least one keratin polypeptide expressed in periderm
cells is different from those in the basal cells, K18
(45 kDa), although it is a marker for Merkel cells [5]. In
contrast to the adult tissue, the filaments in fetal
embryonic epidermis are dispersed in the cytoplasm
or assembled in small, seemingly short, bundles
that are associated primarily with desmosomes and
hemidesmosomes (see Fig.  1.3b). Periderm cells and
basal cells also differ in the expression of many growth
factors, growth factor receptors (Fig. 1.6), cell adhesion
(b)
molecules and other cytoplasmic and cell‐surface
­molecules [6–8].
Two of the immigrant cells that are prominent in
adult epidermis, melanocytes (neural crest in origin)
[2,3], and unique cell‐surface molecules [4]. The latter and Langerhans cells, are present in the embryonic
markers may reflect the differences in environments epidermis among basal cells and associated with
­surrounding each layer. the basement membrane. Sheets of embryonic epider­
The columnar‐shaped basal cells of the embryonic mis immunostained with an antibody that recognizes
epidermis express the keratins, K5 (58 kDa) and K14 m elanocytes specifically (HMB‐45, an inducible,
­
Chapter 1  Embryogenesis of the Skin 5

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
(a)

Fig. 1.6  Section of skin from a 78‐day EGA human fetus showing
differential expression of the A‐chain of platelet‐derived growth factor
(PDGF) in the basal and intermediate cell layers (green) and an absence of
staining in peridermal cells. The receptor for PDGFA, PDGFR‐α (red), is
expressed by cells in the dermis (×350).

(b)

(c)

(d)
Fig. 1.5  Immunostained samples of (a) early (∼50‐day EGA) and (b–d)
later (∼60‐day) human embryonic epidermis showing positive staining of
both periderm and basal layers with the AE1 (a) and AE3 (d) monoclonal
antibodies that recognize keratins. Both layers are negative when reacted
with the AE2 (c) antibody, which recognizes the differentiation‐specific
keratins (×350).

(a)

cytoplasmic antigen common to melanoma and embryonic/


fetal melanocytes [9,10]) show a remarkably high den­
sity (∼1000 cells/mm2) of these cells organized in a
regular pattern of distribution (Fig. 1.7). They are den­
dritic as early as 50 days EGA in general body skin but
there is no evidence of ­melanosomes in the cytoplasm
[11]. Langerhans cells are recognized in embryonic skin
as early as 42 days EGA on the basis of a reaction prod­
uct for membrane‐bound Mg2+ adenosine triphos­
phatase (ATPase) and histocompatibility locus antigen
(HLA‐DR) on the plasma membrane [12–14]. Their
truncated or dendritic morphology is also apparent (b)
(Fig. 1.8). Interestingly, they are present in skin before
Fig. 1.7  Embryonic skin from a 54‐day EGA human embryo immunostained
the bone marrow begins to function leading to a with the HMB‐45 monoclonal antibody, which recognizes an antigen in the
hypothesis that they are derived from the yolk sac or melanocyte. (a) Section of skin. Note the abundance and position of these
fetal liver at this age. At 7 weeks EGA, the density of cells within the two‐layered epidermis. (b) Epidermal sheet. Note the density,
Langerhans cells is about 50 cells/mm2 [13,14]. spacing and dendritic morphology of these cells (a, ×350; b, ×25).
6 Section 1  Development, Structure and Physiology of the Skin

A continuous basal lamina (lamina densa) underlies the


STRUCTURE AND PHYSIOLOGY

two‐layered epidermis and defines, morphologically, one


SECTION 1: DEVELOPMENT,

structural component of the basement membrane zone


[25–27]. The basal lamina is patchy, however, in regions of
the body where the epidermis may be only a single layer,
for example, superior to the spinal cord. The molecules and
antigens characteristic of all basal laminae (type IV colla­
gen, laminin, heparan sulphate proteoglycan, nidogen/
entactin) are present in the earliest recognized basal lamina
of the skin; skin‐specific molecules are recognized later
during the first trimester in accord with the more promi­
nent development of the attachment structures [28,29].
A thin, mat‐like layer of microfilaments lies just inside the
Fig. 1.8  Epidermal sheet from a 53‐day EGA human embryo immunostained
to recognize HLA‐DR antigen in epidermal Langerhans cells (×400). basal plasma membrane of the basal cell keratinocytes
Source: Micrograph courtesy of Dr Carolyn Foster. (Fig. 1.9). It may reinforce this surface of the epidermis and
add to the strength of the DEJ at this stage when the struc­
tural modifications associated with dermoepidermal adhe­
sion (hemidesmosomes, anchoring filaments, anchoring
The third immigrant cell, the Merkel cell, can be rec­ fibrils) are rudimentary [30]. The same organization of fila­
ognized in embryonic palmar skin as early as 55–60 ments is observed in cultured keratinocytes, which do not
days EGA (see Eccrine sweat gland formation) at a den­ typically form hemidesmosomes and anchoring fibrils in
sity of ∼130 cells/mm2 [15], using as a marker any one vivo, and in basal keratinocytes under pathological situa­
of the set of keratins expressed by Merkel cells (K8, K18, tions, such as j­unctional epidermolysis bullosa, in which
K10 and K20) [5,16–18]. K20 is the only keratin found the epidermis separates from the dermis.
exclusively in Merkel cells [18]. At this embryonic age, The antigens associated with the attachment structures
they are distributed randomly and in a suprabasal posi­ (laminin 5/epiligrin/kalinin and 19 DEJ‐1 for hemides­
tion. Merkel cells are neuroendocrine cells that were mosomes and anchoring filaments [31–34]; type VII
originally thought to function primarily as slow‐adapting ­collagen for anchoring fibrils [35]) are not seen by light
mechanoreceptors. Studies that have found soluble microscopic immunostaining methods until early in the
mediators produced by these cells such as nerve growth fetal period. It is likely, however, that keratinocytes begin
factor (NGF) and brain‐derived neurotrophic factor to synthesize these proteins in the embryonic period but
[19,20] suggest that it is likely that Merkel cells are tar­ that the methods used for detection are not sensitive
gets for ingrowing nerve fibres or other cells such as the enough to demonstrate their low levels of expression.
smooth muscle cells of the arrector pili muscle [21,22]. The dermoepidermal boundary is flat in the embryonic
Their presence in selected sites of developing epidermal skin (Figs  1.2, 1.3 and 1.9) and thus presents a limited
appendages (e.g. sweat glands and hair follicles) has surface area for nutrients to traverse between the dermis
also been suggested to stimulate or to correlate with and the epidermis. This may be relatively less important
active proliferation of the tissue. It is generally accepted in the developing skin than in infant and adult skin
that Merkel cells are derived from keratinocytes in situ because the dermis is thin and the small, dispersed bun­
[16,18,21,23,24]. dles of dermal matrix proteins and the hydrated condition

Fig. 1.9  Enlarged view of the DEJ of human embryonic epidermis showing the microfilament network within the basal epidermal cell (arrows), sites where
desmosomes are forming (arrowhead) and the lamina densa. Note collagen fibrils (C) surrounding the dermal fibroblastic cells (×11 625).
Chapter 1  Embryogenesis of the Skin 7

of the interstitial matrix permit more rapid diffusion of

STRUCTURE AND PHYSIOLOGY


substances than the mature skin.

SECTION 1: DEVELOPMENT,
The dermis in the embryo is highly cellular (Figs  1.2
and 1.10), but it also contains the extracellular fibrous
matrix proteins, types I, III, V and VI interstitial collagens,
characteristic of adult dermis [30,36–43]. Small bundles of
collagen accumulate in a thin, dense layer, called the retic­
ular lamina, immediately beneath the dermoepidermal
interface (Figs 1.2b, 1.5 and 1.9). They are also dispersed
throughout the dermis in varying densities according to
the collagen type and age of the embryo. Types I, III and
VI collagen are distributed uniformly throughout the
dermis. Type V collagen is concentrated primarily along
basement membranes (at the DEJ and around blood ves­
sels) and surrounding cells (Fig.  1.11). Fibre bundles
within the interstitial spaces are widely dispersed by a
hydrated, hyaluronic acid‐rich proteoglycan matrix
(a) [44,45] (Figs 1.11 and 1.12). The fluidity of the matrix at
this stage permits migration of mesenchymal cells to sites
of active tissue morphogenesis.
A broader zone of sulphated proteoglycan‐rich matrix,
called the compact mesenchyme, is delineated beneath
the epidermis on the basis of its rich concentration of cells
that express growth factor receptors – the platelet‐derived
growth factor receptor β (PDGFR‐β) and PDGFR‐α (see
Fig. 1.6), nerve growth factor receptor (NGFR) – and cell
adhesion molecules (e.g. neural cell adhesion molecule,
NCAM) [44,45]. Evidence from the skin of non‐human
species during development has shown enlargement of
the composition of growth factors and receptors and
adhesion molecules that are included in this dermal zone
(reviewed in [44] and [46–48]). The compact mesenchyme
may be involved in the exchange of signals between the
epidermis and dermis and may be very important in
stimulating the onset of appendage formation. Many of
the growth factors that correspond to the receptors on the
mesenchymal cells are produced by cells of the develop­
ing epidermis (e.g. PDGF‐AA, PDGF‐BB and NGF)
(Fig. 1.6). The compact mesenchyme may also be the earli­
est evidence of a papillary dermis. In the adult, the modi­
fied composition and structure of the papillary dermis
probably reflects molecular interactions between the epi­
dermal and dermal cells, similar to the situation of the
compact mesenchyme.
Elastic fibres are not formed in the embryonic skin, but
fibrillin (the microfibrils of elastic fibres) (Fig.  1.13) and
elastin proteins of the elastic fibre can be identified immu­
nohistochemically [30,36–40,42] and microfibrils can be
seen by electron microscopy [30].
Fine nerve fibres and capillaries are present within the
compact mesenchyme and deeper dermis (Fig.  1.14a),
and large nerve trunks and vessels are readily apparent in
the subcutaneous region. Reconstructions of vessels from
(b)
serial sections of developing first‐trimester skin have
Fig. 1.10  Transmission (a) and scanning (b) electron micrographs of the
shown that the basic pattern of cutaneous vasculature is
embryonic dermis at 48 days EGA beginning at the DEJ. The matrix is less
evident in the sectioned sample (a) than in the whole‐mount specimen (b)
established in the first trimester [49]. New vessels pre­
(a, ×4500; b, ×1500). sumably both form de novo from dermal mesenchyme
and sprout from deeper, established vessels through a
process that includes endothelial cell migration, capillary
budding and vessel remodelling [50]. Pieces of full‐thickness
8 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a) (b)

(c) (d)
Fig. 1.11  Samples of embryonic skin immunostained with antibodies that recognize type I (a), III (b), V (c) and VI (d) collagens. Note that all of the collagens
are concentrated beneath the DEJ but types III and V, especially, are found in association with all basement membranes. Types I, III and VI are found in the
matrix throughout the dermal and subcutaneous tissue (a–c, ×150; d, ×300). Source: Immunostaining courtesy of Dr Lynne T. Smith.

Fig. 1.12  Section of the body wall from a 57‐day EGA embryo treated
with the Alcian blue/periodic acid–Schiff (PAS) histochemical stains. The
bright pink staining of the epidermis (glycogen) and DEJ (glycoproteins)
indicates a PAS‐positive reaction. The blue dermis reflects the high content
of hyaluronic acid. The dermal–subcutaneous boundary is marked by a
transition to a lighter slightly purple reaction indicating more of the
collagen–glycosaminoglycan complex (×300). Source:
Immunohistochemistry courtesy of Dr Richard Frederickson.

Fig. 1.13  Section of skin from a 57‐day EGA human embryo immu-
nostained with an antifibrillin antibody. Note staining throughout the
dermis (×200). Source: Immunostaining courtesy of Dr Lynne T. Smith.
Chapter 1  Embryogenesis of the Skin 9

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
(a)
Fig. 1.15  Nerves and vessels in the skin of a 79‐day EGA human fetus
immunostained with an anti‐neurofilament antibody. Note the positively
stained nerve network, the immunopositive cells (presumably Merkel cells)
and the vascular network (clear) (×25). Source: Immunostaining courtesy
of Dr Mark Bressler.

fibres become organized in networks throughout the


­ ermis and in relation to developing epidermal append­
d
ages [53]. At 7 weeks EGA, a few calcitonin gene‐related
product (CGRP)‐immunopositive fibres, denoting sen­
sory fibres, are also evident [53], but autonomic nerves
are not yet recognized in the skin. Staining the tissue with
the p75 low‐affinity NGFR antibody also reveals the pat­
terns of nerve fibres and specific concentrations of mesen­
chymal cells (e.g. around developing hair follicles) [54].
(b) Both nerves and vessels are visible in stained, full‐thickness
samples of the nearly transparent skin (Fig. 1.15).

­References
1 Sharp F. A quantitative study of the glycogen content of human fetal
skin in the first trimester. J Obstet Gynaecol Br Commonw
1971;78:981–6.
2 Dale BA, Holbrook KA, Kimball JR et al. Expression of the epider-
mal keratins and filaggrin during fetal human development. J Cell
Biol 1985;101:1257–69.
3 Moll R, Moll I, Wiest W. Changes in the pattern of cytokeratin poly­
peptides in epidermis and hair follicles during skin development in
human fetuses. Differentiation 1983;23:170–8.
4 Dabelsteen E, Holbrook KA, Clausen H et  al. Cell surface carbohy­
drate changes during embryonic and fetal skin development. J Invest
Dermatol 1986;87:81–5.
5 Moll R, Moll I. Early development of human Merkel cells. Exp Dermatol
1992;1:180–4.
(c) 6 Piepkorn M, Underwood RA, Henneman C et  al. Expression of
amphiregulin is regulated in cultured human keratinocytes and in
Fig. 1.14  Sections of human embryonic skin at 42 days EGA (a) and 59
developing fetal skin. J Invest Dermatol 1996;105:802–9.
days EGA (b) immunostained with PGP 9.5, which recognizes all cutaneous 7 Nanney LB, Stoscheck CM, King LE et  al. Immunolocalization of
nerves, and of a sample of 52‐day EGA embryonic skin (c) immunostained ­epidermal growth factor receptors in normal developing human skin.
with p75 antibody, which recognizes the low‐affinity NGFR. Note the large J Invest Dermatol 1990;94:742–8.
nerve trunks deep in the subcutaneous tissue (a), the significant density of 8 Fujita M, Furukawa F, Fujii K et al. Expression of cadherin molecules
the fine fibres in the tangential section of the dermis of (b) and the during human skin development: morphogenesis of epidermis,
distribution of both nerves and vessels (c) (a, ×100; b, ×200; c, ×200). hair follicles and eccrine sweat ducts. Arch Dermatol Res 1992;
284:159–66.
9 Gown AM, Vogel AM, Hoak D et al. Monoclonal antibodies specific
skin and sections of skin immunostained with an anti­ for melanocytic tumors distinguish populations of melanocytes. Am J
Pathol 1986;123:195–203.
body that demonstrates all cutaneous nerves (protein 10 Smoller BR, Hsu A, Krueger J. HMB‐45 monoclonal antibody recog­
gene product 9.5 or PGP 9.5) [51,52] reveal finely beaded nizes an inducible and reversible melanocyte cytoplasmic protein.
nerve filaments distributed in an impressive density in J Cutan Pathol 1991;8:315–22.
11 Holbrook KA, Underwood RA, Vogel AM et al. The appearance, den­
the subepidermal region and in association with blood
sity and distribution of melanocytes in human embryonic and fetal
vessels (Fig. 1.14b and c). The number of fibres recognized skin revealed by the anti‐melanoma monoclonal antibody, HMB45.
by this antibody increases during development as the Anat Embryol 1989;180:443–55.
10 Section 1  Development, Structure and Physiology of the Skin

12 Foster CA, Holbrook KA, Farr AG. Ontogeny of Langerhans cells in of a new skin basement membrane antigenic defect in junctional and
human embryonic and fetal skin: expression of HLA‐DR and OKT‐6 dystrophic epidermolysis bullosa. Arch Dermatol 1989; 125:520–3.
STRUCTURE AND PHYSIOLOGY

determinants. J Invest Dermatol 1986;86:240–3. 35 Burgeson RE. Type VII collagen, anchoring fibrils and epidermolysis
SECTION 1: DEVELOPMENT,

13 Drijkoningen M, DeWolf‐Peeters C, VanDerSteen K et al. Epidermal bullosa. J Invest Dermatol 1993;101:252–5.


Langerhans cells and dermal dendritic cells in human fetal and neo­ 36 Holbrook KA. Structural and biochemical organogenesis of skin
natal skin: an immunohistochemical study. Pediatr Dermatol and cutaneous appendages in the fetus and neonate. In: Polin RA,
1987;4:11–17. Fox WW (eds) Neonatal and Fetal Medicine Physiology and
14 Foster CA, Holbrook KA. Ontogeny of Langerhans cells in human Pathophysiology. New York: Grune & Stratton, 1992:527–51.
embryonic and fetal skin: cell densities and phenotypic expression 37 Holbrook KA, Wolff K. The structure and development of skin. In:
relative to epidermal growth. Am J Anat 1989;84:157–64. Fitzpatrick TB, Eisen AZ, Wolff K et al. (eds) Dermatology in General
15 Kim D‐G, Holbrook KA. The appearance, density and distribution Medicine, 6th edn. New York: McGraw‐Hill, 1993:97–144.
of Merkel cells in human embryonic and fetal skin: their relation to 38 Holbrook KA, Sybert VP. Basic science. In: Schachner L, Hansen R
sweat gland and hair follicle development. J Invest Dermatol (eds) Pediatric Dermatology, 2nd edn. New York: Churchill
1995;104:411–16. Livingstone, 1995.
16 Moll I, Moll R, Franke W. Formation of epidermal and dermal Merkel 39 Smith LT, Holbrook KA, Byers PH. Structure of the dermal matrix
cells during human fetal skin development. J Invest Dermatol during development and in the adult. J Invest Dermatol 1982;
1986;87:779–87. 79:93S–104S.
17 Moll R, Löwe A, Laufer J et al. Cytokeratin 20 in human carcinomas: a 40 Smith LT, Holbrook KA. Development of dermal connective tissue in
new histodiagnostic marker detected by monoclonal antibodies. Am J human embryonic and fetal skin. Scan Electron Microsc 1982:
Pathol 1992;140:427–47. 1745–51.
18 Moll I, Kuhn C, Moll R. Cytokeratin 20 is a general marker of cutane­ 41 Smith LT, Holbrook KA, Madri JA. Collagens types I, III and V in
ous Merkel cells while certain neuronal proteins are absent. J Invest human embryonic and fetal skin. Am J Anat 1986;175:507–22.
Dermatol 1995;104:910–15. 42 Smith LT, Holbrook KA. Embryogenesis of the dermis. Pediatr

19 Vos P, Stark F, Pittman RN. Merkel cells in vitro: production of nerve Dermatol 1986;3:271–80.
growth factor and selective interactions with sensory neurons. Dev 43 Smith LT. Patterns of type VI collagen compared to types I, III, and V
Biol 1991;144:281–300. collagen in human embryonic and fetal skin and in fetal skin‐derived
20 Reed‐Geaghan EG, Wright MC, See LA et al. Merkel cell‐driven BDNF cell cultures. Matrix Biol 1994;14:159–70.
signaling specifies SAI neuron molecular and electrophysiological 44 Holbrook KA, Smith LT, Kaplan ED et al. The expression of mor-
phenotypes. J Neurosci 2016;36:4362–76. phogens during human follicle development in vivo and a model
21 Narisawa Y, Hashimoto K, Nakamura Y et al. A high concentration of for studying follicle morphogenesis in vitro. J Invest Dermatol
Merkel cells in the bulge prior to the attachment of the arrector pili 1993;101:39S–49S.
muscle and the formation of the perifollicular nerve plexus in human 45 Kaplan ED, Holbrook KA. Dynamic expression patterns of tenascin,
fetal skin. Arch Dermatol Res 1993;285:261–8. proteoglycans and cell adhesion molecules during human hair follicle
22 Moore SJ, Munger BL. The early ontogeny of the afferent nerves and morphogenesis. Dev Dyn 1994;199:141–55.
papillary ridges in human digital and glabrous skin. Dev Brain Res 46 Chuong C‐M, Widelitz RB, Jiang T‐X. Adhesion molecules and home­
1989;48:119–41. oproteins in the phenotypic determination of skin appendages.
23 Moll I, Lane AT, Franke WW et al. Intraepidermal formation of Merkel J Invest Dermatol 1993;101:10S–15S.
cells in xenografts of human fetal skin. J Invest Dermatol 1990; 47 Chuong C‐M, Widelitz RB, Ting‐Berreth S et al. Early events during
94:359–64. avian skin appendage regeneration: dependence on epithelial–mes­
24 Tilling T, Wladykowski E, Failla AV et al. Immunohistochemical anal­ enchymal interaction and order of molecular reappearance. J Invest
yses point to epidermal origin of human Merkel cells. Histochem Cell Dermatol 1996;107:639–46.
Biol 2014;141:407–21. 48 Widelitz RB, Jiang T‐X, Noveen A et  al. FGF induces new feather
25 Marinkovich MP, Keene DR, Rimberg CL. Cellular origin of the der­ buds from developing avian skin. J Invest Dermatol 1996;107:
mal–epidermal basement membrane. Dev Dyn 1993;196:255–67. 797–803.
26 Christiano AM, Uitto J. Molecular complexity of the cutaneous base­ 49 Johnson CL, Holbrook KA. Development of human embryonic and
ment membrane zone: revelations through the paradigms of epider­ fetal dermal vasculature. J Invest Dermatol 1989;93:10S–17S.
molysis bullosa. Exp Dermatol 1996;5:1–11. 50 Karelina TV, Goldberg GI, Eisen AZ. Matrix metalloproteinases in
27 Uitto J, Pulkkinen L. Molecular complexity of the cutaneous basement blood vessel development in human fetal skin and in cutaneous
membrane zone. Mol Biol Rep 1996;23:35–46. tumors. J Invest Dermatol 1995;105:411–17.
28 Fine JD, Smith LT, Holbrook KA et al. The appearance of four base­ 51 Dalsgaard CJ, Rydh M, Haegerstrand A. Cutaneous innervation in
ment membrane zone antigens in developing human fetal skin. man visualized with protein gene product 9.5 (PGP 9.5) antibodies.
J Invest Dermatol 1984;83:66–9. Histochemistry 1989;92:385–9.
29 Smith LT, Sakai LY, Burgeson RE et al. Ontogeny of structural compo­ 52 Gulbenkian S, Wharton J, Polak JM. The visualisation of cardiovascu­
nents at the dermal–epidermal junction in human embryonic and lar innervation in the guinea pig using an antibody to protein gene
fetal skin: the appearance of anchoring fibrils and type VII collagen. product 9.5. J Autonom Nerv Syst 1987;18:235–47.
J Invest Dermatol 1988;90:480–5. 53 Terenghi G, Sundaresan M, Moscoso G et  al. Neuropeptides and a
30 Holbrook KA. Structure and function of the developing human skin. neuronal marker in cutaneous innervation during human foetal
In: Goldsmith LA (ed.) Physiology, Biochemistry and Molecular development. J Comp Neurol 1993;328:595–603.
Biology of the Skin, 2nd edn. Oxford: Oxford University Press, 54 Holbrook KA, Bothwell MA, Schatteman G et al. Nerve growth factor
1991:63–110. receptor labelling defines developing nerve networks and stains
31 Hopkinson SB, Riddelle KS, Jones JCR. Cytoplasmic domain of the ­follicle connective tissue cells in human embryonic and fetal skin.
180‐kD bullous pemphigoid antigen, a hemidesmosomal component: J Invest Dermatol 1988;90:609A.
molecular and cell biologic characterization. J Invest Dermatol
1992;99:264–70.
32 Ishiko A, Shimizu H, Kikuchi A et al. Human autoantibodies against ­Embryonic–fetal transition
the 230 kD bullous pemphigoid antigen (BPAG1) bind only to the
intracellular domain of hemidesmosome, whereas those against the The most remarkable time in skin development is the
180 kD bullous pemphigoid antigen (BPAG2) bind along the plasma embryonic–fetal transition, which occurs at approxi­
membrane of hemidesmosome in normal human and swine skin.
J Clin Invest 1993;91:1608–15. mately 2 months’ gestation, when the embryo measures
33 Verrando P, Blanchet‐Bardon C, Pisani A et al. Monoclonal antibody about 31 mm in length (crown–rump), weighs about 2.5 g
GB3 defines a widespread defect of several basement membranes and and has a humanoid appearance. The skin and the under­
a keratinocyte dysfunction in patients with lethal junctional epider­
molysis bullosa. Lab Invest 1991;64:85–92.
lying tissues of the body wall are translucent, revealing
34 Fine J‐D, Horiguchi Y, Couchman JR. 19‐DEJ‐1, a hemidesmosomal– the ribs and solid organs. The skin has a mucoid quality
anchoring filament complex associated monoclonal antibody: definition (Fig. 1.16). In spite of this structural simplicity, the cells in
Chapter 1  Embryogenesis of the Skin 11

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
c

n c
n

(a)

M
Fig. 1.16  Sample of human fetal skin of 80 days EGA held at the tips of a
forceps and demonstrating the mucoid quality of the tissue.

the skin begin to express characteristics of adult skin. The


2‐month age is thus identified as an important landmark
in skin development (see Fig.  1.1). Since this period of
development begins the establishment of adult character­
istics of the skin, it is a stage that is vulnerable to errors in
n
development. The most apparent change in the skin is the
stratification of the epidermis from two to three cell layers
(Fig. 1.17a and b). An intermediate cell layer is added as
the product of basal cell mitoses. Basal cells divide asyn­ (b)
chronously to produce an epidermis that, initially, remains
Fig. 1.17  Skin from a human fetus of approximately 70–89 days EGA
two‐layered at some sites and at others becomes three lay­
shown at the light (a) and electron (b) microscopic levels. Note the
ers thick. intermediate layer of cells between basal and periderm epidermal layers,
Intermediate cells are both similar to and distinct from the distinction between dermis and subcutaneous tissue based on
basal and periderm cells. Keratins are more abundant differences in the orientation of fibroblastic cells, the density of collagenous
and distributed in a more specific distribution than in matrix and the subcutaneous vascular plane. Small nerves (n) and
the cells in the basal and periderm layers; small bundles capillaries (c) are evident in the dermis. Segments of melanocytes (M)
of keratin filaments associated with desmosomes outline are evident within the basal layer and collagen is accumulated beneath
the DEJ (×3675).
the boundaries of the intermediate cells (Fig. 1.17b). The
expression of the major keratin pairs in both basal‐ and
intermediate‐layer keratinocytes in the early fetal epi­ on the surface of intermediate‐layer cells. Like the peri­
dermis is now identical to the expression of the keratins derm and basal cells, the first intermediate cells still con­
in the fully keratinized adult epidermis. The K5 and K14 tain glycogen as the primary cytoplasmic component
basal cell keratins are downregulated in the intermedi­ (Fig. 1.17a and b). Thus, at this stage, when the epidermis
ate cells and a new keratin pair, K1 (56.5 kDa) and K10 is only a few cell layers thick, and has few similarities in
(67 kDa), the high‐molecular‐weight differentiation‐ morphology to adult epidermis, it possesses all of the
specific keratins, is synthesized [1,2] (Fig.  1.18). Other keratins and many of the other markers that are typical
markers of keratinocyte differentiation (e.g. pemphigus of the epidermis throughout life. Therefore, genetic
antigen [3], cornified cell envelope proteins [4,5], blood ­diseases that involve mutations in keratin proteins have
group antigens [6] and cell‐surface glycoproteins [7]; the potential of being expressed as early as the first
reviewed in [8–11]) are also expressed in the cytoplasm or trimester in development.
12 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a)

(b)
Fig. 1.18  Keratin filaments in the cells of the intermediate layer of 77‐day
EGA fetal skin stain positively with the AE2 monoclonal antibody, which
recognizes the K1 and K10 differentiation‐specific keratins (a). The reaction
pattern is even stronger when a second intermediate layer is added at the
beginning of the second trimester (b) (a, ×120; b, ×120).
Fig. 1.20  Scanning electron micrograph of the periderm of a 60‐ to
70‐day EGA human fetus showing the blebs and microvilli that modify the
amniotic surface (×8000).

Fig. 1.19  Skin from a 72‐day EGA fetus immunoreacted with an anti‐epidermal
growth factor receptor showing a reaction pattern on the membranes of
basal and intermediate layer cells and on the basal and lateral borders of
periderm cells (×120).

Fig. 1.21  Section of skin from an 82‐day EGA fetus immunostained with
Initially, the keratinocytes of both basal and intermediate the HMB‐45 antibody, which recognizes melanocytes. Note the high density
cell layers express epidermal growth factor (EGF) recep­ of the cells and their position within the basal epidermal layer (×200).
tors [12] (Fig. 1.19), respond to EGF and retain the ability
to proliferate [13,14]. Near the end of the first trimester, ­ ifferentiation. There must be more processes involved,
d
however, the proliferative cells become restricted primar­ however, than simply the addition of cell layers because
ily to the basal layer [13,14]; only the basal cells express embryonic skin maintained in suspension organ culture
P‐cadherin [15], a marker of proliferative ability. Basal stratifies to become several layers thick but will not dif­
cells change in morphology and cell‐surface properties ferentiate in the manner characteristic of early fetal skin
after stratification. A greater volume of the cytoplasm is in vivo [18,19].
occupied by organelles and keratin filaments than with Melanocytes are easily recognized in sections of fetal
glycogen, and cell‐surface carbohydrates that correlate epidermis at 8 weeks EGA by their position along the
with stratification and differentiation are differentially basement membrane, dense cytoplasm, an absence of
expressed by cells of the basal and intermediate layers glycogen and a heterochromatic nucleus [20]. Around
[7,16]. Selected basal‐ and intermediate‐layer keratino­ 80 days EGA, they are present in the epidermis in maxi­
cytes participate in the formation of the epidermal mal density (∼3000 cells/mm2) [20] compared with all
appendages: the pilosebaceous structures, nails and teeth, other stages of skin development, and in a nonrandom
and the eccrine sweat glands in thick skin. The morpho­ distribution among cells of the basal layer (Fig. 1.21). The
genesis of these structures is described under Unique fea­ numbers decrease towards birth then continue to decline
tures of developing human skin. over the decades of postnatal life. The high numbers of
The cells of the periderm increase in size and develop melanocytes around the embryonic–fetal transition may
microvilli‐covered blebs that extend from the outermost reflect the fact that these cells arrive early in the skin, pro­
surface of the cell into the amniotic cavity (Fig. 1.20). The liferate and remain close together before there is substan­
molecular species of keratins remain the same as they tial growth of the fetus. The labelling index of keratinocytes
were in the embryonic periderm cells, but the cells lose is also high [13] at this stage, suggesting that the paracrine
their ability to divide and to express P‐cadherin [13,17]. interactions between melanocytes and keratinocytes that
Because the epidermal cells that are located in the more occur in the adult skin may be established early in devel­
superficial layers express differentiation‐related antigens, opment [20]. Melanosomes are recognized late in the
it is appealing to link stratification and the onset of third month of development (Fig.  1.22) and show some
Chapter 1  Embryogenesis of the Skin 13

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Fig. 1.24  Section of skin from the palm of an 83‐day EGA fetus
immunostained to recognize keratin 18 (green), an antibody that
recognizes Merkel cells within the basal layer. Note the regular distribution
of cells, presumably marking the sites of primary epidermal ridge location.
The skin of the hand is more advanced in development than that of the
trunk at an equivalent age (×300). Source: Immunostaining courtesy of
Fig. 1.22  Section through late embryonic–early fetal skin showing Dr Dong‐Kun Kim.
developing melanosomes in a melanocyte positioned between keratino-
cytes in the basal layer. The early age of the tissue is confirmed by the
immature structure of the DEJ (×25 000). however, by about 13 weeks EGA Langerhans cells seem
to express both markers with reasonable consistency.
By the end of the first trimester, Merkel cells are located
along the primary epidermal ridges of palmar skin in
regular alignment relative to the sites of origin of the
sweat duct primordia and at a maximum density of ∼1400
cells/mm2 [25,26] (Fig.  1.24). In hair‐bearing skin, they
first become evident in association with the developing
hair germs. At later stages in follicle development, they
concentrate in the infundibulum and bulge regions of the
hair pegs and bulbous hair pegs [27,28]. It is likely that the
dermal Merkel cells originate in the follicular or interfol­
licular epidermis and migrate into the dermis, where they
are suggested to play a role at early stages of develop­
ment in attracting and organizing nerve fibres in the
upper dermis and around developing appendages [29].
Merkel cells in the interfollicular epidermis lack NGF
Fig. 1.23  Epidermal sheet from an 80‐day EGA fetus reacted to demonstrate receptors (and produce NGF [30]), but dermal Merkel
ATPase. Note the regular distribution and density of these highly dendritic cells and Merkel cells of the developing follicle are immu­
cells (×120). nopositive when the tissue is reacted with the p75 NGF
antibody [29]. It must be recognized, however, that nerve
evidence of pigment formation in selected sites of the fibres are already apparent in the embryonic dermis
body. Understanding the density of melanocytes and the before dermal Merkel cells are detectable; thus, other fac­
onset of pigment synthesis had been previously useful in tors must also attract or direct nerves into the skin. Other
prenatal diagnosis of tyrosinase‐negative oculocutaneous morphological markers that are characteristic for Merkel
albinism, a technique no longer used [15,21]. cells in postnatal skin, such as dense core granules, are
Langerhans cells are also abundant in the epidermis at not apparent in dermal Merkel cells at this stage. Merkel
this stage (∼50/mm2) [22,23]. Unlike melanocytes, which cells decrease in number during the later stages of fetal
migrate into the epidermis only during the embryonic development [29].
period, the bone marrow‐derived Langerhans cells The DEJ has acquired all of the adult features that are
migrate into the epidermis continually throughout life; characteristic for this region (Fig. 1.25). Hemidesmosomes,
their numbers do not increase significantly, however, until anchoring filaments and anchoring fibrils are structurally
the third trimester and after birth [22,23]. Langerhans cells complete, and the antigens related to these attachment
at 80 days EGA are highly dendritic (Fig.  1.23), begin to structures, the skin‐specific markers of the DEJ, are also
express CD1a at the surface [22–24] and develop Birbeck expressed [31–33]. Immunostaining with an antibody to
granules in the cytoplasm, suggesting that they may be type VII collagen outlines the DEJ with high intensity [32]
capable of processing and presenting antigen in utero. The and stains basal cell cytoplasm (the primary source of this
number of cells that are HLA‐DR positive is ­significantly protein) with low intensity. Nonetheless, the structural
greater than the number that express CD1a at this stage; organization of the DEJ appears delicate in contrast to the
14 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a)
Fig. 1.25  Transmission electron micrograph of the DEJ of a 78‐day EGA
human fetus. Note the well‐formed hemidesmosomes and associated
keratin filaments, anchoring filaments within the lamina lucida and fine
anchoring fibrils (×47 500).

(b)
Fig. 1.27  Light micrograph of 78‐day EGA human skin showing the
vascular pattern organized in a series of horizontal plexuses and vertical
connecting vessels (a, b). Note that the diameters of the vessels become
increasingly smaller towards the epidermal surface. In the higher‐magnifi-
cation image, the rounder cells of the papillary dermis are distinct from the
more elongated fibroblastic cells of the reticular dermis and subcutaneous
region (a, ×25; b, ×100). Source: Micrographs courtesy of Dr Greg Hébert.

subcutaneous mesenchymal cells still retain glycogen in


the cytoplasm, but they have assumed a distinctly fibro­
blastic morphology and are responsible for the synthesis
of all of the matrix molecules that are characteristic of
adult dermis. There is accumulation of small bundles of
fibrous proteins within the interstitial space and papillary
and reticular regions of the dermis are demarcated on the
Fig. 1.26  Light micrograph of 72‐day EGA fetal dermis stained with the
basis of increased cell density proximal to the epidermis
Alcian blue/periodic acid–Schiff (PAS) histochemical stain. The low‐
magnification image shows the clear demarcation between the dermal
(the papillary region) and larger collagen fibril diameter
and subcutaneous tissue and the different concentrations of fibrous and and fibre bundle size in the reticular region [9,34–36]
glycosaminoglycan matrix proteins in the two regions. A vascular plane (Fig.  1.27b). The position of the subpapillary vascular
also demarcates the two regions. Skeletal muscle is evident in the lower plexus of arterioles and postcapillary venules also forms
right corner of the micrograph (×200). Source: Histochemical staining an approximate boundary between these two dermal
courtesy of Dr Richard Frederickson. zones. In spite of the significant accumulation of matrix
protein, the dermis remains highly cellular, with the
robust structure of the basal lamina and anchoring fibrils matrix accounting for substantially less of the bulk of the
in adult skin. The dermoepidermal interface is still flat skin than it does in the postnatal infant and the adult.
although modifications of individual basal cells begin to The skin is still transparent enough to permit the net­
alter the smoothness of this junction. works of vessels and nerves to be seen through the body
The dermis and subcutaneous tissue are distinguished wall of the fetus (Fig. 1.28). The vessels are organized in
on a morphological basis by differences in the organiza­ the dominant pattern of adult skin, with one plexus
tion and composition of the matrix (Fig. 1.26). Dermal and located at the dermosubcutaneous boundary and another
Chapter 1  Embryogenesis of the Skin 15

lumina of some of these structures suggest that they may

STRUCTURE AND PHYSIOLOGY


belong to the venous side of the vasculature although the

SECTION 1: DEVELOPMENT,
simplicity of the wall structure would suggest they could
be lymphatics.

­References
1 Dale BA, Holbrook KA, Kimball JR et al. Expression of the epidermal
keratins and filaggrin during fetal human development. J Cell Biol
1985;101:1257–69.
2 Moll R, Moll I, Wiest W. Changes in the pattern of cytokeratin poly­
peptides in epidermis and hair follicles during skin development in
human fetuses. Differentiation 1983;23:170–8.
3 Lane AT, Helm HF, Goldsmith LA. Identification of bullous pemphig­
oid, pemphigus, laminin and anchoring fibril antigens in human fetal
skin. J Invest Dermatol 1985;84:27–30.
4 Holbrook KA, Underwood RA, Dale BA et al. Formation of the corni­
Fig. 1.28  Whole‐mount sample of unfixed 74‐day EGA fetal skin showing fied cell envelope in human fetal skin: presence of involucrin, kerato­
the vascular network of the skin through the translucent tissue of the body linin, loricrin and transglutaminase correlated with the onset of
wall (×63). Source: Micrograph courtesy of Dr Carole Johnson. transglutaminase activity. J Invest Dermatol 1991;96:542A.
5 Akiyama M, Smith LT, Yoneda K et al. Expression of transglutaminase
1 (TG1) and cornified cell envelope (CCE) proteins during human epi­
dermal development. J Invest Dermatol 1997;108:598.
6 Dabelsteen E, Holbrook KA, Clausen H et  al. Cell surface carbohy­
drate changes during embryonic and fetal skin development. J Invest
Dermatol 1986;87:81–5.
7 Watt FM, Keeble S, Fisher C et al. Onset of expression of peanut lectin
binding glycoproteins is correlated with stratification of keratinocytes
during human epidermal development in vivo and in vitro. J Cell Sci
1989;94:355–9.
8 Holbrook KA. Structure and function of the developing human skin. In:
Goldsmith LA (ed.) Physiology, Biochemistry and Molecular Biology of
the Skin, 2nd edn. Oxford: Oxford University Press, 1991:63–110.
9 Holbrook KA. Structural and biochemical organogenesis of skin and
cutaneous appendages in the fetus and neonate. In: Polin RA,
Fox WW (eds) Neonatal and Fetal Medicine Physiology and
Pathophysiology. New York: Grune & Stratton, 1992:527–51.
10 Holbrook KA, Wolff K. The structure and development of skin. In:
Fitzpatrick TB, Eisen AZ, Wolff K et al. (eds) Dermatology in General
Medicine, 6th edn. New York: McGraw‐Hill, 1993:97–144.
11 Holbrook KA, Sybert VP. Basic science. In: Schachner L, Hansen R
(eds) Pediatric Dermatology, 2nd edn. New York: Churchill
Livingstone, 1995.
Fig. 1.29  Section of skin from a 77‐day EGA fetus immunostained with
12 Nanney LB, Stoscheck CM, King LE et  al. Immunolocalization of
the PGP 9.5 antibody, which recognizes all cutaneous nerves. Note the
­epidermal growth factor receptors in normal developing human skin.
larger trunks deep in the dermis and the fine network of fibres that J Invest Dermatol 1990;94:742–8.
supplies the epidermis (×100). Source: Immunostaining courtesy of 13 Bickenbach JR, Holbrook KA. Label retaining cells (LRCs) in human
Dr Dong‐Kun Kim. embryonic and fetal epidermis. J Invest Dermatol 1986;88:42–6.
14 Bickenbach JR, Holbrook KA. Proliferation of human embryonic and
fetal epidermal cells in organ culture. Am J Anat 1986;177:97–106.
15 Gershoni‐Baruch R, Benderly A, Brandes JM et al. Dopa reaction test
at the boundary between the papillary and reticular der­ in hair bulbs of fetuses and its application to the prenatal diagnosis of
mis (Fig. 1.27a). Vertically orientated vessels connect the albinism. J Am Acad Dermatol 1991;24:220–2.
16 Fisher C, Holbrook KA. Cell surface and cytoskeletal changes associ­
two horizontal plexuses, and fine capillaries extend into ated with epidermal stratification in organ cultures of embryonic
the papillary dermis [8–11]. Nerves are also readily appar­ human skin. Dev Biol 1987;119:231–41.
ent in both sections and whole‐mount preparations of 17 Piepkorn M, Underwood RA, Henneman C et  al. Expression of

skin that are immunostained with the p75 antibody to amphiregulin is regulated in cultured human keratinocytes and in
developing fetal skin. J Invest Dermatol 1996;105:802–9.
NGFR [37], neurofilament protein, PGP 9.5 (Fig.  1.29), 18 Holbrook KA, Minami SA. Hair follicle morphogenesis in the human:
CGRP and neuropeptide Y (NPY). NPY recognition of characterization of events in vivo and in vitro. NY Acad Sci
certain fibres associated with blood vessels signifies the 1991;642:167–96.
19 Holbrook KA, Smith LT, Kaplan ED et al. The expression of morpho­
presence of autonomic fibres [38]. Like the vessels, large gens during human follicle development in vivo and a model for
subcutaneous nerve trunks branch to finer and finer fibres studying follicle morphogenesis in vitro. J Invest Dermatol
that terminate beneath the DEJ. The nerve and vascular 1993;101:39S–49S.
networks are at times parallel but are also separate from 20 Holbrook KA, Underwood RA, Vogel AM et al. The appearance, den­
sity and distribution of melanocytes in human embryonic and fetal
one another (see Fig. 1.15). skin revealed by the anti‐melanoma monoclonal antibody, HMB‐45.
The hypodermis appears to have markedly fewer cells Anat Embryol 1989;180:443–55.
than either region of the dermis, and smaller bundles of 21 Eady RAJ, Gunner DB, Garner A et al. Prenatal diagnosis of oculocu­
taneous albinism by electron microscopy. J Invest Dermatol
fibrous matrix. Dilated channels course through the hypo­
1983;80:210–12.
dermis, distinguishing it from deeper tissue of the body 22 Drijkoningen M, DeWolf‐Peeters C, VanDerSteen K et al. Epidermal
wall (see Figs 1.26 and 1.27). Red blood cells within the Langerhans cells and dermal dendritic cells in human fetal and
16 Section 1  Development, Structure and Physiology of the Skin

neonatal skin: an immunohistochemical study. Pediatr Dermatol All of these features will be initiated and/or fully acquired
1987;4:11–17.
STRUCTURE AND PHYSIOLOGY

during the second trimester, and changes will continue to


23 Foster CA, Holbrook KA. Ontogeny of Langerhans cells in human
SECTION 1: DEVELOPMENT,

embryonic and fetal skin: cell densities and phenotypic expression occur in the structures and regions of the skin that have
relative to epidermal growth. Am J Anat 1989;84:157–64. been established but not finalized.
24 Foster CA, Holbrook KA, Farr AG. Ontogeny of Langerhans cells in
human embryonic and fetal skin: expression of HLA‐DR and OKT‐6
determinants. J Invest Dermatol 1986;86:240–3.
Second‐trimester fetal skin
25 Moll R, Moll I, Franke W. Identification of Merkel cells in human skin At 12 weeks’ gestation, the fetus measures about 85 mm in
by specific cytokeratin antibodies: changes in cell density and distri­ length (crown–rump) and has a body form similar to that
bution in fetal and adult plantar epidermis. Differentiation 1984; of the newborn. The skin and body wall are opaque. All of
28:136–54.
26 Moll R, Moll I. Early development of human Merkel cells. Exp
the events in skin morphogenesis initiated during the first
Dermatol 1992;1:180–4. trimester continue in the second trimester in parallel with
27 Kim D‐G, Holbrook KA. The appearance, density and distribution of the onset of formation of those structures that were iden­
Merkel cells in human embryonic and fetal skin: their relation to
tified as lacking in first‐trimester skin. Landmark events
sweat gland and hair follicle development. J Invest Dermatol
1995;104:411–16. in the second trimester include completion of the forma­
28 Narisawa Y, Hashimoto K, Nakamura Y et al. A high concentration of tion of the lanugo hair follicle and synthesis of the hair
Merkel cells in the bulge prior to the attachment of the arrector pili (around 17–19 weeks EGA), completion of the formation
muscle and the formation of the perifollicular nerve plexus in human
fetal skin. Arch Dermatol Res 1993;285:261–8.
of the nail (around 20–22 weeks EGA) and keratinization
29 Narisawa Y, Hashimoto K, Pietruk T. Biological significance of der- of the interfollicular epidermis (around 22–24 weeks
mal Merkel cells in development of cutaneous nerves in human EGA). The timing is variable because the formation of the
fetal skin. J Histochem Cytochem 1992;40:65–71. hair follicle and epidermal keratinization are regionally
30 Vos P, Stark F, Pittman RN. Merkel cells in vitro: production of nerve
growth factor and selective interactions with sensory neurons. Dev dependent. Sweat glands on the general body surface
Biol 1991;144:281–300. only begin to form around 17–18 weeks EGA (see Unique
31 Fine JD, Smith LT, Holbrook KA et al. The appearance of four base­ features of developing human skin). The 24‐week‐old
ment membrane zone antigens in developing human fetal skin.
J Invest Dermatol 1984;83:66–9.
fetus is fully formed and has hair on the scalp and body
32 Smith LT, Sakai LY, Burgeson RE et al. Ontogeny of structural compo­ surface. The length is about 228 mm (crown–rump).
nents at the dermal–epidermal junction in human embryonic and One or two additional intermediate cell layers are
fetal skin: the appearance of anchoring fibrils and type VII collagen. added to the epidermis by proliferation of basal keratino­
J Invest Dermatol 1988;90:480–5.
33 Hertle MD, Adams JC, Watt FM. Integrin expression during human cytes and upward migration of the first intermediate cells.
epidermal development in vivo and in vitro. Development 1991; By 100–110 days EGA, there are typically three supraba­
112:193–206. sal, intermediate cell layers, which become progressively
34 Smith LT, Holbrook KA, Byers PH. Structure of the dermal matrix
more flattened towards the epidermal surface (Fig. 1.30).
during development and in the adult. J Invest Dermatol 1982;
79:93S–104S. The cells of the most superficial layer have large bundles
35 Smith LT, Holbrook KA, Madri JA. Collagens types I, III and V in of keratin filaments, which can be seen in stained specimens
human embryonic and fetal skin. Am J Anat 1986;175:507–22. at the light microscopic level as a reticulate cytoskeleton
36 Smith LT, Holbrook KA. Embryogenesis of the dermis. Pediatr

Dermatol 1986;3:271–80.
(Fig.  1.30). Glycogen is still a major constituent of the
37 Holbrook KA, Bothwell MA, Schatteman G et al. Nerve growth factor cytoplasm.
receptor labelling defines developing nerve networks and stains As the epidermis thickens, the interface it forms with
­follicle connective tissue cells in human embryonic and fetal skin. the dermis becomes less flattened and smooth, due largely
J Invest Dermatol 1988;90:609A.
38 Terenghi G, Sundaresan M, Moscoso G et  al. Neuropeptides and a to changes in the basal surface of each keratinocyte rather
neuronal marker in cutaneous innervation during human foetal than to convolutions of the layer itself. Basal cells stain
development. J Comp Neurol 1993;328:595–603. with less intensity than the intermediate‐layer cells
because there is less glycogen, the bundles of keratin
­filaments are smaller and the cytoplasm is more ribosome
­Fetal skin
rich, dense and organelle filled (Fig. 1.30). At the end of
Conclusion of the first trimester the second trimester, the five‐layered interfollicular
The first stages of fetal skin development occur from the ­epidermis keratinizes. Skin of the trunk shows signs of
time of the embryonic–fetal transition at 2 months to the keratinization around 21 weeks EGA in the uppermost
end of the first trimester at 3 months, when a template of intermediate cell layers and the overlying periderm
the adult skin is established. There are notable features of (Fig. 1.31).
adult skin that are still lacking. The epidermis has yet to Changes in the structure and composition of the plasma
keratinize and one of the key proteins of keratinization, membrane mark the formation of a cornified cell enve­
filaggrin, is not yet expressed in any region of the skin. lope [1,2], and lamellar granules are identified in the cyto­
The dermoepidermal interface lacks rete ridges and rete plasm (Fig. 1.31) and in the spaces between intermediate
pegs. The dermis lacks fully formed elastic fibres and an and periderm layers. The modified intermediate cells
elastic fibre network. Ectodermal appendages are just remain associated with the overlying periderm cells by
starting formation in limited areas; for example, sweat infrequent and tenuous‐appearing desmosomal attach­
gland development is initiated only on the palms and ments. Cells are also evident in which the nucleus is pyk­
soles and apocrine glands have not begun to develop. notic and the cytoplasm contains dense bundles of
Hair and nails are not synthesized. Adipose tissue has not filaments, vacuoles and other remnants of the cytoplasm.
differentiated within the mesenchyme of the hypodermis. Cells beneath these seemingly incompletely keratinized
Chapter 1  Embryogenesis of the Skin 17

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
(a) (b)
Fig. 1.30  Light (a) and electron (b) micrographs of a section of skin from a 104‐day EGA human fetus showing additional intermediate cell layers, changes
in periderm morphology, the reticulate keratin cytoskeleton in upper intermediate cells, less intense staining of basal cells (a, b) and the smaller bundles of
collagen fibrils in the papillary dermis (a). A melanocyte is evident (a, arrow). Several capillaries (a) and nerves (b, n) lie within the papillary region (a, ×300;
b, ×3500).

squames contain very small, stellate keratohyalin gran­ although the adult level of approximately 650 cells/mm2,
ules and react immunopositively with an antibody that or about 8500 cells/mm3, is not achieved until after birth
recognizes profilaggrin and filaggrin proteins of the gran­ [7]. Melanosomes are transferred to keratinocytes in the
ule [3] (Fig. 1.31). The two or three subjacent intermediate fifth month of gestation.
cell layers can now be called spinous cells. At this stage, All of the structures of the DEJ were formed in the first
periderm cells are very large in diameter, flattened and trimester, and only a few antigens of the DEJ (AF‐1 and
also display a thickened cell envelope. Each cell covers a AF‐2 associated with anchoring fibrils) remain to be rec­
cluster of underlying epidermal cells. The periderm stains ognized at this age [8]. By 19–21 weeks EGA, the hemides­
differently from the underlying epidermal cells, probably mosomes are present at the basal keratinocyte plasma
owing to a lesser amount of structural protein within the membrane with adult‐like frequency and show a strong
cytoplasm. association with basal cell keratin filaments. Anchoring
A few layers of thin, flattened, keratinized cells, organ­ filaments and banded anchoring fibrils are well formed
ized in the manner of a true stratum corneum, are first (reviewed in [9–12]).
apparent around 22–24 weeks EGA (Fig. 1.32). The granu­ Small bundles of interwoven, fibrous connective tissue
lar cell layer is now more typical of an adult granular occupy the interstitial space within the dermis (Fig. 1.33a),
layer in that keratohyalin granules are larger and the although they remain loosely organized because the sul­
cytoplasm contains less glycogen. The numbers of layers phated proteoglycans and fibrous proteins of the intersti­
of the stratum corneum continue to increase in the third tial matrix are still very hydrated. Elastin is detectable
trimester to reach a more mature appearance by 34 weeks biochemically, and elastic fibres can be recognized as
gestation [4]. Of note, premature birth hastens develop­ granular‐appearing structures along the borders of
ment of mature stratum corneum and epidermal thick­ ­collagen fibre bundles in immunostained samples of skin
ness with histologically similar appearance to term infants and by electron microscopy. The structure of the elastic
seen within 2–3 weeks after birth regardless of gestational fibres, however, even in the deepest portions of the reticu­
age [4]. lar dermis, is similar to that of the elaunin fibres of adult
By 22–24 weeks EGA, 1700 Merkel cells/mm2 can be skin, which have only sparse amounts of elastin associ­
measured in the epidermis [5,6] and the number of ated with the microfibrillar bundles. The extent to which
Langerhans cells begins to increase (∼200 cells/mm2), elastic fibres are developed is dependent upon the region
18 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a)

Fig. 1.32  Electron micrograph of the keratinized epidermis from a fetus at


the end of the second trimester. There are a few layers of cornified cells, a
single granular cell layer and three layers of spinous cells, which retain a
significant quantity of glycogen. Note the greater irregularity in the basal
border of the basal cells at the DEJ (×3000).

Third‐trimester fetal skin


The skin in the third trimester appears structurally similar
to postnatal skin (reviewed in [9–13]) (Fig.  1.34). The
­epidermis is fully keratinized, contours are beginning to
(b) form at the DEJ, the regions of the dermis are well defined,
Fig. 1.31  Electron micrographs of the skin from two 21‐week EGA fetuses
the adnexa are fully formed and reside deeply in the
showing early (a) and late (b) changes in the upper intermediate layers ­dermis, and large fat lobules fill the hypodermis. There
(spinous) at the onset of keratinization. Note the regressed periderm are, however, some notable differences in structure in all
separating from the upper epidermal layers (a), lamellar granules in the top regions: suprabasal epidermal cells retain a significant
few layers (arrows), the small particulate (a) then stellate (b) keratohyalin amount of glycogen in the cytoplasm and the dermis
granules, and the few layers of incompletely keratinized cells (black remains relatively thin. The bundles of collagen matrix
material) to demonstrate the permeability of the epidermis to tracers
are small elastic fibres and are immature in structure and
(a, ×12 150; b, ×9500). Source: Micrograph (b) courtesy of Dr Richard
Frederickson.
composition, and the stratum corneum has fewer cell
­layers than infant or adult skin.
Studies of the function of skin of the premature infant
provide some understanding of the status of the skin dur­
of the skin. In addition to fibroblasts, mast cells, mac­ ing the third trimester. In general, various functions of the
rophages and smooth muscle cells are present in the skin, such as barrier properties, temperature regulation,
­dermis [9–12]. sweating, response to tactile and mechanical stimuli [14],
The hypodermis remains distinct from the dermis by its that have been measured reflect the gestational age more
less dense matrix and cellularity. Around 15–16 weeks than the birthweight.
EGA, mesenchymal cells collect in globular arrays sur­ The epidermis, even though keratinized and possessing
rounded by a capsule‐like assembly of matrix (Fig. 1.33b). several layers of stratum corneum cells, is a less effective
This is the first stage of adipose tissue formation. Small barrier than the infant epidermis. Transepidermal water
vessels are present within these cellular aggregations. By loss, for example, decreases in a steep slope from 26 weeks
18 weeks EGA, lipid droplets are evident within some of EGA to 38 weeks EGA. It decreases even further and with
the mesenchymal cells, and by 20 weeks lobules of fat are a similarly rapid decline over the first 10–15 postnatal
established. days (reviewed in [15,16]). Disorders of keratinization or
Chapter 1  Embryogenesis of the Skin 19

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
(a)

Fig. 1.34  A section through skin obtained from a third‐trimester (34‐week)


human fetus showing all regions of the skin and the presence of hair
follicles and eccrine sweat glands (×40).

(reviewed in [15,16]). These compromised epidermal


­ arrier properties, coupled with the fact that the preterm/
b
neonatal infant’s body surface‐to‐volume ratio is very
high, can place the premature infant at significant risk.
Although the structure and cellular differentiation of
the sweat glands in the preterm infant and term newborn
(see Unique features of developing human skin) appear
little, if any, different from those of the infant, the sweat­
ing function requires a period of maturation after birth,
presumably for the innervation to become fully estab­
lished. The sweating response is limited or absent in the
preterm infant to an extent that correlates with the gesta­
tional age (reviewed in [13]). Apocrine glands begin to
secrete during this trimester [9–12].
Some aspects of the vasculature are less organized in
the third‐trimester fetus and the newborn than is charac­
(b)
teristic in the infant. The marked redness of the newborn
Fig. 1.33  Scanning electron (a) and light (b) micrographs of the human reflects the high density of superficial vessels in the der­
fetal dermis at 15 weeks EGA showing the density of the fibrous matrix,
mis and the thinness of the epidermis. Remodelling of the
the differences in textures between papillary and reticular regions (a), and
the boundary between the dermis and subcutaneous regions (a, b). The
microcirculation occurs as appendages and regions of the
developing hair follicles are evident in cross‐section in (a) and in longitudinal skin are completed. At birth, the capillary network is still
section in (b). A lobule of subcutaneous fat (arrows) is evident in (b) disorganized and will stabilize only after birth (reviewed
(a, ×80; b, ×100). in [9–12]).

­References
infection of the skin may give an even greater disadvan­ 1 Holbrook KA, Underwood RA, Dale BA et  al. Formation of the
tage to the premature newborn in its ability to regulate cornified cell envelope in human fetal skin: presence of involucrin,
substances crossing the skin. The stratum corneum of the keratolinin, loricrin and transglutaminase correlated with the onset
of transglutaminase activity. J Invest Dermatol 1991;96:542A.
preterm infant is more permissive to absorption of sub­ 2 Akiyama M, Smith LT, Yoneda K et al. Expression of transglutaminase
stances from the external environment and those applied 1 (TG1) and cornified cell envelope (CCE) proteins during human epi­
to skin to protect, treat or cleanse it in the neonatal nursery dermal development. J Invest Dermatol 1997;108:598 (Abstract).
20 Section 1  Development, Structure and Physiology of the Skin

3 Dale BA, Holbrook KA, Kimball JR et al. Expression of the epidermal The most remarkable features of the periderm are the
keratins and filaggrin during fetal human development. J Cell Biol
STRUCTURE AND PHYSIOLOGY

morphological changes that the periderm cells undergo


1985;101:1257–69.
SECTION 1: DEVELOPMENT,

4 Evans NJ, Rutter N. Development of the epidermis in the newborn. with progressive stages of development [8]. Studies of the
Biol Neonate 1986;49:74–80. surface of the developing skin using scanning electron
5 Moll R, Moll I, Franke W. Identification of Merkel cells in human skin microscopy, and of corresponding tissue sections exam­
by specific cytokeratin antibodies: changes in cell density and distri­
bution in fetal and adult plantar epidermis. Differentiation
ined by light and transmission electron microscopy from
1984;28:136–54. consistent regions of the body, have established the stages
6 Kim D‐G, Holbrook KA. The appearance, density and distribution of of human skin development [8] (Fig. 1.35).
Merkel cells in human embryonic and fetal skin: their relation to The early embryo is covered by a thin, flattened pave­
sweat gland and hair follicle development. J Invest Dermatol
1995;104:411–16. ment epithelium that is the periderm (Fig. 1.4). Around
7 Foster CA, Holbrook KA. Ontogeny of Langerhans cells in human 8–11 weeks, when the epidermis stratifies, the periderm
embryonic and fetal skin: cell densities and phenotypic expression cells increase in volume and develop a rounded external
relative to epidermal growth. Am J Anat 1989;84:157–64.
8 Lane AT, Helm HF, Goldsmith LA. Identification of bullous pemphig­
surface. By 10–14 weeks, single blebs extend from the
oid, pemphigus, laminin and anchoring fibril antigens in human fetal amniotic surface of each cell and the cell increases in
skin. J Invest Dermatol 1985;84:27–30. diameter (see Fig. 1.20). All of the cell surface, including
9 Holbrook KA. Structure and function of the developing human skin. the blebs, is modified by microvilli. A network of micro­
In: Goldsmith LA (ed.) Physiology, Biochemistry and Molecular
Biology of the Skin, 2nd edn. Oxford: Oxford University Press, filaments is organized beneath the plasma membrane.
1991:63–110. Later in the second trimester, the surfaces of the peri­
10 Holbrook KA. Structural and biochemical organogenesis of skin
derm cells project multiple blebs, the larger of which
and  cutaneous appendages in the fetus and neonate. In: Polin RA,
have the configuration of a blackberry (Figs  1.30 and
Fox  WW (eds) Neonatal and Fetal Medicine Physiology and
Pathophysiology. New York: Grune & Stratton, 1992:527–51. 1.36). The cell diameter continues to increase as the cells
11 Holbrook KA, Wolff K. The structure and development of skin. In: become thinly stretched over the epidermis. By 16–23
Fitzpatrick TB, Eisen AZ, Wolff K et al. (eds) Dermatology in General weeks EGA, the blebs flatten and the periderm regresses
Medicine, 6th edn. New York: McGraw‐Hill, 1993:97–144.
12 Holbrook KA, Sybert VP. Basic science. In: Schachner L, Hansen R
(Fig.  1.37). It once again becomes a very thin layer of
(eds) Pediatric Dermatology, 2nd edn. New York: Churchill cells, which, at this stage, rarely contain a nucleus, have
Livingstone, 1995. few if any organelles and are composed largely of disor­
13 Holbrook KA. A histologic comparison of infant and adult skin. In: ganized, fine filaments [8].
Boisits E, Maibach HI (eds) Neonatal Skin: Structure and Function.
New York: Marcel Dekker, 1982:3–31. The periderm cells do not undergo the events of dif­
14 Andrews K, Fitzgerald M. The cutaneous withdrawal reflex in human ferentiation that are typical for the keratinocyte. The
neonates: sensitization, receptive fields, and the effects of contralat­ ­composition of keratins in the periderm cells remains
eral stimulation. Pain 1994;56:95–101.
15 Cartlidge PAT, Rutter N. Skin barrier function. In: Polin RA, Fox WW
unchanged throughout development and, since neither
(eds) Fetal and Neonatal Physiology, Vol. 1. Philadelphia: W.B. the K1/K10 keratin proteins nor profilaggrin are present
Saunders, 1992:3133–42. in the periderm cells of any stage, it is clear that they do
16 Chiou YB, Blume‐Peytavi U. Stratum corneum maturation. A review not undergo full keratinization. The plasma membrane of
of neonatal skin function. Skin Pharmacol Physiol 2004;17:57–66.
the early second‐trimester fetal periderm cell, however,
appears similar to a cornified envelope (Fig.  1.38a) and
the presence of several cornified cell envelope proteins,
­ nique features of developing human
U
involucrin, loricrin, keratolinin (cystatin), small proline‐
skin
rich proteins (SPRR) 1 and 2, and the transglutaminase 1
Periderm (TG1) enzyme, in the cytoplasm [9,10] of these cells has
The periderm is the outermost, transient cellular layer of been demonstrated (Fig.  1.38b–d). The expression of
the developing skin of some mammals and birds. These cornified envelope proteins in conjunction with ultras­
embryonic epidermal cells are larger than basal keratino­ tructural studies suggests that periderm cells do contrib­
cytes and cover the entire surface of the early epidermis. ute to the cornified envelope.
The origin of the periderm is not fully elucidated in humans Towards the end of the second trimester, individual
but mouse studies may give some clue to its development. periderm cells loosen from the underlying epidermal cells
It is possible that the amnion contributes cells to periderm and are desquamated over the sites of elevated and
that grow over the single‐layered epidermis. This is sup­ exposed hair canals (follicular epidermis). They remain
ported by the fact that the amnion and periderm are similar associated, however, with the interfollicular epidermis
in keratin composition [1,2] and surface morphology and until the stratum corneum is formed. At this time, the
in the expression of other antigens [3,4]. Studies from early periderm is mostly gone from the skin surface. The events
mouse embryos, however, suggest that a continuous sheet that lead to disengagement of this layer are not known.
of tissue may not cover the epidermis as proposed by this Abnormality of periderm shedding is hypothesized to
model, because patches of periderm cells are present in contribute to collodion formation.
some sites [5]. The single, ectodermal layer of the early The structural properties of periderm cells may provide
embryo may divide and give rise to a second cell layer that clues as to the function of the layer. The blebs and micro­
becomes superficial to the basal layer [6]. This is supported villi increase the surface area of the periderm as it faces
by whole‐mount studies showing expression of basal‐layer the amniotic fluid, suggesting that these cells may be
keratinocytes K5 and K14 in both the single‐layer ectoderm important in the exchange of substances between the
and the periderm [7]. fetus and the amniotic fluid, across the skin, in one or
Chapter 1  Embryogenesis of the Skin 21

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
<36 days

35–55 days

95–120 days

55–75 days

110–160 days

65–95 days

Fig. 1.35  Stages of epidermal development


proposed on the basis of periderm structure. 85–110 days >160 days

Fig. 1.36  Scanning electron micrograph of the periderm from a mid‐second‐ Fig. 1.37  Scanning electron micrograph of the surface of the flexor
trimester fetus showing multiple, complex blebs and microvilli extending forearm of a late‐second‐trimester fetus showing the large, thin, regressed
from the amniotic surface (×1500). periderm cells (×800).

both directions. Direct evidence for this role in humans is fetus, the periderm has been shown to be involved in the
limited. A morphological study of the intramembranous uptake of drugs from the amniotic fluid [13]. It has also
modifications of the periderm plasma membrane sug­ been postulated that the periderm is a secretory epithe­
gests that the cells have a role in regulating water trans­ lium that adds material to the amniotic fluid [14] and that
port [11], and Koren [12] has suggested that the skin it serves as a protective layer for the developing epidermis
absorbs nicotine dissolved in amniotic fluid; in the sheep (also reviewed in [15–17]). In mouse models of aberrant
22 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a) (b)

(c)

(d)
Fig. 1.38  Transmission image of the periderm cornified cell envelope from fetal skin of 21 weeks EGA (a). The periderm expresses involucrin early in the
first trimester. (b) Data from a 98‐day EGA fetus. Epidermal transglutaminase (c) is also expressed early in the periderm but does not appear to function
until the end of the first trimester, when it first demonstrates cross‐linking of dansyl cadaverine to substrate in the tissue (d) (a, ×41 250; b, ×300; c, ×300;
d, ×300).

or absent periderm development, pathological adhesions periderm cells. Keratin filaments are aggregated in the
developed at sites of apposed tissues [18]. This is likely second‐trimester skin of fetuses affected with epidermo­
the mechanism behind popliteal pterygium syndrome lytic hyperkeratosis (EHK) [20] and epidermolysis bullosa
and cocoon or fetal encasement syndrome. simplex Dowling–Meara (EBS‐DM) [21], but none of the
The periderm is regionally variable in its properties filaments clump in periderm cells, nor are there other
and timing of development [2]. Periderm of the plantar consequences for the periderm layer. The absence of
surface of the toe, for example, shows a very late stage of clumping would be expected in the case of EHK since the
development at 70 days compared with trunk skin. At keratins involved in this disorder are not keratins that are
this time, the epidermis of the appendage is thicker and present in periderm cells, but basal cells and periderm
more differentiated than trunk skin. This suggests that the cells do share keratins in common. The persistence of
nature of the underlying epidermis determines the rate ­periderm cells with no adverse outcome in an environ­
of modification of the periderm. This is supported by ment of severe cell destruction in layers proximal to the
similarities in keratin patterning between the basal periderm in EHK is surprising and argues for autonomy
keratinocytes and the periderm cells [7]. The pattern of of this layer. To the contrary, however, animal models of
development of the mature stratum corneum follows ­disorders where mature epidermal barrier function is not
periderm disaggregation, suggesting a functional achieved (e.g. ichthyosis) show incomplete shedding of
relationship between these two processes which coin­ the periderm suggesting a complex interplay that is not
cides with the limits of fetal viability between 22 and yet understood fully [22].
26 weeks EGA [19].
The periderm is thus a distinct layer of cells that is defined Regionalization in developing skin
early in development from the remainder of the epidermis. Regional differences in properties of the skin are well
Many genetic disorders, for example, that modify cells of documented in adult skin. Regionalization is also a
the basal and intermediate layers during development ­phenomenon of developing skin even at very early ages
appear to have no direct or indirect consequences for of gestation. Few systematic studies have been done to
Chapter 1  Embryogenesis of the Skin 23

document these regional differences consistently through­

STRUCTURE AND PHYSIOLOGY


out development [8,23]. Without a clear appreciation and

SECTION 1: DEVELOPMENT,
accurate knowledge of differences in normal morphology
at various sites, structural evaluation of skin samples that
may be from unknown regions can be difficult. At the
same time, it is essential to appreciate whether the disease
of concern is also expressed with regional variation not
only in the adult but also at the onset of expression of the
disease during development [24,25]. In at least one situa­
tion, the understanding of regional variability of expres­
sion of a disease at its first presentation was gained from
samples obtained for prenatal diagnosis [26] (Fig.  1.39).
Systematic studies of affected fetal skin from multiple
regions are valuable to undertake when tissue becomes
available. Such efforts also expand our knowledge of the (a)
natural history of the disorder.

Keratinization
Keratinization of the nails, hair follicles (follicular epider­
mis), intraepidermal sweat ducts and the interfollicular
epidermis occurs at different times during gestation. The
nails are the earliest structures of the skin to keratinize
in utero, with the appearance of cornified cells as early as
11–12 weeks EGA. The timing of keratinization of follicu­
lar epidermis is consistent with the cephalocaudal direc­
tion of follicle morphogenesis [25]; the interfollicular
epidermis keratinizes first in thick skin and then in thin
skin, with the latter also proceeding in a regionally
dependent manner. The timing of keratinization for a
given region appears to follow a rigidly specified pro­
gramme during development. The molecular mediators
of keratinization are beginning to be elucidated and in (b)
many instances disruption of these pathways causes Fig. 1.39  Sections of skin obtained in utero by fetal skin biopsy from a
­diseases of abnormal keratinization [26]. Even in these fetus at risk of lamellar ichthyosis. Note the difference in morphology
situations of abnormal keratinization, for example in between the two samples. One sample shows an epidermis of normal
fetuses affected with lamellar ichthyosis, harlequin ich­ thickness and state of development for 19 weeks EGA (a). The second
thyosis and EHK, there is no evidence for early or delayed sample shows a thickened epidermis, still covered by periderm (b). In both
samples, hair canals were excessively keratinized. This disorder is expressed
onset of the keratinization process in either the follicular
in utero with regional variation (a, ×300; b, ×300).
or interfollicular epidermis (reviewed in [27–30]).

Appendage formation After the embryo–fetal transition, around 10–11


The embryonic epidermis gives rise to diverse structures weeks EGA, basal epidermal cells, through various
collectively known as the ectodermal appendages. molecular events, undergo proliferation at specifically
Despite their diverse structure and function in the mature patterned sites to form buds that grow down into the
skin, the hair follicles, nails, sweat glands, mammary dermis as hair germs and sweat ducts, or as a fold of
glands and teeth have similar early inductive events. tissue that establishes the nail fold. These earliest induc­
Interactions between the embryonic ectoderm and under­ tive events lead to the formation of an epidermal thick­
lying dermis are important in the development of the skin ening, the placode. The epithelial layers then proliferate
appendages. Experimental studies of appendage forma­ with growth into the mesenchyme, forming a bud. After
tion have revealed the early molecular events that occur bud formation, specific characteristics of the diverse
stepwise to control various aspects of appendage forma­ appendages emerge. Nerves and vessels, cell adhesion
tion (reviewed in [31]). Epithelial and mesenchymal cells molecules (CAMs), soluble mediators and homeobox
lie in close proximity at the sites of appendage formation, genes (homeoproteins) have been implicated in the pat­
and in some cases make physical contact (Fig. 1.40). These tern formation of certain appendages (reviewed in [32–37).
structures are thought to develop in response to epithe­ Several molecular pathways, including Wnt/β‐catenin,
lial–mesenchymal interactions that initiate the process fibroblast growth factor (FGF), transforming growth
through instructive messages, sustain the process through factor (TGF)β/bone morphogenetic protein (BMP) and
permissive interactions and then support differentiation hedgehog pathways, are well established as mediators
and maintenance of the fully developed appendage. of these early interactions (reviewed in [31]). The
24 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a)

Fig. 1.40  Developing hair germ showing the close proximity between the
epithelial cells of the germ and the mesenchymal cells of the dermis, which
will follow the developing appendage and influence its development and
differentiation (×8250).

e­ vidence for these interactions in the development of


the epidermal appendages in human skin is best docu­
mented for formation of the pilosebaceous apparatus
[38–40].

Nail formation
(b)
The formation of the nail unit is highly dependent on dor­
sal–ventral patterning of the limb bud. As with other ecto­ Fig. 1.41  Developing nail. (a) Scanning electron micrograph of a digit from
dermal appendages, epithelial–mesenchymal pathways an 85‐day EGA fetus showing the boundaries of the nail field recognized
by proximal (identifying the position of the nail fold), lateral and distal
are critical for proper patterning (reviewed in [41]).
folds. (b) A section through the digit of a 70‐day EGA fetus shows the
Mutations in factors critical in dorsal–ventral patterning position of the nail fold and the thicker and more advanced epidermis over
of the mammalian limb, such as Wnt7a, engrailed 1 (EN1) the nail bed (a, ×100; b, ×150).
and LIM homeobox transcription factor 1β (LMX1B), give
rise to developmental nail anomalies such as nail–patella
syndrome. By 90 days EGA, the dorsal ridge is evident superfi­
The distal rays of the digit are evident on the hand of cially and is delineated from the plantar surface of the
the 50‐day EGA embryo, and within the next 7 days the digital pad by a deep constriction (Fig. 1.42a). The nail
digits separate [42]. Formation of the nail on the dorsal fold has invaginated deeply into the dermis and
surface of the digits and the eccrine sweat glands on organized into dorsal (roof of the fold) and ventral
the ventral surface is initiated at approximately the (floor of the fold) layers that are distinguished from
same time after embryo–fetal transition. By 70 days’ one another morphologically and functionally
gestation, the boundaries of the nail field are estab­ (Fig. 1.42b). The ventral fold becomes the nail matrix,
lished externally by proximal, lateral and distal folds which is primarily responsible for the synthesis of the
(Fig. 1.41a) and sections through the digit reveal a shal­ nail plate.
low nail fold (Fig.  1.41b). Development of the epider­ The earliest nail, which is formed late in the first trimes­
mis over the nail bed is furthest advanced at its ter, consists of several layers of keratinized cells that are
distal‐most margin [43,44]. evident primarily at the distal margin of the nail bed and
Chapter 1  Embryogenesis of the Skin 25

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
(b)

(a)

(c)

(d)
Fig. 1.42  Developing digit and nail. (a) Scanning electron micrograph of the ventral surface of the developing digit of an 80‐day EGA fetus showing the
dorsal ridge and the large volar pads. (b) Transverse section through the nail fold of a digit from an 85‐day EGA fetus showing the distinction between
dorsal and ventral (presumptive nail matrix) surfaces of the nail fold. The higher‐magnification section shows more detail of the epidermal surface and the
mesenchyme beneath the nail fold. (c) Transverse sections through the digit and distal‐most tip of the nail bed of a digit from a 105‐day fetus showing
keratinization of the superficial cells forming the ‘preliminary’ nail. (d) Scanning electron micrograph of a nail of a 140‐day EGA fetus showing the fragile
nature of the nail plate (a, ×60; b, ×100; c, ×200; d, ×100).

over the dorsal ridge. By 15 weeks EGA, a thick cornified Eccrine sweat gland formation
layer covers the nail bed (Fig.  1.42c). This ‘preliminary’ Human skin has the highest density of eccrine sweat
nail is easy to slough from the surface and thus may be glands amongst mammals. The development of the digits
composed to a greater extent of keratinized epidermal and the morphogenesis of eccrine sweat glands and nails
cells from the nail bed rather than derived from the matrix occur on the hand in advance of the foot and on the distal
of the nail fold. The nail of a 19‐week EGA fetus is estab­ pads ahead of the middle and proximal phalanges and
lished by both the nail matrix and the nail‐bed epidermis, the palm [32,42]. Eccrine sweat glands form on the gen­
although the nail is still fragile (Fig. 1.42d). The nail that is eral body surface at least 4–6 weeks later than on the
present at birth is actually a composite of layers of cells palms and soles. They are the last of the epidermal
derived from the dorsal nail fold (contributes the outer­ appendages to be formed (reviewed in [32]). The struc­
most layer of the nail) and the nail matrix (contributes the tural events have been much better characterized for the
intermediate layer of the nail); the distal half to two‐thirds ridged skin of the palmar plantar and digital skin than for
of the nail bed contributes the inner layer of the nail. The trunk skin (reviewed in [15–17]). About 8.5–9 weeks EGA,
layers are more evident in the fetus than in the postnatal the shape of the terminal digit is evident. Volar pads, tran­
individual [45]. sient mounds of mesenchyme that accumulate beneath
26 Section 1  Development, Structure and Physiology of the Skin

the epidermis on the ventral surface of the digits, are well


STRUCTURE AND PHYSIOLOGY

formed (Fig. 1.42a). Their presence in the first trimester


SECTION 1: DEVELOPMENT,

is presumed to influence the dermatoglyphic patterns


[46,47] and the development of those flexion creases that
are not considered to be dependent upon movements of
the hand [42,48,49]. Interest in the development of flexion
creases relates to the aberrant patterns they assume in cer­
tain congenital disorders. Volar pads begin to regress
around 10.5–11 weeks EGA and are nearly gone by 12.5–
13 weeks EGA [42] when, presumably, their influence is
no longer needed for morphogenesis.
The primary epidermal ridges are first formed around
10–11 weeks EGA [50]. They are recognized in sectioned
specimens as localized aggregations of basal epidermal (a)
cells on the digits, palms and soles (Fig. 1.43a); in sheets of
epidermis viewed basally, they appear first as discontinu­
ous then as continuous ridges [51]. At this stage, the epi­
dermis on the plantar surface consists of five or six layers
of intermediate cells and the periderm. Merkel cells con­
taining the characteristic granules are distributed along
the primary epidermal ridges, where they may attract
periglandular nerve fibres to this position of the structure
[52] (Fig. 1.43b). Electron microscopy has revealed nerve
fibres associated with basal laminae underlying the ridges
and Merkel cells, and occasionally extending into the
­epidermal tissue [32]. Merkel cell–nerve complexes are
evident in digital skin before the primary ridges begin to
(b)
form, and remain prominent in the primary ridges after
the appearance of the sweat gland anlagen (Fig.  1.43a).
They do not appear to migrate, however, into any region
of the developing appendage.
The sweat gland primordia are recognized around
13–14 weeks at regular sites along the now flattened
ridges as narrowed, solid, epithelial cords of cells that
contain basal cell keratins and express classical carci­
noembryonic antigen (CEA) on all cells [50,52]. There is
no evidence of condensed mesenchyme associated with
the onset of sweat gland development as there is in fol­
licle development, thus suggesting that other sources of
signalling molecules, possibly the volar pad mesenchyme,
or nerves and/or other cell–cell interactions (perhaps
within the epithelium) may instruct the sites for appendage
formation and trigger the onset of gland development.
As the cords of epithelial cells elongate into the der­
mis, a thickening at the terminus defines the glandular
segment from the duct [53] (Fig. 1.44a). Ductal, secre­ (c)
tory, myoepithelial and acrosyringial cell types differ­ Fig. 1.43  Developing sweat glands on the ventral surfaces of the digit.
entiate in the dermal and intraepidermal regions of the (a) Cross‐section through the digit of a 95‐day EGA fetus showing the
gland and duct, and are easily distinguished from one primary epidermis ridges organized from the basal layer of the epidermis.
another by light and electron microscopy (Fig.  1.44) Note the abundance of nerves and vessels in the proximal dermis.
(b) Immunolabelled section through the palm of a 105‐day fetal hand
and by immunostaining patterns using antibodies to
showing the position of Merkel cells (green) marked by an antibody that
keratin intermediate filament proteins [53]. All cells recognizes keratin 18. The red labelling recognizes neurofilaments in
continue to express CEA [52]. The secretory cells bor­ dermal nerve fibres. (Source: Micrograph courtesy of Dr Dong Kun Kim.)
der a central lumen within the gland; myoepithelial (c) Immunolabelled section through the palm of a 163‐day fetal hand
cells are evident at the periphery of the structure. Cells showing the position of Merkel cells (green) marked by an antibody that
of layers of the duct and of the gland are distinct from recognizes keratin 20. The red labelling recognizes neurofilaments in
one another at 15 weeks EGA by their morphological dermal nerve fibres. Note the well‐established sweat ducts and the
secondary epidermal ridges alternating with the primary ridges from which
properties and by differences in expression of keratins
the ducts are formed. (Source: Micrograph courtesy of Dr Dong‐Kun Kim.)
(duct) and vimentin (gland) and CAMs [53,54]. (a, ×300; b, ×300; c, ×100.)
Coexpression of the two intermediate filament proteins
Chapter 1  Embryogenesis of the Skin 27

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
(a) (c)

(d)

(b)
Fig. 1.44  Developing sweat glands and ducts. (a) Scanning electron micrograph of the palm of a 19‐week EGA fetus showing the elongated ducts and the
club‐like terminal gland. (b) A section through the palm of a 147‐day EGA fetus shows the position of the duct and gland in the dermis and the canalization
(keratinization) of the intraepidermal portion of the duct. A higher‐magnification image of the gland and duct in the palmar dermis of a 126‐day EGA fetus
(c) shows the cell layers of the duct and the cells of the gland (arrows). (d) Scanning electron micrograph of the undersurface of palmar epidermis of a
19‐week EGA fetus showing the primary epidermal ridges with the remnants of torn sweat ducts spaced periodically and the secondary ridges that do not
give rise to sweat ducts (a, ×80; b, ×120; c, ×300; d, ×80).

in the same cells of the secretory segment of the devel­ Secondary ridges form between the primary ridges (see
oping gland is unique to this appendage, but it is char­ Figs 1.43c and 1.44d). They do not give rise to sweat glands
acteristic of other glandular tissues such as mammary or contain Merkel cells. Globular keratohyalin granules
and salivary glands [53]. are evident in the cytoplasm of the circumferentially
28 Section 1  Development, Structure and Physiology of the Skin

organized cells of the intraridge, the intraepidermal por­ Pilosebaceous apparatus formation
STRUCTURE AND PHYSIOLOGY

tion of the duct (the acrosyringium) at about 15 weeks The pilosebaceous apparatus is best described as a com­
SECTION 1: DEVELOPMENT,

EGA, signalling the onset of canalization in the acrosyrin­ posite epithelial–mesenchymal structure with critical
gium (Fig.  1.45). The lumen of the duct forms by the molecular ‘cross‐talk’ between the two. Morphogenesis of
fusion of cytoplasmic vesicles within ductal cells. The the hair follicle begins on the head and face at around
duct remains partially occluded even in the third trimes­ 70–80 days EGA, shortly after the epidermis stratifies,
ter [55]. By 22–24 weeks EGA, the sweat glands on the then proceeds in a cephalocaudal direction [20]. The pro­
palms and soles have attained the structure of the adult cess is completed at around 19–20 weeks EGA, when
glands, with a coiled secretory gland. hairs extend from the lanugo follicle through the peri­
The absence of sweat glands is a hallmark of the derm‐covered surface of the skin. Follicles form in regular
ectodermal dysplasias due to mutations in ectodysplasin patterns in all body regions, with the distances separating
pathway genes [56]. The ectodysplasin pathway interacts each dependent upon the specific site (Fig.  1.46). The
with the Wnt/β‐catenin pathway, amongst others, to con­ stages of follicle development, including hair germ, hair
trol eccrine gland formation from induction through peg, bulbous hair peg and lanugo follicle stages of follicle
secretory duct differentiation [57]. While these pathways formation (Fig. 1.47), are based on the vellus hairs on the
are shared with other skin appendages, the molecular trunk [58]. Follicles form only during development and
mechanisms leading to specification of eccrine glands decline in numbers as a function of ageing.
specifically is not well established. The induction of follicles, their stages of development,
maintenance in the adult and the cyclical growth and
regression of the scalp follicles are all dependent upon an
association of the follicle epithelium with dermal mesen­
chymal cells that form a cellular and matrix sheath
around the developing and mature follicle and establish
the dermal papilla as a special collection of mesenchymal
cells that modulate the production and elongation of
hairs [59].

(a)

(b)
Fig. 1.45  Formation of the intraepidermal sweat duct by the development
and coalescence of vesicles and subsequent keratinization of the lining Fig. 1.46  Scanning electron micrograph of the undersurface of the
cells (a). Note the globular keratohyalin granules that are characteristic of epidermis from a 15‐week EGA fetus showing the pattern of hair follicles
acrosyringial keratinization (×9100). Source: Micrograph originally in the hair peg and hair germ stages of development. Note the longitudinal
published in Odland G, Holbrook K. Curr Prob Derm 1981;9:29–49. grooves in the epidermis that mark the position of intraepidermal hair
Reproduced with permission of Karger Publishers. canals (×100).
Chapter 1  Embryogenesis of the Skin 29

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Fig. 1.47  Diagram of the stages of hair follicle
formation including the prefollicle two‐layered
epidermis, pregerm, hair germ, early hair peg,
late hair peg/early bulbous hair peg and
lanugo follicle stages.

The epithelial and mesenchymal cells have been exten­ At around 13–14 weeks EGA, the hair germs elongate
sively characterized at each stage of follicle formation into the dermis as cords of cells called hair pegs (Figs 1.46
with regard to the expression of growth factors, growth and 1.50). The hair peg consists of an inner core of cuboi­
factor receptors, cytokines, other signalling molecules dal cells and outer layer of columnar cells that is associ­
and growth regulators, and structural proteins and ated with the basal lamina surrounding the follicle and
enzymes (reviewed in [60–63]). Experimental studies in continuous with that of the interfollicular epidermis.
animal models, transgenic animals, tissue recombination Cells of the outer layer contain the same keratins as the
preparations and various cell and organ culture systems basal epidermal keratinocytes and the cells of the inner
have revealed the functions of specific populations of core contain intermediate cell keratins (Fig.  1.50a), thus
cells in developing follicles (e.g. dermal papilla and cells implicating the origins of follicle cells from two epider­
of the bulge) and events that signal early and sequential mal layers. Merkel cells are distributed among the outer
steps in the induction of other appendage primordia root sheath keratinocytes.
[31,36,37]. Thus, the data can be used only to infer what Early hair pegs are cylindrical, but as they elongate
may be occurring in utero at the time the human follicles further they develop three regions: (i) a constricted, neck‐
form de novo as most studies are done on postnatal like connection with the epidermis (the presumptive
human hair follicles. infundibulum); (ii) a central, cylindrical region (the
The sites of follicle formation can be recognized, even presumptive isthmus); and (iii) a terminal zone that
­
before the hair germs are visible, in sections of fetal skin by becomes widened at its most distal end (the lower follicle
immunostaining the tissue with an antibody that recog­ and the presumptive bulb) (Fig.  1.50). The length of
nizes the matrix molecule tenascin [33,34]. Patches of reac­ the hair peg and the three zones of the developing follicle
tion product at the basement membrane zone correspond are exaggerated in some regions of the skin but more
to pregerms, or sites where basal keratinocyte nuclei are ­subtle in others.
closely spaced and mesenchymal cells are aggregated Changes occur in all three regions, with the first notable
(Fig. 1.48) [64,65]. Cells from the basal epidermal layer bud events taking place at the proximal and distal ends.
into the dermis to become hair germs (Fig.  1.48b). Elongated core cells in the neck of the follicle continue
Condensed mesenchymal cells associate closely with the into the epidermis, where they form a strand of cells that
germs, often extending processes that contact the basal lies between the basal and intermediate cell layers (see
lamina (Fig. 1.40); this collection of mesenchymal cells is Fig. 1.50). This is the hair tract that marks the position and
intensely immunoreactive with antibodies to NGFR (p75) pathway of the presumptive hair canal (Fig. 1.50c) [66].
(Fig.  1.49), NCAM and other growth factor receptors. The distal end of the hair peg flattens and the epithelial
Little if any collagenous matrix is present around the cells cells along this basal border elongate to form a distinct
as a consequence of either downregulated production or layer that establishes the matrix (Fig.  1.50a and b). The
enhanced degradation (reviewed in [33,34]). Merkel cells flattened end of the follicle begins to invaginate into
are recognized in some of the developing germs. As is the the cord, shaping the bulb with the matrix as the roof of
case with the other appendages in which Merkel cells are the bulb. Mitotic figures are evident in matrix cells, and lon­
prominent, they may play a role in targeting nerve fibres gitudinally orientated cells, presumably the progeny of
towards the developing appendage. the dividing matrix cells, move out of the matrix into the
30 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a)

(b)
Fig. 1.48  Immunolabelled section of human fetal skin at 70–75 days EGA
showing tenascin‐positive sites where hair germs have formed or are (a) (b)
expected to form (a and b). The clustering of basal keratinocytes is
apparent at sites where tenascin is strongly expressed in the basement
membrane zone (a) (a, ×100; b, ×350). Source: Micrograph courtesy of
Dr Beth Kaplan.

(c)
Fig. 1.50  Late‐stage hair pegs (a, b) showing the continuity of the
intermediate‐layer keratins into the upper core cells of the follicle (a), the
regions of the peg and the mesenchymal cells surrounding the peg and
aggregated at its tip (the presumptive dermal papilla) in association with
the presumptive matrix of the follicle. Note the differences in cell
Fig. 1.49  Hair germs in the skin of a 97‐day fetus immunostained to orientation in the inner and outer and the distal and proximal regions of
recognize the p75 neurotrophic receptor, which recognizes NGFR. Note the the peg (b). (c) Section through the upper end of a hair peg showing the
concentration of this immunoreactive material within the mesenchymal continuation of cells into the epidermis as the hair track (a, ×300; b, ×300;
cells surrounding the hair germ (×120). c, ×300).

centre of the cord, thereby establishing the first layers of


the inner root sheath and the hair (the hair cone). Cells of in [27]) (Fig. 1.51). Such samples can be induced to syn­
the outer layer of the follicle located adjacent and lateral thesize melanin by the dihydroxyphenylalanine (DOPA)
to the matrix appear to become more loosely associated reaction if the fetus is normal [67,68].
with one another, perhaps permitting the inward migra­ During these events, the cord is surrounded in its
tion of the cells derived from the matrix. Melanocytes entirety by several layers of elongated mesenchymal cells
aggregate in the matrix and produce melanin ahead of that form a sheath. The connective tissue matrix is sparse
melanocytes in the general body skin, thus making the within this cellular sheath and appears to be devoid of
bulbs of developing hair follicles ideal sites to examine the fibrillar collagens that are present in the surrounding
when evaluating skin biopsy samples from a fetus at risk subepidermal and interstitial matrix (Fig.  1.52). Diffe­
of tyrosinase‐negative oculocutaneous albinism (reviewed rences in matrix molecules are observed at different levels
Chapter 1  Embryogenesis of the Skin 31

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
(a)
(a)

(b)
(b)
Fig. 1.51  The bulb of a hair peg (a) and lanugo follicle (b) in skin from
different regions obtained at ages 115 days and 125 days EGA. Note the
concentration of melanocytes in the matrix of the follicle (a, ×300;
b, ×300).

of the hair peg and when comparing the dermal papilla


with the follicle sheath [33,34].
Between 15 and 17 weeks EGA, bulges of epithelial
cells begin to grow out from the epithelial cord on the
posterior surface of the follicle and the adult layers of the
follicle differentiate into the hair and internal root sheath.
Once these bulges form, the follicle is called a bulbous
hair peg (Fig. 1.53a). The factors that stimulate the devel­
opment of these structures to arise from the follicle, at a
precise stage in the hair peg formation and at precise sites
along the hair peg, are unknown. There are no obvious
landmarks along the hair peg that provide morphological (c)
clues as to how the bulges might originate. Fig. 1.52  Sections of fetal skin immunolabelled with antibodies to
The most superior bulge is the primordium of the collagens of the dermis. Note the decreased staining for types I (a),
sebaceous gland (Fig.  1.53a). Cells begin to produce III (b) and V (c) collagens in the developing hair germs (a, ×300;
sebum soon after this structure is evident. Analysis of b, ×300; c, ×300). Source: Immunolabelling studies by Dr Lynne T.
Smith.
epidermal lipids from fetal skin at this stage reveals a
sterol/wax ester content, which suggests that the mate­
rial is similar to adult sebum [69]. The second bulge, the
‘true bulge’, forms concurrently with and slightly distal
to the sebaceous gland. It is the site of follicular stem cells
32 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a)

(b)
Fig. 1.54  The hair canal. (a) Section through the skin of a 138‐day EGA
fetus showing the floor of an opened hair canal. Keratinization of this
structure stands out in contrast to the non‐keratinized epidermis.
(b) Scanning electron micrograph of the skin of a 21‐week EGA fetus
(a) (b) showing hair canals within (beneath the surface of) the epidermis. Note
that one hair has emerged and others are evident through the thinned
Fig. 1.53  Bulbous hair peg in the skin of a 15‐week EGA fetus. (a) In the epidermal layers above the canal (a, ×100; b, ×185).
longitudinal section through the follicle, note the sebaceous gland, the
bulge located just distal to the sebaceous gland, the cell layers of the inner
root sheath (inner bar) and the outer layers of the outer root sheath (outer (axilla, areola, scalp, external eyelid, auditory meatus
bar). The infundibulum is the region of the follicle that lies between the and anogenital regions).
sebaceous gland and the epidermis. Note the cells of the dermal papilla
The cylindrical layers of the follicle differentiate and
within the bulb. (b) A cross‐section through a region between the two bars
shows the layers of the outer root sheath and the inner root sheath (a,
keratinization begins in several different structures of the
×300; b, ×300). follicle concurrently: the outer layer of cells of the inner
root sheath (layer of Henle); the cuticle and cortex of the
hair (Fig. 1.53b); the sebaceous duct; and the hair canal.
[70] and the point of attachment of the arrector pili mus­ Continued production of cells of the three layers of the
cle (Fig. 1.53a). Multipotent epithelial stem cells reside in inner root sheath and the hair gradually creates the kerati­
the bulge [71]. The stem cell population is maintained nized tube of the inner root sheath and the hair.
throughout development and postnatal life, giving rise Keratinization within the hair tract canalizes the cord of
to the cells responsible for regenerating the cycling folli­ cells and forms a keratin‐lined channel that courses diag­
cle as well as having an important role in wound healing. onally through the epidermis (Figs 1.50c and 1.54a) [69].
Merkel cells also concentrate in the bulge at early stages The granular and cornified cell layers of the hair canal
of bulge formation. They may be important in establish­ form a sharp contrast with the remainder of the, as yet
ing this structure, stimulating proliferation or attracting non‐keratinized, epidermis (Fig. 1.54a). The angle of the
nerve fibres and smooth muscle cells to the site. A third canal with the epidermis and the intraepidermal length of
bulge may form superior to the sebaceous gland as the the canal are regionally variable. On the eyebrow, for
primordium of the apocrine sweat gland. These struc­ example, the hair canals are closely spaced and their
tures are located in the restricted sites of the body where paths are very short. In other regions of the body, such as
apocrine sweat glands are present in the postnatal infant the appendages, the canals can be very long. By examining
Chapter 1  Embryogenesis of the Skin 33

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Fig. 1.55  Cross‐section of the hair canals from a fetal skin sample
obtained at 23 weeks EGA from a fetus affected with harlequin ichthyosis.
Keratinization of these structures is extraordinarily thick (×300).

Fig. 1.57  Skin from a 117‐day EGA fetus, immunostained with the AE13
antibody, which recognizes a hair keratin (×120).

whole‐mount preparations of tissue at this stage by the


presence of melanin‐producing melanocytes in the
matrix and the melanin pigment in the hair (Fig. 1.51b).
Antibodies to hair keratins also highlight the forming
hair in sections of skin (Fig.  1.56). The first hairs of the
fetus are in the anagen phase of the hair cycle. By 24–28
weeks EGA, the fetal follicles enter catagen and then
telogen phase. There is shedding of the first lanugo hairs
into the amniotic fluid prior to re‐entry into second
anagen. Due to synchronous cycling in newborns, a sig­
nificant number of follicles will enter telogen postna­
tally and be shed, ­creating alopecia that can last up to
6 months.
Fig. 1.56  A lanugo follicle associated with an epidermal sheet from a
126‐day EGA fetus. Note the hair, sebaceous gland, bulge and sebum‐ ­References
filled infundibulum (×150). Source: Micrograph courtesy of Dr Carolyn 1 Dale BA, Holbrook KA, Kimball JR et al. Expression of the epidermal
Foster. keratins and filaggrin during fetal human development. J Cell Biol
1985;101:1257–69.
2 Regauer S, Franke WW, Virtanen I. Intermediate filament cytoskele­
the surface of the fetal skin at this stage, even with a hand ton of amnion epithelium and cultured amnion epithelial cells:
lens, it is possible to see these canals (Fig. 1.54b). Scanning expression of epidermal cytokeratins in cells of a simple epithelium.
J Cell Biol 1985;100:997–1009.
electron microscopy reveals sites where the roof of the 3 Nanbu Y, Fujii S, Konishi I et  al. CA 125 in the epithelium closely
canal is eroded and the hair is visible (Fig. 1.54b). These related to the embryonic ectoderm: the periderm and the amnion. Am
sites expose the keratinized lining of the hair canal and J Obstet Gynecol 1989;161:462–7.
4 Lane AT, Negi M, Goldsmith LA. Human periderm: a monoclonal
loose squames attached to the portion of the hair that lies
antibody marker. Curr Prob Dermatol 1987;16:83–93.
within the canal. It is important to understand that kerati­ 5 M’Boneko V, Merker H‐J. Development and morphology of the peri­
nization of the canals occurs earlier in the follicular epi­ derm of mouse embryos (days 9–12 of gestation). Acta Anat
dermis than in the interfollicular epidermis because this 1988;133:325–36.
6 Sanes JR, Rubenstein JLR, Nicolas J‐F. Use of a recombinant retrovirus
information permits the hair canals to be used for evaluat­ to study post‐implantation cell lineage in mouse embryos. EMBO J
ing fetal skin biopsies obtained for prenatal diagnosis of 1986;5:3133–42.
one of the severe disorders of keratinization (Fig. 1.55). 7 Byrne C, Talbsky M, Fuchs E. Programming gene expression in devel­
oping epidermis. Development 1994;120: 2369–83.
Production of the hair marks the establishment of the
8 Holbrook KA, Odland GF. The fine structure of developing human
lanugo follicle at 19–21 weeks EGA (Figs 1.56 and 1.57). epidermis: light, scanning and transmission electron microscopy of
The hair and its cells of origin are well outlined in the the periderm. J Invest Dermatol 1975;65:16–38.
34 Section 1  Development, Structure and Physiology of the Skin

9 Holbrook KA, Underwood RA, Dale BA et al. Formation of the corni­ 34 Kaplan ED, Holbrook KA. Dynamic expression patterns of tenascin,
fied cell envelope in human fetal skin: presence of involucrin, kerato­ proteoglycans and cell adhesion molecules during human hair follicle
STRUCTURE AND PHYSIOLOGY

linin, loricrin and transglutaminase correlated with the onset of morphogenesis. Dev Dyn 1994;199:141–55.
SECTION 1: DEVELOPMENT,

transglutaminase activity. J Invest Dermatol 1991;96:542A. 35 Chuong C‐M, Widelitz RB, Jiang T‐X. Adhesion molecules and home­
10 Akiyama M, Smith LT, Yoneda K et al. Periderm cells form cornified oproteins in the phenotypic determination of skin appendages.
cell envelope in their regression process during human epidermal J Invest Dermatol 1993;101:10S–15S.
development. J Invest Dermatol 1999;111:903–9. 36 Noveen A, Jiang T‐X, Ting‐Berreth SA et al. Homeobox genes Msx‐1
11 Riddle CV. Intramembranous response to cAMP in fetal epidermis. and Msx‐2 are associated with induction and growth of skin append­
Cell Tissue Res 1985;241:687–9. ages. J Invest Dermatol 1995;104:711–9.
12 Koren G. Fetal toxicology of environmental tobacco smoke. Curr
37 Noveen A, Jiang T‐X, Chuong C‐M. Protein kinase A and protein
Opin Pediatr 1995;7:128–31. kinase C modulators have reciprocal effects on mesenchymal con­
13 Mears GJ, Van Petten GR. Fetal absorption of drugs from the amniotic densation during skin appendage morphogenesis. Dev Biol
fluid. Proc West Pharmacol Soc 1977;20:109–14. 1995;171:677–93.
14 Lind T, Kendal A, Hytten FE. The role of the fetus in the formation of 38 du Cros DL. Fibroblast growth factor influences the development and
amniotic fluid. J Obstet Gynaecol Br Commonw 1972;79:289–98. cycling of murine hair follicles. Dev Biol 1993;156:444–53.
15 Holbrook KA. Structural and biochemical organogenesis of skin and 39 Moore GPM, du Cros DL, Isaacs K et al. Hair growth induction: roles
cutaneous appendages in the fetus and neonate. In: Polin RA, Fox of growth factors. Ann NY Acad Sci 1991;624:308–25.
WW (eds) Neonatal and Fetal Medicine Physiology and 40 Millar SE. Molecular mechanisms regulating hair follicle develop-
Pathophysiology. New York: Grune & Stratton, 1992:527–51. ment. J Invest Dermatol 2002;118:216–25.
16 Chu DH. Development and structure of skin. Chapter 7. In: Goldsmith 41 Maito M, Ohyama M, Amagai M. Exploring the biology of the nail:
LA, Katz SI, Gilchrest BA et  al. (eds) Fitzpatrick’s Dermatology in An intriguing but less‐investigated skin appendage. J Dermatol Sci
General Medicine, 8th edn. New York: McGraw‐Hill, 2012. 2015; 79:187–93.
17 Holbrook KA, Sybert VP. Basic science. In: Schachner L, Hansen R 42 Kimura S. Embryologic development of flexion creases. Birth Defects:
(eds) Pediatric Dermatology, 2nd edn. New York: Churchill Original Article Series 1991;27:113–29.
Livingstone, 1995. 43 Zaias N. Embryology of the human nail. Arch Dermatol 1996;

18 Richardson RJ, Hammond NL, Coulombe PA et  al. Periderm pre- 87:37–53.
vents pathological epithelial adhesions during embryogenesis. 44 Hashimoto K, Gross BG, Nelson R et al. The ultrastructure of the skin
J Clin Invest 2014;124:3891–900. of human embryos. III. The formation of the nail in the 16–18 weeks
20 Hardman MJ, Moore L, Ferguson MW, Byrne C. Barrier formation in old embryo. J Invest Dermatol 1996;47:205–17.
the human fetus is patterned. J Invest Dermatol 1999;113:1106–13. 45 Lewis BL. Microscopic studies, fetal and mature nail and surrounding
21 Holbrook KA, Dale BA, Sybert VP et al. Epidermolytic hyperkerato­ soft tissue. Arch Dermatol 1954;70:732–47.
sis: ultrastructure and biochemistry of skin and amniotic fluid cells 46 Mulvihill JJ, Smith DW. The genesis of dermatoglyphics. J Pediatr
from two affected fetuses and a newborn infant. J Invest Dermatol 1969;75:597–9.
1983;81:222–7. 47 Hirsch W, Schweichel JU. Morphological evidence concerning the
21 Holbrook KA, Wapner R, Jackson L et al. Diagnosis and prenatal diag­ problem of skin ridge formation. J Ment Defic Res 1973;17:58–72.
nosis of epidermolysis bullosa herpetiformis (Dowling–Meara) in a 48 Kimura S, Kitagawa T. Embryological development of human palmar,
mother, two affected children and an affected fetus. Prenatal Diagn plantar and digital flexion creases. Anat Rec 1990;226:249–57.
1992;12:725–39. 49 Hale AR. Morphogenesis of volar skin in the human fetus. Am J Anat
22 Okano J, Lichti U, Mamiya S et  al. Increased retinoic acid levels 1952;91:147–81.
through ablation of Cyp26b1 determine the processes of embryonic 50 Metze D, Bhardwaj R, Amann U et al. Glycoproteins of the carcinoem­
skin barrier formation and peridermal development. J Cell Sci bryonic antigen (CEA) family are expressed in sweat and sebaceous
2012;125:1827–36. glands of human fetal and adult skin. J Invest Dermatol 1966;
23 Holbrook KA, Odland GF. Regional development of the human 106:64–9.
epidermis in the first trimester embryo and the second trimester 51 Kim D‐G, Holbrook KA. The appearance, density and distribution of
fetus (ages related to the timing of amniocentesis and fetal biopsy). Merkel cells in human embryonic and fetal skin: their relation to
J Invest Dermatol 1980;80:161–8. sweat gland and hair follicle development. J Invest Dermatol 1995;
24 Akiyama M, Dale BA, Smith LT et al. Regional difference in expres­ 104:411–16.
sion of characteristic abnormality of harlequin ichthyosis in affected 52 Narisawa Y, Hashimoto K, Pietruk T. Biological significance of dermal
fetuses. Prenat Diagn 1998;18:425–36. Merkel cells in development of cutaneous nerves in human fetal skin.
25 Pinkus H. Embryology of hair. In: Montagna W, Ellis RA (eds) The J Histochem Cytochem 1992;40:65–71.
Hair Growth. New York: Academic Press, 1958:1–32. 53 Moll I, Moll R. Changes of expression of intermediate filament pro­
26 Sasaki K, Akiyama M, Yanagi T et al. CYP4F22 is highly expressed at teins during ontogenesis of eccrine sweat glands. J Invest Dermatol
the site and timing of onset of keratinization during skin develop­ 1992;98:777–85.
ment. J Dermatol Sci 2012;65:156–8. 54 Fujita M, Furukawa F, Fujii K et al. Expression of cadherin molecules
27 Holbrook KA, Dale BA, Williams ML et al. The expression of congeni­ during human skin development: morphogenesis of epidermis, hair
tal ichthyosiform erythroderma in second trimester fetuses of the follicles and eccrine sweat ducts. Arch Dermatol Res 1992;284:159–66.
same family: morphologic and biochemical studies. J Invest Dermatol 55 Hashimoto K, Gross BG, Lever WF. The ultrastructure of the skin of
1988;91:521–31. human embryos. I. The intraepidermal eccrine sweat duct. J Invest
28 Holbrook KA, Smith LT, Elias S. Prenatal diagnosis of genetic skin Dermatol 1965;45:139–51.
disease using fetal skin biopsy samples. Arch Dermatol 1993; 56 Itin PH. Etiology and pathogenesis of ectodermal dysplasias. Am J
129:1437–54. Med Genet A 2014;164A:2472–7.
29 Sybert VP, Holbrook KA, Levy M. Prenatal diagnosis of severe derma­ 57 Cui CY, Yin M, Sima J, et  al. Involvement of Wnt, Eda and Shh at
tologic diseases. Adv Dermatol 1992;7:179–209. defined stages of sweat gland development. Development 2014;
30 Sybert VP, Holbrook KA. Antenatal pathology of the skin. In:
141(19):3752–60.
Claireaux AE, Reed GB (eds) Diseases of the Fetus and Newborn: 58 Holbrook KA, Minami SA. Hair follicle morphogenesis in the human:
Pathology, Radiology and Genetics. New York: Cockburn, Chapman characterization of events in vivo and in vitro. NY Acad Sci
& Hall, 1995:755–68. 1991;642:167–96.
31 Biggs LC, Mikkola ML. Early inductive events in ectodermal
59 Reynolds AJ, Oliver RF, Johoda CAB. Dermal cell populations show
appendage morphogenesis. Semin Cell Dev Biol 2014;25‐26:11–21. variable competence in epidermal support: stimulatory effects of hair
32 Moore SJ, Munger BL. The early ontogeny of the afferent nerves and papilla cells. J Cell Sci 1991;98:75–83.
papillary ridges in human digital and glabrous skin. Dev Brain Res 60 Millar SE. Molecular mechanisms regulating hair follicle develop-
1989;48:119–41. ment. J Invest Dermatol 2002;118:216–25.
33 Holbrook KA, Smith LT, Kaplan ED et al. The expression of morpho­ 61 Lee J, Tumbar T. Hairy tale of signaling in hair follicle development
gens during human follicle development in vivo and a model for and cycling. Semin Cell Dev Biol 2012;23:906–16.
studying follicle morphogenesis in vitro. J Invest Dermatol 62 Messenger A. The control of hair growth: an overview. J Invest

1993;101:39S–49S. Dermatol 1993;201:4S–9S.
Chapter 1  Embryogenesis of the Skin 35

63 Jahoda CAB, Reynolds AJ, Forrester JC et al. Hair follicle regeneration


following amputation and grafting into the nude mouse. J Invest
­Conclusion

STRUCTURE AND PHYSIOLOGY


Dermatol 1996;107:904–7.

SECTION 1: DEVELOPMENT,
Human embryonic development is complex and
64 Chiquet‐Ehrismann R, Mackie EJ, Pearson CA et  al. Tenascin: an
extracellular matrix protein involved in tissue interactions during
wrought with opportunity for developmental missteps
fetal development and oncogenesis. Cell 1986;47:131–9. leading to congenital malformation. Skin development
65 Erickson HP, Bourdon MA. Tenascin: an extracellular matrix protein is no exception. The study of skin development has been
prominent in specialized embryonic tissues and tumors. Annu Rev based largely on the foundation of descriptive work.
Cell Biol 1989;5:71–91.
66 Holbrook KA, Odland GF. Structure of the hair canal and the initial Fortunately, many of the methods for developing these
eruption of hair in the human fetus. J Invest Dermatol 1978;71:385–90. data also provide information about composition, and
67 Eady RAJ, Gunner DB, Garner A et al. Prenatal diagnosis of oculocu­ thus the morphological approaches have allowed a rea­
taneous albinism by electron microscopy. J Invest Dermatol
1983;80:210–12.
sonable story of skin development to unfold. Continuing
68 Kikuchi A, Shimizu H, Nishikawa T. Epidermal melanocytes in nor­ advances in molecular techniques and animal models
mal and tyrosinase‐negative oculocutaneous albinism fetuses. Arch have further contributed to understanding the pathways
Dermatol Res 1995;287:529–33. and interactions needed for normal skin development.
69 Williams ML, Hincenbergs M, Holbrook KA. Skin lipid content dur­
ing early fetal development. J Invest Dermatol 1988;91:263–8. In addition, advancing knowledge of genetic skin dis­
70 Cotsarelis G, Sun T‐T, Lavker RM. Label‐retaining cells reside in ease continues to shed further light on these pathways
the bulge area of pilosebaceous unit: implications for follicular and how perturbations in developmental pathways lead
stem cells, hair cycle and skin carcinogenesis. Cell 1990;61:1329–37.
to cutaneous disease. Further advances in understand­
71 Akiyama M, Dale BA, Sun T‐T et al. Characterization of hair follicle
bulge in human fetal skin: the human fetal bulge is a pool of undif­ ing skin development will undoubtedly lead to new
ferentiated keratinocytes. J Invest Dermatol 1995;105:844–50. therapeutic approaches.
36 

CHA PTER 2
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

Molecular Genetics in Paediatric Dermatology


Anna C. Thomas1 & Veronica A. Kinsler1,2
1
 Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
2
 Paediatric Dermatology Department, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK

Introduction, 36 Which samples to take for genetic Concept of personalized medicine, 39


Genetics and inheritance in brief, 36 testing, 39 Molecular genetics techniques, 40
Consent for genetic testing and incidental How to interpret genetic results, 39
findings, 38 What to do with a genetic result, 39

Abstract volume of genetic data generation, and have made the transition
from the research sphere to the diagnostic laboratories. The iden-
tification of disease‐causing genetic mutations in both inherited
Molecular genetics is the study of the structure of genes at molecu-
and sporadic paediatric dermatological conditions has begun to
lar level, and the impact of gene expression and regulation on the
influence clinical management, allowing preimplantation genetic
biology of the organism. The interplay between these two fields,
diagnosis, stratification for targeted personalized medical therapies,
genetics and biology, is a burgeoning area in all disease. In recent
or gene therapy in some cases. This chapter reviews the essential
years the process of disease gene identification and clinical genetic
terminology, key techniques and important advances needed to
diagnostic reporting has been revolutionized by the technologies
update paediatric dermatologists in this field, forming a basis for
discussed in this chapter, most prominently next‐generation
the detailed disease‐specific chapters that follow.
­sequencing (NGS). These techniques have increased the speed and

­Introduction confirm a diagnosis or to subclassify disease, the


i­dentification of disease‐causing genetic mutations in
Molecular genetics is the study of the structure of genes at both inherited and sporadic paediatric dermatologi-
a molecular level, and the impact of gene expression and cal conditions has begun to be clinically relevant in
regulation on the biology of the organism. The interplay clinical management. Preimplantation genetic diagno-
between these two fields, genetics and biology, is a bur- sis (PGD) as part of the in vitro fertilization (IVF) pro-
geoning area in all disease. For essential terminology in cess can be offered for future pregnancies where there is
molecular genetics, see Table 2.1. a family history and the genetic defect is known, for
A large number of skin conditions have now had their example in epidermolysis bullosa and the autosomal
genetic aetiology identified and this information can be recessive ichthyoses. In other instances the genetic
easily accessed through many of the online curated defect will stratify patients towards targeted medical
databases available, for example Online Mendelian therapies, such as in the vascular overgrowth disorders.
Inheritance in Man (www.omim.org) and the Decipher In addition, paediatric dermatology is now in the era
database (https://decipher.sanger.ac.uk), as well as from of personalized genetic therapy, and this will surely
published literature. be an area of tremendous growth in the decade to come.
In recent years, the process of disease gene identifica- Techniques of gene therapy will be discussed in
tion and clinical genetic diagnostic reporting has been Chapter 170.
revolutionized by the technologies discussed in this chap-
ter, most prominently next‐generation sequencing (NGS).
These techniques have increased the speed and volume of
genetic data generation, and have made the transition
­Genetics and inheritance in brief
from the research sphere to the diagnostic laboratories. The human genome is comprised of deoxyribonucleic acid
Storage and analysis of this data however, is still fraught (DNA) neatly contained within 23 pairs of chromosomes
with difficulties, not least in the interpretation of novel in the human, one of each pair inherited maternally and
(undescribed) variants. In addition, functional analysis of one paternally. The sex chromosomes are inherited XX for
the implications of any novel findings still requires vali- a female and XY for a male, and the other 22 pairs are
dation by cell biology and animal model generation. described as autosomes. It should also be noted that cel-
The implications of genetic diagnosis have changed lular mitochondria house 37 genes within a circular
in recent years. Whereas this used to serve only to genome that is inherited maternally. DNA is transcribed

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 2  Molecular Genetics in Paediatric Dermatology 37

Table 2.1  Essential terminology in molecular genetics

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Allele The alternative form of a gene’s sequence
Allele‐specific A test or treatment directed at only one allele
Array An ordered arrangement of probes on a solid surface
Base/nucleotide A DNA or RNA nucleotide (ACTG or ACUG)
Biallelic mutations Two different mutations at the same site in a gene
Chimaera An organism resulting from the fusion of two or more zygotes
Compound heterozygous mutations Two different mutations at different sites in a gene
Copy number Number of copies of a DNA sequence, usually at chromosomal or gene level but can be applied to
repeats in the DNA sequence
Copy number variation A change in the copy number of a portion of DNA, which is described in the normal population and
therefore considered to be a variant rather than pathogenic
De novo mutation Not inherited from either parent, but arisen for the first time in the patient
Digenic mutation A condition which is caused by mutations in two different genes
Exons The parts of the DNA sequence that code for proteins
Frameshift mutation A mutation that causes a shift in the reading frame of the DNA sequence
Gain‐of‐function/activating mutation A mutation that increases the function of the gene
Germline mutation A mutation that affects all the cells of an organism including the gametes
Hemizygous mutation Having only a single copy of a gene, which results in monoallelic expression
Heterozygous mutation The occurrence of a mutation in one copy of a gene but not in the other
Homozygous mutation The occurrence of the same mutation in both copies of a gene
Hybridization Binding of DNA or RNA to complementary strands or probes
Imprinting Natural silencing of one allele, leading to monoallelic expression
Introns The parts of the DNA sequence of the genome that do not code for proteins
In silico The analysis of the effects of a mutation using bioinformatics rather than laboratory experiments
Knockdown The deliberate reduction in expression of a gene (e.g. in an experiment to see what it does)
Knockout The deliberate obliteration of expression of a gene (e.g. in a mouse model)
Ligation Joining together of fragments of DNA
Loss‐of‐function mutation A mutation that reduces or obliterates the function of a gene
Missense mutation A single base change in the DNA sequence that alters an amino acid in the protein but does not
truncate it
Monoallelic expression Expression of only one allele instead of two. This occurs with X inactivation and with imprinted genes
Mosaicism The presence of two or more genotypes in an organism arising from a single zygote
Mutant or minor allele frequency The proportion of a sample or a population affected by a mutation
Mutation A change in the DNA sequence that is thought or known to be pathogenic. Non‐synonymous
mutations are those that alter the amino acid code in an exon whereas synonymous mutations do
not
Nonsense mutation A mutation that truncates the protein product
Novel A mutation that has not previously been described in the literature or in public databases
Oligonucleotide A short synthetic sequence of DNA or RNA bases
Overexpression Increased expression of a gene, usually deliberately to analyse the effects
Panel/targeted panel A selected set of genes for DNA sequencing
Polymorphism A change in the sequence of DNA that is commoner than 1% frequency in a population, and
therefore considered to be a variant rather than pathogenic
Post‐zygotic A mutation that occurs in utero
Segregation The co‐occurrence of a phenotype in members of a family who are affected by a particular genotype,
and the absence of that phenotype in unaffected family members
Sequencing Elucidation of the sequence of the bases in DNA or RNA
Single nucleotide polymorphism (SNP) or variant A single base pair change in DNA that is seen in more than 1% of a normal population
(SNV)
Somatic mutation A mutation within a tissue
Splice site A sequence of DNA that indicates the junction of an exon and intron
Sporadic A disorder that does not appear to be inherited or passed on
Transcription The process of making RNA from a DNA template
Translation The process of making protein from an RNA template
Uniparental disomy (UPD) The inheritance of two copies of a gene or genes from one parent
Validation The use of secondary or functional tests to confirm a DNA mutation

into messenger ribonucleic acid (mRNA) and mRNA is The genetic code of DNA occurs in the form of four
translated into protein. Proteins are the molecules in each chemicals or ‘bases’, namely cytosine (C), thymine (T),
cell that perform the vital functional tasks required in the guanine (G) and adenine (A). Complementary pairing
human body. It is the chemical properties of the amino occurs between C bases and G via three hydrogen bonds,
acids and the exact order in which they are aligned that and between T with A via two hydrogen bonds, at the
causes the protein to fold into a particular shape, and centre of the famous DNA double helix [1,2]. Long runs of
therefore determines its functional properties. these four bases are grouped into regions known as exons
38 Section 1  Development, Structure and Physiology of the Skin

and introns. Exons are known to be the regions that ‘code’ or disrupted, and on a larger scale there can be chromo-
STRUCTURE AND PHYSIOLOGY

for RNA and therefore for protein products, and they somal structural aberrations such as translocations and
SECTION 1: DEVELOPMENT,

make up only around 1% of the whole DNA sequence. inversions.


Introns make up the rest of the sequence and their func- In many cases a disease manifests itself because the
tion is still not completely understood. Early on in the flow of molecular information from DNA to RNA to pro-
study of molecular genetics they were considered to be tein contains a mutation, usually originating from the
‘junk DNA’, however it is now well established that DNA code. Many diseases can be described as ‘dominant’
intronic sequences are crucial for the functioning and con- or ‘recessive’ in inheritance pattern, where either one or
trol of the exonic areas and their protein products. The both copies of the gene are required to be faulty respec-
four bases of the exons and their flanking sequences are tively in order to cause the resultant disease. As males
‘transcribed’ into RNA by the process of transcription. In have one copy of the X chromosome, all genes in males
this process an RNA molecule is made from the DNA that do not lie on the pseudoautosomal regions of the X/Y
sequence transcript, with the intronic sequences removed, chromosomes cannot be described as heterozygous or
and the thymine base of DNA replaced by the uracil base homozygous and in this situation are referred to as
of RNA. The four RNA molecule bases (UCGA) can be ‘hemizygous’. X‐linked disorders in general are therefore
arranged into groups of three in 64 different ways, called more likely to be severe in males as there is only one copy
codons. Sixty one of these codons code for one of 20 amino of the gene.
acids, and three code for a so‐called stop codon. This is A disease is described as monogenic if there is only one
known as the genetic code, and the excess of codons for gene involved in the phenotype; however this is a rapidly‐
amino acids is known as the redundancy or degeneracy of disappearing concept, as the background genotype of the
the genetic code. Amino acids are the building blocks patient will almost inevitably have some phenotype‐
of proteins, and stop codons tell the protein when the modifying effect in any disease situation. Polygenic disor-
exonic instructions are finished. The process of forming a ders are those in which more than one gene is known to
string of amino acids from the RNA code is known as be involved in the phenotype. Mosaic disorders are dealt
translation. with in Section 23.
DNA mutations can occur anywhere in the human
genome, and in some cases lead directly to or contribute ­References
to disease. Broadly speaking the most damaging muta- 1 Watson JD, Crick FH. Molecular structure of nucleic acids; a structure
for deoxyribose nucleic acid. Nature 1953;171:737–8.
tions are found within exons, however they are increas-
2 Watson JD, Crick FH. Genetical implications of the structure of deox-
ingly being discovered outside coding regions, for yribonucleic acid. Nature 1953;171:964–7.
example in important regulatory regions outside the
gene itself such as the promoter. Mutations can be clas-
­ onsent for genetic testing and
C
sified into (i) point mutations, similar to a spelling mis-
incidental findings
take where a single base is replaced by another, and (ii)
insertions or deletions, where part of the DNA message Individual hospitals usually have their own standards
is missing or a part is added. Point mutations can be and protocols for obtaining consent for genetic testing.
subdivided into synonymous or non‐synonymous and, This will vary depending on the method to be used, and
where non‐synonymous, into missense or nonsense. whether the test is a diagnostic laboratory test or being
Synonymous mutations are base changes that do not done on a research basis. Oral consent for genetic testing
change the amino acid which is coded for and therefore may be sufficient for established diagnostic tests, but any
the protein product remains the same. These are usually research testing should have fully informed written con-
but not always benign, and therefore usually without sent recorded in the hospital and research notes.
apparent functional consequence. Non‐synonymous In most diagnostic tests the issue of incidental find-
mutations are those that alter the amino acid code in an ings does not arise as only a single gene is being tested.
exon. Within this group missense mutations are those However, with the increase in gene panel testing and of
that change the amino acid but not to a stop codon, clinical exome testing there may be genes being tested
producing a protein, although this protein is abnormal which are known to affect clinical outcome in other
in sequence. Nonsense mutations on the other hand ­diseases, such as tumour suppressor genes or onco-
lead to an absent or truncated protein product by chang- genes. In these cases, there is no international consen-
ing the codon to a stop codon. For mutations that are sus as yet for whether specific consent needs to be
deletions or insertions of sequence this can be ′in‐frame’ taken in advance, however, a gradual consensus is
or leading to a ′frameshift’. In‐frame insertions or dele- emerging, and in general it is good practice to inform
tions lead to one codon being changed, either to a dif- families that there is a possibility that other mutations
ferent codon or a stop codon, and frameshift alter the may be uncovered which could affect their health in
reading frame of the gene – in other words the three‐ other areas. Ideally there is a specific option after
letter codons are no longer read in the correct groups of informed consent for the patient/family to choose
three, which leads to a nonsense protein product and whether they would like to be told of incidental find-
sometimes to a premature stop codon. Large deletions, ings. Recent US and European publications in this area
duplications and insertions can also occur at gene level, may help with consideration of issues around consent
causing whole exons or multiple exons to be removed for incidental findings [1–6].
Chapter 2  Molecular Genetics in Paediatric Dermatology 39

­References result as reported should be inserted into the patient’s


1 Evans BJ. Minimizing liability risks under the ACMG recommenda-

STRUCTURE AND PHYSIOLOGY


medical notes, as the exact mutation may be extremely
tions for reporting incidental findings in clinical exome and genome

SECTION 1: DEVELOPMENT,
sequencing. Genet Med 2013;15:915–20. important. Reading the results of a DNA sequence muta-
2 Green RC, Berg JS, Grody WW et al. ACMG recommendations for tion is explained in Table 2.2.
reporting of incidental findings in clinical exome and genome
sequencing. Genet Med 2013;15:565–74.
3 Hehir‐Kwa JY, Claustres M, Hastings RJ et al. Towards a European ­What to do with a genetic result
consensus for reporting incidental findings during clinical NGS
testing. Eur J Hum Genet 2015;23:1601–6. In general terms it is best to refer the family to Clinical
4 May T. On the justifiability of ACMG recommendations for report- Genetics for the result to be given. Even if the paediatric
ing of incidental findings in clinical exome and genome sequencing.
J Law Med Ethics 2015;43:134–42. dermatologist feels confident of their knowledge in the
5 Anderson JA, Hayeems RZ, Shuman C et al. Predictive genetic testing area, they are not trained in genetic counselling and may
for adult‐onset disorders in minors: a critical analysis of the arguments not be aware of all the implications for all family mem-
for and against the 2013 ACMG guidelines. Clin Genet 2015;87:301–10.
6 Kalia SS, Adelman K, Bale SJ, et al. Recommendations for reporting of
bers. In referral of the family, mention that the whole fam-
secondary findings in clinical exome and genome sequencing, 2016 ily may need to attend, and certainly both parents where
update (ACMG SF v2.0): a policy statement of the American College of possible, and warn that further testing may be required.
Medical Genetics and Genomics. Genet Med 2017;19:249–55. Clinical geneticists can also deal with the issues surround-
ing incidental findings where relevant.
­ hich samples to take for genetic
W
testing ­Concept of personalized medicine
Blood DNA A rapidly evolving area of science is that of personalized
Methods of DNA extraction from whole blood can vary medicine, or precision medicine [1,2]. This is a broad term
dependent on the local laboratory, and it is always worth to describe the use of genetic data on an individual to tai-
checking before obtaining a sample. Very commonly lor the treatment of a disorder to that individual. There
1–5 mL is sufficient from children, collected into an EDTA‐ are two principal ways in which this research is being
containing vial. The sample is safe overnight at room tem- driven. First, genetic information is being analysed retro-
perature if need be. spectively from cohorts of patients who have had success-
ful or unsuccessful responses to therapy to create a model
Cheek swab or saliva DNA of genotypic association with outcomes including side‐
Increasingly it is possible to get good‐quality DNA from effects. Second, where disorders are found to be caused
a cheek swab, however this will also depend on the local by or driven by a specific mutation, researchers and clini-
laboratory. These samples require a specialized swab (not cians are prescribing therapies targeted to these mutations
a normal skin swab), and instructions for collection or to the known effects of these mutations. In paediatric
should be followed. Generally lower quantities of DNA dermatology this is currently being used in trials such as
are obtainable than from blood. The method is, however, of AKT1 inhibitors in Proteus syndrome, rapamycin in the
particularly suitable for young babies if it is difficult to PIK3CA‐related overgrowth spectrum, and collagen VII
obtain a blood sample, or from family members who are
not able to attend clinic to have blood taken, when a cheek Table 2.2  Terminology used in genetic testing results
swab or saliva sample can be sent remotely by following
the manufacturer’s instructions. _ (Underscore) is a range (e.g. c.76_78delACT)
> Indicates a substitution at DNA level (e.g. c.76A>T)
Skin DNA c. Position of base pair in cDNA
Skin biopsy is required for DNA extraction where it is c.(83G=/83G>C) Describes the two genotypes of a mosaic case
del Indicates a deletion (e.g. c.76delA)
possible that the mutation is not present in the germline
dup Indicates a duplication (e.g. c.76dupA)
and therefore not detectable in blood but only in affected fs Frameshift
tissue. A skin biopsy for DNA extraction must not be put fs*# *# Indicates at which codon position the new
into formalin. It can be transported fresh to the laboratory reading frame ends in a stop codon (*). The
on a saline‐soaked gauze, or snap‐frozen in liquid nitro- position of the stop in the new reading frame is
gen, or placed in a medium that stabilizes nucleic acids. calculated starting at the first amino acid that is
Most diagnostic laboratories when presented with a skin changed by the frameshift, and ending at the first
stop codon (*#)
sample for DNA extraction will culture fibroblasts from it
g. Position of base pair in genomic DNA
by default, and then extract DNA from the fibroblasts. This ins Indicates an insertion (e.g. c.76_77insG)
may not be appropriate in a mosaic disorder as the muta- inv Indicates an inversion (e.g. c.76_83inv)
tion may not be present in that cell type, and therefore DNA m. Position in mitochondrial DNA
extraction from whole skin should be specifically requested. n. Position in noncoding RNA
p. Position of amino acid in protein
p.(Arg97Profs*23) Frameshifting change with arginine‐97 as the first
­How to interpret genetic results affected amino acid, changing into a proline, and the
new reading frame ending in a stop at position 23
Once a clinical or research test has been ordered it is use- r. Position in RNA (e.g. r.76a>u)
ful to be able to understand the result. The full genetic
40 Section 1  Development, Structure and Physiology of the Skin

replacement therapy in recessive dystrophic epidermoly- of complementary sequence in the gene of interest.
STRUCTURE AND PHYSIOLOGY

sis bullosa. This personalized medicine approach will Polymerase chain reaction (PCR) is then performed to
SECTION 1: DEVELOPMENT,

change the face of medicine and is likely to improve amplify DNA fragments followed by dideoxynucleotide
patient outcome in response to drug therapies, reducing chain termination sequencing to obtain a sequencing
the requirement for multiple therapies and avoidable readout that can be compared to the known ‘wild‐type’
side‐effects. reference version. An example is shown in Fig. 2.1.

­References Next‐generation sequencing (NGS) of DNA [3]


1 Ashley EA. Towards precision medicine. Nat Rev Genet 2016;
NGS is the best way to sequence DNA on a large scale.
17:507–22.
2 Collins DC, Sundar R, Lim JS, Yap TA. Towards precision medicine
Sequencing that would previously have taken years
in the clinic: from biomarker discovery to novel therapeutics. Trends can now be done in around a week by hand or 3 days
Pharmacol Sci 2017;38:25–40. when using a robotic automated system. Over the years
there have been different technologies for NGS, and
there are still a wide variety of methods available.
­Molecular genetics techniques
However, the principles behind NGS are similar in all
Key reviews are referenced for further reading. cases, and there are also certain major categories of
experiment. These categories vary in how the DNA of
Karyotype interest is captured:
A karyotype test is used to determine chromosomal num- 1 Whole exome: all predicted coding regions of the genome
ber, structure and integrity. The test has to be performed are sequenced.
on actively dividing live cells, and therefore the speci- 2 Whole genome: all DNA regardless of its known function
mens need to be freshly delivered to the laboratory. is sequenced.
Karyotyping can be performed on blood or skin. Single 3 Targeted capture: only preselected DNA is enriched for
nucleotide polymorphism (SNP) arrays are increasingly and sequenced. This is often in the form of a set of
replacing karyotyping for the assessment of chromosomal known and/or candidate genes added to a screening
number and integrity (detection of duplications and dele- panel or it could be a region on a particular chromo-
tions); however if structural rearrangements are to be some. This is the most commonly used type within a
detected (for example inversions or translocations) then diagnostic context, where only known genes are being
karyotyping is still required. investigated. NGS panels are now validated in many
paediatric dermatology diseases [4–7].
Mutation identification by DNA sequencing There are a number of different protocols available
All methods of DNA sequencing rely on the existence of depending on the concentration of DNA collected, for
what is known as the reference sequence, which is the example a total concentration of 3 μg or as low as 200 ng.
accepted and curated version of the human genome. There This is extremely helpful if there are only small quantities
are various version of the reference sequence, and new of material to begin with. Recently adapted protocols
‘builds’ of these become available at intervals of a few have been optimized to obtain good‐quality sequencing
years, when the current version is updated with the latest data from DNA extracted from formalin‐fixed paraffin‐
data. The reference sequence can be found at various web- embedded (FFPE) blocks. This is advantageous as it
sites such as the University of California Santa Cruz means archived pathology samples can be analysed from
(UCSC) genome browser, https://genome.ucsc.edu. The skin samples (as well as other tissues). This has previ-
production of the reference sequence was made possible as ously been difficult as DNA extracted from FFPE blocks is
a result of the revolutionary Human Genome Project [1]. often of lower quality and often more fragmented than a
fresh sample.
Sanger sequencing How NGS works:
Sanger sequencing is a technique of sequencing DNA that 1 A ‘library’ is prepared by randomly fragmenting a
was discovered by Frederick Sanger and coworkers [2]. genomic DNA sample.
The method is to design numerous pairs of oligonucleo- 2 Ligation adapters are then added to both ends of each
tide DNA ‘primers’ that cover the coding region of the DNA fragment.
gene of interest. Primers are essentially small pieces of 3 Adapter‐ligated fragments are PCR amplified and
DNA of around 20 base pairs in length that bind to DNA subjected to purification.

#70 #80
G C T G C G G G T C C G C G T G C C C A C C A C
C G A C G C C C A G G C G C A C G G G T G G T G

#130 #130
Fig. 2.1  A typical Sanger sequencing trace after importing the data into an appropriate analysis software tool. Bases are represented in different colours.
Chapter 2  Molecular Genetics in Paediatric Dermatology 41

4 At this stage the library is loaded onto a flow cell so that 7 Data analysis is then carried out by sequence‐read

STRUCTURE AND PHYSIOLOGY


the DNA fragments can hybridize to the surface of the alignment to a selected reference genome with the use

SECTION 1: DEVELOPMENT,
flow cell via surface‐bound oligonucleotides that are of specialized software. After alignment, any differ-
complementary in sequence to the attached adapters. ences between patient samples and the reference
5 Each bound fragment goes through an amplification genome are identified (Fig. 2.2).
process to produce a cluster that becomes clonally If hunting for new genes on a research basis, the ideal
identical. scenario is to find gene variants in all/most of the affected
6 Sequencing reagents, including a differently labelled individuals that are not present in the reference genome
fluorescent nucleotide for each base, are used to iden- and other control populations and, if looking for an inher-
tify each base. ited disease, are present in the parental DNA where the

Library preparation Cluster amplification

Genomic DNA

Fragmentation
Row cell

Adapters
Bridge amplification
cycles
Ligation

Sequencing
library
2 4
1 3

Clusters
NGS library is prepared by fragmenting a gDNA sample and Library is loaded into a flow cell and the fragments hybridize
ligating specialized adapters to both fragment ends. to the flow cell surface. Each bound fragment is amplified into
a clonal cluster through bridge amplification.

(a) (b)

Sequencing Alignment & data anaylsis

A
T
C
2 4 G
1 3
ATGGCATTGCAATTTGACAT
TGGCATTGCAATTTG
AGATGGTATTG
Reads GATGGCATTGCAA
Sequencing cycles
GCATTGCAATTTGAC
ATGGCATTGCAATT
AGATGGCATTGCAATTTG
T A
G C
Reference
Digital image AGATGG TATTGCAATTTGACAT
genoma
Data is exported to an output file

Cluster 1 > Read 1: GAGT...


Cluster 2 > Read 2: TTGA...
Cluster 3 > Read 3: CTAG...
Cluster 4 > Read 4: ATAC... Text file

Sequencing reagents, including fluorescently labelled nucleo- Reads are aligned to a reference sequence with bioinformatics
tides, are added and the first base is incorporated. The flow software. After alignment, differences between the reference
cell is imaged and the emission from each cluster is recorded. genoma and the newly sequenced reads can be identified.
The emission wavelength and intensity are used to identify
the base. This cycle is repeated ”n” times to create a read
length of “n” bases.

(c) (d)
Fig. 2.2  Overview of the workflow of an Illumina next‐generation sequencing (NGS) method. Source: Courtesy of Illumina, Inc.
42 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

p15.31 p15.1 p14.1 p13.2 p12 q11.2 q12.2 q13.2 q14.1 q14.3 q15 q21.2 q22.2 q23.2 q31.1 q31.3 q33.1 q34 q35.2

41 bp
140,207,950 bp 140,207,960 bp 140,207,970 bp 140,207,980 bp

T
T
C

C
C

Typical representation of the output of NGS, with 'reads' represented as grey horizontal bands aligned with the reference
genome build using Integrative Genomics Viewer (Broad Institute). Each of these is the equivalent to one of the Sanger
sequencing traces shown in Figure 2.1. A heterozygous mutation is demonstrated by the blue cytosine change.

(e)
Fig. 2.2  (Continued)

inheritance can be explained. However, we also know Copy number changes


that diseases can be sporadic and mutations can appear Copy number mutations or copy number variations
de novo so this has to be considered during the analysis (CNVs) can be challenging to identify when using DNA
phase. sequencing methods, especially if they occur in heterozygous
At this point there is still a requirement for DNA varia- form. CNVs occur if a copy of all or part of a gene is either
tions within candidate genes to be validated by Sanger deleted or duplicated by one or more copies. There are a
sequencing. It is always important to ensure that any number of different methods by which to identify CNVs.
result is not due to an NGS artefact that has happened by
chance. Array techniques [8–14]
One preferred method of detection is array‐comparative
Other types of NGS genomic hybridization (aCGH). In simple terms aCGH
In the last few years NGS protocols have been adapted to has RNA oligonucleotides or ‘oligos’ spaced at specific
perform sequencing of RNA in a process known as RNA‐ distances along the human genome that bind DNA
seq. This has the huge advantage of being able to analyse complementary in sequence. In each case a patient DNA
the sequence of alternatively spliced mRNA transcripts. sample under investigation is labelled with a red fluo-
Splicing is the editing of an early mRNA into a mature rescent dye and a reference DNA sample is labelled
form, where introns (noncoding DNA) are removed and with a different dye which fluoresces green. Differentially
exons (protein‐coding parts of DNA) are joined together. labelled samples are mixed 50 : 50 and then hybridized
In this manner a single gene can give rise to different pro- to the array surface. After a time of hybridization, sam-
teins depending on the composition of the exons ligated ples can be analysed for CNVs. In areas of normal copy
together during the splicing stage. Different tissues of the number, equal binding of red and green labelled DNA
body can contain differentially spliced forms of the same is assumed. However, in cases of a patient deletion the
mRNA. RNA‐seq can capture this information and addi- software would detect more or all green fluorescence
tionally gather quantitative information on how highly a compared with red in a genomic location depending on
particular transcript is expressed at a given time point. the presence of a homozygous or heterozygous dele-
There are different types of RNA that have specialized tion. Conversely, in the case of a patient duplication
biological functions and, depending on the precise meth- there would be a higher detection of red fluorescence
ods used, small RNAs, microRNA (miRNA) and transfer than green, depending on the number of duplicated
RNA (tRNA) as well as total RNA can be analysed using copies (Fig. 2.3).
RNA‐seq. SNP DNA arrays can also be used to read copy number.
Other types of NGS include chromatin immunopre- These work by fragmentation of the patient DNA and
cipitation sequencing (CHIP‐seq), which extracts data labelling with fluorescent dye, and then hybridization
on protein interactions with DNA (DNA‐binding sites), onto a solid surface peppered with allele‐specific oligo-
and methylated DNA immunoprecipitation (MeDIP‐seq), nucleotide probes. These probes are allele‐specific at sites
which extracts data on DNA methylation. of known common variation in the population, and are
Chapter 2  Molecular Genetics in Paediatric Dermatology 43

Ratio plot 143.39 Mbp. 9q34.3, no genes, no CNVs


4.00

STRUCTURE AND PHYSIOLOGY


3.75

SECTION 1: DEVELOPMENT,
3.50
3.25
3.00
2.75
2.50
2.25
2.00
1.75
1.50
1.25
1.00
0.75
0.50
0.25
0.00
–0.25
–0.50
–0.75
–1.00
–1.25
–1.50
–1.75
–2.00
–2.25
–2.50
–2.75
–3.00
–3.25
–3.50
–3.75
–4.00
24.3
24.2
24.1

23
22.3
22.1
21.3
21.2
21.1
13.3
13.2
13.1
12
11.2
11.1

12

13
21.11
21.12
21.13
21.2
21.31
21.32
21.33
22.1
22.31
22.32
22.33
31.1
31.2
31.3
32
33.1
33.2
33.3
34.11
34.13
34.3
Fig. 2.3  Array comparative genomic hybridization ratio plot depicting a deletion of a large part of chromosome 9p (shown in red), and two small areas of
closely related deletion/duplication (red and green). The chromosomal bands are laid out at the bottom of the figure.

therefore able to give information not only on copy (cDNA). cDNA contains all the relevant information on
number but also on genotype at these loci, and on large expression level and is preferable to work with as it is
areas of homozygosity or hemizygosity. more stable than mRNA and therefore less likely to
degrade during the handling process.
Multiplex ligation‐dependent probe It is now possible to extract mRNA directly from blood
amplification [15] using specialized kits, however this is only useful if the
Often the results of aCGH require validation by another gene of interest is expressed in white blood cells and this
method; the gold standard here is multiplex ligation‐ is not always the case. Luckily for dermatologists, skin
dependent probe amplification (MLPA). There are com- can be easily biopsied. Different cell types such as fibro-
panies that provide validated and optimized panels with blasts and keratinocytes can be extracted from a fresh skin
probes annealing at many points along a certain gene. biopsy and then cultured to obtain a large number of cells
MLPA works on a multiplex PCR basis after specific to work with. Additionally, the skin biopsy as a whole can
probes have annealed to the designated DNA regions. be used to extract mRNA, resulting in mRNA from all the
Using specialized analysis software the designed probes cell populations in the skin in one sample.
can identify CNVs in the gene under investigation. A Two methods are used, differing in how the experiment
schematic showing the interpretation of MLPA results is technically works: an intercalated dye method such as
explained in Fig.  2.4. MLPA is the preferred method for SYBR® green, or a probe method such as TaqMan® where
diagnostic reporting where pathological CNVs are known a fluorescent reporter dye is analysed. Both obtain data
to be causative of a particular disease. on the basis that the amount of cDNA (equating to the
original relative amounts of mRNA) can be quantified as
Quantitative real‐time PCR [16] each round of PCR cycle causes the amount of cDNA to
Expression analysis of a particular gene can be extremely double during the exponential amplification phase. By
useful for validating involvement in a particular disease, comparing to the expression of a gene that is expressed to
and quantitative real‐time PCR (qRT‐PCR) is often used. the same level in all cells, known as a ‘housekeeping
Put simply, this works by obtaining mRNA and using the gene’, such as glyceraldehyde‐3‐phosphate dehydroge-
retroviral enzyme reverse transcriptase to convert RNA nase (GAPDH) or beta‐actin (ACTB), the expression level
back to DNA to a form known as complementary DNA of the gene of interest can be calculated. For example, if
44 Section 1  Development, Structure and Physiology of the Skin

2.5
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

1.5
Peak ratio

0.5

100 150 200 250


(a) Size (bps)

2.5

1.5 Fig. 2.4  Schematic representation of multiplex


Peak ratio

ligation‐dependent probe amplification (MLPA) data.


Blue squares on the right are reference probes showing
1 normal copy number over different chromosomes.
Squares on the left indicate probes for a particular
gene. (a) In this case the gene under investigation
0.5 shows probes in red and above the cut‐off for normal
copy number (depicted by the green lines). This
indicates a duplication of the whole gene. (b) Shows
0 partial deletion of the same gene as five probes are in
green and below the normal cut‐off, but the rest of the
100 150 200 250 gene is of normal copy number as shown by three blue
(b) Size (bps) squares within the lines.

the gene of interest has a significantly lower level of


expression or lacks detectable expression in patient sam-
ples compared to control samples, this can provide fur-
ther evidence that the mutation is causative of the disease
under investigation.

Fluorescence in situ hybridization [17]


Fluorescence in situ hybridization (FISH) is another
method that can be used for copy number variations of
genes or indeed whole chromosomes. This works via the
attachment of fluorescently labelled probes to a specified
chromosomal location in an individual cell where the
chromosomes can be visualized microscopically. Fig. 2.5
shows the presence of three copies of chromosome 21,
which is the karyotype of an individual with Down syn-
drome, in a cell during metaphase of cell division.

Genome‐wide association studies [18]


Sometimes it is extremely challenging to find specific
gene candidates associated with a disease. This is espe-
cially the case for disorders that are likely to be polygenic. Fig. 2.5  Fluorescence in situ hybridization showing three copies of
In these cases a genome‐wide association study (GWAS) chromosome 21 during metaphase as depicted by the red fluorescent probe
may be helpful. This works by analysing the genotype of in a single cell of an individual with Down syndrome. The two control green
a set of patients with a particular condition, for example probes are hybridized to chromosome 13. Chromosomes are stained in blue.
Chapter 2  Molecular Genetics in Paediatric Dermatology 45

psoriasis, compared to a control cohort without clinical creating double‐stranded RNA. Owing to a n ­atural

STRUCTURE AND PHYSIOLOGY


signs or a family history of psoriasis. By genotype in this mechanism within cells this double‐stranded RNA is then

SECTION 1: DEVELOPMENT,
example we are interested in single base variations at degraded by the cell. This therefore prevents translation
locations throughout the genome known as SNPs. If there of the RNA into protein, and effectively stops or reduces
is an association with a particular set of SNPs in the dis- expression of the protein. This molecular genetic tech-
ease cohort compared to the control cohort, this may give nique is therefore useful for studying the effects of
valuable information regarding areas of the genome that removal of the protein product from the cell, and by infer-
require further investigation. ence can help with working out what that gene usually
does. Alternatively, siRNA has been suggested to be used
Other commonly‐encountered techniques as therapy for diseases in paediatric dermatology where a
Polymerase chain reaction (PCR) mutation is leading to overexpression of a protein. This is
PCR is a technique whose discovery revolutionized therefore a type of gene therapy for skin disease and is
molecular biology [19,20], earning its inventor Kary being researched in paediatric dermatology conditions.
Mullis a Nobel Prize in 1993. It forms the basis of many
other techniques in molecular genetics. It exploits a class ­References
of enzymes known as DNA polymerases, which will copy 1 Lander ES, Linton LM, Birren B et  al. Initial sequencing and
double‐stranded DNA from single‐stranded DNA. The analysis of the human genome. Nature 2001;409:860–921.
function of PCR is to selectively amplify one area of the 2 Sanger F, Coulson AR. A rapid method for determining sequences
in DNA by primed synthesis with DNA polymerase. J Mol Biol
genome, such as the area carrying a mutation of interest, 1975;94:441–8.
so that large quantities of DNA just from that area are pro- 3 Goodwin S, McPherson JD, McCombie WR. Coming of age: ten
duced. Taq polymerase is combined with the DNA tem- years of next‐generation sequencing technologies. Nat Rev Genet
2016;17:333–51.
plate (from a patient), two specific oligonucleotide
4 Lai‐Cheong JE, McGrath JA. Next‐generation diagnostics for
primers which are designed by the researcher to flank inherited skin disorders. J Invest Dermatol 2011;131:1971–3.
either side of the area of interest, and other reagents 5 Takeichi T, Liu L, Fong K, et al. Whole‐exome sequencing improves
including base pairs for construction of new DNA. The mutation detection in a diagnostic epidermolysis bullosa laboratory.
Br J Dermatol 2015;172:94–100.
reaction involves multiple rounds of increasing the tem- 6 Bhoj EJ, Yu Z, Guan Q et al. Phenotypic predictors and final diagnoses
perature to around 96° C to cause double‐stranded tem- in patients referred for RASopathy testing by targeted next‐generation
plate DNA denaturation (separation of the two strands), sequencing. Genet Med 2017;19:715–8.
lowering of the temperature (variable) to allow the prim- 7 Scott CA, Plagnol V, Nitoiu D et al. Targeted sequence capture and
high‐throughput sequencing in the molecular diagnosis of ichthyosis
ers to stick to their relevant complementary strands, and and other skin diseases. J Invest Dermatol 2013;133:573–6.
increasing the temperature again to 72° C which is the 8 Rodriguez‐Revenga L, Mila M, Rosenberg C et  al. Structural
optimum temperature for Taq polymerase function. This variation in the human genome: the impact of copy number variants
on clinical diagnosis. Genet Med 2007;9:600–6.
process is usually repeated 35 times and the production of 9 Lee C, Iafrate AJ, Brothman AR. Copy number variations and
DNA proceeds exponentially. The DNA produced is clinical cytogenetic diagnosis of constitutional disorders. Nat Genet
known as the PCR product, and is used then in Sanger 2007;39:S48–S54.
sequencing, genotyping or cloning. 10 Carter NP. Methods and strategies for analyzing copy number

variation using DNA microarrays. Nat Genet 2007;39:S16–S21.
11 Shen Y, Wu BL. Microarray‐based genomic DNA profiling technol-
Methods for genotyping specific base ogies in clinical molecular diagnostics. Clin Chem 2009;55:659–69.
pair changes 12 Barnes MR. Genetic variation analysis for biomedical researchers: a
There are various methods used for genotyping specific primer. Methods Mol Biol 2010;628:1–20.
13 Dumanski JP, Piotrowski A. Structural genetic variation in the

base pair changes that avoid the need to sequence the ­context of somatic mosaicism. Methods Mol Biol 2012;838:249–72.
DNA. These rely on the difference in the sequence at the 14 Hall IM, Quinlan AR. Detection and interpretation of genomic
particular base pair in question, either by using a restric- structural variation in mammals. Methods Mol Biol 2012; 838:225–48.
tion enzyme which recognizes one of the alleles (wild‐ 15 Schouten JP, McElgunn CJ, Waaijer R et al. Relative quantification
of 40 nucleic acid sequences by multiplex ligation‐dependent probe
type or mutant), or by recognition of the differing amplification. Nucleic Acids Res 2002;30:e57.
properties of the two alleles during changes of tempera- 16 Mocellin S, Rossi CR, Pilati P et al. Quantitative real‐time PCR: a
ture. Examples of such genotyping methods are high‐ powerful ally in cancer research. Trends Mol Med 2003;9:189–95.
17 Trask BJ. Gene mapping by in situ hybridization. Curr Opin Genet
resolution melt PCR (HRM), restriction fragment length Dev 1991;1:82–7.
polymorphisms (RFLPs) and amplification‐refractory 18 Visscher PM, Brown MA, McCarthy MI, Yang J. Five years of GWAS
mutation system (ARMS). discovery. Am J Hum Genet 2012;90:7–24.
19 Mullis K, Faloona F, Scharf S et al. Specific enzymatic amplification
of DNA in vitro: the polymerase chain reaction. Cold Spring Harb
Small interfering RNA [21] Symp Quant Biol 1986;51:263–73.
Small interfering RNAd (siRNA) are synthetic RNA 20 Saiki RK, Gelfand DH, Stoffel S et  al. Primer‐directed enzymatic
­oligonucleotides which are designed to be complemen- amplification of DNA with a thermostable DNA polymerase.
Science 1988;239:487–91.
tary to the RNA transcript of a DNA sequence of interest. 21 Sarkies P, Miska EA. Small RNAs break out: the molecular cell
When the DNA is transcribed to RNA in the presence of the biology of mobile small RNAs. Nat Rev Mol Cell Biol 2014;15:
siRNA, the siRNA binds to the specific RNA transcript, 525–35.
46 

CHA PTER 3
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

Cutaneous Microbiome
Carrie C. Coughlin1 & William H. McCoy IV2
 Division of Dermatology, Department of Medicine, and Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
1

 Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
2

Introduction, 46 Methods of collection and analysis, 50 Fungal microbiota, virome and parasites, 54
Evolution of the cutaneous microbiome, 47 Cutaneous diseases and the microbiome, 51 Conclusion, 55

Abstract This chapter investigates the composition of the microbiome


throughout childhood, interactions of the microbiome with the skin
Bacteria, fungi and viruses comprise the cutaneous microbiome. barrier and immune system, and microbial contributions to health
Exposures starting in utero shape our initial microbiome, which then and disease. Current skin microbiome research related to several
evolves over time through exposure to external microbial communi- common diseases (atopic dermatitis, acne, psoriasis) suggests that
ties (delivery method, close contacts, environment) and changes in incorporation of this information may be beneficial in disease treat-
our skin’s biochemical nature (adrenarche, hygiene, skin disease). ment, thus underscoring the need for further studies in this field.

Key points • Different skin environments (sebaceous, moist, dry) harbour


distinct microbial communities.
• The composition of the cutaneous microbiome changes from
• The cutaneous microbiome encompasses bacteria, viruses, fungi
infancy to adulthood.
and parasites living on and in the skin.
• Microbes provide targets to influence disease states, as well as
• The microbiome has many different influences, and starts
substrates of potential therapies.
developing in utero.

Introduction (human β‐defensins [HBDs] and cathelicidins), and


The cutaneous microbiome made up of bacteria, fungi, adaptive responses, with the latter potentially depend­
parasites and viruses evolves over time and differs by ent on the commensal microbiota [4]. These host–
body site. Epithelial bacteria on skin and mucosal sur­ microbe interactions are the subject of investigations
faces have been shown to be important contributors to into the effect of the microbiome on cutaneous disease
health and illness [1–3]. This chapter will discuss changes states (see, for example, Atopic dermatitis). Conceivably,
in cutaneous bacterial microbiota from infancy to adoles­ patients with impaired immune systems may have dif­
cence. Specifically, we will explore its establishment, ferent responses to the bacteria they encounter and
general composition, changes in response to external and different microbiome compositions than those with
internal pressures, variations with disease states, interac­ intact immune systems. To  investigate the microbiota
tions with the gut microbiome, and relation to the virome of patients with primary immunodeficiencies, research­
and fungal microbiota. ers have studied primary immunodeficiency patients
Though the classification of many types of bacteria has with eczematous dermatitis and compared them to
changed with the availability of new research techniques, patients with atopic dermatitis. Patients with primary
there are four phyla and a subset of their representative immunodeficiency had decreased site‐to‐site variation
genera that are commonly referenced in discussions of the of bacteria [5]. These authors have postulated that ‘eco­
cutaneous microbiome (Table 3.1). Familiarity with these logical permissiveness” may contribute to the recurrent
bacteria helps frame discussions of their contributions to infections of patients with primary immunodeficiencies [5].
cutaneous health and disease. To understand how these interactions occur and change
Bacteria on the skin play an important role in host over time, we will examine how a human infant is colo­
immunity, affecting both skin barrier function and nized by microbes and how this microbial community
immunological defences. These defences involve both is  moulded by the host, environment and exposures
innate mechanisms, such as antimicrobial peptides (Fig. 3.1; see Table 3.2 for definitions of terminology).

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 3  Cutaneous Microbiome 47

Table 3.1  Commonly encountered cutaneous bacteria

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Phyla Gram staining Common cutaneous genera

Actinobacteria Gram positive Actinomyces


Corynebacterium
Cutibacterium
Micrococcus
Propionibacterium
Bacteroidetes Gram negative Prevotella
Firmicutes Gram positive (predominantly) Enterococcus
Lactobacillus
Staphylococcus
Streptococcus
Proteobacteria Gram negative Escherichia
Pseudomonas

Table 3.2  Common terms used in discussions of the microbiome

Term Definition

General
Metaorganism Microbes and host organism (e.g. human)
Microbiome The microbes and their genes in a particular environment
Microbiota The microbes living in a particular environment
Prebiotics Chemical/nutrient supplementation that selectively stimulates growth/activity of one or more microbes of a
metaorganism to benefit the host organism [43]
Probiotics Microbe supplementation to selectively modify a microbiome of a metaorganism to benefit the host organism [44]
Synbiotics Combinations of prebiotics and probiotics
Virome The viruses living on or with a host
Statistics and methods of identification
Alpha diversity The mean species diversity (Chao1 Index, Simpson’s Diversity, Shannon Index, etc.) in a habitat on a local scale
Beta diversity The differentiation between habitats based on species diversity
Colony‐forming unit (CFU) A measure of viable bacteria or fungi in a sample
Gamma diversity Total species diversity across a landscape (human)
Next‐generation sequencing ‘High‐throughput DNA sequencing’ performed on a variety of platforms. Faster and more comprehensive than the
(NGS) older Sanger sequencing method
Metagenome Set of genes and genomes from a sample. Usually restricted to microbiota
Operational taxonomic units Groups (‘clusters’) of bacteria with similar (often 97% similarity when compared pairwise) 16S rRNA sequences, with
(OTUs) one representative sequence representing the group for analysis
Quantitative PCR (qPCR) Amplification of nucleic acids (DNA or RNA) by polymerase chain reaction that is monitored in real time for quantification
Relative abundance Amount of one organism as compared with other similar organisms.
16S ribosomal RNA (rRNA) Sequencing a portion of a bacterium’s 16S ribosomal subunit to allow identification of the bacterium. This technique
sequencing does not distinguish organism status (alive/dead), unlike culture techniques used to identify bacteria
Relationships between organisms
Colonization Organisms living on/within a host or surface
Commensalism Two separate organisms living in contact with one another where one organism benefits from this interaction without
affecting the other organism
Dysbiosis A change in normal microbial ecology on/inside a host or surface due to microbial community imbalance/
maladaptation leading to disease (opposite of symbiosis)
Mutualism Two separate organisms living in contact with one another where both derive benefit from this interaction
Parasitism Two separate organisms living in contact with one another where one organism benefits from this interaction at the
expense of the other organism
Symbiosis Two separate organisms living in close association with one another (commensalism, mutualism, parasitism)

Evolution of the cutaneous been identified by quantitative PCR (qPCR) from surface


microbiome swabs from the endometrium and upper endocervix [6].
First exposures contribute Further, Lactobacillus and several other species have been
to the microbiome identified in cultured curettings from the endometrium [7].
The dogma that fetuses develop in a sterile environment Interestingly, Propionibacterium spp. have been grown by
has recently been challenged, as bacteria similar to those culture‐based methods from the upper genital tract [6].
found in the vagina (Prevotella and Lactobacillus spp.) have Multiple studies have now shown bacteria in the placenta
48 Section 1  Development, Structure and Physiology of the Skin

Exposure Microbiome Immunity


STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

Prenatal exposure

vs. Immune response


development
Delivery method

‘NATURAL’
microbiome
Home environment

TPN

Nutrition

Microbiome-related
diseases
Hygiene Dysbiosis

Gastrointestinal
• Clostridium difficile colitis
Antibiotics • Inflammatory bowel disease (CD, UC)
ALTERED Cutaneous Mucosal
microbiome • Atopic dermatitis • Bacterial vaginosis
• Acne • Candidiasis
• Psoriasis • Chronic periodontitis
Urbanization
Fig. 3.1  Exposures affecting the human microbiome. Upper panel: exposures proposed to mould the ‘NATURAL’ microbiome of an individual without
clearly favouring development of disease. Lower panel: exposures known to or proposed to perturb established microbial communities to create an
ALTERED microbiome that favours the development of various diseases.

[8–10], but the validity of a ‘placental microbiome’ has bacteria [16]. Samples taken less than 5 minutes after birth
been challenged due to methodological concerns in these from neonates’ skin, oral mucosa and nasopharyngeal
studies, specifically the concern for contamination during aspirate in the study by Dominguez‐Bello et al. showed
sample acquisition [11,12]. Amniotic fluid [8] and meco­ similar microbial composition [15]. This is in contrast to
nium [13,14] also harbour bacteria. Thus, neonates have the diversity that develops later (see the next section,
several potential influences on their microbiome even Neonates and infants). However, in other work, bacteria
before birth. from stool in the first 1–2 days of life were different than
Method of delivery has a clear impact on an infant’s cuta­ those of the oropharynx [16]. Thus, the lower gut flora
neous microbiome. Neonates born via vaginal delivery may be distinct from other sites starting from birth.
have early skin colonization by vaginal flora (Lactobacillus Regardless of the human microenvironment, it appears
and Prevotella spp.), while infants born via caesarean section that initial external exposure has a profound effect on
are colonized by typical skin flora including Staphylococcus, microbial colonization, which may dictate susceptibility
Corynebacterium and Propionibacterium spp. [15]. Further, the to a variety of diseases.
bacterial composition of a mother’s vaginal flora was more A number of studies have demonstrated an association
similar to the microbiome of her vaginally delivered baby between caesarean section delivery and increased risk for
than to other vaginally delivered infants. The uniqueness of allergies [18], asthma [19,20], immunodeficiency [20] and
an infant’s cutaneous microbiome transmitted via vaginal obesity [21] (though there are mixed results in the litera­
delivery was not observed in caesarean deliveries, as there ture [19,22]), thus leading to a recent investigation dem­
was no difference between the skin floras of infants born via onstrating that infants born via caesarean section but
caesarean section [15,16]. While microbial colonization of intentionally exposed to vaginal fluids can be colonized
the skin would seem to be the most pertinent to dermatolo­ by bacteria from these fluids [23]. While this study estab­
gists, the intestinal flora may also affect allergic and inflam­ lishes how a ‘normal’ microbiome can be fostered in an
matory reactions in patients [3,17], thus an examination of infant born via caesarean section through the introduc­
the oropharyngeal and intestinal flora of infants is relevant tion of an available probiotic source, the link between dis­
to paediatric dermatologists. tinct microbiome communities and/or specific bacteria
Much as observed for cutaneous microbial colonization, with inflammatory skin disease requires further study.
the mode of delivery has been shown to dictate oropharyn­ Another external source supporting maintenance and
geal microbial colonization with infants born vaginally expansion of a physiological microbiome is breast milk.
having increased Firmicutes (trending to more Lactobacillus) Diversity of the infant gut microbiome is proportional to
and infants born via caesarean section having increased the amount of daily breast milk intake even after intro­
Actinobacteria (Propionibacterium, notably) and Proteo­ duction of solid foods [24].
Chapter 3  Cutaneous Microbiome 49

The potential links between skin inflammation and cuta­ paralleling the increased sebaceous gland activity seen in

STRUCTURE AND PHYSIOLOGY


neous bacteria include both intrinsic host–microbe and adolescents and adults [35].

SECTION 1: DEVELOPMENT,
extrinsic microbe–microbe interactions. While the intrinsic
link between a specific bacterial strain and a disease state Adults
may seem straightforward (see Atopic dermatitis), extrin­ After the hormonal shifts of adolescence, adults have dis­
sic effects can be less clear. Extrinsic links may include the tinct sebaceous, moist and dry zones on their skin [37,38].
influence of one type of bacteria on the colonization of Researchers have compared the bacterial composition of
[25,26], virulence of [27] and/or host response to [28] an these sites with an aim to inform work on diseases such as
unrelated bacterium. The last link is particularly concern­ acne and atopic dermatitis that tend to have site predilec­
ing in the setting of increasing emergence of multidrug‐ tions [34]. Propionibacteria dominate the microbial com­
resistant organisms, leaving some patients with limited or munities of sebaceous sites, such as the face [38,39]. Of
no therapeutic options. Of note, antibiotic resistance genes note, P. acnes has been reclassified under the genus
carried by maternal bacteria can transfer to neonates and Cutibacterium (C. acnes), which is important when com­
the mode of delivery influences this transfer [14,29]. Thus, paring older and more recent research. Moist locations,
the transfer of bacteria from one host to another can affect such as the axilla and antecubital fossa, harbor corynebac­
the ability of the new host to fight infection. teria and staphylococci [38,39]. Flavobacteria and
Proteobacteria are found at dry sites [38]. Not only the
Neonates and infants dominant phyla, but also the diversity of the microbial
After delivery, environment is the next major influence on community correlates with skin microenvironment, as
a neonate’s microbiome. Broadly, for neonates, environ­ sebaceous sites have the least, and dry sites have the most,
ment can be split into hospital/nursery and home set­ community diversity [38].
tings. Infants requiring prolonged hospital stays have In contrast to the changes seen in the microbiome from
different influences on their microbiota than children dis­ infancy to maturation, recent work shows the adult cuta­
charged to home [30]. Incubators, cribs, bedding, moni­ neous microbiome is relatively constant over time [34].
tors, leads, tubes, catheters, gloves, nutrition, medications, Bacterial communities are maintained by the host, rather
caregivers, family and more all contribute to the environ­ than reacquired over time, and remain particular to the
ment encountered by a hospitalized infant. Due to individual. Cutaneous sites with lower diversity (such as
changes in the epidermal barrier over time, the effects of the face) were more stable than sites with higher diversity
these influences can also change. Infant skin evolves in (such as the feet). Moreover, left/right symmetry of the
structure and barrier function over weeks to months cutaneous microbiota has been noted [39,40], which could
[31,32]. Consideration of these exposures is important have implications for interventional trials. However,
when seeing neonates in the hospital, as their microbial studies looking specifically at the axillae show differences
flora can be quite different from that encountered in the in bacteria at left versus right sites, with handedness
outpatient setting. potentially influencing these results [41,42]. The microbi­
The cutaneous microbiome starts to change within the ome differences revealed through studies of life stage and
first 3 months of life and continues to evolve over the first body site have begun to reveal important considerations
year of life, as shown by Capone et  al. [33]. Diversity for therapeutic targets for diseases associated with per­
increases and, in contrast to the uniformity in oral and turbations in the microbiome (see Cutaneous diseases
skin samples at birth [15], skin flora evolves to be site‐ and the microbiome).
specific. This evolution also is in contrast to the relative
stability of the adult microbiome over time (see Adults) References
[34]. Firmicutes (specifically staphylococci and strepto­ 1 Grice EA. The intersection of microbiome and host at the skin inter­
face: genomic‐ and metagenomic‐based insights. Genome Res 2015;
cocci) predominate, in contrast to the Proteobacteria
25:1514–20.
dominance in adults [33]. Of note, the colonization seen 2 Johnson CL, Versalovic J. The human microbiome and its potential
by Capone et al. was not influenced by mode of delivery importance to pediatrics. Pediatrics 2012;129:950–60.
of the infant, even in the samples from infants 1–3 months 3 Lim ES, Wang D, Holtz LR. The bacterial microbiome and virome
milestones of infant development. Trends Microbiol 2016;24:801–10.
of age. Thus, an infant’s initial cutaneous microbiome 4 Naik S, Bouladoux N, Wilhelm C et al. Compartmentalized control of
(dictated by mode of delivery) may dissipate by 1–3 months skin immunity by resident commensals. Science 2012;337:1115–9.
of age and yet still have lifelong effects (immunity, skin 5 Oh J, Freeman AF, Program NCS et  al. The altered landscape of the
maturation, barrier function). human skin microbiome in patients with primary immunodeficiencies.
Genome Res 2013;23:2103–14.
6 Mitchell CM, Haick A, Nkwopara E et  al. Colonization of the upper
Preadolescents and adolescents genital tract by vaginal bacterial species in nonpregnant women. Am J
Bacterial colonization continues to evolve from early Obstet Gynecol 2015;212:611 e1–9.
childhood through adolescence. Young children have 7 Cowling P, McCoy DR, Marshall RJ et al. Bacterial colonization of the
non‐pregnant uterus: a study of pre‐menopausal abdominal hysterec­
increased microbial diversity compared to adolescents, tomy specimens. Eur J Clin Microbiol Infect Dis 1992;11:204–5.
and their cutaneous bacterial communities resemble 8 Collado MC, Rautava S, Aakko J et al. Human gut colonisation may be
those of infants more than adults (Firmicutes predomi­ initiated in utero by distinct microbial communities in the placenta and
amniotic fluid. Sci Rep 2016;6:23129.
nate, in contrast to Actinobacteria of adolescents and
9 Stout MJ, Conlon B, Landeau M et  al. Identification of intracellular
adults) [35,36]. Furthermore, older children have more bacteria in the basal plate of the human placenta in term and preterm
lipophilic bacteria on their skin than younger children, gestations. Am J Obstet Gynecol 2013;208:226 e1–7.
50 Section 1  Development, Structure and Physiology of the Skin

10 Aagaard K, Ma J, Antony KM et  al. The placenta harbors a unique 37 Findley K, Oh J, Yang J et al. Topographic diversity of fungal and bac­
microbiome. Sci Transl Med 2014;6(237):237ra65. terial communities in human skin. Nature 2013;498:367–70.
STRUCTURE AND PHYSIOLOGY

11 Lauder AP, Roche AM, Sherrill‐Mix S et al. Comparison of placenta 38 Grice EA, Kong HH, Conlan S et  al. Topographical and temporal
SECTION 1: DEVELOPMENT,

samples with contamination controls does not provide evidence for a diversity of the human skin microbiome. Science 2009;324:1190–2.
distinct placenta microbiota. Microbiome 2016;4:29. 39 Costello EK, Lauber CL, Hamady M et al. Bacterial community variation
12 Kliman HJ. Comment on “the placenta harbors a unique microbiome”. in human body habitats across space and time. Science 2009;326:1694–7.
Sci Transl Med 2014;6:254le4. 40 Gao Z, Perez‐Perez GI, Chen Y, Blaser MJ. Quantitation of major
13 Gosalbes MJ, Llop S, Valles Y et al. Meconium microbiota types domi­ human cutaneous bacterial and fungal populations. J Clin Microbiol
nated by lactic acid or enteric bacteria are differentially associated 2010;48:3575–81.
with maternal eczema and respiratory problems in infants. Clin Exp 41 Callewaert C, Kerckhof FM, Granitsiotis MS et al. Characterization of
Allergy 2013;43:198–211. Staphylococcus and Corynebacterium clusters in the human axillary
14 Gosalbes MJ, Valles Y, Jimenez‐Hernandez N et al. High frequencies region. PloS One 2013;8:e70538.
of antibiotic resistance genes in infants’ meconium and early fecal 42 Egert M, Schmidt I, Hohne HM et al. rRNA‐based profiling of bacteria
samples. J Dev Orig Health Dis 2016;7:35–44. in the axilla of healthy males suggests right‐left asymmetry in bacte­
15 Dominguez‐Bello MG, Costello EK, Contreras M et  al. Delivery rial activity. FEMS Microbiol Ecol 2011;77:146–53.
mode shapes the acquisition and structure of the initial microbiota 43 Gibson GR, Roberfroid MB. Dietary modulation of the human colonic
across multiple body habitats in newborns. Proc Natl Acad Sci U S A microbiota: introducing the concept of prebiotics. J Nutr 1995;125(6):
2010;107:11971–5. 1401–12.
16 Brumbaugh DE, Arruda J, Robbins K et al. Mode of delivery deter­ 44 Fuller R. Probiotics in man and animals. J Appl Bacteriol 1989;66(5):
mines neonatal pharyngeal bacterial composition and early intestinal 365–78.
colonization. J Pediatr Gastroenterol Nutr 2016;63:320–8.
17 West CE, Ryden P, Lundin D et  al. Gut microbiome and innate
immune response patterns in IgE‐associated eczema. Clin Exp Allergy Methods of collection and analysis
2015;45:1419–29.
18 Papathoma E, Triga M, Fouzas S, Dimitriou G. Cesarean section When evaluating a microbiome investigation, it is impor­
delivery and development of food allergy and atopic dermatitis in
early childhood. Pediatr Allergy Immunol 2016;27:419–24.
tant to review the experimental design of the authors, as
19 Cuppari C, Manti S, Salpietro A et al. Mode of delivery and risk for this field lacks standardization, leading to varying results
development of atopic diseases in children. Allergy Asthma Proc from different investigators. Identifying the sample
2015;36:344–51. source is the first step in this process, as it will dictate the
20 Sevelsted A, Stokholm J, Bonnelykke K, Bisgaard H. Cesarean section
and chronic immune disorders. Pediatrics 2015;135:e92–8. overall microbial community being assessed. As reviewed
21 Rutayisire E, Wu X, Huang K et al. Cesarean section may increase earlier in the chapter, the skin microbiome community
the risk of both overweight and obesity in preschool children. BMC profiles of the three main skin environments (sebaceous,
Pregnancy Childbirth 2016;16:338.
moist, dry) are quite distinct [1,2]. Recent work has also
22 Robson SJ, Vally H, Abdel‐Latif ME et al. childhood health and devel­
opmental outcomes after cesarean birth in an Australian cohort. shown that fungal colonization is affected by site with
Pediatrics 2015;136:e1285–93. feet harbouring more diversity than the rest of the body
23 Dominguez‐Bello MG, De Jesus‐Laboy KM, Shen N et al. Partial resto­ [3]. Cutaneous disease at the sites sampled can also affect
ration of the microbiota of cesarean‐born infants via vaginal microbial
transfer. Nat Med 2016;22:250–3.
the microbial community even if that is not the focus of
24 Pannaraj PS, Li F, Cerini C et  al. Association between breast milk the investigation.
bacterial communities and establishment and development of the While body site can have a profound effect on the study,
infant gut microbiome. JAMA Pediatr 2017;171:647–54. the method of skin sampling (cutaneous swab, scrape and
25 Bomar L, Brugger SD, Yost BH et  al. Corynebacterium accolens
releases antipneumococcal free fatty acids from human nostril and biopsy) has generally been found to be equivalent on a
skin surface triacylglycerols. mBio 2016;7:e01725–15. ‘phylotype level’ (operational taxonomic units, OTUs)
26 Christensen GJ, Scholz CF, Enghild J et  al. Antagonism between between samples when examining skin microbes [4].
Staphylococcus epidermidis and Propionibacterium acnes and its
genomic basis. BMC Genomics 2016;17:152.
However, more recent work has found differences in
27 Lo CW, Lai YK, Liu YT et al. Staphylococcus aureus hijacks a skin bacteria isolated from the epidermis, dermis and adi­
commensal to intensify its virulence: immunization targeting beta‐ pose tissue [5]. Unlike pilosebaceous units, the dermis
hemolysin and CAMP factor. J Invest Dermatol 2011;131:401–9. and subcutis are not in direct communication with the
28 Cogen AL, Nizet V, Gallo RL. Skin microbiota: a source of disease or
defence? Br J Dermatol 2008;158:442–55. skin surface, and demonstration of varied bacterial com­
29 Alicea‐Serrano AM, Contreras M, Magris M et al. Tetracycline resist­ munities in different levels of biopsy specimens broadens
ance genes acquired at birth. Arch Microbiol 2013;195:447–51. the potential impact of sampling technique. When look­
30 Hartz LE, Bradshaw W, Brandon DH. Potential NICU environmental
ing at the acne microbiome, an alternative method to
influences on the neonate’s microbiome: a systematic review. Adv
Neonatal Care 2015;15:324–35. these three commonly used sampling techniques is the
31 Evans NJ, Rutter N. Development of the epidermis in the newborn. use of pore strips to selectively sample pilosebaceous
Biol Neonate 1986;49:74–80. units [6]. Further, hair‐specific studies have sampled indi­
32 Nikolovski J, Stamatas GN, Kollias N, Wiegand BC. Barrier function
and water‐holding and transport properties of infant stratum corneum
vidual follicular units (‘plucked hairs’) and found higher
are different from adult and continue to develop through the first year amounts of human papillomavirus (HPV) than skin biop­
of life. J Invest Dermatol 2008;128:1728–36. sies [7]. Beyond reviewing sample site and acquisition,
33 Capone KA, Dowd SE, Stamatas GN, Nikolovski J. Diversity of the critically assessing sample preparation and the use of
human skin microbiome early in life. J Invest Dermatol 2011;131:
2026–32. appropriate negative controls (background microbial signal)
34 Oh J, Byrd AL, Park M et  al. Temporal stability of the human skin is critically important, as contaminants can skew results.
microbiome. Cell 2016;165:854–66. The sensitivity of next‐generation sequencing (NGS) tech­
35 Oh J, Conlan S, Polley EC et al. Shifts in human skin and nares micro­
biota of healthy children and adults. Genome Med 2012;4:77.
niques can often reveal microbial signal from even classi­
36 Shi B, Bangayan NJ, Curd E et al. The skin microbiome is different cally ‘sterile’ techniques, such as sterile cotton swabs used
in pediatric versus adult atopic dermatitis. J Allergy Clin Immunol for skin swabs (personal data). Cleaning and sterilizing the
2016;138:1233–6. skin to avoid introducing surface contaminants when
Chapter 3  Cutaneous Microbiome 51

assessing the microbiome of deeper structures and changing difference in staphylococcal colonization at other sites

STRUCTURE AND PHYSIOLOGY


gloves between sample sites can be crucial. (popliteal fossa, nasal tip, cheeks) and no difference at

SECTION 1: DEVELOPMENT,
Beyond sample collection and basic preparation, the other time points in the first year of life. Older patients with
techniques used to analyse the microbial community AD are more often Staphylococcus aureus carriers [2,3].
structure should be considered as well. While culture‐ Staph. aureus can produce toxins that affect mast cell
based methods previously dominated the literature, they degranulation and drive atopic disease, thus mechanisti­
have now largely taken a secondary role. Investigators cally linking the presence of bacteria and the maintenance
are using more powerful techniques, such as 16S ribo­ of disease [2]. In addition, Staph. aureus is associated with
somal DNA (rDNA) sequencing and whole‐genome Th2 inflammation and immunoglobulin E (IgE) response [4].
shotgun (WGS) metagenomic sequences. NGS uses The benefit of commensal bacteria is exemplified by data
hypervariable rDNA regions to differentiate OTUs instead that Staph. epidermidis has counter‐regulatory effects on
of the full rDNA sequence, as was used in classical Sanger cutaneous inflammation [4] and can interfere with Staph.
sequencing. The choice of which hypervariable region to aureus biofilms [5].
use as a proxy for the sequence can affect the data, as In an attempt to utilize bacteria to modify inflamma­
the V1–V3 region was found to be more accurate (closer tion in patients with AD, one group has investigated
to whole metagenomic data) for skin samples than V4, applying lysates of Vitreoscilla filiformis, a bacterium found
which is more helpful in gut microbiome studies [8,9]. in some thermal springs, directly to the skin in a com­
Further, species discrimination for various microbes may pounded cream [6]. Both AD disease activity and pruritus
require particular hypervariable region primer sets or decreased in the intervention group treated with the
may not be able to be resolved using standard 16S rDNA compound versus controls treated with vehicle only. The
NGS. It is important to consider that as techniques evolve, authors then investigated the mechanism behind this
results from studies using different methodologies may improvement using a mouse model, which showed that
no longer be comparable. this treatment resulted in high levels of interleukin (IL)‐10
in dendritic cells via toll‐like receptor (TLR)2 leading
References to increased regulatory T‐cell activity [7]. This study
1 Grice EA, Kong HH, Conlan S et  al. Topographical and temporal proposes that AD therapy could utilize disease‐modifying
diversity of the human skin microbiome. Science 2009;324:1190–2.
bacteria that are not typical cutaneous colonizers or
2 Perez Perez GI, Gao Z, Jourdain R et al. Body site is a more determinant
factor than human population diversity in the healthy skin microbiome. commensals.
PloS One 2016;11:e0151990. Microbial diversity differs between affected and ­normal
3 Findley K, Oh J, Yang J et  al. Topographic diversity of fungal and skin. In patients with AD, microbial diversity decreases at
bacterial communities in human skin. Nature 2013;498:367–70.
4 Grice EA, Kong HH, Renaud G et al. A diversity profile of the human
an affected site during a flare and increases (improves,
skin microbiota. Genome Res 2008;18:1043–50. approaching levels seen in control patients) with treat­
5 Nakatsuji T, Chiang HI, Jiang SB et  al. The microbiome extends to ment [8]. Additionally, even intermittent treatment of
subepidermal compartments of normal skin. Nat Commun 2013;4:1431. a  site before a flare can affect microbial diversity [8].
6 Craft N, Li H. Response to the commentaries on the paper:
Propionibacterium acnes strain populations in the human skin micro­ Moreover, Staph. aureus as well as Staph. epidermidis are
biome associated with acne. J Invest Dermatol 2013;133:2295–7. over‐represented during flares, showing how dysbiosis
7 Schneider I, Lehmann MD, Kogosov V et al. Eyebrow hairs from actinic can contribute to disease.
keratosis patients harbor the highest number of cutaneous human
papillomaviruses. BMC Infect Dis 2013;13:186.
Manipulating the microbiome of these patients is seen
8 Kong HH. Details matter: designing skin microbiome studies. J Invest as a possible new way to treat and/or prevent AD. Dilute
Dermatol 2016;136:900–2. bleach baths are commonly recommended to decrease
9 Meisel JS, Hannigan GD, Tyldsley AS et al. Skin microbiome surveys Staph. aureus counts in children with AD [9–11]. Topical
are strongly influenced by experimental design. J Invest Dermatol
2016;136:947–56. antibiotics such as mupirocin are used to treat infection or
decrease colonization, as well. The effect of topical corti­
costeroids on the microbiome has been investigated more
recently. Early work showed a reduction of Staph. aureus
Cutaneous diseases and the
with topical corticosteroid use [12], and more recently
microbiome
treatment with fluticasone cream was shown to normalize
The contribution of bacteria and fungi to cutaneous the microbiome at lesional sites [13]. As topical corticos­
disease is a booming field of research. Here, we review teroids do not directly target bacteria, this work demon­
the links between atopic dermatitis (AD), acne and psoria­ strates the intimate cross‐talk between an inflammatory
sis to bacterial communities on the skin. dermatosis and the cutaneous flora that can drive disease.
These studies support the use of topical corticosteroids
Atopic dermatitis even on overtly infected lesions.
Bacterial colonization and bacterial/viral superinfection There is an inverse correlation between AD severity
of AD are recognized as challenges in the management and skin colonization with Streptococcus salivarius [14].
of this disease. Interestingly, Kennedy et  al. found that Interestingly, regular use of emollients in infants at risk of
infants without staphylococcal colonization of the antecu­ developing AD was associated with a lower skin pH and
bital fossa at 2 months were more likely to have AD at increased skin colonization with Strep. salivarius [14].
12 months, raising the possibility of early bacteria modu­ These two effects are thought to contribute to the preven­
lating the risk for AD [1]. However, this study showed no tive effect of emollients in high‐risk infants.
52 Section 1  Development, Structure and Physiology of the Skin

The role of dysbiosis in AD has been investigated level is downregulated in C. acnes from acne skin and
STRUCTURE AND PHYSIOLOGY

by  multiple groups leading to attempts to modify the that host supplementation with B12 can further modify
SECTION 1: DEVELOPMENT,

development and course, and/or improve treatment of AD C. acnes transcription of multiple genes possibly leading
through prebiotics [15,16], probiotics [15,17,18] and synbi­ to skin inflammation via increased porphyrin production
otics [19]. One such study is investigating the effect of sup­ [30]. These initial investigations are beginning to illumi­
plementing pregnant women with probiotics to decrease nate the specific role(s) that the cutaneous microbiome
risk of atopy [18]. Unfortunately, these investigations have plays in acne, but further research is required before these
not yielded consistent results nor have they improved care findings can be translated to clinical treatments.
recommendations. At this point, there is not sufficient evi­
dence to recommend routine pre‐/pro‐/synbiotic supple­ Psoriasis
mentation for patients with or at risk for AD. Acute flares of psoriasis can be triggered by infections,
Clothing materials have also been studied as means to including streptococcal infections of the oropharynx or
change the microbiome in patients with AD. For example, perianal skin. In fact, the strength of this association has
chitosan (a biopolymer derived from crustacean shells led to the proposal that the potential life‐threatening
with inhibitory properties against Staph. aureus) was sequelae of streptococcal infections have exerted evolu­
explored as a coating on cotton to alter the microbiome. In tionary pressure to select for the psoriasis phenotype
one study, there was no difference in Staph. aureus between (Fig. 3.2) [31,32], though an alternative evolutionary argu­
participants who slept in chitosan‐coated pyjamas and ment has also been proposed for Mycobacterium leprae
those who did not, but the amount of coagulase‐negative [33]. The potential link between the microbiome and
Staphylococcus (CoNS) increased in the chitosan group [20]. chronic psoriasis is under investigation, with only a
While this might appear to be a negative result, the increased limited number of studies published. Researchers are
CoNS may help to control Staph. aureus (as described previ­ examining psoriasis in relation to both the cutaneous and
ously) and thus decrease disease activity. Additionally, fab­ gut microbiomes at least in part due to the finding that
rics incorporating silver have been investigated for patients with Crohn disease are more likely to develop
treatment of AD. Silver dressings and topical preparations psoriasis. In addition, these diseases have overlapping
have long been used in wound care for their broad antimi­ genetic associations and immune activation pathways,
crobial properties. More recently, fabrics made with silver which has led some authors to suggest that both psoriasis
mesh [21] and cellulose fibres of seaweed‐impregnated sil­ and Crohn’s are related to alterations in immune toler­
ver [22–24] have been shown to decrease microbial counts ance to the microbiota (Fig. 3.2) [31]. This proposed dys­
in patients with AD. Pruritus [24] and disease severity biosis must be differentiated from true infection, as
[21,23,24] also decreased in patients wearing the fabrics. psoriatic lesions have a very low incidence of infection
These studies were performed with small numbers of due to high expression of antimicrobial peptides such as
patients, which limits the ability to generalize the findings. HBDs 2/3 and cathelicidin LL‐37 [31]. Alekseyenko and
Investigations of antimicrobial fabrics are representative of colleagues have recently published microbiome data
ongoing research to further manipulate microbial dysbio­ suggesting that there are distinct cutaneotypes associated
sis, infection and inflammation in AD. with normal skin (cutaneotype 1: Proteobacteria) versus
psoriatic lesional skin (cutaneotype 2: Firmicutes,
Acne Actinobacteria), with the latter carrying an odds ratio for
The predominant bacterium found at sebaceous sites asso­ psoriasis of 3.52 (95% confidence interval [CI] 1.44 to 8.98,
ciated with acne in adults is Cutibacterium acnes (formerly P <0.01). Notably, their work suggested generalized
Propionibacterium acnes) [25,26]. This bacterium has long dysbiosis was present on both lesional and nonlesional
been linked to acne, and the amount of C. acnes recovered skin from psoriatics (decreased alpha diversity), but
from these sites changes over time [27,28]. More recently, that cutaneotype was only informative when sampled
distinct C. acnes ribotypes (RT; a surrogate descriptor for from a lesional site, as nonlesional skin could be either
C. acnes strains) have been significantly associated with cutaneotype [34]. Others have investigated the gut micro­
acne skin (RT4, 5, 8, 10) or normal skin (RT6) [29], which biome link to psoriasis, thinking of it more as a systemic
suggests dysbiosis of the C. acnes community may lead to disease. In a mouse model, gut bacteria can increase
clinically apparent acne. Interestingly, though, younger inflammation and increase the Th17 response of the skin,
children have comedonal acne and more Streptococcus producing psoriasis‐like inflammation [35]. Further,
than C. acnes (personal data), which correlates with their ingestion of a poorly absorbed carbohydrate analogue
tendency to have less sebaceous gland activity. Though (acarbose) in an imiquimod‐based mouse model of
antibiotics have been a mainstay to treat inflammatory psoriasis can reduce skin inflammatory markers (tumour
acne for generations of dermatologists, the increasing necrosis factor [TNF]‐α, IL‐1β, IL‐6, IL‐17A, IL‐22) and
incidence of antibiotic resistance and general movement clinical findings associated with psoriasis [36], thus sug­
to increased antimicrobial stewardship has led some gesting that manipulation of the mouse gut microbiome
researchers to investigate more specific treatment targets. through acarbose can modulate the psoriatic skin pheno­
Understanding the drivers behind bacterial activity type. While this type of research provides evidence for a
may help us better understand acne pathogenesis and link between alterations in both the skin/gut flora and
develop such treatments. To this end, it has been shown cutaneous disease, confirmatory studies are required
that the vitamin B12 synthesis pathway at the mRNA prior to being useful in clinical practice.
Chapter 3  Cutaneous Microbiome 53

Tonsillar microbiota Evolving pathogens


Evolutionary pressure?

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Mycobacterium leprae?
Strep pyogenes?

Microbe–host
interactions
Gene mutations
Skin microbiota CARD14 IL-23 Intestinal microbiota
IL-23 NFκB
NFκB NOD2/CARD15
PGRP-3, -4
PG + TLR2 PG + NOD2
(Staph, Strep) LPS + TLR4

Innate immunity
ACTIVATION

IL-17, IL-23
Immune

IL-23

Adaptive immunity
Th1 cells Th17 cells
IFN-γ IL-17, IL-22

Keratinocyte proliferation
phenotype
Clinical

Psoriasis Crohn disease

Fig. 3.2  Comparison of microbiota influences on psoriasis and Crohn disease. Proposed evolutionary pressures favouring the development of a psoriasis
phenotype and proposed pathways for the triggering of autoimmune disease (psoriasis by the skin microbiota, Crohn’s by intestinal microbiota) in
genetically predisposed individuals. IFN, interferon; IL, interleukin; LPS, lipopolysaccharide; NF, nuclear factor; PG, peptidoglycan; PGRP, PG recognition
protein; Th, T‐helper; TLR, toll‐like receptor. Source: Adapted from Fry et al. 2013 [31], McFadden et al. 2009 [32] and Bassukas et al. 2012 [33].

Research into the importance of the microbiome to cuta­ 8 Kong HH, Oh J, Deming C et al. Temporal shifts in the skin microbi­
ome associated with disease flares and treatment in children with
neous diseases is providing more insight into pathogenesis
atopic dermatitis. Genome Res 2012;22:850–9.
and treatment. Additionally, recent studies are advancing 9 Huang JT, Rademaker A, Paller AS. Dilute bleach baths for
investigations into the relationship between the skin and Staphylococcus aureus colonization in atopic dermatitis to decrease
other organs (the gastrointestinal tract, most commonly). disease severity. Arch Dermatol 2011;147:246–7.
10 Huang JT, Abrams M, Tlougan B et al. Treatment of Staphylococcus
The ideas that ingested prebiotics and probiotics can aureus colonization in atopic dermatitis decreases disease severity.
affect AD, diet influences acne, and microbial‐induced Pediatrics 2009;123:e808–14.
signalling in the gastrointestinal tract can manifest on the 11 Eichenfield LF, Boguniewicz M, Simpson EL et al. Translating atopic
skin as psoriasis are advancing research into novel ways dermatitis management guidelines into practice for primary care pro­
viders. Pediatrics 2015;136:554–65.
to prevent and treat skin disease. 12 Nilsson EJ, Henning CG, Magnusson J. Topical corticosteroids and
Staphylococcus aureus in atopic dermatitis. J Am Acad Dermatol
1992;27:29–34.
References 13 Gonzalez ME, Schaffer JV, Orlow SJ et  al. Cutaneous microbiome
1 Kennedy EA, Connolly J, Hourihane JO et al. Skin microbiome before effects of fluticasone propionate cream and adjunctive bleach baths in
development of atopic dermatitis: Early colonization with commensal childhood atopic dermatitis. J Am Acad Dermatol 2016;75:481–93.e8.
staphylococci at 2 months is associated with a lower risk of atopic 14 Glatz M, Jo JH, Kennedy EA et al. Emollient use alters skin barrier and
dermatitis at 1 year. J Allergy Clin Immunol 2017;139:166–72. microbes in infants at risk for developing atopic dermatitis. PLoS One
2 Nakamura Y, Oscherwitz J, Cease KB et al. Staphylococcus delta‐toxin 2018;13:e0192443.
induces allergic skin disease by activating mast cells. Nature 2013;503: 15 Baquerizo Nole KL, Yim E, Keri JE. Probiotics and prebiotics in der­
397–401. matology. J Am Acad Dermatol 2014;71:814–21.
3 Chng KR, Tay AS, Li C et  al. Whole metagenome profiling reveals 16 Bozensky J, Hill M, Zelenka R, Skyba T. Prebiotics do not influence
skin microbiome‐dependent susceptibility to atopic dermatitis flare. the severity of atopic dermatitis in infants: a randomised controlled
Nat Microbiol 2016;1:16106. trial. PloS One 2015;10:e0142897.
4 Laborel‐Preneron E, Bianchi P, Boralevi F et  al. Effects of the 17 Nermes M, Kantele JM, Atosuo TJ et al. Interaction of orally adminis­
Staphylococcus aureus and Staphylococcus epidermidis secretomes tered Lactobacillus rhamnosus GG with skin and gut microbiota and
isolated from the skin microbiota of atopic children on CD4+ T cell humoral immunity in infants with atopic dermatitis. Clin Exp Allergy
activation. PloS One 2015;10:e0141067. 2011;41:370–7.
5 Iwase T, Uehara Y, Shinji H et  al. Staphylococcus epidermidis Esp 18 Barthow C, Wickens K, Stanley T et  al. The Probiotics in Pregnancy
inhibits Staphylococcus aureus biofilm formation and nasal colonization. Study (PiP Study): rationale and design of a double‐blind randomised
Nature 2010;465:346–9. controlled trial to improve maternal health during pregnancy and pre­
6 Gueniche A, Knaudt B, Schuck E et al. Effects of nonpathogenic gram‐ vent infant eczema and allergy. BMC Pregnancy Childbirth 2016;16:133.
negative bacterium Vitreoscilla filiformis lysate on atopic dermatitis: a 19 Chang YS, Trivedi MK, Jha A et  al. Synbiotics for prevention and
prospective, randomized, double‐blind, placebo‐controlled clinical treatment of atopic dermatitis: a meta‐analysis of randomized clinical
study. Br J Dermatol 2008;159:1357–63. trials. JAMA Pediatr 2016;170:236–42.
7 Volz T, Skabytska Y, Guenova E et al. Nonpathogenic bacteria alleviating 20 Lopes C, Soares J, Tavaria F et  al. Chitosan coated textiles may
atopic dermatitis inflammation induce IL‐10‐producing dendritic cells improve atopic dermatitis severity by modulating skin staphylococcal
and regulatory Tr1 cells. J Invest Dermatol 2014;134:96–104. profile: a randomized controlled trial. PloS One 2015;10:e0142844.
54 Section 1  Development, Structure and Physiology of the Skin

21 Gauger A, Mempel M, Schekatz A et al. Silver‐coated textiles reduce present on infected skin, but it is not typically present on
Staphylococcus aureus colonization in patients with atopic eczema.
STRUCTURE AND PHYSIOLOGY

healthy skin [4]. Researchers have proposed Malassezia as


Dermatology 2003;207:15–21.
SECTION 1: DEVELOPMENT,

22 Hipler UC, Elsner P, Fluhr JW. Antifungal and antibacterial properties the triggering agent for neonatal cephalic pustulosis, but
of a silver‐loaded cellulosic fiber. J Biomed Mater Res B Appl Biomater data are inconclusive at this point [5]. Examining
2006;77:156–63. Malassezia across the lifespan, investigators found changes
23 Fluhr JW, Breternitz M, Kowatzki D et al. Silver‐loaded seaweed‐based
cellulosic fiber improves epidermal skin physiology in atopic derma­
in the proportions of Malassezia species, with M. globosa
titis: safety assessment, mode of action and controlled, randomized predominating after adolescence in Japanese people [6].
single‐blinded exploratory in vivo study. Exp Dermatol 2010;19:e9–15. In Chinese and French patients with dandruff, though,
24 Juenger M, Ladwig A, Staecker S et  al. Efficacy and safety of silver M. restricta is more abundant [7]. Malassezia was found in
textile in the treatment of atopic dermatitis (AD). Curr Med Res Opin
2006;22:739–50. greater amounts and had greater diversity in 1‐month‐
25 Grice EA, Kong HH, Conlan S et  al. Topographical and temporal olds with seborrhoeic dermatitis compared with those
diversity of the human skin microbiome. Science 2009;324:1190–2. without seborrhoea [8]. In this population, M. sympodialis
26 Bek‐Thomsen M, Lomholt HB, Kilian M. Acne is not associated with
yet‐uncultured bacteria. J Clin Microbiol 2008;46:3355–60.
and M. restricta were the dominant species. Thus, similar
27 Leyden JJ, McGinley KJ, Mills OH, Kligman AM. Propionibacterium to previously described bacterial studies, the fungal
levels in patients with and without acne vulgaris. J Invest Dermatol microbiota changes with maturation of the human host,
1975;65:382–4. which requires longitudinal studies to ascertain the role
28 Leyden JJ, McGinley KJ, Mills OH, Kligman AM. Age‐related changes
in the resident bacterial flora of the human face. J Invest Dermatol of various fungal community members in skin disease.
1975;65:379–81. Compared to the bacterial and fungal components of
29 Fitz‐Gibbon S, Tomida S, Chiu BH et al. Propionibacterium acnes the cutaneous human microbiome, work on the cutane­
strain populations in the human skin microbiome associated with
ous human virome is in its infancy. Notably, viruses have
acne. J Invest Dermatol 2013;133:2152–60.
30 Kang D, Shi B, Erfe MC et al. Vitamin B12 modulates the transcrip­ been linked with multiple skin cancers, including HPV
tome of the skin microbiota in acne pathogenesis. Sci Transl Med and squamous cell carcinoma [9], Merkel cell polyomavirus
2015;7:293ra103. and Merkel cell carcinoma [10] and human herpesvirus 8
31 Fry L, Baker BS, Powles AV et al. Is chronic plaque psoriasis triggered
by microbiota in the skin? Br J Dermatol 2013;169:47–52.
(HHV8) and Kaposi sarcoma [11]. While these skin
32 McFadden JP, Baker BS, Powles AV, Fry L. Psoriasis and streptococci: ­cancers can occur in children, more often as paediatric
the natural selection of psoriasis revisited. Br J Dermatol 2009; dermatologists we deal with acute viral infections, such
160:929–37. as those in children with eczema (eczema herpeticum
33 Bassukas ID, Gaitanis G, Hundeiker M. Leprosy and the natural selec­
tion for psoriasis. Med Hypotheses 2012;78:183–90. [HSV] and eczema coxsackium [coxsackie virus, often A6
34 Alekseyenko AV, Perez‐Perez GI, De Souza A et al. Community dif­ or A16]) or those with verrucae (HPV). Interestingly
ferentiation of the cutaneous microbiota in psoriasis. Microbiome enough in the latter case, work on adult skin has revealed
2013;1:31.
35 Zakostelska Z, Malkova J, Klimesova K et  al. Intestinal microbiota
multiple HPV strains on normal skin [4]. While a viral
promotes psoriasis‐like skin inflammation by enhancing Th17 cause has been established for a small subset of skin
response. PloS One 2016;11:e0159539. diseases, far less is known about the normal viral ecology
36 Chen HH, Chao YH, Chen DY et al. Oral administration of acarbose of human skin.
ameliorates imiquimod‐induced psoriasis‐like dermatitis in a mouse
model. Int Immunopharmacol 2016;33:70–82. One reason that viral cutaneous microbiome research
has lagged behind is that the majority of viruses are not
catalogued in genomic databases, thus making compu­
tational analyses of untargeted DNA sequences that
Fungal microbiota, virome
identify unique genomes particularly challenging. Similar
and parasites
to other microbiome efforts, more is currently known
Studies of cutaneous fungal species show site‐specific about the gut virome than the cutaneous virome [12].
diversity similar to that described for bacteria and viruses Recent work has delineated the composition of the cuta­
[1]. Feet have the most diverse fungal community, with neous double‐stranded (ds) DNA virome at multiple sites
Malassezia and many environmental fungi identified [1]. in adults, thus allowing the investigators to investigate
Knowledge of typical fungal species resident on the skin viral diversity and virulence factors [13]. While cutaneous
can aid investigations of infections with opportunistic viromes were found to be stable over a 1‐month time
fungi. For example, Trichosporon asahii is an uncommon period, diversity over this time was present within the
pathogen of immunocompromised/immunosuppressed metagenomes and site diversity was seen [13]. This work
hosts. T. asahii can be found on normal skin, thus provid­ has also provided data allowing the authors to hypoth­
ing a source for this opportunistic infection [2]. Work on esize how the dsDNA virome interacts with other
cutaneous fungal colonization is essential to understand­ members of the microbiota [13]. Delineation of the typical
ing overall cutaneous microbial ecology (bacteria/fungi/ paediatric virome (both healthy and disease‐associated)
viruses), which will advance the treatment of both fungal will be essential as we investigate the causes of both infec­
skin disease and other dermatoses. tious and malignant skin disease.
Similar to bacterial microbiome colonization with par­ Recently there has been more interest in skin ectopara­
turition, colonization of the skin with fungi is present on sites, with Demodex mites implicated by some authors in
day 0 of life with maternal transfer of Malassezia to infants the pathogenesis of adult rosacea, though causation is not
[3]. Studies of the fungal microbiota often centre on confirmed [14–16]. However, in keeping with the thought
Malassezia, a lipophilic yeast, present as a colonizer and in of one microbial community influencing another, Murillo
disease states. Another common yeast, Candida, can be et al. found differences in the local microbiota associated
Chapter 3  Cutaneous Microbiome 55

with Demodex in patients with papulopustular rosacea 10 Feng H, Shuda M, Chang Y, Moore PS. Clonal integration of a polyoma­
virus in human Merkel cell carcinoma. Science 2008;319:1096–100.

STRUCTURE AND PHYSIOLOGY


(less Actinobacteria, and more Proteobacteria and
11 Chang Y, Cesarman E, Pessin MS et al. Identification of herpesvirus‐

SECTION 1: DEVELOPMENT,
Firmicutes) versus patients with erythematotelangiectatic like DNA sequences in AIDS‐associated Kaposi’s sarcoma. Science
rosacea and controls [17]. Demodex is not seen as often on 1994;266:1865–9.
paediatric skin, but immunosuppressed patients (spe­ 12 Lim ES, Wang D, Holtz LR. The bacterial microbiome and virome
milestones of infant development. Trends Microbiol 2016;24:801–10.
cifically those with acute lymphoblastic leukaemia) are 13 Hannigan GD, Meisel JS, Tyldsley AS et al. The human skin double‐
at risk for eruptions triggered by this mite [18,19]. More stranded DNA virome: topographical and temporal diversity,
work is needed to evaluate the potential influence of genetic enrichment, and dynamic associations with the host micro-
Demodex on paediatric rosacea, and its presence on the biome. mBio 2015;6:e01578–15.
14 Holmes AD. Potential role of microorganisms in the pathogenesis of
skin of different immunosuppressed and immunodefi­ rosacea. J Am Acad Dermatol 2013;69:1025–32.
cient patients. 15 Brown M, Hernandez‐Martin A, Clement A et al. Severe demodexfol­
liculorum‐associated oculocutaneous rosacea in a girl successfully
treated with ivermectin. JAMA Dermatol 2014;150:61–3.
References 16 Gupta G, Daigle D, Gupta AK, Gold LS. Ivermectin 1% cream for
1 Findley K, Oh J, Yang J et al. Topographic diversity of fungal and bac­ rosacea. Skin Therapy Lett 2015;20:9–11.
terial communities in human skin. Nature 2013;498:367–70. 17 Murillo N, Aubert J, Raoult D. Microbiota of Demodex mites from
2 Zhang E, Sugita T, Tsuboi R et al. The opportunistic yeast pathogen rosacea patients and controls. Microb Pathog 2014;71‐72:37–40.
Trichosporon asahii colonizes the skin of healthy individuals: analysis 18 Hitraya‐Low M, Ahmad RC, Cordoro KM. Facial eruption in a
of 380 healthy individuals by age and gender using a nested polymer­ 5‐year‐old child with acute lymphoblastic leukemia. Pediatr Dermatol
ase chain reaction assay. Microbiol Immunol 2011;55:483–8. 2016;33:671–2.
3 Nagata R, Nagano H, Ogishima D et  al. Transmission of the major 19 Ivy SP, Mackall CL, Gore L et al. Demodicidosis in childhood acute
skin microbiota, Malassezia, from mother to neonate. Pediatr Int lymphoblastic leukemia; an opportunistic infection occurring with
2012;54:350–5. immunosuppression. J Pediatr 1995;127:751–4.
4 Foulongne V, Sauvage V, Hebert C et al. Human skin microbiota: high
diversity of DNA viruses identified on the human skin by high
throughput sequencing. PloS one 2012;7:e38499.
5 Bergman JN, Eichenfield LF. Neonatal acne and cephalic pustulosis: is Conclusion
malassezia the whole story? Arch Dermatol 2002;138:255–7.
6 Sugita T, Suzuki M, Goto S et al. Quantitative analysis of the cutane­ The cutaneous microbiome is made up of a diverse array
ous Malassezia microbiota in 770 healthy Japanese by age and gender
of bacteria, fungi, viruses and parasites. These micro­
using a real‐time PCR assay. Med Mycol 2010;48:229–33.
7 Wang L, Clavaud C, Bar‐Hen A et  al. Characterization of the major scopic passengers may colonize or infect the skin depend­
bacterial‐fungal populations colonizing dandruff scalps in Shanghai, ing on natural virulence, host maturation or environment
China, shows microbial disequilibrium. Exp Dermatol 2015;24:398–400. pressures. While the general contribution of microbes to
8 Yamamoto M, Umeda Y, Yo A et al. Utilization of matrix‐assisted laser
desorption and ionization time‐of‐flight mass spectrometry for
both healthy and defective skin barriers is well appreci­
identification of infantile seborrheic dermatitis‐causing Malassezia ated in dermatology, specific host–microbe and microbe–
and incidence of culture‐based cutaneous Malassezia microbiota of microbe interactions are still largely unknown. Research
1‐month‐old infants. J Dermatol 2014;41:117–23. into the ‘normal’ microbiome and how dysbiosis contrib­
9 Euvrard S, Chardonnet Y, Pouteil‐Noble C et al. Association of skin
malignancies with various and multiple carcinogenic and noncarcino­ utes to skin disease is leading to seminal investigations
genic human papillomaviruses in renal transplant recipients. Cancer that will undoubtedly change how we treat, and hope­
1993;72:2198–206. fully in the future prevent, cutaneous diseases.
56 

CHA PTER 4
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

Physiology of Neonatal Skin


Peter H. Hoeger
Department of Paediatric Dermatology, Catholic Children’s Hospital Wilhelmstift, Hamburg, Germany

Introduction, 56 Epidermis, 57 Dermis and skin appendages, 61


Vernix caseosa, 56 Transepidermal water loss, 58

Abstract in premature babies leads to potentially life‐threatening water loss


and enhances the risk of infections. It takes more than 12 months
While the skin of the neonate appears to be ‘mature’ with regard for the intraepidermal concentrations of natural moisturizing fac-
to its anatomical appearance, many physiological functions of the tors (NMF) and, hence, the transepidermal water loss (TEWL), to
skin undergo postnatal maturation. Insufficient interlacement normalize. The threshold for eccrine sweating remains elevated
between epidermis and dermis leads to increased susceptibility for months. Conversely, due to stimulation by maternally derived
towards shearing forces. Immaturity of the epidermal lipid barrier ­androgens, sebum production is increased.

Key points maturation in terms of increasing hydration and concentrations


of water‐binding natural moisturizing factors (NMF).
• Vernix caseosa is a mixture of water, protein and lipids whose • In premature babies, transepidermal water loss (TEWL) is
composition reflects epidermal barrier maturation. It protects inversely proportional to gestational age. In very low birthweight
the epidermis from water (amniotic fluid) exposure and has infants (VLBW) TEWL can be as high as 100 g/m2/h which, if
antimicrobial and hydrating properties. not immediately compensated for (by humidified incubators,
• Epidermis and dermis along with its appendages (sweat glands, plastic wraps, topical emollients), is incompatible with life. Poor
sebaceous glands, hair follicles) are fully developed in a term integrity of the stratum corneum puts premature infants at risk
neonate. During the first year of life the skin undergoes functional of mechanical trauma and transcutaneous infections.

­Introduction ­Vernix caseosa


The transition from an aqueous, but sterile, environment During the last trimester of gestation, the fetus is covered
to a dry one rich in pathogens represents a dramatic by a protective biofilm called vernix caseosa. It forms a
challenge to the skin of the newborn infant. Intactness of mechanical ‘shield’ against maceration by amniotic fluid
the epidermal barrier is of utmost importance both for the and bacterial infection. Vernix is composed mainly of
prevention of water loss and for the defence against water (80.5%), proteins and lipids (8–10%) (Fig. 4.1) [1,2].
microorganisms that start to colonize the neonatal skin These lipids are derived from two sources: wax esters
from the moment of birth. The efficacy of this barrier formed in sebaceous glands [3] and epidermal barrier
is  proportional to its thickness and lipid composition. lipids derived from keratinocytes [2]. Vernix contains all
During late gestation, the number of epidermal layers major stratum corneum lipids, including ceramides [4],
and the thickness of the stratum corneum increase with which are not synthesized by sebaceous glands. Similar
fetal age. The extent of transepidermal water loss (TEWL) to postnatal skin, sebum and epidermal lipids apparently
and the risk of infections with skin‐colonizing microor- mix within vernix in order to provide on the fetal skin
ganisms in preterm infants are thus directly proportional surface what has been referred to as the skin surface lipid
to the infant’s degree of prematurity. Although the gross film [5]. Interestingly, the lipid composition of vernix
anatomy of epidermal and dermal structures in neonatal closely resembles that found in fetal skin [2]. Composition
skin is similar to that in older skin (Table 4.1), the process of vernix lipids was found to reflect epidermal barrier
of postnatal maturation and adaptation affects virtually maturation and correlates with gestational age, sex and
all compartments and structures of the skin. maternal diet [6].

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 4  Physiology of Neonatal Skin 57

Table 4.1  A comparison of skin anatomy between preterm and term neonates and older children

STRUCTURE AND PHYSIOLOGY


SECTION 1: DEVELOPMENT,
Premature baby Full‐term neonate Infants/children

Skin thickness (mm) 0.9 1.2 2.1


Diameter of epidermis (μm) 20–25 40–50 >50
Diameter of stratum corneum (μm) (number of layers) 4–5 (5 or 6) 9–10 (≥10–15) 10–15 (≥10–15)
Dermoepidermal junction Flat, no rete ridges Rete ridges start to form Deep rete ridges
Eccrine sweat glands Upper dermis, inactive Upper dermis, barely active Deeper dermis, fully active
Elastic fibres Microfibrils, no elastin Elastic fibre network, immature Elastic fibre network, mature

Water 80.5% ­Epidermis


Proteins 10.3%
The epidermis protects against evaporation, percutaneous
Other lipids 6.4% absorption of toxic substances, physical damage and
microbial infection. These properties depend largely on the
Barrier lipids 2.72% thickness and barrier lipid content of the epidermis, both of
which are directly related to gestational age [1,2]. As shown
Cholesterol 1.1% in Fig. 4.2, the number of epidermal cell layers and, from
about the beginning of the third trimester, the thickness
FFA 0.6%
of the stratum corneum increase progressively with age.
Phospholipids 0.4%
Scanning electron microscopy of the epidermal surface
Ceramides 0.7% showed high anisotropy with irregular corneocyte clusters
in young infants indicating a poorly controlled process of
Fig. 4.1  Composition of human vernix caseosa. FFA, free fatty acid.
Source: Adapted from Pickens et al. 2000 [1] and Hoeger et al. 2002 [2]. corneocyte desquamation while in older children the dis-
tribution of superficial corneocytes was more regular [3].
The most important lipids required for barrier function
Unlike postnatal skin, sebum and keratinocytes are (i.e. ceramides, cholesterol and free fatty acids) are syn-
not shed in the fetal period but adhere to the skin; accu- thesized in the lamellar bodies within the granular layer.
mulation of vernix might thus compensate for the relative There is a patterned succession of epidermal expression of
lack of barrier lipids in fetal skin. Application of vernix to mRNA and of enzymes involved in lipid synthesis preced-
normal adult skin has been shown to increase surface ing the formation of an effective epidermal barrier [4,5].
hydration [7]. Vernix contains antimicrobial peptides, e.g. Among the most important factors regulating the sequence
cathelicidin, lysozyme and lactoferrin, which along with of epidermal differentiation and stratum corneum forma-
free fatty acids provide antimicrobial protection against tion is the peroxisome proliferator‐activated receptor‐α
fungi, bacteria and parasites [8,9]. (PPAR‐α). PPARs are expressed abundantly in early fetal
epidermis; they regulate the activity of key enzymes
required for barrier ontogenesis (e.g. β‐glucocerebrosidase
­References and steroid sulphatase) [5,6]. Similar to what happens in
1 Pickens WL, Warner RR, Boissy YL et  al. Characterization of vernix
maturation of the lung, glucocorticoids, thyroid hormones
caseosa: water content, morphology, and elemental analysis. J Invest
Dermatol 2000;115:875–81. and oestrogens accelerate barrier formation, while andro-
2 Hoeger PH, Schreiner V, Klaassen IA et al. Epidermal barrier lipids in gens retard it [5]. Initiation of skin barrier formation in the
human vernix caseosa: corresponding ceramide patterns in vernix and human fetus starts at around 20–24 weeks’ gestation [7].
fetal epidermis. Br J Dermatol 2002;146:194–201.
3 Stewart ME, Quinn MA, Downing DT. Variability in the fatty acid com- The process of keratinization reveals an interesting tempo-
position of wax esters from vernix caseosa and its possible relation to ral and spatial pattern, starting at and spreading from dis-
sebaceous gland activity. J Invest Dermatol 1982;78:291–5. tinct epidermal initiation sites such as forehead, palms and
4 Oku H, Mimura K, Tokitsu Y. Biased distribution of the branched‐
soles [7,8]. Filaggrin  –  derived from profilaggrin in the
chain fatty acids in ceramides of vernix caseosa. Lipids 2000;35:
373–81. keratohyalin granules – aggregates keratin filaments in the
5 Sheu H‐M, Chao S‐C, Wong T‐W et al. Human skin surface lipid film: stratum corneum. Small molecules known as natural mois-
an ultrastructural study and interaction with corneocytes and inter- turizing factor (NMF) are proteolytically cleaved from
cellular lipid lamellae of the stratum corneum. Br J Dermatol
1999;140:385–91.
filaggrin. They are responsible for hydration and plasticity
6 Checa A, Holm T, Sjödin MOD et al. Lipid mediator profile in vernix (flexibility) of the skin surface and help to prevent TEWL
caseosa reflects skin barrier development. Sci Rep 2015;5:15740. [9]. Forty percent of the NMF are formed by hygroscopic
7 Bautista MIB, Wickett RR, Visscher MO et  al. Characterization of free amino acids such as urocanic acid; their concentration
vernix caseosa as a natural biofilm: comparison to standard oil‐based
ointments. Pediatr Dermatol 2000;17:253–60. at birth is extremely low, and increases slowly over the first
8 Akinbi HT, Narendran V, Pass AK et  al. Host defense proteins in months in parallel with stratum corneum hydration [10,11].
vernix caseosa and amniotic fluid. Am J Obstet Gynecol 2004; Intraepidermal concentrations of the pro‐inflammatory
191:2090–6.
9 Tollin M, Bergsson G, Kai‐Larsen Y et al. Vernix caseosa as a multi‐
cytokine interleukin 1‐alpha (IL‐1α) are higher in infants
component defence system based on polypeptides, lipids and their than in adults, and higher in premature than in mature
interactions. Cell Mol Life Sci 2005;62:2390–9. infants. Its release is stimulated by the rapid transition
58 Section 1  Development, Structure and Physiology of the Skin
STRUCTURE AND PHYSIOLOGY
SECTION 1: DEVELOPMENT,

(a)

(b)

(c) (d)
Fig. 4.2  Embryonic, fetal and neonatal skin: (a) at 13 weeks’ gestation; (b) at 18 weeks’ gestation; (c) at 25 weeks’ gestation; (d) in a mature neonate.

from high to low humidity at birth, and in response to epi- 80


dermal damage in order to induce the recovery process
[12,13]. The same applies to the antimicrobial peptides
60
(human β‐defensins [HBD] and cathelicidins) which are
TEWL (g/m2/hr)

reduced in neonates [14] while the antimicrobial proteins


lysozyme and lactoferrin are present in neonatal epider- 40
mis at levels five times higher than in adults [15].

20
­Transepidermal water loss
The intactness of the epidermal barrier can be assessed by
0
measuring the TEWL. The TEWL is proportional to the
0 5 10 15 20
vapour pressure gradient measured with an evaporimeter
Postnatal age (days)
[16,17]. It is influenced by gestational age, site and ambi-
ent humidity [16–18]. In term neonates, the TEWL ranges Fig. 4.3  The effect of gestational age on transepidermal water loss
(TEWL). Serial measurements from abdominal skin in 17 infants of 25–29
from 4 to 8 g/m2/h. This is slightly lower than in adults
weeks’ gestation. Shaded bar represents TEWL in term infants. Source:
[19] owing to the fact that eccrine sweating is low or absent Cartlidge and Rutter 1998 [18]. Reproduced with permission of Elsevier.
in the newborn infant. In the premature infant, TEWL is
inversely proportional to gestational age (Fig. 4.3). In very
immature infants (24–26 weeks’ gestation), it can be as along a water vapour gradient, it can be prevented by rais-
high as 100 g/m2/h, which means that these infants, if left ing the ambient humidity. It is now common practice to
in a dry atmosphere, could lose 20–50% of their body humidify incubators for premature babies, particularly
weight within 24 hours. This degree of TEWL would rap- those of less than 32 weeks’ gestation [20]. Humidity needs
idly lead to hypernatraemia, polyglobulia and hypother- to be as high as 80–90% within the first days in order to
mia, resulting eventually in periventricular haemorrhage prevent heat and fluid loss. Pre­vention of hypothermia
and death. As TEWL represents passive diffusion of water and TEWL can also be ascertained by using polyethylene
Chapter 4  Physiology of Neonatal Skin 59

caps or wraps immediately after delivery [21]. Randomized infants should therefore be adequately increased during

STRUCTURE AND PHYSIOLOGY


controlled trials in Pakistan and India where incubators phototherapy. Neonatal epidermis can easily be damaged

SECTION 1: DEVELOPMENT,
are not readily available have demonstrated that postnatal (e.g. by removal of plastic adhesives), which induces a
application of topical emollients (sunflower seed oil, coco- measurable disruption of the skin barrier function [29].
nut oil or mineral oils [petrolatum]) can (i) reduce TEWL Interestingly, air exposure leads to acceleration of post-
in VLBW preterm babies [22] and (ii) improve skin integ- natal barrier maturation. As depicted in Fig. 4.3, TEWL in
rity and reduce the risk of bloodstream infections in pre- most premature infants approaches that of term infants
term infants [23] if started immediately after birth [24]. within 10–15 days. Studies in rodents have shown that
The effectiveness of this approach in primary care settings this functional maturation is paralleled by an increase in
needs to be further explored since a meta‐analysis of pre- stratum corneum thickness, the number of lamellar bod-
vious reports could not demonstrate benefits of topical ies in stratum granulosum cells and the barrier lipid con-
emollients with respect to infection rate and mortality [25]. tent of the stratum corneum [30,31]. In ultra‐low‐birthweight
There is a striking regional variability on the skin surface infants (23–25 weeks of gestational age), this process can,
regarding TEWL; it is usually highest through the abdomi- however, take significantly longer [32]. As demonstrated
nal skin, where maturation of the epidermal barrier occurs recently, even in mature babies it takes up 12 months until
latest [7,8]. Preterm infants nursed under a radiant heater TEWL normalizes to levels seen in older children and
exhibit higher rates of evaporation because the level of adults; this process is paralleled by a constant increase of
ambient water vapour is lower [26]. It is likewise increased NMF levels within the epidermis [33].
(by 20%) during phototherapy, even if relative humidity
and ambient temperature are tightly controlled; this is Skin surface pH
probably caused by increased dermal blood flow during Acidification of the skin surface is effected by acidic compo-
phototherapy [27,28]. Maintenance fluid intake of preterm nents in the sweat, sebum and horny layer (Fig. 4.4a) [34].

7.5 140

7.0 120

6.5 100
Corneometer units

6.0 80
pH

5.5 60

5.0 40

4.5 20

4.0 0
3 30 90 3 30 90
(a) RZ Din (b) Ra

160 30

140
25
120
20
100

80 15
μm

μm

60
10
40
5
20

0 0
3 30 90 3 30 90
(c) Age (days) (d) Age (days)
Fig. 4.4  Development of skin surface parameters in neonates and young infants (n = 180 healthy neonates). (a) Skin surface pH. (b) Stratum corneum hydration (both
measured on the frontal area). (c and d) Microtopography (parameters of skin roughness). Rz Din, mean depth of roughness; Ra, arithmetic mean surface roughness.
60 Section 1  Development, Structure and Physiology of the Skin

Three classes of molecules are considered to be the most 15 Walker VP, Akinbi HT, Meinzen‐Derr J et al. Host defense pro-
teins on the surface of neonatal skin: implications for innate
STRUCTURE AND PHYSIOLOGY

likely sources of protons in the epidermis [35]: some


immunity. J Pediatr 2008;152:777.
SECTION 1: DEVELOPMENT,

amino acids and filaggrin‐derived breakdown products 16 Hammarlund K, Sedin G, Stromberg B. Transepidermal water loss
such as urocanic acid and pyrrolidone carboxylic acid; in newborn infants. VIII. Relation to gestational age and postnatal
α‐hydroxy acids such as lactic acid; and acidic lipids age in appropriate and small for gestational age. Acta Paediatr
Scand 1983;72:721–8.
such as cholesterol sulphate and free fatty acids. At 17 Hammarlund K, Sedin G. Transepidermal water loss in newborn
birth, neonates exhibit a characteristic neutral or alkaline infants. III. Relation to gestational age. Acta Paediatr Scand 1979;
skin surface pH of 6.2–7.5 [27,28]. In both term [36] and 68:795–801.
preterm infants [37], the pH declines rapidly in the first 18 Cartlidge PHT, Rutter N. Skin barrier function. In: Polin RA, Fox WW,
eds. Textbook of Fetal and Neonatal Physiology, 2nd edn. Philadelphia:
week, and slowly thereafter up to the fourth week of life, W.B. Saunders, 1998:771–88.
when a range of 5.0–5.5 is reached, which is similar to 19 Cunico RL, Maibach HI, Khan H et al. Skin barrier properties in the
that in older children and adults [34–39]. Acidity of the newborn. Biol Neonate 1977;32:177–82.
20 Harpin VA, Rutter N. Humidification of incubators. Arch Dis Child
stratum corneum facilitates homeostasis of colonizing 1985;60:219–24.
commensal bacteria and inhibits replication of pathogenic 21 McCall EM, Alderdice F, Halliday HL et al. Interventions to prevent
bacteria and fungi. hypothermia at birth in preterm and/or low birthweight infants.
Cochrane Database Syst Rev 2010;3:CD004210.
22 Nangia S, Paul VK, Deorari AK et  al. Topical oil application and
Stratum corneum hydration and skin transepidermal water loss in preterm very low birthweight
infants – A randomized trial. J Trop Pediatr 2015;61:414–20.
roughness 23 Salam RA, Darmstadt GL, Bhutta ZA. Effect of emollient therapy on
Neonatal skin is relatively dry and rough compared clinical outcomes in preterm neonates in Pakistan: a randomised con-
with that of older infants (Fig. 4.4b–d) [38–40]. Stratum trolled trial. Arch Dis Child Fetal Neonatal Ed 2015;100:F210–15.
24 Darmstadt GL, Ahmed S, Ahmed ASMN, Saha SK. Mechanism
corneum hydration and skin roughness are correlated
for prevention of infection in preterm neonates by topical emol-
[34,38]. In healthy term neonates, corneal layer hydration lients: a randomized, controlled clinical trial. Pediatr Infect Dis J
increases and skin roughness decreases proportionate to 2014;33:1124–7.
age. The skin surface of the newborn is rather hydropho- 25 Cleminson J, McGuire W. Topical emollient for preventing infection
in preterm infants. Cochrane Database Syst Rev 2016;(1):CD001150.
bic, which limits epidermal adsorption of water [41]. The 26 Kjartansson S, Arsan S, Hammarlund K et al. Water loss from the skin
heat loss caused by evaporation of amniotic fluid from of term and preterm infants nursed under a radiant heater. Pediatr
the skin of the newborn is thus minimized. Res 1995;37:233–8.
27 Maayan‐Metzger A, Yosipovitch G, Hadad E et  al. Transepidermal
water loss and skin hydration in preterm infants during phototherapy.
­References Am J Perinatol 2001;18:393–6.
1 Evans NJ, Rutter N. Development of the epidermis in the newborn. 28 Grünhagen DJ, de Boer MGJ, de Beaufort AJ et  al. Transepidermal
Biol Neonate 1986;49:74–80. water loss during halogen spotlight phototherapy in preterm infants.
2 Fairley JA, Rasmussen JE. Comparison of stratum corneum thickness Pediatr Res 2002;51:402–5.
in children and adults. J Am Acad Dermatol 1983;8:652–4. 29 Lund C, Nonato LB, Kuller JM et al. Disruption of barrier function in
3 Fluhr JW, Lachmann N, Baudouin C et al. Development and organi- neonatal skin associated with adhesive removal. J Pediatr 1997;131:
zation of human stratum corneum after birth: electron microscopy 367–72.
isotropy score and immunocytochemical corneocyte labelling as 30 Hanley K, Jiang Y, Elias PM et al. Acceleration of barrier ontogenesis
epidermal maturation’s markers in infancy. Br J Dermatol 2014; in vitro through air exposure. Pediatr Res 1997;41:293–9.
17:978–86. 31 Denda M, Sato J, Masuda Y et  al. Exposure to a dry environment
4 Williams ML, Hincenbergs M, Holbrook KA. Skin lipid content enhances epidermal permeability barrier function. J Invest Dermatol
during early fetal development. J Invest Dermatol 1988;91:263–8. 1998;111:858–63.
5 Williams ML, Hanley K, Elias PM et al. Ontogeny of the epidermal 32 Kalia YN, Nonato LB, Lund CH et  al. Development of skin barrier
permeability barrier. J Invest Dermatol Symp Proc 1998;3:75–9. function in premature infants. J Invest Dermatol 1998;111:320–6.
6 Schmuth M, Schoonjans K, Yu QC et al. Role of peroxisome prolifera- 33 Nikolovski J, Stamatas GN, Kollias N, Wiegand BC. Barrier func-
tor‐activated receptor α in epidermal development in utero. J Invest tion and water‐holding and transport properties of infant stratum
Dermatol 2002;119:1298–303. corneum are different from adult and continue to develop through
7 Holbrook KA, Odland GF. Regional development of the human epi- the first year of life. J Invest Dermatol 2008;128:1728–36.
dermis in the first trimester embryo and the second trimester fetus. 34 Abe T, Mayuzumi J, Kikuchi N et  al. Seasonal variations in skin
J Invest Dermatol 1980;74:161–8. temperature, skin pH, evaporative water loss and skin surface lipid
8 Hardman MJ, Moore L, Ferguson MWJ et al. Barrier formation in the values on human skin. Chem Pharm Bull 1980;28:387–92.
human fetus is patterned. J Invest Dermatol 1999;113:1106–13. 35 Seidenari S, Giusti G. Objective assessment of the skin of children
9 Visscher M, Narendran V. The ontogeny of skin. Adv Wound Care affected by atopic dermatitis: a study of pH, capacitance and TEWL in
2014;3:291–303. eczematous and clinically uninvolved skin. Acta Derm Venereol
10 Visscher MO, Utturkar R, Pickens WL et al. Neonatal skin matu­ration— (Stockh) 1995;75:429–33.
vernix caseosa and free amino acids. Pediatr Dermatol 2011;28:122. 36 Behrendt H, Green M. Skin pH pattern in the newborn infant. Am J
11 Chittock J, Cooke A, Lavender T et  al. Development of stratum Dis Child 1958;35:1958.
corneum chymotrypsin‐like protease activity and natural moisturiz- 37 Green M, Carol B, Behrendt H. Physiologic skin pH patterns in infants
ing factors from birth to 4 weeks of age compared with adults. of low birth weight. Am J Dis Child 1968;115:9–16.
Br J Dermatol 2016;175:713–20. 38 Hoeger PH, Enzmann CC. Skin physiology of the neonate and young
12 Ashida Y, Ogo M, Denda M. Epidermal interleukin‐1 alpha genera- infant: prospective study of functional skin parameters during early
tion is amplified at low humidity: implications for the pathogenesis of infancy. Pediatr Dermatol 2002;19:256–62.
inflammatory dermatoses. Br J Dermatol 2001;144:238. 39 Fox C, Nelson D, Wareham J. The timing of skin acidification in very
13 Jiang YJ, Lu B, Crumrine D et  al. IL‐1alpha accelerates stratum low birth weight infants. J Perinatol 1998;18:272–5.
corneum formation and improves permeability barrier homeostasis 40 Saijo S, Tagami H. Dry skin of newborn infants: functional analysis of
during murine fetal development. J Dermatol Sci 2009;54:88. the stratum corneum. Pediatr Dermatol 1991;8:155–9.
1 4 Strunk T, Doherty D, Richmond P et al. Reduced levels of anti- 41 Okah FA, Wickett RR, Pompa K et  al. Human newborn skin: the
microbial proteins and peptides in human cord blood plasma. Arch effect of isopropanol on skin surface hydrophobicity. Pediatr Res
Dis Child Fetal Neonatal Ed 2009;94:F230–1. 1994;35:443–6.
Chapter 4  Physiology of Neonatal Skin 61

­Dermis and skin appendages is hastened postnatally so that nearly all preterm infants

STRUCTURE AND PHYSIOLOGY


are able to sweat at the age of 13 days, although the ther-

SECTION 1: DEVELOPMENT,
The dermis supplies sweat, sebum and, most impor- mal stimulus required is higher and sweat output lower
tantly, nutrients to the epidermis. Dermal vessels are than in term neonates [10]. Emotional sweating, which is
most important for the regulation of skin and body particularly prominent on the palmoplantar regions, rep-
temperature. The dermis connects the epidermal sheath resents a response to hunger or pain independent of
with the underlying fatty tissue and, through a network ambient temperature. It is not present before 36–37 weeks’
of collagenous and elastic fibres, provides stability and gestation [12]. Functional immaturity of the sweat glands
protection against trauma to the skin. With its papillary appears to be without clinical significance in the neonatal
ridges, the epidermis is intertwined with the dermis, thus period. Even in children with anhidrotic ectodermal dys-
protecting against shearing or abrasive forces. The basal plasia, who are completely unable to sweat, hyperpyrexia
cell layer to surface length ratio gives an indication of the does not occur until late infancy/early childhood.
undulation of the epidermal–dermal interface [1]. In term
infants, it increases from 1.07 ± 0.07 to 1.2 ± 0.13 within the Percutaneous respiration
first 4 months [1]. As long as the rete ridges are not or only The absorption of oxygen and excretion of carbon dioxide
incompletely formed, the epidermis is prone to abrasive through the skin is an often quoted and much overrated
injuries caused for instance by shearing movements of phenomenon. In adults and mature neonates, it accounts
the patients’ own hands or by the removal of plasters. for <2% of total respiration. In premature infants of
This effect is augmented by the relative thinness of the <30 weeks’ gestation, however, transcutaneous gas
epidermis. exchange is 6–11 times higher than in term infants. In par-
allel with the postnatal maturation of the transepidermal
Sebaceous gland activity lipid barrier, it tends to normalize within 2–3 weeks after
Sebum is composed of squalenes and monoester waxes
birth [13]. Interestingly, prolonged skin‐to‐skin contact
[2]. Sebum levels during the first month tend to be as
(‘kangarooing’) between premature babies (gestational
high as those in adults [3,4], but they decline significantly
age 30 weeks) and their mothers has been shown to
towards the end of the first trimester and remain low
improve gas exchange independent of postnatal age [14].
until the beginning of puberty. Stimulation of sebaceous
glands by maternal androgens starts before birth [3].
Accordingly, transient sebaceous gland hypertrophy is a
Wound healing
Scars result from cutaneous damage involving the basal
common finding in term neonates. Rates of maternal and
layer and dermis. Scarless wound healing is a much debated
neonatal sebum secretion are correlated [4].
phenomenon associated with fetal skin. During the first and
Thermoregulation second trimester, skin and bone wounds heal in a regenera-
Neonates, and particularly premature babies, are at an tive manner [15]. Cutaneous wound healing in the fetus
increased risk of heat loss. Heat loss is largely caused by differs in many ways from that in children or adults. The
evaporative rather than radiative loss during the first most striking difference is the absence of an acute inflam-
week of life [5]. In term infants thermogenesis is largely matory response to trauma. Degranulation and aggregation
driven by brown adipose tissue which is located around of fetal platelets and the production of fibrogenic platelet‐
the kidney and in the interscapular areas and represents derived growth factor (PDGF), transforming growth factor
about 1% of fetal weight at term [6]. Regional heat loss is (TGF)‐β1 and TGF‐β are decreased. As very few neutrophils
closely related to the external temperature. The vaso- are attracted to the wound site, devitalized tissue is mainly
constrictive response to reduced temperature, which cleared by macrophages and wound fibroblasts instead of
can be assessed by laser Doppler flowmetry, appears to neutrophils [16]. While healing of postnatal wounds involves
be diminished in the newborn infant [7]. Occlusive coordinated activity of chemokines, cytokines, growth
wrapping of VLBW infants has been shown to prevent factors and other soluble mediators [17], pro‐inflammatory
the dangerous postnatal evaporative heat loss [8]. chemokines, such as interleukins (IL)‐6 and 8, are dimin-
Although their density of sweat glands is even higher ished while IL‐10 is increased [18].
than that in adults, thermal sweating is reduced in the The number of stem cells is increased in fetal wounds
term neonate (i.e. the induction threshold for sweating is as well as in unwounded fetal skin [19]. They may be
higher than in adults) [9,10]. Sweating occurs first on the ‘educating’ the stromal cells [15] towards regeneration
forehead and later on the trunk and extremities. The instead of scarring. Proliferation and migration rates of
intensity of sweating in response to a thermal stimulus fetal fibroblasts as well as their rate of synthesis of colla-
depends on gestational age [10]. Unlike term neonates, gen are indeed increased as compared to adults. There are
preterm babies are usually unable to sweat in response also differences in the type of collagen (collagen III > I = 3 : 1
to heat during the first days of life. On the other hand, instead of 1 :3) synthesized [20].
sweating can be induced chemically as early as 32 weeks’
gestation, suggesting that hypohidrosis results from ­References
immaturity of neurological regulation rather than ana- 1 Evans NJ, Rutter N. Development of the epidermis in the newborn.
Biol Neonate 1986;49:74–80.
tomical immaturity [11]. Similar to their adaptation 2 Nikkari T. Comparative chemistry of sebum. J Invest Dermatol 1974;
regarding TEWL, however, the development of sweating 62:257–67.
62 Section 1  Development, Structure and Physiology of the Skin

3 Agache P, Blanc D, Barrand C et al. Sebum levels during the first year 13 Cartlidge PHT, Rutter N. Percutaneous respiration in the newborn
of life. Br J Dermatol 1980;103:643–9. infant: effect of gestation and altered ambient oxygen concentration.
STRUCTURE AND PHYSIOLOGY

4 Henderson CA, Taylor J, Cunliffe WJ. Sebum excretion rates in


Biol Neonate 1987;52:301–6.
SECTION 1: DEVELOPMENT,

mothers and neonates. Br J Dermatol 2000;142:110–11. 14 Föhe K, Kropf S, Avenarius S. Skin‐to‐skin contact improves gas

5 Hammarlund K, Strömberg B, Sedin G. Heat loss from the skin of exchange in premature infants. J Perinatol 2000;5:311–15.
preterm and full term newborn infants during the first weeks after 15 Yates C, Hebda P, Wells A. Skin wound healing and scarring: fetal
birth. Biol Neonate 1986;50:1–10. wounds and regenerative restitution. Birth Defects Res C Embryo
6 Hillman N, Kalapur SG, Jobe A. Physiology of transition from Today 2012;96:325–33.
intrauterine to extrauterine life. Clin Perinatol 2012;39:769–83. 16 Coolen NA, Schouten KC, Middelkoop E et al. Comparison between
7 Karlsson H, Hänel SE, Nilsson K et al. Measurement of skin tempera- human fetal and adult skin. Arch Dermatol Res 2010;302:47–55.
ture and heat flow from skin in term newborn babies. Acta Paediatr 1 7 Wulff BC, Parent AE, Meleski MA et al. Mast cells contribute to
1995;84:605–12. scar formation during fetal wound healing. J Invest Dermatol
8 Rohana J, Khairina W, Boo NY, Shareena I. Reducing hypothermia in 2012;132:458–46.
preterm infants with polyethylene wrap. Pediatr Int 2011;53:468–74. 18 Kiechty KW, Adzick NS, Crombleholme TM. Diminished interleukin
9 Mancini AJ, Lane AT. Sweating in the neonate. In: Polin RA, Fox WW, 6 (IL‐6) production during scarless human fetal wound repair.
eds. Textbook of Fetal and Neonatal Physiology, 2nd edn. Philadelphia: Cytokine 2000;12:671–6.
W.B. Saunders, 1998:767–70. 19 Wagers AJ, Christensen JL, Weissman IL. Cell fate determination from
10 Harpin VA, Rutter N. Sweating in preterm babies. J Pediatr 1982; stem cells. Gene Ther 2002;9:606–12.
100:614–18. 20 Merkel JR, DiPaolo BR, Hallock GG et al. Type I and Type III collagen
11 Behrendt H, Green M. Nature of the sweating deficit in prematurely content of healing wounds in fetal and adult rats. Proc Soc Exp Biol
born neonates. N Engl J Med 1972;286:1376–9. Med 1988;187:493–7.
12 Harpin VA, Rutter N. Development of emotional sweating in the
newborn infant. Arch Dis Child 1982;57:691–5.
 63

C HA PTER   5

Neonatal Skin Care


Peter H. Hoeger

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Department of Paediatric Dermatology, Catholic Children’s Hospital Wilhelmstift, Hamburg, Germany

Introduction, 63 Skin care of the premature infant, 66 Percutaneous absorption, 71


Skin care of the neonate, 63 Skin care of the term neonate and infant, 67

Abstract The benefits of emollient care need to be weighed against poten-


tial side‐effects. Anionic detergents such as sodium lauryl ­sulfate
Due to its structural and functional immaturity, the skin of neo- and emollients containing emulsifiers can emulsify s­tratum
nates is more susceptible to mechanical damage and chemical ­corneum lipids, thus increasing permeability and skin dryness.
­irritation. Neonatal skin care aims at preventing physical injury, Common ­ingredients such as lanolin can cause contact a ­ llergies,
minimizing transepidermal water loss and avoiding infection. others such as parabenes or triclosan can act as endocrine
The  optimal emollient for skin care remains to be determined. ­disrupting agents.

Key points • The rate of inadvertent percutaneous absorption of many agents


applied to the skin surface is increased in infants, but largely
• Due to its immature epidermal barrier, neonatal skin is highly unexplored for cosmetic ingredients.
sensitive to physical and chemical damage. • Commonly used ingredients can lead to contact sensitization.
• Infants are exposed to many cosmetic ingredients at an early • These aspects need to be considered for all aspects of skin care in
age. the neonate and young infant.

­Introduction however not easily answered as there are very few rand-
omized controlled trials (RCTs) addressing these issues.
Due to its structural and functional immaturity, the skin In the absence of scientific evidence, many recommenda-
of full‐term and preterm neonates and infants is different tions are rather based on common sense, traditional prac-
from that of older children. Epidermal barrier and ther- tice or ‘experts’ opinions’, some of which are more or less
moregulatory mechanisms are immature; the skin is thin- openly sponsored by the cosmetic industry.
ner and more susceptible to mechanical damage and
chemical irritation; and the skin surface of neonates is
­References
larger (in proportion to their body mass) than in adults. 1 Nikolovski J, Stamatas GN, Kollias N et  al. Barrier function and
As outlined in Chapter  4, these structural and develop- water‐holding and transport properties of infant stratum corneum
mental differences are associated with decreased epider- are different from adult and continue to develop through the first
year of life. J Invest Dermatol 2008;128:1728–36.
mal concentrations of natural moisturizing factors (NMF) 2 Chittock J, Cooke A, Lavender T et  al. Development of stratum cor-
and increased transepidermal water loss (TEWL) [1,2]. neum chymotrypsin‐likeprotease activity and natural moisturizing
As  a consequence, the skin surface pH is increased and factors from birth to 4 weeks of age compared with adults. Br J
epidermal proteases are activated [2,3]; both factors Dermatol 2016;175:713–20.
3 Hachem JP, Man MQ, Crumrine D et  al. Sustained serine proteases
plus  decreased epidermal concentrations of antimicro- activity by prolonged increase in pH leads to degradation of lipid pro-
bial lipids [4] facilitate bacterial colonization and trans- cessing enzymes and profound alterations of barrier function and stra-
cutaneous infections, particularly in preterm infants. tum corneum integrity. J Invest Dermatol 2005;125:510–20.
4 Drake DR, Brogden KA, Dawson DV et al. Thematic review series: skin
Furthermore, the immature epidermal barrier allows eas- lipids. Antimicrobial lipids at the skin surface. J Lipid Res 2008;49:
ier transcutaneous penetration of allergens and chemi- 4–11.
cals. In conjunction with the larger skin surface of neonates
this transforms into an increased risk for systemic toxicity
­Skin care of the neonate
from topically applied substances. Maturation of the bar-
rier takes at least 12 months [1]. It is therefore obvious that Aims of neonatal skin care
the skin of neonates and young infants requires specific Neonatal skin care aims at:
care in order to avoid damage and, ideally, to compensate • minimizing transepidermal water loss
for the immaturity of the epidermal barrier. The questions • prevention of physical injury and
if, when and how neonates should receive skin care are • avoiding infections.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
64 Section 2  Skin Disorders of the Neonate and Young Infant

Environmental factors affecting epidermal As bathing can lead to neonatal hypothermia (<36.5 °C),
integrity which in itself carries the risk of intraventricular haemor-
The abrupt switch from 100% humidity within the rhage, the first bath should be delayed until the neonate’s
­amniotic fluid to the relatively dry ambient atmosphere vital signs and body temperature have remained stable
represents a maximal challenge to the adaptability of for at least 4–6 hours [7,11,12]. For rewarming after
the  neonatal skin. Various environmental factors can the  first bath, close skin‐to‐skin contact (under protec-
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

adversely affect the epidermal integrity and should be tive covering) is preferred by most mothers and has been
avoided wherever possible (Table 5.1). shown to be safe and effective [13]. In resource‐poor
Soap and anionic detergents such as sodium lauryl sul- countries neonatal mortality can be reduced significantly
fate (SLS) lead to an immediate increase of the skin sur- by postponing the first bath for 72 hours post partum [14].
face pH which in turn activates proteases capable of Blood and meconium should however be gently removed
disturbing the epidermal barrier. ‘Hard’ tap water which after birth.
contains high levels of free calcium is prevalent in many
areas. In these areas, the incidence of atopic dermatitis Washing and bathing
(AD) is increased [1]. Hard water can induce skin irrita- Immersion (tub) bathing is not only a means of cleansing,
tion [2], particularly so because it requires increased use but also a moment of close tactile interaction between
of soap. High levels of free calcium have been shown to baby and parent(s). In an RCT comparing sponge and tub
inhibit skin barrier repair in vitro [3]. immersion bathing in a cohort of late preterm infants, tub
Skin irritation can likewise be induced by too‐frequent bathing was shown to significantly improve thermoregu-
washing, synthetic or inflexible garments causing abra- lation [15]. The water should cover the infant’s entire
sions, or the removal of adhesives [4]. body in order to decrease evaporative heat loss. The opti-
It is well known that disruption of the normal skin mal water temperature is 37–38 °C. Water temperature
microbiome by antibiotic treatment during the first weeks should always be accurately measured before immersion
of life increases the risk to develop AD [5]. Recent evi- as tests just by touch have proved inaccurate to avoid
dence using whole‐metagenome sequencing [6] suggests scalds. Since hyperhydration of the epidermis would ren-
that skin bacteria can influence the skin surface microen- der the skin more fragile, the duration of the bath should
vironment, for instance by production of excessive be limited to 5–10 minutes every other day [9,16].
amounts of ammonia which adds to the increased skin Immediately after bathing, the infant’s skin should be
surface pH in children with AD. gently towelled and a head covering applied [7]. There is
no evidence that delaying the first bath until the umbilical
Skin care immediately after birth cord has separated prevents umbilical cord infection
and in the first 2–3 days [8,9,17].
Immediately after delivery, the neonate should be Surfactants in skin cleansers are tensioactive agents
wrapped in a towel and gently rubbed. The baby should capable of removing fatty substances from the skin sur-
be kept in a room of at least 25–28 °C ambient temperature face by emulsifying them into fine droplets, which can
and no draught. Any remaining vernix should be left in then be rinsed away. Anionic detergents contain mole-
place and allowed to dry and peel off naturally [7–9]. cules with a negatively charged hydrophilic end (e.g.
Owing to its antimicrobial property and its capacity to SLS). They create a foaming effect which is associated
diminish TEWL, vernix would in fact be an ideal emol- with a cleansing action on the stratum corneum. Cationic
lient for the neonate [10], and there is some evidence that detergents such as quaternary ammonium salts contain
retention of vernix is associated with better skin hydra- molecules with a positively charged end. Skin cleansing
tion and lower skin surface pH as compared to newborns with soaps leads to alkalization of the skin surface and
whose vernix was removed immediately after birth [10]. can cause irritation [17,18].
However, in case of maternal human immunodeficiency Surfactants can emulsify stratum corneum lipids, thus
virus (HIV) or hepatitis B infection, early removal is increasing permeability and dryness of the skin surface
advisable due to the risk of transmission of infectious [19]. Detergents should therefore be used cautiously in
agents via maternal blood [7]. neonates, and the skin should be rinsed with water fol-
lowing their use. Although frequently recommended by
Table 5.1  Environmental factors exerting negative effects on the epider- various sponsored panels, the evidence in favour of the
mal barrier use of liquid (or gel) cleansers with or without emollient
is inconclusive. A comparative study in neonates indi-
Factor Effect on epidermal barrier cated that liquid cleansers plus emollients might lead to
increased TEWL as compared to rinsing with water alone
Hard water Hydration of SC↓, skin barrier repair↓ [20]; opposite results were found in another study [21]. It
Detergents Skin surface pH↑, activation of proteases
is strongly advisable to use only mild, non‐irritating liq-
Too‐frequent washing Hydration of SC↓, skin dryness↑
Adhesive plasters Destruction of corneal layer uid cleansers able to maintain the normal surface pH [17];
Synthetic garments Skin irritation in order to minimize their irritation potential, cleansers
Altered skin flora Formation of ammonia, skin surface pH↑ should be fragrance‐free and dye‐free and contain the
least possible concentration of preservatives [8]. The same
↑, increase; ↓, decrease; SC stratum corneum. cleansers can be used for the scalp and hair. Alternatively,
Chapter 5  Neonatal Skin Care 65

shampoos containing very mild surfactant complexes


Box 5.1  List of common potential allergens in baby wipes causing
and buffers in order to achieve a pH and saline concentra-
allergic contact dermatitis [36,37]
tion similar to those of tears have been specifically
designed for infant care [16,22]. • Botanical extracts, including members of the Compositae family
• α‐tocopherol
Umbilical cord care • Fragrances

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
The umbilical cord stump dries out and falls off between • Propylene glycol
5 and 10 days after birth. In countries with poor hygienic • Parabens
standards, the cord stump represents an important focus • Iodopropynyl butylcarbamate
• Lanolin
for neonatal infection and mortality. Umbilical cord care
• Methylchloroisothiazolinone/methylisothiazolinone
varies widely around the world [23–27]. General recom-
• Formaldehyde releaser
mendations issued by the WHO in 2004 [23] include:
• proper hand hygiene
• cutting the cord with a sterile instrument
• washing the cord stump with clean water or soap. to maintain an acidic skin surface pH [34]. This is in
Several agents formerly recommended for cord care accordance with the results of a prospective study show-
(isopropyl alcohol, hexachlorophene, neomycin) can have ing that a higher skin pH in neonates is a predisposing
serious adverse effects and should be avoided [9]. Alcohol factor for napkin dermatitis [35]. However, preservatives
or topical antiseptics delay cord separation by 2 days and fragrances in baby wipes can lead to allergic contact
[23,24]. Meta‐analyses of controlled studies indicate that sensitization which is an underrecognized cause of per-
the application of 4% chlorhexidine solution to the umbil- ineal dermatitis [36,37] (Box 5.1).
ical stump is non‐hazardous and can reduce the risk of
omphalitis by up to 50% and of neonatal mortality by 12% ­References
[25–27]. However, in a large randomized study from 1 Ewence A, Rumsby P, Rockett L et al. A review of skin irritation and
France with 8698 participants published in 2017, dry care tap water quality. Swindon: WRc. Drinking Water Inspectorate Ref.
DWI8375, 2011:1–149.
of the cord proved non‐inferior to the use of antiseptics 2 Warren R, Ertel KD, Bartolo RG et  al. The influence of hard water
[28]. Both approaches may have their justification; how- (calcium) and surfactants on irritant contact dermatitis.Contact
ever, antiseptic prophylaxis may be preferable for deliver- Dermatitis 1996;35:337–43.
3 Lee SH, Elias PM, Proksch E et al. Calcium and potassium are impor-
ies outside a hospital setting and in resource‐limited
tant regulators of barrier homeostasis in murine epidermis. J Clin
populations [29]. Invest 1992;89:530–8.
There is currently no good evidence favouring full‐ 4 Lund CH, Nonato LB, Kuller JM et al. Disruption of barrier function
body skin cleansing with chlorhexidine as a prophylactic in neonatal skin associated with adhesive removal. J Pediatr 1997;131:
367–72.
skin disinfectant in neonates [26]. The same applies to 5 Tsakok T, McKeever TM, Yeo L, Flohr C. Does early life exposure to
maternal vaginal chlorhexidine washes compared to antibiotics increase the risk of eczema? A systematic review. Br J
usual care [26]. Underdeveloped countries with poor Dermatol 2013;169:983–91.
hygienic conditions might nevertheless benefit from these 6 Chng KR, Tay ASL, Li C et al. Whole metagenome profiling reveals
skin microbiome‐dependent susceptibility to atopic dermatitis flare.
approaches to reduce neonatal mortality [9]. All antisep- Nat Microbiol 2016;1:1–10.
tic  solutions required for sterile procedures should be 7 Lund CH, Osborne JW, Kuller J et al. Neonatal skin care: clinical out-
applied only to the immediate skin area and in very small come of the AWHONN/NANN evidence‐based clinical practice
guideline. J Obstet Gynecol Neonatal Nurs 2001;30:41–51.
amounts. They should be removed with water‐soaked 8 Dyer JA. Newborn skin care. Semin Perinatol 2013;37:3–7.
gauze after the procedure to prevent unnecessary absorp- 9 Ness MJ, Davis DMR, Carey WA. Neonatal skin care: A concise
tion due to prolonged exposure in order to avoid serious review. Int J Dermatol 2013;52:14–22.
complications [30]. In neonates, alcohol used as a topical 10 Visscher MO, Barai N, LaRuffa AA et al. Epidermal barrier treatments
based on vernix caseosa. Skin Pharmacol Physiol 2011;24:322–9.
antiseptic is nearly completely absorbed percutaneously 11 Blume‐Peytavi U, Lavender T, Jenerowicz D et al. Recommendations
with a potential for systemic toxicity. It is also drying and from a European Roundtable Meeting on Best Practice Healthy Infant
painful to abraded skin and can cause skin necrosis [31]. Skin Care. Pediatr Dermatol 2016;33:311–21.
12 WHO. Pregnancy, Childbirth, Postpartum and Newborn Care: A
Its use should be completely abandoned in this age group.
guide for essential practice, 3rd edn. Geneva: WHO, 2015.
13 George S, Phillips K, Mallory S et al. A pragmatic descriptive study of
Napkin/diaper care rewarming the newborn after the first bath. J Obstet Gynecol Neonatal
Since permanently moist skin leads to maceration, and Nurs 2015;44:203–9.
14 Akter T, Dawson A, Sibbritt D. What impact do essential newborn
macerated skin is more susceptible to irritation and injury, care practices have on neonatal mortality in low and lower‐middle
napkins should be changed frequently, i.e. every 3–4 income countries? Evidence from Bangladesh. J Perinatol 2016;36:
hours, at least after each feed. Superabsorbent napkins 225–30.
are clearly superior to cloth napkins with respect to sur- 15 Loring C, Gregory K, Gargan B et  al. Tub bathing improves ther-
moregulation of the late preterm infant. J Obstet Gynecol Neonatal
face dryness and prevention of napkin dermatitis [32,33] Nurs 2012;41:171–9.
(Chapter  20). Although water and a wash cloth are the 16 Gelmetti C. Skin cleansing in children. J Eur Acad Dermatol Venereol
traditional standard of care for rinsing the napkin area 2001;15:12–15.
17 Blume‐Peytavi U, Cork MJ, Faergemann J et al. Bathing and cleansing
[8,9], disposable infant wipes have become a popular
in newborns from day 1 to first year of life: recommendations from a
alternative. Comparative studies suggest that they may European round table meeting. J Eur Acad Dermatol Venereol
be equally or even better tolerated, particularly those able 2009;23:751–9.
66 Section 2  Skin Disorders of the Neonate and Young Infant

18 Visscher MO, Adam R, Brink S. Newborn infant skin: Physiology, humidity of ≥80%. Ambient humidity is usually reduced
development and care. Clin Dermatol 2015;33:271–80.
stepwise after a week because of concerns about an
19 Lodén M, Buraczewska I, Edlund F. The irritation potential and reser-
voir effect of mild soaps. Contact Dermatitis 2003;49:91–6. increased risk of infection [2]. Conventional phototherapy
20 Roberta R, Patrizi A, Cocchi G et al. Comparison of two different neo- can increase TEWL, while modern LED phototherapy
natal skin care practices and their influence on transepidermal water units do not [3].
loss in healthy newborns within first 10 days of life. Minerva Pediatr
2014;66:369–74.
Various emollients have been shown to improve the
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

21 Garcia Bartels N, Scheufele R, Prosch F et al. Effect of standardized immature epidermal barrier function in premature
skin care regimens on neonatal skin barrier function in different body infants. Since the first randomized controlled study in
areas. Pediatr Dermatol 2010;27:1–8. 60  premature infants (<33 weeks of gestation) demon-
22 Walters RM, Fevola MJ, LiBrizzi JJ et al. Designing cleansers for the
unique needs of baby skin. Cosm Toil 2008;123:53–60. strated decreased TEWL, improved skin scores and
23 Zupan J, Garner P, Omari AA. Topical umbilical cord care at birth. decreased colonization and infection rates in those treated
Cochrane Database Syst Rev 2004;CD001057. with a preservative‐free petrolatum ointment versus an
24 Ahn Y, Sohn M, Jun Y et  al. Two methods of cord care in high‐risk
newborns: their effects on hydration, temperature, pH, and floras of
untreated control group [4], about 20 similar studies with
the cord area. J Child Health Care 2015;19:118–29. various emollients (mineral oil, lanolin cream, oils from
25 Mullany LC, Shah R, El Arifeen S et al. Chlorhexidine cleansing of sunflower seed, olive, almond, vegetable or coconut) and
the umbilical cord and separation time: a cluster‐randomized trial. involving a total of >3000 infants have confirmed the orig-
Pediatrics 2013;131:708–15.
26 Sinha A, Sazawal S, Pradhan A et  al. Chlorhexidine skin or cord inal findings [e.g. 5–9]. There are however concerns about
care for prevention of mortality and infections in neonates. an increased risk of coagulase‐negative staphylococcal
Cochrane Database Syst Rev 2015;(3):CD007835. infection, nosocomial infection and systemic candidiasis
27 Karumbi J, Mulaku M, Aluvaala J et al. Topical umbilical cord care for
in premature infants treated with topical emollients
prevention of infection and neonatal mortality. Pediatr Infect Dis J
2013;32:78–83. [10,11]. A meta‐analysis of 18 studies published between
28 Gras‐Le Guen C, Caille A, Launay E et al. Dry care versus antisep- 1993 and 2015 revealed no benefit in terms of significant
tics for umbilical cord care: A cluster randomized trial. Pediatrics reduction of mortality in the emollient‐treated groups of
2017;139:e20161857.
29 Stewart D, Benitz W. Umbilical cord care in the newborn infant.
preterm infants [12]. This was probably due to methodo-
Pediatrics 2016;138:e20162149. logical weakness of many studies, specifically uncertainty
30 Upadhyayula S, Kambalapalli M, Harrison CJ. Safety of anti‐infective about adequate allocation concealment methods in many
agents for skin preparation in premature infants. Arch Dis Child and lack of blinding in all of the trials [12].
2007;92:646–7.
31 Harpin VA, Rutter N. Percutaneous alcohol absorption and skin
Therefore, preventive application of ointments to
necrosis in a premature infant. Arch Dis Child 1982;57:477–9. improve the immature epidermal barrier cannot be rec-
32 Heimall LM, Storey B, Stellar JJ, Davis KF. Beginning at the bottom: ommended at present. However, this simple and low‐
evidence‐based care of diaper dermatitis. MCN Am J Matern Child
Nurs 2012;37:10–16.
cost approach to reduce infectious complications and
33 Tüzün Y, Wolf R, Bağlam S, Engin B. Diaper (napkin) dermatitis: A mortality of premature infants has great potential, par-
fold (intertriginous) dermatosis. Clin Dermatol 2015;33:477–82. ticularly for low‐income countries, and deserves further
34 Visscher M, Odio M, Taylor T et  al. Skin care in the NICU patient: well‐designed RCTs.
effects of wipes versus cloth and water on stratum corneum integrity.
Neonatology 2009;96:226–34.
35 Yonezawa K, Haruna M, Shiraishi M et al. Relationship between skin Avoidance of mechanical damage
barrier function in early neonates and diaper dermatitis during the Patients undergoing neonatal intensive care are exposed to
first month of life: a prospective observational study. Pediatr Dermatol
2014;31:692–7.
many diagnostic and therapeutic procedures. Neonates are
36 Yu J, Treat J, Chaney K, Brod B. Potential allergens in disposable particularly susceptible to iatrogenic injury, the incidence
diaper wipes, topical diaper preparations, and disposable dia- of which was calculated in a prospective study at 25.6 per
pers:  under‐recognized etiology of pediatric perineal dermatitis. 1000 patient days [13]. Among 267 iatrogenic events
Dermatitis 2016;27:110–18.
37 Chang MW, Nakrani W. Six children with allergic contact dermatitis observed, cutaneous injuries were the most frequent
to methylisothiazolinone in wet wipes (baby wipes). Pediatrics 2014; (35.2%), but they were mild in 95% [13]. In a more detailed
133:e434–8. analysis in 113 premature infants <33 weeks of gestation,
16.8% of all infants were affected by iatrogenic cutaneous
­Skin care for the premature infant injuries [14]. These were mainly induced by ventilation
equipment, intravenous catheters, electrodes, dressings,
As outlined in Chapter 4, the thickness of the epidermis disinfectants and pressure sores. The main risk factors
correlates inversely with gestational age. In a 25‐week‐ were low birthweight and duration of ventilation [14].
old premature baby, it is only 25 μm instead of 50 μm in a Implementation of a systematic quality improvement
neonate at term. The skin of premature infants is thus approach can lead to a significant reduction in device‐
even more susceptible to trauma, TEWL and transepider- related pressure ulcers on the NICU [15]. This approach
mal intoxication than that of a mature neonate. includes preventive skin care, early detection of immi-
nent damage through systematic assessment of the skin,
Control of transepidermal water loss and identification of strategies to mitigate device‐related
In premature infants younger than 30 weeks of gesta- injury to further reduce ulcer rates [15]. Nasal continuous
tion, transepidermal and evaporative water loss is exces- positive airway pressure frequently (20–60%) leads to
sive and life‐threatening. Immediately after birth, babies nasal injury and trauma secondary to tight‐fitting nasal
are wrapped in polyurethane foil [1] and then placed interfaces [16]. Nasal injury can be reduced using rotating
in  a humidification incubator with an initial ambient mask/prong nasal interfaces [17].
Chapter 5  Neonatal Skin Care 67

Table 5.2  Recommendations for skin care in premature infants 4 Nopper AJ, Horii KA, Soodeo‐Drost S et al. Topical ointment therapy
benefits premature infants. J Pediatr 1996;128:660–9.
5 Darmstadt GL, Badrawi N, Law PA et al. Topically applied sunflower
Problem Recommendation
seed oil prevents invasive bacterial infections in preterm infants in
Egypt: a randomized, controlled clinical trial. Pediatr Infect Dis J
Bathing • No full‐immersion bathing until ≥32 weeks of 2004;23:719–25.
gestation and stable body temperature 6 Darmstadt GL, Ahmed S, Ahmed NU, Saha SK. Mechanism for pre-
• No cleansing agent in the first 2 weeks vention of infection in preterm neonates by topical emollients. A ran-

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
• Surface cleansing with a cotton cloth twice a week domized, controlled clinical trial. Pediatr Infect Dis J 2014;33:1124–7.
Adhesives • Avoid adhesives wherever possible 7 Erdemir A, Kahramaner Z, Yuksel Y et al. The effect of topical oint-
• Place protective barrier (pectin hydrocolloid) between ment on neonatal sepsis in preterm infants. J Matern Fetal Neonatal
Med 2015;28:33–6.
skin and sites of frequent taping (e.g. nose for
8 Salam RA, Darmstadt GL, Bhutta ZA. Effect of emollient therapy on
nasogastric tubes, continuous positive airway pressure) clinical outcomes in preterm neonates in Pakistan: a randomized con-
• Use silicone adhesives wherever possible trolled trial. Arch Dis Child Fetal Neonatal Ed 2015;100:F210–15.
• Use nonadhesive probes 9 Nangia S, Paul VK, Deorari AK et al. Topical oil application and trans‐
Topical agents • Be aware of percutaneous absorption epidermal water loss in preterm very low birth weight infants—a ran-
• Never use alcohol or alcoholic solutions domized trial. J Trop Pediatr 2015;61:414–20.
• Never use triclosan, hexachlorophene and other 10 Conner JM, Soll RF, Edwards WH. Topical ointment for preventing
potentially toxic agents (see Table 5.4) infection in preterm infants. Cochrane Database Syst Rev 2004;(1):
Emollients • Consider preventive/protective skin care with CD001150.
11 Campbell JR, Zaccaria E, Baker CJ. Systemic candidiasis in extremely
emollients when humidified incubators for premature
low birth weight infants receiving topical petrolatum ointment for
infants are not available skin care: A case‐control study. Pediatrics 2000;105:1041–5.
Phototherapy • Use LED phototherapy only 12 Cleminson J, McGuire W. Topical emollient for preventing infection
in preterm infants. Cochrane Database Syst Rev 2016;(1):CD001150.
13 Ligi I, Arnaud F, Jouve E et al. Iatrogenic events in admitted neonates:
a prospective cohort study. Lancet 2008;371:404–10.
14 Roche‐Kubler B, Puzenat E, Mariet AS et  al. Lésions cutanées

Adhesive tapes should be used only sparingly. Removal iatrogènes: étude prospective chez les prématurés de moins de 33
semaines de l´hôpital universitaire de Besançon. Ann Dermatol
requires a special technique with one hand holding the Venereol 2015;142:3–9.
underlying skin while the other peels off the adhesive. 15 Visscher M, King A, Nie AM et al. A quality‐improvement collabora-
The application of a hydrocolloid (pectin) barrier between tive project to reduce pressure ulcers in PICUs. Pediatrics 2013;
131:e1950–60.
the adhesive surface of a temperature probe and the neo-
16 Newnam KM, McGrath JM, Estes T et  al. An integrative review of
nate’s skin or under tape minimizes the trauma of strip- skin breakdown in the preterm infant associated with nasal continu-
ping when the probe is removed [18,19]. The pectin‐based ous positive airway pressure. J Obstet Gynecol Neonatal Nurs
barrier under tape held probes and appliances safely for 2013;42:508–16.
17 Newnam KM, McGrath JM, Salyer J et al. A comparative effectiveness
an average of 5–6 days, and the skin condition remained study of continuous positive airway pressure‐related skin break-
normal in 97% after barrier removal [18]. down when using different nasal interfaces in the extremely low birth
Use of soft silicone adhesives was shown to reduce pain weight neonate. Appl Nurs Res 2015;28:36–41.
severity scores during dressing changes and was atrau- 18 Lund C, Kuller JM, Tobin C et al. Evaluation of a pectin‐based barrier
under tape to protect neonatal skin. J Obstet Gynecol Neonatal Nurs
matic in >99% [20]. For full‐thickness and exudative 1986;15:39–44.
wounds, polyurethane foam may likewise be suitable as 19 OʼNeil A, Schumacher B. Application of a pectin barrier for medical
an atraumatic wound dressing. Silver alginate, however, adhesive skin injury (epidermal stripping) in a premature infant.
J Wound Ostomy Continence Nurs 2014;41:219–21.
is not advisable in premature infants because it can lead 20 Morris C, Emsley P, Marland E et al. Use of wound dressings with soft
to systemic absorption of silver [21]. silicone adhesive technology. Paediatr Nurs 2009;21:38–43.
Pulse oximetry has largely replaced transcutaneous 21 Khattak AZ, Ross R, Ngo T, Shoemaker CT. A randomized controlled
oxygen monitoring. If transcutaneous oxygen monitors evaluation of absorption of silver with the use of silver alginate
(Algidex) patches in very low birth weight (VLBW) infants with cen-
are still required, electrodes should be adjusted to the tral lines. J Perinatol 2010;30:337–42.
lowest effective temperature; in order to prevent skin burns, 22 Villarroel M, Guazzi A, Jorge J et al. Continuous non‐contact vital sign
electrode positions should be changed every 4  hours. monitoring in neonatal intensive care units. Healthc Technol Lett
2014;1:87–91.
An ideal goal for the (near) future is the continuous moni-
toring of vital signs (heart and respiratory rate, oxygen
saturation) in preterm infants with non‐contact methods
such as a digital video camera [22]. ­ kin care of the term neonate
S
Table 5.2 summarizes basic recommendations for skin and infant
care in premature infants. Infants and young children have become a major target of
the cosmetics industry. The use of cosmetic products has
­References increased significantly in this age group over the past
1 Bhandari V, Brodsky N, Porat R. Improved outcome of extremely low years [1–3]. Regular use of ‘leave‐on’ products such as
birth weight infants with Tegaderm application to skin. J Perinatol
2005;25:276–81. facial cream and body lotion is very common in children
2 Lund CH, Osborne JW, Kuller J et al. Neonatal skin care: clinical out- from 0 to 4 years [1,3]. The newborn infant is exposed to
come of the AWHONN/NANN evidence‐based clinical practice an average of 8 ± 3 different skin care products containing
guideline. J Obstet Gynecol Neonatal Nurs 2001;30:41–51.
3 Bertini G, Perugi S, Elia S et al. Transepidermal water loss and cerebral
48 ± 18 different environmental chemicals. On average,
hemodynamics in preterm infants: conventional versus LED photo- infants are bathed four times per week and shampooed
therapy. Eur J Pediatr 2008;167:37–42. three times [4]. Many cosmetic products carry a risk of
68 Section 2  Skin Disorders of the Neonate and Young Infant

percutaneous absorption, irritation or sensitization, and TEWL, and hence increased dryness. Maturation of the
the mere fact that they are advertised for children does epidermal barrier extends throughout the first year of life
not necessarily imply that they are free of these risks [1–4]. and beyond [7]. The extent of skin dryness varies widely
Baby powder containing talcum or corn starch was part of between neonates; if marked and persistent it can be an
the traditional care of infants. However, it neither pos- early sign of dominantly inherited filaggrin deficiency.
sesses moisturizing properties nor is it suitable for the Emollients are cosmetic products intended to make the
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

treatment of oozing umbilical stump infections. Powders skin softer (Latin: mollis) since sufficient hydration is
should be completely abolished from infant care because essential for softness and flexibility of the skin. Some work
of the risk of accidental aspiration with potentially life‐ by reducing dehydration through an occlusive effect and
threatening respiratory disease [5,6]. create a film, thus decreasing imperceptible water loss.
The most occlusive emollients are derivatives of mineral
Emollients oil (Vaseline, liquid paraffin) or those containing waxes
Dry skin is a common phenomenon in term and post‐ (mainly beeswax). More physiological lipid film compo-
term neonates. The instantaneous transition from 100% nents are lanolin, plant oils (evening primrose, sunflower
humidity in utero to a comparatively dry ambient atmos- seed, jojoba, wheat germ, avocado and others) and cera-
phere is frequently followed by a period of transitory mides. Fatty, cetyl or stearyl alcohols are frequently required
postnatal desquamation (Fig. 5.1) which can be quite pro- due to their emulsifying capacity. In addition to lipids,
nounced and is sometimes mistaken for the manifestation many emollients contain humectants (mainly glycerol,
of a keratinization disorder. As outlined previously, urea). In a direct comparison, emollients containing humec-
immaturity of the epidermal barrier leads to increased tants exhibit a greater and more sustained improvement of

(a) (c)

(b)
Fig. 5.1  Postnatal desquamation in 2‐ to 8‐day‐old neonates. (a) Abdomen, (b) upper arm, (c) foot.
Chapter 5  Neonatal Skin Care 69

Table 5.3  Potentially irritant or hazardous components of emollient • Vegetable oils: Vegetable oils share the benefit of being
creams ‘natural’ and are commonly used in baby skin care.
However, owing to different ratios of saturated/unsat-
Component Hazard
urated fatty acids, the physicochemical properties of
vegetable oils on the skin surface are quite different.
Detergents (e.g. SLS) Damage of epidermal barrier, TEWL↑
Emulsifiers Emulsification of endogenous epidermal
Olive oil, with a high percentage of saturated fatty

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
lipids, TEWL↑ acids, is prone to oxidation on the skin surface, which
Lanolin Contact allergy; low‐level contamination with leads to increased formation of reactive oxygen radi-
organophosphates from pesticides cals. As a consequence, and in contrast to sunflower
Mineral oils (petrolatum, Accumulation of mineral oil saturated and seed oil, olive oil was found to damage the epidermal
paraffin) aromatic hydrocarbons in fatty tissue barrier and cause skin irritation and exacerbation of
Olive oil Damage of epidermal barrier, TEWL↑
AD [11]. However, in an RCT in 115 healthy, full‐term
Polyethylene glycols (PEG) Percutaneous absorption in burns patients,
serum osmolarity ↑, high calcium gap,
neonates, both olive and sunflower seed oil treated
imminent renal failure infants had significantly improved hydration but sig-
Propylene glycol Serum hyperosmolarity after excessive use in nificantly less improvement in lipid lamellae structure
burns patients or patients with toxic compared to the no oil group. There were no significant
epidermal necrolysis differences in TEWL, pH or erythema/skin scores
Urea Skin irritation in children <2 years (‘stinging between treated and untreated neonates [19].
effect’)
• Emulsifiers: Emulsifiers are required to link the lipo-
philic and hydrophilic components in creams. They
↑, increase; SLS, sodium lauryl sulfate; TEWL, transepidermal water loss.
permeate through the stratum corneum into deeper
layers of the epidermis where they can interact with
the epidermal barrier function [8]. However, all emollients epidermal lipids [20]. Long‐term use of creams con-
currently available have only transient effects and need to taining high concentrations of emulsifiers can lead to
be applied repeatedly over the day [8]. irritational dermatitis and paradoxically increase skin
Emollients are generally perceived as ‘inert’, and poten- dryness [20,21].
tial side‐effects are neither expected nor systematically • Polyethylene glycols (PEG) are widely used in cosmetic
investigated. However, several reports highlight negative applications because of their emulsifying and viscous
effects of commercially available skin care products on properties and low toxicity. In patients with wide-
the epidermal barrier [9–11]. Potential hazards of emol- spread burns, however, percutaneous absorption of
lients (Table 5.3) should therefore be borne in mind, par- ethylene glycol after topical treatment with a PEG‐
ticularly in infants and young children: based burn cream was described, leading to increased
• Mineral oils (petrolatum, paraffin) contain variable serum osmolalities and a high ‘calcium gap’, i.e.
amounts of mineral oil saturated hydrocarbons and increased serum calcium with a concomitant decrease
mineral oil aromatic hydrocarbons, some of which are in the ionized calcium likely attributable to binding of
strongly accumulated in fatty tissue and can lead to calcium by dicarboxylic acid metabolites of PEG [22].
granuloma formation [12,13]. Mineral oil hydrocarbons All three patients died in acute renal failure. Similar
are considered the greatest contaminant of the human findings were obtained in rabbits treated with PEG.
body, amounting to approximately 1 g per person. Caution is advised with PEG‐containing emollients not
Routes of contamination include inhalation, food only in burn patients but also in children with wide-
intake (e.g. breast milk [12], baby food contaminated by spread barrier defects, e.g. premature babies and epi-
packaging [14]), and dermal absorption from moistur- dermolysis bullosa patients. Hyperosmolarity was
izers or lipsticks. likewise observed after excessive topical use of PEG in
• Lanolin: Lanolin is a component of many ‘natural’ skin burns or toxic epidermal necrolysis patients [23].
care products. With a prevalence of up to 3.8% [15], it is • Urea: Emollients containing urea frequently cause skin
also one of the most common contact allergens. irritation In infants described as a ‘stinging effect’.
Children with AD are particularly prone to develop Glycerol (5–20%) is well tolerated and therefore the
contact allergy to skin care components, most com- preferred moisturizer up to the age of 2 years.
monly to cocamidopropyl betaine, lanolin, tixocortol There are very few RCTs dealing with regular skin care in
pivalate and parthenolide [16]. Since lanolin is derived neonates [24], and virtually all were sponsored by pro-
from sheep wool, low‐level contamination of lanolin ducers of baby skin care products. In a small RCT with 64
wool grease with organophosphates such as diazinon neonates assigned to different skin care modalities and
derived from insecticides is still detectable with highly followed up for 2 months, babies who were bathed twice
sensitive methods [17,18]. a week with a wash gel followed by application of a skin
• Sodium lauryl sulfate (SLS): Detergents such as SLS are care cream were found to have lower TEWL than those
known to exert negative effects on the intactness of the bathed in water alone; not surprisingly, cream application
epidermal barrier. A widely used emollient, aqueous was associated with higher stratum corneum hydration
cream BP, was found to increase TEWL in volunteers [25]. The significance of these findings is limited by the
prone to AD; this effect was attributed to the presence small group of infants studied and the short period of
of 1% SLS in the emollient [10]. follow‐up.
70 Section 2  Skin Disorders of the Neonate and Young Infant

Emollients for primary prevention 5 Silver P, Sagy M, Rubin L. Respiratory failure from corn starch aspi-
ration: a hazard of diaper changing. Pediatr Emerg Care 1996;12:
of atopic dermatitis 108–10.
Although AD rarely manifests before the age of 3–4 6 Matina F, Collura M, Maggio MC et al. Inhaled surfactant in the treat-
months, recent evidence suggests that asymptomatic neo- ment of accidental talc powder inhalation: a new case report. Ital J
nates who later develop AD have an elevated TEWL as Pediatr 2011;37:47.
7 Nikolovski J, Stamatas GN, Kollias N et  al. Barrier function and
early as 2 days and 2 months of life [26]. In filaggrin‐defi- water‐holding and transport properties of infant stratum corneum
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

cient infants, early transcutaneous sensitization to food are different from adult and continue to develop through the first
allergens is a common phenomenon [27]. Correspondingly, year of life. J Invest Dermatol 2008;128:1728–36.
8 Danby SG, Chalmers J, Brown K et  al. A functional mechanistic
early elevation of TEWL also correlates with the presence study of the effect of emollients on the structure and function of the
of food sensitization/food allergy at the age of 2 years skin barrier. Br J Dermatol 2016;175:1011–19.
[28]. It would therefore make perfect sense to prevent AD 9 Man MQ, Sun R, Man G et al. Commonly employed african neonatal
and food allergy by improving the defective skin barrier skin care products compromise epidermal function in mice. Pediatr
Dermatol 2016;33:493–500.
in presymptomatic neonates immediately after birth. The 10 Danby SG, Al‐Enezi T, Sultan A et al. The effect of aqueous cream BP
effectiveness of a preventive barrier therapy in neonates on the skin barrier in volunteers with a previous history of atopic der-
at high risk of developing AD – as deduced from the fam- matitis. Br J Dermatol 2011;165:329–34.
11 Danby SG, Al‐Enezi T, Sultan A et al. Effect of olive and sunflower
ily history – was studied in two RCTs from Japan [29] and
seed oil on the adult skin barrier: implications for neonatal skin
the UK [30], with 118 and 108 healthy neonates, respec- care. Pediatr Dermatol 2013;30:42–50.
tively, who were randomized to either receive daily emol- 12 Concin N, Hofstetter G, Plattner B et al. Evidence for cosmetics as a
lient care for 6 months, or not. The primary endpoint was source of mineral oil contamination in women. J Womens Health
(Larchmt) 2011;20:1713–19.
the cumulative incidence of AD at week 32 and 24, respec- 13 Niederer M, Stebler T, Grob K. Mineral oil and synthetic hydrocar-
tively. This was 32% less in the treated group (P = 0.12) in bons in cosmetic lip products. Int J Cosmet Sci 2016;38:194–200.
the first [29], and 50% less (OR 0.50, CI 0.28–0.9, P = 0.17) 14 Mondello L, Zoccali M, Purcaro G et al. Determination of saturated‐
in the second [30] study. No effect on food sensitization hydrocarbon contamination in baby foods by using on‐line liquid‐gas
chromatography and off‐line liquid chromatography‐comprehensive
could be demonstrated. gas chromatography combined with mass spectrometry. J Chromatogr
The results of these pilot studies have been received A 2012;1259:221–6.
with enthusiasm. However, many questions still need to 15 Miest RY, Yiannias JA, Chang YH, Singh N. Diagnosis and preva-
lence of lanolin allergy. Dermatitis 2013;24:119–23.
be answered before preventive barrier therapy can and 16 Jacob SE, McGowan M, Silverberg NB et  al. Pediatric Contact

should be recommended for all newborns at risk: Dermatitis Registry Data on Contact Allergy in Children With Atopic
• Type of emollient: Different emollients have been used in Dermatitis. JAMA Dermatol 2017;153:765–70.
the studies mentioned. With respect to potential haz- 17 Copeland CA, Raebel MA, Wagner SL. Pesticide residue in lanolin.
JAMA 1989;261:242.
ards associated with the long‐term use of emollients as 18 Wester RC, Sedik L, Melendres J et  al. Percutaneous absorption of
outlined previously, the risks of unnecessarily exposing diazinon in humans. Food Chem Toxicol 1993;31:569–72.
many asymptomatic neonates to agents that can be sen- 19 Cooke A, Cork MJ, Victor S et al. Olive oil, sunflower oil or no oil for
baby dry skin or massage: a pilot, assessor‐blinded, randomized con-
sitizing, accumulate or induce paradoxical skin dry-
trolled trial (the Oil in Baby SkincaRE [OBSeRvE] Study). Acta Derm
ness must be weighed up against noticeable long‐term Venereol 2016;96:323–30
benefits which still need to be demonstrated. 20 Wohlrab J, Klapperstück T, Reinhardt HW, Albrecht M. Interaction of
• Definition of risk group: It is obvious that the more spe- epicutaneously applied lipids with stratum corneum depends on the
presence of either emulsifiers or hydrogentated phosphatidylcholine.
cifically risk group(s) can be defined, the more effective Skin Pharmacol Physiol 2010;23:298–305.
(and justifiable) are the preventive measures. Based on 21 Wolf G, Höger PH. [Hypoallergenic and non‐toxic emollient therapies
recent studies [26,28], it is likely that early assessment for children.] J Dtsch Dermatol Ges 2009;7:50–60.
of TEWL in addition to a positive family history would 22 Bruns DE, Herold DA, Rodeheaver GT, Edlich RF. Polyethylene gly-
col intoxication in burn patients. Burns 1982;9:49–52.
significantly enhance the predictive power and thus 23 European Medicines Agency (EMA). Propylene glycol in medicinal
limit the number needed to treat (number of neonates products for children. Assessment report EMA/175205/2014. London:
needed to receive preventive barrier therapy in order to EMA, 2014.
24 Irvin EJ, Miller HD. Emollient use in the term newborn: a literature
reduce the incidence of AD and food allergy). review. Neonatal Network 2015;34:227–30.
• Duration of protective effects: It is unclear at present 25 Garcia Bartels N, Scheufele R, Prosch F et al. Effect of standardized
whether preventive barrier therapy indeed reduces the skin care regimens on neonatal skin barrier function in different body
manifestation rate of AD or just postpones it to a later areas. Pediatr Dermatol 2010;27:1–8.
26 Kelleher M, Dunn‐Galvin A, Hourihane JO et al. Skin barrier dys-
stage. function measured by transepidermal water loss at 2 days and 2
months predates and predicts atopic dermatitis at 1 year. J Allergy
Clin Immunol 2015;135:930–5.
­References 27 Brown SJ, Asai Y, Cordell HJ et  al. Loss‐of‐function variants in the
1 Gomez‐Berrada MP, Gautier F, Parent‐Massin D, Ferret PJ.
filaggrin gene are a significant risk factor for peanut allergy. J Allergy
Retrospective exposure data for baby and children care products: an Clin Immunol 2011;127:661–7.
analysis of 48 clinical studies. Food Chem Toxicol 2013;57:185–94. 28 Kelleher MM, Dunn‐Galvin A, Gray C et al. Skin barrier impairment
2 Ficheux AS, Dornic N, Bernard A et  al. Probabilistic assessment of at birth predicts food allergy at 2 years of age. J Allergy Clin Immunol
exposure to cosmetic products by French children aged 0‐3 years. Food 2016;137:1111–16.
Chem Toxicol 2016;94:85–92. 29 Horimukai K, Morita K, Narita M et al. Application of moisturizer to
3 Manová E, von Goetz N, Keller C et al. Use patterns of leave‐on per- neonates prevents development of atopic dermatitis. J Allergy Clin
sonal care products among Swiss‐German children, adolescents and Immunol 2014;134:824–830.
adults. Int J Environm Res Publ Health 2013;10:2778–98. 30 Simpson EL, Chalmers JR, Hanifin JM et al. Emollient enhancement of
4 Cetta F, Lambert GH, Ros SP. Newborn chemical exposure from over‐ the skin barrier from birth offers effective atopic dermatitis preven-
the‐counter skin care products. Clin Pediatr (Phila) 1991;30:286–9. tion. J Allergy Clin Immunol 2014;134:818–23.
Chapter 5  Neonatal Skin Care 71

­Percutaneous absorption the endocrine disrupting actions of triclosan, parabenes


and several chemical ultraviolet (UV) filters [3–6] which
Immaturity of the epidermal barrier in infants is not only have been shown to exert mainly oestrogenic activity in
associated with increased TEWL and skin dryness but vitro. Early (or even prenatal) exposure to triclosan, para-
also brings an increased risk of percutaneous resorption benes, phthalates and other chemical compounds can
of toxic substances applied to the skin surface. Skin per- contribute to premature puberty [7]. In addition to avoid-
meability is inversely proportional to gestational age [1].

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
ance of high‐level UV exposure and appropriate clothing,
Even in the term infant, transcutaneous absorption is UV protection in young children should preferably be
more readily achieved because the body surface area to based on mineral agents such as micro‐ (not nano‐) molec-
weight ratio is two‐ to threefold higher than in older chil- ular titanium dioxide and zinc dioxide.
dren and adults [2]. Low‐molecular‐weight chemicals
(<800 Da) penetrate more easily. Topical antiseptics (hexa- ­References
chlorophene, iodine) and antibiotics (particularly neomy- 1 Barker N, Hadgraft J, Rutter N. Skin permeability in the newborn.
cin, which is highly ototoxic), alcohol dressings, salicylates, J Invest Dermatol 1987;88:409–11.
urea and others have all been associated with neonatal 2 Rutter N. Percutaneous drug absorption in the newborn: hazards and
uses. Clin Perinatol 1987;14:911–30.
toxicity caused by transcutaneous resorption, particularly 3 Diamanti‐Kandarakis E, Bourguignon JP, Giudice LC et  al.

in the preterm infant [1]. The likelihood of percutaneous Endocrine‐disrupting chemicals: an Endocrine Society scientific
absorption is higher in areas with high numbers of seba- statement. Endocr Rev 2009;30:293–342.
4 Schlumpf M, Cotton B, Conscience M et al. In vitro and in vivo estro-
ceous glands per cm2 (head, napkin area) and in intertrigi- genicity of UV screens. Environ Health Perspect 2001;109:239–44.
nous areas due to the occlusive effect. The vehicle also 5 Ozáez I, Martínez‐Guitarte JL, Morcillo G. Effects of in vivo exposure
affects percutaneous absorption. Lipophilic agents can to UV filters (4‐MBC, OMC, BP‐3, 4‐HB, OC, OD‐PABA) on endocrine
penetrate the lipid bilayer better than water‐soluble com- signaling genes in the insect Chironomus riparius. Sci Total Environ
2013;456‐457:120–6.
pounds. Their occlusive effects lead to enhancement of 6 Schreurs RH, Sonneveld E, Jansen JH et  al. Interaction of polycyclic
epidermal hydration which in turn widens the intracellu- musks and UV filters with the estrogen receptor (ER), androgen recep-
lar bonds and facilitates absorption. The risk of toxicity is tor (AR), and progesterone receptor (PR) in reporter gene bioassays.
Toxicol Sci 2005;83:264–72.
further compounded by immature metabolic mechanisms
7 Harley KG, Berger KP, Kogul K et al. Association of phthalates, para-
of detoxification. Table 5.4 summarizes potential hazards bens and phenols found in personal care products with pubertal tim-
of common topically applied drugs. Of particular note are ing in girls and boys. Human Reproduction 2019;34:109–17.

Table 5.4  Potential hazards of percutaneously absorbed agents

Compound Function Toxicity

Alcohols Topical antiseptic Cerebral and hepatic damage, cutaneous haemorrhagic necrosis
Benzocaine Topical anaesthetic Methaemoglobinaemia
Calcipotriol Topical vitamin D3 analogue Hypercalcaemia
Clioquinol Topical antiseptic Neurotoxicity (subacute myelo‐ophthalmoneuritis, SMON)
Diphenhydramine Topical antipruritic Sedation, central anticholinergic syndrome
EMLA Topical anaesthetic mixture Methaemoglobinaemia
Gentamicin Topical antibiotic Neuro‐, oto‐, nephrotoxic
Lindane Topical scabicide Neurotoxic
Neomycin Topical antibiotic Neuro‐, oto‐, nephrotoxic
Contact allergen
N,N‐dimethyl‐m‐toluamide (DEET) Insect repellant Neurotoxicity
Parabene Preservative Endocrine disrupting agent
Phenolic compounds (pentachlorophenol, Topical antiseptic Neurotoxicity, tachycardia, metabolic acidosis,
hexachlorophene, resorcinol) methaemoglobinaemia, death
Povidone–iodine Antiseptic Hypothyroidism
Prilocaine Topical anaesthetic Methaemoglobinaemia
Salicylic acid Keratolytic Metabolic acidosis, seizures
Silver sulfadiazine Topical antibiotic Kernicterus, agranulocytosis, argyria
TCS (potent) Anti‐inflammatory Adrenal suppression, Cushing’s syndrome, skin atrophy, acne
Triclosan Antiseptic Endocrine disrupting agent
UV filtersa UV protection Endocrine disrupting agents

TCS, topical corticosteroid.


a
 UV filters with potential oestrogenic side‐effects: benzophenone‐3, ethylhexyl‐methoxy‐cinnamate (EHMC), octocrylene.
72 

CHA PTER 6

Transient Skin Disorders in the Neonate


and Young Infant
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Margarita Larralde1,2 & Maria Eugenia Abad1,2


1
 Dermatology Department, Hospital Alemán, Buenos Aires, Argentina
2
 Pediatric Dermatology Department, Hospital Ramos Mejía, Buenos Aires, Argentina

Introduction, 72 Transient vesicopustular eruptions, 76 Miscellaneous, 80


Physiological skin findings, 72 Oral lesions, 77
Transient vascular physiological changes, 75 Pigmentary skin lesions, 79

Abstract (erythema toxicum neonatorum and transient neonatal ­pustular


melanosis), oral lesions (Epstein pearls, Bohn nodules, dental
lamina cysts, natal and neonatal teeth, eruption cyst and sucking
Cutaneous lesions are common in the neonatal period. Most are
pads of the lips), pigmentary lesions (Mongolian spots or congeni-
physiological, transient and benign. Transient skin disorders may
tal dermal melanocytosis, pigmentary lines of the newborn and
be classified as physiological skin disorders (vernix caseosa, phys-
periungual hyperpigmentation) and miscellaneous lesions (subcu-
iological desquamation, lanugo, sebaceous gland hyperplasia,
taneous fat necrosis, sucking blisters and erosions, adnexal polyps,
milia, physiological jaundice, and the collective effects of maternal
pedal papules of infancy and perineal groove). In this chapter we
or placental hormones resulting in what has been referred to as
discuss the categories mentioned above and also consider that
miniature puberty), transient vascular physiological changes (cutis
serious and potentially life‐threatening diseases or associations
marmorata, salmon patch, harlequin colour change, erythema
should sometimes be ruled out.
neonatorum and acrocyanosis), transient vesicopustular eruptions

Key points eruptions, oral lesions, pigmentary lesions and miscellaneous


lesions.
• Transient skin disorders should be differentiated from serious
• Most cutaneous lesions in the neonatal period are physiological,
and potentially life‐threatening diseases, e.g. infections or
transient and benign.
genodermatoses.
• Transient skin disorders include physiological skin disorders,
transient vascular physiological changes, transient vesicopustular

Introduction Most cutaneous lesions in neonates represent transient


skin disorders characterized by benign and self‐limiting
Cutaneous lesions are common in the neonatal period. conditions (Box 6.1).
Most are benign, transient or physiological conditions
that should be differentiated from serious and potentially
Physiological skin findings
life‐threatening diseases that carry more significant risks
of morbidity or mortality, as might be seen with infections Vernix caseosa
or genodermatoses. For this reason, initial evaluation This white, greasy and lipophilic natural skin barrier is
should include a careful physical examination and a present at birth. It is synthesized by fetal keratinocytes
complete clinical history focusing on family and prenatal and sebaceous glands in late pregnancy, and formed by
history, pregnancy, delivery, gestational age, birthweight, sebaceous secretions and shed epithelial cells and lanugo
and the presence of general symptoms and specific [4]. It may cover the entire cutaneous surface of full‐term
anomalies. When needed, useful diagnostic tools include babies or may be found concentrated in skin folds and on
routine laboratory (blood chemistry and haematological the back. After a few hours or days of life, it disappears.
profile) testing; screening for infections through bacterial, Vernix caseosa may be brownish‐yellow if there was
viral and mycological smears, cultures, antibody detec­ contact with meconium or as might be seen in cases of
tions and polymerase chain reaction (PCR); skin biopsy; haemolytic disease. Vernix caseosa may have a noticeable
and genetic testing [1–3]. typical odour in a case of neonatal sepsis [4–6]. It consists

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 6  Transient Skin Disorders in the Neonate and Young Infant 73

Box 6.1  Transient skin disorders

Physiological skin disorders • Eosinophilic pustular folliculitis


• Vesiculopustular eruption in neonatal transient myeloproliferative disorder
• Vernix caseosa
• Physiological desquamation
Oral lesions

SECTION 2: SKIN DISORDERS


• Lanugo

OF NEONATES AND INFANTS


• Sebaceous gland hyperplasia • Epstein pearls
• Milia • Bohn nodules
• Miniature puberty • Dental lamina cysts
• Physiological jaundice • Natal and neonatal teeth
• Eruption cyst
Transient vascular physiological changes • Sucking pads of the lips

• Cutis marmorata
Pigmentary skin lesions
• Salmon patch
• Harlequin colour change • Mongolian spots
• Erythema neonatorum and acrocyanosis • Pigmentary lines of the newborn
• Periungual hyperpigmentation
Transient vesicopustular eruptions
Miscellaneous
• Erythema toxicum neonatorum
• Transient neonatal pustular melanosis • Subcutaneous fat necrosis
• Miliaria • Sucking blisters, erosions and calluses
• Benign neonatal cephalic pustulosis • Adnexal polyp
• Neonatal acne • Pedal papules of infancy
• Infantile acropustulosis • Perineal groove

mainly of water (81%), but also contains lipids (9%) and


proteins (10%) [7,8]. Vernix caseosa protects neonatal skin
and facilitates the transition from intrauterine to extrau­
terine life. Other functions are skin surface adaptation
after birth, temperature regulation, skin hydration,
wound healing, prevention of water loss and antimicro­
bial action. Vernix caseosa contains several antimicrobial
peptides and proteins (lysozyme, lactoferrin, cathelici­
dins and defensins) that may protect the fetus prenatally
and also facilitate postnatal skin colonization by non­
pathogenic bacteria [4,5,8,9]. Vernix caseosa also acts as a
lubricant to facilitate passage through the birth canal.
Although the traditional practice has been to wipe off the
vernix caseosa after birth, more current recommendations
advocate leaving it in place and avoidance of removal by
wiping or bathing [5,10].

Physiological desquamation
A fine desquamation that occurs in most newborns may Fig. 6.1  Lanugo on the dorsum.
persist during the first 3 months of life [11]. In healthy
full‐term neonates this process begins on the first or second Lanugo is often located on the dorsum, shoulders, face
day of life and in preterm infants it starts at 2–3 weeks of and scalp (Fig.  6.1). The first coat of lanugo normally
age. Desquamation is more evident on hands, feet and sheds in utero during the last trimester of gestation, and
ankles. In post‐term newborns it tends to be more gener­ consequently it is more prominent in preterm newborns
alized and thicker [2,11]. In extreme cases in which des­ [8,12]. Lanugo must be differentiated from congenital
quamation is persistent or severe, consideration should hypertrichosis lanuginosa, gingival fibromatosis with
be given to evaluation for other underlying disorders hypertrichosis, Cornelia de Lange syndrome and other
such as congenital syphilis or ichthyosis [11,12]. rare disorders associated with excess body hair [4,6,8,12].

Lanugo Sebaceous gland hyperplasia


Newborn skin is often covered by fine, soft, nonpig­ Sebaceous gland hyperplasia is found at birth in 50–89%
mented and unmedullated immature hair termed lanugo. of newborns, especially full‐term neonates [10,13,14].
It is replaced by vellus hair during the first months of life. Both sexes are affected equally, with the highest incidence
74 Section 2  Skin Disorders of the Neonate and Young Infant

in African Americans and the lowest in Asians [10,15].


Box 6.2  Genodermatoses associated with milia
It is produced by the increased activity of the sebaceous
gland stimulated by transplacental transfer of maternal
Primary congenital milia
androgens. It is characterized by multiple tiny whitish‐
yellow papules located at the opening of pilosebaceous • Orofaciodigital syndrome type 1
follicles in areas in which sebaceous glands are prominent • Familial milia with absent dermatoglyphics (Basan syndrome)
SECTION 2: SKIN DISORDERS

• Familial profuse congenital milia


OF NEONATES AND INFANTS

such as around the nose, cheeks, forehead and upper lip


[6,7,13]. No treatment is needed since it generally resolves
spontaneously within the first few weeks after birth. Primary milia developing later in life

Differential diagnosis includes milia, miliaria cristallina • Basal cell naevus syndrome (Gorlin syndrome)
and neonatal acne [10]. • Generalized basaloid follicular hamartoma syndrome
• Brooke–Spiegler syndrome
Milia • Basex–Dupré–Christol syndrome
Milia are small superficial keratinous cysts found com­ • Rombo syndrome
monly on newborn skin, produced by retention of keratin • Pachyonychia congenita
within the superficial dermis. They originate from the • Atrichia with papular lesions
pilosebaceous apparatus of vellus hair [6,13]. Milia may • Nicolau–Balus syndrome
be divided into two categories: primary milia which arise • Keratitis–ichthyosis–deafness syndrome (KID syndrome)
spontaneously, and secondary milia due to medications,
Secondary milia
diseases and cutaneous healing after trauma. Neonatal
milia are included in the first subtype [16,17]. They occur • Epidermolysis bullosa
in 40–50% of neonates, with no racial or gender prepon­ • Porphyria
derance. Milia are multiple superficial pinpoint white‐
yellowish papules 1–2 mm in diameter located on the
face, especially the nose, but also cheeks, forehead and
chin (Fig. 6.2). Scalp, upper trunk and upper limbs may
also be affected. There may be single or multiple lesions.
In some cases, a solitary and usually larger milium may
be present on the areola, genitalia or foreskin [13]. They
disappear spontaneously, usually within a few weeks of
life without scarring, although they may persist for several
months. In cases where profuse and persistent milia are
noted or associated with other anomalies, an underlying
genodermatosis must be suspected (Box 6.2) [6,13,17–19].

Miniature puberty
Maternal hormones, especially androgens, are acquired
transplacentally, producing a neonatal transient hormo­
nal elevation that normalizes by 6–8 weeks of age.
Miniature puberty comprises a group of spontaneously
resolving manifestations secondary to this phenomenon
[6,13,20]. Darkening of the linea alba (linea nigra), areolas
and external genitalia is frequently seen, especially in Fig. 6.3  Miniature puberty: hypertrophic and darkened genitalia in a
female newborn.
non‐Caucasian infants [21]. Breast hypertrophy may

occur in both sexes, as may neonatal acne and sebaceous


gland hyperplasia [20]. The enlarged breast gland may
secrete a colostrum‐like substance. Male genitalia may
appear well‐developed with hyperpigmentation of scro­
tum. Female genitalia may appear full with clitoral hyper­
trophy and darkening of the labia and vulva (Fig.  6.3).
Within a few days after birth, a whitish, creamy vaginal
discharge can be seen. Rarely, on the third to fourth day of
life, bleeding similar to menses may occur [6,13,20,21].
Hyperpigmentation of labia may occasionally be misdi­
agnosed as naevi.

Physiological jaundice
Jaundice is produced by the accumulation of bilirubin in
Fig. 6.2  Milia: tiny whitish cysts. the skin, mucous membranes and sclerae that causes a
Chapter 6  Transient Skin Disorders in the Neonate and Young Infant 75

yellowish colouration. Physiological jaundice refers to the Adams–Oliver syndrome, phakomatosis pigmentovas­
common and in general harmless jaundice seen in babies cularis and van Lohuizen syndrome [24,25]. Rarely,
during the first days of life, with no significant underly­ neonatal lupus erythematosus may present with cutis
ing disease. It is common in the first week, affecting marmorata telangiectatica congenita‐like lesions. Reticu­
60% of term and 80% of preterm newborns. Up to 30% late capillary malformations (port wine stains) may also
of predominantly breastfed babies are still jaundiced at resemble cutis marmorata; however, these reticulate cap­

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
1 month of age [4,6,22]. illary malformations are persistent. Extensive reticulate
Red blood cell breakdown produces unconjugated or capillary malformations or congenital livedo reticularis‐
indirect bilirubin which is mostly bound to albumin as it like findings in conjunction with overgrowth or macro­
circulates. Unconjugated bilirubin is metabolized in the cephaly represent some of the cutaneous hallmarks of
liver to produce conjugated or direct bilirubin which macrocephaly–capillary malformation [26,27] and other
passes into the gut and is excreted in stool. Hepatic PIK3‐related overgrowth syndromes.
immaturity associated with a shorter lifespan and higher
concentration of red blood cells is responsible for hyper­ Salmon patch
bilirubinaemia [22]. Also known as naevus simplex, Unna naevus, ‘angel kiss’
Early‐onset or prolonged jaundice may be a sign of or ‘stork bite’, this is the most common vascular stain pre­
pathological jaundice. Pathological jaundice has many sent at birth. It affects almost half of newborns with no
aetiologies including blood group incompatibility gender predilection. It involves the median part of the
(Rhesus or ABO incompatibility), as well as other causes face (glabella, forehead, eyelids, nasal alae and philtrum),
of haemolysis, such as large haematomas (hepatic sub­ the nape and occipital area and in some cases the sacral
capsular or cephalohaematoma), sepsis, in addition region (Fig. 6.4) [26]. Its colour typically varies from pale
to  primary liver disease (Gilbert and Crigler–Najjar pink to bright red with undefined borders that become
syndrome), biliary atresia and glucose‐6‐phosphate more prominent with heat, crying and physical activity.
dehydrogenase deficiency [4,22,23]. Facial lesions usually fade within the first 2 years of life.
Lesions involving the nape may persist [28,29]. Patients
with lumbosacral naevus simplex should have imaging
Transient vascular physiological performed to rule out underlying spinal dysraphism if
changes the naevus is extensive or atypical, or other cutaneous
Cutis marmorata anomalies such as lipoma, hypertrichosis or dermal sinus,
Cutis marmorata is a frequent, benign and generalized among others, are also present [12,29].
cutaneous vasomotor phenomenon induced by hypother­
mia. The immature autonomic nervous system is respon­
Harlequin colour change
This transient and sudden, uncommon phenomenon
sible for an irregular distribution of superficial capillary
consists of sharply demarcated erythema on one half of
blood flow [6,8]. Although it is more common in preterm
the body with simultaneous blanching on the contralat­
infants, full‐term babies may also be affected. It is charac­
eral half. A line of demarcation along the midline is well‐
terized by a transient, blanchable, bluish‐red, reticulated
defined. The change of colour, which generally fades
mottling that in general lasts minutes, aggravated by cold
within 30 seconds to 20 minutes, appears between the
and abated by warm temperatures. Physiological cutis
second and fifth days after birth, but may occur up to the
marmorata improves spontaneously within the first few
third week [8,30,31]. Although harlequin colour change
weeks [4,6,8].
may arise as a single episode, it sometimes reappears. It is
Persistent or severe cutis marmorata has been reported
in several diseases including Down syndrome, trisomy
18, congenital hypothyroidism, homocystinuria, Divry–
Van Bogaert syndrome and Cornelia de Lange syndrome
[4,24,25].
Cutis marmorata telangiectatica congenita is a rela­
tively uncommon capillary malformation that can mimic
physiological cutis marmorata but does not disappear
with warming. It is characterized by deep purple reticular
or stellate areas of mottling, is present at or shortly after
birth, and may have a localized, segmental or generalized
distribution. Atrophy or skin ulceration and loss of
underlying subcutaneous fat, as well as prominent
veins, telangiectasias and hyperkeratosis may be seen.
Extracutaneous findings such as growth and develop­
mental delay, discrepancies in length and circumference
of limbs, glaucoma, macrocephaly, neurological abnor­
malities and other vascular anomalies have been reported
in 20–80% of patients. Cutis marmorata telangiectatica
congenita may be present in some disorders such as Fig. 6.4  Salmon patch: erythematous macules on the nose and philtrum.
76 Section 2  Skin Disorders of the Neonate and Young Infant

most commonly observed when the baby is in a lateral


decubitus position. The colour characteristically changes
location when the baby is rotated [6,30]. Newborns with
harlequin colour change are in good general condition
with no other signs or symptoms associated. Although it
is observed in healthy newborns, it has also been reported
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

in the context of hypoxia, such as prematurity or low


birthweight. Exacerbation of the phenomenon has been
reported in patients with congenital cyanotic cardiomyo­
pathy receiving prostaglandin E1 treatment [30,32]. Its
aetiopathogenesis is poorly understood, but it has been
suggested that hypothalamic immaturity produces a
transient imbalance in cutaneous vessel regulation [30,32].

Erythema neonatorum and acrocyanosis


Generalized erythema or rubor neonatorum is a physio­
logical condition present from the first hours until the sec­ Fig. 6.5  Erythema toxicum neonatorum: erythematous macules and
ond day of life. Its mechanism is related to cutaneous pustules.
vasodilation and hyperaemia. Neonatal polycythaemia is
an associated factor that contributes to erythema [4,6]. Although this rash may be present at birth, it usually
Acrocyanosis is characterized by bilateral and symmet­ begins 2–3 days after birth. Typical lesions consist of
rical purple‐bluish colouration of hands and feet as well erythematous macules 2–3 cm in diameter with a central
as the perioral area. It is intermittent, may intensify with papule or pustule. Lesions can vary in number from one
vigorous crying, cold, polycythaemia or other hypervis­ to several and be located on the face, trunk and proximal
cosity syndromes, and may revert with warmth. In gen­ extremities, without involvement of palms and soles
eral it resolves within the first week of life [4,6,8]. Its (Fig. 6.5). The lesions are highly evanescent and usually
pathogenesis is not well understood, but it is proposed evolve in crops that wax and wane, with spontaneous
that cutaneous vasomotor instability related to immatu­ resolution of individual lesions within hours to days.
rity is involved. Blood flow is reduced in dilated subpap­ Spontaneous involution of the eruption occurs within
illary and papillary venous vessels favouring oxygen 1–2 weeks without sequelae, although lesions may recur
release in peripheral tissues and subsequent formation of [3,34,36,38].
desaturated haemoglobin [4]. It should be distinguished Diagnosis is made clinically. Cytological examination
from Raynaud’s phenomenon and other disorders associ­ of a pustular smear with Giemsa or Wright staining dem­
ated with central cyanosis such as congenital cardiac dis­ onstrates eosinophilia. Skin biopsy, if performed, shows a
ease or respiratory diseases [33]. Since this condition is dense inflammatory infiltrate around hair follicles com­
benign and self‐healing, no treatment is required. posed mostly of eosinophils and subcorneal pustules.
Peripheral eosinophilia may be present [34,37]. Differential
diagnosis includes transient neonatal pustular melanosis,
Transient vesicopustular eruptions miliaria and eosinophilic pustular folliculitis, as well as
Erythema toxicum neonatorum infections including congenital candidiasis, staphylococ­
Erythema toxicum neonatorum is a benign and self‐ cal impetigo and neonatal herpes simplex [3,34,36].
limited condition with no racial, gender or seasonal As erythema toxicum neonatorum is a self‐limiting
predilection. Its incidence varies from 16.7% to 55% in condition, no treatment is required. However, atypical
different reports [34,35]. Higher incidence rates are seen presentations often appropriately prompt further evalua­
in term neonates with birthweight over 2500 g. It is rarely tion to rule out an underlying infection.
seen in preterm infants and its presence in this population
should prompt an evaluation for underlying infection. Transient neonatal pustular melanosis
Different hypotheses have been proposed to explain This benign, transient disorder is found in 0.2‐–4% of full‐
the pathogenesis of erythema toxicum, including allergic term neonates. It is more common in more dark‐skinned
response to transplacental or environmental allergens; infants, but there is no sex predilection. It is characterized
response to mechanical, chemical or thermal irritation; by fragile pustules or vesicles without surrounding ery­
hormonal influences on the extracellular matrix; and a thema that are almost always present at birth. Lesions
graft‐versus‐host reaction triggered by maternal lympho­ measure 1–5 mm in diameter, are single or grouped in
cytes transferred to the infant before or during delivery clusters, and may occur anywhere but are commonly seen
[3,34,36]. Currently it is accepted that pro‐inflammatory on the forehead, mandibular area, neck, trunk, buttocks,
mediators (IL‐1, IL‐8, eotaxin, aquaporins 1 and 3, psoria­ thighs, palms and soles (Fig. 6.6). The vesicopustules are
sin, nitric oxide synthases 1, 2 and 3) identified in the very fragile and break down easily, leaving pigmented
erythema toxicum infiltrate reflect an immunological macules surrounded by fine, white collarettes of scale.
cutaneous reaction to microbial skin colonization of hair Brownish macules may persist for months, but in general
follicles from birth [37]. fade spontaneously within a few weeks. In some cases,
Chapter 6  Transient Skin Disorders in the Neonate and Young Infant 77

Epstein pearls
Epstein pearls are single or multiple, small (1–3 mm),
whitish keratin‐filled cysts, located at the junction of the
hard and soft palates or at the palatal midline (Fig. 6.7).
They have been reported in 65–85% of newborns, and
arise from epithelial remnants along the line of fusion of

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
palatal components during embryogenesis. No treatment
is indicated, as they regress spontaneously within the first
several weeks after birth. They are considered analogous
to milia [10,44,45].

Bohn nodules
Bohn nodules are isolated or multiple firm small white
cystic structures located on the vestibular and lingual
surface of the alveolar ridges (Fig. 6.8). The maxillary arch
is more commonly affected than the mandibular. These
lesions are very common, reported in 85% of neonates.
They are keratin cysts derived from epithelial remnants of
minor salivary glands or from remnants of odontogenic

Fig. 6.6  Transient neonatal pustular melanosis: grouped pustules without


surrounding erythema on the scalp.

the vesicular stage develops in utero and the newborn


presents with only hyperpigmented macules at birth
[3,36,39,40].
The differential diagnosis includes erythema toxicum
neonatorum, staphylococcal pustulosis, congenital can­
didiasis, syphilis, herpes simplex and acropustulosis of
infancy.
Giemsa or Wright staining of pustular content reveals
neutrophils and occasional eosinophils. No organism is
observed and cultures are negative. Skin biopsy of pustu­
lar lesions shows an intracorneal or subcorneal collection
of neutrophils; in contrast to classical erythema toxicum,
there are few eosinophils [3,7,36].
Based on similarities in clinical findings and the pres­
ence of both neutrophils and eosinophils in the skin
biopsy, as well as the coexistence of lesions of erythema
toxicum neonatorum and transient neonatal pustular
melanosis in the same patient, it has been postulated that Fig. 6.7  Epstein pearls: multiple small whitish palatal cysts.
these disorders may be variations of the same disease.
The term sterile transient neonatal pustulosis was pro­
posed by Ferrandiz et  al. to include these overlapping
conditions [41,42].

Oral lesions
Lesions affecting the oral cavity of infants may vary from
benign transient lesions to tumours. Knowledge of these
common conditions is important for appropriate manage­
ment of patients and their families.
Transient inclusion cysts or developmental nodules of
the oral mucosa are very frequent, affecting almost 80% of
newborns. They represent the most common oral disor­
der in infants, and are located in the alveolar ridges or on
the palate. Based on histological origin and location in the
oral cavity, they can be classified as Epstein pearls, Bohn Fig. 6.8  Bohn nodules: small white cystic structures on the surface of
nodules or dental lamina cysts [43,44]. alveolar ridges.
78 Section 2  Skin Disorders of the Neonate and Young Infant

epithelium over the dental lamina [43,46]. Differential


diagnosis includes Epstein pearls, natal or neonatal teeth
and dental lamina cysts. Spontaneous rupture and sub­
sequent involution of the cysts within a few weeks is the
rule. No treatment is needed [43,45–47].
SECTION 2: SKIN DISORDERS

Dental lamina cysts


OF NEONATES AND INFANTS

Also known as gingival cysts of the newborn, they occur


on the crest of alveolar ridges, and derive from remnants
of the dental lamina. They may be single or multiple, and
are located mainly on the maxilla. Clinically, they are
small (1–3 mm), pearly, firm papules observed in 25–53%
of newborns [44,48]. These keratin‐filled cysts disappear
within the first weeks of life.

Natal and neonatal teeth


Normal eruption of primary teeth usually occurs around Fig. 6.9  Partially erupting natal tooth and ulceration of the tongue (Riga–
6 months of age. Premature eruption of teeth is a rare Fede disease).
condition; depending on the time of eruption such teeth
are defined as natal teeth when present at birth, and neo­ Clinically, natal and neonatal teeth are typically poorly
natal teeth when they erupt during the first month. The developed, loose and conical, with yellowish‐brown or
incidence of natal and neonatal teeth varies from 1 : 716 whitish opaque discolouration, and have hypoplastic
to 1 : 3500 live births, with no gender predilection [49,50]. enamel. Occasionally they are normal in size, shape and
Natal teeth are approximately three times more frequent colour (Fig. 6.9). Natal and neonatal teeth may be classi­
than neonatal teeth. Most natal and neonatal teeth repre­ fied as mature when they are nearly or fully developed
sent early eruption of the normal primary deciduous den­ and immature when their structure is incomplete, leading
tition, whereas less than 10% are supernumerary teeth to a poor prognosis [49,50]. The most common location of
[45,49,51]. The exact pathogenesis is not known, but it has natal and neonatal teeth is mandibular central incisors
been suggested that premature eruption of primary teeth (85%), followed by maxillary incisors (11%), mandibular
may be related to superficial location of the developing canines or molars (3%) and maxillary canines or molars
tooth germ in the alveolar bone [51]. It has also been (1%) [45,50,52].
hypothesized that increased resorption of overlying bone Radiographic examination helps to differentiate between
results in premature tooth eruption [49,52]. Autosomal supernumerary primary teeth and teeth of the normal
dominant inheritance has been reported in some cases, dentition, as well as Bohn nodules and dental lamina
as have several predisposing factors such as hormonal cysts, the major differential diagnosis.
disturbances (pituitary, thyroid), poor maternal health, Extraction is the treatment of choice in the case of
febrile episodes during pregnancy and environmental supernumerary teeth as they may interfere with normal
factors (including polyhalogenated aromatic hydrocar­ tooth eruption. Excessively mobile teeth should be
bons) [49–52]. Some syndromes are associated with natal extracted to prevent the risk of exfoliation and subse­
and neonatal teeth (Table 6.1) [10,45,49,51,53]. Natal and quent swallowing or aspiration [45,49]. A major compli­
neonatal teeth have been reported in infants with con­ cation is the development of a traumatic ulceration on
genital hypothyroidism and in the context of cleft lip the ventral surface of the tongue, lips or mother’s breast,
and palate [54,55]. which represents a sign of Riga–Fede disease and associ­
ated pain insensitivity. The lesion begins as an eroded
area, evolving into an enlarged granulomatous ulcerated
Table 6.1  Syndromes associated with natal and neonatal teeth
mass. Discomfort during breastfeeding with subsequent
refusal to eat, malnutrition and dehydration may be asso­
Syndrome Inheritance
ciated. A conservative approach is the first treatment
Pachyonychia congenita AD
option for Riga–Fede disease. Sharp tooth edges may be
Ellis–van Creveld syndrome AR polished using a finishing burr or covered with compos­
Hallermann–Streiff syndrome Isolated cases ite resin. In some cases tooth extraction may be required
Wiedemann–Rautenstrauch syndrome (neonatal AR [49,50,52,56].
progeroid syndrome)
Short‐rib thoracic dysplasia 13 with or without AR Eruption cyst
polydactyly Eruption cyst is characterized by circumscribed fluctuant
Restrictive dermopathy AR
mucosal swelling overlying a tooth during the eruption
Raine syndrome (osteosclerotic bone dysplasia) AR
Osteopathia striata with cranial sclerosis XLD process. Given that eruption cysts are related to both pri­
Epidermolysis bullosa, lethal acantholytic AR mary and permanent tooth eruption, they are rarely
observed in the neonatal period, in which case they are
AD, autosomal dominant; AR, autosomal recessive; XLD, X‐linked dominant. secondary to natal or neonatal teeth [45]. The eruption
Chapter 6  Transient Skin Disorders in the Neonate and Young Infant 79

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Fig. 6.10  Eruption cyst: bluish, dome‐shaped lesion on the alveolar ridge
of the mandible. Fig. 6.11  Mongolian spots: aberrant and superimposed.

cyst appears as a translucent, flesh‐coloured or bluish, in early to mid childhood. Superimposed darker spots on
dome‐shaped, compressible lesion on the alveolar ridge classic Mongolian spots may be present [61–64].
of the mandible or maxilla (Fig.  6.10) [45,57,58]. The Histopathology reveals the presence of scattered mel­
differential diagnosis includes mucocoele, lymphatic anocytes in the lower dermis associated with occasional
malformation, congenital epulis, dental lamina cysts, melanophages [62].
teratoma and Bohn nodules [58]. Radiological examina­ Rarely, Mongolian spots may be associated with lysoso­
tion shows a natal or neonatal tooth. Cystic content mal storage diseases. In these cases Mongolian spots are
obtained by needle aspiration has a yellowish colouration persistent, aberrant in appearance and extensive, affecting
and low viscosity and presents cholesterol crystals. not only the dorsum but also the ventral aspect of the
Although treatment options include marsupialization or trunk, with darker and progressive pigmentation. An
surgical removal, most eruption cysts resolve spontane­ extensive confetti appearance has also been observed in
ously within several weeks [45,58]. some patients. The most common lysosomal storage
disease associated with Mongolian spots is GM1 gangli­
Sucking pads of the lips osidosis 1, followed by mucopolysaccharidosis type 1
Sucking pads or sucking calluses on the lips are present (Hurler disease) and, to a lesser degree, mucopolysaccha­
in both term and preterm neonates, and are considered ridosis type 2 (Hunter disease), mucolipidosis, Niemann–
a physiological adaptive reaction of lip structures to Pick disease and mannosidosis [61,65–68]. Phakomatosis
sucking. They are characterized by painless, whitish and pigmentovascularis consists of the association of a vascu­
hyperkeratotic swelling of the lips, predominantly in the lar malformation and a cutaneous melanocytic lesion.
middle of the upper lip. Skin biopsy, if performed, shows Persis­tent Mongolian spots are part of phakomatosis
hyperkeratosis, epidermal hyperplasia and intracellular pigmentovascularis types II, IV and V, also known as
oedema. They disappear spontaneously after 3–6 months, phakomatosis cesioflammea and phakomatosis cesio­
when breastfeeding is stopped [45,59,60]. marmorata [61,62,65,69].

Pigmentary lines of newborns


Pigmentary skin lesions This rare, transient, pigmentary dermatosis is character­
Mongolian spots ized by horizontal linear hyperpigmentation on the skin
Mongolian spots (congenital dermal melanocytosis) are folds of the abdomen, back and extremities from birth.
congenital homogeneous greyish macules located on Spontaneous resolution occurs between 2 and 6 months
sacral and gluteal areas, resulting from the aberrant pres­ of age. Most reported cases were darkly‐pigmented male
ence in the lower dermis of melanocytes that failed to newborns. Pathogenesis is thought to be nonhormonal,
migrate from the neural crest to the epidermis during possibly induced by mechanical trauma related to flexion
fetal life. The prevalence varies with race, affecting around in utero, which produces slight hyperkeratosis within the
90–100% of Africans and Asians, 50% of Hispanics, and skin folds resulting in postinflammatory hyperpigmenta­
10% of Caucasians [61,62]. tion [70–72].
Clinically Mongolian spots are characterized by single
or multiple grey, blue‐grey, blue‐green or dark blue mac­ Periungual hyperpigmentation
ules of varying sizes and shapes, with irregular borders, Hyperpigmentation of the distal phalanges and the proxi­
located mainly on the sacrogluteal area. Aberrant extrasa­ mal nail fold is an occasional clinical finding in dark‐
cral locations include the lower and upper extremities, skinned newborns, although it has also been described
upper back and shoulders (Fig. 6.11). They typically fade in Caucasian babies. Pigmentation is uniform in each
80 Section 2  Skin Disorders of the Neonate and Young Infant

patient: light‐brown in fair‐skinned patients and intensely


hyperpigmented in dark‐skinned neonates. It occurs in
both fingers and toes, is present at birth in full‐term
neonates, and fades spontaneously within the second
year of life [73,74]. Its pathogenesis is unclear, although it
is speculated to be a result of melanocyte‐stimulating
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

hormone in utero [71,73].

Miscellaneous
Subcutaneous fat necrosis
Subcutaneous fat necrosis of the newborn is a rare and
transient disorder of fat tissue that affects term and post‐
term babies. It is a lobular form of panniculitis that
appears in the first week of life, few cases developing as
late as 6 weeks of age. It has been associated with various
physiological stressors as risk factors, including maternal
factors (gestational diabetes, pre‐eclampsia, smoking or
exposure to passive smoking, intake of calcium channel
blockers, cocaine abuse) and neonatal factors (perinatal
asphyxia, meconium aspiration, umbilical cord prolapse,
hypothermia, obstetrical trauma, Rhesus incompatibility)
[75–79]. Subcutaneous fat necrosis was reported in 1–3%
of neonates managed with moderate therapeutic hypo­ Fig. 6.12  Subcutaneous fat necrosis: erythematous plaques on the back.
thermia for hypoxic ischaemic encephalopathy. Lesions
may develop in areas directly exposed to the cooling
blanket. Induction of therapeutic cooling improves sur­
vival and reduces neurological sequelae in newborns
with hypoxic ischaemic encephalopathy, and has become
common practice in these cases [80–82]. Subcutaneous fat
necrosis was also documented in infants who underwent
whole‐body cooling before cardiovascular surgery [82].
Perinatal asphyxia induces blood shunting from skin and
spleen to brain, heart and adrenal glands, producing
impaired tissue perfusion with local hypoxia [78,82].
Neonatal adipose tissue has a relatively high concentra­
tion of saturated fatty acids (stearic and palmitic acids)
with a high melting point that makes them more prone to
solidify and crystallize under cold stress, resulting in adi­
pocyte necrosis and subsequent formation of granuloma­
tous inflammation [75,76,82]. Also, enzymes involved in
fatty acid metabolism are immature [83].
This condition is characterized by multiple indurated, Fig. 6.13  Subcutaneous fat necrosis (H&E): radially arranged needle‐
shaped birefringent crystals.
erythematous or violaceous painful plaques and nodules
located on the back, shoulders, extremities, buttocks and
cheeks (Fig.  6.12). Some lesions may become fluctuant Skin biopsy in subcutaneous fat necrosis demonstrates
with drainage of liquefied fat from nodules [75,78,82]. lobular panniculitis with fat necrosis, mixed inflammatory
Lesions resolve spontaneously within a few weeks, infiltrate with abundant histiocytes, giant multinucleated
leaving on rare occasions atrophy, fibrosis, scarring, cells with granuloma formation and adipocytes with
ulcers or necrosis. radially arranged needle‐shaped birefringent crystals and
The differential diagnosis most commonly includes, clefts (Fig.  6.13) [75,77]. Fine‐needle aspiration is an
but is not limited to, infection (cellulitis, abscess) and option for diagnosis.
tumours (such as infantile myofibromatosis, rhabdo­ Although subcutaneous fat necrosis is a benign condi­
myosarcoma and deep haemangioma of infancy). tion, complications such as transitory hypoglycaemia,
Sclerema neonatorum is the main differential diagnosis, hypertriglyceridaemia and thrombocytopenia have been
though it is encountered uncommonly. The latter appears reported [75,76,78,79]. Hypercalcaemia is a potentially
in preterm infants with sepsis or underlying disease in life‐threatening complication found in 25–56% of patients.
the first week of life, with generalized hardening of the It usually appears when the cutaneous lesions start to
skin except on palms, soles and genitalia. Its prognosis is regress [75,84]. Although the first 6 weeks of life is the
poor [75,77]. period of highest risk for developing hypercalcaemia,
Chapter 6  Transient Skin Disorders in the Neonate and Young Infant 81

calcium should be evaluated periodically up to 6 months


of age. The pathogenesis of hypercalcaemia is not fully
understood. The mechanism involved includes increased
prostaglandin E2 activity leading to osteoclast activation,
direct calcium release from necrotic fat tissue and secre­
tion of 1,25‐dihydroxyvitamin D3 from granulomas

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
which produces increased intestinal calcium absorption
[76,78,83,85]. Infants with hypercalcaemia present with
irritability, lethargy, hypotonia, vomiting, polyuria, poly­
dipsia, dehydration, constipation and failure to thrive.
Persistent moderate‐to‐severe hypercalcaemia may result
in metastatic calcification of skin, myocardium, falx cer­
ebri, liver, gastric mucosa and kidney, leading to nephro­
calcinosis, nephrolithiasis and, rarely, renal failure [76,78].
Treatment of hypercalcaemia includes conservative
management with formulas that are low in calcium and
vitamin D, increased hydration, calcium‐wasting diuretics Fig. 6.14  Adnexal polyp: small pedunculated flesh‐coloured lesion.
and corticosteroids. In cases of refractory hypercalcaemia,
treatment with pamidronate has been reported as being
effective. Differential diagnostic considerations should include
Treatment of subcutaneous fat necrosis in most cases is skin tags, accessory auricles and supernumerary nipples.
unnecessary because it is a self‐limited condition. Severe Skin tags are multiple acquired tumours common in
fluctuant abscess‐like nodules or plaques should be adults but rare in infants, often located on axilla, groin
aspirated to prevent rupture and infection, and to mini­ and neck. Histologically, adnexal structures are absent in
mize pain and skin sequelae. It is important to prevent skin tags [88,90].
and manage complications [76,82]. Cooled neonates
should be turned regularly to prevent prolonged contact Pedal papules of infancy
with the cooling interface [82]. Pedal papules, also known as precalcaneal congenital
fibrolipomatous hamartomas or benign anteromedial
plantar nodules, were first described by Larralde et al. in
Sucking blisters, erosions and calluses 1990 [91]. They are present at birth and consist of bilateral,
Sucking blisters and erosions are the result of repetitive symmetrical, asymptomatic, subcutaneous flesh‐coloured
vigorous fetal sucking. They occur in 0.4–2% of healthy nodules located on the plantar region of the heel. As only
newborns at birth [86,87]. They present as an intact flaccid relatively few case series have been reported in the lit­
bulla or erosion on non‐inflamed skin of the dorsal aspect erature, the natural history of this entity is unclear.
of forearms, wrists and fingers (thumb and index) and However, one group reported an incidence rate of 5.9% in
rarely on the feet. Although a solitary lesion is the most newborns and 39.4% among infants, with the majority of
common form of presentation, multiple and bilateral lesions lesions appearing by 2–3 months of age and largely
have also been reported. Calluses are the result of chronic showing spontaneous regression by 3 years of age [92].
and less intense sucking [13,86,87]. They spontaneously The pathogenesis is not yet understood. Histopathological
regress without sequelae within a few days to 2 weeks study shows mature adipose tissue enveloped in pre­
after birth. The differential diagnosis with serious neonatal dominantly collagenous fibrous sheaths. Differential
blistering diseases such as bullous impetigo, herpes diagnostic considerations include other lipomas, naevus
simplex, congenital syphilis, mastocytosis or epidermolysis lipomatosus and childhood fibrous hamartoma. They are
bullosa should be effected [13,86,87]. Observation of focal an entity distinct from piezogenic pedal papules, whose
sucking of the area involved as well as failure to develop onset is more frequently in adulthood [91–94].
new lesions postnatally aids diagnosis.
Perineal groove
Adnexal polyp This rare and benign congenital anomaly of the perineum
Adnexal polyp of neonatal skin is a solitary, small, con­ occurs mostly in females, with only one male reported.
genital polypoid tumour occurring most frequently on It is characterized by a wet groove that extends from the
the areola of the nipple (Fig. 6.14). It may also develop in posterior fourchette to the anus with a normal vestibule.
other sites such as eyelid, cheek, scapula, arm, axilla, labia On rare occasions an anogenital or urogenital anomaly
majora and scrotum. Although it usually falls off sponta­ may be associated. It is an asymptomatic lesion, with few
neously within a few days, persistent lesions in a 53‐day‐ reported complications such as constipation or infection.
old and a 2‐year‐old infant have been reported. Its Given that complete epithelialization is achieved by 1–2
incidence is estimated between 0.7 and 4%. years of age, treatment is not generally necessary. Although
Histopathology demonstrates the presence of hair fol­ its pathogenesis remains unclear, some embryological
licles, eccrine glands and vestigial sebaceous glands in the mechanisms have been proposed such as failure to fuse of
centre of the tumour [11,88–90]. the midline perineal raphe [95,96].
82 Section 2  Skin Disorders of the Neonate and Young Infant

References 29 Juern AM, Glick ZR, Drolet BA, Frieden IJ. Nevus simplex: A recon-
1 Hulsmann AR, Oranje AP. Educational paper: Neonatal skin sideration of nomenclature, sites of involvement, and disease
lesions. Eur J Pediatr 2014;173:557–66. associations. J Am Acad Dermatol 2010;63:805–14.
2 Conlon JD, Drolet BA. Skin lesions in the neonate. Pediatr Clin North 30 Januário G, Salgado M. The Harlequin phenomenon. J Eur Acad
Am 2004;51:863–88. Dermatology Venereol 2011;25:1381–4.
3 De Araujo T, Schachner L. Benign vesicopustular eruptions in the 31 Tang J, Bergman J, Lam JM. Harlequin colour change: Unilateral
neonate. An Bras Dermatol 2006;81:359–66. erythema in a newborn. CMAJ 2010;182:E801.
4 Eichenfield L, Lee P. Transient physiologic changes and cutaneous 32 Rao J, Campbell ME, Krol A. The harlequin color change and associa­
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

lesions in the newborn. In: Schachner L, Hansen R (eds) Pediatric tion with prostaglandin E1. Pediatr Dermatol 2004;21:573–6.
Dermatology, 4th edn. Philadelphia: Mosby Elsevier, 2011:301–8. 33 Sharathkumar AA, Castillo‐Caro P. Primary Raynaud’s phenomenon
5 Monteagudo B, Labandeira J, León‐Muiños E et al. Influence of neo­ in an infant: a case report and review of literature. Pediatr Rheumatol
natal and maternal factors on the prevalence of vernix caseosa. Actas Online J 2011;9:16.
Dermosifiliogr 2011;102:726–9. 34 Morgan AJ, Steen CJ, Schwartz RA, Janniger CK. Erythema toxicum
6 Serdaroglu S, Çak B. Physiologic skin findings of newborn. J Turk neonatorum revisited. Cutis 2009;83:13–16.
Acad Dermatol 2008;2:83401r. 35 Monteagudo B, Labandeira J, Cabanillas M et al. Prospective study of
7 Ghosh S. Neonatal pustular dermatoses: An overview. Indian J erythema toxicum neonatorum: Epidemiology and predisposing
Dermatol 2015;60:211. factors. Pediatr Dermatol 2012;29:166–8.
8 Afsar FS. Physiological skin conditions of preterm and term neo- 36 Larralde M, Luna PC. Vesicopustular, bullous and erosive diseases
nates. Clin Exp Dermatol 2010;35:346–50. of the newborn. In: Schachner L, Hansen R (eds) Pediatric
9 Jha AK. Is There a preventive role for vernix caseosa?: An in vitro Dermatology, 4th edn. Philadelphia: Mosby Elsevier, 2011:328–42.
study. J Clin Diagnostic Res 2015;9:13–16. 37 Marchini G, Nelson A, Edner J et al. Erythema toxicum neonatorum
10 Larralde M, Luna PC. Neonatal transient benign cutaneous lesions: is an innate immune response to commensal microbes penetrated
An update. In: Sarkar R, Inamadar A, Palit A (eds) Advances in into the skin of the newborn infant. Pediatr Res 2005;58:613–16.
Pediatric Dermatology‐2. New Delhi: Jaypee Brothers Medical 38 Connor NR, McLaughlin MR, Ham P. Newborn skin: Part I. Common
Pubishers, 2014:14–23. rashes. Am Fam Physician 2008;77:47–52.
11 Boralevi F, Taieb A. Common transient neonatal dermatoses. In: 39 Agusti‐Mejias A, Messeguer F, Febrer I, Alegre V. Transient neonatal
Irvine A, Hoeger P, Yan A (eds) Harper`s Textbook of Pediatric pustular melanosis. Actas Dermosifiliogr 2013;104:84–5.
Dermatology, 3rd edn. Oxford: Blackwell Publishing, 2011. 40 Chia PS, Leung C, Hsu YL, Lo CY. An infant with transient neonatal
12 Giachetti A. Cambios transitorios de la piel de recien nacido. In: pustular melanosis presenting as pustules. Pediatr Neonatol
Larralde M, Abad E, Luna P (eds) Dermatologia Pediatrica. Buenos 2010;51:356–8.
Aires: Ediciones journal, 2010:13–17. 41 Fujisawa Y, Miyazono Y, Kawachi Y, Otsuka F. A case of sterile tran­
13 Larralde M, Luna PC. Benign self‐limiting cutaneous lesions. In: sient neonatal pustulosis presenting with large flaccid pustules.
Schachner L, Hansen R (eds) Pediatric Dermatology, 4th edn. Pediatr Dermatol 2013;30:e238–9.
Philadelphia: Mosby Elsevier, 2011:311–13. 42 Ferrandiz C, Coroleu W, Ribera M et al. Sterile transient neonatal
14 Haveri FTTS, Inamadar AC. A Cross‐Sectional Prospective Study of pustulosis is a precocious form of erythema toxicum neonatorum.
Cutaneous Lesions in Newborn. ISRN Dermatol 2014. http://www. Dermatology 1992;185:18–22.
hindawi.com/journals/isrn.dermatology/2014/360590/ (accessed 6 43 Cambiaghi S, Gelmetti C. Bohn’s nodules. Int J Dermatol 2005;44:
October 2018). 753–4.
15 Kanada KN, Merin MR, Munden A, Friedlander S. A prospective 44 Singh RK, Kumar R, Pandey RK, Singh K. Dental lamina cysts in a
study of cutaneous findings in newborns in the United States: correla­ newborn infant. BMJ Case Rep 2012. http://www.ncbi.nlm.nih.gov/
tion with race, ethnicity, and gestational status using updated classifi­ pubmed/23048002 (accessed 6 October 2018).
cation and nomenclature. J Pediatr 2012;161:240–5. 45 Larralde M, Luna PC. Benign oral neonatal lesions. In: Schachner L,
16 Monteagudo B, Labandeira J, Cabanillas M et al. Prevalence of milia Hansen R (eds) Pediatric Dermatology, 4th edn. Philadelphia:
and palatal and gingival cysts in Spanish newborns. Pediatr Dermatol Mosby Elsevier, 2011:308–11.
2012;29:301–5. 46 Dutta A, Gosh S, Nag S. Bohn’s nodules. Indian Pediatr 2014;51:
17 Berk DR, Bayliss SJ. Milia: A review and classification. J Am Acad 849–50.
Dermatol 2008;59:1050–63. 47 Larralde M. Lesiones orales del recien nacido. In: Larralde M, Abad
18 Luna PC, Larralde M. Profuse congenital familial milia with absent E, Luna P (eds) Dermatologia Pediatrica. Buenos Aires: Ediciones
dermatoglyphics (Basan’s syndrome): Description of a new family. journal, 2010:17–20.
Pediatr Dermatol 2012;29:527–9. 48 Kumar A, Grewal H, Verma M. Dental lamina cyst of newborn: A case
19 Rutter KJ, Judge MR. Profuse congenital milia in a family. Pediatr report. J Indian Soc Pedod Prev Dent 2008;26:175–6.
Dermatol 2009;26:62–4. 49 Mhaske S, Yuwanati MB, Mhaske A et al. Natal and Neonatal Teeth:
20 Adya KA, Inamadar AC, Palit A, Shirasangi A. “Miniature puberty” An Overview of the Literature. ISRN Pediatr 2013 http://www.ncbi.
in an eight‐days‐old female neonate. Indian J Pediatr 2014;81:1266–7. nlm.nih.gov/pubmed/24024038 (accessed 6 October 2018).
21 Monteagudo B, Labandeira J, León‐Muiños E et  al. Estudio pro- 50 Basavanthappa NN, Kagathur U, Basavanthappa RN, Suryaprakash
spectivo de la hiperpigmentación del área genital en recién nacidos ST. Natal and neonatal teeth: A retrospective study of 15 cases. Eur J
españoles: Frecuencia y factores predisponentes. Med Cutan Ibero Dent 2011;5:168–72.
Lat Am 2013;41:3–6. 51 Malki GA, Al‐Badawi EA, Dahlan MA. Natal teeth: A case report and
22 Ng M, How C. When babies turn yellow. Singapore Med J 2015;56: reappraisal. Case Rep Dent 2015. http://www.ncbi.nlm.nih.gov/
599–603. pubmed/25722895 (accessed 6 October 2018).
23 Cohen RS, Wong RJ, Stevenson DK. Understanding neonatal jaundice: 52 Volpato LER, Simões CAD, Simões F et al. Riga‐Fede disease associ-
A perspective on causation. Pediatr Neonatol 2010;51:143–8. ated with natal teeth: two different approaches in the same case.
24 Levy R, Lam JM. Cutis marmorata telangiectatica congenita: a mim­ Case Rep Dent 2015. http://www.ncbi.nlm.nih.gov/pubmed/26421196
icker of a common disorder. CMAJ 2011;183:249–51. (accessed 6 October 2018).
25 Mazereeuw‐Hautier J, Carel‐Caneppele S, Bonafé JL et al. Cutis mar­ 53 Shah S, Boen M, Kenner‐Bell B et al. Pachyonychia congenita in pedi­
morata telangiectatica congenita: Report of two persistent cases. atric patients. JAMA Dermatol 2014;150:146.
Pediatr Dermatol 2013;88:249–51. 54 Venkatesh C, Adhisivam B. Natal teeth in an infant with congenital
26 Happle R. Capillary malformations: A classification using specific hypothyroidism. Indian J Dent Res 2011;22:498.
names for specific skin disorders. J Eur Acad Dermatology Venereol 55 Yilmaz RBN, Cakan D, Mesgarzadeh N. Prevalence and management
2015;29:2295–305. of natal/neonatal teeth in cleft lip and palate patients. Eur J Dent
27 Trevisan F, Cunha PR, Pinto CAL, Cattete FG. Cutaneous neonatal 2016;10:54.
lupus with cutis marmorata telangiectatica congenita‐like lesions. An 56 Khandelwal V, Nayak UA, Nayak PA, Bafna Y. Management of an
Bras Dermatol 2013;88:428–31. infant having natal teeth. BMJ Case Rep 2013. http://www.ncbi.nlm.
28 Cordoro KM, Speetzen LS, Koerper MA, Frieden IJ. Physiologic nih.gov/pubmed/23737593 (accessed 6 October 2018).
changes in vascular birthmarks during early infancy: Mechanisms 57 Alemán Navas R, Martínez Mendoza M. Congenital eruption cyst.
and clinical implications. J Am Acad Dermatol 2009;60:669–75. Pediatr Dermatol 2010;27:671–2.
Chapter 6  Transient Skin Disorders in the Neonate and Young Infant 83

58 Navas RMA, Mendoza MGM, Leonardo MR et al. Congenital erup­ 79 Akin M, Akin L, Sarici D et  al. Follow‐up during early infancy of
tion cyst: A case report. Braz Dent J 2010;21:259–62. newborns diagnosed with subcutaneous fat necrosis. J Clin Res
59 Thariat J, Roth V, Marcy P‐Y. Sucking pads in a full‐term newborn. Pediatr Endocrinol 2011;3:216–18.
J Pediatr 2011;158:166. 80 Strohm B, Hobson A, Brocklehurst P et al. Subcutaneous fat necrosis
60 Heyl T, Raubenheimer J. Sucking pads (sucking calluses) of the lips in after moderate therapeutic hypothermia in neonates. Pediatrics
neonates: a manifestation of transient leukoedema. Pediatr Dermatol 2011;128:e450–2.
1987;4:123–9. 81 Grass B, Weibel L, Hagmann C, Brotschi B. Subcutaneous fat necrosis
61 Gupta D, Thappa D. Mongolian spots: How important are they? in neonates with hypoxic ischaemic encephalopathy registered in

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
World J Clin Cases 2013;1:230–2. the Swiss National Asphyxia and Cooling Register. BMC Pediatr
62 Marcoux D, Durán‐McKinster C. Melanocytes and melanogenesis. 2015;15:73.
In: Schachner L, Hansen R (eds) Pediatric Dermatology, 4th edn. 82 Oza V, Treat J, Cook N et al. Subcutaneous fat necrosis as a com-
Philadelphia: Mosby Elsevier, 2011:700–19. plication of whole‐body cooling for birth asphyxia. Arch Dermatol
63 Gupta D, Thappa DM. Mongolian spots  –  A prospective study. 2010;146:882–5.
Pediatr Dermatol 2013;30:683–8. 83 Samedi VM, Yusuf K, Yee W et al. Neonatal hypercalcemia second­
64 Musumeci M, Lacarrubba F, Santagati C, Micali G. Multiple and ary to subcutaneous fat necrosis successfully treated with
superimposed Mongolian spots. BMJ Case Rep 2013. http://www. pamidronate: A case series and literature review. Am J Perinatol
ncbi.nlm.nih.gov/pubmed/24014338 (accessed 6 October 2018). Rep 2014;4:e93–6.
65 Ma H, Liao M, Qiu S et al. The case of a boy with nevus of Ota, exten­ 84 Shumer D, Thaker V, Taylor G, Wassner A. Severe hypercalcemia
sive Mongolian spot, nevus flammeus, nevus anemicus and cutis due to subcutaneous fat necrosis: presentation, management
marmorata. An Bras Dermatol 2015;90:S10–12. and  complications. Arch Dis Child Fetal Neonatal Ed 2014;99:
66 Hanson M, Lupski JR, Hicks J, Metry D. Association of dermal 419–21.
melanocytosis with lysosomal storage disease: clinical features and 85 Farooque A, Moss C, Zehnder D et al. Expression of 25‐hydroxyvita­
hypotheses regarding pathogenesis. Arch Dermatol 2003;139:916–20. min D3‐1α‐hydroxylase in subcutaneous fat necrosis. Br J Dermatol
67 Hackbart BA, Arita JH, Pinho RS et al. Mongolian spots are not always 2009;160:423–5.
a benign sign. J Pediatr 2013;162:1070. 86 Monteagudo B, Labandeira J, León‐Muiños E et al. Neonatal suck-
68 Mimouni‐Bloch A, Finezilber Y, Rothschild M, Raas‐Rothschild A. ing blisters: prevalence and differential diagnosis. An Pediatr
Extensive Mongolian Spots and Lysosomal Storage Diseases. J Pediatr (Barc) 2011;74:62–4.
2016;170:333–333.e1 87 Monteagudo B, León‐Muiños E. Neonatal sucking blisters. Indian
69 Happle R. Phacomatosis pigmentovascularis revisited and reclassi- Pediatr 2010;47:794.
fied. Arch Dermatol 2005;141:385–8. 88 Hidano A, Kobayashi T. Adnexal polyp of neonatal skin. Br J Dermatol
70 Fosse N, Itin P. Pigmentary lines of the newborn: a case report and 1975;92:659–62.
review of the literature. Dermatology 2014;228:198–201. 89 Koizumi H, Itoh E, Ohkawara A. Adnexal polyp of neonatal skin
71 Martin JM, Jorda E, Alonso V. Transient pigmentary lines of the observed beyond the neonatal period. Acta Derm Venereol
newborn. Pediatr Dermatol 2009;26:768. 1998;78:391–2.
72 Garg G, Bhalla M, Thami G. Transient infantile patterned hyper- 90 Lucas Costa A, Betlloch I, Pérez‐Crespo M et al. Mammary skin tag
pigmentation. Pediatr Dermatol 2012;29:372–3. in a 2‐year‐old girl: a long‐lasting adnexal polyp of neonatal skin?
73 Iorizzo M, Oranje AP, Tosti A. Periungual hyperpigmentation in Pediatr Dermatol 2009;26:618–20.
newborns. Pediatr Dermatol 2008;25:25–7. 91 Larralde de Luna M, Ruiz León J, Cabrera H. Pápulas podálicas en
74 Perez‐Bescos L, Martinez‐Elgarresta M, de la Fuente‐de la Prieta M, el recién nacido. Med Cutan Ibero Lat Am 1990;18:9–12.
Iturralde‐Iriso J. Hiperpigmentación de las falanges distales del 92 Greenberg S, Krafchik BR. Infantile pedal papules. J Am Acad
lactante. An Pediatr 2006;65:384–92. Dermatol 2005;53:333–4.
75 Larralde M, Abad ME, Corbella C et al. Subcutaneous fat necrosis of 93 Rubio‐Flores C, López‐Barrantes González O, Garrido‐Gutiérrez C,
the newborn, report of five cases. Dermatol Argent 2009;15:200–4. Díaz‐Díaz R. Precalcaneal congenital fibrolipomatous hamartoma.
76 Tran JT, Sheth AP. Complications of subcutaneous fat necrosis of Actas Dermosifiliogr 2012;103:649–51.
the newborn: A case report and review of the literature. Pediatr 94 Ortega‐Monzó C, Molina‐Gallardo I, Monteagudo‐Castro C et  al.
Dermatol 2003;20:257–61. Precalcaneal congenital fibrolipomatous hamartoma: a report of four
77 Gomes M, Porro A, Enokihara M, Floriano M. Subcutaneous fat cases. Pediatr Dermatol 2000;17:429–31.
necrosis of the newborn: clinical manifestations in two cases. An Bras 95 Diaz L, Levy M, Kalajian A, Metry D. Perineal groove: a report of 2
Dermatol 2013;88:154–7. cases. JAMA Dermatol 2014;150: 101–2.
78 Rubin G, Spagnut G, Morandi F et al. Subcutaneous fat necrosis of the 96 Harsono M, Pourcyrous M. Perineal groove: a rare congenital midline
newborn. Clin Case Rep 2015;3:1017–20. defect of perineum. Am J Perinatol Rep 2016;6:e30–2.
84 

CHA PTER 7

Congenital and Acquired Infections


in the Neonate
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Scott H. James1, Nico G. Hartwig2, David W. Kimberlin1 & Peter H. Hoeger3


1
 Department of Pediatrics, Division of Infectious Diseases, University of Alabama, Birmingham, AL, USA
2
 Department of Paediatrics, Franciscus Hospital, Rotterdam, The Netherlands
3
 Department of Paediatrics and Paediatric Dermatology, Catholic Children’s Hospital Wilhelmstift, Hamburg, Germany

Introduction, 84 Congenital infections, 84 Acquired neonatal infections, 86

Abstract ­ anifestations. Viral, bacterial, fungal and parasitic aetiologies are


m
reviewed for common congenital, perinatal and postnatal infec-
This chapter provides a brief overview of congenital and ­neonatal tions in neonates. Diagnostic evaluation and clinical management
infections, with particular consideration given to cutaneous are also discussed.

Key points • Due to the potential for significant clinical sequelae, timely
diagnostic evaluation is crucial for the appropriate identification
and management of congenital, perinatal and postnatal
• Cutaneous findings in the neonate may be a sign of an underlying
infections presenting during the neonatal period.
infectious process, but they are often nonspecific.
• Clinicians should maintain a broad differential diagnosis,
including viral, bacterial, fungal, parasitic and noninfectious
aetiologies, when evaluating neonates.

Introduction growth restriction, decrease in muscle tone, hepatosple-


nomegaly, lymph node enlargement, pale skin colour or
Consideration of infectious diseases presenting at or neurological findings. The skin may be involved, but
soon after birth is an important aspect of neonatal care. diagnosis of congenital infections should not be based on
Cutaneous findings can be a sentinel for an underlying cutaneous findings alone.
infectious process, but clinicians should also be aware During the neonatal period, defined as the age from birth
that many unrelated skin disorders can mimic infectious to 4 weeks (the first month of life), postnatally acquired
diseases. Pustules in the neonate, for instance, can repre- infections may present with systemic symptoms and can
sent self‐limiting noninfectious processes such as erythema lead to serious sequelae, depending upon the pathogen.
toxicum, infantile acropustulosis, transient neonatal The skin is frequently involved and may show petechiae,
pustular melanosis, acne and miliaria, or they may be purpura, vesicles, pustules or a maculopapular rash.
indicative of potentially life‐threatening diseases such as This chapter provides an overview of congenital,
bacterial sepsis (Listeria, group B Streptococcus) or herpes perinatal and postnatal infections, with particular
simplex virus (HSV) and varicella zoster virus (VZV) consideration given to cutaneous manifestations.
­
infections [1,2]. Although some cutaneous findings more Diagnostic evaluation and clinical management are
specifically point to an infectious aetiology (e.g. oral vesi- also discussed. Many of the topics addressed here are
cles in HSV infections, scarring after intrauterine VZV also discussed more extensively in other chapters.
infection, or ‘blueberry muffin’ lesions), the clinician must
maintain a broad differential diagnosis when evaluating
neonates.
Congenital infections are transmitted from mother to
Congenital infections
fetus in utero and include a wide range of aetiologies Congenital infections may be due to viral, bacterial, fun-
and clinical presentations. While some congenital infec- gal or protozoal microorganisms. Many of these infec-
tions are asymptomatic, many infants will be born with tions are associated with cutaneous findings, which may
clinically apparent signs of systemic involvement such as aid in diagnosis.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 7  Congenital and Acquired Infections in the Neonate 85

Box 7.1  Causes of blueberry muffin rash

Dermal erythropoiesis

• Congenital infections
Toxoplasmosis

SECTION 2: SKIN DISORDERS


Rubella

OF NEONATES AND INFANTS


Cytomegalovirus
Herpes simplex virus
Coxsackie B2 virus
• Haemolytic disease of the newborn
Rhesus and ABO incompatibility
Hereditary spherocytosis
Twin–twin transfusion syndrome

Neoplastic diseases

• Transitory myeloproliferative disease


• Neuroblastoma
• Langerhans cell histiocytosis
• Congenital leukaemia

One notable cutaneous finding relevant to several con-


genital infections is a blueberry muffin rash. Blueberry
Fig. 7.1  Congenital varicella zoster virus infection with widespread
muffin rash was first described in congenital rubella with
papulovesicles.
thrombocytopenia [3]. It is characterized by bluish‐red
macules representing sites of dermal erythropoiesis,
which occur after intrauterine infections with rubella
virus, cytomegalovirus (CMV), Toxoplasma, or other seri- sequelae are common in infants with severe involvement,
ous systemic illnesses including congenital leukaemia but 6 months of oral valganciclovir has been shown to
[4–7]. A cutaneous biopsy is helpful for rapid diagnosis of improve hearing and neurodevelopmental outcomes in
the underlying disease (Box 7.1). symptomatic congenital CMV infection [11].
Molluscum contagiosum can be transmitted vertically
Viral infections and presents with characteristic umbilicated papules
Except for HSV, VZV and rubella, few congenital viral either at birth or within 6 weeks of age [6]. These and
infections are associated with specific cutaneous signs; in other congenital viral infections associated with cutaneous
fact, many newborns with congenital viral infections are manifestations are listed in Table 7.1.
asymptomatic.
Grouped vesicles on an erythematous base are typical Bacterial infections
of HSV. Congenital HSV infections likewise can present Bacterial infections present at birth can directly cause
with widespread scarring or erosions without any vesi- skin lesions, most commonly pustules (e.g. Staphylococcus
cles or vesiculopustules [8]. Congenital VZV can present aureus, β‐haemolytic streptococci), but also macules, pap-
with disseminated vesicles, vesiculopustules and papules ules and others. Widespread macules and scaly papules
(Fig. 7.1) or, if transmission occurs early during the course are often seen in congenital syphilis. Most of these skin
of gestation, with limb hypoplasia and significant cutane- changes are directly attributable to the presence of bacte-
ous scarring. Disseminated vesicles have been reported ria in the skin and are therefore contagious themselves.
with coxsackievirus infections, which may ultimately This is particularly true for congenital syphilis, which
ulcerate or become nodular [9,10]. classically presents with a papulosquamous rash and
CMV is the most common congenital infection, occur- coryza. Erythema multiforme‐like target lesions have
ring in approximately 0.5–2% of live births. Skin abnor- also been described as a presenting sign; Treponema
malities include petechiae, purpura, maculopapules pallidum DNA can be detected within these lesions [21].
and papulonodules. Vesicles and pustules are unusual Erythema multiforme‐like lesions have also been reported
but have been described. The clinical presentation of as a presenting sign of congenital tuberculosis [22], which
‘blueberry muffin rash’ (dermal extramedullary erythro- is very rare and usually characterized by disseminated
poiesis) can be observed. Diagnosis of congenital CMV papulopustules and cutaneous abscesses not responding
infection often is based on viral urine culture or the detec- to routine antibiotics.
tion of CMV DNA via polymerase chain reaction (PCR) in Bacterial toxins, mainly produced by Staph. aureus
saliva, peripheral blood, cerebrospinal fluid (CSF) or (exfoliative toxin, toxic shock syndrome toxins [23]) or
urine within the first 3 weeks after birth. Long‐term pyogenic streptococci (streptococcal toxic shock syndrome
86 Section 2  Skin Disorders of the Neonate and Young Infant

Table 7.1  Cutaneous signs of congenital viral infection

Infectious agent Time of Cutaneous signs Diagnosis Comments References


maternofetal
transmission

Coxsackievirus A4, Any time Herpangina (A5, B2, B3); mostly Culture, ELISA–IgM Types B5 and B1 [4,5]
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

B1–6 transient maculopapular rash, most commonly


occasionally petechial; vesicular associated with
lesion on erythematous base, cutaneous
ulcerate or become nodular symptoms
Cytomegalovirus Any time Petechiae (76%), jaundice (67%), PCR or culture (urine, saliva), – [12]
(CMV) purpura (13%); blueberry Tzanck smear, CMV IgM/IgE
muffin lesions
Echovirus (types 4, 5, Any time Maculopapular rash, petechiae Culture, ELISA–IgM Rash often very mild [12,13]
7, 9, 11, 16–19, after perinatal
21–22, 33) infection
Herpes simplex virus Any time Vesicles (single, grouped or PCR or culture (vesicles, CSF, – [14]
(HSV) types I and II disseminated), scars. conjunctivae, skin), Tzanck
Widespread erosions smear, direct
immunofluorescence
Human Any time Generalized macular rash (‘acute PCR, serology, viral load – [12]
immunodeficiency retroviral syndrome’)
virus (HIV)
Molluscum Birth to Typical skin‐coloured papules Clinical Vertical transmission [15]
contagiosum 6 weeks with central umbilication likely in early‐onset
lesions
Papillomavirus At birth Condylomata acuminata, Cervical smear (mother), PCR – [12]
laryngeal papilloma (laryngeal swab)
Parvovirus B19 Intrauterine, Transient or recurrent macular PCR, parvovirus IgM – [12]
at birth erythematous rash
Rubella virus Any time Dermal erythropoiesis (blueberry Maternal: four‐fold rise in – [16–18]
muffin syndrome); generalized antibody (HI, CF, LA); fetal:
brown macules and papules; positive rubella IgM (ELISA)
persistent facial rash
Varicella zoster virus Intrauterine, Generalized varicelliform blisters PCR – [19,20]
at birth (Fig. 7.1) or scars; grouped
zosteriform vesicles

CF, complement fixation text; CSF, cerebrospinal fluid; ELISA, enzyme‐linked immunosorbent assay; HI, haemagglutination inhibition; HSV, herpes simplex
virus; IgE and IgM, immunoglobulin E and M; LA, latex agglutination; PCR, polymerase chain reaction.

toxin [24]), can equally cause exanthematous diseases. and upper torso (Fig.  7.2). These pustules have in the
Staphylococcal scalded skin syndrome, which usually past mistakenly been referred to as ‘neonatal acne’ [30].
presents during the neonatal period or later, has been While infection with Malassezia spp. may or may not
reported to start in utero during late gestation. Toxic shock be attributable to immaturity of the neonatal cutane-
syndrome can also induce rashes in the early neonatal ous defence system, widespread cutaneous infections
period [25]. Bacterial pathogens associated with congeni- with Candida spp. or Aspergillus spp. are almost exclu-
tal and neonatal rashes are listed in Table 7.2. As seen in sively observed in premature infants or term neonates
this table, the timing of acquisition of bacterial infections with an underlying immunodeficiency disorder
(intrauterine vs. peri‐ or postnatal) varies widely and, in (Fig. 7.3) [31,32].
certain cases, many of the microorganisms covered here Toxoplasma gondii is a protozoan that can lead to congen-
could rightly be classified as neonatal rather than congen- ital infection in the event of primary maternal infection
ital infections. during the course of gestation. Congenital toxoplasmosis
can be associated with petechiae, maculopapular or pur-
Fungal and protozoal infections puric lesions, or a blueberry muffin rash [33]. Additional
It is often very difficult to decide whether an infection considerations of fungal and protozoal congenital infec-
observed in a young neonate results from intrauterine tions can be found in Table 7.3.
or early postnatal transmission. Given their ubiquitous
distribution, this is particularly difficult with fungal
Acquired neonatal infections
infections. Malassezia spp. can be isolated from the
scalp region of 30–40% of adults and are readily trans- As opposed to congenital infections, which are acquired
mitted to the newborn infant either at or shortly after in utero, the infections considered in this section are
birth, frequently leading to pustules in the facial area transmitted to the newborn infant in the perinatal and
Table 7.2  Cutaneous signs of congenital bacterial infection

Infectious agent Time of Cutaneous signs Diagnosis Comments References


maternofetal
transmission

Gram‐negative septicaemia At birth or Petechiae; erythematous Culture (blood, CSF, – [1]


(Escherichia coli, Klebsiella after PRM, macules; occasionally small urine)
spp., Pseudomonas perinatal nodules or vesicles;
aeruginosa, Haemophilus Pseudomonas aeruginosa:

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
influenzae) ecthyma gangrenosum (rare)
Borrelia burgdorferi Intrauterine May have vasculitis or ‘skin rash’ PCR, specific IgM Congenital infection rare [12]
Listeria monocytogenes Perinatal Pustular petechial lesions Stool microscopy, culture [26]
Mycobacterium Intrauterine, Erythematous papulopustules Microscopy and culture Fundoscopy recommended [22]
tuberculosis diaplacental, with central crusting, multiple from multiple sites (chorioretinitis frequent);
at birth abscesses, multiform lesions (including gastric lavage), maternal infection often
placenta histology subclinical; skin lesions rare
Mycoplasma hominis, Perinatal Subcutaneous abscess Cervical/vaginal swabs Preterm babies at risk [12]
M. pneumoniae formation
Neisseria gonorrhoeae At birth Ophthalmia neonatorum, Microscopy, culture, PCR – [27]
occasionally pustules
Staphylococcus aureus At birth Impetigo bullosa, toxic shock Culture, assays for toxin – [23,25]
syndrome, staphylococcal production
scalded skin syndrome
Streptococcus pyogenes At birth Impetigo, streptococcal toxic As above – [24]
shock syndrome
Group B streptococci Perinatal; rarely Blisters, impetigo, erosions, Culture Skin lesions rare [28,29]
intrauterine superficial blisters
Treponema pallidum Intrauterine, Papulosquamous rash; Microscopy, TPHA, FTA – [21]
diaplacental, desquamation over palms and
at birth soles; EM‐like targetoid lesions

CSF, cerebrospinal fluid; EM, erythema multiforme; FTA, fluorescent treponemal antibody; IgM, immunoglobulin M; PCR, polymerase chain reaction; PRM,
premature rupture of membranes; TPHA, Treponema pallidum haemagglutination test.

Fig. 7.2  Neonatal cephalic pustulosis due to Malassezia spp.


Fig. 7.3  Primary cutaneous aspergillosis.

Table 7.3  Cutaneous signs of congenital fungal and protozoal infection

Infectious agent Time of maternofetal Cutaneous signs Diagnosis References


transmission

Candida albicans Perinatal Congenital cutaneous candidiasis: widespread erythema/ Skin swabs, KOH [32]
erythroderma or localized thrush preparation, culture
Malassezia spp. At birth Folliculitis As above [30]
(Pityrosporum spp.)
Aspergillus spp. At birth, post partum Pustules, necrotic ulceration As above [32,34]
Trichophyton rubrum At birth, post partum Scaling, erosion As above [35]
Sarcoptes scabiei At birth, post partum Generalized papulopustules (including face, palms, soles) KOH, microscopy [36]
Toxoplasma gondii Intrauterine erythropoiesis Petechiae, purpura, jaundice Toxoplasma IgM ELISA [33]

ELISA, enzyme‐linked immunosorbent assay; IgM, immunoglobulin M; KOH, potassium hydroxide.


88 Section 2  Skin Disorders of the Neonate and Young Infant

Varicella is caused by VZV, a member of the herpes-


Box 7.2  Common acquired infections in the neonate
virus family. Its normal route of transmission is airborne.
Viral infections
The incubation time for varicella is approximately
2 weeks (range 10–23 days). Perinatal varicella is poten-
• Herpes simplex infection tially dangerous in the neonate, especially when primary
• Cytomegalovirus infection maternal infection develops any time from 5 days before
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

• Varicella to 2 days after delivery. In this setting, mother‐to‐child


• Enteroviral infections transmission can occur haematogenously or via the air-
Bacterial infections
borne route in a newborn with no protective maternal
antibodies. Haematogenous transmission may shorten
• Pustular staphylococcal infections the incubation period and some children may present
• Neonatal impetigo with vesicles at birth [40].
• Omphalitis Mild prodromal symptoms such as fever, malaise and
• Cellulitis
upper respiratory symptoms may precede the onset of
• Neonatal abscesses
rash by 1–2 days but are generally absent in neonates. The
• Circumcision infections
skin lesions erupt over 1 week and evolve within 12–24 h
Yeast infections of eruption from small red macules to papules, vesicles
and pustules on an erythematous base. The vesicular
• Neonatal candidiasis
lesions are typically umbilicated and differ in their stage
• Malassezia furfur pustulosis
of development. Crusting occurs 1–3 days following the
• Neonatal pityriasis versicolor
appearance of an individual lesion.
Parasitic infections Diagnosis is usually by observation of typical signs.
Tzanck smears from the base of an intact vesicle are no
• Scabies
longer commonly performed. Cytology and histopathol-
ogy are identical to those seen in HSV infections.
Definitive evidence of VZV usually is obtained via a
positive PCR from a swab of the base of one or more
postnatal periods. Common neonatal infections are listed unroofed fresh lesions. Newborns of mothers who
in Box 7.2. developed clinical varicella from 5 days before to 2 days
after delivery should be treated with varicella zoster
Viral infections immunoglobulin (VariZIG) intramuscularly as soon as
Viral exanthemas are discussed in detail elsewhere. Brief possible, preferably within 72 h. If VariZIG is not avail-
mention is made here of neonatally acquired HSV, VZV able, intravenous immune globulin may be used [41].
and enteroviral infections. In  addition, administering parenteral aciclovir may
Neonatal HSV infection is a potentially serious disease reduce symptoms. Mortality in untreated perinatally
that manifests in the first 4–6 weeks of life. Based on the acquired varicella can approach 30%.
incubation period, the onset rarely occurs before the age Enteroviruses include RNA viruses such as groups A
of 1 week. Transmission occurs perinatally during pas- and B coxsackieviruses, echoviruses, and enteroviruses
sage through the birth canal in 85% of infected infants. (types 68–71), which may cause a variety of illnesses in
The risk of transmission is greater than 50% in women neonates. Enteroviral infections are frequently observed
with first‐episode primary genital infections at the time of in infants and children. Parechoviruses, a recently
delivery, whereas women with recurrent genital lesions described genus of viruses, were previously classified
carry a 2% risk increase [37]. In an effort to prevent among the enteroviruses as echovirus 22 and 23, which
mother‐to‐child transmission of HSV, women with active have similar features to other enteroviral infections.
recurrent genital herpes should be offered suppressive These viruses are spread via faecal–oral and oral–oral
antiviral therapy beginning at 36 weeks of gestation and routes. Faecal viral excretion and transmission may
caesarean delivery should be performed in women with continue for weeks or months. The incubation time is
active genital lesions at the time of delivery (whether pri- 3–6 days.
mary or recurrent) [38]. It should be noted, however, that Common skin manifestations of neonatal enteroviral
mother‐to‐child transmission can also occur in asympto- infections include maculopapular, petechial and vesic-
matic women with no reported history of genital herpes, ular rashes. These skin manifestations are frequently
as well as those with a history of genital herpes who are accompanied by a sepsis‐like syndrome and/or respir-
receiving antiviral suppressive therapy [39]. Medical per- atory problems. Gastrointestinal symptoms such as
sonnel and family members with herpetic infection on the stomatitis, vomiting, abdominal pain and diarrhoea
­
fingers or around the mouth also can transmit HSV, and may occur. In rare cases neonates develop pericarditis,
therefore should take appropriate measures to prevent myocarditis, (meningo‐)encephalitis, hepatitis and
transmission. In 45% of cases skin lesions are observed, disseminated intravascular coagulopathy. Diagnosis is
usually vesicles or petechiae. Disseminated disease has a aided by PCR of blood, CSF, saliva and stool. There cur-
high mortality. Treatment with parenteral aciclovir is rently are no licensed antiviral agents for neonatal
advised when neonatal HSV infection is suspected. enteroviral infections.
Chapter 7  Congenital and Acquired Infections in the Neonate 89

Bacterial infections Box 7.3  Neonatal pustular eruptions


Newborns are susceptible to a number of bacterial infec-
tions, many of which are associated with cutaneous Infectious
findings (Table 7.2). Particular infections are covered in
greater detail elsewhere, but the following represents an • Bacterial
overview of common neonatally acquired bacterial • Staphylococcus aureus (bullous impetigo)

SECTION 2: SKIN DISORDERS


• Listeria monocytogenes

OF NEONATES AND INFANTS


infections.
• Streptococcus (group B β‐haemolytic)
Pustular eruptions in the neonate are most commonly
• Pseudomonas aeruginosa
caused by Staphylococcus aureus. Skin lesions may appear
• Haemophilus influenzae
as early as the second or third day of life. Clinical pres-
entation ranges from vesicles to pustules on an erythe- Viral
matous base. The condition may evolve into honeycomb
• Herpesvirus infections (HSV, CMV, VZV)
crusts or into bullous lesions, so‐called bullous impetigo
(Fig. 7.4). The intertriginous and napkin areas are sites Fungal
of predilection. The same symptoms are caused rarely
by group B streptococci, which are of a more invasive • Candidiasis (congenital, neonatal)
nature [29]. • Malassezia furfur folliculitis

In addition to Staphylococcus, pustular eruptions in the Parasitic


neonate include a large range of infectious and nonin-
fectious causes (Box 7.3). Final diagnosis is obtained after • Scabies
culturing for Staph. aureus. Cytological smears and histol-
Noninfectious
ogy may also show polymorphic neutrophils and Gram‐
positive cocci in clusters. Local therapy such as mupirocin • Erythema toxicum neonatorum
is sufficiently effective for limited lesions in an other- • Infantile acropustulosis
wise well‐appearing neonate with no fever. If lesions • Transient neonatal pustular melanosis
are accompanied by lymphadenopathy or fever, empiric • Neonatal acne
systemic antibiotics and further evaluation for dissemi- • Pustular miliaria
• Eosinophilic pustular folliculitis of infancy
nated disease are warranted.
• Incontinentia pigmenti
Neonatal impetigo is another possible manifestation of
• Congenital self‐healing Langerhans cell histiocytosis
staphylococcal infection. Some strains of Staph. aureus
produce an exfoliative exotoxin, which is able to split the Source: Adapted from Praag et al. 1997 [1]. Reproduced with permission
superficial epidermis at the level of the granular layer. from John Wiley & Sons.
When the exotoxins enter the blood circulation, involve-
ment of the entire skin is possible and can lead to staphy-
lococcal scalded skin syndrome (SSSS) [42]. At present,
three exfoliative exotoxins have been described but only impetigo may occur on the second or third day of life,
two, exfoliatin A and B, are associated with neonatal starting with pustules that develop into flaccid bullae
impetigo and SSSS [43,44]. The higher potential for devel- filled with yellow fluid. The bullae burst easily, leaving a
oping disseminated and even systemic disease in neo- circular erosion with a peripheral collarette of scale. The
nates is brought about by less efficient metabolism and infection is limited to the epidermis but may extend very
excretion of the toxin [45]. easily and may result in SSSS.
Cytological smears and histology for neonatal impetigo Neonatal impetigo is treated in the same way as pustu-
are identical to those of pustular infection. Neonatal lar staphylococcal infection, but systemic treatment may
be required more often to prevent the development of
SSSS. However, the infection is mainly limited to the
epidermis and generally does not result in systemic dis-
ease. Localized infections are treated topically, whereas
widespread infection requires systemic treatment with a
penicillinase‐resistant penicillin, a cephalosporin or van-
comycin, depending on drug susceptibility. SSSS is always
treated systemically via intravenous therapy. It is some-
times lethal, especially in the neonatal period.
Omphalitis neonatorum has a low incidence in devel-
oped countries (about 0.7%) but is more frequently
encountered in underdeveloped countries (about 6%)
[46]. Improper severing of the umbilical cord, application
of oily substances to the umbilical stump and other unhy-
gienic practices during the neonatal period are important
predisposing factors. Late separation of the umbilical
Fig. 7.4  Bullous impetigo in the neonate. cord may also lead to infection and inflammation.
90 Section 2  Skin Disorders of the Neonate and Young Infant

In most cases, Gram‐negative organisms are responsible The treatment of choice is the parenteral combination of
for omphalitis, particularly Escherichia coli and Klebsiella ampicillin with an aminoglycoside. Listeria is not sensi-
pneumoniae. Infections with group A streptococci and tive to cephalosporins.
staphylococci have also been observed. Polymicrobial
infection is common. Gram staining is a reliable and easy Fungal infections
method for initial identification of the microorganism. Yeast infections of the neonate are common and include
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

A persistent omphalitis may indicate immunodeficiency thrush, neonatal candidiasis (napkin eruption, paronych-
[47]. Cellulitis occurs when inflammation spreads ium) and Malassezia furfur pustulosis. Greater detail on
beyond the umbilicus. When the inflammation extends each of these can be found elsewhere in this book.
to the subcutaneous tissue and underlying fascia, Two types of Candida infection are seen in neonates:
necrotizing fasciitis develops [48]. Necrotizing fasciitis superficial infections and disseminated infections.
is associated with a mortality rate of 50%. Thus, early Disseminated Candida infections are rare but the incidence
diagnosis is strongly indicated, and should be followed appears to be increasing because of higher survival rates
by surgical debridement. of premature and very‐low‐birthweight neonates [53].
Neonates have a high risk of cellulitis and abscess These disseminated infections may manifest as vesicu-
formation following skin injury. Scalp abscesses are asso- lobullous, pustular or a burn‐like exfoliative dermatitis.
ciated with the use of scalp electrodes. Also, puncture Superficial infections such as oral candidiasis (thrush)
wounds from blood sampling or intravenous catheters and Candida napkin dermatitis are common diseases in
are sites where cellulitis or abscesses are found. The neonates, especially when broad‐spectrum antibiotics
microorganisms involved are those that colonize the neo- have been administered.
nate during passage through the birth canal: Staph. aureus, Superficial candidiasis is seen after 1–2 weeks of life,
E. coli and group A and B streptococci. Gram stain and presenting first as oral thrush and then as perianal napkin
culture of the abscess content will typically reveal the dermatitis. Candida species, usually C. albicans, can be iso-
aetiology. Antimicrobial therapy should be tailored to the lated from the lesions and faeces. Neonatal superficial
causative organism and surgery should be considered candidiasis is characterized by a moist erythematous
when fluctuation or abscesses are diagnosed. (‘beefy red’) napkin dermatitis, located near the anus,
Breast abscess is a special entity that appears in full‐ surrounded by satellite lesions. Pustules and vesicles
term neonates within the first 8 weeks. Usually, unilateral may develop. Inner folds such as the intergluteal fold,
swelling of the breast in combination with redness and perineum and inner thighs are often involved. Direct
warmth appears about 1 week after birth. Transplacentally potassium hydroxide smears show the presence of
acquired maternal hormones stimulate the glandular Candida pseudohyphae. Cultures yield mostly Candida
breast tissue causing it to be more susceptible to infec- albicans and rarely another species. Therapy of superfi-
tions. The disease is seen more often in girls than in boys. cial candidiasis includes local antifungal therapy for
Diagnostic procedures and treatment are the same as 7–10 days. Therapy of disseminated candidiasis requires
described previously [49]. a longer course of a parenteral antifungal agent.
Circumcision has proven to have preventive effects on Neonatal M. furfur pustulosis is another common fungal
the development of penile cancer [50] and also leads to a infection. M. furfur is associated with a nonfollicular
lower risk of urinary tract infections as well as a decreased pustulosis on the face and neck of a neonate. This disease
likelihood of acquisition of human immunodeficiency frequently is misdiagnosed as neonatal acne. Identification
virus (HIV). Pathological phimosis and paraphimosis of M. furfur can be made by direct microscopy of pustular
induce inflammation of the glans penis and the prepuce material with May–Grünwald–Giemsa stain. If treatment
(balanitis, posthitis, balanoposthitis). These infections are is warranted, pustules typically respond to topical keto-
extremely painful and primarily affect uncircumcised conazole therapy. A disseminated M. furfur fungaemia
male babies. Chronic balanoposthitis may result in scar- may occur in rare circumstances, usually in premature
ring and secondary phimosis [51]. The circumcision of a neonates who receive parenteral nutrition [54].
neonate is a low‐risk procedure. The most common com- Pityriasis versicolor can be acquired from the mother in
plications are local infections and bleeding. Any infection the neonatal period, often diagnosed in the first 2 weeks
that develops should be treated immediately. of life. The differential diagnosis of multiple depigmented
Listeriosis is a septicaemic disease caused by Listeria lesions in a neonate may include tuberous sclerosis, nae-
monocytogenes, which is a small aerobic non‐spore‐forming vus anaemicus and naevus depigmentosus (achromic
Gram‐positive rod. Transmission occurs via contami- naevus) [55].
nated food, especially products made from unpasteur-
ized milk. Early‐onset infection is acquired in utero; Parasitic infections
late‐onset infection may be acquired after birth. Late‐ Scabies is a parasitic infection caused by Sarcoptes scabiei.
onset infection usually affects full‐term healthy infants. Scabies in infancy is different from scabies in children and
The infection develops soon after birth to weeks there- adults (Fig.  7.5). Pruritus is often subtle or nonexistent.
after [52]. The infection is accompanied by grey‐white Neonatal scabies presents with papules, vesicles, vesicu-
maculopapules with vesicles or pustules. The organism lar burrows and very typical vesicles on palms and soles
can be recovered in routine cultures, but special tech- [36]. Eczematization and impetiginization are common
niques may be necessary to isolate and identify Listeria. and sometimes lead to misdiagnosis. Mites, ova and faeces
Chapter 7  Congenital and Acquired Infections in the Neonate 91

3 Brough AJ, Jones MD, Page RH et al. Dermal erythropoiesis in neonatal


infants: a manifestation of intrauterine viral disease. Pediatrics 1967;
40:627–35.
4 Groark SP, Jampel RM. Violaceous papules and macules in a newborn:
dermal erythropoiesis associated with congenital cytomegalovirus
infection. Arch Dermatol 1989;125:116–17.
5 Bowden JB, Hebert AA, Rapini RP. Dermal hematopoiesis in neo-
nates: report of five cases. J Am Acad Dermatol 1989;20:1104–10.

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
6 Gottesfeld E, Silverman RA, Coccia PF et  al. Transient blueberry
muffin appearance of a newborn with congenital monoblastic leukemia.
J Am Acad Dermatol 1989;21:347–51.
7 Mehta V, Balachandran C, Lonikar V. Blueberry muffin baby: a picto-
ral differential diagnosis. Dermatol Online J 2008;14:8.
8 Koch LH, Fisher RG, Chen C et al. Congenital herpes simplex virus
infection: two unique cutaneous presentations associated with prob-
able intrauterine transmission. J Am Acad Dermatol 2009;60:312–15.
9 Theodoridou M, Kakourou T, Laina I et al. Vesiculopapular rash as a
single presentation in intrauterine coxsackie virus infection. Eur J Pediatr
2002;161:412–13.
10 Sauerbrei A, Glück B, Jung K et al. Congenital skin lesions caused by
intrauterine infection with coxsackievirus B3. Infection 2000;28:326–8.
11 Kimberlin DW, Jester PM, Sánchez PJ et  al. Valganciclovir for
symptomatic congenital cytomegalovirus disease. N Engl J Med
2015;372:933–43.
12 Remington JS, Klein JO, Wilson BW, Nizet V (eds). Infectious
Diseases in the Fetus and Newborn Infant, 7th edn. Philadelphia:
W.B. Saunders, 2010.
13 Shah SS, Gallagher PG. Neonatal sepsis due to echovirus 18 infection.
J Perinat Med 1997;25:381–4.
14 James SH, Kimberlin DW. Neonatal herpes simplex virus infection.
Infect Dis Clin North Am 2015;29:391–400.
15 Connell CO, Oranje A, Van Gysel D, Silverberg NB. Congenital mol-
Fig. 7.5  Neonatal scabies.
luscum contagiosum: report of four cases and review of the literature.
Pediatr Dermatol 2008;25:553–6.
can be visualized in potassium hydroxide preparations. 16 Penouil MH, Bressieux JM, Mehaut S et  al. Hematopoiese cutanee
neonatale revelant une rubeole congenitale. Ann Dermatol Vénéréol
Skin scrapings should be taken from vesicles and burrows. 1996;123:334–6.
Treatment of neonatally acquired scabies is compli- 17 Seno A, Tada J, Matsuura H et al. Congenital rubella syndrome with
cated by the fact that the therapies of choice in older rubella virus‐associated generalized brownish macules, indurated
infants and children are not approved in neonates. Topical erythemas, papules, and pigmentation. J Dermatol 1994;21:323–8.
18 Ostlere LS, Harris D, Stevens HP et al. Chronic rash associated with
permethrin 5% cream applied head to toe and then congenital rubella. J R Soc Med 1994;87:242–3.
washed off after 8–14 h is the most effective treatment in 19 Querol I, Bueno M, Cebrian A et  al. Connatal herpes zoster. Cutis
older infants and children, but is not approved for chil- 1996;58:231–4.
20 Chiang CP, Chiu CH, Huang YC et al. Two cases of disseminated cuta-
dren younger than 2 months. Permethrin kills the scabies
neous herpes zoster in infants after intrauterine exposure to varicella‐
mite and eggs. Two (or more) applications, each about a zoster virus. Pediatr Infect Dis J 1995;14:395–7.
week apart, may be necessary to eliminate all mites. Oral 21 Wu CC, Tsai CN, Wong WR et al. Early congenital syphilis and ery-
ivermectin is effective for treating scabies, but is less thema multiforme‐like bullous targetoid lesions in a 1‐day‐old new-
born: detection of Treponema pallidum genomic DNA from the targetoid
effective than topical permethrin. Because ivermectin is plaque using nested polymerase chain reaction. J Am Acad Dermatol
not ovicidal, it is given as two doses, 7–14 days apart. 2006;55(2 Suppl):S11–15.
Ivermectin is not approved for treatment of scabies by the 22 Spyridis N, Georgouli H, Tsoukatou T et  al. Severe disseminated
US Food and Drug Administration. Although its use has tuberculosis in a 4‐month‐old infant initially presenting with multi‐
form cutaneous lesions. Scand J Infect Dis 2006;38:306–8.
been reported anecdotally, the safety of ivermectin in 23 Longhead LJ. Congenital staphylococcal scalded skin syndrome.
children weighing less than 15 kg has not been deter- Pediatr Infect Dis J 1992;11:413.
mined. Alternative drugs include 10% crotamiton cream 24 Torres‐Martinez C, Mehta D, Butt A et  al. Streptococcus associated
toxic shock. Arch Dis Child 1992;67:126–30.
or lotion, or unapproved 5% to 10% precipitated sulphur 25 Takahashi N, Nishida H, Kato H et al. Exanthematous disease induced
compounded into petrolatum. by toxic shock syndrome toxin 1 in the early neonatal period. Lancet
1998;351:1614–18.
26 Smith KJ, Yeager J, Skelton HG et al. Diffuse petechial pustular lesions
Acknowledgements in a newborn: disseminated Listeria monocytogenes. Arch Dermatol
The authors acknowledge use of material from previous 1994;130:245–8.
27 Desenclos JC, Garrity D, Scaggs M et al. Gonococcal infection of the
chapters in the 3rd edition by Nico G. Hartwig, Arnold P. newborn in Florida, 1984–1989. Sex Transm Dis 1992;19:105–10.
Oranje, Dirk Van Gysel, Marinus C.G. van Praag, and 28 Lopez JB, Gross P, Boggs TR. Skin lesions in association with β hemo-
Peter H. Hoeger. lytic Streptococcus group B. Pediatrics 1976;58:859–60.
29 Kline A, O’Donnell E. Group B Streptococcus as a cause of neonatal
bullous skin lesions. Pediatr Infect Dis J 1993;12:165–6.
References 30 Bernier V, Weill FX, Hirigoyen V et al. Skin colonization by Malassezia
1 Van Praag MC, Van Rooij RW, Folkers E et al. Diagnosis and treat- species in neonates: a prospective study and relationship with neonatal
ment of pustular disorders in the neonate. Pediatr Dermatol cephalic pustulosis. Arch Dermatol 2002; 138:1215–18.
1997;14:131–43. 31 Darmstadt GL, Dinulos JG, Miller Z. Congenital cutaneous candidiasis:
2 Sahn EE. Vesiculopustular diseases of neonates and infants. Curr Opin clinical presentation, pathogenesis, and management guidelines.
Pediatr 1994;6:442–6. Pediatrics 2000;105:438–44.
92 Section 2  Skin Disorders of the Neonate and Young Infant

32 Smolinski KN, Shah SS, Honig PJ, Yan AC. Neonatal cutaneous 44 Yamasaki O, Yamaguchi T, Sugai M et al. Clinical manifestations of
fungal infections. Curr Opin Pediatr 2005;17:486–93. staphylococcal scalded‐skin syndrome depend on serotypes of exfo-
33 Maldonado YA, Read JS, AAP Committee on Infectious Diseases. liative toxins. J Clin Microbiol 2005;43:1890–3.
Diagnosis, treatment, and prevention of congenital toxoplasmosis 45 LaVoo EJ, Paller AS. Common skin problems during the first year of
in the United States. Pediatrics 2017;139:e20163860. life. Pediatr Clin North Am 1994;41:1105–19.
34 Woodruff CA, Hebert AA. Neonatal primary cutaneous aspergillosis: 46 Fraser N, Davies BW, Cusack J. Neonatal omphalitis: a review of its
case report and review of the literature. Pediatr Dermatol 2002;19:439–44. serious complications. Acta Paediatr 2006;95:519–22.
35 Chang S‐E, Kang S‐K, Choi J‐H et al. Tinea capitis due to Trichophyton 47 Guneser S, Altintas DU, Aksungur P et  al. An infant with severe
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

rubrum in a neonate. Pediatr Dermatol 2002;19:356–8. ­leucocyte adhesion deficiency. Acta Paediatr 1996;85:622–4.
36 Kim D, Teng J. Scabies infection in a neonate. J Pediatr 48 Samuel M, Freeman NV, Vaishnav A et  al. Necrotizing fasciitis: a
2014;165:1266–1266.e1 serious complication of omphalitis in neonates. J Pediatr Surg 1994;
37 Brown ZA, Wald A, Morrow RA et al. Effect of serologic status and 29:1414–16.
cesarean delivery on transmission rates of herpes simplex virus from 49 Faden H. Mastitis in children from birth to 17 years. Pediatr Infect Dis
mother to infant. JAMA 2003;289:203–9. J 2005;24:1113.
38 American College of Obstetricians and Gynecologists Practice Bulletin. 50 Schoen EJ. The relationship between circumcision and cancer of the
Management of herpes in pregnancy. Obstet Gynecol 2007;109:1489–98. penis. CA Cancer J Clin 1991;41:306–9.
39 Pinninti SG, Angara R, Feja KN et al. Neonatal herpes disease fol- 51 Wiswell T. Circumcision circumspection. N Engl J Med
lowing maternal antenatal antiviral suppressive therapy: a multi- 1997;336:1244–5.
center case series. J Pediatr 2012;161:134–8.e131–3. 52 Mylonakis E, Paliou M, Hohmann EL et  al. Listeriosis during
40 Sauerbrei A, Wutzler P. Neonatal varicella. J Perinatol 2001;21:545–9. pregnancy: a case series and review of 222 cases. Medicine
41 Sauerbrei A, Wutzler P. Herpes simplex and varicella‐zoster virus 2002;81:260–9.
infections during pregnancy: current concepts of prevention, diagno- 53 Manzoni P, Stolfi I, Pugni L et al. A multicenter randomized trial of
sis and therapy. Part 2: varicella‐zoster virus. Med Microbiol Immunol prophylactic fluconazole in preterm neonates. N Engl J Med
2007;196:95–102. 2007;356:2483–95.
42 Ginsberg CM. Staphylococcal toxin syndromes. Pediatr Infect Dis J 54 Devlin RK. Invasive fungal infections caused by Candida and
1991;10:319–21. Malassezia species in the neonatal intensive care unit. Adv Neonatal
43 Ladhani S. Understanding the mechanism of action of the exfoliative Care 2006;6:68–77.
toxins of Staphylococcal aureus. FEMS Immunol Med Microbiol 55 Wire HW Jr, Johnson WT. Neonatal pityriasis versicolor. Arch
2003;39:181–9. Dermatol 1981;117:752–3.
 93

C HA PTER   8

Transplacentally Acquired Dermatoses


Paula Carolina Luna

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Hospital Alemán, Buenos Aires, Argentina

Neonatal lupus erythematosus, 93 Transplacental melanoma, 98


Transplacentally acquired neonatal bullous autoimmune Transplacental Behçet disease, 99
disorders, 97

as maternal antibodies wane from the neonatal circulation. This


Abstract group includes neonatal lupus erythematosus and transplacentally
acquired autoimmune bullous diseases (pemphigoid gestationis,
Transplacentally acquired dermatoses are a group of rare diseases pemphigus vulgaris and foliaceus and epidermolysis bullosa acquisita).
that affect the skin and are due to the action of maternal anti- Although Behçet disease has no recognizable antibodies, it shows
bodies that cross the placenta. They all behave in a similar way, a similar behaviour. Transplacental metastasis of maternal melanoma
being present at birth (or developing shortly after) and resolving will also be briefly addressed.

Key points maternal antibody (SSA/Ro, SSB/La or U1RNP). It may affect


the neonatal skin, heart, liver, blood, brain and even the bones.
• Transplacentally acquired dermatoses are rare and typically Cardiac manifestations can be severe and life‐threatening and
caused by transplacental passage of maternal autoantibodies. are the only manifestation that may persist through life.
• Neonatal lupus (NL), being the most frequent of this group, • Manifestations are transient and resolve as maternal antibodies
represents a syndrome caused by the tranplacental passage of clear from the infant’s blood.

Neonatal lupus erythematosus Pathogenesis


According to current knowledge, NL is due mainly to the
Introduction transplacental passage of maternal antibodies, although
Neonatal lupus (NL) is a disease that was first described
several other factors, both maternal and fetal, have also
by McCuiston et al. in 1954. It represents a model of pas-
been implicated [3].
sive autoimmune disease in which pathogenic maternal
The main antibody associated with NL is anti‐Ro/SSA,
antibodies (mainly SSA/Ro, SSB/La and less frequently
followed by anti‐La/SSB, and rarely anti‐U1 RNP [4]. It
U1RNP) are transmitted through the placenta. It is
has been hypothesized that it is the titre of maternal anti-
reported to affect 1% to 2% of children born to mothers
bodies rather than their presence that is associated with
with these antibodies and is characterized by the variable
cardiac injury. Approximately 15% of NL patients with
presence of dermatological, cardiac, haematological or
cardiac issues had Ro‐values of ≥50 U/mL while 85% had
hepatic manifestations. Aside from heart block, most
≥100 U/mL; overall, the rate of complete heart block was
manifestations are transient and completely resolve as the
5%. Those with low titres of antibodies had a 0% rate of
passive antibodies clear from the newborn’s blood.
complete heart block. High anti‐La levels (≥100 U/mL)
Cardiac manifestations are the exception; these may be
were also more related to noncardiac features of NL [5].
life‐threatening and can persist throughout life.
Anti‐U1‐RNP antibodies are less commonly seen,
although they have been reported in around 5% of patients.
Epidemiology
These neonates may develop cutaneous, haematological,
NL has an estimated incidence of around 1 in 12 500 to
hepatic or osteoarticular manifestations, but have never
20 000 live births [1] although this number is probably
been reported to develop cardiac manifestations [6,7].
underestimated because of the self‐healing nature of the
While all these antibodies are necessary to the develop-
extracardiac manifestations that are only diagnosed when
ment of NL, they are not the only determinant of the
there is a high degree of suspicion. It is considered to be
development of NL. Fetal susceptibility may be influ-
the most frequent transplacental immune‐mediated
enced by specific HLA alleles, mainly DQB1*02, DRB1*03
dermatosis [2] as well as the most frequent cause of con-
and a polymorphism in the promoter region of the gene
genital heart block.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
94 Section 2  Skin Disorders of the Neonate and Young Infant

for tumour necrosis factor‐α [8]. Discordance in identical Lesions tend to develop around the sixth week after
twins raises concern about other possible in utero factors. birth, although some cases present as early as at birth or
Animal models have revealed that reactivity to the shortly thereafter. Reports exist that document the skin
p200 region of the Ro52 protein and antibody targeting of findings being triggered following exposure to photo-
L‐type calcium channels seem to be relevant in the devel- therapy for neonatal hyperbilirubinaemia [1].
opment of cardiac NL. In vitro studies support a protec- The most typical lesions are erythematous, arcuate,
SECTION 2: SKIN DISORDERS

tive role of β2 glycoprotein 1 (by preventing anti‐Ro


OF NEONATES AND INFANTS

polycyclic or annular macules, patches and papules.


binding to apoptotic cells) and pathological roles of the Lesions typically show slight central atrophy, with or
urokinase–plasminogen activator/receptor system (lead- without a fine scale, appearing mainly on the scalp and
ing to activation of transforming growth factor [TGF]‐β) periorbital area with a tendency to be confluent around
and endothelin‐I secretion by macrophages in mediating the eyes giving the baby an ‘owl eyes’, ‘raccoon eyes’ or
tissue injury. Genetic studies highlight the fetal major his- ‘owl mask’ appearance [15] (Figs  8.1 and 8.2). These
tocompatibility complex (MHC) in the development of lesions show some clinical and histological similarity to
disease, and a multigenerational study demonstrated that subacute cutaneous lupus erythematosus (SCLE) with
mothers of NL children accumulate genetic risk factors interface changes, vacuolar alteration in the basement
preferentially from the NL child’s grandparents [9]. membrane and dermoepidermal junction, along with epi-
Mothers of patients with NL may suffer from systemic dermal atrophy and perivascular inflammatory infiltrates
lupus erythematosus (SLE) [10], Sjögren syndrome, SLE/ with lymphocyte dominance [16].
SS, undifferentiated connective tissue disease or leukocy- Urticaria‐like lesions (Fig.  8.3) are characterized by
toclastic vasculitis [11], or be completely asymptomatic, polycyclic erythematous plaques that mainly affect the
although when tested mothers always show positive anti- scalp and face but can also appear on the trunk or limbs.
bodies. Although asymptomatic, mothers with positive On histology these lesions may also show interface
antibodies will need to be followed up. Progression to SLE changes or a superficial and deep perivascular and inter-
and SS does not invariably occur. In fact, only about 10% stitial lymphocytic infiltrate [16].
of mothers developed SLE and 14% developed SS during Atrophic lesions [17] (Fig.  8.4), bullae, telangiectasia
a 15‐year follow‐up period. [12]. [18], purpura, erythema multiforme‐like lesions (Fig. 8.5),
Maternal hypothyroidism has been suggested to widespread erosions [19], cutis marmorata telangiectatica
increase the risk nine‐fold of having an affected fetus with congenita (CMTC)‐like [20,21] (Fig. 8.6) lesions as well as
congenital heart block compared to mothers without
hypothyroidism [13].

Clinical manifestations
Neonatal lupus may show a spectrum of clinical manifes-
tations. Affected organs include the skin, the heart, the
blood, the liver, the brain and the bones. Most of the lit-
erature reports a similar incidence of around 50% for both
skin and heart and 15% for all the other symptoms col-
lectively. Any individual patient may show one to several
of these manifestations. According to the Research
Registry for Neonatal Lupus (RRNL; a nationwide US
registry of NL patients), 61% of NL patients present with
cardiac disease alone, 26.9% with cutaneous manifesta-
tions, 8.7% with both cutaneous and cardiac, and 3.2%
with hepatic or haematological abnormalities [3].

Cutaneous manifestations
Cutaneous lesions are highly variable and polymorphous
[14]. The different morphologies are listed in Box 8.1.

Box 8.1  Dermatological manifestations of neonatal lupus

• ‘Raccoon eye’ or ‘owl mask’ erythema


• Erythema multiforme
• Cutis marmorata telangiectatica‐like skin changes
• Bullous skin changes
• Eczematous skin changes
• Telangiectasias
• Extensive capillary malformation
• Targetoid lesions
• Varicelliform lesions Fig. 8.1  Erythematous arcuate plaques over the forehead and around the
eyes, with the typical ‘owl mask’ appearance.
Chapter 8  Transplacentally Acquired Dermatoses 95

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Fig. 8.2  Scaly circinate plaques around the eyes, with the typical
‘owl‐mask’ appearance.
Fig. 8.5  Erythema multiforme‐like lesions over the face.

Fig. 8.3  Urticarial lesions over the chest of a newborn.


Fig. 8.6  Cutis marmorata telangiectatica congenita‐like lesions over the
abdomen.

Fig. 8.4  Congenital atrophic lesions over the face.


Fig. 8.7  Erosions in the napkin area, showing that non‐photo‐exposed
lesions mimicking an extensive capillary malformation [22] areas can also be affected.
have all been described more rarely in affected patients.
A report of one patient that presented with alopecia, atro- Although these lesions tend to be aggravated with sun
phy and erosions has also been published [23]. exposure or phototherapy, they can appear in non‐exposed
In addition to the cutaneous eruption, mucosal ulcera- areas such as the feet or napkin area (Fig.  8.7) [24] and
tion has been documented in several patients. even be present at birth, before there is any sun exposure.
96 Section 2  Skin Disorders of the Neonate and Young Infant

The clinical differential diagnosis includes but is not Haematological manifestations


limited to: a drug eruption, urticaria, bullous pemphi- Any haematological lineage can be affected by the
goid, congenital syphilis, erythema multiforme, cutis maternal passage of antibodies. Thrombocytopenia,
marmorata telangiectatica congenita, tinea corporis and haemolytic or aplastic anaemia and neutropenia have all
annular erythema of infancy, among many others. been described in NL patients. These manifestations
Most of the lesions tend to resolve completely, paralleling tend to appear in 10% [29] of patients and rarely lead to
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

the decline in maternal antibodies around 6–9 months [25] complications such as bleeding or sepsis [14]. Although
although in some instances some residual hypo‐ or hyper- lymphopenia is a relatively common manifestation of
pigmentation, atrophic scars or telangiectasias may remain. lupus in adults, it is not a feature of NL [15].

Cardiological manifestations Neurological manifestations


More than half of the affected patients present different Hydrocephalus and macrocephaly have been described
types of cardiac manifestations. NL accounts for 80–95% in patients with NL. Some degree of neuropsychiatric
of the cases of congenital complete atrioventricular block dysfunction, compared with normal controls, has also
(CAVB) diagnosed in utero [26]. This alteration develops been reported. Abnormal computed tomography (CT)
usually during a vulnerable period from weeks 18 to 24 findings have been reported in affected patients, including
and rarely arises after week 30. Binding of autoantibodies nonspecific white matter changes, calcification of the basal
to the cardiac conduction system at this early stage of ganglia and subependymal cysts [30]. A case of transient
development results in cardiac dysfunction that does not myasthenia gravis has also been reported [31]. These
undergo spontaneous regression. findings are also thought to be transient and reverse as
Although the precise pathogenic mechanism of the the antibodies clear from the neonatal circulation.
cardiac injury remains unknown, it is thought to be a In a study evaluating the potential neurotoxic effect
two‐stage process. First, maternal anti‐Ro antibodies of autoantibodies from NL, no statistical difference in
bind to fetal cardiomyocytes leading to apoptosis. The attention and behavioral problems, depression, anxiety,
subsequent binding of anti‐La antibodies to the apop- developmental delays, learning, hearing, and speech prob-
totic cardiomyocytes starts the inflammatory cascade lems was noted among patients and their siblings [32].
that finally leads to fibrosis and calcification of the
conducting tissue and surrounding myocardium.
In addition to CAVB, several other electrophysiological Chondrodysplasia punctata
abnormalities have been described, including transient The term chondrodysplasia punctata (CDP) describes a
and persistent sinus node dysfunction, long QT interval, heterogeneous group of disorders characterized by the
ventricular and atrial ectopy, ventricular and junctional radiological finding of punctuate calcification of cartilage
tachycardia and atrial flutter. [33]. The different aetiologies responsible for these find-
A more diffuse myocardial disease with ventricular ings include disorders of peroxisome biosynthesis,
dilation, myocardial hypertrophy or endocardiofibroelas- maternal conditions (including anti‐SSA/Ro, SSB/La and
toidosis [27] has also been described. anti‐U1RNP), and teratogens, especially anticoagulants
Structural congenital heart disease including persistent [34]. Several patients with CDP and maternally derived
ductus arteriosus, atrial and ventricular septal defects, antibodies (mainly anti‐U1RNP) have been described.
semilunar and atrioventricular valve abnormalities has These patients may present only with CDP or may also
been reported in 16–42% of patients. These alterations develop cutaneous or haematological manifestations. No
may also present in patients in the absence of CAVB, patient with CDP and associated cardiac manifestations
suggesting the possibility that maternal autoimmune‐ has been reported to date.
mediated pathology may also play a role in other forms of The proposed mechanism for the bony defects and
congenital heart disease. stippling in CDP‐associated maternal lupus is immune‐
Most children with CAVB will require permanent mediated, attributed to maternal autoantibodies crossing
heart pacing before adulthood with 60% paced during the placenta in early to mid gestation, and may involve
the neonatal period [28]. the inhibition of a high‐affinity calcium‐binding protein,
calreticulin, which is a multifunctional protein of the
Liver disease endoplasmic reticulum. Ro/SSA is an autoantigen com-
Liver involvement is said to occur in approximately plex that may include calreticulin.
15% of patients with NL, but its incidence is probably As with other extracutaneous manifestations, the
underestimated [14]. In many patients, liver disease is stippling of the bones can disappear with time.
asymptomatic and transient or may be misdiagnosed as
physiological jaundice [1]. It usually presents as asympto- Other manifestations
matic transaminitis, hepatomegaly, transient hepatitis, Pulmonary complications of NL such as transient self‐
hyperbilirubinaemia or, rarely, liver failure [29]. Most of limited pneumonitis and pulmonary capillaritis and
these manifestations (except that of fulminant liver failure) haemorrhage have been described although they are
are transient and, as seen in the other noncardiac organs exceedingly rare [35]. Nephritis has rarely been described
affected in NL, resolve spontaneously concurrent with [36]. A rare case with unilateral pectoris major atrophy has
waning of maternally derived antibodies. also been reported [37].
Chapter 8  Transplacentally Acquired Dermatoses 97

Diagnosis recurrence rate can be unusually high in some families: in


As there are no diagnostic criteria for NL, the diagnosis is one case report four affected siblings were documented [25].
made in a newborn presenting with one or more of the It is suggested that all pregnant women with autoim-
previously described clinical findings (cutaneous, car- mune symptoms should undergo early first‐trimester
diac, haematological, hepatic, osseous) in the context of screening for SSA/Ro and SSB/La antibodies. If they are
positive antibodies (anti‐SSA/Ro, SSB/La or anti‐U1RNP) positive, fetal PR interval should be monitored by

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
both in the patient and the mother. echocardiogram weekly between weeks 16 and 26 and
biweekly between weeks 26 and 32 [45].
Evaluation All patients should have long‐term follow‐up, because
Once the diagnosis is established, affected patients should although extracardiac manifestations resolve spontane-
be evaluated for other manifestations of NL. A cardiologi- ously early in life, these patients seem to be at a higher
cal evaluation with an electrocardiogram (ECG) and an risk of developing other autoimmune diseases during
echocardiogram should be performed in every patient. A their childhood. A long‐term follow‐up study of NL
blood test including platelet count and liver function is patients and their unaffected siblings [42] showed that six
also advisable at the time of diagnosis. If the patient is out of 49 patients had a definite rheumatic/autoimmune
U1RNP positive, a radiological evaluation is also advisa- disease (two had juvenile rheumatoid arthritis, one had
ble. Neurological evaluation should be suggested if there Hashimoto thyroiditis, one had psoriasis and iritis, one
are clinical symptoms of neurological involvement. had psoriasis and diabetes, and one had diabetes and
nephrotic syndrome) by the age of 8 years or older.
Treatment
As cutaneous lesions are usually transient, sun avoidance
is the only recommended intervention. Under some spe- Transplacentally acquired neonatal
cial circumstances topical calcineurin inhibitors or topical bullous autoimmune disorders
corticosteroids might be considered [38]. Transplacental passage of maternal antibodies to several
Given that complete heart block is irreversible and can autoimmune blistering diseases has been reported to
currently only be treated with a pacemaker, prevention is affect newborns. Neonatal blisters or erosions (or lesions
the most promising strategy. The use of prophylactic developing shortly after birth) have been documented in
intrapartum hydroxychloroquine has been evaluated the infants of mothers with pemphigus vulgaris, pemphi-
through a case‐control study. In this study, exposure to gus foliaceus, gestational pemphigoid and epidermolysis
hydroxychloroquine during pregnancy was demon- bullosa acquisita [46–48] (Table  8.1). Although the true
strated to decrease the risk of fetal cardiac NL among neo- incidence of transplacentally acquired neonatal bullous
nates born to mothers with these antibodies [39]. autoimmune disorders is unknown [49], they seem to be
Hydroxychloroquine inhibits the endosomal acidification exceedingly rare.
required for optimal toll‐like receptor (TLR) signalling, The presumed mechanism of disease is passive transfer
whose activation is known to promote cardiac inflamma- of maternal IgG autoantibodies across the placental tis-
tion and scarring. sues. Hormonal changes of pregnancy can modulate
Systemic fluorinated steroids administered to the mother autoimmune bullous diseases. Vertically transmitted
have been widely used, with questionable efficacy and autoimmune conditions are caused by passive transfer of
risks [3]. When first‐ or second‐degree heart block is IgG antibodies, which are able to cross the placenta begin-
detected in utero, systemic fluorinated steroids (dexameth- ning at week 13 and are present at highest levels in the
asone and betamethasone) may help reverse the cardiac third trimester [50].
alteration. In utero treatment with glucocorticosteroids for Management of these diseases during the neonatal
these degrees of heart block is generally not advised unless period usually involves use of bland emollients and topi-
the patient also presents with myocarditis [28]. cal corticosteroids, if needed, because disease activity will
As it is well known that the rate of recurrence in moth- probably wane as maternal autoantibodies are cleared
ers with a previous affected child rises dramatically, the from the affected neonate. New blisters rarely develop
use of prophylactic intravenous immunoglobulin (IVIG) past the newborn period [50].
in subsequent pregnancies has been evaluated, but
proved not to be as effective as prophylactic therapy for
CAVB in high‐risk mothers [40,41].
Table 8.1  Targets of different immunologically‐mediated bullous neonatal
Long‐term follow‐up diseases
The risk of a second affected neonate after the first NL
child is about 17–36% [42,43]. Cardiac NL recurrence is Type of immune bullous disorder Target
thought to be approximately 17% and seems to be
unaffected by maternal health, use of steroids, antibody Pemphigoid gestationis BP180
Neonatal pemphigus Desmoglein 3
status, severity of cardiac disease in the first affected child
Neonatal epidermolysis bullosa 145‐kDa NC1 domain of
or gender of the fetus [44]. For these reasons, subsequent acquisita collagen VII
pregnancies should be strictly monitored and maternal Neonatal pemphigus foliaceus Desmoglein 1
prophylactic systemic therapy may be considered. The
98 Section 2  Skin Disorders of the Neonate and Young Infant

Gestational pemphigoid keratinocytes [56]. The frequency of neonatal PF is exceed-


Pemphigoid gestationis, previously known as herpes ges- ingly low and this is thought to be due to the fact that in
tationis, is a rare pregnancy‐associated immunobullous neonatal epidermis (in contrast to adult epidermis),
disease. Affected mothers usually develop the skin condi- desmoglein 3 is coexpressed in the superficial epidermis
tion during the second or third trimester of pregnancy and together with desmoglein 1. Therefore, the lack of des-
findings resolve within a few weeks after labour. It has moglein 1 is compensated by desmoglein 3, providing a
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

been shown that the pathogenic antibody to BP180 certain degree of protection [55]. Confirming this, a study
increases immediately before and after delivery in patients of 19 neonates born to mothers with active fogo selvagem
with gestational pemphigoid, presumably in response to (endemic PF) showed that no skin disease was manifested
the release of the BP180 antigen from the chorionic mem- in the neonates despite positive Dsg1 autoantibodies in
brane. The exact role of hormones in accelerating bulla 9 of 19 neonates.
formation or pathogenic antibody titre is unknown. Very few cases of neonatal PF have been reported in the
Pemphigoid gestationis is exceedingly rare, with an literature. In all of them maternal antibody titres against
estimated incidence of 1 in 50 000 pregnancies. Neonatal desmoglein 1 were very high (1 : 320 and 1 : 640) [57]. Clinical
bullous pemphigoid has been rarely reported in around manifestations included superficial erosions and crusts
5–10% of newborns of affected mothers [51]. There is a with no mucosal involvement. Management with topical
strong male predominance with a male : female ratio of corticosteroids seems to be sufficient and lesions have been
4.6, in contrast to adult bullous pemphigoid which has reported to be completely resolved at 2 weeks of age.
female : male ratios ranging from 1.01 to 2.00 [49].
Lesions in newborns can appear as blisters, erosions, Epidermolysis bullosa acquisita
vesicles, urticarial plaques or reticular eruptions [49] and Epidermolysis bullosa acquisita (EBA) is an extremely
yellow plaques with erythematous bases. Lesions typi- rare autoimmune bullous disorder caused by circulating
cally spontaneously resolve within 1–3 weeks [52], with IgG autoantibodies directed against the 145‐kDa NC1
antibody negativization by around 1 month [53]. Babies domain of collagen VII. Only one case of transplacentally
are affected due to transfer of pathogenic antibodies to a acquired EBA has been reported in the literature to date.
subunit of bullous pemphigoid antigen (180 kDa). Apart This patient, born to a mother with a well‐known history
from the rare occurrence of neonatal blistering, pemphig- of EBA, presented at birth with generalized blisters and
oid gestationis has occasionally been associated with other erosions including nares and lips, while sparing the
complications such as adverse birth outcomes, including mucous membranes. At 2 months of age, lesions were
miscarriage, low birthweight and prematurity [48]. completely re‐epithelialized with the development of
numerous milia [46].
Neonatal pemphigus vulgaris
Neonatal pemphigus vulgaris is a rare transient autoim-
Transplacental melanoma
mune blistering disorder caused by the transfer of mater-
nal IgG autoantibodies against desmoglein 3 to the neonate Malignancies during pregnancy are calculated to occur
born from a mother with pemphigus vulgaris [54]. in one every 1000 pregnancies, with a frequency that
Antibody titres in the mother have ranged from 1 : 20 to parallels that of age‐matched nonpregnant women.
1 : 640, and several authors suggest that neither antibody These include breast, cervix, lung, melanoma and hae-
titres nor the clinical severity of the mother’s findings matological malignancies [58]. The estimated incidence
predict the severity of disease in the child. In fact, of all of melanoma during pregnancy is 0.1 to 2.8 per 1000
reported transient bullous transplacentally acquired pregnancies. Fetal metastases are rare, with around 100
dermatoses, only neonatal pemphigus has been reported in cases reported in the literature [59]. Melanoma is the
neonates born from clinically asymptomatic mothers [55]. most common neoplasm with transplacental transmission
It is clinically characterized by transient flaccid blisters to the fetus [60]. Given that 45% of melanoma patients
and erosions on the skin and, very rarely, the mucous are diagnosed in the first four decades of life, and with
membranes because of the compensatory expression of rates of melanoma rising over the last two decades, this
desmoglein 1 in infant mucous membranes. It may be incidence may rise [61].
present at birth or develop during the first 2 weeks of life. Lesions have been identified between birth and 8
It has never been reported to persist beyond the neonatal months of age [62], and they may appear as cutaneous
period or progress to adult disease [47]. Cutaneous lesions pigmented lesions or as internal metastases.
regress in less than 3 weeks and circulating antibodies are Although exceptional cases of spontaneous regression
not detected longer than 2 months after birth [53]. have been reported [58], most neonates with clinical evi-
Treatment may consist of supportive measures alone, dence of maternal metastasis at time of delivery have an
emollients or topical corticosteroids, depending on exceedingly poor prognosis. Death typically occurs before
severity. the age of 3 months. Placental involvement seems to be a
key risk factor among patients with no clinical evidence
Neonatal pemphigus foliaceus of metastasis at birth [62]; for that reason, the placenta
Pemphigus foliaceus (PF) is an autoantibody‐mediated should be evaluated in these cases, and all children born
epidermal blistering disease caused by antibodies against to mothers with evidence of placental involvement should
desmoglein 1 with loss of adhesion between superficial be evaluated and strictly monitored [58].
Chapter 8  Transplacentally Acquired Dermatoses 99

The diagnosis of metastatic melanoma in a child born Given that all cases of neonatal BD have been reported
to a mother with known metastatic melanoma seems among symptomatic mothers, and colchicine reduces the
straightforward when there are pigmentary lesions evi- risk of severe BD flares, the use of this drug could be
dent in a newborn from a mother with known metastatic considered as prophylactic therapy during pregnancy.
melanoma. In some circumstances where congenital Colchicine has an antimitotic effect and crosses the pla-
melanocytic naevi may appear clinically atypical and centa. The safety of this drug was evaluated in one study

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
might even show benign proliferative nodules with high in which 238 colchicine‐exposed pregnancies showed no
mitotic rates on the biopsy, the diagnosis of transplacental increase in teratogenicity or congenital abnormalities [67].
melanoma may be confirmed with the use of modern
genetic techniques such as polymerase chain reaction References
(PCR) with 5 STR polymorphic loci and imaging mass 1 Wisuthsarewong W, Soongswang J, Chantorn R. Neonatal lupus
erythematosus: Clinical character, investigation and outcome. Pediatr
spectrometry analysis [62]. Dermatol 2011;28:115–21.
There is no currently approved treatment for paediatric 2 Aguilera Peiró P, Vicente Villa A, Gonzalez Enseñat MA et  al.
metastatic melanoma, and consultation with a paediatric Espectro clínico del lupus eritematoso neonatal cutáneo. An
Pediatr 2009;70:287–92.
oncologist for further evaluation and treatment is indicated. 3 Izmirly PM, Rivera TL, Buyon JP. Neonatal lupus syndromes. Rheum
Dis Clin North Am 2007;33:267–85.
4 Ortiz‐Santamaria V, Olive A, Martinez‐Caceres E et  al. Neonatal
Transplacental Behçet disease lupus erythematosus: A possible role for anti‐Sm antibodies. Lupus
2010;19:659–61.
Behçet disease (BD) is a rare, chronic, multisystem disease 5 Jaeggi E, Laskin C, Hamilton R et al. The importance of the level of
of unknown cause characterized by mucocutaneous, ocu- maternal anti‐Ro/SSA antibodies as a prognostic marker of the
lar, vascular and central nervous system manifestations development of cardiac neonatal lupus erythematosus a prospec-
tive study of 186 antibody‐exposed fetuses and infants. J Am Coll
and is associated with thrombogenicity [63]. No laboratory
Cardiol 2010;15:2778–84.
diagnostic test is available and the diagnosis remains 6 McGeachy C, Lam J. Anti‐RNP neonatal lupus in a female newborn.
clinical. The International Study Group for Behçet’s Disease Lupus 2009;18:172–4.
has reached agreement on a set of diagnostic criteria [64,65] 7 Heelan K, Watson R, Collins SM. Neonatal lupus syndrome associ-
ated with ribonucleoprotein antibodies. Pediatr Dermatol 2013;30:
(Table 8.2). Although the aetiology remains unknown, it is 416–23.
believed to be an immune‐mediated disease, supported by 8 Clancy RM, Backer CB, Yin X et al. Genetic association of cutaneous
the fact that transient cases of BD in neonates born to moth- neonatal lupus with HLA class II and tumor necrosis factor: implica-
ers with this disease have been reported. tions in pathogenesis. Arthritis Rheum 2004;50:2598.
9 Izmirly PM, Peter M. Buyon JP, Saxena A. Neonatal lupus: advances
In neonates, lesions tend to appear at around 1 week of in understanding pathogenesis and identifying treatments of
age and resolve by 2 months of age. Clinical manifesta- cardiac disease. Curr Opin Rheumatol 2013;24:466–72.
tions include different degrees of orogenital ulcerations 10 Demaestri M, Sciascia S, Kuzenko A et  al. Neonatal lupus in triplet
pregnancy of a patient with undifferentiated connective tissue disease
and pustulonecrotic skin lesions associated with fever. evolving to systemic lupus erythematosus. Lupus 2009;18:368–71.
In all reported cases, mothers were symptomatic during 11 Borrego L, Rodriguez J, Soler E et al. Neonatal lupus erythematosus
pregnancy, although they were not always diagnosed as related to maternal leukocytoclastic vasculitis. Pediatr Dermatol
having BD. A case of life‐threatening transient BD in a 1997;14:221–5.
12 Rivera TL, Izmirly PM, Birnbaum BK et al. Disease progression in
neonate has also been reported [66]. mothers of children enrolled in the Research Registry for Neonatal
Lupus 2009;6:828–35.
13 Spence D, Hornberg L, Hamilton R, Silverman E. Increased risk of
complete congenital heart block in infants born to women with hypo-
Table 8.2  The International Study Group for Behçet’s Disease diagnostic thyroidism and anti.Ro and/or anti‐La antibodies. J Rheumatol
criteria (1990) 2006;33:167–70.
14 Silverman E, Jaeggi E. Non‐cardiac manifestations of neonatal
lupus erythematosus. Scand J Immunol 2010;72:223–5.
Required Recurrent Minor aphthous, major aphthous or
15 Lee L. The clinical spectrum of neonatal lupus. Arch Dermatol Res
criteria oral herpetiform ulceration observed by
2009;301:107–10.
ulcerations physician or patient, which recurred at 16 Peñate Y, Guillermo N, Rodríguez J et al. Histopathologic character-
least 3 times in one 12‐month period istics of neonatal cutaneous lupus erythematosus: description of
Minor Recurrent Aphthous ulceration or scarring observed by five cases and literature review. J Cutan Pathol 2009;36:660–7.
criteria genital physician or patient 17 Diociaiuti A, Paone C, Giraldi L et al. Congenital lupus erythemato-
ulceration sus: case report and review of the literature. Pediatr Dermatol
2005;22:240–2.
Eye lesion Anterior uveitis, posterior uveitis or cells in
18 Guinovart R, Vicente A, Rovira C et al. Facial telangiectasia: an unu-
vitreous on slit‐lamp examination or retinal
sual manifestation of neonatal lupus erythematosus. Lupus
vasculitis observed by ophthalmologist 2012;21:552–5.
Skin lesions Erythema nodosum observed by physician or 19 Lynn Cheng C, Galbraith S, Holland K. Congenital lupus erythemato-
patient, pseudofolliculitis or papulopustular sus presenting at birth with widespread erosions, pancytopenia, and
lesions, or acneiform nodules observed by subsequent hepatobiliary disease. Pediatr Dermatol 2010;27:109–11.
physician in post‐adolescent patients not 20 Trevisan F, Cunha PR, Pinto CAL, Cattete FG. Cutaneous neonatal
on corticosteroid treatment lupus with cutis marmorata telangiectatica congenita‐like lesions. An
Positive (Behçetine test) read by physician 24–48 hours Bras Dermatol 2013;88:428–31.
21 Heughan CE, Kanisberg N. Cutis marmorata telangiectatica congen-
pathergy
ita and neonatal lupus. Pediatr Dermatol 2007;24:320–1.
test
22 Vílchez‐Márquez F, Martín‐Fuentes A, Conejero R et  al. Neonatal
lupus erythematosus mimicking extensive capillary malformation.
Source: Adapted from [65]. Reproduced with permission of Elsevier. Pediatr Dermatol 2013;30:495–7.
100 Section 2  Skin Disorders of the Neonate and Young Infant

23 Crowley E, Frieden I. Neonatal lupus erythematosus: an unusual con- 44 Llanos C, Izmirly PM, Katholi M et  al. Recurrence rates of cardiac
genital presentation with cutaneous atrophy, erosions, alopecia, and manifestations associated with neonatal lupus and maternal/fetal
pancytopenia. Pediatr Dermatol 1998;15:38–42. risk factors. Arthritis Rheum 2009;60:3091–7.
24 Admani S, Krakowski A. Neonatal lupus erythematosus presenting 45 Brucato A, Cimaz R, Caporali R. Pregnancy outcomes in patients with
as atypical targetoid‐like lesions involving genitals and soles of feet autoimmune diseases and anti‐Ro/SSA antibodies. Clin Rev Allergy
following brief sun exposure. J Clin Aesthet Dermatol 2013;6:19–23. Immunol 2011;40:27–41.
25 Monari P, Gualdi G, Fantini F, Giannetti A. Cutaneous neonatal lupus 46 Abrams ML, Smidt A, Benjamin L et al. Congenital epidermolysis bul-
erythematosus in four siblings. Br J Dermatol 2008;158:626–8. losa acquisita. Arch Dermatol 2011;147:337.
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

26 Hornberger LK, Al Rajaa N. Spectrum of cardiac involvement in neo- 47 Panko J, Florell SR, Hadley J et al. Neonatal pemphigus in an infant
natal lupus. Scand J Immunol 2010;72:189–97. born to a mother with serologic evidence of both pemphigus vulgaris
27 Guettrot‐Imbert G, Cohen L, Fermont L et al. A new presentation of and gestational pemphigoid. J Am Acad Dermatol 2009;60:1057–62.
neonatal lupus: 5 cases of isolated mild endocardial fibroelastosis 48 Huilaja L, Mäkikallio K, Sormunen R et  al. Gestational pemphigoid:
associated with maternal Anti‐SSA/Ro and Anti‐SSB/La antibodies. Placental morphology and function. Acta Derm Venereol 2013;93:33–8.
J Rheumatol 2011;38:378–86. 49 Zhao CY, Chiang YZ, Murrell DF. Neonatal autoimmune blistering
28 Buyon JP, Clancy RM, Friedman D. Cardiac manifestations of neona- disease: a systematic review. Pediatr Dermatol 2016;33:367–74.
tal lupus erythematosus: guidelines to management, integrating clues 50 Marathe K, Lu J, Morel KD. Bullous diseases: Kids are not just little
from the bench and bedside. Nat Clin Pr Rheumatol 2009;5:139–48. people. Clin Dermatol 2015;33:644–56.
29 Kim KR, Yoon T. A case of neonatal lupus erythematosus showing 51 Al‐Mutairi N, Sharma AK, Zaki A et al. Maternal and neonatal pem-
transient anemia and hepatitis. Ann Dermatol 2009;21:315–18. phigoid gestationis. Clin Exp Dermatol 2004;29:202–4.
30 Prendiville JS, Cabral DA, Poskitt KJ et al. Central nervous system 52 Erickson NI, Ellis RL. Images in clinical medicine. Neonatal rash due
involvement in neonatal lupus erythematosus. Pediatr Dermatol to herpes gestationis. N Engl J Med 2002;347:660.
2003;20:60–7. 53 Krusinski P, Saurat J. Transplacentally transferred dermatosis. Pediatr
31 Rider LG, Sherry DD, Glass ST. Neonatal lupus erythematosus Dermatol 1989;6:166–77.
simulating transient myasthenia gravis at presentation. J Pediatr 54 Parlowsky T, Welzel J, Amagai M et al. Neonatal pemphigus vulgaris:
1991;118:417–19. IgG4 autoantibodies to desmoglein 3 induce skin blisters in new-
32 Askanase AD, Izmirly PM, Katholi M et  al. Frequency of neuro‐ borns. J Am Acad Dermatol 2003;48:623–5.
psychiatric dysfunction in anti‐SSA/SSB exposed children with and 55 Bonifazi E, Milioto M, Trashlieva V et  al. Neonatal pemphigus vul-
without neonatal lupus. Lupus 2010;19:300–6. garis passively transmitted from a clinically asymptomatic mother.
33 Kozlowski K, Basel D, Beighton P. Chondrodysplasia punctata in sib- J Am Acad Dermatol 2006;55(5 Suppl):113–14.
lings and maternal lupus erythematosus. Clin Genet 2004;66:545–9. 56 Hirsch R, Anderson J, Weinberg JM et al. Neonatal pemphigus folia-
34 Shanske AL, Bernstein L, Herzog R. Chondrodysplasia punctata ceus. J Am Acad Dermatol 2003;49:187–9.
and maternal autoimmune disease: a new case and review of the 57 Wu H, Wang ZH, Yan A et al. Protection against pemphigus folia-
literature. Pediatrics 2007;120:e436–41. ceus by desmoglein 3 in neonates. N Engl J Med 2000;343:31–5.
35 Morton RL, Moore C, Coventry S et  al. Pulmonary capillaritis and 58 Alexander A, Samlowski WE, Grossman D et al. Metastatic melanoma
hemorrhage in neonatal lupus erythematosus. J Clin Rheumatol in pregnancy: Risk of transplacental metastases in the infant. J Clin
2004;10:130–3. Oncol 2003;21:2179–86.
36 Rheault MN. Nephrotic and nephritic syndrome in the newborn. Clin 59 Valenzano Menada M, Moioli M, Garaventa A et  al. Spontaneous
Perinatol 2014;41:605–18. regression of transplacental metastases from maternal melanoma in a
37 Mondal R, Nandi M, Sarkar S MK. Neonatal lupus erythematosus newborn: case report and review of the literature. Melanoma Res
associated with unilateral pectoralis major atrophy. Indian J Pediatr 2010;20:443–9.
2011;78:1415–16. 60 Ferreira CM, Maceira JM, Coelho JM. Melanoma and pregnancy with
38 Boh EE. Neonatal lupus erythematosus. Clin Dermatol 2004;22:125–8. placental metastases. Report of a case. Am J Dermatopathol 1998;20:
39 Izmirly PM, Kim MY, Llanos C et  al. Evaluation of the risk of 403–7.
anti‐SSA/Ro‐SSB/La antibody‐associated cardiac manifestations 61 O’Meara AT, Cress R, Xing G et  al. Malignant melanoma in preg-
of neonatal lupus in fetuses of mothers with systemic lupus nancy: A population‐based evaluation. Cancer 2005;103:1217–26.
­erythematosus exposed to hydroxychloroquine. Ann Rheum Dis 62 Alomari AK, Glusac EJ, Choi J et al. Congenital nevi versus meta-
2010;69:1827–30. static melanoma in a newborn to a mother with malignant mela-
40 Pisoni CN, Brucato A, Ruffatti A et al. Failure of intravenous immuno- noma ‐ Diagnosis supported by sex chromosome analysis and
globulin to prevent congenital heart block findings of a multicenter, imaging mass spectrometry. J Cutan Pathol 2015;42:757–64.
prospective, observational study. Arthritis Rheum 2010;62:1147–52. 63 Iskender C, Yasar O, Kaymak O et al. Behçet’s disease and pregnancy:
41 Friedman DM, Llanos C, Izmirly PM et al. Evaluation of fetuses in a a retrospective analysis of course of disease and pregnancy outcome.
study of intravenous immunoglobulin as preventive therapy for con- J Obstet Gynaecol Res 2014;40:1598–602.
genital heart block: Results of a multicenter, prospective, open‐label 64 Antonelou M, Braha N. Transient neonatal Behcet’s disease. BJM Case
clinical trial. Arthritis Rheum 2010;62:1138–46. Rep 2013;2013:bcr2012007589.
42 Martin V, Lee LA, Askanase AD et al. Long‐term followup of children 65 International Study Group for Behcet’s Disease. Criteria for diagnosis
with neonatal lupus and their unnafected siblings. Arthritis Rheum of Behcet’s disease. Lancet 1990;335:1078–80.
2002;46:2377–83. 66 Stark AC, Bhakta B, Chamberlain MA et al. Life‐threatening transient
43 Izmirly PM, Llanos C, Lee LA et al. Cutaneous manifestations of neo- neonatal Behçet’s disease. Br J Rheumatol 1997;36:700–2.
natal lupus and risk of subsequent congenital heart block. Arthritis 67 Noel N, Wechsler B, Nizard J et al. Behçet’s disease and pregnancy.
Rheum 2010;62:1153–7. Arthritis Rheum 2013;65:2450–6.
 101

C HA PTER   9

Developmental Anomalies
Marion Wobser & Henning Hamm

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany

Introduction, 101 Other abnormalities of the breast and Rudimentary (atretic, sequestrated)
Cutis verticis gyrata, 101 nipple, 107 meningocele, 112
Accessory tragi, 103 Developmental abnormalities of the Nasal glioma (nasal cerebral heterotopia), 112
Congenital cartilaginous rests of the neck umbilicus, 107 Congenital inclusion dermoid cysts, 113
(wattles), 103 Median raphe cysts and canals of the Cutaneous signs of occult spinal
Preauricular cysts and sinuses, 104 penis, 108 dysraphism, 114
Branchial cysts, sinuses and fistulae, 104 Infantile perineal (perianal) protrusion, 108 Aplasia cutis congenita (congenital
Congenital midline cervical cleft, 105 Precalcaneal congenital fibrolipomatous absence of skin), 116
Thyroglossal duct cysts, 105 hamartoma, 109 Amniotic constriction band (amnion
Cutaneous bronchogenic cysts, 105 Congenital smooth muscle rupture sequence), 118
Skin dimples, 106 hamartoma, 110 Congenital abnormalities of
Transverse nasal line, 106 Rhabdomyomatous mesenchymal dermatoglyphics, 119
Congenital lip pits, 106 hamartoma, 110
Supernumerary nipples (polythelia), 106 Cutaneous signs of cranial dysraphism, 111

Abstract of nature. However, some developmental abnormalities may be as-


sociated with severe underlying conditions, such as cranial or spi-
nal dysraphism, or complex syndromes, for example branchial arch
A multitude of developmental anomalies may affect the skin.
malformations. Hence, the purpose of the chapter is to enable the
Some of them are easily visible; others are unimpressive or hidden
specialist to differentiate between harmless variations and cutane-
and can therefore remain overlooked for a while. A few anoma-
ous markers of serious developmental failures. This knowledge is
lies, such as the transverse nasal line and precalcaneal congenital
of ultimate clinical importance as the latter may require rapid di-
fibrolipomatous hamartoma, are merely an insignificant variation
agnostic evaluation and treatment to prevent grave consequences.

• Early recognition of conspicuous cutaneous signs may help to


Key points prevent severe complications as is evidenced by spinal dysraphism.
• Cutaneous malformations often demand an interdisciplinary
• Developmental anomalies of the skin may be associated with diagnostic and therapeutic approach involving radiologists,
further clinical findings within a syndromal context. pathologists and surgeons.

­Introduction of the brain. It has an estimated prevalence of 1 in 100 000


of the male and 0.026 in 100 000 of the female general pop-
Developmental anomalies of the skin comprise a rela- ulation. In psychiatric patients and mentally handicapped
tively large group of conditions that are caused by errors persons the condition is far more frequent with a reported
in embryonic development [1]. Fig. 9.1 demonstrates the prevalence of 0.2–12.5%.
typical site of some important anomalies on the head and
neck. Most of them are apparent at birth. This chapter Aetiology. CVG is not a disease entity but originates
focuses on characteristic minor abnormalities. Major from a multitude of diverse aetiologies. In primary (‘true’)
defects forming part of more complex malformation types underlying soft‐tissue abnormalities are lacking,
syndromes are mentioned in connection with particular apart from hypertrophy of connective tissue and adnexal
cutaneous features but are not presented in detail. structures [2]. The alteration may occur as an independ-
ent condition (primary essential CVG) or in association
­Cutis verticis gyrata with a wide range of neurological and psychiatric disor-
ders, such as mental retardation, developmental delay,
Definition. Cutis verticis gyrata (CVG) is a descriptive cerebral palsy, epilepsy, microcephaly and schizophrenia
term that refers to the clinical appearance of ridges and (primary nonessential CVG) with or without ophthalmo-
furrows on the scalp resembling the convoluted surface logical changes (cataract, strabismus, blindness, retinitis

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
102 Section 2  Skin Disorders of the Neonate and Young Infant

10

9 Predilection sites of
1 Accessory tragus
2 Preauricular sinus

3 4 3 Dermoid cyst
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

2 4 Nasal glioma
5
1 5 Transverse nasal line
6 Congenital lip pits
6 7 Congenital midline cervical cleft
8 Branchial cysts/sinus/fistulae
9 Aplasia cutis congenita
7 10 Cutis verticis gyrata
8
Fig. 9.1  A schematic diagram showing various sites of
developmental abnormalities over the face and neck.

pigmentosa) [3]. There is a strong association between dermal oedema, elastic fibre degeneration and hypertro-
CVG and chromosomal abnormalities and genetic phy of sebaceous glands are present within the context
syndromes (Turner syndrome, Klinefelter syndrome, of pachydermoperiostosis, possibly as a result of prosta-
Noonan syndrome, fragile chromosome syndromes, glandin‐mediated tissue effects due to mutations in pros-
Beare–Stevenson cutis gyrata syndrome, Ehlers–Danlos taglandin‐catalysing enzymes [5]. Histological changes in
syndrome, tuberous sclerosis, insulin resistance syndrome, secondary types depend on the underlying condition.
Apert syndrome and primary hypertrophic osteoar-
thropathy/pachydermoperiostosis) [4]. CVG in Turner
Clinical features. Primary CVG usually presents with
and Noonan syndrome is believed to result from regress-
symmetrical folds that run from anterior to posterior and
ing lymphoedema present in utero.
involve the vertex and occiput, only occasionally the
Secondary types (‘pseudo‐CVG’) result from local
entire scalp. Male patients are affected considerably more
inflammation (eczema, psoriasis, folliculitis, impetigo,
often with a ratio of 4–5 : 1. Onset is usually postpubertal
erysipelas, pemphigus, acne keloidalis, acne inversa),
with the vast majority developing after the third decade
neoplastic processes and hamartomas of the scalp (con-
of life. Congenital occurrence of primary CVG is extremely
genital or acquired intradermal melanocytic naevus,
rare [6]. If an underlying genetic abnormality is suspected,
sebaceous naevus, naevus lipomatosus, connective tissue
molecular genetic or cytogenetic studies are advised.
naevus, neurofibroma, fibroma, cylindroma) or from
In secondary types the clinical appearance is much
systemic illnesses (acromegaly, myxoedema, acanthosis
more variable, and the folds usually do not run in a
nigricans, diabetes mellitus, misuse of anabolic substances,
sagittal direction. Systemic and inflammatory conditions
amyloidosis, syphilis, chronic pulmonary diseases, con-
may present with mild corrugation of the entire scalp
genital cyanotic heart disease, hepatobiliary disorders,
whereas local neoplasms may produce irregular, cerebri-
leukaemia, paraneoplastic syndromes) or certain drugs.
form folding in a circumscribed area. Secondary types
occur at any age, reflecting the manifold causes. Cranial
Pathology. Histopathological examination of a scalp biopsy computed tomography (CT) or magnetic resonance
is mandatory to distinguish between primary and sec- imaging (MRI) may be indicated to rule out a pituitary
ondary types, especially to exclude neoplastic causes. adenoma and acromegaly or involvement of deeper
Findings in primary types range from essentially normal structures. Congenital secondary CVG is mainly caused
skin to thickened connective tissue with hypertrophy or by benign neoplasms such as an intradermal melano-
hyperplasia of adnexal structures. Mucin deposition, cytic naevus (see Table 9.1).

Table 9.1  Differences between primary and secondary cutis verticis gyrata

Primary cutis verticis gyrata Secondary cutis verticis gyrata

Clinical features Furrows and ridges in sagittal orientation Irregular furrows and ridges
Associated conditions • Genetic defects including chromosomal aberrations • Local inflammatory diseases
• Neurological/psychiatric disorders • Local hamartomas/neoplasias
• Sporadic • Systemic illnesses
Pathology Hypertrophy of connective tissue and adnexal structures Variable, depending on aetiology
Time of onset Second decade of life to adulthood, rarely congenital Variable, depending on aetiology
Sex Male predominance Equal sex distribution
Chapter 9  Developmental Anomalies 103

CVG is asymptomatic in most cases but may cause pru-


ritus, burning, tenderness, unpleasant odour or recurrent
infection. The texture and amount of hair are usually
unaffected in primary types whereas neoplastic lesions
may show progressive alopecia.

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Treatment. Good skin and scalp hygiene with optional
application of topical antiseptics to deep furrows is rec-
ommended to avoid intertrigo and bacterial or mycotic
infection. Many conservative treatments have proved
ineffective in flattening the uneven surface. In spite of the
benign nature of CVG, elective surgery may be indicated
for psychological or aesthetic reasons. Possible interven-
tions include excision with primary closure or local flaps
in small lesions or partial resection, serial excision or tissue
expansion procedures for larger areas. In CVG within
the context of pachydermoperiostosis, inhibition of the
underlying pathogenetically relevant prostaglandin syn-
thesis may show benefit if carried out in early childhood.

­Accessory tragi

Aetiology. Accessory tragi are minor developmental


abnormalities of the dorsal portion of the first (mandibu-
lar) branchial arch, which explains their location between
the pretragal and sternoclavicular areas.
Fig. 9.2  Accessory tragi in typical preauricular location.

Pathology. There is histological similarity to the normal


radial dysplasia and other limb defects) syndromes [8].
anatomy of the external ear. Numerous mature vellus hair
Multiple accessory tragi are a relatively constant finding
follicles are present in various phases of the hair cycle.
in the oculo‐auriculo‐vertebral (Goldenhar) syndrome
A prominent connective tissue framework encloses aggre-
featuring ear malformations, hemifacial microsomia, epi-
gates of mature adipose tissue. A central core of cartilage is
bulbar (lipo)dermoids and vertebral anomalies.
occasionally present, helping to distinguish an accessory
The accessory tragus should be differentiated from a
tragus from hair follicle naevus and trichofolliculoma.
macrotragus which shares its histopathological charac-
teristics but represents an enlarged, mostly anteriorly
Clinical features. Accessory tragi present as congenital displaced tragus in a normal anatomical position.
roundish, skin‐coloured papules or nodules, usually
located in front of the auricle (Fig. 9.2). Less common sites Treatment. Accessory tragi can easily be treated by surgical
include the cheek along the line of the mandible to the excision. Pedunculated papules may even be removed by
angle of the mouth, and the lateral neck anterior to the ligation or application of a titanium clip flush to the skin.
sternocleidomastoid muscle down to the suprasternal Care must be taken to remove any protuberant portion of
region. In a cervical location, they may be called wattles the underlying cartilage. Some authors advocate treat-
or congenital cartilaginous rests of the neck (see ment under local anaesthesia in the first days of life.
Congenital cartilaginous rests of the neck (wattles)).
The skin tags may be solitary or multiple, unilateral or
­ ongenital cartilaginous rests
C
bilateral, soft or cartilaginous, sessile or pedunculated.
of the neck (wattles)
They are often covered by vellus hairs. The incidence is
approximately 1 in 500–600 live births. Congenital cartilaginous rests of the neck (wattles) are
Usually, accessory tragi are isolated congenital defects rare, uni‐ or bilateral branchiogenic surplus malforma-
[7]. Familial occurrence has been reported. In rare tions that may be considered the cervical variant of acces-
instances, especially if multiple, they are associated with sory tragi [9]. An irregular, often pedunculated, papule or
other first branchial arch abnormalities, including cleft nodule of ‘springy’, cartilaginous texture is located over
lip, cleft palate, branchial fistulae and hypoplasia of the or near the lower half of the sternocleidomastoid muscle
mandible, and are an inconstant feature of the Treacher– (Fig. 9.3). Other malformations, such as microtia, stenosis
Collins, Wolf–Hirschhorn (4p deletion), Townes–Brocks, of the external ear canal and branchiogenic fistulae, are
Delleman and VACTERL (vertebral anomalies, anal atre- rarely associated. Differential diagnosis includes sentinel
sia, congenital cardiac anomalies, tracheo‐oesophageal tags associated with branchial sinuses or fistulae, simple
fistula and/or oesophageal atresia, renal anomalies, skin tags and benign papillomas. As congenital cartilaginous
104 Section 2  Skin Disorders of the Neonate and Young Infant
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Fig. 9.3  Bilateral congenital cartilaginous rests of the neck (‘wattles’). Fig. 9.4  Inflamed preauricular cyst and sinus.

homologue of the Drosophila eye absent (EYA1) gene.


rests of the neck are not cystic or fistulous, they can easily
Therefore, auditory testing and renal ultrasound are
be excised for cosmetic reasons.
regarded as imperative if a preauricular sinus is accompa-
nied by one or more of the following: other malformations
­Preauricular cysts and sinuses or dysmorphic features, a family history of deafness,
auricular and/or renal malformations or a maternal history
Aetiology. Preauricular cysts and sinuses are believed to of gestational diabetes.
be the result of imperfect fusion of the sixth auditory In most cases preauricular sinuses remain asymptomatic,
hillocks of the first two branchial arches. Global preva- at least until adulthood. However, the seemingly trivial
lence seems to differ considerably (India, 0.17%; USA, and often unnoticed anomaly can give rise to troublesome
0.1–0.9%; England, 0.9%; China, 1.4%; Taiwan 1.6–2.5%; symptoms if infection occurs (Fig. 9.4). In this case, recur-
some areas of Africa, 4–10%) [10,11]. rent purulent discharge and preauricular abscesses causing
oedema, erythema and pain may develop.
Pathology. The cysts and sinuses are lined by stratified Ultrasound of underlying tissue is helpful to confirm
squamous epithelium. The adjacent connective tissue fre- the diagnosis and to determine the extent of the sinus and
quently contains skin appendages. After severe inflam- cysts and their relation to adjacent anatomical structures.
mation, the lining is often replaced by granulation tissue.
Treatment. If asymptomatic, a preauricular sinus does not
Clinical features. Preauricular sinuses usually present as require intervention. Cure of symptomatic sinuses is only
tiny pits or indentations at the anterior margin of the achieved by often difficult, meticulous complete excision
ascending limb of the ear helix (Fig. 9.4). Much less com- of the entire tract and its associated cysts. Thus, most
mon sites are the lateral surface of the helicine crus and authors recommend surgery only after preoperative anti-
the superior posterior margin of the helix, tragus or lob- biotic treatment. Nevertheless, postoperative recurrences
ule [12]. The sinuses may be unilateral, more frequently are reported to occur in 9–42% of cases. General anaesthe-
on the right side, or bilateral; they can be sporadic or sia, magnification employed during surgery, wide local
inherited. Bilateral occurrence is more likely to be heredi- excision with removal of cartilage at the base of the tract,
tary and transmitted as an autosomal dominant trait with demonstrating the sinus with dye injection, and probing
reduced penetrance and variable expression. The narrow at the time of surgery help to reduce the recurrence risk.
sinuses have a cutaneous opening and a blind inner end,
almost always connecting to the perichondrium of the
auricular cartilage. They may arborize, with a subcutane- ­Branchial cysts, sinuses and fistulae
ous network of cysts arranged around their tracts, and
rarely extend into the parotid gland. Aetiology. Branchial cysts, sinuses and fistulae result from
In 3–10% of cases, they are associated with deafness maldevelopment of the branchial apparatus, most often
and with several complex developmental defects, pre- the second branchial arch, during embryonic develop-
dominantly branchio‐oto‐renal (BOR) syndrome and ment. Familial occurrence has repeatedly been reported.
branchio‐otic (BO) syndrome. BOR and BO syndromes
are autosomal dominant conditions characterized by Pathology. The sinus tracts of branchial anomalies are
preauricular pits, conductive, sensorineural or mixed lined by stratified squamous epithelium, which may in
hearing loss, structural defects of the outer, middle or part be replaced by respiratory (ciliated columnar) epi-
inner ear, lateral cervical fistulae, cysts or sinuses, and thelium. There is often abundant subepithelial lymphoid
nasolacrimal duct stenosis or fistulae. Renal anomalies tissue. Thymic or thyroid tissue points to derivation from
are additional features of BOR syndrome [12]. Both syn- the third or fourth branchial arch, respectively. The pres-
dromes are frequently caused by mutations in the human ence of smooth muscle, cartilage and seromucous glands
Chapter 9  Developmental Anomalies 105

in branchial arch remnants may lead to confusion with a ­Thyroglossal duct cysts
bronchogenic cyst (see Cutaneous bronchogenic cysts).
Thyroglossal duct cysts develop as a result of mucus pro-
Clinical features. External branchial sinuses and fistulae duction within an incompletely obliterated thyroglossal
are detectable as openings located on the lateral neck, at duct [16]. Most often, these cysts lie close to the hyoid
the anterior border of the sternocleidomastoid muscle. bone in or near the midline of the neck, but they may be
located at any site along the pathway of the thyroid

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Sinuses end blindly in the deep neck tissue, while fis-
tulae have a continuous communication between skin anlage. They are the commonest cause of midline neck
and mucosa and may drain into the tonsillar area of the swellings in childhood and adolescence although a signifi-
pharynx. Cysts present as painless, mobile swellings and cant minority of cysts are not diagnosed before adulthood.
have neither a cutaneous nor a mucosal opening. The Typically, a painless soft mass of 1–2 cm diameter moves
external orifice of sinuses and fistulae may exude a upwards on swallowing and on protrusion of the tongue.
mucous secretion. Early secondary infection is frequent Less often, there may be a discharging sinus occurring
and may cause significant morbidity. Because of their spontaneously or after incomplete excision. Infection
obvious skin opening and associated drainage and infec- occurs with increasing frequency with age. To avoid infec-
tion, branchial sinuses and fistulae are usually diagnosed tious complications and malignant change, thyroglossal
much earlier than branchial cysts, most often in the first duct cysts should be excised early in life by resection of
few years of life [13]. the mid‐portion of the hyoid bone and en bloc cystectomy
Bilateral lesions are rare; preauricular pits may be an with a cuff of muscle around the duct to the base of the
associated finding. Branchial anomalies may form part of tongue (Sistrunk’s operation) [17]. Postoperative recur-
complex hereditary conditions, such as BOR syndrome, rence rates are in the range of 3–4%. Prior to surgery, the
branchio‐oculo‐facial syndrome and others. normal thyroid gland should be identified by appropriate
Diagnosis of branchial sinuses and fistulae is obvious imaging. On ultrasound, the cysts themselves show an
in light of the cutaneous opening in a typical location. unechoic, hypoechoic, pseudosolid or heterogeneous
In contrast, branchial cysts are part of the extensive pattern. CT, MRI and radioisotope thyroid scanning offer
differential diagnosis of lateral neck masses including more precise information about location, size and relation
lymphadenitis, abscess, lipoma, cystic hygroma, hae- to adjacent structures and together with fine‐needle
mangioma, lymphangioma, lymphoma, ectopic thyroid aspiration help to exclude differential diagnoses such
cyst, thyroglossal duct cyst and cervical thymic cyst [14]. as ectopic thyroid tissue, lipoma, lymphadenopathy
Radiographic imaging may be indicated to establish the and carcinoma. The ducts are frequently multiple and
diagnosis and assess the extension. branched. There are numerous cases reporting malig-
nancy arising in thyroglossal duct remnants with thyroid
Treatment. Due to the lack of spontaneous regression of
papillary carcinoma being by far the most common type
branchial anomalies and common recurrent infections, of associated neoplasm.
complete surgical removal under general anaesthesia
during the first year of life is the treatment of choice. ­Cutaneous bronchogenic cysts
The precise aetiology of cutaneous bronchogenic cysts
­Congenital midline cervical cleft
is disputed. The more common cysts inside the thoracic
This rare congenital anomaly, most likely related to aber- cage are believed to originate from accessory buds that
rant fusion of the first or second branchial arches, is became sequestrated from the primitive tracheobron-
located in the anterior midline of the neck at any point chial tree or primitive foregut. This concept also fits for
between the mandible and the sternum [15]. The charac- presternal cysts that may develop after the intervention
teristic clinical presentation consists of a cephalic nipple‐ of fusing sternal bars, but it is unlikely for bronchogenic
like projection (‘skin tag’) with the inferior margin being cysts in ectopic locations such as the shoulder and scap-
formed by a short sinus of about 1 cm length and an ular regions. Transplantation via lymphatic or haematog-
atrophic mucosal surface dividing the two. An underly- enous routes, metaplasia and maldifferentiation of
ing fibrous cord may impair the movement of the head epithelial components to ciliated epithelium are other
and neck and cause webbing. The cleft is covered by possibilities.
stratified squamous epithelium lacking skin append- In most cases, the lesion becomes evident within the
ages while the sinus tract is usually lined by pseu- first days of life as a swelling or as discharge on the lower
dostratified columnar epithelium and often contains neck, the anterior upper chest near the suprasternal notch
seromucinous salivary glands. The abnormality may be or over the manubrium sterni. Chin, shoulder, scapular
isolated or associated with a broad spectrum of midline region, back and abdomen are less common sites [18].
defects, such as median cleft of the mandible, tongue There is no connection to underlying structures. Males are
and lower lip. Complete excision of the anomaly with predominantly affected. Histological examination of
the underlying fibrous tract within the first year of life is bronchogenic cysts typically reveals a lining of ciliated
critical to avoid scarring contracture and mandibular and mucin‐producing pseudostratified columnar epithe-
deformities. The surgical defect is usually closed with lium of respiratory type. Excision is recommended to pre-
multiple Z‐plasties. vent infection and malignant transformation.
106 Section 2  Skin Disorders of the Neonate and Young Infant

­Skin dimples within the oral cavity at the angle of the mouth and rep-
resent the openings of bilateral, short and blind‐ending
Skin dimples are unilateral or more often bilateral deep sinuses. They are frequently inherited as an isolated
cutaneous depressions commonly seen over bony autosomal dominant anomaly, but may be a feature of
prominences, such as the acromial process of the scapula, the BOR syndrome, associated with branchial, hearing
os sacrum, elbow, patella and tibia [19]. They are thought and renal anomalies. Furthermore, commissural lip
to be caused by early entrapment‐induced fixation of the
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

pits have been described in association with alveolar


skin to the underlying bony prominence, thus preventing synechia, ankyloblepharon filiforme adnatum and
the development of subcutaneous tissue. ectodermal defects.
Usually, skin dimples are an isolated congenital finding In midline sinus of the upper lip, the opening of a
and, as in the case of bilateral acromial or shoulder blind‐ending sinus tract is seen in the midline of the
dimpling, also known as ‘supraspinous fossae’, may upper‐lip philtrum. Hypertelorism, nasal fistula and lip
represent a harmless autosomal dominant anomaly. fistula may be associated.
However, bilateral acromial dimples have also been Congenital lower‐lip pits result from a fusion defect of
reported as a relatively constant feature of the 18q dele- the lower part of the first branchial arch. They usually
tion syndrome, and as an occasional finding in trisomy 9p present as bilateral, symmetrical depressions on the ver-
syndrome, Russell–Silver dwarfism and Say syndrome. milion portion of the lower lip on each side of the midline.
An association of subacromial dimples with recurrent The orifices represent the endpoints of blind sinuses that
posterior dislocation of the shoulder has been postulated. extend inwards into the orbicularis oris muscle to a depth
Dimples overlying the tibia are seen in camptomelic of up to 20 mm or more and may bifurcate. The tracts are
dwarfism, oral–facial–digital syndrome and osteoglo- lined by stratified squamous epithelium. If there is a com-
phonic dysplasia. Dimples over elbows and knees have munication with the ducts of the nearby minor salivary
been observed in prune belly syndrome, Joubert syndrome glands, saliva or mucus may be discharged. Occasionally,
and facial clefting syndrome. Midline sacral dimples only a single paramedian or median sinus may be pre-
are an inconstant minor feature of various congenital sent. Labial conical elevations without fistulous ostia are
malformation syndromes including spinal dysraphism, believed to represent the microform of the anomaly.
especially when they are large, more than 2.5 cm away Congenital lower‐lip pits are the hallmark of Van der
from the anus or combined with other lesions. Sporadic Woude syndrome, an autosomal dominant craniofacial
cases of skin dimpling have been described in association disorder with high penetrance and variable expressivity
with congenital rubella, hypophosphatasia and misoprostol characterized by lower‐lip pits, cleft lip/palate and
exposure in utero. Dimple‐like scars are not uncommon other inconstant features such as hypodontia or diverse
after amniocentesis. malformations [24]. They are also a characteristic
feature of the popliteal pterygium syndrome (popliteal
­Transverse nasal line webs, cleft lip and/or cleft palate and anomalies of the
genitourinary system). Van der Woude syndrome and
The transverse nasal line runs transversely across the skin popliteal pterygium syndrome are allelic conditions
between the upper two‐thirds and lower third of the caused by diverse mutations of the gene encoding the
nose. The clinical appearance varies from a faint reddish interferon regulatory factor 6 (IRF6) [25]. Therefore,
to hyperpigmented line to a more visible groove or crease genetic counselling has to respect the considerable
with a depth and width of up to several millimetres [20]. phenotypic variation and overlap of the two conditions.
It appears early in life and often remains indefinitely. Furthermore, lower‐lip pits are a facultative sign of the
There is a clear familial predisposition, and females seem orofaciodigital syndrome type 1 and the Kabuki make‐
to be affected more often. up syndrome.
Distinct growth kinetics of the alar and triangular The only treatment for symptomatic or unpleasant lip
cartilages of the nose are presumed to be pathogeneti- pits is surgical excision, ensuring complete removal of the
cally relevant. The transverse nasal line may be associ- often bifurcating tracts.
ated with milia, cysts and comedones. Inflammation may
lead to ‘pseudoacne of the nasal crease’ [21].
The transverse nasal line has to be distinguished from ­Supernumerary nipples (polythelia)
the nasal crease or wrinkle developing in patients with With a prevalence of 0.2–5.6%, supernumerary nipples
allergic rhinitis as a response to frequently rubbing the (polythelia) are the most frequently encountered congeni-
nose upward for relief of itching (the ‘allergic salute’) [22]. tal abnormality of the breast. They are remnants of the
embryological milk line which runs from the anterior
axillary fold to the inner thigh. Most often, they are
­Congenital lip pits
located on the chest wall and upper abdomen and present
Congenital lip pits can be divided into three types: com- as small brown or pink, umbilicated or elevated papules
missural pits, midline sinuses of the upper lip, and lower surrounded by a pigmented areola (Fig. 9.5) [26]. A nipple
lip pits (lip sinuses) [23]. may also be seen without an areola, and vice versa.
Commissural pits are the commonest type of lip pits, Usually there is only a single lesion, but multiple and
found in about 0.5–2% of neonates. They are located bilateral nipples are possible.
Chapter 9  Developmental Anomalies 107

include ectodermal dysplasia, Al Awadi–Raas‐Rothschild


syndrome (absence or severe hypoplasia of skeletal parts
of the limbs), Poland syndrome choanal atresia–athelia
syndrome, scalp–ear–nipple syndrome, and ADULT
(acro‐dermato‐ungual‐lacrimal‐tooth) syndrome [28].
Athelia is believed to be caused by a failure in parathy-

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
roid hormone‐related protein production.
Unilateral absence (amastia) or hypoplasia (hypomastia)
of the breast and nipple is most often a feature of Poland
syndrome. This congenital anomaly consists of a spec-
trum of congenital deformities of the chest wall, breast
and upper extremity. Unilateral absence of the sternocos-
tal head of the pectoralis major muscle is the indispensa-
ble prerequisite for diagnosis. In addition, amastia or
mild hypoplasia of the breast, athelia or abnormal nipple,
scarcity of subcutaneous tissue over the pectoral region,
absence of the pectoralis minor muscle, deficiency of
additional chest wall muscles, aplasia/deformity of costal
cartilages or anterior ribs II–V, alopecia of axillary and
mammary regions, and ipsilateral hand anomalies (brach-
ysyndactyly, syndactyly, hypoplasia of middle phalan-
ges) may be present. Less frequent associated defects
include renal malformations, dextrocardia and vertebral
abnormalities. Poland syndrome is thought to be caused
Fig. 9.5  Supernumerary nipple in a male adolescent. by interruption of the embryonic blood supply to the
upper limb bud in the sixth week of gestation with
Familial occurrence without further anomalies suggest- consecutive hypoplasia of the subclavian artery. It occurs
ing autosomal dominant transmission has been observed. in about 1 out of 25 000 live births, mostly sporadically
Polythelia may present within a syndromal context, such and mainly in males. Evaluation of the vascular status is
as part of the Becker naevus syndrome, the X‐linked mandatory prior to reconstructive breast surgery.
Simpson–Golabi–Behmel syndrome or in association Ipsilateral hypoplasia of the breast, areola and/or
with malformations of the urinary tract or with haemato- nipple is the most frequent anomaly in Becker naevus
logical disorders. syndrome. Other associated features include supernumer-
Accessory nipples are occasionally mistaken for mel- ary nipples, scoliosis, vertebral anomalies and shortening
anocytic naevi [27]. They should also be differentiated or other forms of asymmetry of the upper limbs due to
from unilateral or bilateral intra‐areolar polythelia with hypoplastic soft tissue [29]. Hypomastia has also been
two or more nipples located within the areola, caused observed in conjunction with naevus depigmentosus and
by intrauterine division (dichotomy) of the developing phacomatosis cesioflammea in individual cases.
nipple. Excision may be indicated for diagnostic or While functional deficits of the breast and nipple
cosmetic reasons. deformities may need early intervention, final aesthetic
surgery should be postponed until after puberty to obtain
an optimal cosmetic outcome respecting the symmetry of
­ ther abnormalities of the breast
O
the chest anatomy.
and nipple
Accessory breast tissue (polymastia) may underlie a super-
numerary nipple and usually becomes evident only after
­Developmental abnormalities
hormonal stimulation at puberty or in pregnancy.
of the umbilicus
Supernumerary breasts are located along the primitive A variety of congenital umbilical lesions result from
milk line with the axilla being a particularly common site complete or partial failure of obliteration of two embryo-
where they can be mistaken for lymphadenopathy or logical structures: the omphalomesenteric (vitelline) duct
lipoma. Comparable to polythelia, polymastia can be and the urachus. The clinical features depend upon the
associated with other, especially renal, anomalies. Fine‐ site and extent of patency of these structures [30].
needle aspiration may serve as a useful tool for diagnosis. A completely patent omphalomesenteric duct is exceed-
Complete excision of ectopic breast tissue is recom- ingly rare and leads to a fistula between the ileum and
mended due to the risk of malignant transformation [27]. umbilicus. The anomaly is noted soon after birth by faecal
Athelia is defined as congenital complete absence of the discharge severely irritating the adjacent skin. Infections
nipple–areola complex and may be unilateral or bilateral. and sudden ileal prolapse through the umbilicus may
The rare anomaly is almost always combined with complicate the anomaly; the latter represents a surgical
amastia (absence of the breast and nipple) within the emergency because of the danger of strangulation and
frame of syndromic developmental disorders. These necrosis of the affected bowel.
108 Section 2  Skin Disorders of the Neonate and Young Infant

A partially patent omphalomesenteric duct gives rise


to an umbilical polyp of bright‐red colour (remnant
of the peripheral portion), an umbilical sinus (patency of
the peripheral portion) or a vitelline cyst (patency
of  the intermediate portion). The last is usually
asymptomatic whereas polyps and sinuses mostly dis-
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

charge serous or mucinous fluid shortly after birth.


This distinguishes the umbilical polyp from the most
important differential diagnosis, granuloma pyogeni-
cum, which usually has no secretion. Polyps and cysts
are frequently accompanied by potentially serious
internal omphalomesenteric remnants, such as Meckel’s
diverticulum attached to the umbilicus by obstructing
fibrous bands.
A completely patent urachus leads to fistula between
the urinary bladder and umbilicus. It presents soon after
birth by leakage of urine from the abnormal‐appearing Fig. 9.6  Median raphe cyst.
umbilicus, which may cause irritation of periumbilical
skin. An umbilical tumour that may be confused with
pyogenic granuloma and infection of the genitourinary ­ edian raphe cysts and canals
M
tract may occur. of the penis
A partially patent urachus may result in an umbilical Median raphe cysts and their more elongated variants,
urachal sinus (patency of the peripheral portion) or a ura- called raphe canals, are located in a midline position
chal cyst (patency of the intermediate portion). Unless along the genitoperineal raphe anywhere from the ure-
large or infected, they present as asymptomatic cystic thral meatus to the anus but most commonly on the distal
swellings. Adenocarcinoma of the urachus may rarely portion of the penis (Fig. 9.6) [32]. They present as well‐
occur in later life and has a poor prognosis. circumscribed, freely mobile, solitary or multiple cystic
The diagnosis of developmental anomalies can be veri- lesions. On rare occasions, they may be pigmented or
fied by radiographic imaging, especially ultrasound and appear as a perianal polyp. Histologically, they are lined
sinography, and histological demonstration of ectopic with pseudostratified columnar or less often stratified
gastrointestinal or bladder transitional epithelium cover- squamous epithelium, mimicking the epithelial lining of
ing polyps or lining sinuses and cysts. the male urethra and suggesting derivation from urethral
Umbilical enteric and urinary fistulae are absolute or genital folds [33]. Mucous glands may be present.
indications for surgical intervention [31]. Simple exci- Most median raphe cysts and canals remain asympto-
sion of umbilical polyps should be performed only if matic. Medical attention is sought in the first three
underlying intestinal and urinary tract anomalies are decades of life for secondary infections of staphylococcal
ruled out. More often, the lesion has to be completely or gonococcal origin, injury or discomfort upon inter-
excised together with the attached part of the bladder course, or for cosmetic reasons. Simple excision with
or bowel. Total resection of the umbilicus is usually not primary closure is effective because there is no communi-
required. cation with the urethra.
Umbilical anomalies may also form part of complex
malformation syndromes. Redundant periumbilical skin
extending along the cord for an excessive distance is a ­Infantile perineal (perianal) protrusion
characteristic feature of Rieger syndrome, an autosomal
dominant disorder of morphogenesis. Recognition of Definition. Infantile perineal protrusion (IPP) is a harm-
the umbilical stump abnormality enables early diagno- less, often congenital, soft‐tissue lesion on the perineal
sis of associated glaucoma due to goniodysgenesis median raphe of prepubertal children, mostly girls [34].
(abnormal development of the anterior chamber angle of
the eye) and may prevent blindness. Lozenge‐shaped Aetiology. Classification into three different types has
diastasis around the umbilicus and umbilical depression been proposed. The constitutional type is considered to
occur within the scope of the autosomal recessive so‐ result from constitutional weakness of the median raphe
called 3MC syndrome (Malpuech–Michels–Mingarelli– or perineum of females or a remnant of a projected tip of
Carnevale syndrome). Umbilical hernia and abnormal the urogenital septum; it may be familial and/or congeni-
umbilicus are signs of the autosomal dominant Robinow tal. The functional type is related to bowel irregularities
syndrome. Ventral developmental defects, such as (diarrhoea, constipation) or other irritant exposure. A
sternal clefting or supraumbilical abdominal raphe, may third type is associated with genitoanal lichen sclerosus.
be associated with posterior fossa brain malformations,
large or complex haemangiomas of the face, arterial Pathology. The epidermis and dermis are essentially
anomalies, cardiac anomalies and eye abnormalities normal. Inflamed lesions may show acanthosis, vacuo-
(PHACES association). lar changes in basal keratinocytes, thickening and/or
Chapter 9  Developmental Anomalies 109

­Precalcaneal congenital
fibrolipomatous hamartoma

Definition. Precalcaneal congenital fibrolipomatous


hamartoma (PCFH) is a harmless nodular anomaly on
the medial plantar aspects of infants’ feet. It has been

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
described under various terms, including bilateral con-
genital adipose plantar nodules, benign anteromedial
plantar nodules of childhood, bilateral congenital fatty
heel pads, pedal papules in the newborn, congenital pie-
zogenic‐like pedal papules, hypertrophic infantile pedal
papules and congenital fibrolipomata.

Aetiology. The exact aetiology is unknown. PCFH has


been speculated to result from hamartomatous formation
of fat and connective tissue, secondary to incomplete
involution of the physiologically abundant fetal subcu-
taneous plantar tissue, or from herniation of fatty tissue
through congenital defects in the plantar fascia and the
trabecular fibroconnective system of the sole [36]. Rare
familial observations favour a genetic cause.
Fig. 9.7  Infantile perineal protrusion. Although PCFH is considered to be uncommon, a
systematic investigation found an incidence of 5.9% in
newborns and of 39.4% in infants [37]. Underreporting
elongation of the rete ridges, upper dermal oedema,
may be attributed to the small size and the lack of symp-
dilatation of capillary and lymph vessels and mild infil-
toms in most cases.
tration of neutrophils and eosinophils. Histological
signs of lichen sclerosus may be associated.
Pathology. Histological examination reveals lobulated,
Clinical features. The anomaly presents as a single, soft, mature adipose tissue protruding into the reticular
skin‐coloured or reddish papule or nodule with a smooth ­dermis, enveloped by fibrous septa. Blood vessels may
or slightly velvety surface and resembles an outward pro- be prominent. Mucinous deposits have been described
jection of essentially normal, redundant skin (Fig. 9.7). It at the periphery of the lesion and within fat lobules.
is usually located along the midline of the perineum ante-
rior to the anus, only exceptionally posterior to it or both. Clinical features. PCFH usually presents as soft, flesh‐
IPP is almost exclusively seen in girls [35]. Mechanical coloured, painless nodules of 5–10 mm symmetrically
irritation of wiping after defaecation and constipation located on the medial posterior aspects of the soles, just
may cause swelling, inflammation or fissuring, and anterior to the heels of an infant (Fig. 9.8) [38]. Unilateral
induce pain. Spontaneous regression is common in the occurrence and extension onto the heel area are rare. The
functional type, in contrast to only gradual involution in lesions are present at birth or appear in the first few
the constitutional and lichen sclerosus‐associated types. months of life, grow in proportion to the growth of the
infant and are most prevalent at about 1 year of age. They
Differential diagnosis. Skinfolds, haemorrhoids, acro- tend to disappear at 2–3 years of age, but can persist
chordons, midline malformations, genital warts, sentinel
tag of anal fissure, granulomatous lesions of inflamma-
tory bowel disease, haemangioma and rectal prolapse
may be considered in the differential diagnosis [35].
Misinterpretation as a sign of sexual abuse has to be
avoided.

Treatment. The child’s anal region should be wiped


clean from anterior to posterior to avoid inflammation.
Inflamed lesions respond rapidly to topical corticosteroids
whereas the outcome of the lichen sclerosus‐associated
type is determined by the course of the underlying der-
matosis. A fibre‐rich diet and hydration are recommended
in case of associated constipation. As most lesions are
asymptomatic and tend to regress spontaneously, surgical
excision is rarely required. Fig. 9.8  Precalcaneal congenital fibrolipomatous hamartoma.
110 Section 2  Skin Disorders of the Neonate and Young Infant

considerably longer. They almost never cause functional


Differential diagnosis. The clinical diagnosis is difficult
problems, such as interference with standing and walking.
as specific diagnostic criteria are lacking and clinical
There are no reports of associated malformations.
findings are nonspecific. However, histology allows clari-
fication. CSMH should be suspected in any congenital
Differential diagnosis. In contrast to PCFH, piezogenic hairy lesion, especially in the lumbosacral area. Differential
papules usually develop in adulthood, are multiple and diagnosis includes congenital melanocytic naevus, Becker
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

smaller, and accentuate with standing. Further differen- naevus, connective tissue naevus, ­epidermal naevus, café‐
tial diagnoses include juvenile plantar fibromatosis, au‐lait spot, leiomyoma and mastocytoma.
fibrolipomatous hamartoma of the plantar nerve, lipoma,
naevus lipomatosus, haemangioma, neurofibroma, con-
genital solitary histiocytoma, lymphatic malformation, Treatment. No treatment is necessary.
calcified nodules after repeated heel sticks and focal
dermal hypoplasia [36]. ­Rhabdomyomatous mesenchymal
hamartoma
Treatment. In almost all instances treatment is not
required and parents should be reassured about the harm- Definition. Rhabdomyomatous mesenchymal hamartoma
less finding with high propensity for rapid spontaneous (RMH), originally termed striated muscle hamartoma, is
involution. Surgical excision should be confined to cases a rare congenital lesion of the dermis and soft tissues con-
of persistent tenderness or impairment of walking. sisting of a disordered and varied collection of mature
skeletal muscle and other mesenchymal tissues.
­Congenital smooth muscle hamartoma
Aetiology. The aetiology is unknown. Aberrance in the
Definition and  aetiology. Congenital smooth muscle embryonic migration of mesodermally derived tissues, in
hamartoma (CSMH) originates from smooth muscle particular skeletal muscle, has been suggested. RMH pre-
fibres of the musculi arrectores pilorum. dominantly affects areas of superficially located striated
muscles that are derived from the second branchial arch.
Pathology. Histological examination shows a focal prolif-
eration of large, mature, well‐demarcated bundles of Pathology. The central part of the lesion contains dermal
smooth muscle fibres in the reticular dermis which may and subcutaneous bundles of haphazardly arranged stri-
extend to the subcutaneous tissue. The bundles are ated muscle fibres admixed with varying amounts of fat,
haphazardly arranged and interwoven. fibrous tissue, blood vessels and nerves. It is surrounded
by multiple adnexal elements, such as vellus hair follicles
and sebaceous and eccrine glands.
Clinical features. CSMH is usually present at birth or
noted shortly thereafter with an estimated prevalence of
0.5–1 per 1000–2700 live births. Clinical features. Most often, RMH is noted at birth as a
The most frequent presentation of the congenital variant solitary, sporadic lesion in or near the midline of the head
is a single skin‐coloured or slightly hyperpigmented, or neck, in sites where striated muscles are superficially
indurated plaque, often with prominent vellus hairs. It located. Unusual sites include oral cavity, perianal or
occurs on the trunk, particularly the lumbosacral area, or genital area, and digit. The flesh‐coloured, firm, non-
the extensor surfaces of the thighs, more rarely on arms, tender lesion may present as a papillomatous, polypoid
face or mammary region. Another manifestation is an or pedunculated papule or nodule or, less commonly, as a
area of studded follicular papules without prominent sessile plaque or subcutaneous mass. It hardly changes
hairs. Occasionally, large areas may be involved. Stroking in size and appearance over time. Rarely, spontaneous
the lesion often produces transient elevation with pilo- telescopic and retractile movements of the ‘skin tag’ may
erection (pseudo‐Darier sign) [39]. Sometimes, worm‐like be noted. There are few cases with multiple lesions.
movements owing to involuntary contraction of the In about one‐quarter of reported RMH cases associ-
arrector pili muscles may be elicited (myokymia) [40]. ated congenital abnormalities have been observed.
Multiple lesions are rare. Generalized forms may These include ocular abnormalities, preauricular sinuses,
present with follicular dimpling and excess skin folds, nasofrontal meningocoele, dermoid cyst, thyroglossal
representing one of the causes of the striking ‘Michelin duct sinus, haemangioma, cleft lip and palate, and more
tyre baby’ phenotype. They may be associated with complex malformation syndromes such as amniotic
psychomotor and growth retardation, epilepsy, ingui- band syndrome and oculocerebrocutaneous (Delleman)
nal hernia, joint hypermobility, skeletal, dental and syndrome (absent corpus callosum, colobomas, orbital
other anomalies [41]. cysts, porencephalic cysts and facial skin tags) [42].
The clinical course of CSMH is benign. In most lesions,
induration, hyperpigmentation and hypertrichosis Differential diagnosis. RMH lesions may be clinically or
diminish with age. Malignant transformation has not pathologically mistaken for accessory tragi, congenital
been observed. midline cervical cleft, nasal glioma, fibrous and other rare
Chapter 9  Developmental Anomalies 111

hamartomas of infancy, fetal rhabdomyoma, naevus


lipomatosus superficialis, benign Triton tumour, acro-
chordon, infantile myofibromatosis and smooth muscle
hamartoma [43]. Histological examination establishes
the correct diagnosis.

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Treatment. Simple excision is sufficient. Removal of
plaque‐type variants or other atypical presentations is not
obligatory because malignant transformation has never
been observed. Systemic evaluation of patients is advisable
because of possible associated anomalies.

­Cutaneous signs of cranial dysraphism

Definition. The terms cranial dysraphism and cranial


neural tube defect summarize developmental defects of
Fig. 9.9  Hair collar sign, associated with rudimentary meningocoele.
the central nervous system due to failure of normal
midline fusion [44]. Neural tube defects may occur as
an isolated anomaly but also form part of many syn- of congenital nontraumatic scalp nodules are supposed to
dromes and chromosomal aberrations. connect to the underlying central nervous system.
Cephalocoele is the general term for a congenital herni- Additional cutaneous stigmata of cranial dysraphism
ation of intracranial structures through a scalp defect. [3,45] include capillary malformations and large haeman-
An encephalocoele or meningoencephalocoele contains giomas, cutaneous dimples, dermoid cysts and dermal
both meninges and brain tissue, and the sac content sinuses. The latter are 1–5 mm tracts in the midline of the
communicates with cerebrospinal fluid through the bony nose (see Nasal glioma (nasal cerebral heterotopia)) or
defect. A meningocoele is a scalp herniation in which the occipital scalp, connecting a dermoid cyst to the skin sur-
sac contains only meninges and cerebrospinal fluid. For face. They may not be noticed until infection occurs and
neural tube defects in which the central nervous system pus drains from the orifice. Communication with the
connection has been ablated and the underlying bone central nervous system implies the risk of meningitis.
defect has healed, various terms are in use including cuta- Of special significance are circumscribed abnormalities
neous neural heterotopia, heterotopic or ectopic brain tis- of hair growth. The so‐called hair collar sign (Fig. 9.9) is
sue and rudimentary, atretic or vestigial encephalocoele/ characterized by a ring of coarse, long, dark hair encircling
meningocoele. a bald nodule, plaque or patch [46]. It is found around
encephalocoeles and meningocoeles as well as around
Aetiology. Cranial and spinal dysraphism result from their rudimentary counterparts. A tuft of hair may have the
genetic, environmental and dietary factors. In contrast to same implication. Lesions surrounded by a hair collar are
what was formerly believed, neural tube closure initiates most often located at the vertex or parietal regions. About a
at several, probably five, separate sites representing the third of them are associated with an underlying skull defect
clinical ‘hotspots’ of dysraphism. Specific genes may con- [47]. Coexistence of the hair collar sign and a capillary
trol individual closure sites. Reduced red blood cell folate malformation around a congenital scalp lesion is a highly
levels are associated with a higher risk of open neural indicative cutaneous marker of cranial dysraphism [48].
tube defects. Rudimentary meningocoele and nasal glioma are dis-
cussed in more detail later in the chapter.
Clinical features. Large encephalocoeles and meningo-
coeles are noted at birth and usually do not come to the Differential diagnosis. Cephalocoeles may be mistaken
attention of dermatologists. for dermoid and epidermoid cysts, a haemangioma, lipo-
Smaller or rudimentary encephalocoeles and menin- blastoma or lipoma, naevus psiloliparus, pilomatricoma,
gocoeles may be mistaken for cutaneous tumours or may eosinophilic granuloma or cephalohaematoma. The exact
be inconspicuous enough to go unnoticed for a time. nature of the cephalocoele has to be defined by cranial
Usually, minor forms of cranial dysraphism present as a imaging, especially MRI. Bony defects vary greatly in size
soft, cystic, compressible nodule of variable size with a from large, obvious defects easily demonstrated radiologi-
bluish translucent or glistening surface in the midline of cally to small defects evident only on surgery. Elevation of
the frontal, parietal or occipital scalp or 1–3 cm lateral to α‐fetoprotein in maternal serum, a well‐established marker
it. The base of the nodule often has an oval shape and of neural tube defects, is found only in open subtypes.
smooth boundaries. The malformation increases in size
on agitation of the neonate. Occasionally, the lesion may Treatment. Surgical correction of neural and – if necessary –
flatten to an atrophic, parchment‐like, well‐marginated bony structures with plastic reconstruction is the treatment
scar during the first months of life. Between 20% and 37% of choice for all cephalocoeles.
112 Section 2  Skin Disorders of the Neonate and Young Infant

­ udimentary (atretic, sequestrated)


R evaluation including appropriate radiological imaging
meningocoele is required.

Definition. Rudimentary meningocoele is a develop-


mental anomaly in which meningothelial elements are ­ asal glioma (nasal cerebral
N
found ectopically in the skin or subcutis without an heterotopia)
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

overt underlying bone defect.


Definition. Nasal cerebral heterotopia denotes a benign
mass of heterotopic, mainly glial, brain tissue at the nasal
Aetiology. As discussed previously, rudimentary menin- root thought to arise from the abnormal extracranial
gocoele is regarded as a remnant of a neural tube defect sequestration of neural elements during embryogenesis
with abnormal attachment of the developing neural tube to [50]. The term ‘nasal glioma’ is sensu stricto a misnomer in
the skin [49] displaying a continuous spectrum with true that it implies an underlying neoplastic condition with
meningocoele. A few familial cases have been observed. malignant potential.

Pathology. Histological diagnosis is difficult as changes Aetiology. In most cases, a portion of neuroectodermal
are often subtle and meningothelial cells may be mistaken tissue is sequestrated by closure of the craniofrontal
for vascular elements or connective tissue. In the dermis sutures, equivalent to an encephalocoele that has lost
and subcutis a variably dense collagenous stroma con- its intracranial attachment. If closure is incomplete, as
tains scattered strands and small aggregates of menin- it is in 15–25% of cases, a vestigial stalk of fibroglial tissue
gothelial cells lining pseudovascular spaces. The ovoid to remains attached to the neuroectodermal mass through
spindle‐shaped cells have a scant eosinophilic cytoplasm the foramen caecum.
with indistinct cell borders and small oval nuclei with
finely stippled chromatin. Calcification may be present.
Immunohistochemical examination applying a panel of Pathology. Histologically, nasal gliomas are composed
antibodies is necessary to assess lineage and confirm the of nonencapsulated islands of astrocytes and neuroglial
meningothelial derivation. fibres, embedded in varying amounts of fibrovascular
connective tissue stroma [51]. The glial cells stain posi-
tively with antibodies to glial fibrillary acidic protein
Clinical features. The congenital malformation is and S100 protein. Multinucleated giant astrocytes and
observed at birth or in the first few months of life. The gemistocytic astrocytes are often present with a variable
clinical presentation is highly variable, ranging from a inflammatory infiltrate. Occasionally, focal calcifications
dimple, a bald patch, a pink papule or a soft cystic are present. In some cases, definitive distinction from
nodule to an exophytic mass up to several centimetres in encephalocoele is not possible on histological grounds.
diameter. However, most lesions are small and are usu-
ally located over the posterior scalp or cranial suture
lines. The posterior trunk axis and forehead are less Clinical features. Nasal gliomas are slightly more
common sites. Rarely, more than one lesion is noted. ­common in boys than in girls. There is no familial pre-
Rudimentary meningocoele may be accompanied by disposition and malignant degeneration has never been
abnormalities of hair growth, such as a ‘hair collar’ (see reported.
Fig. 9.9), hair tuft or (annular) alopecia. In the majority of Extranasal (60%), intranasal (30%) and combined cases
cases, no underlying skull defect is present. However, (10%) can be distinguished. Extranasal gliomas are
minor rudimentary bony defects, a fibrous tract connect- usually recognized at birth. They present as a smooth,
ing to the dura or other evidence of residual communica- firm, nontender, noncompressible, nontransilluminating
tion to the meninges may occasionally be identifiable in nodule or mass at the root of the nose, often located to
imaging studies or during surgery and point to their one side of the midline. More unusual sites include the
pathogenetic mechanism. junction of the bony and cartilaginous portions of the
nose, and between the frontal, nasal, ethmoid and lacri-
mal bones [50]. The overlying skin may be red, bluish or
Differential diagnosis. Clinically, other types of cranial telangiectatic. In contrast to encephalocoeles, the bulge
dysraphism, aplasia cutis congenita, congenital dermal does not pulsate or expand with straining and crying, or
sinus, dermoid cyst, epithelial and sebaceous naevus, when the jugular vein is compressed (negative Furstenberg
vascular naevi and tumours, lipoblastoma and lipoma, test) [52]. Bilateral occurrence is very exceptional.
neurofibroma and meningioma may be taken into account. Intranasal gliomas are seen as a polypoid mass pro-
Histological differential diagnosis mainly includes classical truding from a nostril or, more commonly, within the
meningocoele, secondary cutaneous meningioma, endothe- nasal cavity or nasopharynx. Nasal obstruction with
lial neoplasms and giant cell fibroblastoma. breathing and feeding difficulties and rhinorrhoea may
result [53].
Treatment. Rudimentary meningocoele is usually cured Although benign, the mass may cause substantial
by complete local excision. To exclude any communica- deformity and displacement of the nasal skeleton.
tion to the central nervous system, careful preoperative In  cases with intracranial communication, leakage of
Chapter 9  Developmental Anomalies 113

c­erebrospinal fluid with consecutive meningitis or ­Congenital inclusion dermoid cysts


encephalitis may occur.
Aetiology. Congenital inclusion dermoid cysts result
Differential diagnosis. Congenital midline nasal masses from the entrapment of skin and adnexal structures along
occur in one in about 20 000–40 000 live births. Differential the lines of embryonic fusion. Although usually sporadic,
diagnosis includes numerous lesions (Box 9.1), of which several familial cases in frontonasal, epibulbar and exter-

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
dermoid cysts, haemangiomas, nasal gliomas and enceph- nal angular sites have been reported. Inclusion dermoid
alocoeles are the most frequent [54]. cysts have to be distinguished from epidermoid and
trichilemmal cysts, and from the more complex ovarian
Treatment. Biopsies and any partial nasal interven- ‘dermoids’, which represent benign cystic teratomas com-
tions should be avoided. Prior to surgery, a thorough prising entodermal, ectodermal and mesodermal tissue.
radiological evaluation is mandatory, also for differ-
entiation from other congenital nasal masses [55]. Pathology. Dermoid cysts are located in the subcutis
Ultrasound and Doppler flow studies help to rule out but are known to form deep tracts that may adhere to
haemangioma. CT scans are useful in visualizing bony subjacent periosteum and erode or penetrate the bony
defects, whereas MRI is better suited to rule out intrac- calvarium. The skin‐like (‘dermoid’) cyst wall is lined
ranial extension. by keratinizing stratified squamous epithelium. The
The treatment of choice is complete surgical resection adjacent dermis may contain all elements of the skin
early in life to prevent facial deformities, secondary vis- including hair follicles, smooth muscle, sebaceous and
ual impairment, meningitis and other complications eccrine sweat glands, and fibroadipose tissue. In contrast
[52]. Owing to intracranial extension of the sequestered to the clinical presentation, many dermoid cysts show
glial tissue in 15–25% of cases, the therapeutic approach signs of inflammation on histological grounds, mainly
is often multidisciplinary and requires the collaboration due to leakage of the keratin contents.
of otorhinolaryngologists and neurosurgeons. Excision
of the skin overlying the tumour may avoid recurrences Clinical features. Most external dermoid cysts are noticea-
if the glial tissue infiltrates the dermis. Endoscopic ble at birth or early in life. If small, the manifestation may
resection of the nasal glioma with immediate repair of be delayed until inflammation and subsequent enlarge-
the skull base defect may be a reliable technique in ment occur. Typically, they present as a firm, roundish,
selected cases. slowly growing, asymptomatic, subcutaneous nodule or
mass of variable size on the head or neck and are nonpul-
sating, nontransilluminating and noncompressible [56].
Box 9.1  Differential diagnosis of congenital midline nasal The orbital and periorbital areas, in particular the lateral
masses eyebrow (Fig.  9.10), are by far the most common sites.
Lateral eyebrow dermoid cysts often adhere to the under-
Benign lying bone but do not exhibit intracranial invasion [57].
• Epidermoid cyst Nasal dermoid cysts appear as a pale, flesh‐coloured,
• Glioma (ectopic neuroglia) sometimes pearly or erythematous nodule or mass located
• Encephalocoele in the midline close to the glabella or on the bridge of the
• Meningocoele nose [50]. In about half of the cases, they are associated
• Dermoid cyst with a sinus tract of variable length connecting the der-
• Juvenile xanthogranuloma moid cyst to the surface of the skin (nasal dermoid sinus
• Haemangioma cyst). The sinus ostium presents as a small punctum or pit
• Lymphangioma anywhere on the midline of the nose between glabella and
• Meningioma
• Neurofibroma
• Teratoma
Malignant

• Rhabdomyosarcoma
• Olfactory neuroblastoma
• Lymphoma
• Metastatic tumour
• Teratoma

Infectious

• Subcutaneous abscess
• Dacryocystitis

Facial trauma

Source: Adapted from Julapalli MR et al. 2006 [54].


Fig. 9.10  Dermoid cyst underlying the lateral eyebrow.
114 Section 2  Skin Disorders of the Neonate and Young Infant

base of the columella, most frequently on the distal two‐ bony or neural elements of the spine in the dorsal midline
thirds of the dorsum of the nose, and may intermittently of the back. Open spinal dysraphism, with non‐neurulated
discharge. A small tuft of hair emerging from the pit is a neural tissue exposed to the environment, is distinguished
characteristic sign. The sinus is mostly confined to superfi- from closed or occult spinal dysraphism, which is cov-
cial structures but may also extend deeply, penetrate the ered by intact skin. The latter may be further subdivided
nasal septum and the base of the skull and – with a reported based on whether a subcutaneous mass is present on
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

incidence of 6–45% – even reach intracranial structures [58]. the lower back or not. Closed spinal dysraphisms with
In these cases, it most often passes through the foramen associated mass comprise lipomyelocoele, lipomye-
caecum or cribriform plate to the base of the frontal fossa lomeningocoele, meningocoele and myelocystocoele.
and adheres to the leaves of the falx cerebri extradurally. Closed spinal dysraphisms without mass include simple
Patients with a clinically evident sinus opening are sug- dysraphic states, such as tight filum terminale, intradural
gested to have a higher risk of intracranial extension. lipoma and dermal sinus, and complex dysraphic states,
Other dermoid cysts are found overlying the anterior such as diastematomyelia (split spinal cord).
fontanelle, in submental, suprasternal and suboccipital
regions. When located on the scalp, there may be focal Aetiology. Separation of the neuroectoderm from the
alopecia surrounded by a collar of hypertrophic hair. epithelial ectoderm occurs between the third and fifth
Dermoid cysts may gradually flatten and resolve over a weeks of intrauterine life. Lack of separation of germi-
number of years leaving behind a scar. nal layers and failure of closure of the caudal and
Common complications of dermoid cysts include cephalic neuropores result in neural tube defects which
recurrent inflammation, e.g. secondary to trauma, and often involve tissues overlying the spinal cord, such as
bacterial infection, particularly if the cyst is connected to meninges, vertebral arches and skin. Most combined
the skin surface via a sinus. Infection may result in local defects are located on or near the midline in the lum-
abscesses, periorbital and mid‐facial cellulitis, and osteo- bosacrococcygeal and suboccipital regions in which
myelitis. Recurrent inflammation and expansion of the fusion of neural folds occurs last. The recurrence risk in
cyst may cause bone atrophy and skeletal distortion. In subsequent pregnancies is increased, however it is less
cases of intracranial connection, brain abscesses, menin- than 25%.
gitis and meningoencephalitis may develop. In submen-
tal and anterior neck locations the cysts may compromise Clinical features. Overt forms of spinal dysraphism
swallowing, speech and respiration. Malignant degener- are easily recognized, whereas occult forms may be
ation is extremely rare. overlooked until neurological deficits occur. A variety of
congenital cutaneous signs which are associated in about
Differential diagnosis. Differential diagnosis essentially three‐quarters of cases (43–95% in different series) may
depends on the location of the dermoid cyst. Other types serve as early clues to the diagnosis of occult spinal
of cyst, such as epidermoid, trichilemmal, glandular and ­dysraphism [59]. Lesions with a high and low index of
other developmental cysts, are the most frequent simula- suspicion may be differentiated (Box 9.2 and Fig. 9.11)
tors. When located on the neck, dermoid cysts may be [60,61]. In general, a combination of two or more con-
mistaken for thyroid neoplasms or enlarged lymph nodes. genital midline skin lesions constitutes a much higher
Box 9.1 provides a comprehensive list of congenital mid- degree of suspicion than isolated anomalies.
line nasal masses. Lipomatous swellings, either isolated or in conjunction
with other suggestive lesions, especially overlying a
Treatment. Complete excision under general anaesthesia vascular naevus, are the most common midline cutane-
is performed to prevent inflammation and for cosmetic ous markers of occult spinal dysraphism. They present
reasons. Endoscopic approaches have been introduced to as soft, nontender, poorly circumscribed masses of fat
avoid prominent scars in aesthetically challenging sites. The of considerably varying size in the lumbosacral area.
best time for intervention is considered to be early child- Histologically, they are not encapsulated and are finely
hood, around 2 years of age. The surgeon should be aware lobulated, much like mature adult fat. The fatty tissue
that dermoid cysts are often fixed to the surrounding tissue. may penetrate through a vertebral defect into the
Prior to surgery, radiological studies are mandatory to visu- intraspinal canal causing a tethered cord (lipomye-
alize putative intracranial and intraorbital extension. CT is lomeningocoele). Deviation of the gluteal furrow is an
best suited to detect bony alterations while MRI is the pre- important feature suggesting an underlying mass.
ferred method for evaluating the anatomy of the cyst and Dorsal dermal sinuses are deep tracts that may connect
sinus tract and potential intracranial connection. In case of the skin directly to the spinal canal. The sinus opening may
the latter, craniotomy is required for adequate resection. be located off the midline. Epidermoid cysts lined by strati-
fied squamous epithelium or dermoid cysts containing, in
­ utaneous signs of occult spinal
C addition, cutaneous appendages in association with the
dysraphism cyst wall may form at any depth along a sinus. Their cuta-
neous openings, usually located in the midline of the
Definition. The term spinal dysraphism encompasses a ­suboccipital and lumbosacral areas, are often accompanied
spectrum of developmental abnormalities caused by by other skin lesions, such as dimpling, hypertrichosis or
incomplete fusion or malformation of mesenchymal, vascular naevus. There is a considerable risk of infection,
Chapter 9  Developmental Anomalies 115

Box 9.2  Cutaneous signs of spinal dysraphism

Depressed lesions • Congenital melanocytic naevus


• Teratoma
• Congenital absence of skin*
• Neurofibroma
• Sinus tract (with or without dermoid cyst)*

SECTION 2: SKIN DISORDERS


• Atypical dimple* (see text)

OF NEONATES AND INFANTS


Polypoid lesions
• Simple dimple (see text)
• True (persistent vestigial) tail*
Dermal lesions • Pseudotail*
• Congenital scar* • Acrochordon*
• Connective tissue naevus
• Hypertrophic skin Subcutaneous masses

• Lipoma*
Dyschromic lesions
• Dermoid cyst*
• Hyperpigmentation • Neural tissue* (ependymoma, lipomeningocoele, lipomyelomeningo-
• Hypopigmentation coele, occult meningocoele)

Hairy lesions Vascular lesions


• ‘Faun tail’* • Infantile haemangioma
• ‘Silky down’* • Vascular naevus
• Other forms of hypertrichosis* • Telangiectasia*

Neoplasms

• Hamartoma, unclassifiable

* Lesions marked with an asterisk are those with a high index of suspicion.
Source: Adapted from Davis et al. 1994 [59].

Neurosurgical
Overt protrusion Open spinal dysraphism intervention,
of neural tissue supportive therapy

Cutaneous signs
if indicated
• ≥ 2 of the following
findings
• Congenital scar/sinus
High index of suspicion of Imaging
• Hypertrichosis occult spinal dysraphism (ultrasound, magnetic
• Vestigial tail/acrochordon resonance tomography)
• Lipoma
• Haemangioma
• Dermoid cyst
• Hyper-/hypopigmentation
Low index of suspicion of
Fig. 9.11  An algorithm describing the approach • Nevus, hamartoma Watch & wait
occult spinal dysraphism
to a child with spinal dysraphism. • Telangiectasia

including meningitis and epidural, subdural or spinal localized just above the gluteal furrow can be considered
cord abscesses. Inflammatory enlargement of cysts and as harmless.
sinuses may exert pressure on neural structures. Abnormal lumbosacral hair growth present at birth is
Skin dimples are roundish depressions caused by fixa- commonly associated with other indicative stigmata of
tion of the skin to underlying fibrous or bony structures. spinal dysraphism and frequently associated with tethered
They are usually located in the midline of the postanal cord and diastematomyelia. A ‘faun tail’, a broad lozenge‐
area. This common finding presents in about 4% of new- shaped patch of coarse terminal hair, and a ‘silky down’, a
borns and is rarely associated with underlying anomalies. line of soft lanugo hairs limited to a discrete midline area,
Only ‘atypical’ dimples that are deep, large (≥ 5 mm), high may be differentiated from other forms of hypertrichosis.
on the back (≥2.5 cm from the anus) or combined with The hairy lesion may hide a dermal sinus opening.
other lesions are associated with a considerable risk of A human tail is a rare, finger‐like, cutaneous appendage
spinal dysraphism. In contrast, ‘simple’ dimples up to protruding from the lumbosacrococcygeal area. It is
5 mm in diameter and 2.5 cm or closer to the anus and believed to represent a persistent caudal vestigium.
116 Section 2  Skin Disorders of the Neonate and Young Infant

Histologically, it contains a central core of mature fat


Treatment. Undetected spinal dysraphism often gives
and bundles of muscle and nerve fibres. In contrast, a
rise to severe orthopaedic (abnormal gait, clubfoot,
pseudotail is a stump‐like projection that may contain
inequality of the lower limbs, scoliosis), neurological
cartilage.
(urinary and faecal incontinence, muscle weakness and
An acrochordon is a small sessile or pedunculated skin
atrophy, awkwardness, paresis, back pain, pain or sen-
tag composed of epidermis and a fibrovascular core. In all
sory deficits in the legs with resultant trophic changes,
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

developmental abnormalities mentioned here, appropri-


recurrent meningitis) and urological complications (uri-
ate radiological imaging is mandatory prior to removal
nary tract infections) in infancy or childhood [58,62]. The
since these findings may be associated with an underlying
sequelae develop insidiously and are often permanent.
dysraphic state, even in the absence of further cutaneous
As posterior midline cutaneous abnormalities are usually
markers.
present at birth, attentive paediatricians and dermatologists
Among vascular lesions, haemangiomas are the most
have the opportunity to forestall irreversible damage.
important cutaneous signs indicating spinal dysraphism.
Early neurosurgical intervention can prevent deteriora-
They may be large and tend to ulcerate. The so‐called
tion, or even improve symptoms. High‐resolution spinal
PELVIS syndrome indicates perianal haemangioma,
ultrasonography is an easy screening method for early
external genitalia malformations, lipomyelomeningo-
detection of occult dysraphic lesions, even prenatally, but
coele, vesicorenal abnormalities, imperforate anus and
its use is limited to the first 4–6 months of life owing to
skin tags. Telangiectasias or vascular naevi, whether in a
skeletal maturation and a relatively high rate of false neg-
nuchal or sacral location, are of minor significance.
ative findings. MRI is accepted as the best noninvasive
Further lesions associated with spinal dysraphism
technique for the detection of occult spinal dysraphism
include congenital absence of skin or scar‐like defects
[59]. Therefore, spinal MRI has to be performed in every
(‘cigarette burn marks’) in the lumbosacral region, tera-
case where there is a high degree of suspicion, even if ultra-
tomas, melanocytic naevi and unusual pigmentation.
sound examination is unremarkable. Plain radiographs
Other occasionally associated cutaneous stigmata are
are not sufficiently sensitive.
listed in Box 9.2.

Differential diagnosis. A variety of other tumours may be ­ plasia cutis congenita (congenital
A
located in the lumbosacrococcygeal area, such as naevus absence of skin)
lipomatosus (Fig.  9.12), rectal duplication, chordoma,
pacinioma and astrocytoma. In contrast to dorsal dermal Definition. The term aplasia cutis congenita (ACC) is
sinuses, pilonidal sinuses represent tracts superficial to used for a heterogeneous group of disorders in which the
the sacral fascia. They are not connected with the spinal skin is absent at birth or a congenital scar is present. In
canal and rarely appear before adulthood. most cases, this condition is localized and solitary, how-
ever multiple or widespread areas may be affected.

Aetiology. Congenital absence of skin areas has an esti-


mated incidence of 1–3 in 10 000 births. It is not attributable
to a single cause. Exogenous factors, such as detached
amniotic adherences, intrauterine trauma, viral infections
and teratogens, as well as endogenous factors such as
vascular abnormalities, associated fetus papyraceus and
genetic factors can be causative. Genetic causes of ACC are
heterogeneous and mutations have been found in genes
previously not known to regulate skin morphogenesis [63].
Numerous familial cases, most of them compatible with
autosomal dominant transmission, have been reported.
The most popular classification of ACC distinguishes nine
groups according to the site involved and the presence or
absence of associated abnormalities [64] (Table 9.2).

Pathology. Healed areas show a flattened re‐epithelialized


epidermis and an underlying dermal fibrosis with absence
of adnexal structures and variable reduction of elastic
fibres.

Clinical features. About 80% of lesions are located on the


scalp, most of them in the midline in close proximity to
the parietal hair whorl [4] (Fig. 9.13). Scalp lesions are
Fig. 9.12  Naevus lipomatosus in the lumbosacral area. single in about 75% of cases, double in about 20% and
Chapter 9  Developmental Anomalies 117

Table 9.2  Frieden’s classification of aplasia cutis congenita (ACC)

Group Specification Body areas affected Associated abnormalities and syndromes Inheritance

1 Scalp ACC without Scalp, particularly vertex Without associations Sporadic or autosomal
multiple anomalies In association with isolated abnormalities including: dominant
• Cleft lip and palate

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
• Tracheo‐oesophageal fistula
• Double cervix and uterus
• Patent ductus arteriosus
• Heart disease
• Omphalocoele
• Polycystic kidneys
• Mental retardation
• Cutis marmorata telangiectatica congenita
2 Scalp ACC with limb Midline scalp Adams–Oliver syndrome Autosomal dominant
reduction abnormalities
3 Scalp ACC with associated Scalp Linear epidermal naevus Sporadic
epidermal and organoid Linear sebaceous naevus (didymosis aplasticosebacea)
naevi Schimmelpenning syndrome
SCALP syndrome
4 ACC overlying Abdomen, posterior axis, Meningomyelocoele Variable, depending on
embryological scalp Cranial stenosis underlying condition
malformations Congenital midline porencephaly
Leptomeningeal angiomatosis
Spinal dysraphism
Omphalocoele
Gastroschisis
5 ACC with associated fetus Multiple, symmetrical Single umbilical artery, developmental delay, spastic Sporadic
papyraceus or placental areas paralysis, nail dystrophy, clubbed hands and feet,
infarcts amniotic constriction band, gastrointestinal atresia
6 ACC associated with Extremities, rarely trunk Blistering of skin and/or mucous membranes, absent or Autosomal dominant or
epidermolysis bullosa (EB) deformed nails, metatarsus varus, congenital absence of recessive, depending
kidney, pyloric or duodenal atresia, abnormal ears and on type of EB
nose, ureteral stenosis, renal abnormalities, arthrogryposis
7 ACC localized to Pretibial areas, dorsal None Autosomal dominant or
extremities without aspects of hands and recessive
blistering feet, extensor areas of
wrists
8 ACC caused by specific Any area Drugs: methimazole, misoprostol, cocaine, valproic acid, Sporadic
teratogens benzodiazepines, heparin
Intrauterine herpes simplex infection
Intrauterine varicella zoster infection
9 ACC associated with Scalp, any area Trisomy 13 (Patau syndrome) Variable, depending on
malformation 4p syndrome (Wolf–Hirschhorn syndrome) underlying condition
syndromes Xp deletion syndromes
Oculocerebrocutaneous syndrome (Delleman–Oorthuys
syndrome)
Johanson–Blizzard syndrome
Focal dermal hypoplasia
Focal facial dermal dysplasia
Bitemporal ACC syndrome (Setleis syndrome)
Oculoectodermal syndrome
Rodrigues syndrome

SCALP, sebaceous naevus syndrome, central nervous system malformations, aplasia cutis congenita, limbal dermoid and pigmented naevus (giant
congenital melanocytic naevus) with neurocutaneous melanosis.
Source: Adapted from Frieden 1986 [64].

triple in about 8%. Typically, they are 1–3 cm in diameter, with areas of preserved skin appendages (nonmembra-
round or oval in shape, sharply marginated and hairless. nous ACC). In approximately 20% of cases lesions extend
At birth, they often show an atrophic, shiny, membrane‐ to the bone, dura or meninges, with possible complica-
like surface, with or without a hair collar (membranous tions of haemorrhage from the eroded sagittal sinus, sinus
ACC). In contrast, larger lesions tend to have an irregular, thrombosis and meningitis. Even if the skull is involved,
stellate outline and to be deeply ulcerated. Healing leaves ulcers tend to heal spontaneously within several months.
a sclerotic, sometimes hypertrophic surface intermingled Membranous ACC has been suggested to result from
118 Section 2  Skin Disorders of the Neonate and Young Infant

larger defects are recommended to be closed early by skin


grafts or local flaps. Application of cultured dermal and
epithelial autografts as a minimally invasive method has
been used to accelerate healing. Bone grafts may be
needed for closure of cranial defects. If acute intervention
is not required, an unsightly scar may later be excised or
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

hidden by hair transplants. Tissue expansion of the adja-


cent hair‐bearing scalp can help to close larger surgical
defects resulting from scar removal.

­ mniotic constriction band


A
(amnion rupture sequence)

Definition. Amniotic constriction band comprises an


extremely varied group of sporadic congenital birth
defects characterized by fibrous constriction bands,
limb deformities and amputations, acrosyndactyly and
multiple craniofacial, visceral and body wall defects. It is
supposed to be secondary to a disruption process of the
amniotic membrane.
Fig. 9.13  Aplasia cutis congenita on the vertex healed with atrophic scarring.
Aetiology. The condition is believed to arise from early
incomplete or faulty closure of ectodermal fusion lines rupture of the amniotic sac, leading to inhibition of its
whereas nonmembraneous ACC appears to have a differ- growth and formation of mesodermic fibrous strands
ent pathogenesis. between the outer surface of the amnion and the denuded
Lesions on the trunk and limbs are often large, irregular chorion (‘extrinsic theory’) [65]. When passing through
in shape and symmetrical in distribution. This unusual the defect in the amniotic membrane, fetal parts may be
pattern may indicate the presence of a fetus papyraceus entangled by these inelastic fibrous strands, resulting in
and also occurs in association with certain severe types of ring constrictions and even amputation of limbs or digits.
epidermolysis bullosa. Healing may lead to dermatogenic Temporary oligohydramnios due to increased amniotic
contractures, especially in the vicinity of the joints. fluid absorption by the bared chorion gives rise to com-
Elevated levels of α‐fetoprotein in midtrimester pression and decreased movement of the fetus. If early
maternal serum and amniotic fluid as well as elevated and acute, this may cause haemorrhage and necrosis,
acetylcholinesterase levels in amniotic fluid may result whereas during organogenesis compression may lead to
from widespread ACC. visceral and neural tube defects and later to clubbing of
In the majority of cases, further abnormalities are the feet, scoliosis and joint contractures. Other, particu-
lacking. However, congenital absence of skin may rarely larly craniofacial, defects may result from vascular dis-
be associated with a large number of anomalies, such as ruption with or without amniotic adhesion to the embryo.
transverse limb malformations (Adams–Oliver syndrome), Early rupture of the amniotic membrane before 45 days
epidermal and organoid naevi, overt or occult develop- gestational age is claimed to produce the most severe
mental abnormalities, fetus papyraceus, epidermolysis defects and even spontaneous abortion. Maternal abdom-
bullosa, specific teratogens (methimazole), intrauterine inal trauma, uterine malformations, dietary deficiencies,
infections, chromosomal abnormalities, ectodermal dys- teratogens and the use of oral contraceptives shortly
plasias and other malformation syndromes [1] (Table 9.2). before conception have all been incriminated in causing
amnion rupture.
Differential diagnosis. Lesions of congenital absence of As the extrinsic theory does not readily explain all pos-
skin have to be differentiated from iatrogenic injuries by sible manifestations, an ‘intrinsic theory’ has been intro-
forceps or scalp electrodes. Further differential diagnoses duced advocating alterations in key germline genes
include congenital abscess of the scalp, encephalocoele implicated in normal development. More probably, dif-
and meningocoele. Healed areas may be mistaken for ferent patterns of presentation result from different aeti-
other causes of cicatricial alopecia, sebaceous naevus, ologies. For example, certain combined defects featuring
naevus psiloliparus or, if hypertrophic, scalp tumours. congenital skin pedicles and cleft lip and palate may cor-
respond to mutations in the mutant mouse ‘disorganiza-
Treatment. There is no general consensus on the manage- tion gene’ [66].
ment of ACC. In most cases, conservative management of Amniotic band syndrome occurs in approximately 1 in
the wound with antiseptics and appropriate dressings is 1200–15 000 live births. Females outnumber males by
sufficient. Deeper lesions require radiological evaluation about 3 : 2. The prevalence has been reported to be 1.76
of the underlying bone. To prevent serious complications, times higher in African Americans than in Caucasians.
such as haemorrhage, local infection and meningitis, Familial occurrence is extremely rare [67].
Chapter 9  Developmental Anomalies 119

dermatoglyphics has also been observed in Kindler


Clinical features. Anomalies occurring within the scope
syndrome.
of amniotic constriction band may be divided into three
Ridge hypoplasia (poorly formed dermal ridges) can
types: disruptions, deformations and malformations.
occur as an isolated autosomal dominant defect or in
They are highly variable and assumed to depend upon
association with chromosomal abnormalities and congen-
the time of amnion rupture. Early rupture is thought to
itally deformed limbs.
give rise to severe craniofacial (encephalocoele, meningo-

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
In ridge dissociation, the dermal ridge is broken into
coele, anencephaly, cleft lip and palate, nasal deformity,
short segments instead of being continuous. This pattern
microphthalmia), visceral and body wall defects and can
can occur sporadically, as an autosomal dominant condi-
be inconsistent with life. More often, constriction bands
tion, or in association with other diseases, such as hypohi-
circumferentially encircle the distal limbs and may be
drotic ectodermal dysplasia, chromosomal abnormalities
deep enough to cause distal lymphoedema, compression
and cystic fibrosis.
of sensory and motor nerves, and ischaemia with result-
The ridges‐off‐the‐end pattern, which is characterized
ant deficits, atrophy and amputations at any level of an
by ridges running off the fingertip instead of looping
extremity or digit. Further defects include hand deformi-
around it, is usually not associated with other anomalies.
ties, clubfoot (talipes equinovarus), atypical syndactyly
Punctate interruptions of the skin ridges indicating
predominantly involving the distal portions of the digits
pitting are a well‐known manifestation of Darier disease.
(acrosyndactyly, or fenestrated syndactyly), absence of
Unusual dermatoglyphic patterns also occur in the
skin or alopecia at sites of band attachment, abnormal
Kabuki make‐up syndrome, a multiple congenital anom-
palmar creases and altered dermatoglyphic pattern.
alies/mental retardation syndrome.
Asymmetrical involvement of multiple extremities to var-
ying degrees is typical. Upper extremities are more often ­References
affected than the legs. Scoliosis is a less common feature. 1 Bellet JS. Developmental anomalies of the skin. Semin Perinatol
2013;37:20–5.
2 Aslan C, Tan O, Hosnuter M, Isik D. Primary cutis verticis gyrata.
Differential diagnosis. As there is virtually no increased J Craniofac Surg 2015;26:974–5.
recurrence risk in successive pregnancies, amniotic 3 Larsen F, Birchall N. Cutis verticis gyrata: three cases with different
constriction band must be differentiated from inherited aetiologies that demonstrate the classification system. Australas J
Dermatol 2007;48:91–4.
congenital deformities. Among these, brachydactylies, 4 Behle V, Hamm H. [Developmental abnormalities and nevi of the
Adams–Oliver, AEC (ankyloblepharon, ectodermal defects scalp]. Hautarzt 2014;65:1022–9.
and cleft lip/palate), EEC (ectrodactyly, ectodermal dys- 5 Tanese K, Niizeki H, Seki A et  al. Pathological characterization of
pachydermia in pachydermoperiostosis. J Dermatol 2015;42:710–14.
plasia and cleft lip/palate), Van der Woude and popliteal
6 Snyder MC, Johnson PJ, Hollins RR. Congenital primary cutis verticis
pterygium syndromes may be taken into account. Specific gyrata. Plast Reconstr Surg 2002;110:818–21.
teratogens, including thalidomide, warfarin, phenytoin, 7 Solak SS, Altunay IK, Demirci GT, Can B. Prevalence of congenital
valproic acid and cocaine, and diagnostic or therapeutic cutaneous anomalies in 1000 newborns and a review of the literature.
Am J Perinatol 2016;33:79–83.
interventions in pregnancy may cause indistinguishable 8 Bahrani B, Khachemoune A. Review of accessory tragus with high-
limb anomalies [67]. lights of its associated syndromes. Int J Dermatol 2014;53:1442–6.
9 Coras B, Hafner C, Roesch A et al. Congenital cartilaginous rests of
the neck (wattles). Dermatol Surg 2005;31:1349–50.
Treatment. Plastic surgery with wound closure by Z‐plasty
10 An S‐Y, Choi HG, Lee JS et al. Analysis of incidence and genetic pre-
may be urgently needed for release of constriction or disposition of preauricular sinus. Int J Pediatr Otorhinolaryngol
compression. Release of acrosyndactylies may require 2014;78:2255–7.
skin grafting. Improvement in prenatal diagnosis, imag- 11 Huang XY, Tay GS, Wansaicheong GK‐L, Low W‐K. Preauricular
sinus: clinical course and associations. Arch Otolaryngol Head Neck
ing and fetoscopic surgical techniques now allows in utero Surg 2007;133:65–8.
intervention in selected cases with risk of amputation [68]. 12 Scheinfeld NS, Silverberg NB, Weinberg JM, Nozad V. The preauricu-
lar sinus: a review of its clinical presentation, treatment, and associa-
tions. Pediatr Dermatol 2004;21:191–6.
­Congenital abnormalities 13 Schroeder JW, Mohyuddin N, Maddalozzo J. Branchial anomalies
in the pediatric population. Otolaryngol Head Neck Surg 2007;137:
of dermatoglyphics 289–95.
14 Acierno SP, Waldhausen JHT. Congenital cervical cysts, sinuses and
Congenital malformations of dermatoglyphics (finger- fistulae. Otolaryngol Clin North Am 2007;40:161–76.
prints) may be divided into four categories: ridge aplasia, 15 Puscas L. Midline cervical cleft: review of an uncommon entity. Int J
ridge hypoplasia, ridge dissociation and ridges‐off‐the‐end Pediatr 2015;2015:209418.
16 Kepertis C, Anastasiadis K, Lambropoulos V et al. Diagnostic and sur-
pattern. gical approach of thyroglossal duct cyst in children: ten years data
Ridge aplasia (complete absence of dermal ridges) is an review. J Clin Diagn Res 2015;9:PC13–15.
essential feature of the rare autosomal dominant Basan 17 Righini C‐A, Hitter A, Reyt E, Atallah I. Thyroglossal duct surgery.
syndrome which is further characterized by profuse con- Sistrunk procedure. Eur Ann Otorhinolaryngol Head Neck Dis
2016;133:133–6.
genital milia on the face, acral bullae and palmoplantar 18 Ozel SK, Kazez A, Koseogullari AA, Akpolat N. Scapular broncho-
hypohidrosis. Heterozygous loss‐of‐function mutations genic cysts in children: case report and review of the literature. Pediatr
in a skin‐specific isoform of SMARCAD1 resulting in per- Surg Int 2005;21:843–5.
19 Kumar A, Kanojia RK, Saili A. Skin dimples. Int J Dermatol

turbed expression of epidermal differentiation‐associated
2014;53:789–97.
genes have been described as the molecular basis 20 Shelley WB, Shelley ED, Pansky B. The transverse nasal line: an
for autosomal dominant adermatoglyphia [69]. Loss of embryonic fault line. Br J Dermatol 1997;137:963–5.
120 Section 2  Skin Disorders of the Neonate and Young Infant

21 Risma KA, Lucky AW. Pseudoacne of the nasal crease: a new entity? 46 Chien M‐M, Chen K‐L, Chiu H‐C. The “hair‐collar” sign. J Pediatr
Pediatr Dermatol 2004;21:427–31. 2016;168:246.
22 Stoevesandt J, Hamm H. Transverse nasal novelties: a critical re‐eval- 47 Stevens CA, Galen W. The hair collar sign. Am J Med Genet A

uation of the “allergic salute”. Eur J Dermatol 2013;23:526–7. 2008;146A:484–7.
23 Shobana R, Aithal S, Srikanth S. Congenital lip pits without associated 48 Herron MD, Coffin CM, Vanderhooft SL. Vascular stains and hair
anomalies. Indian J Dermatol Venereol Leprol 2014;80:459–60. collar sign associated with congenital anomalies of the scalp.
24 Rizos M, Spyropoulos MN. Van der Woude syndrome: a review. Pediatr Dermatol 2005;22:200–5.
Cardinal signs, epidemiology, associated features, differential diag- 49 El Shabrawi‐Caelen L, White WL, Soyer HP et al. Rudimentary menin-
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

nosis, expressivity, genetic counselling and treatment. Eur J Orthod gocele: remnant of a neural tube defect? Arch Dermatol 2001;137:45–50.
2004;26:17–24. 50 Adil E, Robson C, Perez‐Atayde A et al. Congenital nasal neuroglial
25 Pingul MM, Quintos JB. Van der woude syndrome (lip pit‐cleft lip heterotopia and encephaloceles: An update on current evaluation
syndrome). J Pediatr 2014;164:1235. and management. Laryngoscope 2016;126:2161–7.
26 Brown J, Schwartz RA. Supernumerary nipples: an overview. Cutis 51 Kathuria P, Agarwal S. Nasal glial heterotopy in childhood: a case
2003;71:344–6. report highlighting the morphological and immunohistochemical fea-
27 Latham K, Fernandez S, Iteld L et al. Pediatric breast deformity. tures. Indian J Pathol Microbiol 2006;49:612–14.
J Craniofac Surg 2006;17:454–67. 52 Rahbar R, Resto VA, Robson CD et  al. Nasal glioma and encepha-
28 Ishida LH, Alves HRN, Munhoz AM et  al. Athelia: case report and locele: diagnosis and management. Laryngoscope 2003;113:2069–77.
review of the literature. Br J Plast Surg 2005;58:833–7. 53 Gnagi SH, Schraff SA. Nasal obstruction in newborns. Pediatr Clin
29 Danarti R, König A, Salhi A et al. Becker’s nevus syndrome revisited. North Am 2013;60:903–22.
J Am Acad Dermatol 2004;51:965–9. 54 Julapalli MR, Cohen BA, Hollier LH, Metry DW. Congenital, ill‐
30 Kieliszak CR, Pollinger TH, Tollefson MM, Griffin JR. Umbilical and defined, yellowish plaque: the nasal dermoid. Pediatr Dermatol
periumbilical dermatoses. J Am Acad Dermatol 2015;72:1066–73. 2006;23:556–9.
31 Snyder CL. Current management of umbilical abnormalities and
55 Hedlund G. Congenital frontonasal masses: developmental anat-
related anomalies. Semin Pediatr Surg 2007;16:41–9. omy, malformations, and MR imaging. Pediatr Radiol 2006;36:647–62;
32 Krauel L, Tarrado X, Garcia‐Aparicio L et al. Median raphe cysts of quiz 726–7.
the perineum in children. Urology 2008;71:830–1. 56 Cambiaghi S, Micheli S, Talamonti G, Maffeis L. Nasal dermoid sinus
33 Lezcano C, Chaux A, Velazquez EF, Cubilla AL. Clinicopathological cyst. Pediatr Dermatol 2007;24:646–50.
features and histogenesis of penile cysts. Semin Diagn Pathol 57 Pryor SG, Lewis JE, Weaver AL, Orvidas LJ. Pediatric dermoid cysts
2015;32:245–8. of the head and neck. Otolaryngol Head Neck Surg 2005;132:938–42.
34 Khachemoune A, Guldbakke KK, Ehrsam E. Infantile perineal 58 Sreetharan V, Kangesu L, Sommerlad BC. Atypical congenital der-
protrusion. J Am Acad Dermatol 2006;54:1046–9. moids of the face: a 25‐year experience. J Plast Reconstr Aesthetic
35 Konta R, Hashimoto I, Takahashi M, Tamai K. Infantile perineal Surg 2007;60:1025–9.
protrusion: a statistical, clinical, and histopathologic study. Dermatol 59 Davis DA, Cohen PR, George RE. Cutaneous stigmata of occult spinal
Basel Switz 2000;201:316–20. dysraphism. J Am Acad Dermatol 1994;31:892–6.
36 Semadeni BL, Mainetti C, Itin P, Lautenschlager S. Precalcaneal con- 60 Sewell MJ, Chiu YE, Drolet BA. Neural tube dysraphism: review of
genital fibrolipomatous hamartomas: report of 3 additional cases and cutaneous markers and imaging. Pediatr Dermatol 2015;32:161–70.
discussion of the differential diagnosis. Dermatol Basel Switz 61 Guggisberg D, Hadj‐Rabia S, Viney C et al. Skin markers of occult
2009;218:260–4. spinal dysraphism in children: a review of 54 cases. Arch Dermatol
37 Greenberg S, Krafchik BR. Infantile pedal papules. J Am Acad
2004;140:1109–15.
Dermatol 2005;53:333–4. 62 Patnaik A, Mahapatra AK. Complex forms of spinal dysraphism.
38 Cambiaghi S, Galloni C, Restano L, Cavalli R. Precalcaneal congenital Childs Nerv Syst 2013;29:1527–32.
fibrolipomatous hamartoma. Int J Dermatol 2006;45:1202–3. 63 Marneros AG. Genetics of aplasia cutis reveal novel regulators of skin
39 Bilgiç Ö, Tunçez Akyürek F, Altınyazar HC. Pseudo Darier sign: a dis- morphogenesis. J Invest Dermatol 2015;135:666–72.
tinctive finding for congenital smooth muscle hamartoma. J Pediatr 64 Frieden IJ. Aplasia cutis congenita: a clinical review and proposal
2016;169:318. for classification. J Am Acad Dermatol 1986;14:646–60.
40 Kienast A, Maerker J, Hoeger PH. Myokymia as a presenting sign of 65 Kawamura K, Chung KC. Constriction band syndrome. Hand Clin
congenital smooth muscle hamartoma. Pediatr Dermatol 2007;24: 2009;25:257–64.
628–31. 66 Isidor B, Baujat G, Le Caignec C et al. Congenital skin pedicles with
41 Holland KE, Galbraith SS. Generalized congenital smooth muscle or without amniotic band sequence: Extending the human pheno-
hamartoma presenting with hypertrichosis, excess skin folds, and fol- type resembling mouse disorganization. Am J Med Genet A 2009;
licular dimpling. Pediatr Dermatol 2008;25:236–9. 149A:1734–9.
42 Takeyama J, Hayashi T, Sanada T et  al. Rhabdomyomatous mesen- 67 Goldfarb CA, Sathienkijkanchai A, Robin NH. Amniotic constric-
chymal hamartoma associated with nasofrontal meningocele and der- tion band: a multidisciplinary assessment of etiology and clinical
moid cyst. J Cutan Pathol 2005;32:310–13. presentation. J Bone Joint Surg Am 2009;91(Suppl 4):68–75.
43 Rosenberg AS, Kirk J, Morgan MB. Rhabdomyomatous mesenchymal 68 Javadian P, Shamshirsaz AA, Haeri S et  al. Perinatal outcome after
hamartoma: an unusual dermal entity with a report of two cases and fetoscopic release of amniotic bands: a single‐center experience and
a review of the literature. J Cutan Pathol 2002;29:238–43. review of the literature. Ultrasound Obstet Gynecol 2013;42:449–55.
44 McComb JG. Spinal and cranial neural tube defects. Semin Pediatr 69 Nousbeck J, Sarig O, Magal L et al. Mutations in SMARCAD1 cause
Neurol 1997;4:156–66. autosomal dominant adermatoglyphia and perturb the expression
45 Drolet BA. Cutaneous signs of neural tube dysraphism. Pediatr of epidermal differentiation‐associated genes. Br J Dermatol 2014;
Clin North Am 2000;47:813–23. 171:1521–4.
 121

C HA PTER   10

Differential Diagnosis of Neonatal


Erythroderma

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
Hagen Ott1 & Peter H. Hoeger2
 Division of Pediatric Dermatology and Allergology, Epidermolysis bullosa Centre Hannover, Children’s Hospital AUF DER BULT, Hannover, Germany
1

 Department of Paediatric Dermatology, Catholic Children’s Hospital Wilhelmstift, Hamburg, Germany


2

Introduction, 121 Adverse drug reactions, 129 Diagnostic work‐up of neonatal


Cutaneous disorders, 123 Immunological disorders, 130 erythroderma, 133
Infections and toxicities, 128 Inborn errors of metabolism, 132

Abstract critically depends on swift diagnosis and early therapeutic inter-


vention. Therefore, this chapter focuses on clinical clues and further
Neonatal erythroderma occurs during the first 28 days of life and diagnostic steps that are helpful in the differentiation of conditions
is defined as a generalized erythema covering more than 90% of regularly associated with neonatal erythroderma. For practical rea-
body surface area. It can be the manifestation of various clinical sons, these are classified into acquired and hereditary skin diseases,
syndromes ranging from benign, transient skin diseases to poten- infections, immunodeficiency disorders, drug reactions and inborn
tially fatal systemic disorders. The prognosis of affected patients errors of metabolism.

Key points • Irrespective of its cause, neonatal erythroderma is associated


with complications due to impaired epidermal barrier function
such as hypothermia, dehydration or infection.
• Neonatal erythroderma is characterized by a generalized
• To differentiate nonsyndromic forms with good prognosis from
erythema covering more than 90% of the neonate’s body
more severe variants of neonatal erythroderma, a thorough
surface area.
diagnostic work‐up is mandatory in every affected patient.
• The underlying illnesses range from benign, erythematosquamous
• Besides routine laboratory tests such as whole blood count, total
disorders such as atopic dermatitis to fatal diseases such as
serum IgE and blood gas analysis, further molecular genetic and
severe combined immunodeficiencies.
other analyses are often required.

­Introduction
increased transcutaneous loss of fluid leading to hyper­
By definition, neonatal erythroderma has its onset during natraemic dehydration, increased energy consumption
the first 28 days of life and refers to a generalized ery­ and hypothermia.
thema with or without scaling that covers more than 90% Thus, in order to avoid a critical delay in diagnosis,
of the newborn’s body surface area [1]. Although reliable definitive identification of the underlying condition is
epidemiological data are still not available, neonatal imperative and often requires further laboratory, histologi­
erythroderma can be considered a rare disorder as dem­ cal, microbiological or molecular genetic analyses [3–5]
onstrated by a single‐centre study of 19 000 paediatric (Fig. 10.1). The historical term ‘Leiner’s disease’ (synonyms:
dermatology patients yielding a 0.11% incidence of eryth­ erythrodermia desquamativa Leiner, Leiner–Moussous
rodermic neonates and infants during a 6‐year period [2]. syndrome) originally denoted infants with exfoliative
Neonatal erythroderma can be the manifestation of erythroderma, diarrhoea and failure to thrive. Because this
various clinical syndromes ranging from benign, tran­ condition can be attributed to a wide range of different dis­
sient skin diseases to potentially fatal systemic disorders. eases it is recommended that the term be abandoned.
The prognosis of affected patients is serious due to a per­ Many of the diseases that need to be considered in this
sistence of severe skin symptoms in 70% after 3 years and, context are discussed in detail elsewhere in this textbook.
more importantly, a mortality rate of up to 25%. Therefore, the focus of this chapter is on clinical clues and
Irrespective of its cause, erythroderma per se is a life‐ further diagnostic steps that are helpful in the differentia­
threatening condition in neonates, mainly due to an tion of conditions regularly associated with neonatal

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
122 Section 2  Skin Disorders of the Neonate and Young Infant

Basic diagnostic work-up


- Patient history
- Clinical examination Neonatal/infantile erythroderma
- Skin smear
- Capillary blood gas analysis
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

CSF, blood, urine + Congenital cutaneous


Candida?
cultures candidiasis
no

Blood ammonia level, Molecular


Ketoacidosis? + + Holocarboxylase synthetase def.
HLCS-activity genetics (HLCS)
no

+ Re-exposure, if + Red man syndrome


Vancomycin? Discontinue
indicated

Ophthalmology, Molecular + Chondrodysplasia punctata II


+
no

BERA/audiometry genetics (EBP)

Immunohistochemistry, Mol. genetics


+ + Netherton syndrome
trichogram (SPINK5)
Ichthyosîs?
Mol. genetics
Electron microscopy + + NCIE/BCIE
(TGM1, K1/10)

Funduscopy, FALDH- Mol. genetics


no

+ + Sjögren–Larsson syndrome
activity (fibroblasts) (ALDH3A2)

Exfoliation? Histology (cleavage level) Subepidermal? Toxic epidermal necrolysis

no

Subcorneal? PCR
+ SSSS
(sea, seb)
no

Total lgE, Mol. genetics


+ + Omenn syndrome
lymphocyte typing (RAG1/2, others)
Dystrophy,
Alopecia?
Histology, Chimerism
+ + Maternofetal GvHD
lymphocyte typing analysis
no

Atopic dermatitis
Consider Psoriasis vulgaris
‘transient erythroderma’ Seborrheic dermatitis
Pityriasis rubra pilaris

Fig. 10.1  Proposal for a diagnostic algorithm to be followed in the management of neonatal and early infantile erythroderma. Source: Ott et al. 2008 [1].
Reproduced with permission of John Wiley & Sons. EBP, emopamil‐binding protein; FALDH, fatty aldehyde dehydrogenase; NCIE/BCIE, nonbullous
congenital ichthyosiform erythroderma/bullous congenital ichthyosiform erythroderma; PCR, polymerase chain reaction.
Chapter 10  Differential Diagnosis of Neonatal Erythroderma 123

Box 10.1  Neonatal erythroderma: differential diagnosis


­Cutaneous disorders
Retrospective hospital‐based investigations have identified
Cutaneous disorders primary skin diseases as the most common cause of
Erythematosquamous skin diseases neonatal and infantile erythroderma, accounting for as
• Seborrhoeic dermatitis many as 80% of all cases. The underlying cutaneous
disorders comprise erythematosquamous diseases, geno­

SECTION 2: SKIN DISORDERS


• Atopic dermatitis

OF NEONATES AND INFANTS


• Psoriasis dermatoses and further skin diseases, particularly scabies
• Pityriasis rubra pilaris and generalized cutaneous mastocytosis [1–3].

Genodermatoses
• Nonsyndromic ichthyosis (nonbullous congenital ichthyosiform Erythematosquamous skin diseases
erythroderma, epidermolytic ichthyosis) Seborrhoeic dermatitis (see Chapters 15 and 21)
• Syndromal ichthyosis (Chanarin–Dorfman syndrome, Conradi– Infantile seborrhoeic dermatitis (SD) represents a tran­
Hünermann–Happle syndrome, Sjögren–Larsson syndrome) sient skin disease of good prognosis affecting up to 70%
• Disorders of epidermal homeostasis (Netherton syndrome, peeling of newborns and infants during the first months of life [4].
skin syndrome type B, SAM syndrome) While pruritus is usually mild or completely absent, skin
• Ectodermal dysplasia lesions typically correspond to erythematous patches
with a yellowish hue and greasy scaling which are most
Other skin diseases prominent on the scalp (cradle cap), the napkin (diaper)
• Diffuse cutaneous mastocytosis area and other intertriginous zones [5]. In most patients,
• Scabies skin symptoms respond rapidly to topical treatment with
low‐potency glucocorticoids or antifungal agents, but in
Infections and toxicities
some cases neonatal and infantile SD may develop into
• Staphylococcal scalded skin syndrome erythroderma.
• Congenital and neonatal candidiasis As pathognomonic laboratory parameters or histological
features do not exist, the diagnosis of infantile SD is
Adverse drug reactions established on clinical grounds. In contrast to atopic
• Stevens–Johnson syndrome, toxic epidermal necrolysis
­dermatitis, erythematous patches in SD are clearly demar­
• Red man syndrome cated, usually less pruritic, and tend to predominate in
the flexural folds including the anogenital area.
Immunological disorders In patients with systemic symptoms such as diarrhoea
or failure to thrive, skin biopsy and further diagnostic
• Omenn syndrome
steps are warranted to rule out more severe conditions
• Graft‐versus‐host disease
such as immunodeficiencies or metabolic defects.
• Other primary immunodeficiencies (DiGeorge syndrome,
Wiskott–Aldrich syndrome, IPEX syndrome)
Atopic dermatitis (see Chapter 15)
Inborn errors of metabolism Despite a period prevalence of up to 28% within the first
• Holocarboxylase synthetase deficiency year of life, atopic dermatitis (AD) does not usually occur
• Amino acid disorders (maple syrup urine disease, methylmalonic in infants younger than 3 months of age and only very
acidaemia) rarely causes severe skin symptoms or even erythroderma
in neonates [6]. If present, however, acute lesions are char­
acterized by widespread pruritic erythematous papules
and patches, often with marked exudation. Infantile AD
typically involves the scalp, the face, the trunk and the
erythroderma. For practical reasons, these can be classified
extensor surfaces of the extremities whereas, in contrast
into cutaneous disorders, infectious diseases, immunode­
to infantile psoriasis and seborrhoeic dermatitis, the nap­
ficiency disorders, drug reactions and inborn errors of
kin area is frequently spared (Fig. 10.2) [7]. Moreover, in
metabolism (Box 10.1).
contrast to more serious causes of neonatal erythroderma,
newborns affected by AD without concurrent food allergy
­References
1 Ott H, Hutten M, Baron JM et al. Neonatal and infantile erythrodermas.
usually thrive well and do not exhibit persisting alopecia
J Dtsch Dermatol Ges 2008;6:1070–85. or additional clinical signs of extracutaneous pathology
2 Sarkar R, Basu S, Sharma RC. Neonatal and infantile erythrodermas. (e.g. lymphadenopathy, diarrhoea, atypical infections).
Arch Dermatol 2001;137:822–3. The lesions respond rapidly to topical therapy with cor­
3 Al‐Dhalimi MA. Neonatal and infantile erythroderma: a clinical and
follow‐up study of 42 cases. J Dermatol 2007;34:302–7. ticosteroids, calcineurin inhibitors and emollients. Due to
4 El Euch D, Zeglaoui F, Benmously R et  al. Erythroderma: a clinical a considerable clinical overlap and rather uncharacteristic
study of 127 cases and review of the literature. Exog Dermatol 2003;2: histological findings upon skin biopsy, differentiation of
234–9.
5 Pruszkowski A, Bodemer C, Fraitag S et  al. Neonatal and infantile
erythrodermic AD from other causes of neonatal erythro­
erythrodermas: a retrospective study of 51 patients. Arch Dermatol derma is challenging. Although elevated total serum IgE
2000;136:875–80. levels indicate early‐onset AD, they are nonspecific and
124 Section 2  Skin Disorders of the Neonate and Young Infant
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Fig. 10.2  Erythroderma sparing the napkin (diaper) area in an otherwise


healthy male infant with atopic dermatitis.

may also be associated with more serious disorders such


as Omenn syndrome, Wiskott–Aldrich syndrome or
Netherton syndrome. Conversely, normal total serum IgE
levels do not rule out neonatal or infantile AD due to a
high prevalence of the intrinsic variant of AD in this age
group [8]. Likewise, recent prospective birth cohort
studies have disclosed that the presence of food‐specific
IgE is not a reliable indicator of infantile AD (as was
previously postulated), because it can be detected equally
often in affected and healthy children [9–11]. Hence, if
severe AD with concurrent cow’s milk allergy is sus­ Fig. 10.3  Noncongenital psoriasis with widespread lesions in the napkin
pected in a non‐breastfed neonate with erythroderma, the and abdominal area of a male infant revealing sharply demarcated,
patient should first be fed with an extensively hydrolysed infiltrated, bright red plaques.
milk‐ or amino acid‐based formula for a limited period of
time, e.g. 2 weeks. Likewise, in breastfed infants, maternal
avoidance of potential food allergens may be considered, ichthyosiform erythroderma, severe AD or Netherton
but only after adequate dietary counselling. If a clear syndrome. Hence, correct diagnosis may require skin
amelioration is achieved by allergen avoidance, the infant biopsy that reveals a psoriasiform reaction pattern with
should undergo an oral food challenge (OFC) with the laminated parakeratosis, elongated rete ridges often con­
incriminated allergen. If symptoms recur upon OFC, the taining neutrophils and little or no dermal lymphocytic
diagnosis of food allergy in the context of severe AD is infiltrate while LEKTI staining is negative [15]. Once the
highly probable. diagnosis of psoriasis is safely established, systemic
treatment with acitretin (0.5–1 mg/kg/day) has proved to
Psoriasis (see Chapter 30) be well‐tolerated and effective even in young infants and
Up to 37.5% of children suffering from plaque psoriasis children with GPP [16–18].
and up to 50% of children with pustular psoriasis develop
symptoms during the first year of life [12]. Neonatal or Pityriasis rubra pilaris (see Chapter 32)
congenital psoriasis is, however, very rare. Neonatal Pityriasis rubra pilaris (PRP) represents a rare papulo­
erythroderma due to psoriasis is likely to evolve into squamous disorder of unknown origin which can mani­
generalized pustular psoriasis (GPP). Erythrodermic fest at any age. In the first month of life, PRP is almost
GPP requires a thorough diagnostic work‐up in order to exclusively encountered in those extremely rare patients
exclude other serious differential diagnoses, especially with congenital disease onset. These individuals may be
infections with herpes simplex virus (HSV), varicella zoster affected by familial PRP, which accounts for about 5% of
virus (VZV), Staphylococcus aureus or Candida spp. More­ all PRP cases and has recently been shown to be due to
over, GPP demands immediate treatment to avoid poten­ activating mutations in CARD14, the gene encoding
tially severe complications such as sterile lytic bone caspase recruitment domain family member 14, a known
lesions, bacterial superinfection or sepsis [13,14]. activator of nuclear factor kappa B (NF‐κB) signalling [19].
Infantile psoriasis often starts with an unusually severe Clinically resembling atypical juvenile PRP (Griffiths
napkin rash that is better demarcated and brighter red than classification type V), skin lesions consist of widespread
that of seborrhoeic dermatitis (Fig.  10.3). Subse­ quently, follicular, keratotic papules and erythematous patches
rapid dissemination leads to generalized erythemato­ coalescing into erythroderma. As in typical juvenile PRP,
squamous skin lesions, but in neonates and young infants neonates display salmon‐coloured erythema with islands
psoriatic erythroderma may also mimic nonbullous of unaffected skin (‘nappes claires’) (Fig.  10.4) whereas
Chapter 10  Differential Diagnosis of Neonatal Erythroderma 125

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
(a)

Fig. 10.4  Atypical juvenile pityriasis rubra pilaris in a male infant with
follicular, erythematous papules and patches coalescing into erythroderma.
Of note, the typically salmon‐coloured erythema reveals islands of
unaffected skin (‘nappes claires’) and marked palmar hyperkeratosis.

palmoplantar keratoderma is usually discrete or com­


pletely lacking. Histological features of neonatal PRP have
only been reported casuistically and consist of alternating
ortho‐ and parakeratosis with lipping of follicular ostia
and associated follicular plugging [20,21].

Genodermatoses
Neonatal erythroderma is a presenting sign of several
(b)
monogenic skin diseases, particularly ectodermal dyspla­
sia and hereditary disorders of keratinization. The latter Fig. 10.5  (a) Collodion baby phenotype in a female neonate with
nonbullous congenital erythroderma.Source: Ott et al. 2008 [24].
include nonsyndromic ichthyoses with exclusively cutane­
Reproduced with permission of John Wiley & Sons. (b) Same patient as in
ous symptoms as well as syndromic ichthyoses associated (a) who reveals ‘self‐improving’ ichthyosiform erythroderma with fine
with potentially severe metabolic, neurological, ophthal­ scaling and discrete palmoplantar hyperkeratosis after shedding of the
mological or other abnormalities [22,23]. If suspicion arises, collodion membrane.
skin biopsy is mandatory and further diagnostic steps
(e.g. molecular genetics, immunohistochemistry) should
optimally be coordinated in cooperation with national ischaemia due to constricting skin bands [26]. At first
reference centres such as the Network for Ichthyoses and presentation, it is important to rule out syndromic
Related Keratinization Disorders: NIRK (https://www. ­differential diagnoses of the collodion baby phenotype
medizin.uni‐muenster.de/nirk/network‐for‐ichthyoses‐ such as Sjögren–Larsson syndrome, Netherton syndrome,
and‐related‐keratinization‐disorders/willkommen/) or the trichothiodystrophy, Chanarin–Dorfman syndrome or
Centre de référence des maladies rares de la peau et des type 2 Gaucher disease [25].
muqueuses d’origine génétique, MAGEC (http://www. Autosomal dominant epidermolytic ichthyosis (EI, OMIM
maladiesrares‐necker.aphp.fr/magec/), for example. #113800), also designated as bullous congenital ichthy­
osiform erythroderma or epidermolytic hyperkeratosis,
Nonsyndromic ichthyoses (see Chapter 129) belongs to the group of keratinopathic ichthyoses and is
Nonbullous congenital ichthyosiform erythroderma (NCIE, caused by mutations in genes encoding the suprabasal
OMIM #242100) is an autosomal recessive keratinization keratins 1 and 10 (KRT1, KRT10) [27]. Clinical presenta­
disorder that is caused by mutations in various genes tion varies greatly, but affected patients usually suffer
(ABCA12, ALOXE3, ALOX12B, CERS3, CYP4F22, LIPN, from severe blistering immediately after birth, sometimes
NIPAL4/ICHTHYIN, PNPLA1, TGM1) [22]. As a clinical with widespread erosions, accompanied by variously
hallmark, up to 90% of affected patients present with a severe ichthyosiform erythroderma. During the following
collodion membrane directly post partum which is shed weeks, hystrix‐like hyperkeratoses appear. Interestingly,
during the neonatal period, revealing erythroderma with marked palmoplantar hyperkeratosis is characteristically
generalized fine scaling (Fig. 10.5a and b) [24,25]. While encountered in patients with KRT1 mutations, although
systemic symptoms are usually absent, further cutaneous individuals with EI due to KRT10 mutations may also be
symptoms can include ectropion or eclabium, obstruction of affected. Initially, other bullous disorders of neonatal onset
the external auditory canal, scarring alopecia or peripheral have to be considered as possible differential diagnoses,
126 Section 2  Skin Disorders of the Neonate and Young Infant

especially epidermolysis bullosa hereditaria, superficial oligodendrocytes, retinal cells), resulting in the character­
epidermolytic ichthyosis (formerly ichthyosis bullosa istic clinical triad of ichthyosis, psychomotor retardation
Siemens), staphylococcal scalded skin syndrome and and spastic diplegia. In neonates and young infants, SLS
diffuse cutaneous mastocytosis [28]. Affected family tends to manifest as ichthyosiform erythroderma while
members may also facilitate diagnosis, although a nega­ the full clinical picture is usually not apparent until early
tive family history does not rule out EI because >50% childhood. Definitive diagnosis can be established by the
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

of patients have spontaneous mutations that are readily assessment of FALDH activity in cultured fibroblasts,
detected in EDTA blood samples or skin biopsy specimens. the characteristic elevation of leukotriene B4 (LTB4) in
urine specimens and, finally, targeted sequencing of the
Syndromic ichthyoses (see Chapter 129) ALDH3A2 gene. Pathognomonic crystalline deposits
Autosomal recessive Chanarin–Dorfman syndrome (CDS, within the retina (‘glistening dots’) and reduced levels of
OMIM #275630), also referred to as neutral lipid storage mean foveal macular pigment are seen upon funduscopy
disease with ichthyosis, represents a multiorgan disorder and macular pigment reflectometry, respectively [33,34].
elicited by mutations in the CGI85 (ABDH5) gene. These
induce pathological alterations in the metabolism of Disorders of epidermal homeostasis
endogenous triglycerides leading to the accumulation of The autosomal recessive Netherton syndrome (NS, OMIM
neutral lipids in multiple cell types, particularly leuko­ #256500), a syndromic ichthyosis, is caused by inactivat­
cytes, myocytes, hepatocytes, fibroblasts and keratino­ ing mutations in the epidermal serine protease inhibitor
cytes. The resulting cutaneous phenotype resembles Kazal‐type 5 (SPINK5) gene which codes for the lym­
other forms of congenital ichthyosiform erythroderma phoepithelial Kazal‐type 5 related inhibitor (LEKTI). As a
only occasionally associated with the presenting sign of result, severe defects in epidermal barrier function and a
a collodion membrane. Systemic manifestations in neo­ persistent inflammatory reaction lead to severe neonatal
nates and infants may consist of liver involvement such erythroderma (Fig. 10.6) while the typical polycyclic, ser­
as hepatosteatosis or cirrhosis in 70% of patients, whereas piginous plaques with erythematous borders and double‐
ocular symptoms (e.g. cataract, nystagmus, myopia) and edged scaling (ichthyosis linearis circumflexa) usually do
sensorineural deafness can occur in about 40% and 25% of not manifest before early childhood. Affected neonates
patients, respectively. Additionally, developmental delay and infants often have bacterial skin infections which are
and growth retardation are frequently observed in CDS also due to an underlying immunodeficiency associated
patients. Lipid vacuoles are present in numerous organs
and can be seen in a skin biopsy specimen or in granulo­
cytes and monocytes of a peripheral blood smear on
Pappenheim staining (Jordan sign) [29,30].
Also known as Conradi–Hünermann–Happle syndrome,
type 2 chondrodysplasia punctata (CDPX2, OMIM #302960)
is an X‐linked dominant disorder caused by mutations in
the emopamil‐binding protein (EBP) gene. These entail
increased serum levels of pathological cholesterol metabo­
lites due to an impaired function of 8‐7‐sterol isomerase.
CDPX2 is nearly always fatal for male fetuses. In contrast,
affected female neonates suffer from severe ichthyosiform
erythroderma immediately post partum which is usually
associated with hyperkeratosis and scaling following
Blaschko’s lines. Extracutaneous features which may not
become apparent until later in childhood include short stat­
ure and kyphoscoliosis, hearing loss, sectorial cataracts,
punctate bone calcifications and mild‐to‐moderate mental
retardation. Moreover, asymmetrical shortening of the
limbs is observed in up to 80% of affected girls. While
elevated serum levels of 8‐dehydrocholesterol and a dimin­
ished stratum granulosum with cytoplasmic vacuoles upon
skin biopsy hint at CDPX2, definitive diagnosis is reached
by molecular genetic analysis of the EBP gene [31,32].
As a severe neurocutaneous disease, the autosomal
recessive Sjögren–Larsson syndrome (SLS, OMIM #270200)
is caused by mutations in the ALDH3A2 gene that codes
for fatty aldehyde dehydrogenase (FALDH), an enzyme
of crucial importance for cutaneous and extracutaneous
lipid metabolism. Diminished FALDH activity leads
to  a pathological accumulation of essential fatty acids Fig. 10.6  Severe ichthyosiform erythroderma, hypotrichosis and failure
in  the membrane of various cell types (keratinocytes, to thrive in an infant with Netherton syndrome.
Chapter 10  Differential Diagnosis of Neonatal Erythroderma 127

with an immature natural killer cell phenotype and a Severe dermatitis, multiple allergies and metabolic wasting
decrease of nonswitched memory B cells or CD27+ mem­ syndrome (SAM, OMIM #615508) has recently been shown
ory B cells [35]. The clinical distinction of NS from other to be caused by homozygous mutations in DSG1, the
causes of ichthyosiform erythroderma is facilitated by desmoglein‐1‐encoding gene (DSG1). These mutations
microscopy of hair shafts (scalp hair, eyebrows, eyelashes) lead to desmosome dysfunction, impaired cell adhesion
revealing trichorrhexis invaginata (‘bamboo hair’) and and aberrant epidermal differentiation. As a consequence,

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
hypotrichosis. Skin biopsy with immunohistochemical affected neonates reveal early‐onset, ichthyosiform eryth­
staining reveals a psoriasiform epidermal hyperplasia roderma, multiple food allergies and a severe failure to
with absence of the stratum granulosum, a mixed perivas­ thrive. Additional clinical features consist of recurrent
cular infiltrate and reduced or absent LEKTI expression. skin or pulmonary infections, hypotrichosis and palmo­
Additionally, total and allergen‐specific IgE levels are fre­ plantar keratoderma, which may help to distinguish SAM
quently elevated while other traits of atopic disease such syndrome from NS and PSS‐B clinically. While histology
as bronchial asthma or allergic rhinitis do not become reveals subcorneal separation associated with acantholysis,
apparent until late infancy or early childhood. Associated a definitive diagnosis can be established by DSG1
systemic symptoms are subject to marked phenotypic sequencing [38].
variability and include hypernatraemic dehydration, failure
to thrive and chronic diarrhoea which are responsible for Ectodermal dysplasia (see Chapter 134)
a fatality rate of up to 20% in the first year of life [35,36]. Erythroderma can also be seen in neonates suffering from
Peeling skin syndrome type B (PSS‐B, OMIM #270300), a ectodermal dysplasias, a heterogenous group of inherited
nonsyndromic ichthyosis, results from loss‐of‐function disorders involving absence or dysplasia of the ectodermal
mutations in CDSN, the gene encoding for corneodes­ appendages. In particular, neonates with ankyloblepha­
mosin. Phenotypically, neonates and infants with PSS‐B ron, ectodermal dysplasia and cleft lip/palate syndrome
develop a clinical picture mimicking NS with ichthyosi­ (AEC, OMIM #106260), a very rare autosomal dominant
form erythroderma, recurrent staphylococcal skin infec­ genodermatosis caused by TP63 mutations, may present
tions, severe pruritus and frequent food anaphylaxis in with widespread erythema. In these patients, neonatal
the context of elevated total and food allergen‐specific erythroderma with extensive erosions and diffuse depig­
serum IgE levels. However, unlike in NS, hair shaft abnor­ mented patches in previously eroded areas can be observed
malities are absent and, even more importantly, PSS‐B besides further clinical signs of ectodermal dysplasia such
patients show increased skin fragility and spontaneous as palmoplantar keratoderma, partial anhidrosis, nail
peeling (Fig. 10.7) corresponding to subcorneal cleavage dystrophy, patchy alopecia and hypodontia [39,40].
upon skin biopsy [37,38].
Other skin diseases
Diffuse cutaneous mastocytosis (see Chapter 92)
As the rarest subtype of cutaneous mastocytosis (CM),
diffuse cutaneous mastocytosis (DCM) affects up to 5% of
CM patients and usually manifests with a peculiar type
of neonatal erythroderma either at birth or shortly there­
after. Characteristically, infiltration of mast cells through­
out the entire cutis leads to widespread, yellow‐reddish
erythema accompanied by extensive bulla formation.
Other skin manifestations attributable to mediator release
from mast cells comprise flushing, urticaria or pachyder­
mia. Extracutaneous symptoms occur in virtually all
affected patients and may consist of abdominal pain,
diarrhoea, headache or even anaphylaxis. Hence, erythro­
dermic neonates with DCM require interdisciplinary
management including bone marrow biopsy if suspicion
of systemic mastocytosis arises, as in patients with hepat­
osplenomegaly, lymphadenopathy, abnormal blood count
and/or serum tryptase levels above 100  ng/mL [41–43].

Scabies (see Chapter 59)


Neonatal erythroderma due to infestation with the mite
Sarcoptes scabiei hominis has only been very rarely reported.
In these cases, physical examination revealed scaly
erythroderma, multiple micropustules and papules on
the trunk, extremities, scalp, face and the palmoplantar
region with secondary eczematization and bacterial super­
Fig. 10.7  Severe erythroderma and spontaneous, superficial exfoliation in infection. Peripheral blood cell counts showed marked
a newborn with peeling skin syndrome type B. eosinophilia. In a recent multicentre trial, anamnestic
128 Section 2  Skin Disorders of the Neonate and Young Infant

relapse, the presence of skin nodules and the involvement 28 Avril M, Riley C. Management of epidermolytic ichthyosis in the
newborn. Neonatal Netw 2016;35:19–28.
of soles and scalp were found to be highly characteristic
29 Emre S, Unver N, Evans SE et  al. Molecular analysis of Chanarin‐
signs of scabies infestation in infants and young children. Dorfman syndrome (CDS) patients: Identification of novel mutations
Extracutaneous features such as hepatosplenomegaly or in the ABHD5 gene. Eur J Med Genet 2010;53:141.
lymphadenopathy were not observed [44–46]. 30 Nur BG, Gencpinar P, Yuzbasıoglu A et  al. Chanarin‐Dorfman
syndrome: genotype‐phenotype correlation. Eur J Med Genet 2015;
58:238–42.
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

­References 31 Kolb‐Maurer A, Grzeschik KH, Haas D et  al. Conradi‐Hunermann‐


1 Al‐Dhalimi MA. Neonatal and infantile erythroderma: a clinical and Happle syndrome (X‐linked dominant chondrodysplasia punctata)
follow‐up study of 42 cases. J Dermatol 2007;34:302–7. confirmed by plasma sterol and mutation analysis. Acta Derm
2 El Euch D, Zeglaoui F, Benmously R et  al. Erythroderma: a clinical Venereol 2008;88:47–51.
study of 127 cases and review of the literature. Exog Dermatol 32 Cañueto J, Girós M, Ciria S et al. Clinical, molecular and biochemical
2003;2:234–9. characterization of nine Spanish families with Conradi‐Hünermann‐
3 Pruszkowski A, Bodemer C, Fraitag S et  al. Neonatal and infantile Happle syndrome: new insights into X‐linked dominant chondro­
erythrodermas: a retrospective study of 51 patients. Arch Dermatol dysplasia punctata with a comprehensive review of the literature.
2000;136:875–80. Br J Dermatol 2012;166:830–8.
4 Foley P, Zuo Y, Plunkett A et al. The frequency of common skin condi­ 33 Nagappa M, Bindu PS, Chiplunkar S et al. Child Neurology: Sjögren‐
tions in preschool‐aged children in Australia: seborrheic dermatitis Larsson syndrome. Neurology 2017;88:e1–4.
and pityriasis capitis (cradle cap). Arch Dermatol 2003;139:318–22. 34 Fuijkschot J, Theelen T, Seyger MM et al. Sjögren‐Larsson syndrome
5 Alexopoulos A, Kakourou T, Orfanou I et al. Retrospective analysis of in clinical practice. J Inherit Metab Dis 2012;35:955–62.
the relationship between infantile seborrheic dermatitis and atopic 35 Hannula‐Jouppi K, Laasanen SL, Ilander M et al. Intrafamily and
dermatitis. Pediatr Dermatol 2014;31:125–30. interfamilial phenotype variation and immature immunity in
6 Draaisma E, Garcia‐Marcos L, Mallol J et  al.; EISL Study Group. A patients with Netherton Syndrome and Finnish SPINK5 Founder
multinational study to compare prevalence of atopic dermatitis in the Mutation. JAMA Dermatol 2016;152:435–42.
first year of life. Pediatr Allergy Immunol 2015;26:359–66. 36 Renner ED, Hartl D, Rylaarsdam S et al. Comel‐Netherton syndrome
7 Deleuran M, Vestergaard C. Clinical heterogeneity and differential defined as primary immunodeficiency. J Allergy Clin Immunol
diagnosis of atopic dermatitis. Br J Dermatol 2014;170(suppl 1):2–6. 2009;124:536–43.
8 Ott H, Stanzel S, Ocklenburg C et al. Total serum IgE as a parameter 37 Oji V, Eckl KM, Aufenvenne K et al. Loss of corneodesmosin leads
to differentiate between intrinsic and extrinsic atopic dermatitis in to severe skin barrier defect, pruritus, and atopy: unraveling the
children. Acta Derm Venereol 2009;89:257–61. peeling skin disease. Am J Hum Genet 2010;87:274–81.
9 Ballardini N, Bergström A, Wahlgren CF et  al. IgE antibodies in 38 Samuelov L, Sprecher E. Peeling off the genetics of atopic dermatitis‐
relation to prevalence and multimorbidity of eczema, asthma, and like congenital disorders. J Allergy Clin Immunol 2014;134:808–15.
rhinitis from birth to adolescence. Allergy 2016;71:342–9. 39 Berk DR, Crone K, Bayliss SJ. AEC syndrome caused by a novel p63
10 Eller E, Kjaer HF, Host A et al. Food allergy and food sensitization in mutation and demonstrating erythroderma followed by extensive
early childhood: results from the DARC cohort. Allergy 2009;64:1023–9. depigmentation. Pediatr Dermatol 2009;26:617–18.
11 Eigenmann PA. Clinical features and diagnostic criteria of atopic der­ 40 Yoo J, Berk DR, Fabre E et  al. Ankyloblepharon‐ectodermal dyspla­
matitis in relation to age. Pediatr Allergy Immunol 2001;12(Suppl sia‐clefting (AEC) syndrome with neonatal erythroderma: report of
14):69–74. two cases. Int J Dermatol 2007;46:1196–7.
12 Burden‐Teh E, Thomas KS, Ratib S et al. The epidemiology of child- 41 Méni C, Bruneau J, Georgin‐Lavialle S et al. Paediatric mastocytosis:
hood psoriasis: a scoping review. Br J Dermatol 2016;174:1242–57. a systematic review of 1747 cases. Br J Dermatol 2015;172:642–51.
13 Lehman JS, Rahil AK. Congenital psoriasis: case report and literature 42 Heide R, Zuidema E, Beishuizen A et  al. Clinical aspects of diffuse
review. Pediatr Dermatol 2008;25:332–8. cutaneous mastocytosis in children: two variants. Dermatology
14 de Oliveira ST, Maragno L, Arnone M et al. Generalized pustular pso­ 2009;219:309–15.
riasis in childhood. Pediatr Dermatol 2010;27:349–54. 43 Lange M, Niedoszytko M, Nedoszytko B et  al. Diffuse cutaneous
15 Leclerc‐Mercier S, Bodemer C, Bourdon‐Lanoy E et  al. Early skin mastocytosis: analysis of 10 cases and a brief review of the literature.
biopsy is helpful for the diagnosis and management of neonatal and J Eur Acad Dermatol Venereol 2012;26:1565–71.
infantile erythrodermas. J Cutan Pathol 2009;37:249–55. 44 Haim A, Grunwald MH, Kapelushnik J et  al. Hypereosinophilia in
16 Chao PH, Cheng YW, Chung MY. Generalized pustular psoriasis in a red scaly infants with scabies. J Pediatr 2005;146:712.
6‐week‐old infant. Pediatr Dermatol 2009;26:352–4. 45 Hortala M, Vicente A, Abellaneda C et  al. Erythroderma in a 1‐

17 Haug V, Benoit S, Wohlleben M, Hamm H. Annular pustular psoriasis month‐old boy. Eur J Pediatr 2007;166:979–80.
in a 14‐month‐old girl: a therapeutic challenge. J Dermatolog Treat 46 Boralevi F, Diallo A, Miquel J et al; Groupe de Recherche Clinique
2017:28:520–2. en Dermatologie Pédiatrique. Clinical phenotype of scabies by age.
18 Posso‐De Los Rios CJ, Pope E, Lara‐Corrales I. A systematic review of Pediatrics 2014;133:e910–16.
systemic medications for pustular psoriasis in pediatrics. Pediatr
Dermatol 2014;31:430–9.
19 Fuchs‐Telem D, Sarig O, van Steensel MA et al. Familial pityriasis ­Infections and toxicities
rubra pilaris is caused by mutations in CARD14. Am J Hum Genet
2012;91:163–70. Staphylococcal scalded skin syndrome
20 Thomson MA, Moss C. Pityriasis rubra pilaris in a mother and two (see Chapter 37)
daughters. Br J Dermatol 2007;157:202–4.
21 Allison DS, El‐Azhary RA, Calobrisi SD, Dicken CH. Pityriasis rubra Staphylococcal scalded skin syndrome (SSSS), also known
pilaris in children. J Am Acad Dermatol 2002;47:386–9. as Ritter disease, staphylogenic Lyell syndrome or
22 Takeichi T, Akiyama M. Inherited ichthyosis: Non‐syndromic forms. pemphigus acutus neonatorum, is caused by phage group
J Dermatol 2016;43:242–51.
2 staphylococci that produce the exfoliative toxins A or B
23 Yoneda K. Inherited ichthyosis: Syndromic forms. J Dermatol 2016;
43:252–63. (ETA, ETB). As epidermotropic serine proteases, ETA and
24 Ott H, Hutten M, Baron JM et al. Neonatal and infantile erythroder­ ETB induce subcorneal cleavage by proteolysis of the
mas. J Dtsch Dermatol Ges 2008;6:1070–85. desmosomal adhesion protein desmoglein‐1, thereby
25 Prado R, Ellis LZ, Gamble R et al. Collodion baby: an update with a
focus on practical management. J Am Acad Dermatol 2012;67:
causing exfoliative dermatitis.
1362–74. The disease tends to affect neonates, infants and young
26 Craiglow BG. Ichthyosis in the newborn. Semin Perinatol 2013;37: children because of immature renal toxin clearance
26–31. and low serum levels of toxin‐neutralizing antibodies.
27 Hotz A, Oji V, Bourrat E et al. Expanding the clinical and genetic spec­
trum of KRT1, KRT2 and KRT10 mutations in keratinopathic ichthyosis. Following a superficial staphylococcal infection (e.g.
Acta Derm Venereol 2016;96:473–8. purulent conjunctivitis, omphalitis), patients exhibit
Chapter 10  Differential Diagnosis of Neonatal Erythroderma 129

paronychia has also frequently been reported. Unlike


neonatal candidiasis, which is acquired during passage
through the birth canal, the oral cavity and nappy area are
usually spared.
In neonates with CCC, further diagnostic and therapeu­
tic measures are determined by the patient’s gestational

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
stage. In full‐term infants the disease is generally benign,
running a self‐limiting course without complications.
Oral fluconazole (3–6 mg/kg/day) is a safe option; in
milder cases, the condition can be treated with topical
agents alone (clotrimazole, miconazole) under careful
clinical surveillance. In contrast, premature infants with
CCC, especially those born before 27 weeks’ gestation
and/or a birthweight of <1000 g, may develop invasive
candidal infections that are associated with a mortality
rate of up to 40%. Therefore, a thorough diagnostic work‐up
Fig. 10.8  Widespread erythema with flaccid, rapidly eroding bullae in a should be performed in these high‐risk patients includ­
male neonate with staphylococcal scalded skin syndrome (SSSS). ing blood, urine and cerebrospinal fluid (CSF) cultures.
Depending on test results, systemic antimycotic therapy
with liposomal amphotericin B or fluconazole should be
flexural and facial erythema which may eventually initiated under close clinical surveillance. Moreover, vari­
coalesce into erythroderma. Other symptoms include ous transient and infectious pustular diseases of the new­
extremely tender skin, fever and a rapid decline of the born must be considered as possible differential diagnoses
infant’s general condition. Sepsis‐like symptoms are not (see Chapters 6 and 11). In particular, Listeria monocytogenes
uncommon in this initial erythematous stage of the infection should be excluded as it may also present with
disease. Shortly thereafter, large, flaccid, rapidly eroding vesiculopustular eruptions in the n ­ eonate and whitish
bullae appear over the entire body (Fig. 10.8). A positive macules on the umbilical cord and placenta [4,5].
Nikolsky sign can regularly be elicited in this exfoliative
stage of SSSS, whereas the mucous membranes are usually ­References
spared. 1 Handler MZ, Schwartz RA. Staphylococcal scalded skin syndrome:
Neonates and especially preterm infants with sus­ diagnosis and management in children and adults. J Eur Acad
Dermatol Venereol 2014;28:1418–23.
pected SSSS should be hospitalized, especially because 2 Aalfs AS, Oktarina DM, Diercks GF et al. Staphylococcal scalded skin
there is a reported mortality risk of about 2.5–11% in this syndrome: loss of desmoglein 1 in patient skin. Eur J Dermatol
age group [1]. If exfoliation is extensive, affected neonates 2010;20:451–6.
3 Saida K, Kawasaki K, Hirabayashi K et al. Exfoliative toxin A staphy­
and infants should be treated with nonadhesive, polyure­
lococcal scalded skin syndrome in preterm infants. Eur J Pediatr
thane wound dressings in analogy to burn care or epider­ 2015;174:551–5.
molysis bullosa patients. In addition, after microbiological 4 Darmstadt GL, Dinulos JG, Miller Z. Congenital cutaneous candidia­
samples have been obtained, systemic antibiotic treatment sis: clinical presentation, pathogenesis, and management guidelines.
Pediatrics 2000;105:438–44.
is warranted, preferably consisting of penicillinase‐resistant 5 Aguin TJ, Sobel JD. Vulvovaginal candidiasis in pregnancy. Curr Infect
penicillins. For definitive diagnosis, isolated Staph. aureus Dis Rep. 2015;17:462.
strains may be sent for molecular genetic analysis of the
genes sea and seb. If other blistering or exfoliative skin
disorders such as toxic epidermal necrolysis, epidermolysis
­Adverse drug reactions
bullosa hereditaria or ichthyosis bullosa of Siemens cannot Stevens–Johnson syndrome and toxic epidermal
be ruled out with certainty, a lesional skin biopsy should necrolysis (see Chapters 66 and 67)
be taken. Subcorneal acantholysis is the characteristic Stevens–Johnson syndrome (SJS) and toxic epidermal
feature of SSSS and distinguishes it from toxic epidermal necrolysis (TEN) represent a spectrum of severe, usually
necrolysis with subepidermal blistering [1–3]. drug‐induced skin changes that arise from massive apop­
tosis of epidermal keratinocytes and an interaction of
Congenital cutaneous candidiasis (see Chapter 7) additional pathogenetic factors which are discussed more
Candidal chorioamnionitis, which occurs more often in thoroughly in Chapters 66 and 67. Both entities are
pregnant women who have had cerclage or placement of extremely rare during the first 2 months of life and only a
an intrauterine pessary, can manifest immediately post few cases of neonatal or very early infantile TEN have
partum as congenital cutaneous candidiasis (CCC). been published so far.
Whitish macules appear on the placenta and umbilical These neonates and young infants revealed irritability,
cord whereas the affected neonate has a maculopapular, poor feeding and hypothermia as nonspecific prodromal
sometimes pustular or bullous exanthema that may rap­ symptoms before the onset of cutaneous lesions. As in adults,
idly evolve into exfoliative erythroderma (‘white dots on skin involvement consisted of rapidly coalescing, tender,
the placenta, red dots on the baby’). Characteristically, the erythematous macules progressing to extensive exfoliative
palmoplantar and umbilical regions are involved initially; erythroderma with pluriorificial mucosal involvement.
130 Section 2  Skin Disorders of the Neonate and Young Infant

Histopathological analysis of lesional skin revealed ­Immunological disorders (see Chapters


characteristic subepidermal blistering and complete 53 and 56)
necrosis of the epidermis, whereas the main differential
diagnosis SSSS was ruled out by the absence of subcorneal Omenn syndrome
splitting. In contrast, concurrent systemic infection with Omenn syndrome (OS, OMIM #603554) represents a
Klebsiella pneumoniae could be diagnosed in two individ­ genetically heterogeneous, autosomal recessive primary
immunodeficiency characterized by lymphadenopathy,
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

uals, whereas Candida albicans/dubliensis, Escherichia coli


and coagulase‐negative streptococci were found to be hepatosplenomegaly, elevated serum IgE and eosinophilia.
relevant pathogens in other affected individuals. Thus, It is caused predominantly by mutations in the recombi­
the majority of patients had received multiple antimicro­ nation activating genes 1 and 2 (RAG‐1/RAG‐2), the
bial and other medications prior to the onset of blistering interleukin‐7Rα chain or the nonhomologous end‐joining
which hampered the identification of a single culprit factor Artemis, which interfere with somatic diversifica­
drug. Unfortunately, all reported patients died despite tion of T‐ and B‐cell receptor‐encoding genes. This leads
the limitation of applied medications to vitally indicated to oligoclonal expansion of autoreactive T cells and a
drugs and intensive care support [1–4]. However, whether marked reduction in circulating B cells (‘T+B‐ SCID’,
neonatal TEN generally carries a fatal prognosis and ‘leaky SCID’).
whether concurrent sepsis is a relevant co‐factor or even Affected neonates and infants initially reveal severely
the main prerequisite for TEN in newborns remains to be pruritic eczematous lesions resembling atopic dermatitis,
elucidated. which can coalesce to form erythroderma, pachydermia,
onychodystrophy and alopecia while eyebrows and eye­
Red man syndrome lashes may appear normal (Fig.  10.9a and b). Over the
Rapid intravenous administration (<60 min) of vancomy­ course of the disease, the majority of patients develop
cin and other antimicrobial agents, such as rifampicin, lymphadenopathy, hepatosplenomegaly, profuse diar­
teicoplanin, ceftriaxone, amphotericin B or ciprofloxacin, rhoea and failure to thrive. Furthermore, patients often
can lead to nonspecific histamine release in up to 14% of present in a reduced general state with hypothermia,
treated patients. Within seconds to several minutes after hypernatraemic dehydration and clinical signs of bacte­
the initiation of therapy, patients of any age may develop rial infection, particularly pneumonia, or even sepsis.
generalized flushing, with or without anaphylactoid More than 90% of children show a massive increase in
symptoms, and consecutive transient erythroderma giving total serum IgE levels and nearly all patients display
patients a ‘red man’ appearance. Studies of vancomycin marked peripheral eosinophilia. Lesional skin biopsies
have also shown that the most severe reactions tend usually show epithelial hyperplasia, spongiosis, focal
to occur in younger patients, particularly children. Further­ basal vacuolation and parakeratosis as well as an inflam­
more, concomitant application of further histamine liber­ matory infiltrate in the upper dermis mainly consisting of
ators such as opioid analgesics or radiological contrast lymphocytes with few interspersed eosinophils.
media may enhance the risk of vancomycin‐induced red OS is known to have a lethal outcome if left untreated,
man syndrome. mostly due to recurrent severe infections. In the presence
The syndrome is easily recognizable, given the short of the characteristic clinical triad of erythroderma, hepat­
interval between drug administration and onset of initial osplenomegaly and lymphadenopathy in conjunction
symptoms. As in other immediate‐type reactions, acute with typical laboratory changes such as marked eosino­
therapy consists of intravenous administration of an philia and elevated total IgE levels, immunomodulatory
antihistamine and, if necessary, epinephrine in patients therapy with ciclosporin (cyclosporine) can be initiated
with circulatory disturbances. As a preventive measure, rapidly to suppress autoreactive T‐cell clones. This has
vancomycin should be given in an adequate dilution been shown to provide quick symptom relief, particularly
and the infusion rate should be less than 10 mg/min. In with regard to the often debilitating pruritus. Nevertheless,
the absence of alternative agents and if vancomycin is curative therapy can only be achieved with bone marrow‐
urgently indicated, desensitization with the culprit drug or umbilical cord blood‐derived haematopoietic stem cell
is possible but requires close clinical surveillance [5,6]. transplantation yielding survival rates that range from
less than 50% (haploidentical donor) to 75% (HLA‐identical
­References
donor) [2–5].
1 de Groot R, Oranje AP, Vuzevski VD, Mettau JW. Toxic epidermal

necrolysis probably due to Klebsiella pneumoniae sepsis. Dermatologica
1984;169:88–90.
Graft‐versus‐host disease
2 Hawk RJ, Storer JS, Daum RS. Toxic epidermal necrolysis in a 6‐week‐ Along with Omenn syndrome, there are several other
old infant. Pediatr Dermatol 1985;2:197–200. severe combined immunodeficiencies (SCID) that are
3 Lohmeier K, Megahed M, Schulte KW et al. Toxic epidermal necrolysis inherited in an autosomal recessive or X‐linked pattern
in a premature infant of 27 weeks’ gestational age. Br J Dermatol
2005;152:150–1.
and are very rare with an estimated incidence of 1 in
4 Islam S, Singer M, Kulhanjian JA. Toxic epidermal necrolysis in a 50 000 to 1 in 500 000 live births. Owing to variable defects
neonate receiving fluconazole. J Perinatol 2014;34:792–4. in both T‐ and B‐cell immunity, classic symptoms such as
5 Myers AL, Gaedigk A, Dai H et al. Defining risk factors for red man profuse diarrhoea, failure to thrive, mucocutaneous can­
syndrome in children and adults. Pediatr Infect Dis J 2012;31:464–8.
6 Wazny LD, Daghigh B. Desensitization protocols for vancomycin
didiasis and orogenital ulcerations can occur as early as
hypersensitivity. Ann Pharmacother 2001;35:1458–64. the first 4 weeks of life.
Chapter 10  Differential Diagnosis of Neonatal Erythroderma 131

Upon analysis of the affected baby’s peripheral blood,


the identification of accessory HLA haplotypes, high
numbers of maternal lymphocytes or an XX genotype in a
male neonate are highly suggestive of GVHD. Interest­
ingly, skin histopathology in neonates with SCID and
maternal engraftment has been reported to differ from

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
that obtained in patients with SCID after bone marrow
transplantation or after transplant for other diseases.
Whereas a vacuolar interface pattern is usually observed
in the latter disorders, GVHD due to maternal engraft­
ment is characterized by psoriasiform epidermal hyper­
plasia, dermal lymphocytic infiltrates, parakeratosis and
variable spongiosis [8].
Given the otherwise grave prognosis, haematopoietic
(a) stem cell transplantation should be performed promptly,
as it allows full recovery in 52% (HLA‐different, nonre­
lated donors) to 92% (HLA‐identical, related donors) of
patients [6].

Other primary immunodeficiencies


While OS and GVHD due to maternal engraftment are
regularly associated with neonatal erythroderma, other
primary immunodeficiencies only occasionally induce
widespread erythema in newborns or very young
infants.
For example, patients with DiGeorge syndrome (DGS,
OMIM #188400), which is caused by a hemizygous
deletion of chromosome 22q11.2, suffer from conotrun­
cal cardiac defects, hypoparathyroidism and immune
dysfunction due to thymic hypoplasia. Additionally,
affected infants and children frequently develop eczem­
atous skin lesions closely resembling those of atopic
dermatitis. Never­theless, generalized eczematous erup­
tions have only very rarely been observed in newborns
with DGS [9,10].
Wiskott–Aldrich syndrome (WAS, OMIM #301000), an
X‐linked immunodeficiency with thrombocytopenia,
recurrent infections and secondary autoimmune disorders,
is associated with recalcitrant eczematous dermatitis in
nearly 90% of affected patients. However, only very few
WAS cases have been reported to present with neonatal
erythroderma [11–13]. Besides inflammatory skin lesions
and recurrent cutaneous infections, petechiae and ecchy­
(b) mosis in a boy with eczema should raise the suspicion
Fig. 10.9  (a) Ichthyosiform erythroderma, pachydermia and alopecia in a of WAS [14,15].
female infant with Omenn syndrome. (b) Same patient as in (a) after The immune dysregulation, polyendocrinopathy, enteropathy,
5 days of incubator care, intravenous rehydration and topical therapy with
X‐linked syndrome (IPEX, OMIM #304790) represents a
panthenol ointment.Source: Ott et al. 2008 [1]. Reproduced with
permission of John Wiley & Sons.
very rare disorder caused by mutations in the FOXP3
gene, the master transcriptional regulator for the devel­
opment and function of CD4+ regulatory T cells. IPEX
In addition, up to 50% of patients with T‐ SCID have patients reveal disrupted immune homeostasis resulting
maternal T cells in the peripheral blood leading to graft‐ in early‐onset autoimmunity with neonatal diabetes mel­
versus‐host‐disease (GVHD) in nearly 60% of these cases. litus and hypothyroidism, recurrent infections, cytope­
Affected infants may develop alopecia and severe neona­ nias and villous atrophy leading to enteropathy and
tal erythroderma with a scaly erythematous rash spread­ severe failure to thrive. Cutaneous symptoms include
ing craniocaudally, often also affecting the palms and eczematous skin lesions in up to 65% of patients that may
soles [6,7]. appear as severe and generalized ichthyosiform dermatitis.
If suspicion arises, a comprehensive immunological Additionally, serum total IgE levels are clearly elevated in
work‐up including differential blood counts, lymphocyte the majority of neonates and infants with IPEX who also
phenotyping and chimerism analysis should be performed. have marked peripheral eosinophilia [16–18].
132 Section 2  Skin Disorders of the Neonate and Young Infant

­References with the disease also have eczematous erythroderma with


1 Ott H, Hutten M, Baron JM et al. Neonatal and infantile erythroder­
marked periorificial involvement and alopecia. IF HSD is
mas. J Dtsch Dermatol Ges 2008;6:1070–85.
2 Cassani B, Poliani PL, Moratto D et al. Defect of regulatory T cells in not included in the neonatal screening programme, rapid
patients with Omenn syndrome. J Allergy Clin Immunol 2010;125: diagnosis and effective therapy may be missed unless
209–16. carboxylase activity is analysed in cultured fibroblasts.
3 Bai X, Liu J, Zhang Z et al. Clinical, immunologic, and genetic charac­
teristics of RAG mutations in 15 Chinese patients with SCID and
On the other hand, if a high clinical index of suspicion is
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Omenn syndrome. Immunol Res 2016;64:497–507. supported by characteristic laboratory parameters such as
4 Caglayan Sozmen S, Isik S, Arikan Ayyildiz Z et al. Cyclosporin treat­ severe ketoacidosis, hyperammonaemia and hypoglycae­
ment improves skin findings in omenn syndrome. Pediatr Dermatol mia, neonatal intensive care including biotin substitution
2015;32:e54–7.
5 Sharapova SO, Guryanova IE, Pashchenko OE et  al. Molecular
at an oral dose of 10 mg daily is mandatory [1–3].
Characteristics, Clinical and Immunologic Manifestations of 11 Methylmalonic acidaemias (MMA, OMIM #251110,
Children with Omenn Syndrome in East Slavs (Russia, Belarus, #243500 and others) represent genetically heterogeneous
Ukraine). J Clin Immunol 2016;36:46–55.
6 Muller SM, Ege M, Pottharst A et al. Transplacentally acquired mater­
disorders of methylmalonate and cobalamin metabolism.
nal T lymphocytes in severe combined immunodeficiency: a study of Patients affected by the infantile, non‐vitamin B12‐responsive
121 patients. Blood 2001;98:1847–51. subtype may appear healthy at birth, but rapidly develop
7 Wahlstrom J, Patel K, Eckhert E et al. Transplacental maternal engraft­ severe symptoms such as muscular hypotonia, respira­
ment and posttransplantation graft‐versus‐host disease in children
with severe combined immunodeficiency. J Allergy Clin Immunol tory distress and encephalopathy which are associated
2017;139:628–33. with marked metabolic ketoacidosis, hyperammonaemia
8 Leclerc‐Mercier S, Bodemer C, Bourdon‐Lanoy E et  al. Early skin and hyperglycinaemia. Cutaneous symptoms are initially
biopsy is helpful for the diagnosis and management of neonatal and
similar to the rash observed in acrodermatitis enteropathica
infantile erythrodermas. J Cutan Pathol 2009;37:249–55.
9 Archer E, Chuang TY, Hong R. Severe eczema in a patient with comprising acrofacial, psoriasiform dermatitis and alope­
DiGeorge’s syndrome. Cutis 1990;45:455–9. cia. However, untreated infants may develop widespread
10 Minakawa S, Nakano H, Takeda H et al. Chromosome 22q11.2 dele­ erythema and, finally, exfoliative erythroderma [4–6].
tion syndrome associated with severe eczema. Clin Exp Dermatol
2009;34:410–11.
Similar acrodermatitis‐like eruptions have been
11 Sarkar R, Basu S, Sharma RC. Neonatal and infantile erythrodermas. encountered in newborns and infants suffering from
Arch Dermatol 2001;137:822–3. maple syrup urine disease (MSUD, OMIM #248600), another
12 Al‐Dhalimi MA. Neonatal and infantile erythroderma: a clinical and organic aminoacidopathy elicited by an impaired
follow‐up study of 42 cases. J Dermatol 2007;34:302–7.
13 Pruszkowski A, Bodemer C, Fraitag S et  al. Neonatal and infantile branched‐chain alpha‐keto acid dehydrogenase complex
erythrodermas: a retrospective study of 51 patients. Arch Dermatol (BCKD). MSUD is characterized by elevated tissue, blood
2000;136:875–80. and urine concentrations of valine, leucine and isoleucine
14 Massaad MJ, Ramesh N, Geha RS. Wiskott‐Aldrich syndrome: a com­
prehensive review. Ann N Y Acad Sci 2013;1285:26–4.
which are responsible for potentially severe symptoms
15 Loyola Presa JG, de Carvalho VO, Morrisey LR et  al. Cutaneous such as respiratory distress, muscular hypotonia and
manifestations in patients with Wiskott‐Aldrich syndrome submit­ seizures. The only effective therapy currently consists of
ted to haematopoietic stem cell transplantation. Arch Dis Child restricted dietary intake of branched‐chain amino acids.
2013;98:304–7.
16 Chen CA, Chung WC, Chiou YY et al. Quantitative analysis of tissue Interestingly, generalized erythematous eruptions have
inflammation and responses to treatment in immune dysregulation, only been described in treated neonates, so far, and have
polyendocrinopathy, enteropathy, X‐linked syndrome, and review of been attributed to extremely low plasma levels of iso­
literature. J Microbiol Immunol Infect 2016;49:775–82.
leucine. Accordingly, neonatal erythroderma resolved
17 Bin Dhuban K, Piccirillo CA. The immunological and genetic basis
of immune dysregulation, polyendocrinopathy, enteropathy, X‐linked after supplementation of the deficient amino acid in the
syndrome. Curr Opin Allergy Clin Immunol 2015;15:525–32. majority of affected patients [7,8].
18 Xavier‐da‐Silva MM, Moreira‐Filho CA, Suzuki E et  al. Fetal‐onset
IPEX: report of two families and review of literature. Clin Immunol
2015;156:131–40.
­References
1 Donti TR, Blackburn PR, Atwal PS. Holocarboxylase synthetase

­Inborn errors of metabolism deficiency pre and post newborn screening. Mol Genet Metab Rep
2016;7:40–4.
(see Chapter 152)
2 Van Hove JL, Josefsberg S, Freehauf C et al. Management of a patient
Case studies have reported a number of hereditary meta­ with holocarboxylase synthetase deficiency. Mol Genet Metab 2008;95:
201–5.
bolic disorders that present with cutaneous symptoms in 3 Tammachote R, Janklat S, Tongkobpetch S et  al. Holocarboxylase
neonates and infants. Of these, only holocarboxylase synthetase deficiency: novel clinical and molecular findings. Clin
synthetase deficiency, methylmalonic acidaemia and Genet 2010;78:88–93.
4 Fraser JL, Venditti CP. Methylmalonic and propionic acidemias: clinical
maple syrup urine disease appear to be frequently asso­ management update. Curr Opin Pediatr 2016;28:682–93.
ciated with neonatal erythroderma. 5 Sasaki M, Aikoh H, Sugai K et  al. Picture of the month. Cutaneous
Holocarboxylase synthetase deficiency (HSD, OMIM lesions associated with isoleucine deficiency. Arch Pediatr Adolesc
#253270) is a very rare autosomal recessive metabolic Med 1998;152:707–8.
6 Bodemer C, De Prost Y, Bachollet B et al. Cutaneous manifestations
disorder arising from a defect in incorporation of biotin of methylmalonic and propionic acidaemia: a description based on
(vitamin B7 or H) as a prosthetic group in biotin‐dependent 38 cases. Br J Dermatol 1994;131:93–8.
enzymes such as acetyl‐CoA carboxylase and pyruvate 7 Koch SE, Packman S, Koch TK, Williams ML. Dermatitis in treated
maple syrup urine disease. J Am Acad Dermatol 1993;28:289–92.
carboxylase. While the major clinical findings include
8 Flores K, Chikowski R, Morrell DS. Acrodermatitis dysmetabolica
severe neurological symptoms such as lethargy and sei­ in an infant with maple syrup urine disease. Clin Exp Dermatol
zures as well as potentially lethal ketoacidosis, infants 2016;41:651–4.
Chapter 10  Differential Diagnosis of Neonatal Erythroderma 133

­ iagnostic work‐up of neonatal


D (blood culture) should be performed to detect the exfo­
erythroderma liative toxins A and B.
Whitish maculae on the placenta and umbilical cord
So far, no prospective multicentre studies of neonatal or warrant mycological investigation for Candida. Very pre­
infantile erythroderma have been performed and, there­ mature infants who are diagnosed with congenital cutane­
fore, evidence‐based diagnostic algorithms are still not ous candidiasis should be tested for an invasive candidal
available.

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
infection (CSF/blood culture).
However, even in the case of a complex differential If a clearly elevated serum IgE level is observed in a
diagnosis, a thoughtful stepwise approach to neonatal neonatal patient or a young infant with erythroderma,
erythroderma will lead to a straightforward clinical lymphocyte phenotyping is warranted. In patients with
diagnosis in most cases (see Fig. 10.1). The first step is to B‐lymphocytopenia, a mutation analysis, especially of
determine whether the physical examination and history the RAG‐1/RAG‐2 gene loci, should be obtained; in T‐cell
are consistent with psoriasis, atopic or seborrhoeic der­ immunodeficiency, a chimerism analysis should be per­
matitis and, if so, to treat appropriately. If the answer to formed to detect engraftment of maternal T‐lymphocytes.
the first step is ‘no’ or there is no or insufficient response If the results of immunological tests are normal and
to therapy, the next step is to consider alternative diagnoses, hair shaft anomalies such as trichorrhexis invaginata
i.e. pustules with candidiasis, periorificial distribution in are not present, epidermal LEKTI expression should be
metabolic disorders. The third step is to assess whether determined by immunohistochemistry so as to exclude
the patient is ill or exhibits failure to thrive. If so, the Netherton syndrome.
patient is more likely to have an underlying infectious, A blood gas analysis showing ketoacidosis with con­
immunological or metabolic disorder. comitant hypoglycaemia and hyperammonaemia has to
Irrespective of the underlying disease, neonates and be considered a metabolic emergency. Thus, patients
young infants with erythroderma are at greater risk of should be referred to a specialized centre for substitution
potential complications due to impaired epidermal barrier therapy or dietary counselling and further diagnostic
function such as hypothermia, hypernatraemia, dehydra­ testing (holocarboxylase‐synthetase activity in cultured
tion or hypoproteinaemia. Therefore, at the initial visit, fibroblasts, organic acids in urine).
these patients need to undergo a thorough clinical paedi­ Ichthyosiform erythroderma is particularly difficult
atric examination including neurological assessment and to distinguish from other diagnoses. The previously‐
measurement of bodyweight and core body temperature. mentioned molecular genetic and hair shaft analyses as
Initial capillary blood gas analysis and assessment of well as ophthalmological examinations, including fundus­
serum electrolytes in combination with a limited number copy (glistening dots, sectoral cataract?), and if necessary,
of other screening tests such as differential blood count assessment of sterol isomerase and fatty aldehyde dehy­
and serum total IgE level are also advisable. In desquama­ drogenase activity in cultured fibroblasts are needed.
tive and bullous erythrodermas affecting neonates, a skin
biopsy should be taken to locate the cleavage level and ­Reference
exclude toxic epidermal necrolysis. If subcorneal blister­ 1 Ott H, Hutten M, Baron JM et al. Neonatal and infantile erythrodermas.
ing is present, polymerase chain reaction (PCR) analysis J Dtsch Dermatol Ges 2008;6:1070–85.
134 

CHA PTER 1 1

Vesiculopustular, Bullous and Erosive Diseases


of the Neonate
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Caroline Mahon1 & Anna E. Martinez2


1
 Department of Dermatology, Bristol Royal Infirmary, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
2
 Paediatric Dermatology Department, Great Ormond St Hospital for Children NHS Trust, London, UK

Introduction, 134 Autoimmune causes of vesicular and Other causes of neonatal blistering, 149
Benign and/or physiological neonatal bullous lesions in the neonate, 146 Erosive lesions in the newborn infant, 150
vesiculopustular and bullous skin Genodermatoses associated with neonatal
lesions, 134 blistering, 147

Abstract becomes accustomed to a low‐humidity environment, is colonized


with commensal organisms and matures as an environmental barrier.
The skin of the neonate must make a rapid adaptation to extrau- Distinguishing between cutaneous eruptions that represent benign
terine life. A number of benign physiological vesiculopustular and or transient physiological cutaneous phenomena, infections, inflam-
bullous skin lesions present in the newborn period and these matory dermatoses and genodermatoses is essential to guide clinical
probably represent adaptive events as the skin of the neonate management.

Key points • Most transient and benign vesiculopustular eruptions require


no intervention when typical clinical features present in a well
• Self‐limiting physiological and benign vesiculopustular skin newborn infant, however there should be a low threshold for
eruptions are very common in the neonatal period. further investigation if lesions progress.
• Blistering and erosions are most commonly due to infectious
causes, however genodermatoses and autoimmune skin disorders
should be considered in the differential diagnosis.

Introduction Benign and/or physiological


The skin of the neonate must make a prompt adaptation neonatal vesiculopustular
to extrauterine life. A number of benign and physiological and bullous skin lesions
vesiculopustular and bullous skin lesions may present Milia (see also Chapter 6)
in the newborn period. These eruptions probably rep- Milia are 1‐ to 2‐mm firm, pearly‐white papular lesions
resent adaptive physiological events while the skin with a pseudopustular appearance.
of the neonate becomes accustomed to a low‐humidity
environment, is colonized with commensal organisms
and matures to establish itself as an environmental barrier. Epidemiology and  pathogenesis. In prospective
Distinguishing between skin eruptions that represent observational studies documenting cutaneous lesions
benign and/or transient physiological phenomena, in otherwise well full‐term neonates the incidence of
infections, inflammatory dermatoses and genodermatoses primary or congenital milia ranged between 7.5%
is essential to avoid the unnecessary investigation and and  50% [1–4]. One study found that milia were
treatment of newborn infants with benign skin lesions, ­significantly more common in male than female infants
to appropriately investigate those with likely signifi- at less than 48 hours of age [4]. Milia are subepidermal
cant pathology and to counsel parents appropriately keratin‐containing cysts that represent an accumula-
(Tables 11.1 and 11.2). For further differential diagnoses tion of keratin within vellus hair‐associated sebaceous
please refer to Chapter 73. glands.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 135

Table 11.1  Differential diagnosis of vesiculopustular and bullous eruptions in the neonate

Pathophysiology Typical age of onset Clinical features Typical distribution Diagnostic investigations and
pathological features

Benign, self‐limiting and/or physiological vesiculopustular and bullous disorders


Milia At birth Pinpoint 1‐ to 2‐mm milky Cheeks, nasal bridge and Clinical diagnosis

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
papules alae, forehead Histopathology shows inclusion
cysts containing keratinized
stratum corneum
Miliaria crystallina At birth, or days to Confluent sheets or clusters of Forehead and trunk are most Clinical diagnosis
weeks of age 1‐ to 2‐mm fragile watery commonly involved Histopathology shows sub‐or intra‐
vesicles without inflammatory corneal eccrine duct obstruction
component
Erythema toxicum Typical onset at 1–2 days Small pinhead‐sized pustules Trunk and limbs excluding Clinical diagnosis
neonatorum (ETN) of age. May be seen as and vesicles associated with acral skin and mucous Pustule smear microscopy reveals
late as 2 weeks of age diffuse erythematous flares membranes predominant eosinophils
Miliaria rubra First days to weeks of Localized crops of erythematous Forehead, posterior neck, Clinical diagnosis
age papules and pustules trunk and proximal limbs Histopathology as for miliaria crystallina
Benign (or transient) Usually after the first Scattered superficial fragile Trunk, limbs, hands and feet Clinical diagnosis
neonatal pustular week of life and up pustules which erode leaving Mucous membranes are not Wright‐stained pustule smear
melanosis to 6 weeks of age a characteristic collarette of involved microscopy reveals predominant
fine scale and macular May infrequently occur on neutrophils (in contrast to ETN)
hyperpigmentation acral surfaces
Benign cephalic Onset within 1–2 Crops of fragile pustules and Face and scalp predominate. Clinical diagnosis
pustulosis weeks of age, up to vesicles associated with May occur over the neck
6 weeks of age erythema and often fine scale and upper chest
Neonatal eosinophilic At birth or weeks to Recurrent, intensely pruritic Scalp and acral sites Clinical diagnosis
pustulosis/pustular months of age pustulovesicular eruption Giemsa‐ or Wright‐stained smear
folliculitis microscopy of pustule contents
reveals abundant eosinophils
Acropustulosis of Typical onset within the Crops of inflammatory pustules Palmoplantar surfaces Clinical diagnosis
infancy first 8 weeks of age predominate Smears of pustule contents reveal
Occasionally present Dorsa of the hands and feet either neutrophil‐ or eosinophil‐
from birth may be involved predominant infiltrate
Infantile acne Onset usually from weeks Crops of comedonal and small Cheeks, chin and forehead Clinical diagnosis
to months of age cystic lesions.
Scarring may occur
Infective causes of vesiculopustular and bullous lesions in the neonate
Staphylococci At birth, or days to Superficial flaccid or fragile May occur at any site. Skin swab bacterial culture
(bullous impetigo and weeks of age blisters associated with The umbilical stump is a
staphylococcal desquamation and crusting common focus of bacterial
scalded skin May present as an exfoliative infection/colonization
syndrome) erythroderma with flexural
accentuation (staphylococcal
scalded skin syndrome)
Streptococcus At birth, or days to Superficial bullae and erosions May occur at any site Skin swab bacterial culture
pyogenes weeks of age associated with yellow crusts
Congenital or neonatal At birth, or days to Pustules and/or erosive skin Pustular lesions are generalized Skin swab microscopy and culture
candidiasis weeks of age lesions Congenital candidiasis may Microscopic examination of the
May be complicated by present as an erosive placenta and umbilical cord
pneumonia erythroderma The presence of microabscesses is
Skinfolds and the napkin highly suggestive of Candida
area are common sites of placentitis
involvement
Herpes simplex virus At birth, or days to Presents with either congenital Vesicles, bullae and erosions Viral swab HSV DNA PCR
(HSV) (types 1 and 2) weeks of age erythroderma and extensive may occur anywhere on Tzanck smear of blister base: balloon‐
skin loss or, in cases of the body and be localized like multinucleated giant cells and
postnatal infection, multiple or generalized eosinophilic inclusion bodies
clusters of monomorphic Mucosal lesions are common Direct immunofluorescence assay
vesicles and necrotic erosions (DFA) smear of vesicle contents or
on a background erythema mucocutaneous lesions
  Histopathology shows acantholysis,
Infants may present with solitary keratinocytes within an
disseminated HSV infection intradermal blister cavity and viral
without skin lesions nuclear inclusion bodies
Clinical diagnosis

(Continued )
136 Section 2  Skin Disorders of the Neonate and Young Infant

Table 11.1  Continued

Pathophysiology Typical age of onset Clinical features Typical distribution Diagnostic investigations and
pathological features

Neonatal varicella At birth or within days Polymorphous fragile May occur at any site Viral swab VZV DNA PCR from skin
zoster virus (VZV) to 2 weeks of age (if haemorrhagic vesicles and Neonatal VZV infection is lesions or CSF
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

mother develops superficial erosions on an generally associated with Tzanck smear of blister base reveals
primary varicella erythematous base disseminated disease and multinucleate giant cells in the
infection 1 week may be severe and epidermis with marginated
before or after life‐threatening chromatin
delivery) DFA testing of smear of vesicle
contents
Histopathology reveals intraepidermal
vesiculation, multinucleated giant
cells and nuclear inclusions
Congenital Listeria At birth Haemorrhagic and purpuric skin Generalized skin lesions Bacterial skin swabs and blood
monocytogenes lesions with associated associated with sepsis is cultures
pustular and necrotic lesions typical
Congenital syphilis At birth Generalized and nonspecific Palmoplantar pustules and T. pallidum observed in skin
annular, erythematous or desquamation are classical scrapings or skin biopsy by
purpuric lesions when present dark‐field microscopy, Giemsa or
Localized pustules, blisters and silver staining
erosions (‘pemphigus Serological testing has variable
syphiliticus’) sensitivity and specificity
Rhinitis, periostitis metaphyseal Serum VDRL is nonspecific and may
erosions, hepatosplenomegaly be falsely positive
and lymphadenopathy Placental histology may be helpful
Scabies Days to weeks of age Urticated papules and plaques, Scalp typically involved in Clinical diagnosis
pustules and vesicles infants as well as trunk, Microscopy of skin scraping reveals
Absence of excoriation in limbs, palms and soles scabies mites and/or ova
neonates is characteristic Examination of close Dermoscopy reveals linear scabetic
May present with nodules and contacts is mandatory burrows
may mimic urticaria Skin biopsy reveals intraepidermal
pigmentosa and subcorneal pustules
associated with eosinophils
Coxsackie‐type Typically after the first Crops of acral perioral and genital Oral mucosa, perioral skin, Clinical diagnosis
enterovirus week to 1 month papules and grey‐blue vesicles palms and soles as well as Viral swab enterovirus DNA PCR
of age on an erythematous base dorsal hands and feet
May be koebnerized to flexural Not infrequent in the genital
skin, especially in infants with skin, buttocks and
eczema inguinal folds
Autoimmune causes of vesiculopustular and bullous lesions in the neonate
Maternally‐transmitted autoimmune blistering diseases
Pemphigus vulgaris Onset at birth or during Flaccid bullae and erosions Typically generalized Histopathology reveals
(PV) and pemphigus the first 2 weeks of associated with crusting affecting the skin and the intraepidermal blister
foliaceus (PF) age affecting the skin and mucous mucous membranes Direct IF: intercellular deposits of
membranes in both PV and PF (neonates in contrast to IgG and/or C3 in the epidermis
adults have diffusely Indirect IF: may be performed on
distributed desmoglein‐3 serum
in both the skin and
mucous membranes)
Bullous pemphigoid Onset within days or Urticated plaques and tense Widespread tense vesicles Histopathology shows eosinophilic
(BP) weeks of age serous fluid‐filled or and bullae spongiosis with subepidermal
haemorrhagic bullae on an Mucous membranes are not blistering/clefting
inflammatory base affected Direct IF: linear deposition of IgG and/or
C3 along the basement membrane
Indirect IF: may be performed on
serum or blister fluid
NaCl salt‐split skin reveals IgG
deposition on the epidermal side
(blister roof)
Pemphigoid gestationis Within the first week of Annular crops of vesicles and Blistering may occur Direct IF: linear deposition of IgG and
(PG) age bullae on a background of anywhere C3 along the basement membrane
Neonates affected in polycyclic or urticarial Mucous membranes are not NaCl salt‐split skin reveals IgG
3–5% of pregnancies erythema affected deposition on the epidermal side
complicated by PG (blister roof)

(Continued ) 
Table 11.1  Continued

Pathophysiology Typical age of onset Clinical features Typical distribution Diagnostic investigations and
pathological features

Linear immunoglobulin Onset of blistering Crops of tense vesicles and Generalized blistering Skin biopsy reveals subepidermal
A bullous dermatosis reported as early as bullae Mucosal involvement is blistering with linear IgA
1–10 days of life common in neonates and deposition along the epidermal
may lead to life‐threatening basement membrane

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
aerodigestive complications
Paraneoplastic causes of vesiculopustular and bullous lesions in the neonate
Pustular dermatosis of First weeks to the first Crops of superficial Facial rash is commonly Histopathology reveals infiltrate of
myelodysplasia in month of life pustulovesicular lesions on a described but may occur immature myeloid cells within
Down syndrome background urticated anywhere epidermal spongiotic
erythema vesiculopustular cavities
Other causes of vesiculopustular and bullous lesions in the neonate
Langerhans cell At birth, or within the Multiple haemorrhagic Lesions usually are Histopathology reveals dense dermal
histiocytosis (LCH) first days of life papulovesicles generalized with infiltrate of Langerhans cells
May mimic miliary predominance of lesions Staining for CD1a, S100 and
haemangiomatosis in the scalp, inguinal folds Langerin are classically positive
Solitary congenital LCH tumours and genital areas Histopathology as above
or ulcers have been described May occur anywhere on the
and are frequently self‐resolving body
Bullous mastocytosis At birth, or within days Sparse or generalized bullae Lesions may occur anywhere Histopathology reveals dense
to weeks of life associated with widespread on the body dermal mast‐cell infiltrate with
red‐brown plaques and plane of blister cleavage through
macules the lamina lucida
CD117 positive
Bullous neonatal lupus At birth or at days to Bullae on an inflammatory base May occur anywhere on the Histopathology reveals subepidermal
erythematosus (NLE) weeks of age healing with milia body blistering and vacuolar
Sun‐exposed sites are degeneration of the basal layer
thought to be more prone Maternal anti‐Ro/SSA and/or
to NLE lesions anti‐La/SSB antibody positive
Hyperimmunoglobulin At birth, days, weeks or Generalized sterile papulopustular Face and scalp involvement Histopathology reveals eosinophil‐
E syndrome (HIES) months of age eruption occurring within 2 initially, progressing in rich infiltrate similar to that
months of life in 67% of cephalo‐ caudal direction seen in EPF
infants with HIES Markedly elevated serum IgE levels
Mucocutaneous candidiasis and Three known gene mutations
fungal paronychia are common (TYK2, STAT3, DOCK8)
Deficiency of At birth or days to Sheet‐like micropustular eruption Usually generalized pustular Due to deficiency of interleukin‐1
interleukin‐1 weeks of age on background erythema lesions scattered anywhere receptor antagonist (DIRA)
receptor antagonist resembling pustular psoriasis on the body Skin biopsy shows neutrophilic
(DIRA) Fever, arthritis and/or subcorneal pustules
osteomyelitis may be Molecular genetic testing for
associated mutations in ILRN1
Neonatal bullous At 1–2 months of age Multiple indurated annular May be localized or Histopathology reveals a dense
neutrophilic plaques associated with generalized dermal neutrophilic infiltrate
dermatosis vesicles and bullae or A florid peripheral neutrophilia is
(Sweet syndrome) pustulation on an common
erythematous base Often heralds an immunodeficiency
syndrome, leukaemia or
autoimmune/inflammatory disorder
Genodermatoses associated with neonatal blistering
Epidermolysis bullosa At birth, or within days Bullae, erosions and/or May occur anywhere, but IF of the skin biopsy may be
(EB) or weeks of age ulceration extremities and sites of diagnostic; confirm with genetic
May present with large areas trauma most common testing if available; rarely EM may
of congenital skin loss Aplasia cutis congenita may be required
(aplasia cutis) occur in all forms of EB
Congenital At days to weeks of Generalized severe Skin fragility, bullae and Histopathology reveals
erythropoietic age photosensitivity superficial erosions in subepidermal blistering with
porphyria Severe blistering may result from light‐exposed areas of skin cleavage within the lamina lucida
phototherapy treatment for May be associated with Markedly decreased erythrocyte
neonatal jaundice profound haemolytic uroporphyrinogen synthase
anaemia activity and/or markedly increased
Pink urine and urinary levels of urinary uroporphyrin I
fluorescence under and coproporphyrin I isomers
Wood’s lamp is Detection of biallelic pathogenic
characteristic variant in UROS, or rarely GATA1

(Continued )
138 Section 2  Skin Disorders of the Neonate and Young Infant

Table 11.1  Continued

Pathophysiology Typical age of onset Clinical features Typical distribution Diagnostic investigations and
pathological features

Incontinentia pigmenti At birth or up to 6 Blaschkolinear crusted May occur anywhere on the Histopathology shows
weeks thereafter vesicopustules on an body, although the face is intraepidermal blistering with an
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

(skin lesions may first inflammatory base almost always spared eosinophilic infiltrate
present as late as 1 Cicatricial scalp alopecia is Molecular genetic testing: IKBKG
year of age) not infrequent reveals deletion of exons 4–10 in
80% of cases
Superficial At birth or within a few Bullae and erosions with Intermittent blistering and Skin biopsy reveals hyperkeratosis,
epidermolytic days of age minimal associated desquamation may occur vacuolated keratinocytes and
ichthyosis (previously background erythema anywhere on the body keratohyalin granules in the
ichthyosis bullosa of Superficial erosions and No mucosal involvement epidermal granular and spinous
Siemens) desquamation, so‐called   layers
‘Mauserung phenomenon’ No associated acral Molecular genetic testing of KRT2
keratoderma
Epidermolytic At birth or within a few Congenital erythrodema and Generalized erythema and Clinical diagnosis
ichthyosis (previously days of age fragile superficial bullae blistering resulting in Skin swab bacterial culture
called epidermolytic appearing at or within days superficial erosions with Molecular genetic testing of
hyperkeratosis or of birth desquamation KRT1 and KRT10
bullous congenital Annular subtype described Secondary bacterial infection Skin biopsy shows variable
ichthyosiform May be confused with is common hyperkeratosis and acanthosis
erythroderma) staphylococcal scalded skin No mucosal involvement with vacuolar change and
syndrome and suprabasal Palmoplantar keratoderma thickening of the epidermal
forms of EB may be present from birth granular layer
(more commonly
associated in infants with
mutations in KRT1)

CSF, cerebrospinal fluid; DEJ, dermoepidermal junction; DFA, direct fluorescent antibody; IF, immunofluorescence; NaCl, sodium chloride; PCR, polymerase
chain reaction (in vitro nucleic acid amplification); VDRL Venereal Disease Research Laboratory (serological treponemal antibody test).

Table 11.2  Differential diagnosis of erosive or ulcerative skin lesions in the neonate

Pathophysiology Age at onset Clinical features Typical distribution Pathological features and
diagnostic investigations

Mechanical and/or iatrogenic causes of erosions in the neonate


Iatrogenic scalp At birth Localized erosions on the Anywhere on the scalp Clinical diagnosis
birth injuries vertex of the scalp due to Usually linear or crescenteric lesions
pH scalp electrode, ventouse
or forceps placement
May be confused with aplasia
cutis capitis
Sucking blisters At birth Isolated annular erosions, Typically present on the thumb, fingers or Clinical diagnosis
usually solitary medial aspect of the forearms
Infective causes of erosions in the neonate
Staphylococcal At birth or days to Erythroderma with widespread Generalized desquamation Skin swab bacterial culture
scalded skin weeks of age superficial desquamation Perioral, genital and flexural accentuation Skin biopsy shows subcorneal plane
syndrome (SSSS) classical of cleavage
The umbilical stump is a common focus of
infection
Congenital or At birth, days to Erythema associated with May be present with erythroderma, Clinical diagnosis
neonatal weeks of life generalized pustules and generalized micropustulation, scaling Skin swab bacterial culture
candidiasis genital erosions with or without ulceration (congenital
candidiasis) or be localized to the
napkin area, trunk and/or the skinfolds
Herpes simplex virus At birth or at a few Erythroderma with erosions May present with either localized Tzanck smear
(HSV) infection days of age and ulceration ulceration or extensive areas of DFA testing of skin smear
(types 1 and 2) denuded skin Viral swab HSV DNA PCR
from skin lesions or CSF
Skin biopsy

(Continued ) 
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 139

Table 11.2  Continued

Pathophysiology Age at onset Clinical features Typical distribution Pathological features and
diagnostic investigations

Fetal varicella At birth (if mother Stellate, well‐circumscribed May occur anywhere on the body Clinical diagnosis
(VZV) syndrome infected in the ulcerations (aplasia cutis) May be associated with limb hypoplasia, PCR swab for VZV DNA

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
first 20 weeks of often in a segmental or microcephaly, ocular abnormalities from skin lesions or CSF
pregnancy) dermatomal distribution (microphthalmia, chorioretinitis, Serum IgM (25% positive)
corneal alterations, cataracts) and
growth retardation
Primary varicella At birth, days or Eroded or ulcerated papules Usually generalized PCR swab for VZV DNA
zoster virus (VZV) weeks of life and vesicles
infection
Ecthyma Days and weeks Necrotic plaques and May be solitary or multiple and occur Bacterial skin swabs/blood cultures
gangrenosum after birth ulceration anywhere on the body for Pseudomonas aeruginosa
localized ulcerations
Congenital Listeria At birth Haemorrhagic areas, May occur anywhere on the skin Bacterial skin swabs/blood cultures
monocytogenes purpura, pustules and
infection bullae
Primary cutaneous Days to weeks after Erosions, ulcerations and Skin may be primarily inoculated Skin biopsy
aspergillosis birth pustulation superimposed through areas of macerated skin, or Fungal hyphae may be visible in
Particularly on background erythema infected at sites of adhesive skin fresh tissue stained with calcofluor
associated with taping or central or peripheral venous Haematoxylin and eosin staining
prematurity and catheter devices variably demonstrates Aspergillus
central venous hyphae
access Period acid–Schiff and Grocott silver
stains may add sensitivity
PCR detection of Aspergillus DNA in
fresh tissue
Autoimmune causes of erosive lesions in the neonate
Maternally‐transmitted autoimmune blistering disease
Pemphigus Onset at birth or In both PV and PF, infants Erosions may involve both the skin and Skin biopsy reveals epidermal
vulgaris during the first 2 present with localized or mucous membranes in neonates with acantholysis with intraepidermal
(PV), pemphigus weeks of life extensive flaccid fragile PF or PV plane of blister cleavage
foliaceus (PF) bullae and crusted erosions IF: intraepidermal IgG antibodies
Other causes of erosive lesions in the neonate
Congenital erosive At birth Mixed erosions, vesicles and Typically generalized skin involvement Clinical diagnosis
and vesicular bullae in early neonatal sparing mucous membranes Almost Skin biopsy findings depend on
dermatosis with period all cases involve >75% of skin surface disease stage
reticulated Reticulated atrophic but
supple scarring subtle scarring evolves over
weeks to months
Langerhans cell At birth, or within Multiple small haemorrhagic Seborrhoeic distribution is classical, but Skin biopsy shows dense dermal
histiocytosis days of life erosions and ulcerations may occur anywhere infiltrate of Langerhans cells
(LCH) CD1a, S100 and Langerin positive
staining
Congenital At birth Solitary eroded or ulcerated May occur at any site Solitary lesions are generally self‐
self‐healing papule and/or nodule healing and regress spontaneously
reticulo­- over 3–6 months
histiocytosis
Bullous and At birth, or within Sparse or generalized erosions Lesions may be single or multiple and Skin biopsy reveals a dense dermal
diffuse days to weeks of and ulcers admixed with occur anywhere on the body mast‐cell infiltrate with plane of
cutaneous life bullae and vesicles blister cleavage through the
mastocytosis Skin may thicken markedly in lamina lucida
early postnatal life CD117 positive staining
Aplasia cutis At birth Well‐demarcated, often Localized or widespread ulcers or scars Skin biopsy reveals absence of
congenita stellate areas of absent or May occur anywhere, but most common epidermal and dermal structures
scarred skin on the vertex of the scalp and skin appendages
Lesions may be linear and
appear blaschkoid or
dermatomal in distribution
Methylmalonic At birth or within days Extensive erythema and Marked perioral accentuation Skin biopsy is nondiagnostic
acidaemia of birth superficial erosions Blood count often reveals neutropenia
and thrombocytopenia
Plasma amino acid profile is charac­
teristic and confirms the diagnosis

(Continued )
140 Section 2  Skin Disorders of the Neonate and Young Infant

Table 11.2  Continued

Pathophysiology Age at onset Clinical features Typical distribution Pathological features and
diagnostic investigations

Genodermatoses associated with erosions and ulceration


Epidermolysis At birth, days or Bullae, erosions and/or May present with large areas of Skin biopsy
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

bullosa (EB) weeks of ulceration, particularly denuded skin or skin loss IF and EM may confirm the
postnatal life extremities Nail changes may be present at birth diagnosis
May present with localized Genetic testing may be appropriate
area of aplasia cutis on one
or more limbs
Epidermolytic At birth Erythroderma with occasional Generalized superficial exfoliation and Clinical diagnosis
ichthyosis fragile bullae and erosions Skin biopsy shows variable
superficial exfoliation Congenital palmoplantar keratoderma hyperkeratosis and acanthosis
Desquamation is often may be evident with vacuolar change and
annular and may mimic thickening of the epidermal
SSSS and other bullous granular layer
disorders Mutational analysis of KRT1 and
KRT10 genes
Superficial At birth Occasional bullae and Intermittent blistering and Skin biopsy
epidermolytic minimal erythema desquamation may occur anywhere Mutational analysis of KRT2 gene
ichthyosis Superficial erosions and on the body
desquamation No mucosal involvement
Incontinentia At birth or up to Blaschkolinear erosions/ May occur anywhere on the body, Clinical diagnosis
pigmenti (IP) 6 weeks ulcerations although face is almost always spared Mutational analysis of IKBKG gene
thereafter
(may first occur as
late as 1 year
of age)
Focal dermal At birth Blaschkolinear aplastic streaks May occur anywhere on the body Clinical diagnosis
hypoplasia and/or dermal hypoplasia Sparse scalp hair and/or patchy Mutational analysis of
(Goltz syndrome) often associated with fat cicatricial alopecia are common PORCN gene
herniation and
telangiectasia
Limb malformations, ocular
abnormalities and
craniofacial dysmorphism
are common
Restrictive At birth Tight, shiny translucent Severe generalized skin restriction Clinical diagnosis
dermopathy skin associated with Erosions typically on extensor surfaces of Skin biopsy reveals hypergranulosis
localized erosions and the joints and absent sebaceous and
ulceration eccrine structures
Joint contractures, pinched Mutational analysis of ZMPSTE24
facial features and or LMNA
microstomia are
characteristic features

CSF, cerebrospinal fluid; DEJ, dermoepidermal junction; DFA, direct fluorescent antibody; EM, electron microscopy; IF, immunofluorescence; PCR,
polymerase chain reaction (in vitro nucleic acid amplification).

bridge. Bohn nodules may be mistaken for natal teeth.


Clinical features. Milia are typically present at birth and
Profuse congenital facial milia are a prominent feature of
their facial distribution is characteristic. Most commonly
Basan syndrome, a rare autosomal dominant dermatosis
they are found on the face, typically on the forehead and
also associated with transient acral bullae, lack of derma-
chin and in the nasolabial folds. Mucosal lesions, either
toglyphs, and palmoplantar hypohidrosis [5]. Other rare
on the palate (Epstein pearls) or alveolar surfaces (Bohn
disorders associated with persistent or widespread milia
nodules) are also common.
include oro‐facial‐digital syndrome, hereditary trichod-
ysplasia (Marie Unna hypotrichosis) (OMIM #146550),
Differential diagnosis. The appearance and distribution Loeys–Dietz syndrome (OMIM #609192) [6], follicular
of milia is usually typical and does not cause diagnostic atrophoderma–basal cell carcinoma (Bazex–Dupré–Christol)
confusion. Milia most closely resemble sebaceous hyper- syndrome (OMIM 301845) and Rombo syndrome (OMIM
plasia, also common in neonates, but in contrast, seba- 180730) characterized by vermiculate atrophoderma, milia,
ceous hyperplasia is almost always confined to the nasal hypotrichosis, trichoepitheliomas and basal cell carcinomas.
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 141

adhesive medical monitoring leads and tapes. Congenital


Laboratory/histology findings. The diagnosis is clinical.
lesions are uncommon, but have been reported [10].
The histological appearance of milia is classical. Typically a
In some cases, a history of intrapartum fever is suspected
globular cyst lined with several layers of stratified squa-
to have been the trigger [11].
mous epithelium containing keratinous material is associ-
Miliaria rubra is characterized by crops of nonfolli-
ated with the infundibular portion of a hair follicle or
cular vesicles on an inflammatory base. Most often the
eccrine sweat duct. There is no associated inflammation [7].

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
flexural skin and sites of friction such as the neck folds
are most prominently affected, but miliaria rubra also
Treatment and  prevention. Primary milia in newborns commonly involves the trunk. Type 1 pseudoaldoster-
resolve spontaneously over weeks to months and require onism, a disorder of mineralocorticoid resistance, results
no treatment. in excess loss of salt through eccrine secretions and
appears to characteristically cause a recurrent pustular
Miliaria form of miliaria rubra [12].
Miliaria is a pustulovesicular eruption caused by eccrine
duct obstruction leading to subcutaneous sweat reten- Differential diagnosis. Vesicular lesions may evolve to
tion. Three forms are described: miliaria crystallina become pustular and mimic the infectious lesions of
(sudamina), miliaria rubra and miliaria profunda. These Staphylococcus aureus, Candida albicans and herpes simplex
forms are clinically and histopathologically distinct, the virus (HSV). Miliaria rubra may be mistaken for erythema
appearances depending on the level of eccrine duct toxicum neonatorum, however unlike erythema toxicum
obstruction. miliaria rubra has a predilection for flexural skin and
tends to recur.
Epidemiology and pathogenesis. In a retrospective study
of 5387 Japanese infants examined on a postnatal ward,
Laboratory/histology findings. Miliaria crystallina is
miliaria crystallina was diagnosed in 4.5% of newborns,
characterized by sterile subcorneal vesicles, miliaria rubra
with a peak incidence at 1 week of age [8]. Obstruction of
by intraepidermal vesicles and, in cases of miliaria pustu-
the epidermal component of the sweat ducts leads to
losa, vesicles and pustules are present at the level of the
accumulation of eccrine secretions within the epidermis
dermoepidermal junction. Miliaria can be distinguished
and subsequently superficial vesicle formation.
from infective lesions by the absence of inflammatory
cells, negative bacterial and viral swabs and the presence
Clinical features. Miliaria crystallina is the most super-
of giant keratinocytes on cytological examination of
ficial form of miliaria. Fragile sheets of superficial trans-
vesicle contents.
lucent noninflammatory vesicles measuring 1–3 mm in
diameter and containing watery fluid are most commonly
Treatment and prevention. Cooling measures are gener-
seen in the neck folds and axillae and on the back. Lesions
ally all that is required. Infants should be unwrapped if
on the face are not uncommon (Fig. 11.1). Onset is typically
tightly swaddled and removed from warm or excessively
in the first days of life, and lesions are exacerbated by
humid environments. If care in an incubator is required,
high‐humidity environments, phototherapy and occlusive
the humidity and temperature should be reduced and the
clothing and emollients [9]. Lesions may be found under
infant dressed in loose clothing.

Erythema toxicum neonatorum


(see also Chapter 6)
Erythema toxicum neonatorum (ETN) is characterized by
pinpoint vesicles and pustules appearing on a blotchy
background of ill‐defined macular erythema.

Epidemiology and pathogenesis. The incidence of ETN


in population studies of infants in neonatal units and
postnatal wards varies between 8% and 44% [3,4,11–13].
Incidence is higher in Caucasian infants [10,14] but ETN
is seen in all ethnic groups with incidence rates appearing
to depend on the timing of the neonatal skin examination.
Some authors have reported a preponderance of female
infants [15,16], but others have not [10,11]. The incidence
of ETN appears to increase with gestational age with most
studies [6,11,17–19] reporting a higher incidence in term
Fig. 11.1  Miliaria crystallina on the forehead of a neonate.
infants than in those born prematurely. Associations with
Source: © Crown copyright [2000‐2005] Auckland District Health Board. mode of delivery are unclear: some authors have observed
Image courtesy of Auckland District Health Board, Department of Newborn higher rates of ETN in infants born vaginally [10,11,13]
Services. New Zealand. and others in those born by caesarean section [4,12,20].
142 Section 2  Skin Disorders of the Neonate and Young Infant

The cause of ETN is not known. Many have speculated


that ETN represents a type of cutaneous hypersensitivity
reaction and a normal physiological response to coloni-
zation of neonatal skin with commensal bacteria and
yeasts [21].
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Clinical features. Lesions appear as early as day 2 of life


and as late as 10 days. Congenital lesions have rarely been
reported [22,23]. Typically an infant with ETN presents
with multiple macular areas of blotchy ill‐defined ery-
thema with a central 1‐ to 2‐mm papule or pustule, most
commonly on the trunk [10] but possibly involving any
Fig. 11.2  Transient pustular melanosis on the sole of the foot of a
area of the skin except acral sites. Occasionally infants newborn infant.Source: © Crown copyright [2000‐2005] Auckland District
present with florid ETN with marked erythema and urti- Health Board. Image courtesy of Auckland District Health Board,
carial papules or pustules. Infants are characteristically Department of Newborn Services. New Zealand.
asymptomatic. Lesions spontaneously resolve over 2–4 days
and tend not to recur.
Clinical features. Lesions are typically present at birth.
Differential diagnosis. Confusion between the skin Milky‐white to yellow, fragile pustular lesions without
lesions of ETN, miliaria rubra and neonatal cephalic pus- background erythema are most commonly seen on the
tulosis may arise, or they may occur concurrently. The forehead, bitemporal regions, cheeks, neck and back.
lesions of ETN can generally be clinically distinguished Involvement of the palms and soles is less frequent
from those of herpes simplex and varicella zoster virus or (Fig. 11.2). The fragile pustules rupture with minor trauma
Candida albicans. Eosinophilic pustular folliculitis may leaving pea‐sized pigmented macules with a peripheral
closely resemble ETN both clinically and histologically, collarette of fine scale. The hyperpigmentation may
although large pustules rather than papules are the pre- persist for weeks to months. In some cases neonates
dominant morphological finding in eosinophilic pustular present with annular hyperpigmented macules associ-
folliculitis. ated with a peripheral halo of fine scale soon after birth,
having presumably developed pustular lesions in utero.
Laboratory/histology findings. Microscopic evaluation
of a pustule smear stained with Giemsa or Wright stain Differential diagnosis. Infectious diseases, in particular
reveals an eosinophil‐dominant exudate. A peripheral staphylococcal and streptococcal pyoderma, candidiasis,
blood eosinophilia may be observed in 7–15% [24] of primary varicella zoster and herpes simplex virus and
affected neonates. Skin biopsy reveals dermal oedema syphilitic infections may all mimic TNPM. Other benign
with a primarily eosinophilic perivascular infiltrate. In neonatal eruptions confused with pustular melanosis
papular lesions an eosinophilic infiltrate centred in the include eosinophilic pustulosis, benign cephalic pustulosis
pilosebaceous unit may be observed. and ETN.

Treatment and prevention. Parents should be reassured Laboratory/histology findings. Microscopic examination


that the natural history is spontaneous resolution without of a Wright or Giemsa‐stained pustule smear reveals a
specific intervention. No treatment is required. predominance of neutrophils with only occasional eosin-
ophils. A skin biopsy is rarely required for diagnosis.
Transient neonatal pustular melanosis Histopathology reveals epidermal pustules with either
(see also Chapter 6) intra‐ or sub‐corneal collections of neutrophils with scat-
Transient neonatal pustular melanosis (TNPM), also tered eosinophils [22] and a basal and suprabasal increase
called benign pustular melanosis, is a self‐resolving in pigmentation, without pigmentary incontinence.
pustular eruption that typically presents in the first
­
month of life. Treatment and  prevention. TNPM is a self‐limited
disease in newborns, and no treatment is required.
Epidemiology and  pathogenesis. TNPM affects both
sexes equally with an overall incidence of approximately Neonatal and infantile eosinophilic
0.5–1% [4] and is more common in Afro‐Caribbean infants, pustulosis
in whom the incidence is 4.4% [25]. The aetiology is Neonatal eosinophilic pustulosis (NEP) is a rare eruption
unclear. It has been speculated that TNPM is a variant that appears to be associated with prematurity [27]. It is
form of ETN, and therefore a physiological response to probably a variant of eosinophilic pustular folliculitis
skin colonization with commensal microorganisms, (EPF), a relapsing and remitting eosinophil‐predominant
because multiple cases of infants with pustular melanosis pruritic pustular dermatosis of unknown aetiology first
followed by the subsequent appearance of classical described in Japanese adults by Ofuji et  al. in 1970 [28]
lesions of ETN have been described [26]. (also referred to as Ofuji’s disease), but subsequently also
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 143

reported to occur in infancy and childhood [29]. An infan-


tile form of EPF was first described in 1981 [30], followed
by the publication of a case series of five children [31] in
1984. Infantile forms of EPF do not consistently present
with histological evidence of folliculitis. Although the
clinical history, lesion morphology and distribution are

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
similar in eosinophilic pustulosis of infancy (EPI) [32]
and NEP [33], rather than being distinct disorders, these
entities are likely to fall within the spectrum of EPF.
Additionally, a number of authors have commented on
the clinicopathological similarity between eosinophilic
pustulosis of infancy and acropustulosis of infancy [28,34]
and have suggested that this entity also falls within the
spectrum of EPF.

Epidemiology and  pathogenesis. There are a handful


of case reports describing nonfollicular eosinophilic
pustulosis and eosinophilic folliculitis in neonates, all
of which were boys born at 26–28 weeks’ gestation with
onset at between 7 and 13 weeks of age [24,35,36]. Two
infants had a preceding history of Candida sepsis.
A large series of 15 infants with EPI accompanied by a
review of an additional 46 cases from the literature Fig. 11.3  Infantile eosinophilic pustulosis of the scalp in a child aged
found that EPI is more common in boys, with a 36 months.
male : female ratio of 4 : 1, with a mean age of onset
of  6 months with 95% of children presenting before In neonates, NEP is characterized by sterile pustules
14 months of age [37]. There appears to be no racial primarily or exclusively involving the scalp with less
predilection, although EPF appears to be more com- prominent involvement of the face, trunk and extremities.
monly reported and probably more readily recognized Although EPI is characterized by remissions and relapses,
in Japan than in other countries [38]. only one of the neonatal cases described developed
The pathogenesis is unknown. The clinicopathological recurrence.
similarities between hyperimmunoglobulin E syndrome,
EPI and NEP, and the fact that eosinophilic folliculitis is Differential diagnosis. Staphylococcal and streptococcal
well described in patients with human immunodeficiency impetigo should be excluded, as should neonatal herpes
virus (HIV) [39], haematological malignancies [40] and simplex and varicella zoster virus infection. Scabies mites
immunodeficiency syndromes [41] suggests that a dys- commonly infest the head and neck in young infants who
regulated or immature immune response to an as yet may present with a florid vesiculopustular eruption in
unknown trigger is responsible for the vigorous cutane- the scalp. Hyperimmunoglobulin E syndrome may pre-
ous eosinophilic response seen in these infants. In both sent in the neonatal period [42] and this diagnosis,
children and adults EPF should be considered a potential although rare, should also be considered, as there is
cutaneous marker of immunosuppression. considerable overlap in the clinical presentation and labo-
ratory and histopathological findings in both disorders.
Clinical features. Pustular lesions are typically clustered Other diagnoses that may mimic NEP or EPI include
in groups (in contrast to adult EPF in which lesions are Langerhans cell histiocytosis, neonatal pustular melanosis
classically annular) and favour the scalp and bitemporal and neonatal cephalic pustulosis.
areas (Fig.  11.3). In 65% of cases other body sites are
involved and may include the palms and soles. Pruritus Laboratory/histology findings. Eighty percent of EPI
is very common (84%) and many younger infants are cases and all the cases of affected neonates have an
noted to be unsettled and irritable [34]. Crops of pustules associated peripheral eosinophilia with leucocytosis
with an associated erythema mature and crust over a [34]. A Wright‐stained smear of pustular contents dem-
matter of days, then spontaneously heal without scarring onstrates abundant eosinophils. Histopathology reveals
over subsequent weeks. Recurrence is the rule (intervals perifollicular (54%) or interfollicular (32%) inflammatory
between flares ranging from 1 to 12 weeks) with multiple infiltrates in upper and mid dermis, comprising pre-
stereotyped episodes of pustulation occurring over dominantly eosinophils together with neutrophils and
months to years following which there is spontaneous mononuclear cells [33].
resolution in all cases. Eighty percent of infants experi-
ence spontaneous resolution by 3 years of age [34]. Treatment and prevention. Although spontaneous reso-
Affected infants are otherwise well and no systemic lution is the rule, and treatment not necessarily indicated,
involvement, beyond a florid blood eosinophilia with pruritus may be troublesome. Numerous treatments have
leucocytosis, has been reported. been utilized in EPI, including topical corticosteroids [43],
144 Section 2  Skin Disorders of the Neonate and Young Infant

oral antihistamines [26], dapsone [29] and erythromycin


[26] with mixed results. In adult cases, indometacin [44] is
the treatment of choice with response rates of 80–90%,
however in young infants this agent has significant
adverse effects, particularly on renal perfusion. Oral
indometacin has been used successfully in childhood [45].
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Topical tacrolimus [46] and indometacin [47] have both


been used successfully, and these may be appropriate
first‐line treatment options.

Infantile acropustulosis

Epidemiology and pathogenesis. Infantile acropustulo-


sis (IA) was first described in 1979 by Kahn et  al. [48]
and Jarrat et al. [49] who proposed this term to describe Fig. 11.4  Infantile acropustulosis involving the foot of an 18‐month‐old infant.
a series of 15 infants with recurrent crops of pruritic pap-
ulopustules on acral skin and the distal extremities pre-
treated for scabies prior to presentation, only one infant
senting within the first 2 months of life and responding
having confirmed scabies by microscopic examination of
rapidly to treatment with dapsone. In some cases lesions
a skin scraping. Infants were followed up for 2–24 months,
were present at birth. It appeared initially that acropustulo-
the majority (18) improving significantly with topical
sis was almost exclusively seen in Afro‐Caribbean infants;
corticosteroid therapy alone. A further series of 25 cases
however, not long after, IA was reported in a number of
reported by Dromy et  al. reported similar findings
countries in infants of various ethnic groups [50]. Male
although of note two children in this series with very late
infants have predominated in the cases reported in some
onset IA presented at 8 and 9.5 years with classical clinical
[45–47,51] but not all [52,53] of the series reported.
features [53].
A number of authors have highlighted similarities
between scabies infestation and AI because in most cases
Differential diagnosis. The primary differential diagnosis
the clinical findings are indistinguishable [54,55]. Indeed,
of AI is scabies infestation. Other diagnoses to consider
some authors feel that AI does not represent a separate
are pompholyx‐type dyshidrotic eczema, an id dermatitis
clinical entity, and have documented high rates of EPI in
associated with dermatophyte infection. A viral exan-
immigrant children and infants from developing countries
them, such as hand, foot and mouth disease (coxsackie
with high rates of endemic scabies [48,56]. It has been
virus), recurrent herpes simples virus infection or impe-
postulated that in fact most cases of IA are in fact a sequela
tigo should also be considered. In those with involvement
of scabies infestation in which a hypersensitivity reaction
beyond acral surfaces, (particularly if the face and scalp
to the scabies mite and its detritus perpetuates cyclic
are affected), the diagnosis of EPI should be entertained.
recurrent acral pustulation. Diagnostic difficulty is com-
Transient neonatal pustular melanosis and congenital
pounded by the fact that infants are often diagnosed and
cutaneous candidiasis should be considered if onset is
treated empirically for scabies, albeit appropriately, early
at birth.
in the course, and may present to their paediatrician or
dermatologist many months after detection of mites or
ova is possible through skin scrapings. At the same time, Laboratory/histology findings. Direct smears from
there also appears to be considerable clinicopathological pustule contents reveal either neutrophils or eosinophils
overlap between some cases of EPI and IA, leading to fur- although the infiltrate is often mixed. Biopsy of acral sites
ther diagnostic confusion, particularly in infants in whom is rarely performed but, where reported, histopathology
pustular lesions are widespread [26,28,31]. shows intraepidermal vesiculation with either most com-
monly neutrophilic [57] or mixed neutrophil and eosino-
Clinical features. Intensely pruritic palmoplantar papules phil [58] or eosinophil‐predominant infiltrate [50]. Biopsy
evolve into 3‐ to 4‐mm fragile superficial pustules over of a precursor acral lesion has revealed intraepidermal
1–2 days followed by desquamation leaving multiple necrolysis without a cellular infiltrate [59]. A peripheral
collarettes of peeling skin (Fig. 11.4). Typically cyclic recur- eosinophilia may accompany the acral eruption [50,56,60].
rences tend occur 3‐ to 4‐weekly and flares are reported to In general, although the histopathological features are
occur up to the age of 3 years [49]. The dorsal surfaces of distinctive, skin biopsy is not required to make the diag-
the hands and feet are also frequently involved. Some nosis of AI once characteristic lesions are seen and the
infants have generalized involvement including lesions typical clinical course is established.
in the scalp, strongly suggesting overlap with EPI [28,31].
Extreme pruritus is a universal finding and infants are Treatment and prevention. Potent (class I and II) topical
often very unsettled because of itch. In a case series of 21 corticosteroids are the first‐line treatment and effective in
infants diagnosed with AI reported by Mancini et al. [52] aborting an exacerbation if used early and aggressively
lesions were first noted at between 1 and 30 months of during flares [49,50]. Some authors have however
age (mean age 9.7 months). Fourteen infants (70%) were reported poor responses to topical corticosteroids and
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 145

questioned their usefulness, although many have not age, 29 of these were culture positive [62]. In a more recent
stated the potency of the corticosteroids used [45,46,54]. similar study of 104 neonates, positive lesional culture
Scabicides are generally not helpful once recurring pustu- was demonstrated in only 6 of 26 infants with cephalic
lation is established and are not indicated unless there is pustulosis [60]. Thus, while there appears to be a strong
documented infection of a first‐degree relative or close association between Malassezia colonization in newborns
contact, the eruption is localized and dermoscopic skin and the development of cephalic pustulosis, a causative

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
examination and/or scrapings are positive. Dapsone at relationship has not been conclusively demonstrated.
1–2 mg/kg/day has been used with rapid improvement
in pruritus and sustained remission without relapse Clinical features. Infants typically present with non-
[45,54,61] and may be appropriate in severe cases unre- follicular inflammatory papules, pustules and pseudove-
sponsive to topical treatment. A recent case reported com- sicular lesions on a background erythema distributed on
plete responses using a topical D3 analogue, Maxacalcitol the forehead, cheeks and chin. Lesions may less frequently
0.0025% ointment, twice daily in the acute phase, and 3–4 involve the neck and upper chest. The typical age of onset
days per week thereafter [55]. is 2–3 weeks, but infants may present as late as 6–8 weeks
Acropustulosis of infancy eventually remits spontane- of age. Lesions are characteristically asymptomatic, but
ously with less florid and frequent recurrences over time. parents are often very distressed about the appearance.
Parents need to be reassured that this often distressing The natural history is spontaneous resolution over
condition improves gradually with time and does eventu- 7–14 days in the majority of infants [60].
ally remit even without treatment.
Differential diagnosis. Lesions may mimic or indeed
Benign or neonatal cephalic pustulosis
coexist with those of miliaria rubra and seborrhoeic
Benign cephalic pustulosis is a very common self‐resolving
dermatitis. Benign cephalic pustulosis has been called
pustular eruption that favours the face and scalp. Lesions
‘neonatal acne’, causing confusion between this common
consist of inflammatory papules, pustules and pseu-
neonatal eruption and true infantile acne vulgaris, a rela-
dovesicles. The role of Malassezia furfur colonization in
tively rare condition in which children usually present at
causing or perpetuating this inflammatory eruption is
6–9 months of age with comedonal, papulopustular and
unclear. This entity differs both clinically and histopatho-
sometimes cystic lesions typically confined to the cheeks
logically from infantile acne, and the term ‘neonatal acne’
and thought to be primarily androgen‐driven.
should therefore be discouraged.

Epidemiology and  pathogenesis. Neonatal cephalic Laboratory/histology findings. Skin scrapings of affected
pustulosis (NCP) is common, the prevalence varying areas or of pustule contents may be examined using
between 10% and 66% of healthy term newborns [62,63]. potassium hydroxide (KOH) or KOH–Calcofluor wet
Clinical overlap with, or concomitant miliaria rubra and mounts. Stains such as Giemsa or periodic acid–Schiff
seborrhoeic dermatitis may be frequently observed. (PAS), Grocott, and Gomori’s methenamine silver may
Infants of all ethnicities are affected and there is no be used to identify yeasts (and dermatophyte fungi, as
gender predilection. indicated). Culture of Malassezia spp. requires specific
Aractingi et al. first described this entity in 1991 [64] in media (Dixon agar) and a positive growth of Malassezia
a 4‐week‐old infant with a cephalic vesiculopustular generally correlates well with positive smear microscopy.
eruption from which M. furfur was isolated. Rapelanoro However the identification of Malassezia species neither
et al. proposed a set of diagnostic criteria in 1996, which confirms nor refutes the diagnosis of NCP, given that
included the isolation of M. furfur from microscopy of colonization is common, and diagnostic swabs or smears
pustular contents [65]. Other criteria were: age at onset, are generally not recommended unless there is a need to
the presence of lesions distributed on the head and neck investigate an alternative infectious diagnosis.
and clearance with topical 2% ketoconazole therapy.
However, in many cases of NCP Malassezia spp. cannot Treatment and prevention. As infants are typically symp-
be detected on microscopy of pustule smear or culture tom‐free and the eruption self‐limiting, no treatment is
leading to uncertainty about whether colonization or required. Parents should be counselled that the eruption is a
indeed infection with this organism is responsible for benign and transient phenomenon thought to be related to
NCP [59,60,66,67]. the colonization of newborn skin with nonpathogenic com-
Malassezia is a dimorphic lipophilic commensal yeast mensal organisms and that it will resolve without scarring.
and can be isolated from 90% of healthy adults [68], If deemed necessary, topical treatment with a combination
11–50% of term infants in the first days of life and 52–80% of an imidazole (such as 2% ketoconazole or clotrimazole)
after 1 week of age [59,69]. Two studies following term and 1% hydrocortisone cream can be applied twice daily for
infants from birth observing Malassezia colonization rates 3–5 days, which typically leads to rapid clearance.
over time and documenting whether the subsequent
development of cephalic pustulosis relates to positive Erosive pustular dermatosis of the scalp
identification of a Malassezia species have been published. Erosive pustular dermatosis of the scalp (EPDS) is a rare
In a study by Bernier et  al. of 102 neonates, 56 were cause of scarring alopecia and is primarily reported in
diagnosed with cephalic pustulosis at 3 weeks postnatal elderly patients and associated with chronic sun‐damaged
146 Section 2  Skin Disorders of the Neonate and Young Infant

skin. In most cases a traumatic or topically applied ­irritant quantities. IgG transfer to the fetus represents an impor-
trigger precedes the eruption [70]. tant adaptive mechanism conferring short‐lived passive
immunity to the neonate. At the same time, placental
Epidemiology and pathogenesis. Pye et al. first described immunoglobulin transfer from mothers with IgG‐mediated
EPDS in 1979 in a series of older women with sterile pus- autoimmune blistering diseases may result in clinical
tules, erosions and scarring alopecia that resolved with expression of these diseases in the infant at birth or
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

topical corticosteroid therapy [71]. Since then there have during the first days of life. Pemphigus vulgaris (PV),
been multiple case reports and series of adult patients, pemphigus foliaceus (PF), bullous pemphigoid (BP),
and a handful of case reports in neonates with associated pemphigoid gestationis (PG) as well as epidermolysis
scalp trauma sustained during instrumental or difficult bullosa acquisita (EBA) have all been reported in babies
vaginal delivery [72] and in one case of an infant with of affected mothers. In almost all cases blistering has
Klippel–Feil syndrome who at 3 months of age developed developed in babies whose mothers had active disease in
scarring alopecia with chronic scalp ulceration with no pregnancy, however occasionally neonatal disease has
antecedent history of injury to the head [73]. presented in cases in which the mother’s disease was well
controlled, or who had no disease manifestations prior
Clinical features. At birth infants were noted to have to delivery but subsequently developed autoimmune
either annular ulceration or, in one case, necrotic crescen- bullous disease [76].
teric scalp erosion secondary to vacuum extraction and,
in one case, ulceration in the scalp following prolonged Epidemiology and pathogenesis. The autoimmune‐mediated
labour. Days and weeks after the initial injury, a pro- blistering diseases are extremely rare in neonates but
longed chronic phase develops in which inflammation, should be considered in the differential diagnosis of gen-
crust, scaling and an associated alopecia persists for many eralized or localized blistering or erosive lesions present
months. In the case series reported by Siegel et  al. [72], either at birth or soon after. Transplacental transfer of
inflammation, crusting and in some cases recurrent pus- maternal IgG results in disease expression in the new-
tulation persisted for between 4 months and 2 years. born infant. There are multiple case reports of neonatal
PV and BP presenting in the newborns of affected moth-
Differential diagnosis. A diagnosis of EPDS is primarily ers [77]. PG is thought to affect 5–10% of infants of affected
clinical as there are no specific histological features. mothers [78]. Reports of maternally transmitted EBA [79]
Dermatophyte infections closely mimic EPDS and these and pemphigus foliaceus (PF) [80] are extraordinarily
should be excluded, particularly in infants presenting rare and the incidence is unknown. Overall, only a minor-
beyond the immediate neonatal period. The differential ity of infants of mothers with an antenatal diagnosis of
diagnosis includes scalp aplasia cutis congenita. Other con- autoimmune blistering disease develop disease manifes-
ditions associated with congenital or early and persistent tations [81].
neonatal scalp ulceration include ankyloblepharon–­ Male and female infants are affected equally. The
ectodermal defects–cleft lip/palate (AEC) [74] and occurrence and severity of maternally transmitted PV
ectrodactyly–ectodermal dysplasia–cleft lip/palate (EEC). and BP appear to correlate with disease activity and
serum antibody titres during pregnancy, however there
Laboratory/histology findings. Skin biopsy is nonspe- are a number of reports of neonatal PV and BP presenting
cific. Most of the pathological features are consistent with in infants of mothers with well‐controlled or inactive
chronic inflammation and/or scar tissue [68]. disease [82]. Stillbirths and neonatal deaths have been
reported and thus far have occurred exclusively in cases
Treatment and  prevention. Bacterial superinfection is of PV, all occurring in infants whose mothers had severe
common, but antibiotic therapy generally fails to result in or poorly controlled disease during pregnancy [83].
healing of the ulceration [68]. Potent topical corticos-
teroids are the mainstay of treatment in most cases Clinical features. Blistering or localized skin erosions are
once fungal and bacterial infection has been treated or usually present at birth or within the first few days of life,
excluded. In adults, topical tacrolimus 0.1%, dapsone 5% although they may first manifest as late as 2 weeks after
gel, calcipotriol, intralesional and oral corticosteroids and birth [84] (Fig. 11.5). The diagnosis is usually straightfor-
oral isotretinoin have all been reported to be helpful in ward once a history of a maternal autoimmune blistering
isolated case reports [75]. A degree of scarring alopecia is disease is established. Blisters are usually generalized and
the inevitable long‐term sequela in most cases. morphologically similar to adult manifestations of the
respective bullous diseases. However, important differ-
ences warrant mentioning. In contrast to adult patients,
Autoimmune causes of vesicular
neonates with PV present with generalized blistering and
and bullous lesions in the neonate
erosions as well as mucosal blistering because neonates
Bullae in the newborn due to maternal have more diffusely distributed desmoglein‐3, the pri-
autoimmune blistering disorders mary target antigen in PV in nonmucosal skin [85].
(see also Chapter 6)
Of the human immunoglobulins, only IgG and its sub- Differential diagnosis. Infections  –  bacterial, viral and
classes cross the placenta to the fetus in significant fungal – are the important differential diagnoses, followed
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 147

problems for the affected neonate [89]. Of the neonatal


cases reported, the age at presentation ranged from 1 to
10 days. Most had a clinical course complicated by signifi-
cant respiratory and/or feeding problems and required
systemic treatment. In one case, severe ocular scarring
resulted in blindness [90]. Although cases described in

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
the literature have generally been severe, the author has
personal experience of infants in whom blistering was
mild and self‐limiting, requiring only topical treatment.
However, close follow‐up of all infants is advised. In con-
trast to neonatal maternally‐transmitted autoimmune
blistering diseases which spontaneously improve over
weeks, infants with LABD tend to have a prolonged
course, suggesting that the origin of the IgA autoantibodies
Fig. 11.5  Bullous pemphigoid involving the sole of the foot in a 2‐month‐
is not maternal [83].
old infant

Differential diagnosis. Other diagnoses to consider


by toxin‐mediated eruptions such as staphylococcal scalded include staphylococcal and streptococcal pyoderma,
skin. Pemphigus lesions may closely mimic impetigo. bullous pemphigoid, neonatal lupus erythematosus and
bullous erythema multiforme.
Laboratory/histology findings. If autoimmune blistering
is suspected in a neonate, the diagnosis can be confirmed Laboratory/histology findings. LABD is characterized by
by indirect immunofluorescence testing of a cord blood subepidermal blistering with abundant neutrophilic and
sample or by obtaining lesional smear and/or biopsy for eosinophilic dermal infiltrates. The demonstration of lin-
histopathology as well as a biopsy of perilesional fresh ear deposition of IgA along the basement membrane
skin for direct immunofluorescence studies. through immunofluorescence studies is diagnostic.

Treatment and  prevention. The prognosis is generally Treatment. Aggressive treatment with systemic corticos-
excellent. The natural history is generally spontaneous teroids is indicated if respiratory, ocular or oesophageal
resolution over 1–3 weeks as maternal IgG is cleared from involvement intercedes. Early ophthalmology assessment
the infant circulation [79]. Neonatal PG typically resolves is strongly recommended. Dapsone is generally consid-
more rapidly than PV, PF and BP; PG generally clears ered the mainstay of therapy for LABD in the longer term,
completely and spontaneously within days of delivery. and is considered an appropriate first‐line steroid‐sparing
Topical therapy with moderate‐potency corticosteroids is agent. Screening for glucose‐6‐phosphate deficiency
usually all that is required. Systemic corticosteroids are should always be performed before commencing therapy.
rarely indicated. In those with localized disease or blistering confined to
the skin, a mild to moderately potent topical corticoster-
Neonatal linear immunoglobulin A (IgA) oid could be safely employed.
bullous dermatosis

Epidemiology and pathogenesis. Linear immunoglobulin


Genodermatoses associated
IgA bullous dermatosis (LABD) is extremely rare in
with neonatal blistering
infancy. There are seven reported cases of LABD presenting Incontinentia pigmenti
in the neonatal period to date [86]. None of the affected
infants had mothers with linear IgA disease. The major Epidemiology and pathogenesis. Incontinentia pigmenti
target antigen of IgA autoantibodies is linear IgA disease (IP, OMIM #308300) is a rare X‐linked dominant genoder-
antigen‐1 (LAD‐1) a component of BP antigen 180 of the matosis resulting from mutations in the inhibitor of the
basement membrane zone [87]. Genetic susceptibility is kappa B kinase gamma (IKBKG) (previously known as
likely as HLA‐DR3, HLA‐B8 and HLA‐DQW2 subtypes NEMO or Nuclear factor kappa B Essential MOdulator)
are found more frequently in patients with LABD [88]. gene. Mutations in IKBKG lead to aberrant expression of
an essential modulator protein NF‐κB in ectodermal tis-
Clinical features. LABD presents at birth or within days sues, resulting in disturbed transcriptional regulation of
of birth as multiple crops of tense, serous, fluid‐filled genes involved in apoptotic pathways and inflammatory
vesicles and bullae. There may be background erythema, as well as immune responses [91].
but this is not seen in all cases. Lesions may be clustered The estimated population prevalence is 0.7 in 100 000
or appear ‘targetoid’, but the classical annular ‘string‐of‐ [92]. IP is seen almost exclusively in females and is usu-
pearl’ lesions associated with linear IgA disease of child- ally lethal in males. Males represent 6% of all reported
hood are usually seen in older children. Neonatal LABD cases (largely due to sex chromosome aneuploidies or
may result in severe and extensive mucosal and respira- somatic mosaicism) and infants of undetermined sex
tory tract involvement and therefore pose life‐threatening make up 2–3% of cases [93]. The phenotypic expression of
148 Section 2  Skin Disorders of the Neonate and Young Infant

IKBKG mutations is highly variable even within families. Other differential diagnoses include the autoimmune
This is thought to be due to skewed X‐chromosome inac- blistering diseases. It is often helpful to properly undress
tivation combined with highly pleiotropic IKBKG expres- the infant with neonatal blistering as the characteristic
sion resulting in generally poor genotypic–phenotypic blaschkoid patterning of the vesicles in IP may not be
correlation [94]. Many cases of IP are sporadic, but 55% immediately obvious if only localized areas of affected
have an affected mother, who is often unaware of her skin are examined.
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

diagnosis [95].
Laboratory/histology findings. Peripheral blood eosino-
Clinical features. IP may be thought of as a unique form philia is a characteristic finding in the neonatal period.
of ectodermal dysplasia with a characteristic neonatal A skin biopsy may be diagnostic, but is often not neces-
phenotype. The skin is almost invariably involved in sary to make the diagnosis. The classical features are a
neonates with IP and is the defining clinical feature. spongiotic epidermis with intraepidermal eosinophilic
Classical linear vesiculopustular lesions are almost vesiculation, dyskeratotic keratinocytes and pigmentary
always present at birth, although they may first appear incontinence.
as late as the sixth week of life, and represent the first of
four classically‐described phases of cutaneous involve- Treatment and prevention. The natural history is spon-
ment. Blaschkolinear vesiculopustular lesions are most taneous evolution of blistering to warty hyperkeratotic
commonly seen on the arms, legs, trunk and scalp lesions and eventually macular, linear, whorled or retic-
(Fig. 11.6), almost always sparing the face [92]. ulate hyperpigmented streaks over months to years.
IP may be associated with neurological and/or ocular Most adult patients have very subtle skin findings [99].
sequelae. Neurological complications are the most com- No specific intervention is known to modify this course.
mon cause of death and are present in approximately Basic skin care measures may prevent superinfection and
30% of cases. Seizures due to cerebral ischaemia and/or bland emollients may be helpful for crusting and hyper-
structural parenchymal anomalies are the most common keratosis. Ophthalmological assessment in neonates
presentation in the neonatal period [96]. More than 90% with IP is mandatory as sight‐threatening ocular pathology
of neurological problems are evident by the age of 2 years may evolve rapidly after birth [100]. Routine imaging of
[97]. Ophthalmic disease is common, occurring in 36–77% the brain of all neonates with a diagnosis of IP has
of IP patients, and may evolve rapidly in early life. been advocated by some authors [101] but consensus is
Without early intervention almost 60% of these abnor- lacking. Genetic counselling and prenatal molecular
malities may result in irreversible visual loss [98]. genetic testing in subsequent pregnancies should be
offered to parents.
Differential diagnosis. The blistering skin lesions of IP
in newborns are most frequently confused with herpes
simplex infection, but also with congenital varicella. Focal dermal hypoplasia (Goltz syndrome)

Epidemiology and  pathogenesis. Focal dermal hypo-


plasia (FDH; OMIM #305600) is a rare X‐linked dominant
multisystem disorder associated with cutaneous, skeletal,
ocular and vascular anomalies. FDH is due to mutations
in PORCN, a gene coding for the PORCN protein, impor-
tant in Wnt signalling pathways involved in the early
fetal development of both mesodermal and ectodermal
tissues [102].

Clinical features. Characteristic cutaneous lesions are


present at birth, and often blaschkolinear typically
atrophic or cicatricial streaks associated with telangiec-
tasia are found on the trunk and limbs (Fig. 11.7). The
streaks may be accompanied by vesicular lesions, fat
herniation through depressed linear areas of atrophic
skin (soft yellow nodules), erosions or areas of frank
aplasia cutis. Macular hypopigmented and/or hyper-
pigmented streaks may coexist, as may other ectodermal,
skeletal and eye abnormalities.

Differential diagnosis. Other X‐linked dominant syn-


dromes in which skin, ocular and distal limb anomalies
are a prominent feature include X‐linked dominant
Fig. 11.6  Incontinentia pigmenti. Blaschkolinear vesiculopustular and chondrodysplasia punctata type 2 (Conradi–Hünermann–
eroded streaks on the arm of a 2‐week‐old infant. Happle syndrome) and congenital hemidysplasia with
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 149

typically born prematurely with tight, rigid, shiny and


translucent skin. The facial gestalt is distinctive and char-
acterized by a pinched nose and a fixed open mouth.
Generalized arthrogryposis is common. Erosions and
ulceration of the skin are frequently described, particu-
larly over joint prominences. Pulmonary hypoplasia is

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
common. Infants are often stillborn or born prematurely,
and die early in the neonatal period, primarily because of
severe pulmonary insufficiency.

Differential diagnosis. The typical clinical features of RD


do not usually cause diagnostic confusion. However the
presence of widespread skin erosions or ulceration in a
low‐birthweight newborn may lead to the consideration
of congenital infection or lethal acantholytic forms of
epidermolysis bullosa in the differential diagnosis.

Laboratory/histology findings. Skin biopsy shows hyper-


granulosis with large keratohyalin granules, a paucity of
dermal elastic fibres and absence of pilosebaceous and
eccrine glands [106].

Treatment and  prevention. RD caused by recessive


Fig. 11.7  Focal dermal hypoplasia. Blaschkolinear atrophic and cicatricial (typically null) mutations in ZMPSTE24 causes a severe
streaks associated with telangiectasia.
phenotype and is generally lethal either in utero or early
in the neonatal period [104–106]. Treatment is support-
ichthyosiform erythroderma and limb defects (CHILD ive. Genetic counselling and prenatal molecular genetic
syndrome). testing in subsequent pregnancies should be offered to
parents.
Laboratory/histology findings. FDH may be diagnosed
clinically based on the presence of characteristic major
findings in the skin and skeletal systems. Clinical diag-
Other causes of neonatal blistering
nostic criteria have been established [103]. Molecular Bullous and diffuse cutaneous mastocytosis
genetic testing may be helpful in cases in which the diag-
nosis is uncertain. Skin biopsy is nonspecific. Epidemiology and  pathogenesis. Blistering caused by
localized mastocytosis or diffuse cutaneous mast cell
Treatment and prevention. No specific therapy alters the infiltration of the skin rarely manifests as widespread
natural history of FDH. Dressing of eroded areas may be blistering at birth. Classically three forms of cutaneous
required in the newborn period. mastocytosis in children are described: mastocytoma
(solitary or up to three isolated skin lesions), maculopap-
Restrictive dermopathy ular mastocytosis (urticaria pigmentosa), multiple discrete
skin lesions and diffuse cutaneous mastocytosis (DCM).
Epidemiology and pathogenesis. Restrictive dermop- Mastocytosis is caused by clonal mast cell proliferation
athy (RD, OMIM #275210) is a very rare autosomal associated with somatic activating mutations in c‐kit [107].
recessive disorder and typically lethal in the newborn
period. RD results from homozygous null mutations in Clinical features. All forms of cutaneous mastocytosis
ZMPSTE24 which codes for zinc metallopeptidase, a pro- may be associated with blistering in infancy, but DCM,
tease enzyme responsible for cleaving prelamin A to form the rarest subtype, presents with neonatal erythroderma
mature lamin A, and less commonly, heterozygous muta- and generalized bullae [108]. Approximately 25 cases
tions in LMNA coding for proteins lamin A and C. LMNA have been reported. The male : female ratio was 2.5 : 1.
gene mutations and missense ZMPSTE24 appear to con- Almost all infants presented with bullae at birth [109].
fer a milder phenotype [104]. Five infants died before the age of 6 months due to systemic
involvement. Transformation to mast cell leukaemia has
Clinical features. RD results in profound fetal hypokine- been described [110]. Initially the skin is erythematous
sia due to skin restriction in late gestation. There is often a and dermographism is a prominent feature. Gradual
history of polyhydramnios, growth restriction and poor generalized thickening of the skin then develops (pachy-
fetal movements evident after 22 weeks’ gestation. Early dermia). In contrast to urticaria pigmentosa and mastocy-
fetal ultrasound scanning is often unremarkable, and the toma, neonatal DCM is often associated with systemic
severe restrictive effect of the dermopathy not evident symptoms, notably recurrent flushing, dyspnoea, vomit-
until the late second or third trimester [105]. Infants are ing and diarrhoea [94]. Neonates with DCM may present
150 Section 2  Skin Disorders of the Neonate and Young Infant

with prolonged bleeding or develop purpuric lesions, A number of hypotheses have been put forward in an
probably due to heparin release. The tendency to blister attempt to explain nonsyndromic forms of ACC including
usually improves over 3–4 years. incomplete fusion of midline mesodermal tissues [115],
local ischaemic or thrombotic events in utero and focal
Differential diagnosis. Bullous mastocytosis may mimic pressure necrosis. More recently the potential role of genes
all infectious and autoimmune causes of neonatal blister- regulating skin morphogenesis has been emphasized,
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

ing as well as epidermolysis bullosa. particularly given the very strong association between
ACC and multisystem and dysmorphic syndromes [116].
Laboratory/histology findings. Skin biopsy reveals a This subject is covered in detail in Chapter 9.
plane of blister cleavage within the lamina lucida and a
dense dermal mast cell infiltrate. Serum tryptase is ele- Congenital erosive and vesicular
vated in infants with extensive cutaneous disease as well dermatosis with reticulated supple
as those with systemic involvement and has no utility as scarring
a prognostic marker [111]. Consideration should be given
to investigating infants with extensive or symptomatic Epidemiology and pathogenesis. Congenital erosive and
disease for systemic involvement. vesicular dermatosis with reticulated supple scarring
(CEVD) is very rare. To date 29 cases have been reported.
Treatment and prevention. Antihistamines (both H1 and CEVD affects mostly preterm infants (79%) and neonates
H2 receptor blocking agents) and sodium cromoglicate whose mothers had chorioamnionitis [117]. The cause is
are the cornerstones of treatment. Tyrosine kinase inhibi- unknown.
tors targeting the c‐kit ligand are promising novel thera-
pies for infants with severe systemic manifestations [112]. Clinical features. CEVD often causes diagnostic confu-
Neonates with DCM are at increased risk of developing sion in the neonatal period. Infants present at birth with
anaphylaxis‐like episodes [113] and parents should be widespread vesicles, multiple erosions and/or extensive
informed and educated in managing these. Provision of areas of ulceration. Typically most of the body surface is
an adrenaline autoinjector pen should be considered. involved. Alopecia of the scalp (43%) and nail hypoplasia
Immunization reactions are common and have been (46%) is common. Acral skin involvement is infrequent
reported to trigger systemic symptoms as well as circula- (7%). Reported associated anomalies include seizures,
tory collapse. The administration of routine childhood microcephaly, ocular anomalies, hepatomegaly and
immunizations may require specialist supervision [114]. delayed development. A third of infants develop new
vesicular skin lesions after birth, although in all cases the
eroded and ulcerated areas gradually heal in the months
Erosive lesions in the newborn infant after birth leaving soft, reticulated, often hypopigmented
Aplasia cutis congenita scarring. Hypohidrosis commonly develops due to
Aplasia cutis congenita (ACC) describes any congenital eccrine gland loss secondary to scarring and heat intol-
absence of skin which may be isolated or associated with erance and compensatory hyperhidrosis in non‐scarred
a host of syndromic disorders. The most common presen- areas is well described. In the longer term some (18%)
tation is a localized congenital area of absent skin or scar infants develop recurrent herpes simplex skin infections
on the scalp in an otherwise healthy neonate (Fig. 11.8). to which these children may be vulnerable. There is
emerging evidence to suggest that in utero infection with
herpes simplex virus is the cause of CEVD, which explains
the high incidence of post-natal recurrence.

Differential diagnosis. The primary differential diagnosis


is congenital infection. Lesions may closely mimic those
of disseminated congenital herpes simplex and varicella
zoster virus as well as Candida albicans infection. Eroded
or ulcerated lesions may at birth resemble those seen
in forms of aplasia cutis congenita. Transient bullous
dermolysis of the newborn may present with similar
extensive erosions, but these lesions are superficial
and heal rapidly without scarring.

Laboratory/histology findings. Skin biopsy findings are


very nonspecific and depend on the age of the lesions.
Early histopathological features include dermal oedema
and a mixed infiltrate consisting mainly of neutrophils.
No features of vasculitis or thrombosis have been noted.
Fig. 11.8  Aplasia cutis congenita of the scalp associated with the ‘hair Direct immunofluorescence reveals nonspecific deposits
collar’ sign in a 3‐month‐old infant. of IgG, IgA, IgM, C3 and fibrin [118]. Biopsies of older
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 151

lesions show characteristic scarring with either decreased 26 Ferrándiz C, Coroleu W, Ribera M et  al. Sterile transient neonatal
pustulosis is a precocious form of erythema toxicum neonatorum.
or absent follicular and eccrine structures [119].
Dermatology 1992;185:18–22.
27 Finelt N, Kristal L. Patient characteristics of neonatal eosinophilic
Treatment and  prevention. No specific treatment is pustulosis. Pediatr Dermatol 2013;30:e204–7.
known to be effective in improving the natural history of 28 Ofuji S, Ogino A, Horio T et al. Eosinophilic pustular folliculitis. Acta
Derm Venereol 1970;50:195–203.
the scarring and associated alopecia. One case report has 29 Giard F, Marcoux D, McCuaig C et al. Eosinophilic pustular folliculitis

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
described the successful use of silicone dressings and skin (Ofuji disease) in childhood: a review of four cases. Pediatr Dermatol
massage in the neonatal period to prevent cutaneous con- 1991;8:189–93.
30 Kasai Y, Uchida S, Sakuma M. Eosinophilic pustular folliculitis. Jpn J
tractures [120].
Dermatol 1981;91:1029–35.
31 Lucky AW, Esterly NB, Heskel N et al. Eosinophilic pustular folliculi-
References tis in infancy. Paediatr Dermatol 1984;1:202–6.
1 Moosavi Z, Hosseini T. One year survey of cutaneous lesions in 1000 32 Taieb A, Basan‐Andrieu L, Maleville J. Eosinophilic pustulosis of the
consecutive newborns. Pediatr Dermatol 2006;23:61–3. scalp in childhood. J Am Acad Dermatol 1992;27:55–60.
2 Rivers JK, Frederiksen PC, Dibdin C. A prevalence survey of derma- 33 Asgari M, Leiferman KM, Piepkorn M et  al. Neonatal eosinophilic
tosis in the Australian neonate. J Am Acad Dermatol 1990;23:77–81. pustulosis. Int J Dermatol 2006;45:131–4.
3 Ferahbas A, Utas S, Akcakus M et al. Prevalence of cutaneous findings 34 Vicente J, España A, Idoate M et al. Are eosinophilic pustular follicu-
in hospitalized neonates: a prospective observational study. Pediatr litis of infancy and infantile acropustulosis the same entity? Br J
Dermatol 2009;26:139–42. Dermatol 1996;135:807–9.
4 Ekiz Ö, Gül Ü, Mollamahmutoğ lu L et  al. Skin findings in new- 35 Picone Z, Madile BM. Eosinophilic pustular folliculitis (Ofuji’s
borns and their relationship with maternal factors: observational disease) in a newborn. Pediatr Dermatol 1992;9:178.
research. Ann Dermatol 2013;25:1–4. 36 Buckley DA, Munn SE, Higgins EM. Neonatal eosinophilic pustular
5 Luna PC, Larralde M. Profuse congenital familial milia with absent folliculitis. Clin Exp Dermatol 2001;26:251–5.
dermatoglyphics (Basan’s Syndrome): description of a new family. 37 Hernandez–Martin A, Nuno‐Gonzalez A, Colmenero I et  al.
Pediatr Dermatol 2012;29:527–9. Eosinophilic pustular folliculitis in infancy: a series of 15 cases and
6 Lloyd BM, Braverman AC, Anadkat MJ. Multiple facial milia in review of the literature. J Am Acad Dermatol 2012;68:150–5.
patients with Loeys‐Dietz syndrome. Arch Dermatol 2011;147:223–6. 38 Nomora T, Katoh M, Yamamoto Y et al. Eosinophilic pustular follicu-
7 Epstein W, Klingman AM. The pathogenesis of milia and benign litis: The transition in sex differences and interracial characteristics
tumors of the skin. J Invest Dermatol 1956;26:1–11. between 1965 and 2013. J Dermatol 2015;42:343–52.
8 Hidano A, Purwoko R, Jitsukawa K. Statistical survey of skin changes 39 Ramdial PK, Morar N, Dlova et  al. HIV‐associated eosinophilic
in Japanese neonates. Pediatr Dermatol 1986;3:140–4. folliculitis in an infant. Am J Dermatopathol 1999;126:29–31.
9 Shelley WB, Horvath PN. Experimental miliaria in man; production 40 Bull RH, Harland CA, Fallowfield ME et al. Eosinophilic folliculitis: a
of sweat retention anidrosis and miliaria crystallina by various kinds self‐limiting illness in patients being treated for haematological
of injury. J Invest Dermatol 1950;14:9–20. malignancy. Br J Dermatol 1993;129:178–82.
10 Haas N, Henz BM, Weigel H. Congenital miliaria crystallina. J Am 41 Rybojad M, Guibal F, Vignon‐Pennamen MD et al. [Eosinophilic pus-
Acad Dermatol 2002;47(5 Suppl):S270–2. tulosis in an infant accompanied by immune deficit]. Ann Dermatol
11 Babu TA, Sharmila V. Congenital miliaria crystallina in a term neo- Venereol 1999;126:29–31.
nate born to a mother with chorioamnionitis. Pediatr Dermatol 2012;29:3. 42 Eberting CL, Davis J, Puck JM et al. Dermatitis and the newborn rash
12 Argoubi H, Fitchner C, Richard O et al. [Pustular miliaria rubra and of hyper‐IgE syndrome in infants and children. Arch Dermatol
systemic type 1b pseudohypoaldosteronism in a newborn]. Ann 2004;140:1119–25.
Dermatol Venereol 2007;134(3 Pt 1):253–6. 43 Miura H, Shoda Y, Adachi J. Eosinophilic pustular folliculitis in
13 Monteagudo B, Labandeira J, Cabanillas M et al. Prospective study of infancy in a Japanese neonate. Pediatr Dermatol 2004;21:615–16.
erythema toxicum neonatorum: Epidemiology and predisposing fac- 44 Ota T, Hata Y, Tanikawa A et  al. Eosinophilic pustular folliculitis
tors. Pediatr Dermatol 2012;29:166–8. (Ofuji disease): indomethacin as a first choice of treatment. Clin Exp
14 Haveri FTTS, Inamadar AC. A cross‐sectional prospective study of Dermatol 2001;26:179–81.
cutaneous lesions in newborn. ISRN Dermatology 2014:360590. 45 Toyonaga E, Abe R, Moriuchi R et  al. Indomethacin for refractory
15 Zagne V, Fernandes NC. Dermatoses in the first 72 h of life: A clinical infantile eosinophilic pustular folliculitis. JAMA Dermatol 2013;149:
and statistical survey. Indian J Dermatol Venereol Leprol 2011;77: 367–8.
470–6. 46 Patel NP, Laguda B, Roberts NM et al. Treatment of eosinophilic pus-
16 Sadana DJ, Sharma YK, Chaudhari ND et al. A clinical and statisti- tulosis of infancy with topical tacrolimus. (Letter). Br J Dermatolol
cal survey of cutaneous changes in the first 120 hours of life. Indian 2012;167: 184–95.
J Dermatol 2014;59:552–7. 47 Imai M, Katho S, Sakamoto K et  al. A case of eosinophilic pustular
17 Kulkarni ML, Singh R. Normal variants of skin in neonates. Indian J folliculitis in a child started initially only with plantar pustular
Dermatol Venereol Leprol 1996;62:83–6. lesions. Rinsho Hifuka 2011;65:227–330.
18 Sachdeva M, Kaur S, Nagpal M et al. Cutaneous lesions in newborn. 48 Kahn G, Rywlin AM. Acropustulosis of infancy. Arch Dermatol
Indian J Dermatol Venereol Leprol 2002;68:334–7. 1979;115:831–3.
19 Liu C, Feng J, Qu R et al. Epidemiologic study of the predisposing 49 Jarrat M, Ramsdell W. Infantile acropustulosis. Arch Dermatol.
factors in erythema toxicum neonatorum. Dermatology 2005;210: 1979;115:834–6.
269–72. 50 Kahn G. Reply. (Letter). J Am Acad Dermatol 1984;11:896.
20 Gokdemir G, Erdogan HK, Koslu A et al. Cutaneous lesions in Turkish 51 Good L, Good T, High W. Infantile acropustulosis in internationally
neonates born in a teaching hospital. Indian J Dermatol Venereol Leprol adopted children. J Am Acad Dermatol 2011;65:763–71.
2009;75:638. 52 Mancini AJ, Frieden IJ, Paller AS. Infantile acropustulosis revisited:
21 Menni S, Boccardi D, Crosti C. Neonatal toxic erythema: clinico‐ history of scabies and response to topical corticosteroids. Pediatr
epidemiologic characteristics and recent pathogenic hypothesis. Dermatol 1998;15:337–41.
Pediatr Med Chir 2005;27:22–5. 53 Dromy R, Raz A, Metzker, MD. Infantile acropustulosis. Pediatr
22 Leung AK. Erythema toxicum neonatorum present at birth. Dermatol 1991;8:284–87.
J Singapore Paediatr Soc 1986;28:163–6. 54 Elpern DJ. Infantile acropustulosis and antecedent scabies. (Letter).
23 Maffei FA, Michaels MG, Wald ER. An unusual case of erythema J Am Acad Dermatol 1984;11:895.
toxicum scrotal pustules present at birth. Arch Pediatr Adolesc 55 Prendiville JS. Infantile acropustulosis – how often is it a sequela of
Med;150:649–50. scabies? Pediatr Dermatol 1995;12:275–6.
24 Plantin P et la Société française de dermatopédiatrie. Érythème 56 Bjornberg A, Friis B. Persistent pustulosis in children adopted from
toxique du nouveau‐né. Ann Dermatol Venereol 2010;137:150–2. Asia; a sequel of scabies? Int J Dermatol 1978;17:69–73.
25 Ramamurthy RS, Reveri M, Esterly NB et  al. Transient neonatal 57 Truong AL, Truong MD, Esterly N. Atypical acropustulosis of infancy.
pustular melanosis. J Pediatr;88:831–5. Int J Dermatol 1997;36:677–99.
152 Section 2  Skin Disorders of the Neonate and Young Infant

58 Kimura M, Higuchi T, Yoshida M. Infantile acropustulosis treated 88 Sachs JA, Leonard J, Awad J et  al. A comparative serological and
successfully with topical maxacalcitol. Acta Derm Venerol 2010;91: molecular study of linear IgA disease and dermatitis herpetiformis.
363–4. Br J Dermatol 1988;118:759–64.
59 Lucky AW, McGuire JS. Infantile acropustulosis with eosinophilic 89 Gluth MB, Witman PM, Thompson DM. Upper aerodigestive tract
pustules. J Pediatr 1982;100:428–9. complications in a neonate with linear IgA bullous dermatosis. Int J
60 Falanga V. Infantile acropustulosis with eosinophilia. J Am Acad Pediatr Otorhinolaryngol 2004;68:965–70.
Dermatol 1985;13:826–8. 90 Hurza LL, Mallory SB, Fitzgibbons J et  al. Linear IgA bullous
61 Vignon‐Pennamen MD, Wallach D. Infantile acropustulosis. Arch dermatitis in a neonate. Pediatr Dermatol 1993;10:171–6.
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Dermatol 1986;122:1155–60. 91 Smahi A, Courtois G, Vabres P et  al. Genomic rearrangement in


62 Bernier V, Weill FX, Hirigoyen V et al. Skin colonization by Malassezia NEMO impairs NF‐kappaB activation and is a cause of incontinentia
species in neonates: a prospective study and relationship with neona- pigmenti. The International Incontinentia Pigmenti (IP) Consortium.
tal cephalic pustulosis. Arch Dermatol 2002;138:215–18. Nature 2000;405:466–72.
63 Ayhan M, Sancak B, Karaduman A et al. Colonisation of neonate skin 92 Orphanet Report Series. Prevalence and incidence of rare diseases:
by Malassezia species: Relationship with neonatal cephalic pustulosis Bibliographic data. 2018. Listed in alphabetical order of disease or
J Am Acad Dermatol 2007;57:1012–18. group of diseases: http://www.orpha.net/orphacom/cahiers/
64 Aractingi S, Cadranel S, Reygagne P et  al. Pustulose neonatale docs/GB/Prevalence_of_rare_diseases_by_alphabetical_list.pdf
induite par Malazessia furfur. Ann Dermatol Venereol 1991;118: (accessed 16 October 2018).
856–8. 93 Minić S, Trpinac D, Obradović M. Incontinentia pigmenti diagnostic
65 Rapelanoro R, Mortureux P, Couprie B et  al. Neonatal Malazessia criteria update. Clin Genet 2014;85:536–42.
furfur pustulosis. Arch Dermatol 1996;132:190–3. 94 Fusco F, Bardaro T, Fimiani G et al. Molecular analysis of the genetic
66 Bardazzi F. Transient cephalic neonatal pustulosis. Arch Dermatol defect in a large cohort of IP patients and identification of novel
1997;133:528–30. NEMO mutations interfering with NF‐kappaB activation. Hum Mol
67 Niamba P, Weill FX, Sarlangue J et al. Is common neonatal cephalic Genet 2004;13:1763–73.
pustulosis (neonatal acne) triggered by Malassezia sympoialis? Arch 95 Carney RG Jr. Incontinentia pigmenti: a world statistical analysis.
Dermatol 1998;134:995–8. Arch Dermatol 1976;112:535–42.
68 Roberts SO. Pityrosporum orbiculare: incidence and distribution on 96 Minic S, Trpinac D, Obradovic M. Systematic review of central
clinically normal skin. Br J Dermatol 1969;81:264–9. nervous system anomalies in incontinentia pigmenti. Orphanet J
69 Koseki S, Takahashi S. Serial observations on the colonization of Rare Dis 2013;8:25.
Pityrosporum obiculare (ovale) on the facial skin surface of newborn 97 Pinheiro A, Mathew MC, Thomas M et al. The clinical profile of children
infants. Jpn J Med Mycol 1998;29:209–15. in India with pigmentary anomalies along the lines of Blaschko and cen-
70 Grattan CE, Peachey RD, Boon A. Evidence for a role of local trauma tral nervous system manifestations. Pediatr Dermatol 2007;24:11–17.
in the pathogenesis of erosive pustular dermatosis of the scalp. Clin 98 Minic S, Obradovic M, Kovacevic I et al. Ocular anomalies in incon-
Exp Dermatol 1988;13:7–10. tinentia pigmenti: literature review and meta‐analysis. Srp Arh
71 Pye RJ, Peachey RD, Burton JL. Erosive pustular dermatosis of the Celok Lek 2010;138:408–13.
scalp. Br J Dermatol 1979;100:559–66. 99 Phan TA, Wargon O, Turner AM. Incontinentia pigmenti case series:
72 Siegel D, Holland K, Philips R et al. Erosive dermatosis of the scalp clinical spectrum of incontinentia pigmenti in 53 female patients and
after perinatal scalp injury. Pediatr Dermatol 2006;23:533–6. their relatives. Clin Exp Dermatol 2005;30:474–80.
73 Shimada R, Masu T, Hanamizu H et  al. Infantile erosive pustular 100 François J. Incontinentia pigmenti (Bloch‐Sulzberger syndrome) and
dermatosis of the scalp associated with Klippel Feil syndrome. Acta retinal changes. Br J Ophthalmol 1984;68:19–25.
Derm Venerol 2010;90:200–1. 101 Swinney CC, Han DP, Karth PA. Incontinentia pigmenti: a com-
74 Sheckter C, Rommer E, Francis C et al. Scalp erosion in ankyloblepharon‐ prehensive review and update. Ophthalmic Surg Lasers Imaging
ectodermal defect‐cleft lip and/or palate (AEC syndrome): treatment Retina 2015;46:650–7.
with a cellular dermal matrix. J Craniofac Surg 2013;24:e28–30. 102 Wang X, Reid‐Sutton V, Omar‐Peraza‐Llanes J et al. Mutations in
75 Broussard KC, Berger TG, Rosenblum et al. Erosive pustular dermato- X‐linked PORCN, a putative regulator of Wnt signalling, cause of
sis of the scalp: A review with a focus on dapsone therapy. J Am Acad focal dermal hypoplasia. Nat Genet 2007;39:836–38.
Dermatol 2012;66:680–6. 103 Bostwick B, Van den Veyver IB, Sutton VR. Focal dermal hypoplasia.
76 Turrentine JE, Sokumbi O, Agim NG. Blisters and erosions in a In: Adam MP, Ardinger HH, Pagon RA et  al (eds) GeneReviews®
neonate. JAMA Dermatol 2014;150:1223–4. [Internet]. Seattle: University of Washington, Seattle:1993–2019
77 Zhao CY, Murrell DF. Blistering diseases in neonates. Curr Opin (updated 21 July 2016).
Pediatr 2016;28:500–6. 104 Navarro CL, Esteves‐Vieira V, Courrier S et  al. New ZMPSTE24
78 Al‐Mutairi N, Sharma AK, Zaki A et  al. Maternal and neonatal (FACE1) mutations in patients affected with restrictive dermopathy
pemphigoid gestationis. Clin Exp Dermatol 2004;29:202–4. or related progeroid syndromes and mutation update. Eur J Hum
79 Abrams ML, Smidt A, Benjamin L et al. Congenital epidermolysis Genet 2014;22:1002–11.
bullosa acquisita: vertical transfer of maternal autoantibody from 105 Ahmad Z, Phadke SR, Arch E et al. Homozygous null mutations in
mother to infant. Arch Dermatol 2011;147:337–41. ZMPSTE24 in restrictive dermopathy: evidence of genetic heteroge-
80 Lorente Lavirgen AI, Bernabeu‐Wittel J, Dominguez‐Cruz J et  al. neity. Clin Genet 2012;81:158–64.
Neonatal pemphigus foliaceus. J Pediatr 2012;161:768. 106 Matulevičienė A, Meškienė R, Morkū nienė A et al. Frame shift muta-
81 Daneshpazhooh M, Chams‐Davatchi C, Valikhani M et al. Pemphigus tions of the ZMPSTE24 gene in two siblings with restrictive dermop-
and pregnancy: a 23‐year experience. Indian J Dermatol Venereol athy. Clin Dysmorphol. 2016; 25(1): 7–11.
Leprol 2011;77:534. 107 Bodemer C, Hermine O, Palmérini F et al. Pediatric mastocytosis is a
82 Zhao CY, Chiang YZ, Murrell DF. Neonatal autoimmune blistering clonal disease associated with D816V and other activating c‐KIT
disease: a systematic review. Pediatr Dermatol 2016;33:367–74. mutations. J Invest Dermatol 2010;130:804–15.
83 Kardos M, Levine D, Gürcan HM et al. Pemphigus vulgaris in preg- 108 Hartmann K, Escribano L, Grattan C et al. Cutaneous manifesta-
nancy: analysis of current data on the management and outcomes. tions in patients with mastocytosis: Consensus report of the
Obstet Gynecol Surv 2009;64:739–49. European Competence Network on Mastocytosis; the American
84 Itsukaichi M, Takakuwa K, Yamaguchi M et al. Twins with neonatal Academy of Allergy, Asthma & Immunology; and the European
pemphigus vulgaris born to a mother with pemphigus vulgaris: a Academy of Allergology and Clinical Immunology. J Allergy Clin
case report. Pediatr Dermatol 2013;30:e59–60. Immunol 2016;137:35–45.
85 Wu H, Wang ZH, Yan A et al. Protection against pemphigus foliaceus 109 Koga H, Kokubo T, Akaishi M et al. Neonatal onset diffuse cutane-
by desmoglein 3 in neonates. N Engl J Med 2000;343:31–5. ous mastocytosis: a case report and review of the literature. Pediatr
86 Julapalli MR, Brandon KL, Rosales CM et al. Neonatal linear immu- Dermatol 2011;28:542–6.
noglobulin a bullous dermatosis: a rare presentation. Pediatr 110 Waters WJ, Lacson PS. Mast cell leukemia presenting as urticaria
Dermatol 2012;29:610–13. pigmentosa. Pediatrics 1957;19:1033–42.
87 Zone JJ, Taylor TB, Meyer LJ et al. The 97 kDA linear IgA bullous dis- 111 Brockow K, Akin C, Huber M et  al. Assessment of the extent of
ease antigen is identical to a portion of the extracellular domain of cutaneous involvement in children and adults with mastocytosis:
the 180kDa bullous pemphigoid antigen BPAg2. J Invest Dermatol relationship to symptomatology, tryptase levels, and bone marrow
1998;110:207–10. pathology. J Am Acad Dermatol 2003;48:508–16.
Chapter 11  Vesiculopustular, Bullous and Erosive Diseases of the Neonate 153

112 Arock M, Akin C, Hermine O, Valent P. Current treatment options in 117 Tlougan BE, Paller AS, Schaffer JV et  al. Congenital erosive and
patients with mastocytosis: status in 2015 and future perspectives. vesicular dermatosis with reticulated supple scarring: unifying
Eur J Haematol 2015;94:474–90. clinical features. J Am Acad Dermatol 2013;69:909–15.
113 Brockow K, Jofer C, Behrendt H et al. Anaphylaxis in patients with 118 Sadick NS, Shea CR, Schlessel JS. Congenital erosive and vesicular
mastocytosis: a study on history, clinical features and risk factors in dermatosis with reticulated supple scarring: a neutrophilic dermato-
120 patients. Allergy 2008;63:226–32. sis. J Am Acad Dermatol 1995;32:44–8.
114 Renke J, Lange M. Routine vaccinations in diffuse cutaneous masto- 119 Sidhu‐Malik NK, Resnick SD, Wilson BB. Congenital erosive and vesicu-
cytosis. J Allergy Clin Immunol Pract 2016;4:190. lar dermatosis healing with reticulated supple scarring: report of three

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
115 Stephan M J, Smith D W, Powzi J W et  al. Origin of scalp aplasia new cases and review of the literature. Pediatr Dermatol 1998;15:214–18.
cutis. J Pediatr 1982;101:850–3. 120 De Lange A, Bayet B, Debauche C et al. Congenital erosive and vesicu-
116 Marneros AG. Genetics of aplasia cutis reveal novel regulators of lar dermatosis with reticulated scarring in a newborn: an innovative
skin morphogenesis. J Invest Dermatol 2015;135:666–72. treatment using a silicone dressing. Pediatr Dermatol 2009;26:735–8.
154 

CHA PTER 1 2

Iatrogenic Disorders of the Newborn


Elia F. Maalouf 1 & Wilson Lopez2
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

1
 Neonatal Unit, Homerton University Hospital NHS Foundation Trust, London, UK
2
 Neonatal Unit, Barking, Havering and Redbridge University Hospitals NHS Trust, UK

Introduction, 154 Iatrogenic injury during pregnancy, 156 Iatrogenic skin disorders after birth, 159
Iatrogenic disorders of the newborn, 154 Iatrogenic injuries during labour, 157

Abstract profile, weak inflammatory response, unique extracellular matrix


c­ haracteristics and attenuated biochemical stress.
In spite of significant technical advances in neonatal care, skin
Iatrogenic injuries to the fetus and infant have longlasting
­injuries continue to occur. Although severe injuries are less frequent,
consequences. Healing occurs without scar formation during
this is an ongoing problem facing neonatal care, especially as
early gestation as opposed to the postnatal period. Various
greater numbers of very preterm babies are surviving. Adherence to
factors play a role in scarless wound healing during early gestation.
standard neonatal procedures and careful monitoring can ­prevent
Fetal wound healing involves a distinct growth factor and cytokine
most of these complications.

Key points anti‐inflammatory cytokine profile and distinct extracellular


matrix rich in specific collagen and hyaluronan, and decreased
biochemical stress.
• There are distinct differences between fetal and postnatal
• Different types of iatrogenic injuries occur in the antenatal and
wound healing.
postnatal periods.
• Fetal wound healing has a unique growth factor profile,
weakened inflammatory response with characteristic

Introduction in the next few days. Monocytes differentiate into tissue


macrophages after entering the wound site and subse-
Iatrogenic skin disorders can occur before labour, during quently are important regulators of repair. The inflamma-
delivery or after birth. They are most commonly caused tory stage is responsible for the secretion of numerous
by skin injury, which may lead to infection and scarring. growth factors and cytokines. Cytokines are very impor-
tant in wound healing. Interleukin (IL)‐1β and tumour
necrosis factor (TNF)‐α have been shown to regulate the
Iatrogenic disorders of the newborn
recruitment and function of neutrophils [2].
Scar formation in postnatal skin Tissue macrophages continue the debridement process
Tissue injury initiates bleeding, coagulation, inflamma- begun by granulocytes, but they also secrete substances
tion, cell replication, angiogenesis, epithelialization and that initiate the development of granulation tissue in the
matrix synthesis [1]. Shortly after injury, vessels rapidly next phase of wound repair. If the conditions in the
constrict. Coagulation then limits loss of blood and gener- wound warrant it, macrophages recruit additional inflam-
ates biologically active substances that begin the early matory cells. Lymphocytes contribute in much the same
and late inflammatory phases of tissue repair. Coagulation manner, but probably to a lesser degree. Wound fibro-
also releases biologically active products that convert blasts are derived from cells surrounding the wound that
fibroblasts and endothelial cells into their reparative change their differentiated phenotypes and become
mode. This forms new local tissue and capillary cells. mobile during the process of replication. As they migrate
The next phase of healing, inflammation, begins with into the wound during fibroplasia, they proliferate and
activation of complement and the initiation of the classic secrete extracellular matrix.
molecular cascade, which leads to infiltration of the During angiogenesis, growth and migration of
wound by granulocytes. These cells consume contaminat- cells take place. The growth of vascular endothelial
ing bacteria and debris and are largely replaced by monocytes cells occurs simultaneously with fibroplasia during

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 12  Iatrogenic Disorders of the Newborn 155

granulation tissue formation, stimulated in much the Characteristic features of fetal wound healing include
same manner as fibroblasts, namely by platelets and a weakened inflammatory response, decreased neutrophil
activated macrophage products. Angiogenesis results and macrophage infiltration, poor platelet aggregation
in the rich microvasculature that gives granulation tis- with decreased production of platelet‐derived growth
sue its deep red colour. Granulation tissue is a loose factor (PDGF) and TGF‐β and decreased production
matrix that appears in open wounds and is composed of pro‐inflammatory cytokines such as IL‐6 and IL‐8

SECTION 2: SKIN DISORDERS


of collagen, fibronectin and hyaluronic acid with dense [12,13].

OF NEONATES AND INFANTS


infiltration by macrophages, fibroblasts and capillary Fetal fibroblasts respond weakly to IL‐6 and IL‐8 compared
endothelial cells. In wounds exposed to the external to adult skin [14]. Fetal skin during early gestation
environment, re‐epithelialization begins within min- expresses high levels of IL‐10, which deactivates
utes after injury. Epithelial cells surrounding the defect macrophages and decreases the production of pro‐
become mobile and move into the wound. Within inflammatory cytokines [15].
1–2 days, epithelial cells at the wound edge begin pro- Fetal wound also has an abundance of type III collagen
liferating, and the new cells migrate either along an which is deposited in a fine reticular pattern. There is an
undisrupted basement membrane or across a provisional increased level of high‐molecular‐weight hyaluronan,
matrix of fibrin and fibronectin, which is then replaced which has a suppressive effect on aggregation of platelets
with basement membrane. Necrotic tissue and foreign and their release of growth factors [13,16–18].
bodies are gradually separated from the wound as the Biochemical forces may play a part in wound healing. It
epithelial cells migrate under them. Once epithelial is postulated that increased shear force induces local pro-
integrity is completed, the epithelium becomes attached duction of profibrotic growth factor [19]. The amniotic
to the new basement membrane, thus creating a b ­ arrier fluid provides the fetus with a unique environment. Fetal
to further contamination and water loss. skin wounds are continually bathed in the warm sterile
Repair is not complete for some time after the epithe- amniotic fluid, which is known to be rich in growth fac-
lium covers the wound and there is no longer a risk of tors that are crucial to fetal development [20]. The fluid is
wound separation. Remodelling is largely concerned also a rich source for extracellular matrix components
with the removal of loose extracellular matrix materials such as hyaluronic acid and fibronectin [21–23]. Amniotic
such as fibronectin and the slow deposition of type I col- fluid could modulate fetal skin wound repair simply by
lagen in fibrils, and complete remodelling takes months. allowing hyaluronic acid and fibronectin to come into
During remodelling, the tissue becomes stronger and direct contact with the wounds and by providing growth
more flexible. The end result is scar formation. A scar factors to stimulate fetal wound cells to make a unique
lacks some components of normal tissue and is therefore wound matrix [24].
less functional than the surrounding uninjured area. The Some of the properties of fetal skin wound healing may
major benefit of repair by scar is the rapid return of tissue reflect the development of fetal skin. Between 4 and 24
integrity. weeks’ gestation, the outermost layer of human skin is
called the periderm, which has the appearance of multi-
Fetal wound healing ple microvilli projecting from the surface of the skin. It is
Despite many reports of cutaneous scars caused by possible that the periderm has an absorptive function as
antenatal procedures performed for diagnostic or ther- an amniotic fluid dialysate membrane. What effect the
apeutic purposes [3,4], there have been several obser- changing histology of fetal skin during development has
vations that fetal skin wounds heal without scarring on wound healing remains to be fully determined. The
[5,6]. Recent experimental work demonstrates distinct fetus lies in a sterile environment and has a minimal
differences between fetal and postnatal skin wound inflammatory response to injury. This may play a vital
healing [7–9]. The nature of these differences remains role in the unique fetal repair process, considering the
unknown, but there are two general ideas that could important role that inflammation plays in adult tissue
provide an explanation: differences in fetal wound wound repair.
environment and differences in fetal cells, or a combi- The fetus grows and develops at an astounding rate. It
nation of the two. can be physically wounded but physiologically does not
Fetal wound healing differs from postnatal healing in respond to injury in an adult‐like fashion.
having a unique growth factor profile, weakened inflam- Fetal tissue repair may be a process closely resembling
matory response with characteristic anti‐inflammatory regeneration and growth rather than healing by scar for-
cytokine profile and distinct extracellular matrix rich in mation [25]. Consequent to injury, embryos mount a poor
specific collagen and hyaluronan, and decreased bio- inflammatory response. The levels of TGF‐ β1 and β2 are
chemical stress much lower in embryonic wounds when compared to
A different spectrum of growth factor subgroups is adult wounds [26]. Genetic analysis of fetal and postnatal
believed to be at play in fetal wound healing. Transforming cutaneous wounds sought to identify genes with differen-
growth factor (TGF)‐β and its isoforms secreted by many tial expression and proregenerative function. It was sug-
cell types are involved in tissue repair [10]. The ratio of gested that skin regeneration during wound repair in the
TGF‐β3 expression in a fetal wound is high in comparison fetus may be coordinated by multiple gene products
with TGF‐β1. Exactly the opposite pattern is observed in and that the pattern of expression of these genes declines
adults with hypertrophied scar [11]. with time [27].
156 Section 2  Skin Disorders of the Neonate and Young Infant

Iatrogenic injury during pregnancy Chorionic villus sampling


Chorionic villus sampling (CVS) involves the use of a
Amniocentesis cannula or biopsy forceps under ultrasound guidance to
The aspiration of amniotic fluid is now a common
take a small biopsy of villi from the chorion fundosum.
procedure in obstetric practice. Its regular use was first
The villi are a good source of DNA and are (nearly always)
advocated by Bevis [28] in the management of rhesus
genetically, chromosomally and biochemically identical
­disease. Amniocentesis is performed under ultrasound
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

to the fetus. The procedure can be performed using either


guidance using a 21‐gauge disposable spinal needle
the transcervical or the transabdominal route (Fig. 12.1).
with a stylet and a disposable syringe. Indications for
The main advantage of CVS is that it enables first‐trimester
amniocentesis are shown in Box 12.1 [29].
fetal diagnosis, therefore providing early reassurance,
Early amniocentesis (before 15 weeks of pregnancy) is
­termination or treatment [29].
associated with higher fetal loss and increased complica-
As with amniocentesis, the main complication is
tion rates [30]. Direct fetal injury can be minimized by
pregnancy loss, occurring in about 2% [41]. There have
ultrasound‐guided amniocentesis [31]. Skin injuries
been reports of an increased incidence of cutaneous
during amniocentesis have been greatly reduced since
haemangiomas in infants exposed to CVS antenatally.
the introduction of ultrasound guidance. There is an
In one study, the reported incidence was 21%, which
increased risk of fetal injury during transplacental
was three times higher than the incidence in the general
amniocentesis therefore needle site should be selected
population. It was found that the incidence of cutaneous
to avoid the placenta where possible [32–36].
haemangiomas, defined as strawberry birth marks, was
Some studies have suggested that there is increased
significantly higher after CVS compared with second‐
incidence of indirect fetal injuries such as talipes equi-
trimester amniocentesis. This increased incidence was
novarus, congenital dislocation of hips or respiratory
largely confined to patients who underwent a transcer-
problems in infants born after amniocentesis compared
vical CVS procedure [42]. The mechanism by which
with controls who did not undergo amniocentesis
haemangiomas develop in the general population is not
[31,37,38]. A case of patellar disruption following
understood and it would therefore be difficult to explain
­amniocentesis has been described [39]. Mid trimester
the above findings.
amniocentesis has been reported to cause fetal injury,
primarily in isolated case reports, and these attributions
Other antenatal procedures
have mostly been based on association rather than
Other procedures that may leave scars or needle marks on
direct evidence. These fetal injuries include exsanguina-
the skin of the newborn include:
tion, skin dimples, ocular injury and intracranial and
• fetal skin biopsies, which may be carried out for prenatal
bowel abnormalities [40].
diagnosis of the severe genodermatoses; with the
The likelihood of injury increases with the number of
development of DNA‐based prenatal diagnosis, this
attempts to obtain fluid [3]. In third‐trimester amnio-
technique is now rarely required
centesis, fetal skin injury has been reported with an
• fetal liver biopsy, which is performed for the diagnosis
incidence of 0.6–2% [3]. Total pregnancy loss has been
of rare inborn errors of metabolism when other tests are
reported to be about 1.9% with a 95% confidence inter-
not suitable, e.g. defects of the urea cycle enzymes
val of 1.4–2.5 [41].
• fetal tumour biopsy, which can be used to confirm the
diagnosis of teratomas or congenital adenomatoid mal-
formations of the lung

Box 12.1  Indications for amniocentesis

Before 20 weeks

• Chromosome abnormalities (maternal age, translocation carrier,


previous affected child)
• Fetal sexing in X‐linked disorders
• Neural tube defects (raised α‐fetoprotein or acetylcholinesterase
levels)
• Inborn errors of metabolism
• DNA analysis

After 20 weeks Syringe


• Bilirubin level estimation
• Phospholipids (for fetal pulmonary maturity)
• Fetal maturity
• Amniography or fetography

Speculum Cervix Portex catheter


Source: Adapted from Rodeck & Pandya 2001 [29].
Fig. 12.1  The transcervical approach for chorionic villus sampling.
Chapter 12  Iatrogenic Disorders of the Newborn 157

• aspiration of fluid collections, such as fetal urine from a involves the breech and scrotum and very unpleasant in a
dilated bladder or a pleural effusion, which can be face presentation. The bruising normally clears sponta-
­performed with ultrasound guidance neously in a few days. No specific treatment is needed,
• therapeutic procedures, such as intrauterine shunting but if there is extensive bruising early phototherapy
of obstructive uropathy, hydrocephalus or hydrotho- for hyperbilirubinaemia may be indicated.
rax, and aspiration of fluid from renal cysts, ovarian A cephalhaematoma (Fig.  12.4) is a subperiosteal

SECTION 2: SKIN DISORDERS


cysts or ascites, all of which can be performed antena- haematoma that produces a lump over the scalp limited

OF NEONATES AND INFANTS


tally using ultrasound guidance. by the cranial sutures. Swelling is usually not visible until
several hours after birth, because subperiosteal bleeding
is a slow process. It is of little importance, although it may
Iatrogenic injuries during labour be many months before it finally disappears. The swelling
Fetal monitoring may begin to calcify by the end of the second week and a
Continuous fetal heart rate monitoring is indicated ­during few may remain for years as bony protrusions. No treat-
labour to monitor fetuses at risk of hypoxia [43]. This is ment is required, although phototherapy may be necessary
achieved by using an ultrasonic transducer placed on the for jaundice.
maternal abdominal wall or by using an electrode Rarely, a bleed may occur under the aponeurosis (sub-
attached to the fetal scalp (Fig.  12.2). The latter method aponeurotic haemorrhage); the bleeding is not confined
may produce bleeding at various levels in the scalp, to a single cranial bone like a cephalhaematoma but
including perforation of the periosteum and subperi- spreads rapidly over the head and down towards the
osteal haemorrhage. Whenever fetal heart rate monitor-
ing is being employed, it is vital that fetal acid–base status
can be established from a scalp blood sample. A falling
blood pH level indicates an impending problem and that
action aimed at delivering the mother will probably be
necessary. Scalp blood samples are obtained by perform-
ing a small incision in the scalp using the instrument
shown in Fig.  12.3. These injuries are a potential site of
serious neonatal infection, which may be acquired during
delivery. Persistent scalp bleeding due to fetal coagulopa-
thy has been reported following fetal blood sampling
[44]. Scalp gonococcal abscess following intrapartum fetal
monitoring is another rare complication [45].

Injuries acquired during normal or


operative deliveries
During labour, a caput succedaneum may develop. This
is defined as soft tissue oedema and bruising of the pre- Fig. 12.3  Lancet used to obtain fetal scalp blood for pH monitoring. The
senting part. The swelling looks uncomfortable when it blade measures 2 mm.

Fig. 12.2  Fetal scalp electrode for heart rate monitoring (note the hook
that penetrates the skin). Fig. 12.4  Left parietal cephalhaematoma in a term baby.
158 Section 2  Skin Disorders of the Neonate and Young Infant

eyes. A significant fraction of the blood volume may be


lost into this haematoma.
Bruising may be seen after manipulative deliveries and
often in preterm infants. Vaginal breech deliveries also
predispose to bleeding into the muscles of the legs and
buttocks resulting from hypoxic capillary damage and
SECTION 2: SKIN DISORDERS

increased hydrostatic pressure in the fetal dependent


OF NEONATES AND INFANTS

parts while awaiting delivery of the head [46].

Forceps deliveries
In addition to the above, forceps deliveries may also
cause erythema, abrasions, ecchymosis and subcutane-
ous fat necrosis of facial or scalp soft tissues. Their loca-
tion depends on the area of application of the forceps.
Moreover, in circumstances in which there is reduced (a)
area of contact, there is a predisposition to slipping of
the forceps, which can result in visible facial and cra-
nial markings [47]. Bruising of the skin after a forceps
delivery may be so extensive as to produce anaemia or
jaundice.

Vacuum extractors
Delivery by the vacuum extractor (ventouse) is occa-
sionally used instead of forceps when there is slow pro-
gress of labour. The vacuum is created using either a
metal or a soft (plastic or silicone rubber) cup applied to
the scalp and the baby is extracted by traction on the
handle (Fig. 12.5). When air is evacuated from the cup,
the scalp is sucked into the space inside the cup, so
­producing the chignon, which has a greater diameter
(b)
above the rim than the rim itself (Fig. 12.6). Apart from
Fig. 12.6  (a) Severe skin trauma and chignon following ventouse extraction.
producing jaundice, the chignon is a benign lesion in the
The ventouse is more usually applied over the vertex. (b) Later appearance
majority of cases and it heals over a few days leaving no of the lesion.
scars, but rarely it becomes lacerated and infected.
Neonates delivered by vacuum have a higher incidence
of cephalhaematomas [48].
Abrasions to the scalp may also occur as a result of
34 weeks’ gestation because of a significant incidence of
rotation or movement of the cap on the skin, caused by a
complications [49]. Alopecia is a rare but important
badly placed cap. Sudden detachment of the cap can also
­complication of ventouse deliveries [50].
cause skin damage. Ventouse extraction (vacuum
extraction) can be used only for cephalic presentations
Caesarean section
and is contraindicated in preterm deliveries before
Scalpel lacerations occurring during caesarean section
may be deep enough to require suturing and may occa-
sionally become infected.

References
1 Jennings RW, Hunt TK. Overview of postnatal wound healing. In:
Adzick NS, Longaker MT (eds) Fetal Wound Healing. New York:
Elsevier, 1992:25–52.
2 Vegesna V, McBride WH, Taylor JMJ, Withers HR. The effect of
interleukin‐1 beta or transforming growth factor beta on radiation‐
impaired murine skin wound healing. J Surg Res 1995;59:699–704.
3 Galle PC, Meis PJ. Complications of amniocentesis: a review. J Reprod
Med 1982;27:149–55.
4 Morrison WA, Hurley JV, Ahmad TS, Webster HR. Scar formation
after skin injury to the human foetus in utero or the premature
­neonate. Br J Plastic Surg 1999;52:6–11.
5 Rowlatt U. Intrauterine wound healing in a 20‐week human fetus.
Virchow’s Arch 1979;381:353–61.
6 Bullard KM, Longaker MT, Lorenz HP. Fetal wound healing: current
biology. World J Surg 2003;27:54–61.
7 Viljanto J, Thomasson B, Pikkarainen T et  al. Fetal connective tissue
Fig. 12.5  Ventouse cup and handle. regeneration. Acta Chir Scand 1975;141:85–9.
Chapter 12  Iatrogenic Disorders of the Newborn 159

8 Adzick NS, Harrison MR, Glick PL et al. Comparison of fetal, new- 35 Reid KP, Gurrin LC, Dickinson JE et al. Pregnancy loss rates following
born and adult wound healing by histologic, enzyme histochemical, second trimester genetic amniocentesis. Aust N Z J Obstet Gynaecol
and hydroxyproline determination. J Pediatr Surg 1985;20:315–19. 1999;39:281.
9 Hallock GG, Rice DC, McClure HM. In utero lip repair in the rhesus 36 Kalogiannidis I, Prapa S, Dagklis T et  al. Amniocentesis‐related
monkey: an update. Plast Reconstr Surg 1987;80:855–8. adverse outcomes according to placental location and risk factors
10 Whitby DJ, Ferguson MW. Immunohistochemical localization of for fetal loss after midtrimester amniocentesis. Clin Exp Obstet
growth factors in fetal wound healing. Dev Biol 1991;147:207–15. Gynecol 2011;38:239.
11 Lin RY, Sullivan KM, Argenta PA et  al. Exogenous transforming 37 Sant‐Cassia LJ, MacPherson MB, Tyack AJ. Midtrimester amniocente-

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
growth factor beta amplifies its own expression and induces scar forma- sis: is it safe? A single centre controlled prospective study of 517 con-
tion in a model of human fetal skin repair. Ann Surg 1995;222:146–54. secutive amniocenteses. Br J Obstet Gynaecol 1984;91:736
12 Mast BA, Albanese CT, Kapadia S. Tissue repair in the fetal intesti- 38 Cederholm M, Haglund B, Axelsson O. Infant morbidity following
nal tract occurs with adhesions, fibrosis, and neovascularization. amniocentesis and chorionic villus sampling for prenatal karyotyp-
Ann Plast Surg 1998;41:140–4. Discussion 144–7. ing. BJOG 2005;112:394.
13 Olutoye OO, Alaish SM, Carr ME et al. Aggregatory characteristics 39 Epley SL, Hanson JW, Cruikshank DP. Fetal injury with midtrimester
and expression of the collagen adhesion receptor in fetal porcine diagnostic amniocentesis. Obstet Gynecol 1979;53:77–80.
platelets. J Pediatr Surg 1995;30:1649–53. 40 Seeds JW. Diagnostic mid trimester amniocentesis: how safe? Am J
14 Peranteau WH, Zhang L, Muvarak N et  al. IL10 over expression Obstet Gynecol 2004;191:607.
decreases inflammatory mediators and promotes regenerative heal- 41 Mujezinovic F, Alferirevic Z. Procedure‐related complications of
ing in an adult model of scar formation. J Invest Dermatol amniocentesis and chorionic villous sampling: a systematic review.
2008;128:1852–60. Obstet Gynecol 2007;110:1425–6.
15 Gordon A, Kozin ED, Keswani SG et al. Permissive environment in 42 Burton KB, Schultz JC, Burd AB. An increased incidence of haeman-
postnatal wounds induced by adenoviral‐mediated overexpression of giomas in infants born following chorionic villous sampling. Prenat
the anti‐inflammatory cytokine interleukin 10 prevents scar forma- Diagn 1995;15:209–14.
tion. Wound Repair Regen 2008;16:70–9. 43 Whittle MJ, Martin WL. Fetal monitoring in labour. In: Chamberlain
16 Cuttle L, Nataatmadja M, Fraser JF et  al. Collagen in the scarless G (ed.) Turnbull’s Obstetrics. London: Churchill Livingstone,
fetal skin wound: detection with picrosirius‐polarization. Wound 2001:439–50.
Repair Regen 2005;13:198–204. 44 Pachydakis A, Belgaumkar P, Sharmah A. Persistent scalp bleeding
17 Merkel JR, DiPaolo BR, Hallok GG, Rice DC. Type 1 and type 3 colla- due to fetal coagulopathy following fetal blood sampling. Int J
gen content of healing wounds in fetal and adult rats. Proc Soc Exp Gynaecol Obstet 2006;92:69–70.
Biol Med 1988;187:493–7. 45 Asnis DS, Brennessel DJ. Gonococcal scalp abscess: a risk of intrauter-
18 Burd DA, Longaker MT, Adzick NS et al. Foetal wound healing in a ine monitoring. Clin Pediatr 1992;31:316–17.
large animal model: the deposition of collagen is confirmed. Br J Plast 46 Ralis ZA. Birth trauma to muscles in babies born by breech delivery
Surg 1990;43:571–7. and its possible fetal consequences. Arch Dis Child 1975;50:4–13.
19 Cass DL, Bullard KM, Sylvester KG et  al. Wound size and gesta- 47 Foster FMC. Robert Barnes and his obstetric forceps. Aust NZ J Obstet
tional age modulate scar formation in fetal wound repair. J Pediatr Gynecol 1971;11:139.
Surg 1997;32:411–15. 48 Johnson JH, Figueroa R, Garry D et  al. Immediate maternal and
20 Mulvihill SJ, Stone MM, Fonkalsrud EW. Trophic effect of amniotic neonatal effects of forceps and vacuum assisted deliveries. Obstet
fluid on fetal gastrointestinal development. J Surg Res 1986;40:291–6. Gynecol 2004;103:513–18.
21 Dahl L, Hopwood JJ, Laurent BG et al. The concentration of hyaluro- 49 Chang AMZ, Lau TK. Operative vaginal delivery. In: Chamberlain G
nate in amniotic fluid. Biochem Med 1983;30:280–3. (ed) Turnbull’s Obstetrics. London: Churchill Livingstone, 2001:
22 Harris MC, Mennuti MT, Kline JA et  al. Amniotic fluid fibronectin 581–99.
concentrations with advancing gestational age. Obstet Gynecol 50 Aron R, Gordon W. Cicatricial alopecia: a complication of vacuum
1988;72:593–5. extraction. S Afr Med J 1972;19:671.
23 Kratz G, Palmer B, Haegerstrand A. Effects of keratinocyte condi-
tioned medium, amniotic fluid and EGF in reepithelialization of
human skin wounds in vitro. Eur J Plastic Surg 1995;18:209–13. Iatrogenic skin disorders after birth
24 Longaker MT, Adzick NS, Hall J et al. Studies in fetal wound healing.
VII. Fetal wound healing may be modulated by elevated hyaluronic The widespread availability of intensive care facilities for
acid stimulating activity in amniotic fluid. J Pediatr Surg 1990;25:430–3. the newborn has resulted in the survival of many infants,
25 Adzick NS, Longaker MT. Characteristics of fetal tissue repair. In:
Adzick NS, Longaker MT (eds) Fetal Wound Healing. New York:
particularly preterm babies of low birthweight, who
Elsevier, 1992:53–70. would previously have died. Effective treatment may,
26 Ferguson MW. Wound healing‐scar wars. Ulster Med J 1998;(Suppl unfortunately, bring in its wake serious problems in the
1):37–40. form of iatrogenic disease which may cause or contribute
27 Colwell AS, Longaker MT, Peter Lorenz H. Identification of differ-
entially regulated genes in fetal wounds during regenerative repair. to rapid demise, or whose effects may accompany these
Wound Repair Regen 2008;16:450–9. infants for the rest of their lives.
28 Bevis DC. The antenatal prediction of haemolytic disease of the new- Most reports of iatrogenic damage in the literature
born. Lancet 1952;23:395–8.
29 Rodeck CH, Pandya PP. Prenatal diagnosis. In: Chamberlain G (ed.)
concentrate on tragic accidents which are potentially
Turnbull’s Obstetrics. London: Churchill Livingstone, 2001:169–96. avoidable by more careful use of equipment.
30 American College of Obstetricians and Gynecologists. ACOG Practice Fortunately, however, these major incidents are rare.
Bulletin No. 88, December 2007. Invasive prenatal testing for aneu- The less serious but more common iatrogenic damage,
ploidy. Obstet Gynecol 2007;110:1459.
31 Tabor A, Philip J, Madsen M et  al. Randomised controlled trial of mainly to the skin, is usually wrongly considered the
genetic amniocentesis in 4606 low‐risk women. Lancet 1986;1:1287. inevitable price to be paid for survival in some pre-
32 Kappel B, Nielsen J, Brogaard Hansen K et al. Spontaneous abortion term infants.
following mid‐trimester amniocentesis. Clinical significance of pla-
Major risk factors of iatrogenic skin injuries after birth
cental perforation and blood‐stained amniotic fluid. Br J Obstet
Gynaecol 1987;94:50. are low birthweight, lower gestational age, prolonged
33 Bombard AT, Powers JF, Carter S et al. Procedure‐related fetal losses stay in the neonatal intensive care unit, surfactant use,
in transplacental versus nontransplacental genetic amniocentesis. Am mechanical ventilation, insertion of umbilical arterial
J Obstet Gynecol 1995;172:868.
34 Andreasen E, Kristoffersen K. Incidence of spontaneous abortion after
catheters, use of ionotropes for circulatory support, pat-
amniocentesis: influence of placental localization and past obstetric ent ductus arteriosus, bronchopulmonary dysplasia and
and gynecologic history. Am J Perinatol 1989;6:268. positive microbiology culture results [1].
160 Section 2  Skin Disorders of the Neonate and Young Infant

Damage produced in ensuring adequate


respiratory function
The skin of the preterm infant is not as well keratinized as
that of the term infant or adult, and is thus very sensitive
to damage by pressure or friction. Unsightly excoriation
may be produced around the nose or mouth by contact
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

with endotracheal tubes or facemasks. Although injury of


this sort is not life‐threatening, it may induce considera-
ble parental anxiety. Many infants on neonatal intensive
care units are managed using nasal continuous positive
airway pressure (CPAP), which is delivered via nares‐
occluding prongs or via masks. This mode of respiratory
support has been associated with the development of
columella nasi necrosis [2]. Inappropriately fitted CPAP
devices can cause mild to severe nasal septum damage Fig. 12.7  Gangrene of the toes in a preterm baby who had an umbilical
[3]. Prolonged use of nasal CPAP in preterm babies can catheter.
cause nasal vestibular stenosis, which may require surgi-
cal correction in later life [4].
Newborns who develop a pneumothorax may require
the immediate insertion of a percutaneous drain. Drains
inserted in the second or third intercostal space in the mid-
clavicular line are more effective than those sited in the
anterior axillary line. This is because the tip of the drain is
more likely to be in the anterior third of the hemithorax
(the preferred position) if inserted through the anterior
chest wall [5]. Permanent breast tissue damage is the most
serious complication of anterior chest drains. In one study,
four (three girls) out of nine children in whom chest drains
were inserted in the neonatal period suffered cosmetically
important scars on the anterior chest wall. In the other five
children, mild scars over the anterior chest wall were
related to the site of insertion of the chest drain [6–8]. In
none of these reported patients was there any damage to
the nipple or areola. It is preferable to use the lateral
approach initially, especially in girls, and leave the ante-
rior approach for use if pneumothorax drainage is unsatis-
factory. Just as important as the site of insertion of the
chest drain is the method used to close the wound. Purse‐
string sutures inevitably pucker the skin and result in an
ugly scar that grows with the infant. Another method is to (a)
cover the wound with an occlusive dressing but, because
the wound edges are not apposed, healing is by granula-
tion; thus, the scar is still likely to be marked. The best way
of closing the wound is by apposing the wound edges
using simple suturing or thin strips of adhesive [6].

Conditions related to arterial


catheterization
Catheterization of the umbilical vessels is commonly per-
formed in the sick neonate because of the ready accessibility
of relatively large vessels. Umbilical arterial catheterization
may lead to thrombosis in the abdominal aorta or iliac arter-
ies. Major aortic thrombosis is infrequent but may lead to
serious skin complications such as gangrene of the limbs
(Fig.  12.7). Gluteal skin necrosis has been described as a
complication of umbilical arterial catheterization [9], but
very similar lesions may be produced by pooling of alcohol‐ (b)
containing cleaning solutions used to prepare the skin prior Fig. 12.8  (a) Skin necrosis in a preterm baby caused by alcoholic
to the insertion of the umbilical line (Fig. 12.8). disinfectant solutions. (b) Later appearance of the lesion.
Chapter 12  Iatrogenic Disorders of the Newborn 161

Peripheral arterial catheterization is a widely used the early neonatal period, indicating that their skin is
technique for blood sampling and blood pressure moni- defective as a barrier. Both drug absorption and water
toring in neonatal intensive care units. The brachial artery loss in preterm infants fall steadily and by about 2 weeks
is an end artery to the lower arm, and thus damage or of age the skin of the most immature infants functions like
thrombosis of that artery caused by cannulation may lead that of mature infants. The varying barrier properties can
to ischaemia or gangrene of the lower part of the limb. be explained by the poor development of the stratum cor-

SECTION 2: SKIN DISORDERS


Radial artery sampling or cannulation may be accompa-

OF NEONATES AND INFANTS


neum in the more premature infants at birth, and its rapid
nied by gangrene of the hand but more commonly the maturation after birth [27,28].
ischaemia caused is reversible after removal of the line.
Catheterization of the posterior tibial artery may lead to
the minor complication of cyanosis of the toes, which Extravasation injuries
responds to removal of the catheter [10]. Irreversible Total parenteral nutrition (TPN) consists of solutions con-
ischaemia of the foot following posterior tibial arterial taining a mixture of amino acids, sugar, electrolytes, min-
catheterization leading to gangrene and subsequent erals and trace elements. Intralipid emulsion, a mixture of
amputation has been described [11]. fats and fatty acids suitable for intravenous administra-
tion, can be added to TPN solutions. TPN is used in neo-
nates in whom feeding via the enteral route is impossible,
Complications related to drugs inadequate or hazardous and in whom this state is likely
and nutrients to be prolonged. The preferred route of administration of
Exposure to environmental toxins is also a concern in the TPN is via a central venous catheter placed in the supe-
infant. All infants, regardless of their gestational age at rior vena cava or inferior vena cava outside the cardiac
birth, are uniquely vulnerable to the toxic effects of chem- silhouette. Incorrect placement of the central line may
ical exposure because of the immaturity of their organs lead to serious thrombosis in the large veins of either
and a relatively high surface area to bodyweight ratio. upper or lower limbs. This would in turn lead to swelling
The vulnerability of neonates to cutaneous poisonings is of the affected limb. TPN may also be given via a periph-
thought to be related to their immature hepatic, renal and eral venous catheter. This route may also be used to
central nervous systems and high body surface to volume administer other infusions such as dopamine, which is
ratio, in addition to their accelerated growth and meta- used to improve blood pressure and renal perfusion,
bolic rates [12]. Percutaneous absorption of topical agents, sodium bicarbonate to correct metabolic acidosis or a dex-
while especially hazardous to the preterm baby, remains a trose solution of over 10% concentration to maintain ade-
danger throughout infancy. Conversely, the relative per- quate blood sugar levels. However, these fluids are very
meability of the skin of premature babies makes it a irritant and frequently thrombose the vessels.
potential route for drug delivery [13]. If extravasation occurs, permanent scarring may result.
Methaemoglobinaemia due to aniline dye absorption has Extravasation of lipid‐containing TPN solutions through
been well documented [14]. Percutaneous absorption of neo- a peripheral cannula will initially appear as a white, well‐
mycin has been associated with deafness [15]. Skin sloughing demarcated area over the affected site (Figs  12.9–12.11).
and blister formation with lethal blood ethanol levels have
been reported from the topical application of isopropyl alco-
hol in infants of less than 27 weeks’ gestation [16–19].
Hexachlorophene and iodine absorbed through the
skin are detectable in infant blood [20,21]. These com-
pounds need to be used with great caution in the new-
born because systemic absorption causes toxic side‐effects
[22,23]. Systemic absorption of iodine compounds may
produce metabolic acidosis, hypernatraemia and impair-
ment of renal function. Routine povidone–iodine applica-
tion in the umbilical cord has been reported to interfere
with neonatal screening for hypothyroidism [24]. The use
of iodine alcoholic solutions is contraindicated in very‐
low‐birthweight infants. Systemic absorption of 3% hexa-
chlorophene has been associated with spongiform
changes in the brains of newborn infants [25,26]. The use
of 3% hexachlorophene in the newborn should be avoided.
Ster‐Zac powder, used as an antiseptic applied to the
umbilical cord, contains only 0.33% hexachlorophene.
In general, infants born at 37 weeks’ gestation or later
show little or no drug absorption and have low skin water
losses, indicating that their skin is an effective barrier. By
contrast, infants born at 32 weeks’ gestation or before
show marked drug absorption and high water losses in Fig. 12.9  The acute phase of an extravasation injury.
162 Section 2  Skin Disorders of the Neonate and Young Infant

Area of
chemical burn
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

Punctures
surrounding Verres
wound site needle
Fig. 12.12  Saline flush‐out technique of chemical burns. Source: Adapted
from Rutter 1987 [13].

plastic surgery. Extravasation from intravenous cannulas


inserted in the scalp may cause alopecia [6]. Extravasation
Fig. 12.10  Later phase of an extravasation injury.
of calcium‐containing TPN solution may lead to subcuta-
neous calcification. Spinal cord compression and necrosis
following TPN extravasation has been reported [29].

Management
Although every effort should be made to detect extrava-
sation injuries early by using clear adhesives to hold
peripheral cannulas in situ and thereby keeping the site of
potential injury in full view, inevitably some injuries may
not become apparent until a late stage. There is a continu-
ing debate about whether treatment of such injuries pro-
duces any long‐term functional or cosmetic benefits. A
final conclusion is difficult without a large controlled trial
to compare the results of treated and untreated groups.
A technique to deal with such injuries has been
described [30] and can be performed by neonatal unit
staff after appropriate training. Under strict aseptic condi-
tions, the discoloured area and surrounding skin are
cleaned and infiltrated with 1% lidocaine. Hyaluronidase,
500–1000  U, is injected into the subcutaneous tissue
beneath the damaged skin. Four small punctures are
made in the tissue plane with a scalpel blade around the
area to be treated. The central blunt cannula of a Verres
needle is inserted subcutaneously through one of the
Fig. 12.11  The granulation stage of an extravasation injury. puncture sites. (This is a needle used most commonly for
insufflation of air at gynaecological laparoscopy; Downes
Surgical Ltd, Sheffield; Rocket London Ltd, Watford.)
This will then gradually develop into a black area of Using a syringe attached to a three‐way tap, saline is
chemical burn. The immediate danger of extravasation injected; this should flow out freely from the other three
injuries is extension of the burn to involve the full thick- incisions (Fig.  12.12). The process is repeated, injecting
ness of the skin and the formation of an abscess, which through each incision and using in total up to 500 mL of
may lead to septicaemia. saline. If during the procedure the limb becomes oedema-
Scars caused by extravasation injuries are typically tous, excess fluid can be removed by gentle massage
larger than needle marks and are most commonly found towards the incisions. The damaged area is then dressed
over the hands and wrists. Extravasation injuries are with Jelonet or a similar dressing and kept covered for
more likely to occur when intravenous cannulas are 24–48 hours. The above technique appeared to be effec-
inserted in the lower limbs, particularly above the ankle. tive in preventing long‐term tissue damage if performed
The majority of extravasation injuries leave minor scars, early (within 24 hours) in a series of 96 patients. There
but some may result in permanent disfiguring scars were no complications reported from the technique in the
and fixed contractures of the limbs which may require study [31,32].
Chapter 12  Iatrogenic Disorders of the Newborn 163

Problems related to intensive care


monitoring
Previously, immature infants survived the first few days
of life only to succumb to overwhelming sepsis and/or
life‐threatening fluid and electrolyte imbalances second-
ary to multiple areas of skin breakdown. Improved sur-

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
vival rates among smaller and more immature babies and
improving technology mean that more monitoring equip-
ment has to be anchored onto the infant’s fragile skin.
Skin surface probes are widely used to monitor heart rate,
respiratory rate, temperature and blood gases in sick pre-
term infants. Such monitors, although not strictly inva-
sive, need to be secured to the skin with adhesive tape, Fig. 12.13  Scar that resulted from the use of adhesive taping for a
nasogastric tube in the neonatal period.
which strips off the superficial layer of the stratum cor-
neum when it is removed.
Although this is of little consequence in a term infant, it To prevent damage caused by adhesives, the necessity of
is of tremendous importance in the very preterm infant every piece of tape or adhesive should be evaluated care-
below 30 weeks’ gestation, whose stratum corneum is fully, especially if applied directly to the infant’s skin. Skin
thin and poorly developed. Transepidermal water loss protection may be achieved by using a pectin‐based skin
increases, absorption of topically applied chemicals barrier such as the type used in ostomy care. This is applied
occurs more readily and the stripped area may provide a directly to the infant’s skin on all areas where tape will be
site for the entry of microorganisms [33]. The finding of used. For example, in neonatal units, where endotracheal
atrophic macules, possibly secondary to the use of elec- tubes are taped to the skin, protective barriers can be used
trodes or other adhesives in extremely preterm infants, underneath the tape. This will allow the endotracheal tube
has been termed ‘anetoderma of infancy’ [34]. to be changed as often as necessary without removing the
barrier, thereby maintaining skin integrity. Other uses of
skin barriers are underneath the tape used to secure an
Transcutaneous monitoring umbilical catheter, chest drains, nasogastric tubes or a tem-
Transcutaneous electrodes can be used for continuous perature probe from a radiant heater. Pectin‐based skin bar-
monitoring of the partial pressure of oxygen (PO2) and riers can also be used directly over exfoliated skin [35,36].
the partial pressure of carbon dioxide (PCO2). These tech- Another method of preventing skin damage caused by
niques are an invaluable adjunct to intermittent arterial adhesives is the use of karaya gum electrodes, which have
sampling during the acute course of respiratory disease. been demonstrated to be gentle to sensitive skin and very
The monitoring technique employs an electrode, gener- easy to remove or replace, and effectively monitor the cardi-
ally heated to 42–44 °C, which is attached to the skin and orespiratory status. These electrodes should be removed
measures the oxygen and carbon dioxide tensions in the and rotated at least once a day to assess the skin underneath
gas that has diffused from the arterialized capillary bed to and to prevent any damage from the silver chloride gel [37].
the skin surface and through a gas‐permeable membrane. Gentle and careful removal of adhesives is mandatory.
The skin in contact with the heated electrode suffers the Alcohol and/or adhesive removers should be avoided for
changes of an early burn, the surrounding skin is dam- routine use because of their potential for irritation and
aged by stripping when the adhesive ring is removed absorption. A gauze soaked in warm water is effective in
and, as the electrode site is usually changed every 4 hours, gently soaking off adhesives. Alcohol or adhesive remov-
a large area of skin is subject to trauma. In one follow‐up ers should be reserved for emergency situations or when
study, children were found to have lesions on the anterior the adhesives have softened and melted on to the skin. If
abdomen and chest consisting of normally textured skin used, they must be washed off with water [9].
with altered pigmentation caused by transcutaneous
monitors. Most of these lesions were not present at the Transillumination burns
time of discharge from the neonatal unit [30]. Transillumination has been used in neonatology in help-
One method that was described to reduce skin damage ing to identify vascular access and also in the diagnosis of
from transcutaneous oxygen electrodes is the use of plas- pneumothoraces. Blisters secondary to thermal burns
tic spray‐on dressings such as Op‐Site [33]. have been attributed to faulty devices [38,39].

Adhesive tape damage Needle marks


Skin damage may be caused by removal of adhesive tape In a follow‐up study of toddlers who had received neonatal
(Fig. 12.13). This may lead to a highly characteristic lesion intensive care, needle marks consisted of white pinhead‐
that resembles a healed skin graft donor site. Skin slough- sized lesions which individually were barely perceptible
ing, blister formation and haemorrhagic skin necrosis but, where grouped together, gave a speckled appearance.
with lethal blood ethanol levels have been reported after They are principally caused by the insertion of intravenous
attempts to remove adhesive tape using isopropyl alcohol cannulas and less often by venepuncture, arterial cannulas,
in infants of 27 weeks’ gestation or less [17]. arterial puncture and lumbar puncture needles [6].
164 Section 2  Skin Disorders of the Neonate and Young Infant

Heelprick marks References


1 Csoma ZR, Meszes A, Abraham R et al. Iatrogenic skin disorders
Heelprick marks may give the heel a faintly granular and related factors in newborn infants. Paediatr Dermatol 2016;
appearance that most commonly develops months after 33:543–8.
discharge from the neonatal intensive care unit. Some 2 Robertson NJ, McCarthy LS, Hamilton PA et  al. Nasal deformities
infants may develop small nodular lesions that intermit- resulting from flow driver continuous positive airway pressure. Arch
Dis Child Fetal Neonatal Ed 1996; 75:F209–12.
tently discharge a white material. These are thought to be 3 McCoskey L. Nursing care guidelines for prevention of nasal break-
SECTION 2: SKIN DISORDERS
OF NEONATES AND INFANTS

implantation dermoid cysts and they rarely interfere with down in neonates receiving nasal CPAP. Adv Neonatal Care 2008;
function, usually disappearing by the age of 2.5 years. 8:116–24.
4 Smith LP, Roy S. Treatment strategy for iatrogenic nasal vestibular
Rarely, infants may have pronounced dimpling of their stenosis in young children. Int J Pediatr Otorhinolaryngol 2006;
heels [36]. 70:1369–73.
5 Allen RW, Lung AL, Lester PD. Effectiveness of chest tube evacuation
Complications of phototherapy of pneumothorax in neonates. J Pediatr 1981;99:629–34.
6 Cartlidge PHT, Fox PE, Rutter N. The scars of newborn intensive
Phototherapy is one of the treatment modalities com- care. Early Hum Dev 1990;21:1–10.
monly used in neonatal jaundice. The principle involved 7 Fox PE, Rutter N. The childhood scars of newborn intensive care.
is transcutaneous photoisomerization of unconjugated Early Hum Dev 1998;51:171–7.
8 Rainer C, Gardetto A, Frühwirth M et al. Breast deformity in adoles-
bilirubin to less toxic polar isomer. A macular erythema- cence as a result of pneumothorax drainage during neonatal intensive
tous rash has been described as a result of phototherapy; care. Pediatrics 2003;111:80–6.
this is self‐limiting. Darkening of the treated skin in black 9 Mann NP. Gluteal skin necrosis after umbilical artery catheterization.
and minority ethnic babies due to increased melanogen- Arch Dis Child 1980;55:815–17.
10 Sphar RC, Macdonald HM, Holzman IR. Catheterization of the poste-
esis has been described. Ultraviolet burns and phototoxic rior tibial artery in the neonate. Am J Dis Child 1979;133:945–6.
reaction to ingested drugs, especially furosemide, have 11 Abrahamson EL, Scott RC, Jurges E et  al. Catheterization of
been reported. posterior tibial artery leading to limb amputation. Acta Paediatr
1992;82(6–7):618–19.
Bronze baby syndrome is a rare complication of photo-
12 West D, Worobec S, Solomon L. Pharmacology: toxicology of infant
therapy. Babies with hepatic disease are predisposed to skin. J Invest Dermatol 1981;76:147–50.
develop this condition as they accumulate porphyrin, 13 Rutter N. Percutaneous drug absorption in the newborn: hazards
copper and photoisomers of bilirubin. Photodestruction and uses. Clin Perinatol 1987;14:911–30.
14 Howarth BE. Epidemic of methaemoglobinaemia in newborn infants.
of these complexes results in dark grey‐brown pigmenta- Lancet 1951;i:934.
tion of the skin, serum and urine [40]. 15 Morrall P, Hey E, Mackee IW et al. Deafness in a preterm baby associ-
ated with topical antibiotic spray containing neomycin. Lancet
1985;i:1167.
Surgical scars 16 Happin V, Rutter N. Percutaneous alcohol absorption and skin necro-
Surgical procedures such as laparotomies and insertion of sis in a preterm infant. Arch Dis Child 1982;57:477–9.
Broviac catheters may leave horrific scars (Fig.  12.14). 17 Schick J, Milstein J. Burn hazard of isopropyl alcohol in the neonate.
Alopecia may occur at the site of surgical incisions. Pediatrics 1981;68:587–8.
18 Al‐Jawad S. Percutaneous alcohol absorption and skin necrosis in a
preterm infant. Arch Dis Child 1983;58:395–6.
Vaccinations 19 Reynolds PR, Banerjee S, Meek JH. Alcohol burns in extremely low
The bacille Calmette–Guérin (BCG) vaccine is currently birthweight infants: still occurring. Arch Dis Child Fetal Neonatal
Ed 2005;90:10.
offered to all newborn babies in many hospitals across the
20 Kopelman A. Cutaneous absorption of hexachlorophene in lower
UK, Ireland and many other countries. As in older chil- birth weight infants. J Pediatr 1973;83:972–5.
dren, an incorrect administration technique may lead to 21 Pyati S, Ramamurthy R, Krauss M et al. Absorption of iodine in the
devastating consequences, including BCGosis and disfig- neonate following topical use of iodine. J Pediatr 1977;91:825–8.
22 Goutières F, Aicardi J. Accidental percutaneous hexachlorophane
uring scars. intoxication in children. BMJ 1977;2:663–5.
23 Ansotegui AJ, Knörr JI, Vege MJ et  al. Iodine overload in newborn
infants caused by the use of PVP‐iodine for perineal preparation of
the mother in vaginal delivery. An Esp Pediatr 1989;30:23–6.
24 Lin CP, Chen W, Wu KW. Povidone‐iodine in umbilical cord care
interferes with neonatal screening for hypothyroidism. Eur J Pediatr
1994;153:756–8.
25 Powell H, Swarner O, Gluck L et al. Hexachlorophene myelinopathy
in preterm infants. J Pediatr 1973;82:976–81.
26 Shuman RM, Leech RW, Alvord EC. Neurotoxicity of hexachloro-
phene in the human. I. A clinicopathologic study of 248 children.
Pediatrics 1974;54:689–95.
27 Haprin VA, Rutter N. Barrier properties of the newborn infant’s skin.
J Pediatr 1983;102:419–25.
28 Chiou YB, Blume‐Peytavi U. Stratum corneum maturation. A review
of neonatal skin function. Skin Pharmacol Physiol 2004;17:57–66.
29 Knobel RB, Meetze W, Cummings J. Case report: total parenteral
nutrition extravasation associated with spinal cord compression and
necrosis. J Perinatol 2001;21:68–71.
30 Davies J, Gault D, Buchdahl R. Preventing the scars of neonatal
intensive care. Arch Dis Child 1994;70:50–1.
31 Gault D. Extravasation injuries. Br J Plastic Surg 1993;46:91–6.
32 Wilkins CE, Emmerson AJ. Extravasation injuries on regional
Fig. 12.14  Surgical scars following an operation for necrotizing enterocol-
­neonatal units. Arch Dis Child Fetal Neonatal Ed 2004;89:F274–5.
itis in the neonatal period.
Chapter 12  Iatrogenic Disorders of the Newborn 165

33 Evans NJ, Rutter N. Reduction of skin damage from transcutaneous 37 Malloy MB, Lambert GH. Neonatal skin care. Compr Ther 1993;
oxygen electrodes using a spray on dressing. Arch Dis Child 1986;61:881–4. 19:286–90.
34 Prizant TL, Lucky AW, Frieden IJ et al. Spontaneous atrophic patches 38 Perman, Marissa J, Kauls, Lynda S. Transilluminator burns in the
in extremely premature infants. Anetoderma of prematurity. Arch neonatal intensive care unit: a mimicker of more serious disease.
Dermatol 1996;12:671–4. Pediatr Dermatol 2007;24:168–71.
35 Lund C, Kuller J, Tobin C et al. Evaluation of a pectin‐based barrier 39 Sajben P, Gibbs NF, Friedlander SF. Transillumination blisters in a
undertape to protect neonatal skin. J Obstet Gynecol Neonatal Nurs neonate. J Am Acad Dermatol 1999;41:264–5.
1986;15:39–44. 40 Siegfried EC, Stone MS, Madison KC. Ultraviolet light burn: a cutane-

SECTION 2: SKIN DISORDERS


OF NEONATES AND INFANTS
36 Dollison EJ, Beckstrand J. Adhesive tape vs pectin‐based barrier use ous complication of visible light phototherapy of neonatal jaundice.
in preterm infants. Neonatal Netw 1995;14:35–9. Pediatr Dermatol 1992;9:278–80.
 167

C HA PTER   13

Epidemiology of Atopic Dermatitis


Carsten Flohr1, Jonathan I. Silverberg2, Joy Wan3 & Sinéad M. Langan4
1
 Unit for Population‐Based Dermatology Research, St John’s Institute of Dermatology, Guy’s & St Thomas’ NHS Foundation Trust and King’s College, London, UK
2
 Northwestern University Feinberg School of Medicine, Chicago, IL, USA
3
 Department of Biostatistics and Epidemiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
4
 Faculty of Epidemiology and Population Health, London, School of Hygiene and Tropical Medicine, London, UK

Why study the epidemiology of atopic Descriptive epidemiology of atopic Prevention of atopic dermatitis, 182

ATOPIC DERMATITIS
dermatitis?, 167 dermatitis, 171 Conclusion, 183
Defining atopic dermatitis for Specific risk factors, 175

SECTION 3:
epidemiology research, 168

Abstract implications for research findings. We discuss what we currently know


about AD epidemiology, focusing on prevalence, incidence and natu­
ral history. In the final sections of the chapter, we assess the ­evidence
Atopic dermatitis (AD) is a critical disease to study due to its high
for risk factors for AD and what we currently know about preventing
prevalence and morbidity. In this chapter, we review AD definitions
AD. Prevention of disease is the ultimate aim for epidemiology and is
in epidemiology studies, including the challenges of identifying the
a noble ambition for AD, given the rising global prevalence.
disease and the heterogeneity in currently used definitions and their

Key points • Mixed data surround the role of breastfeeding, specialized


formulas and solid foods in the predisposition to AD.
• The ‘hygiene hypothesis’ suggests that AD may be due to lower
Descriptive epidemiology of atopic dermatitis (AD)
exposure to certain viral and bacterial pathogens, but there is no
• Reported prevalence rates of AD vary greatly; surveys suggest current evidence to suggest that exposure to a specific infection
that 10–30% of children in developed countries have AD with reduces the risk of AD.
plateauing prevalence rates in some countries. Lower, but rising, • Irritant exposure can exacerbate AD and increase sensitization,
rates are reported in developing countries. particularly in the context of an impaired skin barrier.
• AD starts in early life in most cases but may begin later in childhood • The strength of association between allergic sensitization and
or adulthood. AD varies widely across studies, and the aetiological role of
• AD is a costly disease that leads to impairments in quality of life. sensitization in AD remains uncertain.
• More recent studies suggest that many patients have persistent • Evidence suggests that specific atmospheric pollutants such as
or relapsed AD in adulthood. Severe childhood disease, early tobacco smoke may be associated with AD.
onset disease and concomitant or family history of atopy are
associated with worsened prognosis. Prevention of AD
• Environmental factors associated with urbanization and socio­
• Allergen avoidance may not prevent AD; emerging evidence suggests
economic advantage are thought to contribute to the aetiology of AD.
that early allergenic food exposure may reduce food allergy.
• Prenatal probiotic exposure appears to be associated with a
Specific risk factors
reduction in AD, although further research is required addressing
• Genetic factors that predispose to an impaired skin barrier and timing and formulations.
immune dysregulation are associated with AD. • Limited evidence supports the use of prebiotics, synbiotics
• Environmental influences operating in utero, including maternal and omega‐3 long‐chain polyunsaturated fatty acids although
stress, antibiotic use and allergen exposure, have been proposed prebiotics may be helpful in formula‐fed babies.
to increase the risk of AD. • Emerging evidence suggests that early‐life emollient use may be
• Alterations in gut and skin microflora have been observed in beneficial in the primary prevention of AD.
children with AD; additional studies are needed before a causal • The use of water softeners was tested in a UK‐wide trial in
relationship can be established. children, but showed no benefit over usual care.

Why study the epidemiology of atopic geographical residence. Although this kind of information
dermatitis? may be important in terms of quantifying the burden of
Epidemiology, if mentioned at all in dermatological texts, disease and suggesting hypotheses about disease
is usually confined to brief descriptions of how the causation, it is of little practical use to those who suffer
frequency of skin diseases varies with age, sex and area of from skin diseases such as atopic dermatitis (AD). Modern

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
168 Section 3  Atopic Dermatitis and Related Disorders

clinical epidemiology goes much further than simply typically flexural, but possibly also limited to extensor
describing the burden of disease. It includes the study of or head and neck areas, or generalized. AD lesions can
causes or risk factors of AD, as well as the evaluation of be challenging to characterize owing to variable mor-
interventions to prevent and treat the disease [1]. Previous phology, including acute oozing and vesicles, subacute
studies relied more on questionnaires and physical exami- scaling, chronic lichenification and prurigo nodules,
nation, whereas more recent work often uses genetic and ill‐demarcated versus more psoriasiform lesions and
immunological tools to better understand the interplay variable intensity. AD is particularly difficult to define in
between genetic and environmental factors. adults because many have a nonflexural distribution of
Interest in the epidemiology of AD has risen in recent lesions, with more involvement of the head/neck and
years because the prevalence of AD is on the increase [2,3], hands, and disease onset in adolescence and adulthood
and because AD can be a very miserable and costly long‐ in a substantial proportion [2,3]. The time course of AD
term condition that may be difficult to treat. Furthermore, is also highly variable, with intermittent or chronic per-
because AD tends to be the first allergic disease to develop sistent disease, seasonal variation, flares secondary to
ATOPIC DERMATITIS

in early life, an increasingly large number of birth cohort emotional factors and episodic exposures. Unfortunately,
studies have investigated AD alongside food allergies, there are no widely accepted biomarkers or objective
SECTION 3:

hay fever and asthma in order to understand the relation- diagnostic tests for AD.
ship between these diseases. This chapter provides an Reliance on flexural dermatitis alone as a defining char-
overview of the basic epidemiology of AD, highlighting acteristic of AD is a reasonable approach to defining AD.
some of the important developments and gaps in current However, this approach would miss a large subset of AD
knowledge. patients with nonflexural disease. The Hanifin and Rajka
criteria are the gold‐standard diagnostic criteria for AD
References [4] and are commonly used in clinical trials, but they are
1 Williams HW. Epidemiology of skin disease. In: Griffiths C, Barker J, too cumbersome for large‐scale population studies and
Bleiker T et al. (eds) Rook’s Textbook of Dermatology, 9th edn. Oxford: were found to have low specificity [5,6]. There are a num-
Wiley‐Blackwell, 2016: vol.1, section 5.1.
2 Williams H, Stewart A, von Mutius E et al. Is eczema really on the
ber of other diagnostic criteria used to define AD interna-
increase worldwide? J Allergy Clin Immunol 2008;121:947–54. tionally (e.g. Japanese Dermatological Association,
3 Williams HC. Is the prevalence of atopic dermatitis increasing? Clin Korean Dermatological Association, American Academy
Exp Dermatol 1992;17:385–91. of Dermatology, etc.). However, these definitions of AD
are not well validated for use in epidemiological research.
Defining atopic dermatitis In 1990, a UK working party (UKWP) of 16 doctors
for epidemiology research selected a minimum list of six reliable diagnostic criteria
for diagnosing AD as a modification to the Hanifin and
• AD is difficult to define for epidemiological study Rajka criteria (shown in Box 13.1) [7]. Visible flexural der-
because of heterogeneity of nomenclature and clinical matitis is required to be ascertained by a trained observer,
manifestations. and a training protocol and quality control test have been
• Survey‐based approaches are relatively low cost, feasible developed for this sign [8] (Fig. 13.1). Nineteen validation
and scalable for population‐based studies. studies of the UKWP diagnostic criteria in both hospital
• There is a lack of consensus as to the best way to define and community populations in a number of different geo-
AD in population surveys. graphical settings, including Romania [9] and China [10],
• Self‐ or caregiver‐reported or healthcare‐diagnosed have found a sensitivity ranging between 10% and 100%
AD, the International Study of Allergies and Asthma and a specificity between 89% and 99% [11].
in Children (ISAAC) criteria and other approaches
may be sufficiently valid definitions of AD for epide-
miological study.
• Physician assessment is often costly, labour‐intensive, Box 13.1  The UK diagnostic criteria for atopic dermatitis for use
not feasible in large‐scale population‐based studies, in epidemiological studies
and might overlook disease activity between visits.
• There are no ‘objective’ tests or biomarkers that are Must have:
sufficiently valid for the epidemiological study of AD. • An itchy skin condition (or parental report of scratching or
AD is difficult to define for epidemiological study rubbing) in the last 12 months
because of the considerable heterogeneity of nomencla- Plus three or more of the following:
ture and clinical manifestations. AD is also referred to as • History of involvement of the skin creases (fronts of elbows,
eczema, atopic eczema, childhood eczema, etc. The term behind knees, fronts of ankles, around neck or around eyes)
eczema has several different uses, including as the most • Personal history of asthma or hay fever (or history of atopic disease
in first‐degree relative if child aged under 4 years)
commonly used lay synonym for AD, as a descriptive
• A history of a generally dry skin in the last year
morphological and/or histological term encompassing
• Onset under the age of 2 years (not used if child aged under 4 years)
multiple aetiologies, and as a diagnostic term for AD [1].
• Visible flexural dermatitis (this includes dermatitis affecting cheeks
The lack of standardized nomenclature for AD interna-
or forehead and outer aspects of limbs in children under 4 years)
tionally makes it difficult to develop consistent and valid
questionnaires for epidemiology research. Moreover, Source: Williams et al. 1994 [7].
there is a variable distribution of AD with lesions being
Chapter 13  Epidemiology of Atopic Dermatitis 169

Several approaches have been used to define AD for sometimes incorporated. In clinical epidemiology
epidemiology research, depending on the study design ­studies, physical examination and clinical criteria are
and objectives. In population‐based studies, questionnaires more commonly used. The various definitions used
are most commonly used, with physical examination to  define AD in different studies make it difficult to

Observer’s protocol for recording the sign of


“visible flexural dermatitis”

A. Subjects aged 4 years and over


Your task is to record as consistently as possible the presence/absence

ATOPIC DERMATITIS
of one physical sign - ‘visible flexural dermatitis’.
To decide whether this sign is present or not, there are two components to consider:

SECTION 3:
Step 1 What dermatitis looks like
Definition of dermatitis: Poorly demarcated erythema (redness) with surface change
‘Surface change’ can mean fine scaling, vesicles, oozing, crusting or lichenification.
Here are some photographs to help you:

1. This is dermatitis. Note it is red, has an indistinct margin 4. This is lichenification in a white skin. Lichenification means
and there is surface change (in this case fine scaling). a thickening of the skin in response to scratching. The skin
markings are exaggerated and the skin feels thickened.

2. This is dermatitis showing another type of surface change, 5. This is lichenification in a black skin. Note the exaggerated
in this case oozing (clear fluid leaking from the skin) and skin creases and post-inflammatory pigmentation.
crusting (scabs).

6. This is also lichenification in a black skin. In this case,


3. These are vesicles (tiny clear “water” blisters). the thickening is comprised of smaller flat topped bumps
corresponding to the hair follicles – so called “follicular
lichenification”.

Fig. 13.1  The photographic protocol to determine the physical sign of visible flexural dermatitis. Source: Adapted from Williams et al. 1995 [8]. See also
https://www.nottingham.ac.uk/~mzzfaq/dermatology/eczema.
170 Section 3  Atopic Dermatitis and Related Disorders

compare prevalence estimates and disease associa- diagnostic tests for AD, the best way to define AD for
tions worldwide. epidemiological research remains via clinical diagnostic
The advantages of questionnaires include relatively criteria (e.g. UKWP) or analogous questionnaires, or
low cost to implement, good feasibility and scalability questionnaires assessing for physician‐diagnosed AD or
for population‐based studies. However, there is a lack of eczema. It is imperative that AD be defined using validated
consensus as to the best way to define AD in population assessments that are repeatable, easy to use, acceptable to
surveys. Some studies rely on a single question based on the population under study and correspond well to our
a self‐ or caregiver‐report of ‘eczema’ (the validity of clinical concept of AD [5].
which is usually unknown). Some rely on a history of
healthcare‐diagnosed AD or eczema in the past year. This
approach was previously validated in a dermatology References
1 Kantor R, Thyssen JP, Paller AS, Silverberg JI. Atopic dermatitis,
practice setting and found to have good validity [12]. atopic eczema, or eczema? A systematic review, meta‐analysis, and
However, such questions may underestimate the preva- recommendation for uniform use of ’atopic dermatitis’. Allergy
ATOPIC DERMATITIS

lence of AD because they require access to and utilization 2016;71:1480–5.


2 Katsarou A, Armenaka M. Atopic dermatitis in older patients:
of healthcare, which varies widely between countries
SECTION 3:

particular points. J Eur Acad Dermatol Venereol 2011;25:12–18.


and regions. The ISAAC and National Health and 3 Hanifin JM, Reed ML; Eczema Prevalence and Impact Working
Nutrition Examination Survey in the USA used a com- Group. A population‐based survey of eczema prevalence in the
posite definition of AD based on three questions about United States. Dermatitis 2007;18:82–91.
4 Hanifin J, Rajka G. Diagnostic features of atopic dermatitis. Acta
the presence of an itchy rash for at least 6 months, the Derm Venereol (Stockh) 1980;92(suppl):44–7.
presence of such a rash in the past 12 months, and flex- 5 Williams HC. What is atopic dermatitis and how should it be
ural distribution. These are analogous to having three defined in epidemiological studies? In: Williams HC (ed.) Atopic
Dermatitis: The Epidemiology, Causes and Prevention of Atopic
major Hanifin and Rajka or UKWP criteria for AD. This
Eczema. Cambridge: Cambridge University Press, 2000:3–24.
approach has been previously validated in children from 6 Schultz‐Larsen F. The epidemiology of atopic dermatitis. In: Burr ML
multiple countries and found to have high specificity (ed.) Epidemiology of Clinical Allergy. Basel: Karger, 1993:9–28.
and variable sensitivity [13–17]. However, it has some 7 Williams HC, Burney PG, Pembroke AC, Hay RJ. The U.K. Working
Party’s Diagnostic Criteria for Atopic Dermatitis. III. Independent
limitations as sensitivity has not been adequately tested hospital validation. Br J Dermatol 1994;131:406–16.
in adults [18,19]. 8 Williams HC, Forsdyke H, Boodoo G et al. A protocol for recording
The advantages of physician’s examination include the sign of flexural dermatitis in children. Br J Dermatol 1995;133:
pseudo‐objective assessment and the potential to distin- 941–9.
9 Popescu CM, Popescu R, Williams H, Forsea D. Community valida-
guish between AD and similar‐appearing dermatoses. tion of the United Kingdom diagnostic criteria for atopic dermatitis in
However, there are several limitations of using physician Romanian schoolchildren. Br J Dermatol 1998;138:436–42.
examination in epidemiology studies of AD. Physician 10 Gu H, Chen XS, Chen K et  al. Evaluation of diagnostic criteria for
atopic dermatitis: validity of the criteria of Williams et al. in a hospital‐
assessment is often costly and labour‐intensive and not based setting. Br J Dermatol 2001;145:428–33.
feasible in a large‐scale population‐based study. Moreover, 11 Brenninkmeijer EE, Schram ME, Leeflang MM et al. Diagnostic
AD often has a waxing and waning course and active criteria for atopic dermatitis: a systematic review. Br J Dermatol
disease might not be present at a study visit despite mani- 2008;158:754–65.
12 Silverberg JI, Patel N, Immaneni S et  al. Assessment of atopic
festing as classic AD at other times. Many of the different dermatitis using self‐report and caregiver report: a multicentre vali-
diagnostic criteria may not be suitable for population‐ dation study. Br J Dermatol 2015;173:1400–4.
based studies because they may not be accurately reflected 13 Strina A, Barreto ML, Cunha S et  al. Validation of epidemiological
tools for eczema diagnosis in Brazilian children: the ISAAC’s and UK
in a patient‐ or caregiver‐reported survey, low disease
Working Party’s criteria. BMC Dermatol 2010;10:11.
prevalence may markedly diminish the predictive value 14 Chalmers DA, Todd G, Saxe N et al. Validation of the U.K. Working
of the criteria, and misclassification is common due to an Party diagnostic criteria for atopic eczema in a Xhosa‐speaking
increase in borderline cases [5]. Moreover, formal criteria African population. Br J Dermatol 2007;156:111–16.
15 Yamada E, Vanna AT, Naspitz CK, Sole D. International Study of
are rarely used in routine clinical practice and there is Asthma and Allergies in Childhood (ISAAC): validation of the writ-
considerable heterogeneity about how AD is defined even ten questionnaire (eczema component) and prevalence of atopic
among physicians, depending on their medical back- eczema among Brazilian children. J Investig Allergol Clin Immunol
ground or country of practice [20]. These are particular 2002;12:34–41.
16 Chan HH, Pei A, Van Krevel C et  al. Validation of the Chinese
limitations for retrospective studies of health records or translated version of ISAAC core questions for atopic eczema. Clin Exp
claims‐based data [21]. Allergy 2001;31:903–7.
Some ‘objective’ tests such as raised total or specific 17 Firooz A, Davoudi SM, Farahmand AN et al. Validation of the diag-
nostic criteria for atopic dermatitis. Arch Dermatol 1999;135:514–16.
immunoglobulin E (IgE) and positive skinprick tests are 18 Haileamlak A, Lewis SA, Britton J et al. Validation of the International
often included in stricter definitions of ‘atopic’ dermatitis Study of Asthma and Allergies in Children (ISAAC) and U.K. criteria
(e.g. Millennium criteria [22]). However, requirement of for atopic eczema in Ethiopian children. Br J Dermatol 2005;152:
these factors, depending on the population studied, can 735–41.
19 Oien T, Storro O, Johnsen R. Assessing atopic disease in children two
exclude up to about two‐thirds of individuals with the to six years old: reliability of a revised questionnaire. Prim Care
disease phenotype [23,24]. Over‐reliance on laboratory Respir J 2008;17:164–8.
measures that may reflect epiphenomena related to the 20 Williams HC. Diagnostic criteria for atopic dermatitis: where do we
go from here? Arch Dermatol 1999;135:583–6.
study of sick individuals with severe disease in a hospital 21 Dizon MP, Yu AM, Singh RK et al. Systematic review of atopic derma-
setting should be avoided in population studies until titis disease definition in studies using routinely collected health
better disease markers are found. Without any objective data. Br J Dermatol 2018;178:1280–7.
Chapter 13  Epidemiology of Atopic Dermatitis 171

22 Bos JD, Van Leent EJ, Sillevis Smitt JH. The millennium criteria for the Severity
diagnosis of atopic dermatitis. Exp Dermatol 1998;7:132–8.
23 Flohr C, Weiland SK, Weinmayr G et al. The role of atopic sensiti-
In public health terms, the severity distribution of AD is
zation in flexural eczema: findings from the International Study more significant than the total prevalence of AD (which
of Asthma and Allergies in Childhood Phase Two. J Allergy Clin may include many mild cases), as severity is likely to
Immunol 2008;121:141–7. determine those who use or need to use available health
24 Schafer T, Heinrich J, Wjst M et  al. Association between severity of
atopic eczema and degree of sensitization to aeroallergens in school- services. Few studies have rigorously examined the severity
children. J Allergy Clin Immunol 1999;104:1280–4. distribution of AD in the community. A population‐based
study conducted in Nottinghamshire, England, found
Descriptive epidemiology of atopic that the severity distribution of AD (defined by a derma-
dermatitis tologist) in 1760 children living around Nottingham,
England, was 84% mild, 14% moderate and 2% severe
Prevalence and incidence [43]. In a US survey of 10 386 children with AD, mild
With so many different time periods and methods of disease accounted for 67% of the cases, moderate 26%

ATOPIC DERMATITIS
measuring AD, making comparisons of the prevalence and severe 7% [35]. Geographical variation in AD sever-
of AD between countries is very difficult. For most ity distribution has also been reported [44].

SECTION 3:
­epidemiological studies, the authors recommend that
the 1‐year period prevalence measure is used, in order Morbidity
to reflect the intermittent nature of the disease and to AD ranked first among common skin diseases with
overcome the effect of seasonality. A number of com- respect to disability‐adjusted life‐years and years lived
munity‐based and population‐based prevalence sur- with a disease in the World Health Organization (WHO)
veys have been conducted in various countries around 2010 Global Burden of Disease survey [45,46]. The impact
the world, reporting a wide range of prevalence rates of AD on health‐related quality of life in children is com-
across countries [1–30]. Together, these surveys sug- parable to that of other chronic illnesses such as asthma
gest that around 10–30% of children in developed and diabetes [47]. Furthermore, impairment of quality of
countries suffer from AD, with some studies demon- life appears to be directly related to the severity of AD
strating a plateau in these prevalence estimates over [48]. A US population‐based study found that 25–33% of
recent years [2,4,12,31]. In contrast, the prevalence of adults with AD reported fatigue, daytime sleepiness
AD in developing countries has been generally lower and/or insomnia, which were important predictors of poorer
but is increasing [8,32]. overall health status, number of sick days and doctor visits
To date, the ISAAC has published the most compre- [49]. The psychological morbidity associated with a life-
hensive prevalence and trend data on AD [4,31,33]. The time of scratching, sleep loss and the stigma of a visible
three phases of the ISAAC have included nearly 2 million skin disease can also affect families to a considerable
children from over 100 countries. Using the revised degree [50,51]. Recent studies have found that children
UKWP criteria via questionnaires to identify AD, the and adults with AD are at greater risk for attention‐deficit
ISAAC has found highly variable AD prevalence rates hyperactivity disorder, depression, anxiety and conduct
across different countries, ranging from 0.9% in India to disorder, particularly when AD is severe [52,53]. Children
22.5% in Ecuador among 6‐ and 7‐year‐olds and from with AD also have a greater number of comorbid medical
0.2% in China to 24.6% in Colombia among 13‐ to 14‐year‐ conditions and poorer overall health [35]. In two popula-
olds [31]. Data from ISAAC Phases 1 and 3 have demon- tion‐based surveys in the USA, approximately one in five
strated rising prevalences of AD among 13‐ to 14‐year‐olds adults with AD either met clinical criteria for major
in developing nations over a period of 5–10 years but no depressive disorder or were diagnosed by a physician
significant change or even decreases in prevalence in with depression [54–57]. The association of AD with non-
many developed areas [32]. In contrast, the prevalence of allergic comorbidities is a growing area of investigation.
AD symptoms among 6‐ to 7‐year‐olds has continued to Early studies suggest that AD may be associated with an
increase in both developed and developing nations over increased risk for alopecia areata, vitiligo, rheumatoid
this time period. arthritis, inflammatory bowel disease and extracutaneous
A systematic review of 69 studies examining international infections, but a decreased risk for type 1 diabetes and
trends in AD between 1990 and 2010 also found lifetime selected cancers [58–62].
AD prevalence rates of over 20% in some developed
nations, with increasing rates of AD in Africa, eastern Cost
Asia, western Europe and parts of northern Europe [8]. AD is a costly disease and carries an economic burden
In two population‐based survey studies in the USA, the similar to that of other chronic diseases such as emphy-
mean prevalence rate of AD among children under sema, asthma and epilepsy [63,64]. Although costs‐
18 years of age increased from 10.2% in 2003–2004 to of‐illness studies are relatively few, previous studies from
12.98% in 2007–2008 [34,35], but prevalence estimates also the UK, USA, Germany, Australia and the Netherlands
varied widely, from 8.7% to 18.1%, across different states found highly variable costs across countries, which are
and districts [34]. Finally, prevalence data on adult AD probably due to differences in the study populations
are sparse; to date, studies from the USA, Japan, Australia, (hospital vs. community‐based studies) and the cost com-
Russia and Scotland have found prevalence rates of ponents included [64,65]. In one study, the estimated
2–10% [36–42]. direct costs of AD were nearly 3.8 billion US dollars per
172 Section 3  Atopic Dermatitis and Related Disorders

year [64]. Moreover, these estimates often do not include AD‐genetic risk score as well as personal and parental
costs associated with comorbid diseases or indirect costs, history of atopic disease. A newly defined subset of mid‐
both of which can be significant [66,67]. Thus, the eco- onset‐resolving AD, not associated with FLG mutations,
nomic burden of AD is considerable and probably much was strongly associated with asthma [81].
greater than previously reported. Factors that may indicate a worse prognosis include
severe childhood disease, early onset and a concomitant
Age, sex and natural history or family history of asthma or hay fever [77,78,82]. One
Childhood AD starts in early life in the majority of cases birth cohort study of 1314 children followed to the age of
[17,68], but may also begin later in childhood or even 7 years in Germany (The Multicenter Allergy Study)
in  adulthood [69,70]. In a sub‐sample of the Avon deserves special mention because it included sequential
Longitudinal Study of Parents and Children (ALSPAC), physical examinations, parental interviews and specific
1509 randomly selected children were followed from IgE levels. Of the 21.5% of children with AD in the first
birth until 5 years of age; 33% had onset of AD before 2 years of life, 43.2% were in complete remission by the
ATOPIC DERMATITIS

18 months of age and 18% had onset after this age [71]. age of 3 years, 38.3% had an intermittent pattern of dis-
Adult‐onset AD is also becoming increasingly recognized ease and 18.7% had symptoms of AD every year. A poorer
SECTION 3:

[69,72]. Two studies in Singapore and Nigeria found that prognosis was related to IgE sensitization (adjusted
13.6–24.5% of AD patients in a tertiary dermatology clinic cumulative odds ratio 2.76; 95% confidence interval [CI],
had onset of AD after the age of 21 years [73,74]. 1.29–5.91) and early disease severity (adjusted cumula-
A systematic review and meta‐analysis showed that tive odds ratio 5.86; 95% CI, 3.04–11.29). Interestingly,
AD characteristics are heterogeneous and vary by region early AD without early wheezing did not increase the risk
and age. The most prevalent AD features were pruritus, of subsequent wheeze at school entry (adjusted odds
lichenification and xerosis. Studies from Africa reported ratio 1.11; 95% CI, 0.56–2.20) [83]. Children with loss‐of‐
more papular lichenoid lesions, palmar hyperlinearity, function mutations in filaggrin, an essential epidermal
ichthyosis and orbital darkening [75]. barrier protein (see Chapter 14), have also been found to
Minor sex differences in AD, namely a slightly higher have earlier onset and more persistent AD [79,84].
prevalence among females, have been noted previously.
The Phase 3 ISAAC evaluated over 1 million children in Ethnic group and migrant studies
two distinct age groups (6‐ to 7‐ and 13‐ to 14‐year‐olds) Several studies have addressed ethnicity as an aetiological
and found slightly higher rates of current eczema symp- factor in AD, and particularly helpful are studies from
toms in girls than boys, especially in the older age group communities of different ethnic origin in the same coun-
[31]. While the ISAAC authors suggested that misclassifi- try. For instance, black Caribbean children born in the UK
cation of irritant or allergic dermatitis might explain and children of Chinese origin born in Australia are at an
their observed gender disparities, another study of 1456 increased risk of developing AD in comparison with their
children in the Isle of Wight birth cohort from the UK also (mainly) white Caucasian peers [85, 86]. As these studies
found higher AD prevalence among girls than boys after recorded point prevalence of AD, it is difficult to say
the age of 10 years [76]. Nevertheless, gender differences whether these differences were due to disease chronicity
have not been a consistent finding across studies [34]. or disease incidence between ethnic groups. Another
Few longitudinal studies have examined the natural study of Asian children in Leicester (UK) found no differ-
history of AD, and it should not be assumed that the ence in the prevalence of AD between different ethnic
prognosis of AD cases studied in a hospital setting (which groups, although Asian children were three times as likely
are usually quite severe by definition) is the same as that to be seen in the local dermatology department, perhaps
for the majority of milder AD cases in the community. reflecting lack of parental familiarity with the condition
Studies of well‐defined AD populations suggest that [87]. In contrast, in a US population‐based study, individ-
patients have a clearance rate of around 60% by the age of uals of black or African‐American race were significantly
16 years [77,78]. True clearance rates are probably even more likely to have AD, even after controlling for socio-
lower, as many individuals relapse at some stage in adult- economic factors [34]. Moreover, AD severity distribution
hood (e.g. by developing irritant hand eczema). In two may also vary with respect to race and ethnicity [44].
studies using a paediatric AD registry, most patients Migrant studies have been helpful in suggesting a
reported active disease at every age of childhood and into strong role for the environment in the aetiology of AD.
early adulthood, suggesting that persistence of mild and Studies of Chinese immigrants in Hawaii and children
moderate paediatric AD into adulthood is not insignifi- from Tokelau who migrated to New Zealand found large
cant [79]. Older patients also sought medical attention increases in the prevalence of AD among migrant children
less often, which may contribute to the erroneous percep- compared with similar genetic groups in their country of
tion that AD tends to resolve with age [80]. origin [88]. Another study found that black Caribbean
In two birth cohort studies, 9894 children from the UK children living in London were around three times more
(ALSPAC) and 3652 from the Netherlands (PIAMA), dis- likely to have AD than similar children living in Kingston,
tinct subclasses were identified. The most prevalent was Jamaica, regardless of the method used to diagnose AD
early‐onset‐early‐resolving AD, which was associated [86]. Findings of lower AD prevalence among immigrants
with male sex. Early‐onset‐persistent and early‐onset‐ versus native‐born persons have been observed in a
late‐resolving AD were most strongly associated with the number of countries [3,89–92]. One US study found that
Chapter 13  Epidemiology of Atopic Dermatitis 173

foreign‐born children, particularly those with parents for studies of skinprick tests, wheezing and hay fever,
who were also foreign‐born, had lower odds of AD com- which may be less prone to diagnostic labels. An increased
pared to US‐born children [93]. However, this protective prevalence of AD has been found in comparative cross‐
effect declined over time, as prolonged residence in the sectional surveys [13,103] and, more convincingly, in
USA for 10 or more years was associated with a greater population‐based longitudinal studies in Japan and the
risk of AD compared to foreign‐born children who had USA [30,104] and two large sequential worldwide eczema
resided in the USA for less than 2 years. Together, these surveys conducted as part of the ISAAC [32]. The ISAAC
migrant studies support the notion that environmental surveys showed that eczema prevalence is still on the rise
factors associated with urbanization and ‘western’ life- among younger children (aged 6–7 years) in both devel-
style are important in the aetiology of AD. oped and developing nations, whereas eczema symptoms
seem to be levelling off in older children (aged 13–14
Social class and family size years) in some countries with previously high rates of
Reported eczema has exhibited a strong positive social AD. In a systematic review examining international time

ATOPIC DERMATITIS
class gradient (i.e. it is more common in higher socio- trends in AD, the prevalence rates of AD in Africa, eastern
economic groups) [94]. Although such a trend could be Asia, western Europe and parts of northern Europe

SECTION 3:
due to enhanced reporting of eczema by mothers in appeared to increase from 1990 to 2010 [8]. The reasons
advantaged socioeconomic groups or increased labelling for this increase in AD are not clear, but environmental
by doctors, an identical trend was found among children factors in genetically predisposed individuals are the
with examined eczema in a UK national birth cohort most likely explanation (see Specific risk factors).
study for children in the 1970s [95]. In further studies, Moreover, the ISAAC survey findings suggest that the
higher household incomes and parental education levels plateauing prevalence rates of AD in some countries may
have continued to be independent risk factors for the soon reach their highest limits [32].
development of AD [34,91,96]. A reduced risk of AD has
also been observed for children in larger families, espe- References
cially if older siblings are present [34,97]. Similar findings 1 Amouri M, Masmoudi A, Borgi N et al. Atopic dermatitis in Tunisian
schoolchildren. Pan Afr Med J 2011;9:34.
have been reported for hay fever, asthma, skin test reac- 2 Annesi‐Maesano I, Mourad C, Daures JP et al. Time trends in preva-
tivity and allergen‐specific IgE levels [97], and these lence and severity of childhood asthma and allergies from 1995 to
observations led to the formulation of the so‐called 2002 in France. Allergy 2009;64:798–800.
3 Apfelbacher CJ, Diepgen TL, Schmitt J. Determinants of eczema:
‘hygiene hypothesis’, the notion that reduced exposure
population‐based cross‐sectional study in Germany. Allergy 2011;
­
to certain microbes might be responsible for the increase 66:206–13.
in allergy prevalence over past decades (see The hygiene 4 Asher MI, Montefort S, Bjorksten B et al. Worldwide time trends in
hypothesis). the prevalence of symptoms of asthma, allergic rhinoconjunctivitis,
and eczema in childhood: ISAAC Phases One and Three repeat
multicountry cross‐sectional surveys. Lancet 2006;368:733–43.
Geographical variation 5 Augustin M, Radtke MA, Glaeske G et al. Epidemiology and comor-
Although up to about 30% of children in developed coun- bidity in children with psoriasis and atopic eczema. Dermatology
tries suffer from AD, the prevalence of AD is lower in 2015;231:35–40.
6 Belyhun Y, Amberbir A, Medhin G et al. Prevalence and risk factors of
many developing countries [33]. However, the notion wheeze and eczema in 1‐year‐old children: the Butajira birth cohort,
that AD is rare in developing countries is now being Ethiopia. Clin Exp Allergy 2010;40:619–26.
challenged by data from the ISAAC that suggest that 7 Chalmers DA, Todd G, Saxe N et al. Validation of the U.K. Working
Party diagnostic criteria for atopic eczema in a Xhosa‐speaking
symptoms of AD are becoming increasingly common in
African population. Br J Dermatol 2007;156:111–16.
cities in Latin America and Africa [32]. Furthermore, there 8 Deckers IA, McLean S, Linssen S et al. Investigating international
tends to be a gradient between urban and rural areas time trends in the incidence and prevalence of atopic eczema
within the same country. A clear urban–rural gradient 1990‐2010: a systematic review of epidemiological studies. PLoS
One 2012;7:e39803.
has, for instance, been demonstrated in studies conducted 9 Farajzadeh S, Esfandiarpour I, Sedaghatmanesh M, Saviz M.
in the USA, China, Sweden and Ethiopia [28,34,98,99]. Epidemiology and clinical features of atopic dermatitis in kerman, a
Finally, climatic factors may also contribute to geo- desert area of iran. Ann Dermatol 2014;26:26–34.
graphical variation in AD. Two ecological studies have 10 Finnbogadottir AF, Ardal B, Eiriksson H et al. A long‐term follow‐up
of allergic diseases in Iceland. Pediatr Allergy Immunol 2012;23:
found lower AD prevalence in areas with increased sun 181–5.
exposure, warmer temperatures and greater humidity 11 Govaere E, Van Gysel D, Verhamme KM et  al. The prevalence,
[100,101]. However, in contrast, another study observed characteristics of and risk factors for eczema in Belgian schoolchil-
dren. Pediatr Dermatol 2009;26:129–38.
that among patients with AD, higher temperatures and 12 Grize L, Gassner M, Wuthrich B et al. Trends in prevalence of asthma,
greater sun exposure were associated with worse AD allergic rhinitis and atopic dermatitis in 5‐7‐year old Swiss children
disease control [102]. from 1992 to 2001. Allergy 2006;61:556–62.
13 Hansen TE, Evjenth B, Holt J. Increasing prevalence of asthma,
allergic rhinoconjunctivitis and eczema among schoolchildren:
Secular trends three surveys during the period 1985–2008. Acta Paediatr 2013;102:
There is evidence to suggest that the prevalence of AD has 47–52.
increased two‐ to three‐fold over the last 30 years [32,103]. 14 Harris JM, Cullinan P, Williams HC et  al. Environmental associa-
tions with eczema in early life. Br J Dermatol 2001;144:795–802.
Some of these changes reflect secular trends in increased
15 Henriksen L, Simonsen J, Haerskjold A et al. Incidence rates of atopic
recognition of the disease by parents and doctors. dermatitis, asthma, and allergic rhinoconjunctivitis in Danish and
However, similar trends over time have been observed Swedish children. J Allergy Clin Immunol 2015;136:360–6.e2.
174 Section 3  Atopic Dermatitis and Related Disorders

16 Kanwar AJ, De D. Epidemiology and clinical features of atopic der- 42 Silverberg JI, Hanifin JM. Adult eczema prevalence and associations
matitis in India. Indian J Dermatol 2011;56:471–5. with asthma and other health and demographic factors: a US population‐
17 Kay J, Gawkrodger DJ, Mortimer MJ, Jaron AG. The prevalence of based study. J Allergy Clin Immunol 2013;132:1132–8.
childhood atopic eczema in a general population. J Am Acad Dermatol 43 Emerson RM, Williams HC, Allen BR. Severity distribution of
1994;30:35–9. atopic dermatitis in the community and its relationship to secondary
18 Kjaer HF, Eller E, Host A et al. The prevalence of allergic diseases in an referral. Br J Dermatol 1998;139:73–6.
unselected group of 6‐year‐old children. The DARC birth cohort 44 Silverberg JI, Simpson EL. Associations of childhood eczema severity:
study. Pediatr Allergy Immunol 2008;19:737–45. a US population‐based study. Dermatitis 2014;25:107–14.
19 Lee YL, Li CW, Sung FC et al. Environmental factors, parental atopy 45 Murray CJ, Vos T, Lozano R et  al. Disability‐adjusted life years
and atopic eczema in primary‐school children: a cross‐sectional study (DALYs) for 291 diseases and injuries in 21 regions, 1990‐2010: a sys-
in Taiwan. Br J Dermatol 2007;157:1217–24. tematic analysis for the Global Burden of Disease Study 2010. Lancet
20 Lee YL, Su HJ, Sheu HM et  al. Traffic‐related air pollution, climate, 2012;380:2197–223.
and prevalence of eczema in Taiwanese school children. J Invest 46 Vos T, Flaxman AD, Naghavi M et  al. Years lived with disability
Dermatol 2008;128:2412–20. (YLDs) for 1160 sequelae of 289 diseases and injuries 1990‐2010: a
21 Lowe AJ, Hosking CS, Bennett CM et al. Skin prick test can identify systematic analysis for the Global Burden of Disease Study 2010.
eczematous infants at risk of asthma and allergic rhinitis. Clin Exp Lancet 2012;380:2163–96.
ATOPIC DERMATITIS

Allergy 2007;37:1624–31. 47 Beattie PE, Lewis‐Jones MS. A comparative study of impairment of


22 Martin PE, Koplin JJ, Eckert JK et al. The prevalence and socio‐demo- quality of life in children with skin disease and children with other
graphic risk factors of clinical eczema in infancy: a population‐based chronic childhood diseases. Br J Dermatol 2006;155:145–51.
SECTION 3:

observational study. Clin Exp Allergy 2013;43:642–51. 48 Ben‐Gashir MA, Seed PT, Hay RJ. Are quality of family life and
23 Sangsupawanich P, Chongsuvivatwong V, Mo‐Suwan L, disease severity related in childhood atopic dermatitis? J Eur Acad
Choprapawon C. Relationship between atopic dermatitis and wheeze Dermatol Venereol 2002;16:455–62.
in the first year of life: analysis of a prospective cohort of Thai chil- 49 Silverberg JI, Garg NK, Paller AS et al. Sleep disturbances in adults
dren. J Investig Allergol Clin Immunol 2007;17:292–6. with eczema are associated with impaired overall health: a US
24 Schultz Larsen F, Diepgen T, Svensson A. The occurrence of atopic population‐based study. J Invest Dermatol 2015;135:56–66.
dermatitis in north Europe: an international questionnaire study. 50 McKenna SP, Doward LC, Meads DM et al. Quality of life in infants
J Am Acad Dermatol 1996;34(5 Pt 1):760–4. and children with atopic dermatitis: addressing issues of differential
25 Smidesang I, Saunes M, Storro O et al. Atopic dermatitis among item functioning across countries in multinational clinical trials.
2‐year olds; high prevalence, but predominantly mild disease  –  the Health Qual Life Outcomes 2007;5:45.
PACT study, Norway. Pediatr Dermatol 2008;25:13–18. 51 Misery L, Finlay AY, Martin N et al. Atopic dermatitis: impact on the
26 Tanaka K, Miyake Y, Arakawa M et  al. Prevalence of asthma and quality of life of patients and their partners. Dermatology 2007;
wheeze in relation to passive smoking in Japanese children. Ann 215:123–9.
Epidemiol 2007;17:1004–10. 52 Strom MA, Fishbein AB, Paller AS, Silverberg JI. Association between
27 Vinding GR, Zarchi K, Ibler KS et al. Is adult atopic eczema more com- atopic dermatitis and attention deficit hyperactivity disorder in U.S.
mon than we think? ‐ A population‐based study in Danish adults. children and adults. Br J Dermatol 2016;175:920–9.
Acta Derm Venereol 2014;94:480–2. 53 Smirnova J, von Kobyletzki LB, Lindberg M et al. Atopic dermatitis,
28 Xu F, Yan S, Li F et al. Prevalence of childhood atopic dermatitis: an educational attainment and psychological functioning: a national
urban and rural community‐based study in Shanghai, China. PLoS cohort study. Br J Dermatol 2019;180(3):559–64.
One 2012;7:e36174. 54 Dalgard FJ, Gieler U, Tomas‐Aragones L et  al. The psychological
29 Yu JS, Lee CJ, Lee HS et al. Prevalence of atopic dermatitis in Korea: burden of skin diseases: a cross‐sectional multicenter study among
analysis by using national statistics. J Korean Med Sci 2012;27:681–5. dermatological out‐patients in 13 European countries. J Invest
30 Yura A, Kouda K, Iki M, Shimizu T. Trends of allergic symptoms in Dermatol 2015;135:984–91.
school children: large‐scale long‐term consecutive cross‐sectional 55 Schmitt J, Buske‐Kirschbaum A, Roessner V. Is atopic disease a risk
studies in Osaka Prefecture, Japan. Pediatr Allergy Immunol factor for attention‐deficit/hyperactivity disorder? A systematic
2011;22:631–7. review. Allergy 2010;65:1506–24.
31 Odhiambo JA, Williams HC, Clayton TO et al. Global variations in 56 Yaghmaie P, Koudelka CW, Simpson EL. Mental health comorbidity
prevalence of eczema symptoms in children from ISAAC Phase in patients with atopic dermatitis. J Allergy Clin Immunol 2013;131:
Three. J Allergy Clin Immunol 2009;124:1251–8.e23. 428–33.
32 Williams H, Stewart A, von Mutius E et al. Is eczema really on the 57 Yu SH, Silverberg JI. Association between atopic dermatitis and
increase worldwide? J Allergy Clin Immunol 2008;121:947–54.e15. depression in US Adults. J Invest Dermatol 2015;135:3183–6.
33 Williams H, Robertson C, Stewart A et al. Worldwide variations in 58 Deckert S, Kopkow C, Schmitt J. Nonallergic comorbidities of atopic
the prevalence of symptoms of atopic eczema in the International eczema: an overview of systematic reviews. Allergy 2014;69:37–45.
Study of Asthma and Allergies in Childhood. J Allergy Clin 59 Mohan GC, Silverberg JI. Association of vitiligo and alopecia areata
Immunol 1999;103:125–38. with atopic dermatitis: a systematic review and meta‐analysis. JAMA
34 Shaw TE, Currie GP, Koudelka CW, Simpson EL. Eczema prevalence Dermatol 2015;151:522–8.
in the United States: data from the 2003 National Survey of Children’s 60 Schmitt J, Schwarz K, Baurecht H et al. Atopic dermatitis is associated
Health. J Invest Dermatol 2011;131:67–73. with an increased risk for rheumatoid arthritis and inflammatory
35 Silverberg JI, Simpson EL. Association between severe eczema in bowel disease, and a decreased risk for type 1 diabetes. J Allergy Clin
children and multiple comorbid conditions and increased health- Immunol 2016;137:130–6.
care utilization. Pediatr Allergy Immunol 2013;24:476–86. 61 Silverberg JI, Silverberg NB. Childhood atopic dermatitis and warts
36 Herd RM, Tidman MJ, Prescott RJ, Hunter JA. Prevalence of atopic are associated with increased risk of infection: a US population‐based
eczema in the community: the Lothian Atopic Dermatitis study. Br J study. J Allergy Clin Immunol 2014;133:1041–7.
Dermatol 1996;135:18–19. 62 Strom MA, Silverberg JI. Association between atopic dermatitis
37 Katsarou A, Armenaka M. Atopic dermatitis in older patients: par- and  extracutaneous infections in US adults. Br J Dermatol 2017;
ticular points. J Eur Acad Dermatol Venereol 2011;25:12–18. 176:495–7.
38 Marks R, Kilkenny M, Plunkett A, Merlin K. The prevalence of com- 63 Ellis CN, Drake LA, Prendergast MM et al. Cost of atopic dermatitis
mon skin conditions in Australian school students: 2. Atopic dermati- and eczema in the United States. J Am Acad Dermatol 2002;46:
tis. Br J Dermatol 1999;140:468–73. 361–70.
39 Muto T, Hsieh SD, Sakurai Y et al. Prevalence of atopic dermatitis in 64 Mancini AJ, Kaulback K, Chamlin SL. The socioeconomic impact of
Japanese adults. Br J Dermatol 2003;148:117–21. atopic dermatitis in the United States: a systematic review. Pediatr
40 Ronmark EP, Ekerljung L, Lotvall J et al. Eczema among adults: preva- Dermatol 2008;25:1–6.
lence, risk factors and relation to airway diseases. Results from a large‐ 65 Verboom P, Hakkaart‐Van L, Sturkenboom M et  al. The cost of
scale population survey in Sweden. Br J Dermatol 2012;166:1301–8. atopic dermatitis in the Netherlands: an international comparison.
41 Saeki H, Tsunemi Y, Fujita H et  al. Prevalence of atopic dermatitis Br J Dermatol 2002;147:716–24.
determined by clinical examination in Japanese adults. J Dermatol 66 Emerson RM, Williams HC, Allen BR. What is the cost of atopic
2006;33:817–19. dermatitis in preschool children? Br J Dermatol 2001;144:514–22.
Chapter 13  Epidemiology of Atopic Dermatitis 175

67 Suh DC, Sung J, Gause D et al. Economic burden of atopic manifesta- 93 Silverberg JI, Simpson EL, Durkin HG, Joks R. Prevalence of allergic
tions in patients with atopic dermatitis  –  analysis of administrative disease in foreign‐born American children. JAMA Pediatr 2013;
claims. J Manag Care Pharm 2007;13:778–89. 167:554–60.
68 Spergel JM, Paller AS. Atopic dermatitis and the atopic march. 94 Williams HC, Strachan DP, Hay RJ. Childhood eczema: disease of the
J Allergy Clin Immunol 2003;112(6 Suppl):S118–27. advantaged? BMJ 1994;308:1132–5.
69 Bannister MJ, Freeman S. Adult‐onset atopic dermatitis. Australas J 95 Golding J, Peters TJ. The epidemiology of childhood eczema: I. A
Dermatol 2000;41:225–8. population based study of associations. Paediatr Perinat Epidemiol
70 Williams HC, Strachan DP. The natural history of childhood eczema: 1987;1:67–79.
observations from the British 1958 birth cohort study. Br J Dermatol 96 Weber AS, Haidinger G. The prevalence of atopic dermatitis in
1998;139:834–9. children is influenced by their parents’ education: results of two
71 Perkin MR, Strachan DP, Williams HC et al. Natural history of atopic cross‐sectional studies conducted in Upper Austria. Pediatr Allergy
dermatitis and its relationship to serum total immunoglobulin E in Immunol 2010;21:1028–35.
a population‐based birth cohort study. Pediatr Allergy Immunol 97 von Mutius E, Martinez FD, Fritzsch C et al. Skin test reactivity and
2004;15:221–9. number of siblings. BMJ 1994;308:692–5.
72 Kulthanan K, Samutrapong P, Jiamton S, Tuchinda P. Adult‐onset 98 Schram ME, Tedja AM, Spijker R et al. Is there a rural/urban gradient
atopic dermatitis: a cross‐sectional study of natural history and clinical in the prevalence of eczema? A systematic review. Br J Dermatol

ATOPIC DERMATITIS
manifestation. Asian Pac J Allergy Immunol 2007;25:207–14. 2010;162:964–73.
73 Nnoruka EN. Current epidemiology of atopic dermatitis in south‐ 99 Yemaneberhan H, Flohr C, Lewis SA et al. Prevalence and associated
eastern Nigeria. Int J Dermatol 2004;43:739–44. factors of atopic dermatitis symptoms in rural and urban Ethiopia.

SECTION 3:
74 Tay YK, Khoo BP, Goh CL. The profile of atopic dermatitis in a tertiary Clin Exp Allergy 2004;34:779–85.
dermatology outpatient clinic in Singapore. Int J Dermatol 1999;38: 100 Osborne NJ, Ukoumunne OC, Wake M, Allen KJ. Prevalence
689–92. of eczema and food allergy is associated with latitude in Australia.
75 Yew YW, Thyssen JP, Silverberg JI. A systematic review and meta‐ J Allergy Clin Immunol 2012;129:865–7.
analysis of the regional and age‐related differences in atopic dermatitis 101 Silverberg JI, Hanifin J, Simpson EL. Climatic factors are associated
clinical characteristics. J Am Acad Dermatol 2019;80(2):390–401. with childhood eczema prevalence in the United States. J Invest
76 Ziyab AH, Raza A, Karmaus W et al. Trends in eczema in the first Dermatol 2013;133:1752–9.
18  years of life: results from the Isle of Wight 1989 birth cohort 102 Sargen MR, Hoffstad O, Margolis DJ. Warm, humid, and high sun
study. Clin Exp Allergy 2010;40:1776–84. exposure climates are associated with poorly controlled eczema:
77 Illi S, von Mutius E, Lau S, et al. The natural course of atopic derma- PEER (Pediatric Eczema Elective Registry) cohort, 2004‐2012. J Invest
titis from birth to age 7 years and the association with asthma. Dermatol 2014;134:51–7.
J Allergy Clin Immunol 2004;113:925–31. 103 Williams HC. Is the prevalence of atopic dermatitis increasing?
78 Peters AS, Kellberger J, Vogelberg C et al. Prediction of the incidence, Clin Exp Dermatol 1992;17:385–91.
recurrence, and persistence of atopic dermatitis in adolescence: a pro- 104 Jackson KD, Howie LD, Akinbami LJ. Trends in allergic condi-
spective cohort study. J Allergy Clin Immunol 2010;126:590–5.e1–3. tions among children: United States, 1997‐2011. NCHS Data Brief
79 Margolis JS, Abuabara K, Bilker W et  al. Persistence of mild to 2013(121):1–8.
moderate atopic dermatitis. JAMA Dermatol 2014;150:593–600.
80 Abuabara K, Hoffstad O, Troxel A et  al. Atopic dermatitis disease
control and age: A cohort study. J Allergy Clin Immunol 2015;136: Specific risk factors
190–2.e3.
81 Paternoster L, Savenije OEM, Heron J et al. Identification of atopic Genetics
dermatitis subgroups in children from 2 longitudinal birth cohorts.
There is strong evidence to suggest that genetic factors are
J Allergy Clin Immunol 2018;141:964–71.
82 Mortz CG, Andersen KE, Dellgren C et  al. Atopic dermatitis from important in the predisposition to AD. In addition to fam-
adolescence to adulthood in the TOACS cohort: prevalence, persis- ily studies, twin studies have shown much higher disease
tence and comorbidities. Allergy 2015;70:836–45. concordance for monozygotic than for dizygotic twins [1].
83 Rystedt I. Long term follow‐up in atopic dermatitis. Acta Derm
Venereol Suppl (Stockh) 1985;114:117–20. A systematic review of population‐based twin studies of
84 Rupnik H, Rijavec M, Korosec P. Filaggrin loss‐of‐function mutations AD has estimated the heritability of AD to be approxi-
are not associated with atopic dermatitis that develops in late child- mately 75% [2]. Genome‐wide screens of families with
hood or adulthood. Br J Dermatol 2015;172:455–61.
AD have identified chromosomal regions that overlap
85 Mar A, Tam M, Jolley D, Marks R. The cumulative incidence of atopic
dermatitis in the first 12 months among Chinese, Vietnamese, and with other skin diseases, in particular psoriasis, and with
Caucasian infants born in Melbourne, Australia. J Am Acad Dermatol inflammatory and autoimmune diseases [3]. These,
1999;40:597–602. together with candidate gene studies, suggest that genetic
86 Williams HC, Pembroke AC, Forsdyke H et  al. London‐born black
Caribbean children are at increased risk of atopic dermatitis. J Am
predisposition to an impaired skin barrier, for instance
Acad Dermatol 1995;32(2 Pt 1):212–17. due to loss‐of‐function mutations in the filaggrin (FLG)
87 Neame RL, Berth‐Jones J, Kurinczuk JJ, Graham‐Brown RA. gene located within the epidermal differentiation com-
Prevalence of atopic dermatitis in Leicester: a study of methodology plex on chromosome 1q21, is of particular importance for
and examination of possible ethnic variation. Br J Dermatol 1995;
132:772–7. AD pathogenesis [4,5]. Moreover, genetic predisposition
88 Waite DA, Eyles EF, Tonkin SL, O’Donnell TV. Asthma prevalence in to immune dysregulation also contributes to the patho-
Tokelauan children in two environments. Clin Allergy 1980;10:71–5. genesis of AD. To date, genome‐wide association studies
89 Leung RC, Carlin JB, Burdon JG, Czarny D. Asthma, allergy and
atopy in Asian immigrants in Melbourne. Med J Aust 1994;161:
and meta‐analyses have identified a total of 31 suscepti-
418–25. bility loci for AD, many of which are implicated in innate
90 Marcon A, Cazzoletti L, Rava M et  al. Incidence of respiratory and immune signalling and T‐cell function [6]. However,
allergic symptoms in Italian and immigrant children. Respir Med genetics alone cannot explain the rise in AD prevalence
2011;105:204–10.
91 Schmitz R, Atzpodien K, Schlaud M. Prevalence and risk factors of over past decades. It is also difficult to explain the ten-
atopic diseases in German children and adolescents. Pediatr Allergy dency of AD to clear spontaneously in genetic terms.
Immunol 2012;23:716–23. Genetic and environmental factors for AD probably work
92 Tedeschi A, Barcella M, Bo GA, Miadonna A. Onset of allergy and
in concert rather than in competition, and the social class
asthma symptoms in extra‐European immigrants to Milan, Italy:
possible role of environmental factors. Clin Exp Allergy 2003;33: gradient, geographical variation and migrant studies
449–54. described earlier all point quite strongly to environmental
176 Section 3  Atopic Dermatitis and Related Disorders

factors playing a critical role in the expression of the AD e­ nvironmental factors other than parasites, one study has
phenotype. (Genetic factors are discussed in more detail suggested that there can be an inverse relationship
­
in Chapter 14.) between cord blood cat‐specific IgG and cat‐specific IgE
during later childhood [16]. Overall, these observations
Fetal predictors suggest that high allergen exposure in the mother leads to
Care should be taken in distinguishing between genetic low allergy risk in the offspring, but these are probably
factors and environmental factors operating in utero or only some of the factors involved. For instance, little is
during early life. As the vast majority of AD cases present currently known about the role of viral and bacterial
in the first year of life, and heritability is more strongly infections during pregnancy and AD risk in the off-
linked to the maternal side [7], it seems logical to hypoth- spring, and the few studies to date have not shown a con-
esize that environmental influences operating in utero or vincing association [17,18]. However, increased exposure
in early infancy may be important in determining disease to antibiotics in utero is associated with an increased risk
expression. In one systematic review, prenatal maternal of AD in a dose‐related manner [19], and frequent use of
ATOPIC DERMATITIS

stress was found to be associated with a higher risk of AD antibiotics within the first 2 years of life can have a simi-
and other atopy in the offspring [8]. Fetal immunoglobu- lar promoting effect on the clinical expression of allergic
SECTION 3:

lin can be detected as early as 26 weeks’ gestation, and the disease [20,21]. A meta‐analysis of studies examining
house dust mite allergen Der p 1 has been detected in postnatal antibiotic exposure estimated a 40% overall
amniotic fluid [9]. Amniotic fluid levels of IgE are also increased odds of AD in children with antibiotic use
higher in atopic than in nonatopic mothers [9]. Amniotic within the first year of life and observed increasing odds
fluid is predominantly absorbed via the fetal gut [10], and (+7%) with each additional antibiotic course [22].
because the fetal gut, in contrast to the fetal lungs and
skin, has mature antigen‐presenting cells, it is the most The role of the gut microflora and skin
likely site of sensitization [9]. In addition, allergen bound microbiome
to IgG can cross the placenta during the third trimester It has become evident that the commensal microflora of
[11]. It is thought that such early sensitization represents a the gut plays a major role in the ‘education’ of the immune
crucial first step in the development of the child’s immune system of the newborn [23]. Several researchers have
system and its ability to distinguish between friend and suggested that the use of antibiotics both in utero and
foe. In evolutionary terms, helminth parasites are likely to during early life may have a direct influence on the com-
have been important threats to child survival, and human position of the gut microflora. This, in turn, could prevent
host immune responses to parasites are in principle aller- the normal shift from type 2 T‐helper cell dominance at
gic, marked by raised total and specific IgE, overexpres- birth towards a type 1 T‐helper cell‐rich milieu, predispos-
sion of type 2 T‐helper cell cytokines and eosinophilia. It ing to allergic disease [24]. Prenatal factors such as length
has now been shown that in endemic areas fetal exposure of gestation and mode of delivery, as well as other envi-
to helminth antigens occurs in utero through maternal ronmental exposures such as diet and family size, have
infection, and cord blood lymphocytes from babies born also been associated with the altered gut microflora
to helminth‐infected mothers produce parasite‐specific observed in children with AD [25]. Indirect evidence also
immunoglobulins. This not only allows future recogni- comes from studies comparing the gut microflora in chil-
tion of parasite antigen in the host but also generates a dren with and without AD. The latter have significantly
degree of immune tolerance to parasite antigen [12]. more lactobacilli but fewer enterobacteria than their
Through the known cross‐reactivity between certain counterparts with AD, and these changes precede the
helminth antigens and environmental allergens, such as clinical manifestation of AD [26]. Lower bacterial diversity
house dust mites, this may also convey protection against has also been observed in the early gut microbiota of chil-
atopy and allergic disease [13]. Supporting evidence for dren who subsequently develop AD and allergic sensiti-
this hypothesis comes from a birth cohort study among zation [27]. A change in gut microflora composition could
103 mother–infant pairs in Uganda, where cumulative not only change the stimulation of the infant’s immune
eczema risk was decreased by 74% until the age of 15 months system across the intestinal mucosa but also enhance the
among children whose mothers were infected with exposure to allergens, further contributing to the devel-
­helminths during the last trimester of pregnancy (adjusted opment of allergic disease. Indeed, there is some evidence
OR = 0.26, 0.08–0.83 [13]). Our reduced exposure to para- that the permeability of the gut mucosa is increased in AD
sites might therefore be one of the reasons why AD has [28]. However, all of these results need to be treated with
become increasingly common over past decades in the caution, and further longitudinal studies that sequen-
developed world [13]. In addition, the fetal immune tially measure gut microflora composition in early life
system is generally biased towards a type 2 T‐helper cell‐ alongside careful phenotyping of AD are still required.
dominated milieu, marked by overexpression of IL‐4, The skin microbiome, which normally consists of
IL‐13 and IL‐10, probably in order to suppress a potential hundreds of different bacterial species but most abundantly
rejection of the fetus by the maternal immune system [14]. Propionibacterium, Staphylococcus and Corynebacterium
In children who develop allergic disease, the type 2 T‐ spp., has also been implicated as a potential driver in AD
helper cell dominance remains postnatally, as decreased pathogenesis (see Chapter 3). There is continuous cross‐
interferon‐γ (IFN‐γ) production is a consistent feature in talk between the skin microbiota and the skin immune system,
neonates born to atopic mothers [15]. With regard to with skin commensal bacteria playing an important role in
Chapter 13  Epidemiology of Atopic Dermatitis 177

host defence pathways, regulatory T‐cell function and A few intervention studies have thus been undertaken in
expression of innate immune factors in the skin [29]. The an attempt to resolve such partially contradicting evi-
composition of the microbiota in AD skin differs signifi- dence. In the Learn Early About Peanut (LEAP) study,
cantly from that of normal skin, particularly with its over 600 infants with severe AD and/or egg allergy were
overgrowth of Staphylococcus aureus [30]. Greater skin randomized to either consume or avoid peanuts until the
colonization with Staph. aureus has been associated age of 5; 17% of the children who avoided peanut devel-
with increased eczema severity and greater skin barrier oped peanut allergy by the age of 5 compared to only 3%
dysfunction as measured by transepidermal water loss of those who consumed peanut [48]. In the Enquire About
[31–33]. Skin dysbiosis and Staph. aureus colonization Tolerance (EAT) study, over 1300 exclusively breastfed
have also been shown to drive eczematous inflammation infants were randomly assigned at 3 months of age to the
in mouse models [34]. More recently, the commensal bac- early introduction of six allergenic foods (peanut, cooked
terium Staph. epidermidis has also been implicated in the egg, cow’s milk, sesame, cod and wheat) or exclusive
development of AD, as AD flares have been associated breastfeeding until 6 months of age; the rate of food

ATOPIC DERMATITIS
with an overabundance of both Staph. aureus and Staph. allergy to one or more of the intervention foods did not
epidermidis in addition to the decreased diversity of other differ between the two arms in the intention‐to‐treat

SECTION 3:
skin microflora [35]. Nevertheless, while these findings analysis but was significantly lower in the early introduc-
suggest an association between the skin microbiome and tion group (2.4%, vs. 7.3% in the standard group) in the
AD, it remains unknown whether dysbiosis of the skin per‐protocol analysis [49]. While these results support the
directly causes AD or simply reflects the impaired epider- notion that early introduction of allergenic foods may
mal barrier and immune dysregulation of AD. induce tolerance, the study found no effect on the risk of
AD development in relation to the length of exclusive
Nursing and infant feeding breastfeeding [49].
Breastfeeding is thought to play an important part in the
establishment of a normal postnatal gut microflora and The hygiene hypothesis
by many is also considered an important strategy to pre- AD is more common among children growing up in
vent AD development, in particular in children from smaller families and families of higher socioeconomic
high‐risk families. However, a meta‐analysis concluded status, and it has been suggested that this may be due to
that there was no convincing evidence for a protective a lower exposure to certain viral and bacterial pathogens
effect of exclusive breastfeeding for at least 3 months on [50]. In other words, allergic disease is thought to occur
AD [36]. For instance, a randomized controlled trial in when the developing immune system is deprived of the
Belarus failed to find any beneficial effect of prolonged or obligatory stimulation through certain microbial antigens
exclusive breastfeeding on the development of AD or [51], and the protective effect on AD development seen
other allergies. Even when comparing only studies that with daycare attendance during infancy, endotoxin expo-
looked at formula versus exclusive breastfeeding, the sure, consumption of unpasteurized farm milk and being
protective effect was negligible [36]. brought up with a dog during early life could all be
There is, however, some evidence from a large German mediated by nonpathological microbial stimulation of the
interventional birth cohort to suggest that certain cow’s infant’s immune system [52]. For instance, a systematic
milk hydrolysates can reduce AD risk in high‐risk children review and meta‐analysis found that farm exposure
[37], but a Cochrane review published prior to the German within the first year of life conferred a significant protec-
report had concluded that there was no such effect, having tive effect on allergic sensitization [53]. Immunologically,
consulted a large body of evidence [38]. There is also no this could be explained by a lack of stimulation of T‐cell‐
evidence to suggest that dietary antigen avoidance during mediated anti‐inflammatory cytokines, such as IL‐10 and
pregnancy or lactation in high‐risk children or soy formula transforming growth factor‐β [54,55]. However, there is
feeding alters the risk of AD [39,40]. Furthermore, there currently no clear epidemiological evidence to suggest
does not appear to be any benefit of an egg‐ and milk‐free that exposure to a specific infection reduces the risk of AD.
diet in unselected children with AD [41]. However, one In fact, there is good evidence from prospective studies
study has suggested that using an egg‐free diet in infants that some childhood infections, such as measles, are
with AD who also have elevated specific IgE levels to egg associated with an increased risk of AD development [52].
may lead to significant improvement in disease severity In addition, the decreased risk of AD seen with increased
[41]. (See Chapter 19 for further details.) number of siblings appears to persist even after adjust-
Two studies have suggested an increased risk for AD in ment for early childhood infections, suggesting that early
infants exposed to solid foods during the first few months postnatal or even prenatal factors play an important role
of life [42,43], but three other studies have found the in allergy development [56]. In this context, alteration of
opposite effect [44–46], i.e. that delayed introduction of the gut microflora, for instance secondary to frequent
solids is associated with AD development. Reverse causa- antibiotic use, has been suggested to programme AD
tion has been proposed as an explanation, but no evidence development [26]. However, the large majority of inter-
of parental allergy playing a role in feeding practices was vention studies with pre‐ and probiotics to prevent and
found [46]. There is also mounting evidence from animal treat AD have been disappointing [57,58], and neither can
research that early introduction of potentially allergenic currently be recommended for routine clinical practice.
foods might induce tolerance rather than allergy [47]. Further research with different probiotic strains is required.
178 Section 3  Atopic Dermatitis and Related Disorders

Irritants and washing serum IgE in AD, and in vitro research has shown that
Unlike allergic rhinitis, clinically relevant allergens are eczematous skin is rich in IgE‐bearing antigen‐presenting
seldom documented in AD, and irritants may be more dendritic cells, considered instrumental in the capture of
prominent in triggering skin inflammation [59]. Low environmental allergens [67]. Indeed, inhalation of house
humidity, excessive exposure to soaps and other primary dust mite allergen can provoke eczematous skin lesions in
household irritants, and possibly airborne pollution may predisposed patients, and IgE receptors can act as media-
be important by directly exacerbating skin disease, and tors in the clinical expression of AD following skin
indirectly by increasing vulnerability to sensitization, application of aeroallergens in the atopy patch test [68].
especially in children who are genetically predisposed A systematic review and meta‐analysis of birth cohort
to have an impaired skin barrier [59]. Excessive use of studies also identified a 2.7‐fold increased risk of infantile
chemical household products and frequent washing at eczema among children with food sensitization in the
15 months of age were associated with an increase in AD first 2 years of life [69]. Many therefore see allergic sensi-
at 30 –42 months of age in a birth cohort in the UK [60]. tization as an integral part and potential cause of AD in
ATOPIC DERMATITIS

However, the optimal bathing frequency for children children. However, despite detailed knowledge of the
with AD remains unknown. In a review of the literature, underlying immunological mechanisms in childhood
SECTION 3:

the studies favouring bathing at least once a day outnum- AD, the aetiological role of sensitization in childhood
bered those favouring bathing less than once a day, but eczema remains uncertain [70]. Indeed, a systematic
the evidence still remains limited [61]. More prospective review has shown that the strength of the association
data are also required to see how genetic and environ- between allergic sensitization and AD varies widely
mental factors interact. between studies [65]. Up to 50% of eczema sufferers in
hospital settings are not sensitized, and the proportion of
Pets sensitized individuals tends to be even lower in commu-
Although pets have often been blamed for causing AD nity settings, a finding that is partly explained by differ-
and other allergic diseases, a systematic review of cross‐ ences in disease severity between hospital and community
sectional and cohort studies failed to find any evidence to cases [65]. The strength of the association between allergic
suggest that contact with furry pets before or at birth sensitization and AD phenotype also varies between
increased the subsequent risk of AD. Indeed, another developing countries and industrialized nations.
systematic review and meta‐analysis of birth cohort Population‐based data collected from 31 000 8‐ to 12‐year‐
studies has even shown a protective effect of dogs and old children from 22 countries as part of the ISAAC has
pets overall on the risk of AD in infancy or childhood [62]. shown that even if the same rigid diagnostic criteria and
physical examination are employed across paediatric
Infection populations, the association between allergic sensitiza-
The dry skin and cytokine milieu characteristic of AD tion and flexural eczema is weaker in low‐ versus high‐
patients favours colonization by viruses, such as herpes income countries [71]. One explanation is that the
simplex virus [63], and Staph. aureus, an organism that is association between sensitization and AD in high‐income
implicated in maintaining cutaneous inflammation, as countries is primarily due to shared causes rather than
well as being capable of inciting allergic responses in such direct causality [71]. In addition, an Australian birth
individuals [30]. More than 90% of AD sufferers are colo- cohort study among at‐risk children found that allergic
nized with Staph. aureus compared to 5–30% of the normal sensitization at 18 months was not associated with eczema
population [30]. In addition to increased colonization, at 5 years of age [72]. However, in the same study, AD
about half of the Staph. aureus strains found in AD suffer- phenotype at 18 months was a significant predictor of
ers are toxin producing [30]. Despite this, interventions to skinprick test positivity at 5 years (adjusted OR = 1.67,
reduce Staph. aureus carriage as a treatment of AD have 95% CI [1.20–2.33]), suggesting that sensitization is a
not proven successful to date [64]. However, growing secondary rather than primary phenomenon in AD.
research on the skin microbiome suggests that dysbiosis This idea that allergic sensitization is a secondary phe-
of the skin beyond just increased Staph. aureus may be nomenon in AD is invoked in the concept of the ‘atopic
involved in the pathogenesis of AD. AD flares have been march’, which is characterized by the progression of AD
associated with a significant decrease in overall skin early in life to the subsequent development of asthma and
microbiota diversity as well as increased abundance of allergic rhinitis. It is posited that transcutaneous allergic
Staph. epidermidis [35]. Some studies have also suggested a sensitization occurs through the defective skin barrier in
possible role for Pityrosporum yeasts in inducing exacer- AD, thus invariably leading to the development of the
bations of AD through autoantibody formation [65,66]. other allergic disorders [73]. However, only one‐third of
AD patients develop asthma and two‐thirds develop
Allergic sensitization and the ‘atopic allergic rhinitis, and even among those patients who
march’ appear to follow the atopic march, it remains uncertain
There has been a lot of debate in the dermatological litera- whether the atopic march represents a causal relation-
ture as to whether allergic sensitization, i.e. skinprick test ship, even if atopic dermatitis, rhinitis and asthma often
positivity or elevation of specific IgE levels to common coexist in the same patient [73]. The atopic march
environmental allergens, is an essential feature of AD ­concept was originally based on cross‐sectional study
[65]. Early studies demonstrated very high levels of total evidence, but its existence has since been questioned as
Chapter 13  Epidemiology of Atopic Dermatitis 179

few prospective cohort studies have been conducted and and carbon monoxide, suggested that air pollution may
the longitudinal studies to date suggest significant heter- contribute to atopic eczema risk, but this association,
ogeneity among AD patients in the development of albeit statistically significant, was very weak with odds
asthma and rhinitis [70,74,75]. For instance, in a study ratios just above 1 (OR = 1.12, 95% CI 1.04–1.22 [81]).
of two UK population‐based birth cohorts, fewer than 7% Similarly, a cohort study among 3000 schoolchildren in
of children had progression of allergic symptoms follow- West Germany with repeat objective pollutant measure-
ing the typical atopic march pattern by the age of 11 [74]. ments suggested that NO2 exposure was positively asso-
Interestingly, of the children who did follow the typical ciated with physician‐diagnosed AD at the age of 6
atopic march, 71% had allergic sensitization on skinprick (OR = 1.18, 95% CI 1.00–1.39 [82]). Furthermore, a French
testing, in contrast to only 14% of children who had only cross‐sectional survey among more than 5000 schoolchil-
eczema without any asthma or rhinitis [74]. Some studies dren in six cities showed a stronger positive relationship
have shown that only children with AD plus sensitization with fine particle pollution (OR = 2.51, 95% CI 2.06–3.06
have a higher risk of allergic respiratory disease [76], but [83]). In a systematic review of birth cohort studies exam-

ATOPIC DERMATITIS
similar observations have also been made in those with ining traffic‐related air pollution and eczema, two of three
allergic sensitization alone [65,72]. These findings are cohorts found increased rates of eczema with exposure to

SECTION 3:
consistent with the idea that skin barrier impairment may NO2 and black carbon and living near a major road, while
be the key primary event that leads to allergic sensitiza- one cohort found no association [84]. Previous question-
tion across a ‘leaky’ epidermis, allowing contact between naire‐based studies from Sweden and East Germany
environmental allergens and antigen‐presenting cells in found that AD risk increased with living close to heavy
the epidermis. FLG loss‐of‐function mutations show traffic [85,86], but similar studies in West Germany, Malta,
much stronger associations with atopic compared to Russia and Japan did not confirm these findings [87–90].
nonatopic eczema, and only skin barrier gene mutation As studies have also shown that allergic sensitization is
carriage in the context of established AD is significantly enhanced in the presence of fine particle pollution [83],
associated with later asthma [4]. it is conceivable that allergic sensitization across the
Well‐conducted large prospective studies with clear epidermis is enhanced in the presence of certain outdoor
diagnostic criteria are needed to demonstrate whether pollutants, and it will be interesting to see whether future
eczema sufferers with allergic sensitization have a differ- studies can explain part of these contradicting results by
ent disease from those without positive skinprick tests differences in skin barrier impairment/skin barrier gene
or elevated specific IgE levels. Disease outcomes, such as status.
prognosis and response to treatment, with adjustment for Some indoor pollutants also appear to be risk factors
disease severity, duration, sex, skin barrier gene/phenotype for AD, including certain types of gas heaters and radia-
as well as early allergic respiratory disease, will be par- tors [91] and pre‐ and post‐natal reported exposure to
ticularly important to study. In addition, further research tobacco smoke [92–94]. One study found a positive
needs to address whether and how allergic respiratory correlation between maternal and cord blood nicotine
disease contributes to AD flares and chronicity. metabolite levels and AD risk [94]. Other indoor environ-
mental factors associated with AD risk are dampness and
Other environmental risk factors moulds [95–98], but all studies were questionnaire‐based
Experimental work with mice has suggested that and allergic sensitization to moulds not tested. More
exposure to atmospheric pollutants may enhance the IgE refined studies are therefore needed to examine the
immune response [77], and passive smoking has also effects of specific environmental pollutants in popula-
been linked with IgE responsiveness and increased allergic tions using standardized disease definitions. Another
airways disease [77]. It has been suggested that pollutants, potentially important risk factor is water hardness. Three
such as passive smoking, cause low‐grade airway inflam- cross‐sectional population‐based studies in the UK, Japan
mation, which enhances sensitization, and this is likely to and Spain have shown a positive association between
be one of the factors accounting for the increase in asthma questionnaire‐diagnosed AD and domestic water hard-
seen over the last 30 years [77]. A meta‐analysis of studies ness [99], but a randomized controlled trial of water
examining exposure to tobacco smoking and AD found softeners for treating established AD (the Softened Water
an association between AD and both active and passive Eczema Trial [SWET] study) did not find any additional
smoking, but not maternal exposure to tobacco smoke improvement in AD severity with the use of water soften-
during pregnancy [78]. Similarly, a meta‐analysis of stud- ers compared to usual care alone [100]. The negative find-
ies examining exposure to tobacco smoking and allergic ings from the SWET trial do not, however, exclude the
conditions has found an association between AD and possibility that domestic water hardness is involved in
both active and passive smoking [79]. Whether atmos- initiating eczematous skin inflammation in early life, and
pheric outdoor pollutants are important in increasing a strong positive association between calcium carbonate
vulnerability to sensitization in AD is less clear, and studies levels and eczema risk at 3 months has been found in a
that employ objective air pollution measures have more population‐based cohort of 1300 children from England
recently been conducted [80]. For instance, a population‐ and Wales, with a stronger effect seen in those carrying an
based cross‐sectional survey among more than 300 000 FLG mutation [101]. A prevention trial is currently under-
Taiwanese schoolchildren with objective measurement of way to test whether fitting a water softener around the
traffic‐related air pollutants, including nitrogen oxides time of birth is able to prevent eczema.
180 Section 3  Atopic Dermatitis and Related Disorders

There is also a need to study the potential interaction 22 Tsakok T, McKeever TM, Yeo L, Flohr C. Does early life exposure to
antibiotics increase the risk of eczema? A systematic review. Br J
between indoor and outdoor environmental and climatic
Dermatol 2013;169:983–91.
factors [102]. Other additional risk factors that warrant 23 Bjorksten B. The intrauterine and postnatal environments. J Allergy
further research are pesticide residues (such as organo- Clin Immunol 1999;104:1119–27.
phosphorus compounds, which are secreted in breast 24 Prescott SL, Macaubas C, Holt BJ et  al. Transplacental priming of
the human immune system to environmental allergens: universal
milk) [103,104], drinking water microbial content, increase skewing of initial T cell responses toward the Th2 cytokine profile.
in maternal age, use of oral contraceptives and dietary J Immunol 1998;160:4730–7.
changes such as a reduction in the consumption of free 25 Chan CW, Wong RS, Law PT et al. Environmental factors associated
radical scavengers, which might be important in maintain- with altered gut microbiota in children with eczema: a systematic
review. Int J Mol Sci 2016;17.
ing the homeostasis of inflammatory mediators [105–108]. 26 Bjorksten B, Sepp E, Julge K et  al. Allergy development and the
intestinal microflora during the first year of life. J Allergy Clin
References Immunol 2001;108:516–20.
1 Larsen FS, Holm NV, Henningsen K. Atopic dermatitis. A genetic‐ 27 Melli LC, do Carmo‐Rodrigues MS, Araujo‐Filho HB et al. Intestinal
ATOPIC DERMATITIS

epidemiologic study in a population‐based twin sample. J Am Acad microbiota and allergic diseases: A systematic review. Allergol
Dermatol 1986;15:487–94. Immunopathol (Madr) 2016;44:177–88.
28 Majamaa H, Isolauri E. Evaluation of the gut mucosal barrier: evi-
SECTION 3:

2 Elmose C, Thomsen SF. Twin studies of atopic dermatitis: interpretations


and applications in the filaggrin era. J Allergy (Cairo) 2015;2015:902359. dence for increased antigen transfer in children with atopic eczema.
3 Morar N, Willis‐Owen SA, Moffatt MF, Cookson WO. The genetics J Allergy Clin Immunol 1996;97:985–90.
of atopic dermatitis. J Allergy Clin Immunol 2006;118:24–34; quiz 5–6. 29 Belkaid Y, Segre JA. Dialogue between skin microbiota and immunity.
4 Baurecht H, Irvine AD, Novak N et al. Toward a major risk factor Science 2014;346:954–9.
for atopic eczema: meta‐analysis of filaggrin polymorphism data. 30 De Benedetto A, Agnihothri R, McGirt LY et al. Atopic dermatitis: a
J Allergy Clin Immunol 2007;120:1406–12. disease caused by innate immune defects? J Invest Dermatol 2009;
5 Palmer CN, Irvine AD, Terron‐Kwiatkowski A et al. Common loss‐ 129:14–30.
of‐function variants of the epidermal barrier protein filaggrin are 31 Gong JQ, Lin L, Lin T et  al. Skin colonization by Staphylococcus
a major predisposing factor for atopic dermatitis. Nat Genet aureus in patients with eczema and atopic dermatitis and relevant
2006;38:441–6. combined topical therapy: a double‐blind multicentre randomized
6 Paternoster L, Standl M, Waage J et al. Multi‐ancestry genome‐wide controlled trial. Br J Dermatol 2006;155:680–7.
association study of 21,000 cases and 95,000 controls identifies new 32 Jinnestal CL, Belfrage E, Back O et al. Skin barrier impairment corre-
risk loci for atopic dermatitis. Nat Genet 2015;47:1449–56. lates with cutaneous Staphylococcus aureus colonization and sensiti-
7 Ruiz RG, Kemeny DM, Price JF. Higher risk of infantile atopic derma- zation to skin‐associated microbial antigens in adult patients with
titis from maternal atopy than from paternal atopy. Clin Exp Allergy atopic dermatitis. Int J Dermatol 2014;53:27–33.
1992;22:762–6. 33 Baurecht H, Rühlemann MC, Rodríguez E et  al. Epidermal lipid
8 Andersson NW, Hansen MV, Larsen AD et al. Prenatal maternal stress composition, barrier integrity, and eczematous inflammation are
and atopic diseases in the child: a systematic review of observational associated with skin microbiome configuration. J Allergy Clin
human studies. Allergy 2016;71:15–26. Immunol 2018;141:1668–76.
9 Warner JO. The early life origins of asthma and related allergic disor- 34 Kobayashi T, Glatz M, Horiuchi K et al. Dysbiosis and Staphylococcus
ders. Arch Dis Child 2004;89:97–102. aureus colonization drives inflammation in atopic dermatitis.
10 Jones CA, Vance GH, Power LL et al. Costimulatory molecules in the Immunity 2015;42:756–66.
developing human gastrointestinal tract: a pathway for fetal allergen 35 Kong HH, Oh J, Deming C et al. Temporal shifts in the skin micro-
priming. J Allergy Clin Immunol 2001;108:235–41. biome associated with disease flares and treatment in children with
11 Casas R, Bjorksten B. Detection of Fel d 1‐immunoglobulin G immune atopic dermatitis. Genome Res 2012;22:850–9.
complexes in cord blood and sera from allergic and non‐allergic 36 Yang YW, Tsai CL, Lu CY. Exclusive breastfeeding and incident
mothers. Pediatr Allergy Immunol 2001;12:59–64. atopic dermatitis in childhood: a systematic review and meta‐analysis
12 Cooper PJ. The potential impact of early exposures to geohelminth of prospective cohort studies. Br J Dermatol 2009;161:373–83.
infections on the development of atopy. Clin Rev Allergy Immunol 37 von Berg A, Koletzko S, Filipiak‐Pittroff B et al. Certain hydrolyzed
2004;26:5–14. formulas reduce the incidence of atopic dermatitis but not that
13 Flohr C, Quinnell RJ, Britton J. Do helminth parasites protect against of  asthma: three‐year results of the German Infant Nutritional
atopy and allergic disease? Clin Exp Allergy 2009;39:20–32. Intervention Study. J Allergy Clin Immunol 2007;119:718–25.
14 Power LL, Popplewell EJ, Holloway JA et  al. Immunoregulatory 38 Osborn DA, Sinn J. Formulas containing hydrolysed protein for pre-
­molecules during pregnancy and at birth. J Reprod Immunol 2002; vention of allergy and food intolerance in infants. Cochrane Database
56:19–28. Syst Rev 2006(4):Cd003664.
15 Warner JA, Miles EA, Jones AC et al. Is deficiency of interferon gamma 39 Kramer MS, Kakuma R. Cochrane in context: Maternal dietary anti-
production by allergen triggered cord blood cells a predictor of atopic gen avoidance during pregnancy or lactation, or both, for preventing
eczema? Clin Exp Allergy 1994;24:423–30. or treating atopic disease in the child. Evid Based Child Health
16 Jenmalm MC, Bjorksten B. Cord blood levels of immunoglobulin G 2014;9:484–5.
subclass antibodies to food and inhalant allergens in relation to 40 Osborn DA, Sinn J. Soy formula for prevention of allergy and food
maternal atopy and the development of atopic disease during the first intolerance in infants. Cochrane Database Syst Rev 2006(4):Cd003741.
8 years of life. Clin Exp Allergy 2000;30:34–40. 41 Bath‐Hextall F, Delamere FM, Williams HC. Dietary exclusions
17 Sugiyama M, Arakawa H, Ozawa K et  al. Early‐life risk factors for for improving established atopic eczema in adults and children:
occurrence of atopic dermatitis during the first year. Pediatrics systematic review. Allergy 2009;64:258–64.
2007;119:e716–23. 42 Fergusson DM, Horwood LJ. Early solid food diet and eczema in
18 Zutavern A, von Klot S, Gehring U et  al. Pre‐natal and post‐natal childhood: a 10‐year longitudinal study. Pediatr Allergy Immunol
exposure to respiratory infection and atopic diseases development: a 1994;5(6 Suppl):44–7.
historical cohort study. Respir Res 2006;7:81. 43 Forsyth JS, Ogston SA, Clark A et al. Relation between early introduc-
19 McKeever TM, Lewis SA, Smith C, Hubbard R. The importance of tion of solid food to infants and their weight and illnesses during the
prenatal exposures on the development of allergic disease: a birth first two years of life. BMJ 1993;306:1572–6.
cohort study using the West Midlands General Practice Database. Am 44 Bergmann RL, Diepgen TL, Kuss O et al. Breastfeeding duration is a
J Respir Crit Care Med 2002;166:827–32. risk factor for atopic eczema. Clin Exp Allergy 2002;32:205–9.
20 Droste JH, Wieringa MH, Weyler JJ et al. Does the use of antibiotics in 45 Sears MR, Greene JM, Willan AR et  al. Long‐term relation between
early childhood increase the risk of asthma and allergic disease? Clin breastfeeding and development of atopy and asthma in children and
Exp Allergy 2000;30:1547–53. young adults: a longitudinal study. Lancet 2002;360:901–7.
21 Farooqi IS, Hopkin JM. Early childhood infection and atopic disorder. 46 Zutavern A, von Mutius E, Harris J et al. The introduction of solids in
Thorax 1998;53:927–32. relation to asthma and eczema. Arch Dis Child 2004;89:303–8.
Chapter 13  Epidemiology of Atopic Dermatitis 181

47 Meglio P, Bartone E, Plantamura M et al. A protocol for oral desen- 75 Illi S, von Mutius E, Lau S et al. The natural course of atopic dermatitis
sitization in children with IgE‐mediated cow’s milk allergy. Allergy from birth to age 7 years and the association with asthma. J Allergy
2004;59:980–7. Clin Immunol 2004;113:925–31.
48 Du Toit G, Roberts G, Sayre PH et  al. Randomized trial of peanut 76 Lowe AJ, Hosking CS, Bennett CM et al. Skin prick test can identify
­consumption in infants at risk for peanut allergy. N Engl J Med eczematous infants at risk of asthma and allergic rhinitis. Clin Exp
2015;372:803–13. Allergy 2007;37:1624–31.
49 Perkin MR, Logan K, Tseng A et al. Randomized trial of introduc- 77 Brunekreef B, Holgate ST. Air pollution and health. Lancet 2002;
tion of allergenic foods in breast‐fed infants. N Engl J Med 360:1233–42.
2016;374:1733–43. 78 Kantor R, Kim A, Thyssen JP, Silverberg JI. Association of atopic
50 Harris JM, Cullinan P, Williams HC et al. Environmental associations dermatitis with smoking: A systematic review and meta‐analysis.
with eczema in early life. Br J Dermatol 2001;144:795–802. J Am Acad Dermatol 2016;75:1119–25.e1.
51 Strachan DP. Hay fever, hygiene, and household size. BMJ 79 Saulyte J, Regueira C, Montes‐Martinez A et  al. Active or passive
1989;299:1259–60. exposure to tobacco smoking and allergic rhinitis, allergic dermatitis,
52 Flohr C, Pascoe D, Williams HC. Atopic dermatitis and the ’hygiene and food allergy in adults and children: a systematic review and
hypothesis’: too clean to be true? Br J Dermatol 2005;152:202–16. meta‐analysis. PLoS Med 2014;11:e1001611.
53 Campbell BE, Lodge CJ, Lowe AJ et  al. Exposure to ’farming’ and 80 Weiland SK, Husing A, Strachan DP et al. Climate and the prevalence

ATOPIC DERMATITIS
objective markers of atopy: a systematic review and meta‐analysis. of symptoms of asthma, allergic rhinitis, and atopic eczema in chil-
Clin Exp Allergy 2015;45:744–57. dren. Occup Environ Med 2004;61:609–15.
54 Bach JF. The effect of infections on susceptibility to autoimmune and 81 Lee YL, Su HJ, Sheu HM et  al. Traffic‐related air pollution, climate,

SECTION 3:
allergic diseases. N Engl J Med 2002;347:911–20. and prevalence of eczema in Taiwanese school children. J Invest
55 Yazdanbakhsh M, Kremsner PG, van Ree R. Allergy, parasites, and Dermatol 2008;128:2412–20.
the hygiene hypothesis. Science 2002;296:490–4. 82 Morgenstern V, Zutavern A, Cyrys J et  al. Atopic diseases, allergic
56 McKeever TM, Lewis SA, Smith C et al. Early exposure to infections sensitization, and exposure to traffic‐related air pollution in children.
and antibiotics and the incidence of allergic disease: a birth cohort Am J Respir Crit Care Med 2008;177:1331–7.
study with the West Midlands General Practice Research Database. 83 Annesi‐Maesano I, Moreau D, Caillaud D et al. Residential proximity
J Allergy Clin Immunol 2002;109:43–50. fine particles related to allergic sensitisation and asthma in primary
57 Lee J, Seto D, Bielory L. Meta‐analysis of clinical trials of probiotics for school children. Respir Med 2007;101:1721–9.
prevention and treatment of pediatric atopic dermatitis. J Allergy Clin 84 Bowatte G, Lodge C, Lowe AJ et  al. The influence of childhood
Immunol 2008;121:116–21.e11. traffic‐related air pollution exposure on asthma, allergy and sensi-
58 Osborn DA, Sinn JK. Prebiotics in infants for prevention of allergic tization: a systematic review and a meta‐analysis of birth cohort
disease and food hypersensitivity. Cochrane Database Syst Rev studies. Allergy 2015;70:245–56.
2007(4):Cd006474. 85 Montnemery P, Nihlen U, Goran Lofdahl C et al. Prevalence of self‐
59 Cork MJ, Robinson DA, Vasilopoulos Y et  al. New perspectives on reported eczema in relation to living environment, socio‐economic
epidermal barrier dysfunction in atopic dermatitis: gene‐environment status and respiratory symptoms assessed in a questionnaire study.
interactions. J Allergy Clin Immunol 2006;118:3–21; quiz 2–3. BMC Dermatol 2003;3:4.
60 Sherriff A, Golding J. Hygiene levels in a contemporary population 86 Schafer T, Vieluf D, Behrendt H et al. Atopic eczema and other mani-
cohort are associated with wheezing and atopic eczema in preschool festations of atopy: results of a study in East and West Germany.
infants. Arch Dis Child 2002;87:26–9. Allergy 1996;51:532–9.
61 Cardona ID, Stillman L, Jain N. Does bathing frequency matter in 87 Dotterud LK, Odland JO, Falk ES. Atopic diseases among schoolchil-
pediatric atopic dermatitis? Ann Allergy Asthma Immunol 2016;117: dren in Nikel, Russia, an Arctic area with heavy air pollution. Acta
9–13. Derm Venereol 2001;81:198–201.
62 Pelucchi C, Galeone C, Bach JF et al. Pet exposure and risk of atopic 88 Montefort S, Lenicker HM, Caruna S, Agius Muscat H. Asthma, rhinitis
dermatitis at the pediatric age: a meta‐analysis of birth cohort studies. and eczema in Maltese 13‐15 year‐old schoolchildren  –  prevalence,
J Allergy Clin Immunol 2013;132:616–22.e7. severity and associated factors [ISAAC]. International Study of Asthma
63 Hsu DY, Shinkai K, Silverberg JI. Epidemiology of eczema herpeti- and Allergies in Childhood. Clin Exp Allergy 1998;28:1089–99.
cum in hospitalized U.S. children: analysis of a nationwide cohort. 89 Weiland SK, Mundt KA, Ruckmann A, Keil U. Self‐reported wheezing
J Invest Dermatol 2018;138:265–72. and allergic rhinitis in children and traffic density on street of resi-
64 Birnie AJ, Bath–Hextall FJ, Ravenscroft JC, Williams HC. Interventions dence. Ann Epidemiol 1994;4:243–7.
to reduce Staphylococcus aureus in the management of atopic eczema. 90 Yura A, Shimizu T. Trends in the prevalence of atopic dermatitis in
Cochrane Database Syst Rev 2008(3):Cd003871. school children: longitudinal study in Osaka Prefecture, Japan, from
65 Flohr C, Johansson SG, Wahlgren CF, Williams H. How atopic is 1985 to 1997. Br J Dermatol 2001;145:966–73.
atopic dermatitis? J Allergy Clin Immunol 2004;114:150–8. 91 Schafer T, Heinrich J, Wjst M et  al. Indoor risk factors for atopic
66 Tang TS, Bieber T, Williams HC. Does “autoreactivity” play a role in eczema in school children from East Germany. Environ Res 1999;81:
atopic dermatitis? J Allergy Clin Immunol 2012;129:1209–15.e2. 151–8.
67 Bieber T. Atopic dermatitis. N Engl J Med 2008;358:1483–94. 92 Kramer U, Lemmen CH, Behrendt H et al. The effect of environmental
68 Ring J, Darsow U, Behrendt H. Role of aeroallergens in atopic eczema: tobacco smoke on eczema and allergic sensitization in children. Br J
proof of concept with the atopy patch test. J Am Acad Dermatol Dermatol 2004;150:111–18.
2001;45(1 Suppl):S49–52. 93 Schafer T, Dirschedl P, Kunz B et  al. Maternal smoking during
69 Alduraywish SA, Lodge CJ, Campbell B et al. The march from early pregnancy and lactation increases the risk for atopic eczema in the
life food sensitization to allergic disease: a systematic review and offspring. J Am Acad Dermatol 1997;36:550–6.
meta‐analyses of birth cohort studies. Allergy 2016;71:77–89. 94 Wang IJ, Hsieh WS, Wu KY et al. Effect of gestational smoke exposure
70 Williams H, Flohr C. How epidemiology has challenged 3 prevailing on atopic dermatitis in the offspring. Pediatr Allergy Immunol
concepts about atopic dermatitis. J Allergy Clin Immunol 2006;118: 2008;19:580–6.
209–13. 95 Lee YL, Li CW, Sung FC et al. Environmental factors, parental atopy
71 Flohr C, Weiland SK, Weinmayr G et al. The role of atopic sensiti- and atopic eczema in primary‐school children: a cross‐sectional study
zation in flexural eczema: findings from the International Study of in Taiwan. Br J Dermatol 2007;157:1217–24.
Asthma and Allergies in Childhood Phase Two. J Allergy Clin 96 Miyake Y, Ohya Y, Tanaka K et al. Home environment and suspected
Immunol 2008;121:141–7.e4. atopic eczema in Japanese infants: the Osaka Maternal and Child
72 Almqvist C, Li Q, Britton WJ et  al. Early predictors for developing Health Study. Pediatr Allergy Immunol 2007;18:425–32.
allergic disease and asthma: examining separate steps in the ’allergic 97 Wang IJ, Guo YL, Weng HJ et al. Environmental risk factors for early
march’. Clin Exp Allergy 2007;37:1296–302. infantile atopic dermatitis. Pediatr Allergy Immunol 2007;18:441–7.
73 Dharmage SC, Lowe AJ, Matheson MC et al. Atopic dermatitis and 98 Tsakok T, Weinmayr G, Jaensch A et al. Eczema and indoor environ-
the atopic march revisited. Allergy 2014;69:17–27. ment: lessons from the International Study of Asthma and Allergies in
74 Belgrave DC, Granell R, Simpson A et al. Developmental profiles of Childhood (ISAAC) Phase 2. Lancet 2015;385(suppl 1):S99.
eczema, wheeze, and rhinitis: two population‐based birth cohort 99 McNally NJ, Williams HC, Phillips DR et  al. Atopic eczema and
studies. PLoS Med 2014;11:e1001748. domestic water hardness. Lancet 1998;352:527–31.
182 Section 3  Atopic Dermatitis and Related Disorders

100 Thomas KS, Dean T, O’Leary C et al. A randomised controlled trial of be helpful in formula‐fed babies [10,11]. There is no con-
ion‐exchange water softeners for the treatment of eczema in chil-
sistent evidence for a protective effect with the use of
dren. PLoS Med 2011;8:e1000395.
101 Perkin MR, Craven J, Logan K et al. Association between domestic hydrolysed formula in high‐risk babies [12,13], although
water hardness, chlorine, and atopic dermatitis risk in early life: some studies suggest a protective effect [14]. Another area
A population‐based cross‐sectional study. J Allergy Clin Immunol that warrants further investigation is whether prevention
2016;138:509–16.
102 Langan SM, Silcocks P, Williams HC. What causes flares of eczema in
of skin barrier breakdown in genetically predisposed
children? Br J Dermatol 2009;161:640–6. infants, for instance with intensive emollient therapy, can
103 Karmaus W, Kuehr J, Kruse H. Infections and atopic disorders in reduce the risk of AD development. A small pilot trial in
childhood and organochlorine exposure. Arch Environ Health UK and US high‐risk neonates (n = 124) suggested that
2001;56:485–92.
104 Reichrtova E, Ciznar P, Prachar V et al. Cord serum immunoglobulin early emollient use might be beneficial to prevent the
E related to the environmental contamination of human placentas onset of eczema [12,15]. A large trial is currently recruit-
with organochlorine compounds. Environ Health Perspect 1999; ing in the UK with plans to follow children for 5 years to
107:895–9.
test the effectiveness of early emollient use in the preven-
ATOPIC DERMATITIS

105 Peters TJ, Golding J. The epidemiology of childhood eczema: II.


Statistical analyses to identify independent early predictors. Paediatr tion of eczema. As for secondary prevention, the use of
Perinat Epidemiol 1987;1:80–94. water softeners was tested in a UK‐wide trial in children
SECTION 3:

106 Seaton A, Godden DJ, Brown K. Increase in asthma: a more toxic with moderate to severe eczema but did not show a ben-
environment or a more susceptible population? Thorax 1994;49:
171–4. efit over usual care [16] (see also Other environmental
107 von Hertzen L, Laatikainen T, Pitkanen T et al. Microbial content of risk factors).
drinking water in Finnish and Russian Karelia  –  implications for All such studies need to assess not only the short‐term
atopy prevalence. Allergy 2007;62:288–92.
effect on disease severity but whether such interventions
108 Xu B, Jarvelin MR, Pekkanen J. Prenatal factors and occurrence of
rhinitis and eczema among offspring. Allergy 1999;54:829–36. can alter the natural history of AD. In such a chronic and
intermittent disease, longer‐term studies looking at
disease‐free periods or time series analysis are needed.
Prevention of atopic dermatitis
References
Given that factors operating early in utero might be critical 1 Schafer T, Borowski C, Reese I et al. Systematic review and evidence‐
in determining subsequent AD, manipulation of the early based consensus guideline on prevention of allergy and atopic
environment offers a promising way forward to prevent eczema of the German Network on Allergy Prevention (ABAP).
Minerva Pediatr 2008;60:313–25.
AD if the risk factors responsible can be identified and are
2 Corver K, Kerkhof M, Brussee JE et  al. House dust mite allergen
amenable to public health manipulation. Pregnant mothers reduction and allergy at 4 yr: follow up of the PIAMA‐study. Pediatr
with a family history of atopic disease are usually only Allergy Immunol 2006;17:329–36.
too willing to comply with even quite elaborate interven- 3 Custovic A, Simpson BM, Simpson A et  al. Effect of environmental
manipulation in pregnancy and early life on respiratory symptoms
tions. Some have even gone as far as producing national and atopy during first year of life: a randomised trial. Lancet
guidelines for prevention of AD [1]. Unfortunately, while 2001;358:188–93.
earlier, less rigidly controlled studies seemed promising, 4 Horak F Jr, Matthews S, Ihorst G et al; SPACE study group. Effect of
seven randomized intervention studies have failed to mite‐impermeable mattress encasings and an educational package
on the development of allergies in a multinational randomized,
show a reduction in AD risk and severity with house dust controlled birth‐cohort study  –  24 months results of the Study of
mite avoidance [2–6]. Given that allergic sensitization is Prevention of Allergy in Children in Europe. Clin Exp Allergy
likely to be a secondary phenomenon in AD, this may not 2004;34:1220–5.
5 Koopman LP, van Strien RT, Kerkhof M et al. Placebo‐controlled trial
be entirely surprising. Indeed, allergen avoidance may of house dust mite‐impermeable mattress covers: effect on symptoms
after all not be the right approach, as supported by a in early childhood. Am J Respir Crit Care Med 2002;166:307–13.
Cochrane systematic review of dietary avoidance during 6 Oosting AJ, de Bruin‐Weller MS, Terreehorst I et al. Effect of mattress
pregnancy. A completed intervention birth cohort study encasings on atopic dermatitis outcome measures in a double‐blind,
placebo‐controlled study: the Dutch mite avoidance study. J Allergy
looked at whether the early introduction of allergenic Clin Immunol 2002;110:500–6.
food protein could reduce AD and food allergy risk (see 7 Perkin MR, Logan K, Tseng A et  al. Randomized trial of intro-
Nursing and infant feeding) [7]. The published findings duction of allergenic foods in breast‐fed infants. N Engl J Med
2016;374:1733–43.
to date show no significant benefits in terms of food
8 Cuello‐Garcia CA, Brozek JL, Fiocchi A et al. Probiotics for the pre-
allergy for early allergen exposure in the intention‐to‐ vention of allergy: A systematic review and meta‐analysis of rand-
treat group, but a significantly lower prevalence of food omized controlled trials. J Allergy Clin Immunol 2015;136:952–61.
allergy in the per protocol analysis; the effects on eczema 9 Zuccotti G, Meneghin F, Aceti A et al. Probiotics for prevention of
atopic diseases in infants: systematic review and meta‐analysis.
are eagerly awaited. Other dietary interventions that have Allergy 2015;70:1356–71.
been extensively studied include interventions to manip- 10 Chang YS, Trivedi MK, Jha A et  al. Synbiotics for prevention and
ulate the maternal diet during pregnancy and infant diet treatment of atopic dermatitis: a meta‐analysis of randomized clinical
in early life. Of such interventions, the most promising is trials. JAMA Pediatr 2016;170:236–42.
11 Gunaratne AW, Makrides M, Collins CT. Maternal prenatal and/
probiotic exposure, which appears to be associated with a or postnatal n‐3 long chain polyunsaturated fatty acids (LCPUFA)
reduction in eczema, although further research is required supplementation for preventing allergies in early childhood.
addressing timing and formulations [8,9]. There is cur- Cochrane Database Syst Rev 2015(7):Cd010085.
12 Foisy M, Boyle RJ, Chalmers JR et  al. Overview of Reviews The
rently limited evidence to support the use of prebiotics, prevention of eczema in infants and children: an overview of
synbiotics and omega‐3 long‐chain polyunsaturated fatty Cochrane and non‐Cochrane reviews. Evid Based Child Health
acids although there are suggestions that prebiotics may 2011;6:1322–39.
Chapter 13  Epidemiology of Atopic Dermatitis 183

13 Boyle RJ, Ierodiakonou D, Khan T et al. Hydrolysed formula and epidemiology and long‐term implications are of great
risk of allergic or autoimmune disease: systematic review and meta‐
interest. As AD is predominantly a disease of early onset,
analysis. BMJ 2016;352:i974.
14 von Berg A, Filipiak‐Pittroff B, Schulz H et al. Allergic manifestation birth cohort studies are not prohibitively expensive to set
15 years after early intervention with hydrolyzed formulas—the GINI up and current work is attempting to combine genetic,
Study. Allergy 2016;71:210–19. immunological and classic epidemiological tools to further
15 Simpson EL, Chalmers JR, Hanifin JM et  al. Emollient enhance-
ment of the skin barrier from birth offers effective atopic dermatitis
our understanding of how genetically predetermined skin
prevention. J Allergy Clin Immunol 2014;134:818–23. barrier impairment and environmental risk factors interact
16 Thomas KS, Sach TH. A multicentre randomized controlled trial of to produce the phenotype of AD. We need to understand
ion‐exchange water softeners for the treatment of eczema in chil- better where exactly allergic sensitization comes into this
dren: protocol for the Softened Water Eczema Trial (SWET)
(ISRCTN: 71423189). Br J Dermatol 2008;159:561–6. equation, and it would also be important to know whether
there is a distinct AD phenotype with an underlying immu-
nological defect in the skin and whether its clinical presen-
Conclusion tation differs from AD with a primary skin barrier defect.

ATOPIC DERMATITIS
Over the past few years, the field of AD epidemiology has Understanding these relationships in turn promises new
taken great leaps forward. The information we have opportunities for more intervention studies, which should

SECTION 3:
suggests that environmental factors seem to be critical in not only assess the effect on disease prevention but also
disease expression, which is good news for epidemiolo- evaluate the cost‐effectiveness of preventing disease and
gists as it makes the possibility of disease prevention a reducing the frequency of disease exacerbations.
reality. In addition, the discovery of the filaggrin loss‐
of‐function mutations has rekindled interest in the role of Acknowledgement
skin barrier impairment in the development of AD. Exciting Prof. Hywel Williams cowrote the previous edition of this
new developments in the field of eczema prevention with chapter. We are most grateful for his permission to reuse
barrier enhancement have the potential to transform AD some of this material.
184 

CHA PTER 1 4

Genetics and Aetiology of Atopic Dermatitis


Elke Rodriguez & Stephan Weidinger
Department of Dermatology, Allergology and Venereology, University Hospital Schleswig‐Holstein, Campus Kiel, Kiel, Germany

Introduction, 184 Conclusions and future directions, 190


Genes implicated in the aetiology of eczema, 187
ATOPIC DERMATITIS

Abstract by approximately 20% of patients, and they are neither necessary


SECTION 3:

nor sufficient to cause the disease. Most of the other loci harbour
candidate genes with functions related to immune regulation, in
Atopic dermatitis or eczema represents a typical multifactorial dis-
particular innate signalling and T‐cell specification and activation,
ease in that there is an individual hereditary predisposition that is
but the causative genes or gene products as well as the underlying
triggered by environmental and lifestyle factors. The heritability
molecular mechanisms remain to be identified and characterized.
of atopic dermatitis, i.e. the genetically determined proportion of
Together, the established susceptibility loci explain about 20% of
the total variance observed in a population, is estimated to be 70–
the total estimated heritability. This ‘missing heritability’ may be
80%. The possibilities for deciphering inherited risk factors have
explained by gene interactions, rare gene variants not yet discov-
impressively increased during the last years. As a result, more than
ered and epigenetic changes, but may also relate to an overestima-
30 susceptibility loci have been successfully identified. Null vari-
tion of the heritable disease component and the phenotypic com-
ants in the gene encoding the key epidermal barrier protein filag-
plexity of the disease.
grin are the strongest single risk factor, but they are carried only

Key points • Two notable exceptions are loss‐of‐function variants in the gene
FLG, which cause the dry skin condition ichthyosis vulgaris due
to a deficiency of the epidermal structural protein filaggrin, and
• Atopic dermatitis has a very strong heritable component.
infrequent variants in the gene encoding GARP, which lower the
• Genetic risk loci for atopic dermatitis are being discovered
expression of this receptor for latent transforming growth factor
through a range of strategies from linkage studies to genome‐
(TGF)‐β on the surface of regulatory T cells.
wide association studies.
• Known risk loci explain about 20% of the total estimated
• Thus far, more than 30 susceptibility loci for atopic dermatitis
heritability of atopic dermatitis.
have been identified. For most of these regions the causative
genes or gene products remain to be identified and characterized.

been suggested that eczema is part of a syndrome of


Introduction ‘atopic diseases’, in which patients may develop food
Challenges in defining (atopic) dermatitis/ allergy, eczema, asthma and rhinitis in any order and in
eczema any combination over time [4]. Atopy itself is defined as
Eczema (atopic dermatitis, atopic eczema) is the most ‘personal or familial tendency to become sensitized and
common chronic skin disease in infants and children, produce IgE antibodies in response to low doses of
with prevalence rates of up to 30%. It is estimated that allergens, usually proteins’ and to ‘develop typical
approximately 60% of patients with childhood eczema symptoms such as asthma, rhinoconjunctivitis, or
undergo spontaneous remission in early adolescence, but eczema/dermatitis’ [5]. Based on the fact that eczema
up to 50% may have recurrences in adulthood, and the symptoms often precede those of asthma and because
disease can persist into or start in adulthood, making it eczema represents a well‐established risk factor for
one of the most common skin disorders across all ages [1]. asthma, it has been further hypothesized that a suscepti­
Eczema, asthma and rhinitis tend to cluster in the ble child commonly passes through a sequential/over­
same individuals and families [2], and are often accom­ lapping series of phenotypes from eczema and food
panied by elevated levels of total serum IgE antibodies allergy to asthma and subsequently allergic rhinitis
and aberrant IgE‐mediated responses to otherwise (‘atopic march”) [6–9]. While allergic sensitization
harmless environmental agents [3]. Therefore it has ­represents one possible and plausible mechanistic link

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 14  Genetics and Aetiology of Atopic Dermatitis 185

between asthma and eczema, it is notable that a signifi­ Evidence for a genetic basis for eczema
cant proportion of patients with eczema are not ‘atopic’, There is a longstanding recognition that atopic diseases
i.e. they have normal total serum IgE concentrations and cluster in families and are hereditary disorders [18–20].
no specific IgE responses [3,10]. Furthermore, epidemi­ Twin studies showed distinctly higher concordance rates
ological research indicates that sensitization might sim­ among monozygotic than dizygotic twin pairs (for
ply be a shared epiphenomenon and suggests that the eczema 0.23–0.86 vs. 0.15–0.50), and segregation analyses
association between asthma and eczema may occur suggested that genetic factors account for more than 70%
much earlier, i.e. early co‐occurrence of eczema and of the variance in the susceptibility to eczema [21–24].
wheeze progressing to asthma [10–12]. Thus, the role However, unlike monogenic disorders, which are caused
and temporal significance of elevated IgE in eczema is by mutations in a single gene, eczema and atopic disor­
as yet unclear, and the concept of an ‘allergic march’ ders are likely to be complex poly‐ or oligogenetic traits,
may represent an oversimplification of complex disease thought to be the result of a complicated network of
co‐associations. Other discovered mechanisms such as numerous susceptibility loci that exert additive or syner­

ATOPIC DERMATITIS
epidermally produced thymic stromal lymphopoietin gistic effects but may have only a small role when considered
(TSLP)‐mediated lung inflammation may provide in isolation [25]. In addition, interacting environmental

SECTION 3:
­alternative explanations for an early co‐association of factors are thought to precipitate the polygenic risk back­
eczema and asthma [13,14]. ground into disease manifestation [26]. The importance
Eczema has a wide spectrum of clinical presentations of  environmental influences is emphasized by the wide
and it is unclear whether it is a single disorder with dif­ ranges in concordance rates, the incomplete concordance
ferent clinical manifestations or a group of syndromes among monozygotic twins and the changing and variable
with unique or overlapping pathophysiological path­ prevalences of atopic symptoms both between countries
ways that open out into a rather uniform clinical pres­ with similar ethnic groups and within countries [27,28].
entation. However, a rigorous definition of patient The dissection of complex traits is further hampered by
subgroups is highly desirable in order to facilitate epi­ phenocopy, incomplete penetrance and genetic heterogeneity
demiological, genetic and clinical investigations. In the [26]. In addition, a set of studies suggested that, particu­
absence of a gold standard or adequate laboratory tests larly in atopic disorders such as eczema, inheritance of
to diagnose eczema, numerous lists of diagnostic crite­ certain polymorphisms from the mother is more likely to
ria have been developed in order to establish a defini­ be associated with allergic diseases in the child than
tion. At present, the UK diagnostic criteria have been inheritance from the father [29]. This parent‐of‐origin
the most widely validated and appear to be applicable effect is supported by epidemiological studies that indi­
and repeatable across all ages and many ethnicities [15]. cate a crucial role of environmental exposures during
However, the ideal set of diagnostic criteria still has to pregnancy [30] and a more close relationship of the
be established [16]. infant’s disease risk to maternal than paternal disease sta­
The confusing terminology for atopic diseases, with tus [31–33]. Genomic imprinting is one important epige­
terms such as atopic eczema, atopic dermatitis, childhood netic mechanism that might explain such parent‐of‐origin
eczema, atopiform dermatitis and flexural dermatitis fre­ effects. Imprinting is a phenomenon in which disease‐
quently used synonymously in the literature, reflects the predisposing alleles only show their effects in a particular
complexity of the diseases and the as yet insufficient epigenetic context, for example methylation induced by
knowledge on their pathophysiology. It has previously the parental origin of the variant, leading to mono‐allelic
been suggested that ‘eczema’ be defined as the disease for­ expression in the somatic cells of the offspring. Differential
merly called ‘atopic eczema’ or ‘atopic dermatitis’, expression can occur in all cells, or in specific tissues or
whereas the term ‘atopic eczema’ be reserved for those developmental stages [34].
patients with eczema and evidence for IgE involvement
[5,17]. However, as pointed out in a review, this division Methods used in the genetic dissection
might not adequately reflect the natural history of this dis­ of eczema
ease [1], and it has to be considered that so far most, if not For elucidation of the genetic background of complex dis­
all, studies on the genetics of eczema were performed eases, genetic association studies and linkage analyses
prior to these suggestions. Also, most existing DNA collec­ play an important role. The DNA sequence of human
tions have been assembled using older definitions. beings is on average 99.9% identical. The remaining 0.1%
It is anticipated that difficulties in defining eczema and of DNA sequence differences between any two individu­
its phenotypes may be overcome or at least improved by als are mainly based on common single nucleotide poly­
studying the genetics, and that the results of molecular morphisms (SNPs), in which a single nucleotide within
studies will be of great nosological significance, enabling the DNA sequence is replaced with an alternative one,
a classification of atopic disorders based on the underly­ and where each variation is present to some appreciable
ing genetic effects rather than on hypothetical concepts degree within a population (e.g. >1%). SNPs have become
and clinical symptoms as is the case today. A similar a favourite genetic tool in the investigation of complex
development has already taken place in other medical diseases, although many are nonfunctional and therefore
fields such as in neurodegenerative diseases, many of result in neutral phenotypic outcomes. However, func­
which are now defined by their mutated genes (e.g. spi­ tional SNPs can predispose individuals to disease, or
nocerebellar ataxias). influence its severity, progression or individual response
186 Section 3  Atopic Dermatitis and Related Disorders

to medicine. At the molecular level, these SNPs can affect inadequate assessment of the trait of interest and
the human phenotype by interfering on both levels of the i­nappropriate controls, variable study designs, inap­
protein synthesis machinery: noncoding SNPs may dis­ propriate statistical modelling and failure to correct
rupt transcription factor binding sites, splice sites and for multiple comparisons, genetic and environmental
other regulatory elements on the transcriptional level, heterogeneity, publication bias (more positive reports
whereas coding SNPs can cause an amino acid change are submitted to and accepted by journals) and lack of
and alter the functional or structural properties of the independent replication [26,41].
translated protein. Since the turn of the millennium, association studies
Whole genome linkage analysis aims at the identifica­ have been revolutionized through newly developed
tion of chromosomal regions associated with a disease high‐throughput SNP genotyping platforms and
by identifying genetic markers, e.g. microsatellites or knowledge gained from the HapMap project, which
SNPs, co‐segregating with the disease within families. showed that the human genome is organized into
Susceptibility regions identified in such hypothesis‐free blocks of haplotypes with limited diversity within each
ATOPIC DERMATITIS

screens typically encompass several megabases of DNA of these blocks [42]. Association studies aim at the
sequence and might contain hundreds of genes. Thus, detection of proxy variants, which are in linkage dise­
SECTION 3:

intensive follow‐up analyses are needed to narrow down quilibrium (LD) with the causal variant. LD refers to
the region of interest and to determine single disease the nonindependent inheritance of alleles, e.g. SNPs, at
genes. While this traditional approach has been extremely two or more loci, which extend over relatively short
successful in the identification of monogenic disease distances and may contain either a single gene or a few
genes [35], it has only limited power to detect risk genes genes only [38]. As a consequence of this genomic
for complex traits due to epistasis, incomplete penetrance, architecture a limited set of SNPs can capture the vast
polygeneity or phenotypic heterogeneity. Very few link­ majority of common variations and ‘tag’ the haplotype
age approaches have led to the identification of causal pattern sufficiently.
disease variants in this setting. For eczema several link­ Since 2005, commercial SNP‐typing arrays have been
age studies have identified various putative disease loci available and allow the simultaneous investigation of
in the past [36]. However, with the exception of the filag­ up  to 4 million SNPs. The markers used are selected to
grin (FLG) gene, which partly explains the linkage signal provide maximal coverage of all common variations via
observed on chromosome 1q21 [37], it has not been pos­ LD‐based tagging, or even spacing within the genome
sible to assign a disease gene to any of the linkage regions [43]. These hypothesis‐free genome‐wide association
identified so far. studies (GWAS) were successfully used to identify com­
Association studies are thought to provide a more mon disease‐associated variants (defined as those present
powerful method for detecting complex disease alleles, in more than 5% of the population). Another useful source
in particular alleles that only confer a modest disease for genetic association studies was provided by the
risk. These studies statistically analyse the correlation 1000 Genomes project (http://www.internationalgenome.
between a genetic marker, usually an SNP, and the dis­ org), which between 2008 and 2015 sequenced the
ease in unrelated individuals (case‐control and case‐ genomes of more than 2500 individuals from 26 different
cohort studies) or their transmission in families populations, thereby providing a comprehensive data set
(family‐based design) [38]. An association between the of common as well as low‐frequency variants.
genotype and the phenotype is assumed if the genetic Technical improvements in recent years have also led
marker and the disease occur together more often than to a substantial decrease in costs of next‐generation
expected by chance. Due to technical limitations, sequencing technologies. In contrast to genetic associa­
genetic association studies have been limited to the tion studies that analyse a limited number of prese­
study of candidate genes, i.e. genes located in a region lected single nucleotide variants distributed over the
showing linkage in previous screening studies and/or genome, DNA sequencing provides information on
because of their function and expression patterns each single base constituting an individual human DNA
(positional and/or functional candidate genes) [39]. sequence. Whereas whole genome sequencing still is
Based on the assumption that the primary defect in rather costly, whole exome sequencing approaches,
eczema is immunological and prior to the discovery of which exclusively target the coding DNA regions of the
FLG (see Filaggrin), for many years candidate genes human genome, already allow the identification of as
involved in antigen presentation and cell‐mediated/ yet unknown rare functional variants within larger
humoral immune response, and cell signalling/cellular numbers of samples at reasonable costs. This approach
movement have been investigated, as well as genes has already been very successfully applied within fami­
that had been associated with asthma and/or atopy. lies affected by rare monogenic diseases, and will
More than 100 associated risk genes for eczema were undoubtedly increase our knowledge of genetic risk
reported up to 2009 by candidate gene association factors for common complex diseases like eczema.
studies [40], but most of these studies lack stringent Due to genetic studies the aetiological concept of
replication and have to be interpreted with caution. eczema underwent a profound change within recent
Potential reasons for irreproducibility of results years from a primarily immune‐mediated disorder to one
include low power (as a result of small sample sizes), that is equally, or may even be primarily, based on a skin
inappropriate selection of candidate loci and markers, barrier malfunction.
Chapter 14  Genetics and Aetiology of Atopic Dermatitis 187

Genes implicated in the aetiology highly specific for eczema [48,54–56]. Further, for most of
of eczema these loci the causative gene or gene product as well as
the underlying molecular mechanisms remain to be
Association studies, in particular those using genome‐ identified and characterized. Some notable exceptions are
wide or targeted high‐throughput approaches, have presented here.
robustly identified more than 30 susceptibility regions for
eczema (Table  14.1) [44–52]. These loci together explain
approximately 20% of the estimated heritability. Most of Filaggrin
them harbour candidate genes that contribute to immune Filaggrin (filament aggregating protein; FLG) was first
mechanisms, in particular to innate immune signalling described in 1977 as an insoluble protein purified from rat
and T‐cell activation and specification (Table  14.1, epidermis that interacted with intermediate filaments in
Fig.  14.1), and appear to impact the susceptibility for a vitro [57]. It represents a structural protein with key func­
range of immune‐mediated diseases rather than being tions in the formation and maintenance of the cornified

ATOPIC DERMATITIS
SECTION 3:
Table 14.1  Susceptibility loci and most plausible candidate genes identified through genome‐wide association studies for atopic eczema

Associated Candidate gene Known/proposed function


locus

1q21.2 CIART Circadian transcriptional repressor


1q21.3 FLG Terminal epidermal differentiation; aggregation of keratin intermediate filaments
1q21.3 IL6R Subunit of the IL‐6 receptor; differentiation of multiple immune cells, especially B cells
2p13.3 CD207 C‐type lectin with mannose binding specificity; major receptor on Langerhans cells; antigen
uptake, processing and presentation
2p16.1 PUS10 Posttranscriptional modification of structural RNAs; involved in apoptosis
2p25.1 LINC00299 Long noncoding RNA; no known function
2q12.1 IL1RL1 Subunit of the IL‐33 receptor; Th cell function
IL18R1/IL18RAP Subunits of the IL‐18 receptor; regulation of inflammatory response via NF‐κB
2q24.3 XIRP2 Actin cytoskeleton and cell–cell junction organization; protection of actin filaments from
depolymerization
3p21.1 RFT1 Oligosaccharide transporter; protein N‐glycosylation
3p22.3 CCR4 C‐C chemokine receptor; leucocyte trafficking
3q13.2 CCDC80 Cell adhesion and matrix assembly
4q27 IL2/IL21 Differentiation, proliferation and activation of multiple immune cells (T cells, B cells,
macrophages, natural killer cells)
5p13.2 IL7R Subunit of the IL‐7 and TSLP receptor; proliferation of lymphoid progenitors; release of T‐cell‐
attracting chemokines; promotion of Th2‐cell response
5q22.1 TSLP Release of T‐cell‐attracting chemokines; promotion of Th2‐cell response; AMP activity in the oral
cavity and on the skin
5q31.1 IL13/IL4 Mainly produced by activated Th2 cells; B‐cell proliferation; IgE isotype switching
6p21.32 HLA‐DRB Antigen processing and presentation MHC class II
6p21.33 MICB Stress‐induced self‐antigen; activation of cytolytic response of NK cells, αβ T cells, and γδ T cells
7p22.2 CARD11 TCR‐mediated T cell activation; NF‐κB activation
8q21.13 ZBTB10 Transcription regulation
9p21.3 DMRTA1 Transcription regulation
10p15.1 IL15RA/IL2RA IL‐15 and IL‐2 receptor subunits; proliferation and stimulation of different lymphocytes
10q21.2 ZNF365 Mitotic cytokinesis; maintenance of genome stability
11p13‐12 PRR5L Regulation of protein kinase C phosphorylation; survival and organization of the cytoskeleton;
cell migration
11p15.4 NLRP10 Regulation of the innate immune system; pro‐inflammatory cytokine release; inhibition of
apoptosis; anti‐inflammatory activity
11q13.1 OVOL1 Transcription factor; hair formation and spermatogenesis
11q13.5 LRRC32 Regulation of Treg function and TGF‐β activation
11q24.3 ETS1 Transcription factor; direct control of cytokine and chemokine expression
14q13.2 PPP2R3C Regulation of protein phosphatases; activation‐induced cell death of B cells
16p13.13 CLEC16A C‐type lectin; regulation of mitophagy/autophagy and mitochondrial health
17q21.2 STAT3 Signal transducer and transcription activator; mediation of cellular responses to interleukins
17q21.32‐33 ZNF652 Transcription regulation
19p13.2 ACTL9 Cytoskeletal functions
ADAMTS10 Metalloprotease; assembly of extracellular matrix components
20q13.2 CYP24A1 Vitamin D metabolism
20q13.33 TNFRSF6B TNF receptor superfamily; protection against apoptosis

Sorted by chromosome; AMP, antimicrobial peptide; IL, interleukin; MHC, major histocompatibility complex; NF‐κB, nuclear factor ‘kappa‐light‐chain‐enhancer’
of activated B cells; NK cells, natural killer cells; RNA, ribonucleic acid; TCR, T‐cell receptor; TGF‐β, transforming growth factor β; Th cell, T‐helper cell; TNF,
tumour necrosis factor; TSLP, thymic stromal lymphopoietin.
188 Section 3  Atopic Dermatitis and Related Disorders

Epidermal barrier Environmental sensing


FLG, OVOL1, CARD11,
ACTL9/ADAMTS10 HLA-DRB, MICB
ATOPIC DERMATITIS
SECTION 3:

Tissue response
Immune regulation CCDC80, CYP24A1,
IL1RL1/IL18R1/IL18RAP LRRC32, NLRP10, PRR5L,
IL2/IL21, IL4/IL13, TNFRSF6B,
IL6R, CLEC16A ZNF365, ZNF652
Fig. 14.1  Functional overview for selected candidate
genes identified through genome‐wide association
studies. Source: Adapted from Weidinger and Novak
(2016) [53].

envelope within the outermost layer of the human reduced in the epidermis of individuals with this disease
epidermis, and is essential for the prevention of penetra­ [63,64]. However, in‐depth analysis of the protein‐encoding
tion of environmental agents such as microbes or allergens gene transpired to be technically difficult because of its
into the organism and also, on the other hand, for controlling repetitive structural nature, and the sequence of the entire
transepidermal water loss (TEWL) [58,59]. FLG gene was finally published in 2006, identifying two
Profilaggrin is expressed as a large inactive precursor loss‐of‐function variants (p.R501X and c.2282del4) to be
protein of 400 kDa in the stratum granulosum of the epi­ the cause of IV [65]. Subsequently the same group
dermis, where it represents the main constituent of the detected significant associations between these variants
keratohyalin F granules (Fig.  14.2). It is composed of and eczema [66].
10–12 tandem repeats, each encoding a functionally active Since that time more than 50 recurrent and family‐
FLG monomer. Upon cornification of keratinocytes, pro­ specific variants have been identified within the FLG
filaggrin is dephosphorylated and proteolytically cleaved gene in European and Asian populations [67]. These are
into its active subunits. These monomers lead to a dense present in up to 10% of the general European population,
bundling of keratin intermediate filaments, leading to and up to 40% of eczema patients are carriers of at least
collapse of the keratinocyte’s cytoskeleton during their one of the five most recurrent null variants (p.R501X,
differentiation into corneocytes, and finally to desquama­ c.2282del4, p.S3247X, p.R2447X and c.3702delG), which
tion of the stratum corneum. Concurrently, the amino account for about 95% of all known variants within FLG
(N)‐terminus of the protein is translocated into the cell [67]. Interestingly, frequency appears to increase from
nucleus, where it is believed to fulfill an additional but as southern to northern Europe, which might reflect eth­
yet unknown role in regulation of terminal differentia­ nic differences. In populations from UK and Ireland,
tion. Moreover, degradation products of FLG, free hygro­ FLG loss‐of‐function variants seem to be more prevalent
scopic amino acids and their derivatives, constitute a compared to continental Europe [65,68–72]. Of the more
large part of the pool of so‐called natural moisturizing than 20 described loss‐of‐function variants found in
factors (NMFs) within the skin [61]. NMFs play an impor­ the European population, the majority could not be found
tant role in regulation of skin hydration and skin pH, the in Asian individuals [65,73,74] and vice versa. Similarly,
latter of which is closely linked to protease activity and no FLG loss‐of‐function variants predominate in
antimicrobial defence [62]. The importance of FLG‐ African‐American children; however, uncommon
derived breakdown products for skin barrier function is FLG loss‐of‐function variants in African‐American chil­
supported by the remarkably short half‐life of FLG, dren have been identified and shown to be associated
which exists only for 6 hours before its full proteolysis with more persistent AD [75]. Each ethnic group seems to
into NMFs. have its own exclusive null variants showing the same
FLG was suspected of involvement in keratinization strong impact on the development of eczema. All reported
disorders such as ichthyosis vulgaris (IV) more than variants are frameshift or nonsense variants leading to a
25 years ago, and its expression was found to be clearly premature translation stop and hence truncation of the
Chapter 14  Genetics and Aetiology of Atopic Dermatitis 189

Putative filaggrin functions

Outer stratum corneum


Hydration through hygroscopic
amino acids UCA and PCA
Inner stratum corneum Possible contribution to acid mantle
through acid degradation products
Lipid bilayer
Filament binding, barrier integrity
Granular layer Profilaggrin in granular layer:
Pro-protein, nonfunctional

(a) (b)

ATOPIC DERMATITIS
SECTION 3:
PCA
H2O

PADs 1+3 Caspase 14


+PCA
TGMs Profilaggrin A domain
“NMF”
Profilaggrin B domain
Ca2+
Matriptase Profilaggrin tail domain
Dephosphorylation LEKTI
CAP1/Prss Filaggrin

Keratohyalin F granule
Nucleus
Free amino acid

(c)
Fig. 14.2  Filaggrin expression and putative functions in the skin barrier. (a) The precursor pro‐protein profilaggrin is strongly expressed within keratohyalin
granules, tightly limited to and accounting for the typical appearance of the granular layer. The stratum corneum stains strongly positive for filaggrin. (b)
Filaggrin has several proposed site‐specific functions under the influence of the epidermal terminal differentiation programme through the outer granular
layer (cleavage of profilaggrin to filaggrin), lipid bilayer of the inner stratum corneum (filament compaction, contribution to barrier integrity) and during
desquamation of the outer stratum corneum (production of amino acid degradation products that contribute to the hydration of these outer layers and
probably contribute to the ‘acid mantle’). (c) Current knowledge of molecular control of filaggrin homeostasis. Profilaggrin is dephosphorylated in
conditions of increasing calcium concentration and then proteolytically cleaved by the proteases matriptase (inhibited by the protease inhibitor LETKI) and
CAP1/Prss. Post proteolysis, the filaggrin tail domain locates to the nucleus as part of the terminal differentiation process. Free filaggrin protein is cross‐
linked to keratin filaments by transglutaminases (TGMs) and subsequently deiminated by peptidylarginine deiminases (PADs) 1 and 3. Further posttransla-
tional modification is undertaken by caspase 14 to produce the free amino acid hygroscopic degradation products urocanic acid (UCA) and pyrrolidone
carboxylic acid (PCA), collectively known as natural moisturizing factor (NMF), which contributes to stratum corneum hydration. Source: O’Regan et al.
2008 [60]. Reproduced with permission of Elsevier.

profilaggrin molecule. The two most common variants, Th2 cytokine cluster
p.R501X (a nonsense variant of the arginine codon 501 to One of the most consistent associations with eczema and
a stop codon) and c.2282del4 (a frameshift variant at posi­ a variety of other immune‐related disorders such as
tion 2282 due to a four base pair [bp] deletion), in the first Crohn’s disease, asthma and psoriasis has been observed
repeat of the gene impede any FLG synthesis from these for the T‐helper 2 (Th2) cytokine cluster on chromosome
alleles. As the 3′ gene sequence seems to be important for 5q31 [77,78], a locus that harbours the genes encoding
posttranslational processing into functional FLG subu­ interleukin (IL)‐3, IL‐4, IL‐5 and IL‐13. These cytokines
nits, even variants located in this region prevent or reduce are mainly produced by differentiated CD4+ Th2 cells
production of free FLG in the stratum corneum [62,69]. that are responsible for the antibody‐mediated immune
Scientific evidence for this gene as one of the strongest response against parasites, allergens and bacteria. Th2
risk factors for eczema is compelling due to numerous cytokines show a great functional overlap and are
replication studies and meta‐analyses, which revealed an involved in Th2 cell development and polarization, B‐cell
overall odds ratio of >3.0 for eczema [76]. Due to the dis­ proliferation and differentiation, IgE antibody produc­
covery of FLG, susceptibility genes for eczema are now tion, and activation and recruitment of certain immune
divided into two major functional groups: genes contrib­ cell subsets such as mast cells and eosinophils to inflam­
uting to epidermal or epithelial structures, and genes matory sites. Different potential risk SNPs have been
encoding immune regulatory proteins. identified throughout the whole region, and functionally
190 Section 3  Atopic Dermatitis and Related Disorders

disruptive variants within IL4 and IL13 genes are con­ by targeted next‐generation sequencing revealed a
vincingly associated with total IgE levels and atopic dis­ number of low‐frequency variants within the coding
eases including eczema (reviewed in [79]). Additionally, region of LRRC32 to be strongly associated with eczema.
various GWAS identified associated SNPs within RAD50, LRRC32 encodes the protein glycoprotein A repetitions
a gene situated within the cytokine cluster between predominant (GARP), a cell surface receptor mainly
IL4/IL13 and IL5, encoding a DNA repair protein that is expressed on activated regulatory T cells (Tregs), which
ubiquitously expressed and therefore lacks any direct bio­ binds and activates latent TGF‐β, thereby regulating the
logical connection to eczema. However, in mouse models bioavailability of this multifunctional cytokine [87]. All of
RAD50 has been shown to contain an evolutionarily con­ the identified variants lead to an amino acid exchange
served locus control region (LCR), which is supposed to within the primary structure of the protein, and structural
tightly control expression of the neighbouring Th2 modelling of GARP predicted a defective posttransla­
cytokines [80]. Hence, associated polymorphisms in tional modification due to the identified variants, finally
RAD50 influence regulation of Th2 cytokine gene tran­ leading to incorrect folding and constrained transporta­
ATOPIC DERMATITIS

scription rather than RAD50 gene function itself. tion of GARP to the outer cell membrane. Overexpression
Regulation through the LCR within RAD50 occurs via a experiments of mutated GARP indeed showed reduction
SECTION 3:

complex recruitment of transcription factors and other of the membrane‐bound protein, and a reduced conver­
regulatory molecules and includes epigenetic remodelling sion rate of CD4+/CD25− T cells into Tregs was observed
of the three‐dimensional structure of chromatin within when obtained from individuals carrying the identified
this region. An intrachromosomal loop allows direct variants [86]. Tregs are of high importance for suppres­
interaction of the transcription factor machinery bound to sion of effector T cells and other leucocytes and for con­
the LCR with the Th2 cytokine gene promoter regions in trol of inflammatory immune responses and autoimmunity
order to activate or repress their expression [81,82]. The [88], and experimental deactivation of GARP has been
LCR has also been shown to be rapidly demethylated in shown to lead to a reduction in the suppressor activity of
murine cell lines of stimulated Th2 cells [83], linking the these cells [89]. It is possible to speculate that the missing
epigenetic mechanism of DNA methylation with activa­ membrane expression of GARP on activated Tregs leads
tion of Th2 cytokine expression. Inactivation of the LCR to a decreased activation rate of TGF‐β and thereby affects
in mice leads to a reduction of Th2 cytokine and IgE levels downstream function of this important cytokine, finally
[84], and variants within the LCR impact its transcrip­ resulting in an overshooting inflammatory immune reac­
tional activity [85]. tion. Moreover, Tregs are suspected to be involved in a
Together, genetic as well as epigenetic elements seem to variety of other diseases, and association of the previ­
regulate expression of this important immune gene clus­ ously mentioned eczema marker has also been detected
ter, and variants might interfere with both mechanisms, in a GWAS on Crohn’s disease, implying that the 11q13.5
thereby promoting Th2‐dominated immune reactions locus and GARP are also important within the disease
characteristically for atopy‐related disorders like eczema. pathophysiology of further (auto)immune diseases.
Given the number of immune‐mediated diseases that
have been linked to the Th2 cytokine cluster, it seems very
Conclusions and future directions
likely that variants within this region are more relevant
for the atopic state sensu lato than for any individual Despite much progress over the past two decades, our
distinct atopic disease. understanding of the complex genetic susceptibility to
Identification of further causal variants that probably eczema is still incomplete. In particular, there is a lack of
exist within this important risk locus is hampered by the follow‐up studies in order to determine affected genes and
complex marker–marker correlation in this genomic downstream mechanisms. Thus far only the 1q21.3, 5q31
region and the gene–gene interactions through overlap­ and 11q13.5 loci have been functionally explored. The
ping functional pathways. important role of the skin barrier within eczema has been
clearly evidenced with the identification and validation of
GARP (glycoprotein A repetitions FLG variants as the strongest known genetic risk factors for
predominant)/LRRC32 (leucine‐rich the disease. This paradigm change has now also led to early
repeat‐containing protein 32) intervention studies of primary prevention of the disease by
Another consistently‐replicated eczema risk locus has improving the skin barrier function with daily emollient
been identified on chromosome 11q13.5, with the strong­ applications. The results of these randomized intervention
est association observed for an SNP located in an inter­ studies have been an approximate 50% reduction of eczema
genic region between the two annotated genes chromosome incidence in high‐risk newborns [90, 91], lending further
11 open reading frame 30 (C11orf30) and leucine rich repeat support for the key importance of a functionally intact skin
containing 32 (LRRC32). Assignment of the signal to one of barrier. Likewise, genetic findings that imply an important
its neighbouring genes could not be accomplished, as role of variation in genes affecting (auto‐)immune regula­
both are attractive candidates due to their known func­ tion, in particular innate signalling and T‐cell activation and
tions in epithelial immunity and differentiation and acti­ specification, have contributed to the development of drugs
vation of regulatory T cells, respectively, and the causative modifying immune pathways, some of which are already in
risk gene marked by the associated SNP has more recently advanced stages of development and showed encouraging
been identified [86]. Genetic fine mapping of the region results in clinical trials [92–94].
Chapter 14  Genetics and Aetiology of Atopic Dermatitis 191

In order to deepen our understanding of the genetic 19 Sneddon IB. The management of infantile eczema. Med Press 1951;
226:329–33.
architecture of eczema, there is a need for further large‐
20 Schaffer N. Atopic dermatitis in the older child. J Asthma Res
scale and detailed whole‐genome studies, follow‐up 1966;3:189–91.
studies for fine‐mapping and functional investigations, 21 Larsen FS, Holm NV, Henningsen K. Atopic dermatitis. A genetic‐
and holistic approaches that integrate phenotypic varia­ epidemiologic study in a population‐based twin sample. J Am Acad
Dermatol 1986;15:487–94.
bles and information from different molecular levels. In 22 Schultz Larsen F. Atopic dermatitis: a genetic‐epidemiologic study in
addition, it will be necessary to revisit traditional pheno­ a population‐based twin sample. J Am Acad Dermatol 1993;
type definitions based on familial components, heritable 28:719–23.
endophenotypes and genetic markers. To better define 23 Thomsen SF, Ulrik CS, Kyvik KO et al. Importance of genetic factors
in the etiology of atopic dermatitis: a twin study. Allergy Asthma Proc
pathophysiological mechanisms and to identify novel 2007;28:535–9.
therapeutic targets and biomarkers that predict therapeu­ 24 van Beijsterveldt CE, Boomsma DI. Genetics of parentally reported
tic response, there has been increasing interest in develop­ asthma, eczema and rhinitis in 5‐yr‐old twins. Eur Respir J 2007;
29:516–21.
ing computational analysis and predictive models of AD

ATOPIC DERMATITIS
25 Phillips PC. Epistasis—the essential role of gene interactions in the
[95]. The final frontier will certainly be to translate genetic structure and evolution of genetic systems. Nat Rev Genet
findings into an improved classification and the develop­ 2008;9:855–67.

SECTION 3:
ment of more targeted interventions. 26 Glazier AM, Nadeau JH, Aitman TJ. Finding genes that underlie com­
plex traits. Science 2002;298:2345–9.
27 Williams H, Robertson C, Stewart A et al. Worldwide variations in the
References prevalence of symptoms of atopic eczema in the International Study
1 Bieber T. Atopic dermatitis. N Engl J Med 2008;358:1483–94. of Asthma and Allergies in Childhood. J Allergy Clin Immunol
2 Wuthrich B. The natural history of atopic dermatitis. In: Ring J, 1999;103:125–38.
Przybilla B, Ruzicka T (eds) Handbook of Atopic Dermatitis, 2nd edn. 28 Asher MI, Montefort S, Bjorksten B et al. Worldwide time trends in the
Berlin: Springer Verlag, 2006:150–6. prevalence of symptoms of asthma, allergic rhinoconjunctivitis, and
3 Novak N, Bieber T. Allergic and nonallergic forms of atopic diseases. eczema in childhood: ISAAC Phases One and Three repeat multi­
J Allergy Clin Immunol 2003;112:252–62. country cross‐sectional surveys. Lancet 2006;368:733–43.
4 Wuthrich B, Schmid‐Grendelmeier P. The atopic eczema/dermatitis 29 Moffatt MF, Cookson WO. The genetics of asthma. Maternal effects in
syndrome. Epidemiology, natural course, and immunology of the atopic disease. Clin Exp Allergy 1998;28(suppl 1):56–61.
IgE‐associated (“extrinsic”) and the nonallergic ("intrinsic") AEDS. 30 Warner JA, Jones CA, Jones AC, Warner JO. Prenatal origins of allergic
J Investig Allergol Clin Immunol 2003;13:1–5. disease. J Allergy Clin Immunol 2000;105:S493–8.
5 Johansson SG, Bieber T, Dahl R et al. Revised nomenclature for allergy 31 Dold S, Wjst M, von Mutius E et al. Genetic risk for asthma, allergic
for global use: Report of the Nomenclature Review Committee of the rhinitis, and atopic dermatitis. Arch Dis Child 1992;67:1018–22.
World Allergy Organization, October 2003. J Allergy Clin Immunol 32 Ruiz RG, Kemeny DM, Price JF. Higher risk of infantile atopic
2004;113:832–6. dermatitis from maternal atopy than from paternal atopy. Clin Exp
6 Bergmann RL, Edenharter G, Bergmann KE et al. Atopic dermatitis in Allergy 1992;22:762–6.
early infancy predicts allergic airway disease at 5 years. Clin Exp 33 Diepgen TL, Blettner M. Analysis of familial aggregation of atopic
Allergy 1998;28:965–70. eczema and other atopic diseases by ODDS RATIO regression models.
7 Gustafsson D, Sjoberg O, Foucard T. Development of allergies and J Invest Dermatol 1996;106:977–81.
asthma in infants and young children with atopic dermatitis—a pro­ 34 Reik W, Walter J. Genomic imprinting: parental influence on the
spective follow‐up to 7 years of age. Allergy 2000;55:240–5. genome. Nat Rev Genet 2001;2:21–32.
8 Hakansson K, Thomsen SF, Ulrik CS et al. Increase in the prevalence 35 McKusick VA. Mendelian Inheritance in Man and its online version,
of rhinitis among Danish children from 1986 to 2001. Pediatr Allergy OMIM. Am J Hum Genet 2007;80:588–604.
Immunol 2007;18:154–9. 36 Lee YA, Wahn U, Kehrt R et al. A major susceptibility locus for atopic
9 Spergel JM. From atopic dermatitis to asthma: the atopic march. Ann dermatitis maps to chromosome 3q21. Nat Genet 2000;26:470–3.
Allergy Asthma Immunol 2010;105:99–106. 37 Morar N, Edster P, Street T et al. Fine mapping of susceptibility genes
10 Illi S, von Mutius E, Lau S et al. The natural course of atopic dermatitis for atopic dermatitis in the epidermal differentiation complex on
from birth to age 7 years and the association with asthma. J Allergy chromosome 1q21. Br J Dermatol 2007;157:3.
Clin Immunol 2004;113:925–31. 38 Altshuler D, Daly MJ, Lander ES. Genetic mapping in human disease.
11 Williams H, Flohr C. How epidemiology has challenged 3 prevailing Science 2008;322:881–8.
concepts about atopic dermatitis. J Allergy Clin Immunol 2006; 39 Tabor HK, Risch NJ, Myers RM. Candidate‐gene approaches for stud­
118:209–13. ying complex genetic traits: practical considerations. Nat Rev Genet
12 Flohr C, Weiland SK, Weinmayr G et al. The role of atopic sensitiza­ 2002;3:391–7.
tion in flexural eczema: findings from the International Study of 40 Barnes KC. An update on the genetics of atopic dermatitis: scratching
Asthma and Allergies in Childhood Phase Two. J Allergy Clin the surface in 2009. J Allergy Clin Immunol 2010;125:16–29.e1–11.
Immunol 2008;121:141–7.e4. 41 Lohmueller KE, Pearce CL, Pike M et  al. Meta‐analysis of genetic
13 He R, Oyoshi MK, Garibyan L et al. TSLP acts on infiltrating effector association studies supports a contribution of common variants to
T cells to drive allergic skin inflammation. Proc Natl Acad Sci U S A susceptibility to common disease. Nat Genet 2003;33:177–82.
2008;105:11875–80. 42 The International HapMap Consortium. The International HapMap
14 Demehri S, Morimoto M, Holtzman MJ, Kopan R. Skin‐derived TSLP Project. Nature 2003;426:789–96.
triggers progression from epidermal‐barrier defects to asthma. PLoS 43 Grant SF, Hakonarson H. Microarray technology and applications in
Biol 2009;7:e1000067. the arena of genome‐wide association. Clin Chem 2008;54:1116–24.
15 Williams HC, Burney PG, Hay RJ et  al. The U.K. Working Party’s 44 Esparza‐Gordillo J, Weidinger S, Folster‐Holst R et al. A common
Diagnostic Criteria for Atopic Dermatitis. I. Derivation of a minimum variant on chromosome 11q13 is associated with atopic dermatitis.
set of discriminators for atopic dermatitis. Br J Dermatol 1994;131: Nat Genet 2009;41:596–601.
383–96. 45 Sun LD, Xiao FL, Li Y et al. Genome‐wide association study identifies
16 Brenninkmeijer EE, Schram ME, Leeflang MM et al. Diagnostic crite­ two new susceptibility loci for atopic dermatitis in the Chinese Han
ria for atopic dermatitis: a systematic review. Br J Dermatol 2008; population. Nat Genet 2011;43:690–4.
158:754–65. 46 Hirota T, Takahashi A, Kubo M et al. Genome‐wide association study
17 Johansson SG, Hourihane JO, Bousquet J et al. A revised nomencla­ identifies eight new susceptibility loci for atopic dermatitis in the
ture for allergy. An EAACI position statement from the EAACI Japanese population. Nat Genet 2012;44:1222–6.
nomenclature task force. Allergy 2001;56:813–24. 47 Paternoster L, Standl M, Chen CM et al. Meta‐analysis of genome‐
18 Cooke R, van der Veer A. Human sensitization. J Immunol wide association studies identifies three new risk loci for atopic
1916;1:201–5. dermatitis. Nat Genet 2012;44:187–92.
192 Section 3  Atopic Dermatitis and Related Disorders

48 Ellinghaus D, Baurecht H, Esparza‐Gordillo J et al. High‐density 73 Nomura T, Sandilands A, Akiyama M et al. Unique mutations in the
genotyping study identifies four new susceptibility loci for atopic filaggrin gene in Japanese patients with ichthyosis vulgaris and atopic
dermatitis. Nat Genet 2013;45:808–12. dermatitis. J Invest Dermatol 2007;119:434–40.
49 Esparza‐Gordillo J, Schaarschmidt H, Liang L et al. A functional IL‐6 74 Nomura T, Akiyama M, Sandilands A et al. Prevalent and rare muta­
receptor (IL6R) variant is a risk factor for persistent atopic dermatitis. tions in the gene encoding filaggrin in Japanese patients with ichthyo­
J Allergy Clin Immunol 2013;132:371–7. sis vulgaris and atopic dermatitis. J Invest Dermatol 2009;129:1302–5.
50 Weidinger S, Willis‐Owen SA, Kamatani Y et  al. A genome‐wide 75 Margolis DJ, Mitra N, Gochnauer H et al. Uncommon filaggrin vari­
association study of atopic dermatitis identifies loci with overlapping ants are associated with persistent atopic dermatitis in African
effects on asthma and psoriasis. Hum Mol Genet 2013;22:4841–56. Americans. J Invest Dermatol 2018;138:1501–6.
51 Paternoster L, Standl M, Waage J et al. Multi‐ancestry genome‐wide 76 Rodriguez E, Baurecht H, Herberich E et al. Meta‐analysis of filag-
association study of 21,000 cases and 95,000 controls identifies new grin polymorphisms in eczema and asthma: robust risk factors in
risk loci for atopic dermatitis. Nat Genet 2015;47:1449–56. atopic disease. J Allergy Clin Immunol 2009;123:1361–70.e7.
52 Schaarschmidt H, Ellinghaus D, Rodriguez E et  al. A genome‐wide 77 Rioux JD, Daly MJ, Silverberg MS et al. Genetic variation in the 5q31
association study reveals 2 new susceptibility loci for atopic dermati­ cytokine gene cluster confers susceptibility to Crohn disease. Nat
tis. J Allergy Clin Immunol 2015;136:802–6. Genet 2001;29:223–8.
53 Weidinger S, Novak N. Atopic dermatitis. Lancet 2016;387:1109–22. 78 Li Y, Chang M, Schrodi SJ et  al. The 5q31 variants associated with
ATOPIC DERMATITIS

54 Ellinghaus D, Jostins L, Spain SL et al. Analysis of five chronic inflam­ psoriasis and Crohn’s disease are distinct. Hum Mol Genet
matory diseases identifies 27 new associations and highlights disease‐ 2008;17:2978–85.
specific patterns at shared loci. Nat Genet 2016;48:510–18. 79 Vercelli D. Discovering susceptibility genes for asthma and
SECTION 3:

55 Ferreira MAR, Vonk JM, Baurecht H et al. Eleven loci with new repro­ allergy. Nat Rev Immunol 2008;8:169–82.
ducible genetic associations with allergic disease risk. J Allergy Clin 80 Lee GR, Fields PE, Griffin TJ, Flavell RA. Regulation of the Th2
Immunol 2019;143(2):691–9. cytokine locus by a locus control region. Immunity 2003;19:
56 Devos M, Mogilenko DA, Fleury S et al. Keratinocyte expression of 145–53.
A20/TNFAIP3 controls skin inflammation associated with atopic der­ 81 Spilianakis CG, Flavell RA. Long‐range intrachromosomal interac­
matitis and psoriasis. J Invest Dermatol 2019;139:135–45. tions in the T helper type 2 cytokine locus. Nat Immunol 2004;
57 Dale BA. Purification and characterization of a basic protein from the 5:1017–27.
stratum corneum of mammalian epidermis. Biochim Biophys Acta 82 Lee GR, Spilianakis CG, Flavell RA. Hypersensitive site 7 of the TH2
1977;491:193–204. locus control region is essential for expressing TH2 cytokine genes
58 Candi E, Schmidt R, Melino G. The cornified envelope: a model of cell and for long‐range intrachromosomal interactions. Nat Immunol
death in the skin. Nat Rev Mol Cell Biol 2005;6:328–40. 2005;6:42–8.
59 Proksch E, Brandner JM, Jensen JM. The skin: an indispensable bar­ 83 Kim ST, Fields PE, Flavell RA. Demethylation of a specific hypersensi­
rier. Exp Dermatol 2008;17:1063–72. tive site in the Th2 locus control region. Proc Natl Acad Sci U S A
60 O’Regan GM, Sandilands A, McLean WH, Irvine AD. Filaggrin in 2007;104:17052–7.
atopic dermatitis. J Allergy Clin Immunol 2008;122:689–93. 84 Koh BH, Hwang SS, Kim JY et al. Th2 LCR is essential for regulation
61 Rawlings AV, Harding CR. Moisturization and skin barrier function. of Th2 cytokine genes and for pathogenesis of allergic asthma. Proc
Dermatol Ther 2004;17(suppl 1):43–8. Natl Acad Sci U S A 2010;107:10614–19.
62 Sandilands A, Sutherland C, Irvine AD, McLean WH. Filaggrin in the 85 Kretschmer A, Moller G, Lee H et  al. A common atopy‐associated
frontline: role in skin barrier function and disease. J Cell Sci variant in the Th2 cytokine locus control region impacts transcrip­
2009;122:1285–94. tional regulation and alters SMAD3 and SP1 binding. Allergy 2014;
63 Fleckman P, Holbrook KA, Dale BA, Sybert VP. Keratinocytes cul­ 69:632–42.
tured from subjects with ichthyosis vulgaris are phenotypically 86 Manz J, Rodriguez E, ElSharawy A et al. Targeted resequencing and
abnormal. J Invest Dermatol 1987;88:640–5. functional testing identifies low‐frequency missense variants in
64 Sybert VP, Dale BA, Holbrook KA. Ichthyosis vulgaris: identification the gene encoding GARP as significant contributors to atopic der-
of a defect in synthesis of filaggrin correlated with an absence of kera­ matitis risk. J Invest Dermatol 2016;136:2380–6.
tohyaline granules. J Invest Dermatol 1985;84:191–4. 87 Wang R, Zhu J, Dong X et al. GARP regulates the bioavailability and
65 Smith FJ, Irvine AD, Terron‐Kwiatkowski A et al. Loss‐of‐function activation of TGFbeta. Mol Biol Cell 2012;23:1129–39.
mutations in the gene encoding filaggrin cause ichthyosis vulgaris. 88 Zhang H, Kong H, Zeng X et al. Subsets of regulatory T cells and their
Nat Genet 2006;38:337–42. roles in allergy. J Transl Med 2014;12:125.
66 Palmer CN, Irvine AD, Terron‐Kwiatkowski A et al. Common loss‐ 89 Fridrich S, Hahn SA, Linzmaier M et al. How soluble GARP enhances
of‐function variants of the epidermal barrier protein filaggrin are a major TGFbeta activation. PLoS One 2016;11:e0153290.
predisposing factor for atopic dermatitis. Nat Genet 2006;38:441–6. 90 Horimukai K, Morita K, Narita M et al. Application of moisturizer to
67 Irvine AD, McLean WH, Leung DY. Filaggrin mutations associated neonates prevents development of atopic dermatitis. J Allergy Clin
with skin and allergic diseases. N Engl J Med 2011;365:1315–27. Immunol 2014;134:824–30.e6.
68 Barker JN, Palmer CN, Zhao Y et al. Null mutations in the filaggrin 91 Simpson EL, Chalmers JR, Hanifin JM et al. Emollient enhancement of
gene (FLG) determine major susceptibility to early‐onset atopic the skin barrier from birth offers effective atopic dermatitis preven­
dermatitis that persists into adulthood. J Invest Dermatol 2007; tion. J Allergy Clin Immunol 2014;134:818–23.
127:564–7. 92 Beck LA, Thaci D, Hamilton JD et al. Dupilumab treatment in adults
69 Sandilands A, Terron‐Kwiatkowski A, Hull PR et  al. Com­ with moderate‐to‐severe atopic dermatitis. N Engl J Med 2014;
prehensive analysis of the gene encoding filaggrin uncovers prevalent 371:130–9.
and rare mutations in ichthyosis vulgaris and atopic eczema. Nat Genet 93 Simpson EL, Bieber T, Guttman‐Yassky E et al. Two Phase 3 trials of
2007;39:650–4. dupilumab versus placebo in atopic dermatitis. N Engl J Med
70 Brown SJ, Relton CL, Liao H et al. Filaggrin null mutations and child­ 2016;375:2335–48.
hood atopic eczema: a population‐based case‐control study. J Allergy 94 Thaci D, Simpson EL, Beck LA et al. Efficacy and safety of dupilumab
Clin Immunol 2008;121:940–46.e3. in adults with moderate‐to‐severe atopic dermatitis inadequately
71 Rice NE, Patel BD, Lang IA et al. Filaggrin gene mutations are associ­ controlled by topical treatments: a randomised, placebo‐controlled,
ated with asthma and eczema in later life. J Allergy Clin Immunol dose‐ranging phase 2b trial. Lancet 2016;387:40–52.
2008;122:834–6. 95 Eyerich K, Brown SJ, Perez White BE et al. Human and computational
72 Weidinger S, O’Sullivan M, Illig T et  al. Filaggrin mutations, atopic models of atopic dermatitis: a review and perspectives by an expert
eczema, hay fever, and asthma in children. J Allergy Clin Immunol panel of the International Eczema Council. J Allergy Clin Immunol
2008;121:1203–9. 2019;143(1):36–45.
 193

C HA PTER   15

Clinical Features and Diagnostic Criteria


of Atopic Dermatitis
Sinéad M. Langan1 & Hywel C. Williams2
 Faculty of Epidemiology and Population Health, London, School of Hygiene and Tropical Medicine, London, UK
1

 The Centre of Evidence Based Dermatology, University of Nottingham, Nottingham, UK


2

ATOPIC DERMATITIS
Clinical features of atopic dermatitis, 194 Diagnostic criteria for atopic dermatitis, 203

SECTION 3:
Abstract define reliably and too infrequent to be of use when examining
groups of individuals, especially when it adds very little to the
This chapter is divided into two sections: the first lists the common predictive ability of existing cardinal features such as flexural
and less common clinical features of atopic dermatitis (AD) with involvement and dry skin. Likewise, diagnostic criteria that may
the aim of helping those less familiar with diagnosing atopic der- produce acceptable results for estimating the prevalence of AD in
matitis in a clinical setting. The second part deals with diagnostic a country may not be suitable when trying to diagnose AD in an
criteria for atopic dermatitis that may be used in research studies. individual, since every set of diagnostic criteria will have limita-
The two sections are set apart deliberately since the requirements tions in sensitivity (proportion of true cases correctly identified)
of a disease definition for making a clinical diagnosis in an individual and specificity (proportion of non‐cases correctly identified). All
may be somewhat different from making a group definition for current diagnostic criteria will therefore misclassify some
research participants. A clinical sign such as thinning of the lateral individuals who have AD as not having AD, and some who do not
eyebrows may be associated with AD, and possibly useful in the have AD as having AD. Such misclassification may be acceptable
context of tipping a clinician to make a diagnosis of AD in a child in research studies, providing the degree of misclassification is
with indeterminate skin inflammation. Yet it is too difficult to understood in relation to the objective of the study.

Key points • Previous definitions of an incident case of AD have paradoxically


included chronicity.
• What is recognized as AD today may turn out to be four or five
Clinical features of atopic dermatitis
different phenotypes (subtypes) in years to come. Any proposals
• Although atopic dermatitis (AD) can affect any part of the body, it has for new subtypes of AD should be based on strong evidence that
a predilection for the skinfolds, the reason for which is still unclear. they increase predictive ability in terms of prognosis, disease
• The distribution of eczema changes from involvement of the face, associations or response to treatments.
scalp and extensor surfaces in infancy to flexural involvement in • While a clinical diagnosis is appropriate in clinical practice,
childhood and later life. standardized diagnostic criteria for AD are essential when
• The morphological appearance of AD can vary greatly from acute comparing groups of people in clinical studies.
red vesicular eczema to chronic thickened purple/grey lichenified • A good disease definition should be valid, reliable, acceptable
patches. to the population, coherent with prevailing clinical concepts and
• AD can be associated with serious complications including comprehensive in its application.
secondary infections and exfoliative dermatitis. • The Hanifin and Rajka list of diagnostic features was an important
• Other differential diagnoses should be considered when infants landmark in suggesting major and minor criteria for AD.
present with a severe eczematous rash with failure to thrive, • A systematic review suggested that the most extensively tested
recurrent infections or petechiae. criteria (the UK Working Party’s refinement of the Hanifin and
Rajka criteria) should be used in future intervention studies,
Diagnostic criteria for atopic dermatitis although there is room for improvement.
• Ideally, diagnostic criteria should be tested beforehand
• AD is a difficult disease to define since it is variable in morphology
for validity in the setting where they are to be used. If
and distribution according to age, skin type and whether the
translation or transcultural interpretation becomes a
skin is acutely or chronically affected.
problem, then question‐free methods of assessing visible
• Although the term ‘atopic dermatitis’ is used throughout this
flexural dermatitis in a standardized way are an option for
book, most cases of ‘atopic’ dermatitis are not atopic when
comparative research.
tested in populations. As a consequence there is debate as to
• It is crucial to consider the type of clinical study before deciding
whether the term ‘atopic dermatitis’ should be reserved for
which diagnostic criteria can be used. Surveys will need a trade‐
cases with evidence of IgE reactivity.
off between sensitivity and specificity whereas clinical trials will
• The positive predictive value of diagnostic criteria will fall with
require specific criteria.
low disease prevalence even though sensitivity and specificity
might appear to be high.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
194 Section 3  Atopic Dermatitis and Related Disorders

Clinical features of atopic dermatitis


The evidence for the first section of this chapter is largely
based on classical descriptions in textbooks and scholarly
articles by pioneers in the field of atopic dermatitis (AD),
supplemented by clinical experience. An excellent historical
account of the concepts that formed the notion of what
is recognized today as AD may be found elsewhere [1].
Multiple factors including age, ethnic origin, level of
disease activity, therapeutic interventions and infectious
complications influence both distribution and morphology
of AD, leading to variations in disease presentation. The
characteristic clinical features of AD distribution and
ATOPIC DERMATITIS

morphology described here aid clinical diagnosis in the


absence of a specific laboratory test for AD.
SECTION 3:

Distribution
The distribution of AD is dependent on the age of onset
and disease activity. Infantile eczema usually starts at
3  months or younger and typically affects the face and
scalp first (Fig. 15.1a and b), then spreads to involve the
limbs and trunk in a symmetrical distribution (Fig. 15.2)
[2]. Truncal lesions are diffuse while limb lesions tend to
be discrete and localized (Fig.  15.3). The napkin area is
frequently spared for reasons that are unclear, although
local humidity has been suggested.
Fig. 15.2  Facial eczema and diffuse truncal erythema in an infant. Source:
Courtesy of Dr Paula E. Beattie.

(a)

Fig. 15.3  Symmetrical eczema involving the trunk and extensor surfaces
in atopic dermatitis in a child. Source: Courtesy of Professor Hywel C. Williams.

In childhood, AD distribution is seen most commonly in


a flexural pattern. The tendency for skin inflammation and
the effects of chronic rubbing (lichenification) to become
accentuated in the flexures is one of the classical hallmarks
of AD (Fig. 15.4) [3]. Flexures refer to bends in the articular
(b) skeleton where skin apposes skin. What exactly consti-
Fig. 15.1  Scalp involvement in atopic dermatitis in an infant. Source: tutes a flexure varies from text to text, but most agree that
Courtesy of Dr Paula E. Beattie. the fronts of the elbows, backs of the knees, around the
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 195

neck and in front of the ankles are the commonly affected can affect any part of the body. The fact that a child
flexures. All of the flexure is not always involved – presents with an itchy scalp and diffuse patches of skin
lichenification as a result of prolonged rubbing typically inflammation on extensor parts of the limbs should not
occurs most prominently behind the knees along the most put one off diagnosing AD since it is the most common
prominent tendons (semimembranosus and semitendino- inflamed itchy chronic skin eruption in children in
sus) and likewise overlying the brachioradialis muscle developed countries.
group on the antecubital fossae (Fig. 15.5a and b). From puberty onwards, AD tends to affect the face,
Other affected areas include the wrists and folds hands, back, wrists and dorsal feet. The hands and fin-
beneath the buttocks (infragluteal folds). Why the most gers are frequently involved, probably as a result of con-
flexural sites of the body, i.e. inguinal folds and apex stant irritant exposure, which may result in oedema
of  the axillae, are infrequently involved is unclear. and fissures overlying the finger joints (Fig.  15.6) and
Eczematous inflammation at these sites may be more scaling on the palms adjoining the fingers (apron sign).
likely to be a marker of seborrhoeic dermatitis of infancy Adults may only have persisting hand or facial eczema

ATOPIC DERMATITIS
or early‐onset psoriasis. Two points are worthy of note
with regards to flexural involvement; the first is that AD

SECTION 3:
Fig. 15.4  Thickened lichenified atopic dermatitis on the lower leg and Fig. 15.6  Chronic hand eczema with loss of cuticles, nail dystrophy,
dorsal foot in darkly pigmented skin. Source: Courtesy of Professor Hywel fissuring and scaling. Source: © Diepgen TL, Yihune G et al. Dermatology
C. Williams. Online Atlas (www.dermis.net). Reprinted with permission.

(a) (b)
Fig. 15.5  (a and b) Flexural subacute atopic dermatitis involving the antecubital and popliteal fossae with erythema and excoriations. Source: Courtesy of
Professor Hywel C. Williams.
196 Section 3  Atopic Dermatitis and Related Disorders

and skin that is sensitive to irritant exposure, or they may


have persistent chronic AD.
Specific sites that are frequently involved by AD are as
follows: lips with cheilitis in childhood (Fig. 15.7), which
can lead to secondary ‘lip‐lick cheilitis’ or exfoliating
cheilitis with perlèche (single or multiple fissures and
cracks at the labial commissures) and involvement of the
vermillion border in adult AD. The infra‐auricular,
supra‐auricular and retroauricular region is frequently
affected by fissures (rhagades) in children (Fig.  15.8)
while eyelids and periorbital skin are frequently
involved in adolescents and adults, resulting in promi-
nent infraorbital creases from skin oedema and postin-
ATOPIC DERMATITIS

Fig. 15.7  Acute cheilitis in a child with atopic dermatitis with erythema flammatory pigmentation resulting in the affected
and swelling of the vermillion border and perioral region and fissuring of individual appearing fatigued. Some sites of predilec-
SECTION 3:

the commissures. Source: © Diepgen TL, Yihune G et al. Dermatology tion of AD such as around the eyes, mouth or nose
Online Atlas (www.dermis.net). Reprinted with permission. (Fig. 15.9) or under the ear also occur probably as a result
of influences other than skin‐to‐skin contact such as air-
borne allergens and irritants, or irritant contact dermati-
tis around the mouth from saliva and wet foods in early
age. Nipple eczema is frequently seen from puberty
onwards, but is also seen in infants and can present with
nipple swelling (Fig. 15.10a and b).

Fig. 15.8  Fissures of the infra‐auricular region in childhood atopic Fig. 15.9  Subacute eczema with fissuring and oedema. Source: ©
dermatitis. Source: © Diepgen TL, Yihune G et al. Dermatology Online Diepgen TL, Yihune G et al. Dermatology Online Atlas (www.dermis.net).
Atlas (www.dermis.net). Reprinted with permission. Reprinted with permission.

(a) (b)
Fig. 15.10  (a) Acute nipple eczema with erythema, erosions, oozing and secondary impetiginization. Source: © Diepgen TL, Yihune G et al. Dermatology
Online Atlas (www.dermis.net). Reprinted with permission. (b) Chronic nipple eczema with lichenification and swelling. Source: Courtesy of Dr Diana Purvis.
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 197

ATOPIC DERMATITIS
SECTION 3:
(a) (b)

(c)
Fig. 15.11  (a) Acute, (b) subacute and (c) chronic eczema. Source: Courtesy of Professor Hywel C. Williams.

Morphology
The morphology of AD can be variable and depends on
the stage of the lesion. Acute lesions are characterized by
ill‐defined erythema, papules, papulovesicles, erosions,
oedema and weeping (Fig. 15.11a). Subacute lesions are
erythematous scaly excoriated plaques and papules
(Fig.  15.11b) while thickened purple/grey lichenified
plaques and fibrotic papules (prurigo) are seen in chronic
lesions (Fig. 15.11c). During infancy, AD tends to be acute
and exudative with chronic forms of AD developing later
in childhood. In longstanding AD, individuals may mani-
fest all three stages of AD concurrently or at different
times depending on the level of disease activity, reflecting
the relapsing and remitting nature of eczema.
Fig. 15.12  Symmetrical infraorbital Morgan–Dennie folds. Source:
© Diepgen TL, Yihune G et al. Dermatology Online Atlas (www.dermis.net).
Associated physical signs Reprinted with permission.
A number of clinical signs frequently accompany AD and
these may be clues to the diagnosis when the distribu-
tion and morphology are not classical, although they 50–60% of cases of AD. However, this sign appears to
may not be pathognomonic of AD. A number of these will vary depending on ethnicity, being more common in
be briefly discussed here. darkly pigmented ethnic groups regardless of AD, and
shows significant variability even within individuals [4]
Periocular and ocular signs (Fig.  15.12). Periorbital pigmentation, ‘atopic shiners’,
Infraorbital folds (Dennie–Morgan lines) are frequently describes a periorbital brown to grey discolouration [5].
seen in AD, with most studies reporting their presence in As previously discussed, thinning or absence of the outer
198 Section 3  Atopic Dermatitis and Related Disorders

(blanching of the skin at the site of stroking with a blunt


instrument rather than developing a partial or full triple
response of Lewis with reflex erythema), are frequently
described in AD and are thought to represent abnormal
vascular responses (Fig. 15.15).
Dry skin (xerosis) is seen in all age groups with AD and
is considered a hallmark of AD (Fig. 15.16). Atopic xerosis
ATOPIC DERMATITIS

Fig. 15.13  Thinning of the outer eyebrows (Hertoghe’s sign) with extensive
scaling and lichenification of the forehead. Source: © Diepgen TL, Yihune G
SECTION 3:

et al. Dermatology Online Atlas (www.dermis.net). Reprinted with permission.

eyebrows (Hertoghe’s sign), a sign originally described in


hypothyroidism, may also be seen although its preva-
lence has not been extensively studied (Fig. 15.13). Specific
ocular signs such as keratoconjunctivitis, keratoconus
and anterior subcapsular cataracts may be associated with
AD, although their population‐based frequency is diffi-
cult to identify from the literature. A population‐based
survey from Korea reported a 50% increased odds of cata-
ract in individuals with AD [6], while an Israeli study
reported no increased risk of keratoconus with AD,
although associations were observed for other atopic
diseases [7]. A population‐based study of ocular infec-
tious diseases from Hawaii suggested that individuals Fig. 15.15  White dermographism with blanching of the skin at the site of
with AD had a five‐fold increased risk of ocular herpes stroking with a blunt instrument rather than developing a partial or full triple
simplex virus and a two‐fold increased risk of herpes response of Lewis with reflex erythema. Source: © Diepgen TL, Yihune G et al.
zoster ophthalmicus [8]. Dermatology Online Atlas (www.dermis.net). Reprinted with permission.

Other cutaneous ‘minor’ signs


Two features are described in the neck region, namely the
anterior neck folds and atopic ‘dirty neck’ (Fig.  15.14).
The ‘dirty neck’ refers to rippled hyperpigmentation of
the anterior and lateral neck, which clinically resembles
macular amyloid. Facial pallor and white dermographism

Fig. 15.14  Rippled hyperpigmentation of the lateral neck (atopic ‘dirty


neck’). Source: © Diepgen TL, Yihune G et al. Dermatology Online Atlas Fig. 15.16  Xerosis in infantile atopic dermatitis. Source: Courtesy of
(www.dermis.net). Reprinted with permission. Dr Paula E. Beattie.
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 199

ATOPIC DERMATITIS
SECTION 3:
(a)

Fig. 15.17  Hyperlinear palms with exaggerated wrinkling of the palms.


Source: Courtesy of Professor Alan Irvine.

may be generalized and is frequently most noticeable on


the shins. Xerosis is characterized by fine scaling without
associated inflammation and roughness on palpation. Dry
skin may be associated with ichthyosis vulgaris, an autoso-
mal dominant condition seen in 8% of individuals with AD
[9,10]. However, studies show that dry skin is also inde-
pendently associated with AD in the absence of ichthyosis
vulgaris [11]. Hyperlinear palms are a frequent finding in
AD (Fig.  15.17). More recently, studies have shown that
palmar hyperlinearity is more frequently seen in AD com-
pared to ‘atopiform’ dermatitis (AD without raised IgE lev- (b)
els) and that this sign is strongly associated with filaggrin Fig. 15.18  (a and b) Pityriasis alba with poorly defined slightly scaly
null mutations and ichthyosis vulgaris [12,13]. hypopigmented patches on the face. (a) Source: Courtesy of Professor
Pityriasis alba describes a condition frequently asso- Nanette Silverberg. (b) Source: Courtesy of Dr Diana Purvis.
ciated with AD characterized by poorly defined slightly
scaly hypopigmented patches on the cheeks and upper
arms (Fig.  15.18). This clinical sign is more obvious in
Nummular or discoid atopic dermatitis
dark skin and can be mistaken for tinea corporis.
This type of AD presents with sharply demarcated c­ ircular
Atopic dermatitis in racially pigmented lesions (Fig. 15.20) often located on the trunk and distal
skin types extensor limbs including dorsal hands, lower legs and
The diagnosis of eczema in dark skin is complicated by the forearms. The individual lesions are frequently dry and
lack of visible erythema. The diagnosis is made based on infiltrated plaques. This pattern of AD is reported to
the presence of a typical history accompanied by lesions worsen in winter time. It is often misdiagnosed as ring-
demonstrating a distribution and morphology consistent worm by general practitioners and is almost always asso-
with AD. Other features classically seen in pigmented skin ciated with concurrent or past flexural AD in children.
are enhanced follicular lichenification with a papular Nummular AD often requires stronger and longer treat-
appearance and postinflammatory hypo‐ and hyperpig- ment with potent topical corticosteroids. In adults, num-
mentation (Fig. 15.19). mular eczema may be independent of atopy.

Clinical features of specific subtypes Diffuse ‘dry type’ AD


of atopic dermatitis This type of AD was first described in Japan and refers to
Specific subtypes of childhood AD deserve a mention as a diffuse dry type AD, sometimes called patchy pityriasi-
their morphology and distribution have not been previously form lichenoid eczema, which is slightly irritating and
discussed in this chapter. clinically consists of confluent scaly nonhyperkeratotic
200 Section 3  Atopic Dermatitis and Related Disorders
ATOPIC DERMATITIS
SECTION 3:

Fig. 15.20  Nummular atopic dermatitis affecting the torso in a young


child. Source: Courtesy of Professor Hywel C. Williams.

Fig. 15.19  Postinflammatory hypo‐ and hyperpigmentation in darkly IgE levels) eczema with palmar hyperlinearity and ker-
pigmented skin. Source: Courtesy of Professor Hywel C. Williams.
atosis pilaris [13,17]; however, filaggrin null mutations
are absent in >50% of individuals with eczema [18–22].
Population‐based studies from Denmark and Sweden
skin‐coloured papules or ‘chicken‐skin spots’, mainly on have reported associations between the presence of
the trunk region [14]. Seasonal variations are described, filaggrin mutations and persistent hand dermatitis in
with worsening in winter. adults, with a further Danish study suggesting addi-
tional associations with self‐reported foot dermatitis
Eczema subtypes/phenotypes [22,23].
There is increasing evidence supporting the existence of AD with eczema herpeticum is another proposed
discrete clinical phenotypes of eczema. This encom- clinical phenotype, reported to be associated with a
passes heterogeneity in the clinical disease course, asso- greater risk of atopic diseases and associated bacterial
ciations with specific infections, disease distribution infections [24].
and responses to treatment. Usual phenotypic group-
ings are based on disease trajectories, atopy status and Complications
associated comorbidities with different groupings in Bacterial infections
published papers. For example Leung et  al. describe Staph. aureus colonization can be demonstrated in 90% of
eight categories: onset in infancy and (1) transient or (2) eczema lesions and its density is associated with disease
persistent; onset in adolescence and (3) mild to moder- severity [25]. Clinically infected AD is characterized by
ate or (4) persistent and severe; (5) associated with high honey‐coloured crusted impetigo, folliculitis and pyo-
IgE level and sensitization; (6) non‐IgE associated; (7) derma or as worsening of AD with increased erythema,
associated with Staphylococcus aureus infections; and (8) oozing and pain (Fig. 15.21). Infections are usually caused
associated with disseminated viral infections [15]. by Staph. aureus but β‐haemolytic streptococci may also
Garmhausen et  al. report the existence of five pheno- be isolated from infected AD. There is increasing research
typic groups based on disease trajectories and associa- into the mechanisms of increased cutaneous infection
tions with IgE. Most studies concur that persistent AD risk in individuals with AD. This research has focused on
is frequently associated with early‐onset eczema with the roles of the defective skin barrier (genetic and
associated rhinitis and childhood hand eczema [16]. acquired) and staphylococcal colonization, and the role
Recent advances in our understanding of the genetic of staphylococcal superantigens acting in concert with
basis for AD are likely to lead to other subtypes of AD. defects in adaptive and innate immunity, the latter high-
Specific emerging subtypes include eczema associated lighted by the reduced capacity to upregulate cutaneous
with null filaggrin mutations. These mutations appear to antimicrobial peptides including cathelicidin and
be associated with severe early‐onset extrinsic (­elevated defensins [26].
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 201

ATOPIC DERMATITIS
SECTION 3:
Fig. 15.23  Molluscum contagiosum presenting as infraorbital umbilicated
and crusted papules. Source: © Diepgen TL, Yihune G et al. Dermatology
Online Atlas (www.dermis.net). Reprinted with permission.
Fig. 15.21  Clinically infected infantile atopic dermatitis affecting the
extensor aspects of the limbs and trunk. Source: Courtesy of Professor
Hywel C. Williams
mutations [27]. A clinically similar eruption (eczema
vaccinatum) can be seen following smallpox vaccina-
tion in susceptible individuals, hence AD or house-
hold AD contacts is a contraindication for smallpox
vaccination.
Molluscum contagiosum (MC) is a common childhood
cutaneous poxvirus infection that presents with single or
grouped skin‐coloured papules and is frequently seen in
children with AD (Fig.  15.23). Studies have reported an
increased incidence of MC with AD, with a recent cross‐
sectional study showing that 24% of children with MC
from a tertiary centre had AD [28].
Genetic variants involved in regulation of the immune
response in combination with genetic and acquired skin
barrier defects may increase the risk of viral infections;
staphylococcal toxins may also play a role in the dissemi-
nation of viral infections in AD [26].
Fig. 15.22  Perioral eczema herpeticum displaying grouped punched‐out
erosions with secondary impetiginization. Source: Courtesy of Professor Exfoliative dermatitis
Hywel C. Williams. Exfoliative dermatitis is a life‐threatening dermatosis that
rarely develops in AD (<1% of cases). In this condition,
intense erythema and peeling of more than 90% of the
Viral infections skin surface is accompanied by fever, systemic malaise
Infections with herpes simplex virus (HSV) are more and lymphadenopathy. This usually develops secondary
common in individuals with AD and can result in to either a bacterial infection (e.g. Staph. aureus) or eczema
eczema herpeticum (also known as Kaposi varicelliform herpeticum.
eruption). This is a serious but infrequent complication
occurring in 3% of AD patients. It is characterized by Differential diagnosis
disseminated HSV infection and associated with sig- Although AD is one of the most common causes of an
nificant morbidity, including multiorgan involvement, itchy rash in childhood, and in the vast majority of indi-
and rarely mortality. Clinically this presents with viduals the diagnosis is straightforward, it is important
grouped or widespread umbilicated vesicopustules and to consider other differential diagnoses. Table 15.1 lists
eroded vesicles (Fig.  15.22). This complication is usu- diagnoses that can present in a similar manner to AD.
ally secondary to HSV‐1 infection and recent studies A specific situation in which other differential diagnoses
suggest that there may be an association with filaggrin should be considered is when infants present with a
202 Section 3  Atopic Dermatitis and Related Disorders

Table 15.1  Differential diagnosis of atopic dermatitis (AD)

Other forms of dermatitis


Seborrhoeic dermatitis Red, shiny, relatively well‐demarcated eruption typically involving napkin (diaper) area in first 4 months of life. Lower abdomen,
of infancy neck and armpits may also be involved, along with scalp scaling (cradle cap). Non‐itchy – child happy but parents are not.
Good prognosis – clears within a few months
Adult‐type seborrhoeic Poorly defined erythema due to overgrowth/sensitivity to Pityrosporum yeasts in seborrhoeic areas, i.e. sides of nose, eyebrows,
dermatitis external ear canal, scalp, front of chest, axillae and groin creases
Discoid (nummular) Circular ‘cracked’ areas of erythema, 1–5 cm in diameter, on limbs initially, often with secondary infection (Fig. 15.20). In
eczema children, most commonly associated with AD and often confused with tinea (ringworm). In adults may be associated with
Staphylococcus infection and excessive skin dryness
Irritant contact Cumulative damage to the skin barrier from irritants such as soaps and detergents. Clinical appearance of dermatitis can be
dermatitis (ICD) identical to AD, but location at sites of maximal exposure, e.g. fingers, may be helpful. Some degree of ICD is common in AD
sufferers, e.g. around mouth in babies due to constant saliva and wet food, and in napkin area due to urine
ATOPIC DERMATITIS

Allergic contact A hypersensitivity reaction to specific substances to which individuals have been sensitized, such as nickel (jewellery), rubber
dermatitis (gloves) or glues (some shoes). Linear cut‐off and distribution may suggest diagnosis, but patch tests are needed to establish
SECTION 3:

diagnosis. May coexist with AD


Frictional lichenoid Shiny papules occurring in sites of repeated frictional trauma such as outer forearms in children. Probably quite common
dermatitis
Hyper‐IgE syndrome Features of AD within a few weeks of birth accompanied by staphylococcal infections of the skin, sinuses and lungs,
characteristic facies and abnormal dentition. Laboratory findings include high IgE and eosinophils
Wiskott–Aldrich The rash of Wiskott–Aldrich is identical to AD. It occurs usually in male infants in the first month of life and is accompanied by
syndrome petechiae, purpura and bloody diarrhoea
Other immunological A range of other immunological disorders may have eczematous features, including severe combined immunodeficiency,
disorders OMENN and IPEX syndromes
Inherited ichthyosis Inherited ichthyotic disorders may be associated with eczema and dry skin, including ichthyosis vulgaris, Netherton syndrome,
autosomal recessive congenital ichthyosis and X‐linked ichthyosis
Other inflammatory and Diverse inflammatory and bullous skin diseases including psoriasis, dermatitis herpetiformis, graft‐versus‐host disease, urticaria
bullous dermatoses pigmentosa and pityriasis alba may display features that might mimic eczema and the last may be associated with eczema
Other exogenous skin conditions
Scabies May produce nonspecific eczematous changes on the entire body. Burrows and pustules on palms, soles, genitalia and between
fingers help to establish diagnosis
Onchocercarial dermatitis In its chronic phase, may produce a widespread lichenification of the skin similar to that seen in chronic AD
Insect bites May often develop secondary eczematization and be confused with AD, especially on the limbs of children in tropical countries
Nutritional deficiencies A range of nutritional deficiencies can be associated with eczematous features, including zinc, pyridoxine, biotin, niacin,
phenylketonuria, lipid storage disease and cystic fibrosis
Tinea corporis May resemble eczema, particularly in acute florid phase where vesicles are present. Usually asymmetrical

severe eczematous rash with failure to thrive, recurrent 8 Borkar DS, Gonzales JA, Tham VM et al. Association between atopy
and herpetic eye disease: results from the pacific ocular inflammation
infections or petechiae [29]. In this scenario, it is impor-
study. JAMA Ophthalmol 2014;132:326–31.
tant to exclude immunodeficiency states. For example, 9 Wells R. Ichthyosis. Br Med J 1966;2:1504–6.
Omenn, IPEX (immune dysregulation, polyendo- 10 Smith F, Irvine A, Terron‐Kwiatkowski A et  al. Loss‐of‐function
crinopathy, enteropathy, X‐linked), Wiskott–Aldrich and mutations in the gene encoding filaggrin cause ichthyosis vulgaris.
Nat Genet 2006;38:337–42.
Job syndromes are associated with eczema. In children 11 Fartasch M, Diepgen T, Hornstein O. Atopic dermatitis – ichthyosis
from the Caribbean and Africa, severe infected AD may vulgaris – hyperlinear palms – an ultrastructural study. Dermatologica
be a  manifestation of human T‐lymphotropic virus‐1 1989;178:202–5.
(HTLV1) infection [30–32]. 12 Brenninkmeijer E, Spuls P, Legierse C et  al. Clinical differences
between atopic and atopiform dermatitis. J Am Acad Dermatol
2008;58:407–14.
13 Brown SJ, Relton CL, Liao H et  al. Filaggrin haploinsufficiency is
References highly penetrant and is associated with increased severity of eczema:
1 Taïeb AWD, Tilles G. The history of atopic eczema/dermatitis. In: Ring Further delineation of the skin phenotype in a prospective epidemio-
JPB, Ruzicka T (eds) Handbook of Atopic Eczema. Berlin: Springer, logical study of 792 school children. Br J Dermatol 2009;161:884–9.
2006:10–20. 14 Wuthrich B. Minimal variants of atopic eczema. In: Ring JPB, Ruzicka T
2 Rudikoff D, Lebwohl M. Atopic dermatitis. Lancet 1998;351:1715–21. (eds) Handbook of Atopic Eczema. Berlin: Springer, 2006:74–86.
3 Aoki T, Fukuzumi T. Atopy‐related skin rash in a normal population 15 Leung DY, Guttman‐Yassky E. Deciphering the complexities of
of infants: Distribution and rates for 50 separate skin regions: Cohort atopic dermatitis: shifting paradigms in treatment approaches.
study from 4 months to 3.5 years. J Dermatol 2015;42:453–60. J Allergy Clin Immunol 2014;134:769–79.
4 Williams HC, Forsdyke H, Pembroke AC. Infraorbital crease, ethnic 16 Garmhausen D, Hagemann T, Bieber T et  al. Characterization of
group and atopic dermatitis. Arch Dermatol 1996;132:51–4. different courses of atopic dermatitis in adolescent and adult patients.
5 Lobitz WJ, Dobson R. Physical and physiological clues for diagnosing Allergy 2013;68:498–506.
eczema. J Am Med Assoc 1956;161:1226–9. 17 Weidinger S, Rodríguez E, Stahl C et al. Filaggrin mutations strongly
6 Rim TH, Kim DW, Kim SE, Kim SS. Factors associated with cataract predispose to early‐onset and extrinsic atopic dermatitis. J Invest
in  Korea: a community health survey 2008‐2012. Yonsei Med J Dermatol 2007;127:724–6.
2015;56:1663–70. 18 Flohr C, England K, Radulovic S et  al. Filaggrin loss‐of‐function
7 Merdler I, Hassidim A, Sorkin N et  al. Keratoconus and allergic mutations are associated with early‐onset eczema, eczema severity
­diseases among Israeli adolescents between 2005 and 2013. Cornea and transepidermal water loss at 3 months of age. Br J Dermatol
2015;34:525–9. 2010;163:1333–6.
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 203

19 Brown SJ, Asai Y, Cordell HJ et  al. Loss‐of‐function variants in the systematic review of diagnostic criteria for AD conducted
filaggrin gene are a significant risk factor for peanut allergy. J Allergy
by Brenninkmeijer and colleagues [5].
Clin Immunol 2011;127:661–7.
20 van den Oord RA, Sheikh A. Filaggrin gene defects and risk of
developing allergic sensitisation and allergic disorders: systematic References
review and meta‐analysis. BMJ 2009;339:b2433. 1 Kendell RE. The Role of Diagnosis in Psychiatry. Oxford: Blackwell
21 Henderson J, Northstone K, Lee S et al. The burden of disease associ- Scientific, 1975;5.
ated with filaggrin mutations: a population‐based, longitudinal 2 Williams HC, Burney PGJ, Hay RJ et al. The UK Working Party’s diag-
birth cohort study. J Allergy Clin Immunol 2008;121:872–7. nostic criteria for atopic dermatitis I: Derivation of a minimum set of
22 Heede NG, Thyssen JP, Thuesen BH et  al. Anatomical patterns of discriminators for atopic dermatitis. Br J Dermatol 1994;131:383–96.
dermatitis in adult filaggrin mutation carriers. J Am Acad Dermatol 3 Williams HC, Burney PGJ, Strachan D, Hay RJ. The UK Working
2015;72:440–8. Party’s diagnostic criteria for atopic dermatitis II: Observer variation
23 Thyssen JP, Carlsen BC, Menné T et  al. Filaggrin null mutations of clinical diagnosis and signs of atopic dermatitis. Br J Dermatol
increase the risk and persistence of hand eczema in subjects with atopic 1994;131:397–405.
dermatitis: results from a general population study. Br J Dermatol 4 Williams HC, Burney PGJ, Pembroke AC, Hay RJ. The UK Working
2010;163:115–20. Party’s diagnostic criteria for atopic dermatitis III: Independent

ATOPIC DERMATITIS
24 Beck LA, Boguniewicz M, Hata T et al. Phenotype of atopic dermatitis hospital validation. Br J Dermatol 1994;131:406–16.
subjects with a history of eczema herpeticum. J Allergy Clin Immunol 5 Brenninkmeijer EEA, Schram M, Leeflang MMG et  al. Diagnostic
2009;124:260–9, 269. criteria for atopic dermatitis: a systematic review. Br J Dermatol

SECTION 3:
25 Lever R, Hadley K, Downey D, Mackie R. Staphylococcal colonization 2008;158:754–65.
in atopic dermatitis and the effect of topical mupirocin therapy. Br J
Dermatol 1988;119:189–98.
26 Ong PY, Leung DY. Bacterial and viral infections in atopic dermatitis: Nomenclature
a comprehensive review. Clin Rev Allergy Immunol 2016;51:329–37. Strictly speaking, the term ‘atopic dermatitis’ or its syn-
27 Gao P, Rafaels N, Hand T et al. Filaggrin mutations that confer risk of onymous term ‘atopic eczema’ should only be used to
atopic dermatitis confer greater risk for eczema herpeticum. J Allergy
Clin Immunol 2009;124:507–13. denote those with the phenotype of eczema who also
28 Dohil M, Lin P, Lee J et al. The epidemiology of molluscum contagio- have evidence of allergen‐specific circulating IgE anti-
sum in children. J Am Acad Dermatol 2006;54:47–54. bodies as demonstrated by high serum levels and/or
29 Hochreutener H, Wüthrich B, Huwyler T et al. Variant of hyper‐IgE
syndrome: the differentiation from atopic dermatitis is important
positive skin prick tests [1]. Yet evidence from the
because of treatment and prognosis. Dermatologica 1991;182:7–11. International Study of Asthma and Allergies in Childhood
30 La Grenade L, Manns A, Fletcher V et  al. Clinical, pathologic, and (ISAAC) Phase Two  –  the largest sample of AD cases,
immunologic features of human T‐lymphotrophic virus type I‐associ- defined according to physical examination, in the
ated infective dermatitis in children. Arch Dermatol 1998;134:439–44.
31 Purvis D. Guidelines for the diagnosis and assessment of eczema. world – suggests that around 50% of AD cases in devel-
DermNet NZ 2014. http://www.dermnetnz.org/topics/guidelines‐ oped countries are in fact not atopic, and an even greater
for‐the‐diagnosis‐and‐assessment‐of‐eczema/ (accessed 20 October proportion in developing countries are not atopic [2]. The
2018).
study concluded that any association between atopy and
32 Eichenfield LF, Tom WL, Chamlin SL et al. Guidelines of care for
the management of atopic dermatitis: section  1. Diagnosis and examined flexural eczema is weak and more variable than
assessment of atopic dermatitis. J Am Acad Dermatol 2014; previously suggested, and that the strength of this asso-
70:338–51. ciation is positively linked to gross national income. Some
researchers have suggested that there are two types of
AD  –  atopic (or extrinsic) and nonatopic (intrinsic or
Diagnostic criteria for atopic atopiform) [3,4]  –  but this division may have arisen in
dermatitis part by the fact that atopy is more common in people with
more severe disease, who typically make up the hospital‐
Although disease definition may sound a potentially
based populations featured in studies trying to separate
boring topic, it is a fundamental aspect of all compara-
atopic from nonatopic eczema. Indeed, some have even
tive knowledge that was summarized perfectly by
argued that raised IgE could be an epiphenomenon of
Kendell [1]:
disease severity [5]. It is not surprising therefore that
those who have mainly studied individuals with severe
It is probably more exciting for an architect to design para- disease have a firm belief that AD is atopic, whereas the
bolic canopies or baroque façades than it is to calculate the
reality is that nonatopic eczema can sometimes form the
size and shape of the concrete slab on which his building will
rest. But theories of causation and therapeutic claims have no
majority of cases in population surveys. It is also worth
more chance of surviving than buildings if they are not built on pointing out that nonatopic and atopic dermatitis are still
secure foundations. Developing reliable diagnostic criteria not clearly distinguishable without recourse to skinprick
may be as tedious as filling in muddy holes with concrete but tests or estimation of circulating IgE antibodies to com-
both provide the foundation on which all else depends. mon and relevant environmental allergens.
Kendell 1975 [1]. Reproduced with In order to overcome confusion about the correct use
permission of John Wiley & Sons of the term ‘atopy’ and different synonyms for AD, the
World Allergy Organization (WAO) nomenclature com-
Much of the evidence discussed in this section has mittee recommended that the term ‘eczema’ be used to
arisen from work that one of the authors (HW) led, that denote what we typically refer to as the phenotype of AD,
culminated in the UK refinement of Hanifin and Rajka’s and that the prefix atopy only be used when defining a
diagnostic criteria [2–4]. Because other criteria for AD subset that is truly atopic as indicated in Fig. 15.24 [1]. The
have also been proposed, more emphasis has been term eczema in the WAO scheme is quite specific to the
placed in this chapter on an independent and thorough AD phenotype, and clearly separated from other types of
204 Section 3  Atopic Dermatitis and Related Disorders

Dermatitis communication. The author uses the term ‘eczema’ in his


own publications and clinics as it is based on the most
scientific rationale to date, whereas he is comfortable
using the term ‘atopic dermatitis’ or ‘atopic eczema’ when
Other forms Contact talking to US or other European audiences respectively, as
Eczema
of dermatitis dermatitis
in this chapter. The author is not aware of any evidence
of harm caused by using these terms synonymously.
In future, better classification of dermatitis into various
subtypes may result in a scientific advancement beyond
the WAO classification. Trying to force the whole world to
adopt the term ‘atopic dermatitis’ (which may be neither
atopic nor dermatitis) is unlikely to help and may hinder
Atopic Nonatopic Allergic Nonallergic
harmonious international scientific endeavour to improve
ATOPIC DERMATITIS

eczema eczema CD CD
outcomes for patients with AD.
Fig. 15.24  World Allergy Organization revised classification for atopic Just for consistency with the rest of this chapter, we will
SECTION 3:

dermatitis showing that the term eczema is now the agreed term to use the term atopic dermatitis (AD) to refer to the clinical
replace the transitional term atopic eczema/dermatitis syndrome (AEDS). phenotype without implying that such people are truly
Atopic eczema is eczema in a person of the atopic constitution. Source:
atopic as per the more scientific rationale developed by
Johansson et al. 2004 [1]. Reproduced with permission of Elsevier.
the WAO.

dermatitis such as seborrhoeic, asteatotic, venous, num- References


mular and photosensitive (classified under ‘other forms 1 Johansson SGO, Bieber T, Dahl R et  al. Revised nomenclature for
of dermatitis’) and the two forms of contact dermatitis as allergy for global use. Report of the Nomenclature Review Committee
in Fig.  15.24. The WAO proposition makes good sense of the World Allergy Organization, October 2003. J Allergy Clin
Immunol 2004;113:832–6.
and it obviates the need for yet more sets of diagnostic 2 Flohr C, Weiland SK, Weinmayr G et  al.: the ISAAC Phase Two
criteria such as the millennium criteria [6], which look Study Group. The role of atopic sensitization in flexural eczema:
very similar to the original Hanifin and Rajka criteria [7] findings from the International Study of Asthma and Allergies in
Childhood Phase Two. J Allergy Clin Immunol 2008;121:141–7.
with IgE reactivity added as a necessary criterion.
3 Eedy DJ, Graham‐Brown RA. Atopiform dermatitis: what’s in a name?
Geographical variations in the use of names for AD Br J Dermatol 2002;147:415–17.
occur. ‘Atopic dermatitis’ is the most frequently used 4 Tokura Y. Extrinsic and intrinsic types of atopic dermatitis. J Dermatol
term used by clinicians in the USA, yet the term ‘eczema’ Sci 2010;58:1–7.
5 Flohr C, Johansson SGO, Wahlgren CF, Williams H. How ‘atopic’ is
is used by the National Eczema Association (https:// atopic dermatitis? J Allergy Clin Immunol 2004;114:150–8.
nationaleczema.org/eczema/) and in large population‐ 6 Bos JD, Van Leent EJ, Sillevis Smitt JH. The millennium criteria for
based surveys in the USA [8]. AD is also the most widely the diagnosis of atopic dermatitis. Exp Dermatol 1998;7:132–8.
used term in Japan and other Far Eastern countries such 7 Hanifin JM, Rajka G. Diagnostic features of atopic eczema. Acta
Dermatol Venereol (Stockh) 1980;92:44–7.
as Korea and Taiwan, whereas the phrase ‘atopic eczema’ 8 Kathuria P, Silverberg JI. Association of pollution and climate with
is used more commonly in Europe and just ‘eczema’ in atopic eczema in US children. Pediatr Allergy Immunol 2016;27:478–85.
the UK to denote AD. A review of AD nomenclature by 9 Kantor R, Thyssen JP, Paller AS, Silverberg JI. Atopic dermatitis, atopic
eczema, or eczema? A systematic review, meta‐analysis, and recommen-
Kantor et  al. found that of 33 060 relevant publications, dation for uniform use of ’atopic dermatitis’. Allergy 2016;71:1480–5.
64.4% used the term ‘atopic dermatitis’ and 46.9% used
the term ‘eczema’, with many papers using both terms
[9]. The authors (all of whom lived in countries where the Relative importance of sensitive versus
standard term was AD) strongly concluded that the term specific criteria in different study types
AD should be used in future studies. They claimed that It is easy to dive into the topic of diagnostic criteria for AD
the term eczema is a nonspecific one that denotes several in the belief that there may be a universal ‘one size fits all’
forms of skin inflammation, but this statement is simply definition, yet it is critical to think about the purpose of
not the case when the term eczema is used scientifically as the criteria before choosing which set of criteria or which
per the WAO classification in Fig. 15.24. It is worth point- version of the criteria to use. The need for a disease defini-
ing out that the purpose of a name is to increase commu- tion usually arises in the context of research or clinical
nication and understanding – whether this is with patients audit when groups of people need to be described and
and the public or in scientific communications. Insisting compared. In a population survey, for example, where the
that the phrase ‘atopic dermatitis’ should be used hence- main purpose is to estimate the true prevalence of AD,
forth in the UK (for example) for what is widely under- some individuals with AD will always be misclassified as
stood by the public as ‘eczema’ is likely to result in more not having AD and vice versa. Whatever criteria are used,
misunderstanding and possible harm, especially as the no single set of criteria can be perfect with 100% sensitivity
UK public’s understanding of the term ‘dermatitis’ has and specificity. When undertaking a prevalence survey
connotations of occupationally acquired hand eczema. therefore, the ideal criteria represent a trade‐off between
Much scientific energy has been wasted debating whether sensitivity (the proportion with true disease that is cor-
the term eczema or dermatitis should be used. The key rectly classified) and specificity (the proportion without
is to use the term that is most likely to result in clear AD that is correctly identified). Some misclassification is
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 205

Systematic

y
T or

ey e
r ic
Screening for

T t ic

r v nc
review of all

ho s t
RC nat

RC m a
c o gno

s u ale
Case-control possible

Pr t
a
possible

ag

ev
pl

o
study inclusion in study

Ex

Pr

Pr
studies of AD

Fig. 15.25  Schematic illustration of the different


requirements of the trade‐off between sensitivity
and specificity of diagnostic criteria for atopic
dermatitis in relation to different study designs.
AD, atopic dermatitis; RCT, randomized More specific More sensitive
controlled trial.

ATOPIC DERMATITIS
tolerable at a group level for the purpose of the study. The the positive predictive values (proportion of those who

SECTION 3:
requirements are different in a case‐control study that test positive who really have the disease) for the UK diag-
wishes to explore possible disease causes, because only nostic criteria vary according to underlying true disease
cases and a proportion of controls are needed. Here, spec- prevalence assuming a sensitivity of 80% and specificity
ificity of the criteria is critically important: criteria with of 97%. Even though the values for sensitivity and speci-
low sensitivity that may miss some true cases are toler- ficity sound quite good, the positive predictive value of
ated if the benefit is more certainty that those included 21% is very low when the true prevalence of disease is
cases and controls are accurately classified. Similarly, for only 1%. Yet it is much higher (90%) when the true popu-
clinical trials or genetic studies, it is also important that all lation prevalence is 25%, even though the sensitivity and
cases are ‘true cases’. In such circumstances, a two‐stage specificity parameters have remained constant. Guidance
process may be employed: cases might be defined first by on the effects of disease misclassification and which ver-
a clinician (in order to identify only ‘true cases’), followed sion of diagnostic criteria to use for different study
by a further filter of widely used criteria in order to char- designs is to be found elsewhere [1].
acterize the phenotype for comparison with other studies.
Cohort studies that follow up cases of AD to evaluate Including a measure of disease severity
prognosis must also be relatively specific in order not to When estimating disease prevalence, it is often important
overestimate disease clearance due to contamination with to include a measure of disease severity in order to deter-
non‐cases, but still have reasonable sensitivity so that mine disease burden and likely use of healthcare resources.
they are representative of most cases rather than a subset For example, Saeki et al. conducted a validation survey of
with more severe disease. The trade‐off between sensitiv- 2137 adults in Hokkaido and Osaka and found that of the
ity and specificity in relation to different study design 6.9% that were deemed to have AD on examination,
requirements is depicted in Fig. 15.25. 76.7%, 18.5%, 3.4% and 1.4% of those affected had mild,
moderate, severe and very severe disease respectively [2].
Effects of low disease prevalence The ISAAC global prevalence surveys of over 1 million
Another important point to consider if using diagnostic children used current eczema that was associated with
criteria to estimate disease prevalence is whether the sleep disturbance of one or more nights per week as a
disease is fairly common or rare. Table  15.2 shows how surrogate method of measuring more severe eczema [3].

Table 15.2  The effect of prevalence of atopic eczema (AE) on predictive values of the UK Diagnostic Criteria for Atopic Dermatitis, assuming a sensitivity
of 80% and specificity of 97%

True prevalence of AE Prevalence of AE by criteria Positive predictive value Negative predictive value

25% 22.2% 90.1 93.6


20% 18.4% 87.0 95.1
15% 14.5% 82.8 96.5
12% 12.2% 78.7 97.3
10% 10.7% 74.8 97.8
9% 9.9% 72.7 98.0
8% 9.2% 69.6 98.2
7% 8.4% 66.7 98.5
6% 7.6% 63.2 98.7
5% 6.8% 58.8 98.9
4% 6.1% 52.5 99.1
3% 5.3% 45.3 99.4
2% 4.5% 35.6 99.6
1% 3.8% 21.1 99.8
206 Section 3  Atopic Dermatitis and Related Disorders

Applying diagnostic criteria in the clinic insensibly into normality [1], but many clinicians and
There is a dilemma to be met when diagnostic criteria are researchers still have difficulties in viewing diseases as
applied to individuals in a clinical setting as these criteria continuous processes. Yet in a population setting, even for
were not developed for this purpose. It is important that diseases like scalp ringworm, which might at first appear
those who use the criteria in a clinical setting as a diag- to conform well to a dichotomous disease definition of
nostic aid realize that the criteria refer to probability of diseased or not diseased, one sees a gradation of sickness
disease, and that they should be used as a guide and not ranging from those who are apparently healthy (many of
a rigid rule. Thus it is possible to quote exceptional indi- whom are carriers of the responsible fungus or have sub-
viduals such as a child who has an itchy skin condition clinical infection) to those who have isolated patches of
that began at 3 years of age, who also has a history of hair loss and some who are quite ill with severe inflam-
flexural involvement and visible flexural dermatitis, but matory reactions affecting the entire scalp. Perhaps the
no history of dry skin or other atopic disease, who, most appropriate question therefore is not ‘Has he/she
although not fulfilling the UK diagnostic criteria or some got this disease: yes/no?’ but rather ‘How much of this
ATOPIC DERMATITIS

other criteria, might well have true AD. However, when disease does he/she have?’. Measuring the total amount
applied to a population of children where AD affects 12% of disease in a population on a quantitative scale may
SECTION 3:

of children, the probability that a child does not have AD if sound attractive in that it provides information on all of
he/she does not fulfil the criteria is over 97%. the individuals in that population, but it also presents
some serious difficulties in interpretation. It is important
Defining new cases to return to the main purpose of disease definition, i.e. to
Definition of an incident case is also a tricky area, because aid communication and increase our predictive abilities.
many definitions of AD incorporate having a chronically Whereas a log odds score of AD of 4.321 might mean
relapsing course as a component [4]. A systematic review something to a researcher trying to predict the degree to
of 102 prevention studies found that no definition of an which a hairdressing apprentice is likely to be incapaci-
incident case was given in 27 studies, and that the Hanifin tated by irritant hand dermatitis [2], such a score might
and Rajka criteria (which include disease chronicity as a have little utility to clinicians who wish to describe the
major criterion) had been used in 75 studies [5]. With the disease pattern in their population. Continuous scoring
advent of more birth cohort studies [6] and randomized methods for assessing the degree of AD are probably non-
controlled trials of disease prevention starting at birth [7], linear, i.e. double the score does not mean twice as much
there is a clear need for a uniform definition for an inci- AD, like doubling height and weight. In addition, the
dent case of AD that provides a good trade‐off between weights applied to individual disease features derived
not including all transient irritant eczematous eruptions from regression models are highly dependent upon the
in infancy and not excluding more transient forms of true population that was selected to derive the criteria, and 10
AD [8]. Such a definition has been proposed in a system- different studies could produce 10 different sets of weight-
atic review on this topic [5]. ing, leading to international disputes on which weighting
was ‘correct’. Others have expressed enthusiasm for
References revisiting quantitative methods for assessing AD [3].
1 Williams HC, Flohr C. So How Do I Define Atopic Eczema? A Practical Dichotomous or categorical disease definitions, on the
Manual for Researchers Wishing to Define Atopic Eczema. University other hand, require a line to be drawn between disease
of Nottingham, 1995 (https://www.nottingham.ac.uk/research/groups/
cebd/resources/uk‐diagnostic‐criteria‐for‐atopic‐dermatitis.aspx).
and non‐disease. Indeed the word ‘diagnosis’, which is
2 Saeki H, Oiso N, Honma M et al. Prevalence of atopic dermatitis in derived from the Greek words διá (the number two) and
Japanese adults and community validation of the U.K. diagnostic cri- γιγνώσκειν (to perceive), implies a dichotomous outcome.
teria. J Dermatol Sci 2009;55:140–1. Such dichotomous definitions are far more widely used
3 Odhiambo JA, Williams HC, Clayton TO et al.; ISAAC Phase Three Study
Group. Global variations in prevalence of eczema symptoms in children and easily understood in public health settings, and are
from ISAAC Phase Three. J Allergy Clin Immunol 2009;124:1251–8. therefore logical choices for promoting international com-
4 Hanifin JM, Rajka G. Diagnostic features of atopic eczema. Acta munication. Their main drawback is that the imposition
Dermatol Venereol (Stockh) 1980;92:44–7.
5 Simpson EL, Keck LE, Chalmers JR, Williams HC. How should an inci-
of boundaries between those who are sick and those who
dent case of atopic dermatitis be defined? A systematic review of primary are apparently healthy almost always results in the mis-
prevention studies. J Allergy Clin Immunol 2012;130:137–44. classification of some people. Unless the disease in ques-
6 Yang YW, Tsai CL, Lu CY. Exclusive breastfeeding and incident atopic tion has an abrupt natural cut‐off between normal and
dermatitis in childhood: a systematic review and meta‐analysis of pro-
spective cohort studies. Br J Dermatol 2009;161:373–83. abnormal, the imposition of an arbitrary dividing line
7 Simpson EL, Chalmers JR, Hanifin JM et al. Emollient enhancement of will always be subject to a trade‐off between sensitivity
the skin barrier from birth offers effective atopic dermatitis prevention. and specificity. On balance, a clinical approach of sum-
J Allergy Clin Immunol 2014;134:818–23.
8 Yates VM, Kerr RE, MacKie RM. Early diagnosis of infantile sebor-
marizing a constellation of symptoms and signs seems to
rhoeic dermatitis and atopic dermatitis – clinical features. Br J Dermatol be the most clinically relevant to the study of AD, as other
1983;108:633–8. groups have concluded [4].

Approaches to disease definition Progressive nosology


Binary versus continuous definitions It is worth noting that new discoveries about disease
Pickering et al. suggested as long ago as 1960 that essen- aetiology can lead to changes in disease classification.
tial hypertension was a graded characteristic that shaded The aetiology of AD is not yet fully understood, and
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 207

because the pursuit of an objective test has been fruitless Table 15.3  Between‐observer agreement for 18 signs in atopic dermatitis
to date, a clinical syndrome based on a constellation of from a specially designed repeatability study of UK consultant dermatologists
symptoms and signs remains the most appropriate start-
Substantial (kappa Truncal dermatitis
ing point currently. This statement does not imply that
>0.61)
this clinical syndrome could not be redefined in genetic
Moderate Flexural dermatitis, hand/foot dermatitis, facial
or aetiological terms in the future consistent with the
(kappa 0.41–0.60) dermatitis, hypopigmented patches, orbital fold,
concept of progressive nosology, such as the division of periorbital dermatitis, ear fissure, hyperlinear
‘pemphigus’, which formerly referred to several diseases palms
in which blistering was a feature, into pemphigoid, pem- Fair (kappa Follicular accentuation, periaural dermatitis,
phigus and linear IgA disease on the basis of immuno- 0.21–0.40) cheilitis
logical discoveries. Changes of this sort are not a problem Slight (kappa Keratosis pilaris, fine hair, periorbital pigmentation,
0.01–0.20) dry skin, extensor dermatitis (visible dermatitis)
providing they are explicit, and that they confer benefits
to patients. By analogy, what is recognized as a clinical

ATOPIC DERMATITIS
syndrome of AD today may in time be shown to be com-
posed of three or four different diseases such as a diffuse Repeatability

SECTION 3:
dry type of AD associated with filaggrin gene defects, a Between‐observer and within‐observer variation in
nummular form associated with defective Staph. aureus recording the diagnostic criteria should be kept to a mini-
handling, an early‐onset more severe form and an eczema/ mum. Agreement over dichotomous features, such as the
respiratory form, based on genetic, immunological or presence or absence of periorbital pigmentation, is usu-
aetiological discoveries [5,6]. This does not imply that the ally expressed in terms of a chance corrected measure
original older criteria were ‘wrong’ at the time, provided known as the kappa statistic, which usually varies from 0
they measured something useful or were instrumental (no better than chance) to 1 (perfect agreement), with val-
in stimulating further research into the aetiology of that ues of 0.6 or more indicating substantial agreement. As
syndrome. can be seen from the data on signs of AD recorded by UK
dermatologists in Table 15.3, even experienced physicians
References may exhibit poor agreement over classical features of a
1 Oldham PD, Pickering G, Fraser Roberts JA, Sowry GSC. The nature of skin disease [1]. Many minor signs, such as infraorbital
essential hypertension. Lancet 1960;i:1085–93. crease, can vary according to a number of factors, such
2 Fartasch M, Diepgen TL. Atopic diathesis and occupational derma-
toses. J Invest Dermatol 1994;102:620–1.
as the time of day (Fig.  15.26) or ethnic group [2].
3 Andersen RM, Thyssen JP, Maibach HI. Qualitative vs. quantitative Repeatability can be improved by keeping the number of
atopic dermatitis criteria – in historical and present perspectives. J Eur criteria small, and by clear instructions and training of
Acad Dermatol Venereol 2016;30:604–18. field workers. Although it is true that good validity usu-
4 Darsow U, Wollenberg A, Simon D et al. for the European Task Force
on Atopic Dermatitis/EADV Eczema Task Force. ETFAD/EADV ally implies good repeatability, the converse is not always
Eczema Task Force 2009 position paper on diagnosis and treatment of true; for example, two clocks that are both 10 minutes
atopic dermatitis. J Eur Acad Dermatol Venereol 2010;24:317–28. slow may well agree with each other, but neither is telling
5 Rodríguez E, Baurecht H, Herberich E et al. Meta‐analysis of filaggrin
the correct time.
polymorphisms in eczema and asthma: robust risk factors in atopic
disease. J Allergy Clin Immunol 2009;123:1361–70.
6 Mortz CG, Andersen KE, Dellgren C et  al. Atopic dermatitis from Acceptability to the population
adolescence to adulthood in the TOACS cohort: prevalence, persistence
Although it may be possible and appropriate for phy-
and comorbidities. Allergy 2015;70:836–45.
sicians to perform quite invasive tests on patients in a
hospital setting, even relatively noninvasive procedures
Key requirements of a disease definition such as examining the skin in covered areas or taking
for atopic dermatitis nail clippings may result in a low response rate when
Validity attempted in a population setting. The criteria should
Validity refers to the ability of a diagnostic test or set of also be easy and rapid to apply. Complicated and time‐
criteria to measure what it purports to measure. Validity, consuming procedures will lead to observer fatigue,
in the context of defining a case with a skin disease, has errors and lower response rates if conducted as part of a
two components: large study.
• sensitivity, i.e. the definition should catch as many
cases as possible Coherence with prevailing clinical concepts
• specificity, i.e. the definition should exclude as many Criteria should demonstrate a degree of face validity, i.e.
non‐cases as possible. they should contain features that clinicians identify as
Sensitivity and specificity are measured in relation to a key elements for the disease syndrome. It is here that the
­reference or ‘gold’ standard. Choosing a gold standard is seminal work of Hanifin and Rajka excels [3]. They sug-
relatively easy for some skin diseases such as malignant gested a list of major features and a long list of minor
melanoma where histology is used, but for many skin features that characterize the clinical syndrome of AD
diseases (including AD) no such objective test is available. based on clinical experience. Although the long list is too
In these circumstances, diagnosis by dermatologist may cumbersome and imprecisely defined to be used as a
serve as a clinical gold standard, providing dermatologists research instrument, the features listed have formed a
can be shown to agree on what constitutes a typical case [1]. good basis for subsequent scientific refinements such as
208 Section 3  Atopic Dermatitis and Related Disorders
ATOPIC DERMATITIS
SECTION 3:

Fig. 15.26  Images of the same atopic person


taken 12 hours apart. Although signs such as
infraorbital crease (defined as one or more
prominent creases that extend beyond the midline
when the pupil gazes forward) may sound
straightforward to ascertain, these images show
how unreliable such a sign can be.

the UK Working Party’s minimum list of reliable criteria [4]. scientifically. Many groups have suggested diagnostic
It is also worth pointing out that diagnostic criteria for criteria based on a hunch or consensus but the validity of
AD should also reflect some degree of morbidity; for many diagnostic criteria remains untested. It is also worth
example, it may be possible to develop criteria that noting that instead of focusing on the really important
measure very minor degrees of eczematous inflamma- questions about the diagnostic criteria for AD, i.e. which
tion, but if those identified by such criteria are not itchy criteria are the best discriminators and which can be lost
or bothered by those features, then it is questionable in order to make a minimum list of reliable ones, lots of
whether such a disease is worth studying. This is the studies have focused on the long list of minor features
main reason for making ‘an itchy skin condition’ a neces- and whether or not they apply to a particular study popu-
sary criterion for the UK diagnostic criteria. lation [1–3]. Although these studies are helpful in show-
ing some possible variation in the minor phenotypic
Comprehensiveness features that may be more or less prevalent in some coun-
Criteria need to be applicable to a wide range of groups tries and ethnic groups, they add little to the main quest
such as children and adults, those with dark skins (where for a minimum list of reliable and valid major criteria that
erythema may be less obvious) and different ethnic permit international comparisons.
groups who may have a different cultural understanding At least 10 different sets of diagnostic criteria for
of terms like ‘itching’, especially when compounded by AD  had been proposed by 2007 [4–13], prompting
difficulties in translation [5]. Brenninkmeijer and colleagues to undertake a system-
atic review of the validity of such studies [14]. Following
References an extensive search of three databases and citations from
1 Williams HC, Burney PGJ, Strachan D, Hay RJ. The UK Working retrieved papers, they identified 27 validation studies
Party’s diagnostic criteria for atopic dermatitis II: Observer variation
of clinical diagnosis and signs of atopic dermatitis. Br J Dermatol
that were then quality‐assessed by two independent
1994;131:397–405. data extractors using the Quality Assessment of
2 Williams HC, Forsdyke H, Pembroke AC. Infraorbital crease, ethnic Diagnostic Accuracy (QUADAS) tool, which assesses
group and atopic dermatitis. Arch Dermatol 1996;132:51–4. the risk of bias by means of 14 different items [15]. The
3 Hanifin JM, Rajka G. Diagnostic features of atopic eczema. Acta
Dermatol Venereol (Stockh) 1980;92:44–7. authors also compared sensitivity and specificity param-
4 Williams HC, Burney PGJ, Pembroke AC, Hay RJ. The UK working eters and optimum cut‐offs for different criteria using
party’s diagnostic criteria for atopic dermatitis III: Independent receiver‐operator curves. They found that some of the
hospital validation. Br J Dermatol 1994;131:406–16.
proposed diagnostic criteria such as those of the Japanese
5 Chalmers DA, Todd G, Saxe N et  al. Validation of the U.K. Working
Party diagnostic criteria for atopic eczema in a Xhosa‐speaking African Dermatological Association, the Danish Allergy Research
population. Br J Dermatol 2007;156:111–16. Centre criteria and the Lillehammer criteria had not
undergone any form of validation at all. The original
A systematic review of diagnostic criteria Hanifin and Rajka criteria had only been validated in
for AD two hospital studies, those of Schultz Larsen in two
The last section highlighted the need for a short list of studies, those of Diepgen et  al. and those of Kang and
reliable and valid criteria that can be used widely. Very Tian in one study each, and the UK refinement of Hanifin
few studies have developed and tested diagnostic criteria and Rajka’s criteria in 19 studies.
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 209

Overall, sensitivity and specificity of the various criteria


Box 15.1  Attributes of a good validation study for diagnostic
ranged from 10% to 95% and from 77.6% to 100%, respec-
criteria
tively, in hospital‐based studies, and from 42.8% to 100%
and from 44.7% to 96.6%, respectively, in population 1. Use the criteria questions and examination protocol exactly as
studies. The two hospital‐based studies that evaluated recommended.
the Hanifin and Rajka criteria showed a sensitivity of 2. Ensure that translation follows a strict format.
87.9% and 96.0% respectively, and specificity of 77.6% and 3. Test the criteria in the setting in which they are intended to be used.
93.8%. The eight hospital‐based studies validating the UK 4. Ensure that those acting as assessors or as the clinical criterion
refinement of the Hanifin and Rajka criteria showed a standard are blinded to the criteria being tested.
sensitivity ranging from 10% to 95.5% and specificity 5. Ensure that the clinician who is the clinical criterion standard is
from 90.4% to 98.3%. The 13 population‐based validation comparable to other clinicians through interobserver repeatability
studies showed a sensitivity ranging from 42.8% to 100% testing.
and specificity from 89.3% to 99.1%. No meta‐analytic

ATOPIC DERMATITIS
Source: Williams 1999 [22]. Reproduced with permission of the American
summary estimates could be given due to the major Medical Association.
differences between studies in terms of participants,

SECTION 3:
setting and study design. Validity was particularly poor
in one community validation study of the UK criteria that References
had been translated into the Xhosa language [16], 1 Mevorah B, Frenk E, Wietlisbach V et  al. Minor clinical features
of  atopic dermatitis. Evaluation of their diagnostic significance.
although other studies of translated versions in China, Dermatologica 1988;177:360–4.
Italy and Romania seemed to show good results [17–19], 2 Nagaraja, Kanwar AJ, Dhar S et  al. Frequency and significance of
suggesting that cultural factors rather than just translation minor clinical features in various age‐related subgroups of atopic
dermatitis in children. Pediatr Dermatol 1996;13:10–13.
issues are important. One study from Ethiopia showed
3 Hiletework M. Evaluation of Hanifin and Rajka atopic eczema
that the questionnaire used in the ISAAC showed a posi- diagnostic guidelines for reduced minor criteria. Ethiop Med J
­
tive predictive value of 48.8% and a negative predictive 2009;47:39–47.
value of 91.1% [20]. 4 Asher MI, Keil U, Anderson HR et al. International Study of Asthma
and Allergies in Childhood (ISAAC): rationale and methods. Eur
The methodological quality of the validation studies Respir J 1995;8:483–91.
varied considerably, making comparisons difficult. 5 Bos JD, Van Leent EJ, Sillevis Smitt JH. The millennium criteria for the
Common methodological problems included an inap- diagnosis of atopic dermatitis. Exp Dermatol 1998;7:132–8.
propriate reference standard, lack of blinding of those 6 Diepgen TL, Sauerbrei W, Fartasch M. Development and validation of
diagnostic scores for atopic dermatitis incorporating criteria of data
determining the index test results from the reference test quality and practical usefulness. J Clin Epidemiol 1996;49:1031–8.
results, lack of reporting of intermediate or uninterpreta- 7 Hanifin JM, Rajka G. Diagnostic features of atopic dermatitis. Acta
ble results, and failure to provide details on participant Derm Venereol Suppl (Stockh) 1980;92:44–7.
8 Johnke H, Vach W, Norberg LA et al. A comparison between criteria for
withdrawals. The authors of the systematic review also diagnosing atopic eczema in infants. Br J Dermatol 2005;153:352–8.
point to problems such as the reference standard of 9 Schultz Larsen F, Hanifin JM. Secular change in the occurrence of
clinical diagnosis residing with just one clinician whose atopic dermatitis. Acta Derm Venereol Suppl (Stockh) 1992;176:7–12.
perception of what constitutes AD might be very different 10 Schultz Larsen F, Diepgen T, Svensson A. Clinical criteria in diagnos-
ing atopic dermatitis: the Lillehammer criteria 1994. Acta Derm
from others [21,22]. Another problem when validating Venereol Suppl (Stockh) 1996;96:115–19.
criteria that refer to symptoms over the course of a year 11 Williams HC, Burney PG, Hay RJ et al. The U.K. Working Party’s
(to overcome seasonal variations when making compar- Diagnostic Criteria for Atopic Dermatitis. I. Derivation of a mini-
mum set of discriminators for atopic dermatitis. Br J Dermatol
isons) was that they were often validated by clinical
1994;131:383–96.
examination at just one point in time, a procedure that 12 Kang KF, Tian RM. Criteria for atopic dermatitis in a Chinese popula-
will always underestimate genuine cases that are not tion. Acta Derm Venereol Suppl (Stockh) 1989;144:26–7.
active at the time of examination. The authors concluded 13 Tagami H. Japanese Dermatology Association criteria for the diagno-
sis of atopic dermatitis. J Dermatol 1995;22:966–7.
that the UK diagnostic criteria are the most extensively 14 Brenninkmeijer EEA, Schram M, Leeflang MMG et al. Diagnostic
validated in hospital and community settings, and that criteria for atopic dermatitis: a systematic review. Br J Dermatol
this set of criteria for AD should be recommended in 2008;158:754–65.
future intervention studies. They pointed out that the 15 Whiting P, Rutjes AW, Reitsma JB et al. The development of QUADAS:
a tool for the quality assessment of studies of diagnostic accuracy
ideal set of diagnostic criteria still has to be established, included in systematic reviews. BMC Med Res Methodol 2003;3:25.
and there is considerable scope for improvement of meth- 16 Chalmers DA, Todd G, Saxe N et al. Validation of the U.K. Working
odological design for validation studies. Party diagnostic criteria for atopic eczema in a Xhosa‐speaking
African population. Br J Dermatol 2007;156:111–16.
Guidance on how to design a good validation study for 17 Gu H, Chen XS, Chen K et  al. Evaluation of diagnostic criteria for
diagnostic criteria for AD is summarized in Box 15.1 [22] atopic dermatitis: validity of the criteria of Williams et al. in a hospital‐
although it has rarely been followed. Guidance on what based setting. Br J Dermatol 2001;145:428–33.
makes a good prevalence study has been suggested by 18 Girolomoni G, Abeni D, Masini C et al. The epidemiology of atopic
dermatitis in Italian schoolchildren. Allergy 2003;58:420–5.
Radalescu et al. [23]. An interactive website is available in 19 Popescu CM, Popescu R, Williams H et al. Community validation of
the public domain for those interested in finding out more the United Kingdom diagnostic criteria for atopic dermatitis in
about using the UK Working Party’s diagnostic refine- Romanian school children. Br J Dermatol 1998;138:436–42.
20 Haileamlak A, Lewis SA, Britton J et al. Validation of the International
ment of the Hanifin and Rajka criteria, including transla- Study of Asthma and Allergies in Children (ISAAC) and U.K. criteria
tions, detailed field instructions, training images and a for atopic eczema in Ethiopian children. Br J Dermatol 2005;
quality control test [24]. 152:735–41.
210 Section 3  Atopic Dermatitis and Related Disorders

21 Firooz A, Davoudi SM, Farahmand AN et al. Validation of the diag- however, that the original instructions on the use of the
nostic criteria for atopic dermatitis. Arch Dermatol 1999;135:514–16.
UK diagnostic criteria in adults in the online manual
22 Williams HC. Defining atopic dermatitis  –  where do we go from
here? Arch Dermatol 1999;135:583–6. clearly state ‘Recall of onset of eczema under the age of 2
23 Radalescu M, Diepgen T, Williams H. What makes a good prevalence is likely to be inaccurate in adults’, and should be replaced
survey? In: Williams HC, Bigby M, Diepgen T et al. (eds) Evidence‐ by ‘Did your eczema start when you were a child?’ as few
Based Dermatology, 2nd edn. Oxford: Blackwell Publishing and BMJ
Publishing Group, 2008:61–7.
adults will know what happened in the first 2 years of
24 Williams HC, Flohr C. So How Do I Define Atopic Eczema? A Practical their life [5]. Another study of 518 children in Spain found
Manual for Researchers Wishing to Define Atopic Eczema. University of that the UK criteria administered in a telephone interview
Nottingham, 1995 (https://www.nottingham.ac.uk/research/groups/ agreed with physician diagnosis in 75.3% of cases [6].
cebd/resources/uk‐diagnostic‐criteria‐for‐atopic‐dermatitis.aspx).
Within Phase Two of the ISAAC, 30 358 schoolchildren
aged 8–12 years from 18 countries were examined for
Other studies that have emerged since flexural eczema and their parents completed the ISAAC
the systematic review eczema symptom questionnaire [7]. As might be expected,
ATOPIC DERMATITIS

A few other relevant validation studies have emerged the point prevalence for flexural eczema based on a single
since the systematic review by Brenninkmeijer et al. [1]. examination was lower than the questionnaire‐based 12‐
SECTION 3:

The first is a study of validation of the UK diagnostic cri- month period prevalence (mean centre prevalence 3.9%
teria in adults in Japan [2] – a useful study as nearly all vs. 9.4%), yet both were highly correlated (r = 0.77,
work to date has involved children. Saeki et al. compared P  <0.001). When analysed at an individual level, ques-
the validity of the UK criteria with clinical examination tionnaire‐derived symptoms of ‘persistent flexural
[2]. Such a comparison will always underestimate the true eczema in the past 12 months’ missed less than 10% of
validity of the criteria because a 1‐year period prevalence cases of flexural eczema detected on physical examina-
is being compared against point prevalence. In other tion, although between 33% and 100% of questionnaire‐
words, many of those who were ‘positive’ for the UK based symptoms of ‘persistent flexural eczema in the past
criteria in the last year may not have had active eczema at 12 months’ were not confirmed on examination. The
the time of examination. Nevertheless, the study found authors concluded that prevalences derived using the
that the UK criteria showed a sensitivity of 68.8% (88/128) ISAAC questionnaires were sufficiently robust for com-
and specificity of 93.5% (1863/1992) in an adult Japanese paring disease prevalence between populations. They
population attending college and university. In an earlier went on to say that when diagnostic precision at the indi-
study of community validation of diagnostic criteria for vidual level is important, questionnaires should always
AD in schoolchildren, Saeki and colleagues illustrated be validated first, or a standardized skin examination
how validity of the UK criteria can appear to improve if protocol should be used such as that used by the UK
the time period for assessing symptoms is restricted to the working party (see Fig. 13.1) [8].
last week to make them more like a point prevalence One study has tried to see whether there are differences
measure, which could then be more realistically com- in diagnostic features in children with AD (i.e. truly atopic
pared with a clinical examination at one point in time [3]. with IgE antibodies) versus those who look similar but
They found in 2002 that when the UK diagnostic criteria who do not have evidence of IgE reactivity (called ‘atopi-
were used as a 1‐year prevalence measure, sensitivity and form’ by some). The authors found that the atopiform
specificity were 71.8% and 89.3%, respectively, with a children had later disease onset, an absence of other atopic
positive predictive value of 44.7%. When used as a point diseases, fewer Dennie–Morgan creases, less hand/foot
prevalence estimate in 2004/5 in a different sample, eczema and a variety of other features, yet some of these
sensitivity had dropped slightly to 58.9% but specificity differences could be due to the fact that the atopiform
had increased to 95.4%, which led to an increase in the cases were milder, an important confounder that was not
positive predictive value to 59.9% [3]. taken into account in the selection of cases or analysis [9].
A further validation study concerning adults was Schram and colleagues in 2011 revisited the millennium
conducted on 1131 nursing staff in a Taiwanese teaching criteria for AD for which allergen‐specific IgE is a manda-
hospital; this achieved a good 93% response rate [4]. The tory criterion, accompanied by three additional criteria,
Taiwanese study compared the validity of the UK criteria two of which need to be positive [10]. They removed the
and the ISAAC criteria in the last year against a derma- requirement for allergen‐specific IgE and derived a set of
tologist’s opinion of whether the person had AD in the clinical predictors for AD based on a sample of 210 clinic
last year (using guidance that broadly followed the patients with a clinical diagnosis of AD. They then applied
Hanifin and Rajka list). They found that sensitivity and these criteria to the same patients from which they were
specificity of the UK criteria were 42.2% and 99.6%, derived and reintroduced specific IgE testing to further
respectively, and for the ISAAC criteria they calculated a subdivide them into truly atopic dermatitis and those
sensitivity and specificity of 36.7% and 92.9%, respec- who had the AD phenotype but who were not atopic
tively. Further analysis by means of receiver‐operator (which they called atopiform dermatitis). They found that
curves indicated that dropping the criterion of ‘onset the refined millennium criteria (without IgE requirement)
under the age of 2 years’ resulted in better discrimination had a sensitivity and specificity of 81.8% and 98.8% respec-
for the UK criteria (sensitivity and specificity of 82.2% tively, compared with corresponding values of 97.7% and
and 94.2%, respectively). It should be pointed out, 72.9% for the UK refinement of the Hanifin and Rajka
Chapter 15  Clinical Features and Diagnostic Criteria of Atopic Dermatitis 211

criteria, and 100% and 48.8% for the original Hanifin and References
1 Brenninkmeijer EEA, Schram M, Leeflang MMG et al. Diagnostic
Rajka criteria. Relative value (sensitivity plus specificity
criteria for atopic dermatitis: a systematic review. Br J Dermatol
minus 100) was 0.7 for the revised millennium criteria, 2008;158:754–65.
0.72 for the UK criteria and 0.51 for the Hanifin and 2 Saeki H, Oiso N, Honma M et al. Prevalence of atopic dermatitis in
Rajka criteria for phenotypic AD. The study was well Japanese adults and community validation of the U.K. diagnostic
criteria. J Dermatol Sci 2009;55:140–1.
reported, yet it tested the criteria on the same popula- 3 Saeki H, Iizuka H, Mori Y et  al. Community validation of the UK
tion from which they were derived, a procedure that diagnostic criteria for atopic dermatitis in Japanese elementary
may overestimate test performance. Further independent schoolchildren. J Dermatol Sci 2007;47:227–31.
studies are awaited. 4 Lan CC, Lee CH, Lu YW et al. Prevalence of adult atopic dermatitis
among nursing staff in a Taiwanese medical center: a pilot study
Silverberg and colleagues in a US study in 2015 found on  validation of diagnostic questionnaires. J Am Acad Dermatol
that self‐ and caregiver‐reported diagnosis of eczema in a 2009;61:806–12.
hospital clinic setting was good when compared with 5 Williams HC, Flohr C. So How Do I Define Atopic Eczema? A Practical
Manual for Researchers Wishing to Define Atopic Eczema. University
physician diagnosis of AD, with positive predictive val-

ATOPIC DERMATITIS
of Nottingham, 1995 (https://www.nottingham.ac.uk/research/groups/
ues of 0.87 and 0.76 for caregiver 1‐year reported history cebd/resources/uk‐diagnostic‐criteria‐for‐atopic‐dermatitis.aspx);
of childhood eczema and self‐report of eczema in the last Section 5‐3.

SECTION 3:
year by adults, respectively [11]. The report is well‐­written 6 García‐Díez A, Puig L, Ortiz J, Blanco A. [Validity of a telephone
survey for determining the prevalence of atopic dermatitis and its
but their use of the term ‘eczema’ is at odds with a call seasonal variation in Spain]. Actas Dermosifiliogr 2009;100:298–306.
from the same group not to use such a term [12]. 7 Flohr C, Weinmayr G, Weiland SK et al. and the ISAAC Phase Two
Yet another set of criteria has been suggested by the Study Group. How well do questionnaires perform compared with
physical examination in detecting flexural eczema? Findings from
Korean Atopic Dermatitis Association’s atopic dermati-
the International Study of Asthma and Allergies in Childhood
tis diagnostic criteria group. This set is composed of 11 (ISAAC) Phase Two. Br J Dermatol 2009;161:846–53.
questions (two major and nine minor), many of which 8 Williams HC, Forsdyke H, Boodoo G et al. A protocol for recording
are remarkably similar to those used in the UK criteria the sign of visible flexural dermatitis. Br J Dermatol 1995;133:941–9.
9 Brenninkmeijer EE, Spuls PI, Legierse CM et al. Clinical differences
[13]. They derived their criteria from a training sample between atopic and atopiform dermatitis. J Am Acad Dermatol 2008;
(n = 1129) and tested them on an independent commu- 58:407–14.
nity sample of children (n = 1191) and found a sensitivity 10 Schram ME, Leeflang MM, DEN Ottolander JP et  al. Validation
and specificity of 75.2% and 96.1% respectively when and  refinement of the Millennium Criteria for atopic dermatitis.
J Dermatol 2011;38:850–8.
compared with dermatologist examination performed 11 Silverberg JI, Patel N, Immaneni S, Rusniak B et al. Assessment of
twice over the course of a year. These values were better atopic dermatitis using self‐report and caregiver report: a multicentre
than corresponding values for the simple questions used validation study. Br J Dermatol 2015;173:1400–4.
12 Kantor R, Thyssen JP, Paller AS, Silverberg JI. Atopic dermatitis,
in the ISAAC (sensitivity of 68.8 and specificity of atopic eczema, or eczema? A systematic review, meta‐analysis, and
92.9%), although it is unclear if these values were statis- recommendation for uniform use of ’atopic dermatitis’. Allergy
tically different. The Korean criteria were not compared 2016;71:1480–5.
against the UK or millennium or Hanifin and Rajka cri- 13 Lee SC, Bae JM, Lee HJ et al.; Korean Atopic Dermatitis Association’s
Atopic Dermatitis Criteria Group. Introduction of the reliable estimation
teria in this study and further independent studies are of atopic dermatitis in childhood: novel, diagnostic criteria for childhood
awaited. atopic dermatitis. Allergy Asthma Immunol Res 2016;8:230–8.
212 

CHA PTER  1 6

Severity Scoring and Quality of Life Assessment


in Atopic Dermatitis
Christian Apfelbacher1, Cecilia A.C. (Sanna) Prinsen2, Daniel Heinl1 & Hywel C. Williams3
1
 Medical Sociology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
2
 VU University Medical Center, Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
3
 The Centre of Evidence Based Dermatology, University of Nottingham, Nottingham, UK
ATOPIC DERMATITIS

Introduction, 212 Global measurement instruments of atopic Evaluation of quality‐of‐life instruments


SECTION 3:

How has disease severity and quality of life dermatitis severity, 218 for children with atopic dermatitis, 222
been measured in clinical trials in atopic Other ways of measuring severity of atopic Towards standardization of outcome
dermatitis?, 213 dermatitis, 218 reporting for quality of life in atopic
Choosing adequate measurement Evidence‐based recommendation for the dermatitis, 224
instruments, 213 measurement of subjective severity of atopic Instruments to measure family impact of
Evidence‐based recommendations for dermatitis (symptoms) in clinical trials, 219 atopic dermatitis, 224
the measurement of atopic dermatitis Measuring quality of life in children with Conclusion, 225
severity in clinical trials, 215 atopic dermatitis, 219

Abstract the Patient‐Oriented Eczema Measure (POEM) for symptoms


are the preferred instruments to be measured in all clinical trials
In the absence of a reliable biomarker, instruments to measure in AD. There is no well‐validated way to measure AD disease
AD disease severity are questionnaires, which are completed by ­severity by a global assessment. The Childhood Atopic Dermatitis
patients, significant others or physicians. There is substantial Impact Scale (CADIS) is a promising proxy instrument to meas-
heterogeneity in the way in which objective disease severity ure quality of life in infants and their family with one s­ ingle
(i.e. clinical signs), subjective disease severity (i.e. symptoms) ­instrument. Valid and feasible ways of measuring disease s­everity
and quality of life are measured. The Harmonising Outcome and quality of life in clinical practice are unclear; guidance
Measures for Eczema (HOME) initiative has reached the consen- should be sought from the recommendations for clinical trials
sus that the Eczema Area and Severity Index (EASI) for signs and for the time being.

Key points trials. The SCORing Atopic Dermatitis (SCORAD) index also has
satisfactory measurement properties.
°° For the symptoms domain, the Patient‐Oriented Eczema
• There is substantial heterogeneity in the way in which objective
Measure (POEM) is the preferred instrument to be used in AD
disease severity (clinical signs), subjective disease severity
trials.
(symptoms) and quality of life are measured in AD.
°° For quality of life, the Childhood Atopic Dermatitis Impact Scale
• The Harmonising Outcome Measures for Eczema (HOME)
(CADIS) and the Dermatitis Family Impact (DFI) questionnaire can
initiative has reached international consensus that the core
be used.
outcome set for clinical trials in AD includes clinical signs,
• There is no international consensus regarding the measurement
symptoms, long‐term control and quality of life:
of disease severity and quality of life in clinical practice.
°° For the clinical signs domain, the Eczema Area and Severity
Index  (EASI) is the preferred instrument to be used in AD

questionnaires) [2]. The latter are also referred to as


Introduction patient‐reported outcome measures (PROMs). Measuring
In the absence of an accepted, reliable biomarker for disease severity by physician assessment of visible clini-
measuring atopic dermatitis (AD or atopic eczema, or cal signs is often referred to as ‘objective’ whereas patient
eczema) severity [1], consistent and replicable assessment self‐assessment of symptoms or quality of life is referred
of AD disease severity remains a clinical activity that to as ‘subjective’. However, it should be realized that even
requires questionnaires. These are completed either by though the word ‘objective’ implies an accurate measure-
physicians, nurses or relatives (interviewer‐administered ment of an outcome on a numeric scale, all such objective
questionnaires) or by patients themselves (self‐administered measures require subjective judgement by observers, e.g.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 16  Severity Scoring and Quality of Life Assessment in Atopic Dermatitis 213

grading the intensity of erythema as mild, moderate or the SCORing of Atopic Dermatitis (SCORAD) index [8]
severe, or guessing the amount of surface area involved. and the Simple Scoring System (SSS) [9]. It illustrates the
Therefore, the notion that there is a very clear difference variation in the way the construct ‘disease severity’ is
in objectivity between so‐called objective clinician‐ interpreted in disease severity assessments. Of note, some
assessed and subjective patient‐assessed measures is a instruments include symptoms in their conceptual model
fallacy. Conceptually, it is better to think about who is and some do not. Four further systematic reviews
making the assessment and how it is being done. [4,5,10,11] have been conducted following the cited origi-
nal review by Charman et  al. [3], all of which corrobo-
rated the findings of the original systematic review that
there is wide variation in the way in which disease sever-
How has disease severity and quality
ity and quality of life have been measured.
of life been measured in clinical trials
Concerning subjective disease severity, another system-
in atopic dermatitis?
atic review investigated how symptoms were measured

ATOPIC DERMATITIS
A systematic review investigated the use of outcome in RCTs performed in AD between 2000 and 2014 and
measurement instruments used in trials of therapeutic likewise found substantial variation [12]. Seventeen dif-

SECTION 3:
interventions for AD conducted between 1994 and 2001 ferent symptoms (most frequently itch and sleep loss)
[3]. It was found that 85 of the included 93 studies used an were reported in 78% of the investigated 378 trials, with
objective measurement of clinical signs, but only 23 used only 37% of the trials reporting stand‐alone symptom
a published severity scale. Overall, 56 different scales measurements. The remainder used composite measure-
were identified and there was no consensus on which fea- ment instruments such as SCORAD. SCORAD was also
tures should be assessed: 31 different descriptions of the most frequently used instrument, but only 23% of
clinical signs were used across all scales. Erythema, RCTs reported the part C symptom score separately.
lichenification and excoriation were most frequently Overall, 30 different instruments were identified.
assessed. Table  16.1 shows the contents of the most fre- Similar observations concerning the significant varia-
quently used AD disease severity measurement instru- tion in measurement instruments used can be made with
ments: the Eczema Area and Severity Index (EASI) [6], the respect to quality of life (QoL). A 2016 systematic review
Six Area, Six Sign Atopic Dermatitis (SASSAD) score [7], examined how QoL was captured in RCTs performed in
AD between 2000 and 2014 [13]. Only 63 of 303 included
studies assessed QoL, using 22 different QoL measure-
Table 16.1  Content comparison of selected, frequently used measure- ment instruments. This finding is also in accordance with
ment instruments of atopic dermatitis (AD) disease severity an earlier review looking at the time period of 1985–2010,
which found that 14 different QoL instruments were used
AD disease severity measurement in AD trials [10].
instrument The variation identified in the measurement of clinical
signs, symptoms and QoL means that the comparison and
EASI SASSAD SCORAD SSS meta‐analysis of efficacy studies within systematic
reviews and meta‐analyses is hampered at best and
Clinical signs
impossible at worst. Consequently, it is difficult to iden-
Erythema X X X X
Oedema/induration/ X X X tify the most efficacious therapies [14], which in turn
papulation means that recommendations from clinical research for
Oozing/crusting X X X clinical practice are difficult to make and evidence‐based
Excoriation X X X X healthcare for people with AD cannot be implemented. In
Lichenification X X X X other words, the failure to identify and agree what to
Dryness X X
measure and how to measure AD severity ultimately
Scaling X
Cracking/fissuring X
results in poor patient information and care.
Vesicles X
(De)pigmentation X
Affected body regions Choosing adequate measurement
Extent of affected skin X X instruments
areas (in %)
Inspection of defined x X In order to make an evidence‐based decision on the choice
body areas (e.g. arms) for a specific measurement instrument of AD disease
Symptoms severity or QoL, it is important to compare the quality of
Pruritus x X existing instruments. The measurements generated by
Sleep x X AD severity measurement instruments should, as indeed
should any measurement, be robust and the scores gener-
EASI, Eczema Area and Severity Index; SASSAD, Six Area, Six Sign Atopic
ated will be the more trustworthy the higher the quality
Dermatitis; SCORAD, SCORing Atopic Dermatitis; SSS, Simple Scoring
System.
of the instrument used.
Source: adapted from Deckert et al. 2015 [2], Schmitt, Langan and Williams Generally, systematic reviews of measurement instru-
2007 [4], Schmitt et al. 2013 [5]. Reproduced with permission of Elsevier. ments are important tools that can be used to select the
214  Section 3 Atopic Dermatitis and Related Disorders

best instrument available. In such reviews, the content intensity of these signs and not of another ­combination of
and the measurement properties of all instruments signs. Reliability relates to the precision of measurements.
available are being critically appraised and compared. If two different dermatologists using the same AD sever-
Systematic reviews of measurement instruments focus on ity measurement instrument in a standardized way gen-
two different quality aspects: (i) the methodological qual- erate different scores, inter‐rater reliability will be low.
ity of the included studies on measurement properties, Also if one dermatologist rated a patient with the same
and (ii) the quality of the instrument (i.e. its measurement level of disease severity at two different points in time
properties) itself. How to evaluate the methodological and generated different scores there would be low intra‐
quality of studies on measurement properties is beyond rater reliability. Lastly, responsiveness is defined as the
the scope of this chapter and we refer to the methodology ability of a measurement instrument to detect change
that has been developed by the COnsensus‐based over time in the construct to be measured. If an instru-
Standards for the selection of health Measurement ment is used to evaluate patients’ disease severity over
INstruments (COSMIN) initiative, see: www.cosmin.nl. time, the instrument at issue should be able to pick up
ATOPIC DERMATITIS

In the evaluation of the quality of the instruments three changes over time.
dimensions can be distinguished: reliability, validity and The relevant measurement properties and aspects of
SECTION 3:

responsiveness [15]. Validity means that the instrument measurement properties that are summarized in Table 16.2
measures what it purports to measure. If, for instance, were adapted from the consensus‐based t­axonomy that
disease severity is thought to be something that should be was developed by COSMIN [16].
defined by intensity of erythema, lichenification and exco- For the assessment of a measurement instrument, it is
riation, then the selected instrument should be measuring the also important to assess aspects of feasibility. Feasibility

Table 16.2  Taxonomy and definitions of measurement domains, measurement properties and aspects of measurement properties

Domain Measurement Aspect of a Definition


property measurement
property

Reliability The degree to which the measurement is free from measurement error
Reliability The extent to which scores for patients who have not changed are the same for repeated
(extended measurement under several conditions: for example, using different sets of items from the same
definition) HR‐PROs (internal consistency), over time (test–retest) by different persons on the same occasion
(inter‐rater) or by the same persons (i.e. raters or responders) on different occasions (intra‐rater)
Internal The degree of interrelatedness among the items
consistency
Reliability The proportion of total variance in the measurements which is because of ‘true’a differences
among patients
Measurement The systematic and random error of a patient’s score that is not attributed to true change of
error the construct to be measured
Validity The degree to which an HR‐PRO instrument measures the construct(s) it purports to measure
Content validity The degree to which the content of an HR‐PRO instrument is an adequate reflection of the
construct to be measured
Face validity The degree to which (the items of) an HR‐PRO instrument looks as though it is an adequate
reflection of the construct to be measured
Construct validity The degree to which the scores of an HR‐PRO instrument are consistent with hypotheses (for
instance with regard to internal relationships, relationships to scores of other instruments, or
differences between relevant groups) based on the assumption that the HR‐PRO instrument
validly measures the construct to be measured
Structural validity The degree to which the scores of an HR‐PRO instrument are an adequate reflection of the
dimensionality of the construct to be measured
Hypothesis testing See Construct validity
Cross‐cultural The degree to which the performance of the items on a translated or culturally adapted HR‐
validity PRO instrument are an adequate reflection of the performance of the items of the original
version of the HR‐PRO instrument
Criterion validity The degree to which the scores of an HR‐PRO instrument are an adequate reflection of a ‘gold
standard’
Responsiveness The ability of an HR‐PRO instrument to detect change over time in the construct to be measured
Responsiveness See Responsiveness
Interpretability The degree to which one can assign qualitative meaning – that is, clinical or commonly
understood connotations – to an instrument’s quantitative scores or changes in scores

a
 The word ‘true’ must be seen in the context of classical test theory (CCT) which states that any observation has two components – a true score and error
associated with the observation.
HR‐PRO, health‐related patient‐reported outcome.
Source: Mokkink et al. 2010 [16]. Reproduced with permission of Elsevier.
Chapter 16  Severity Scoring and Quality of Life Assessment in Atopic Dermatitis 215

refers to the ease of application of the instrument in its Detailed information on the measurement properties
intended setting, given constraints such as time or and their quality criteria is beyond the scope of this chap-
money [17]. The COSMIN initiative together with ter. We refer to textbooks (e.g. the book by De Vet et  al.
the Core Outcomes Measures in Effectiveness Trials [15]) and additional medical literature on consensus‐
(COMET) (http://www.comet‐initiative.org/) con- based quality criteria of measurement properties [20] for
ducted a Delphi study in which consensus was reached further information.
that important aspects of feasibility are [16]: patient’s
comprehensibility, interpretability, ease of administra-
tion, length of measurement instrument, completion
Evidence‐based recommendations
time, patient’s mental ability level, ease of standardiza-
for the measurement of atopic
tion, clinician’s comprehensibility, type of instrument,
dermatitis severity in clinical trials
cost of instrument, required equipment, type of admin- The mission of the international, multiprofessional
istration, availability in different settings, copyright, Harmonising Outcome Measures for Eczema (HOME)

ATOPIC DERMATITIS
patient’s physical ability level, regulatory agency’s initiative (www.homeforeczema.org) is to develop a core
requirement for approval, ease of score calculation. set of outcomes and corresponding outcome measure-

SECTION 3:
Interpretability denotes the degree to which one can ment instruments to be measured as a minimum in all
assign qualitative meaning to the quantitative scores or future eczema trials [21]. Based on an e‐Delphi and face‐
changes in scores generated by a particular instrument to‐face consensus voting, four outcome domains (aspects
[16]. For commonly used instruments, such as a ther- of disease that should be measured) were consented as
mometer that measures body temperature in degrees core: clinical signs, symptoms, quality of life and long‐
Celsius or Fahrenheit, interpretability usually causes no term control of flares. Once the core set of outcome
problems once familiarity with the numbers (‘a body tem- domains has been agreed, it is then important to identify
perature of 40 degrees centigrade feels hot’) is established. appropriate measurement instruments for the different
For scores of multi‐item instruments, the interpretation domains, according to step 3 of the HOME roadmap [22]
might be challenging as the meaning of such a score is not (Fig. 16.1).
immediately straightforward (e.g. what does a change in Instruments that objectively assess disease severity in
EASI score from 60 to 52 mean?). Aspects of interpretabil- AD were predominantly created between 1989 and 2009
ity of scores include the distribution of the (total) scores, [4,5]. A systematic review investigated instruments that
floor and ceiling effects, and information on the minimal assess the clinical signs of AD, synthesizing quality grad-
important change (i.e. the smallest change in score which ing of measurement properties and methodological
patients perceive as important [15,18]). study quality to generate recommendations [5]. Sixteen
As an example of feasibility and interpretability issues, such instruments were identified. The Severity Scoring of
a feasibility study was performed for the Eczema Area Atopic Dermatitis (SCORAD) Index [8] was found to
and Severity Index (EASI) [19]. It was shown that it was have adequate validity, responsiveness and interpretabil-
acceptable concerning ease of use with a completion time ity. Although interobserver reliability was adequate,
by trained investigators of approximately 6 minutes. intraobserver reliability was unclear. Internal consist-
Further, bands were suggested guiding the interpretabil- ency of the objective SCORAD, i.e. the part of the
ity of obtained scores, with scores of 0 denoting clear, SCORAD in which clinical signs are assessed, was ade-
0.1–1.0 almost clear, 1.1–7.0 mild, 7.1–21.0 moderate, 21.1– quate. The Eczema Area and Severity Index (EASI) [6]
50.0 severe and 50.1–72.0 very severe disease. was found to have adequate intraobserver reliability,
Just as taking a car driving test without passing is unac- intermediate interobserver reliability, internal consist-
ceptable, it is not sufficient for the measurement proper- ency, validity and responsiveness. However, interpreta-
ties (validity, reliability, responsiveness to change) of an bility and feasibility were unclear. The Three Item
instrument to be simply estimated  –  they also need to Severity Score (TIS) [23] and the SASSAD [7] met only
meet certain standards. The COSMIN initiative proposed some criteria and in particular were only given an ‘inter-
quality criteria to judge the measurement properties of mediate’ rating for content validity. The remaining
health status measurement instruments [20]. For instance, 12 scales had either (almost) not been validated or per-
according to these, a measurement instrument has ade- formed inadequately. Among the measures with unclear
quate content validity when all items are considered to be measurement properties are the patient‐assessed ver-
relevant for the construct to be measured (disease sever- sions of SCORAD and EASI, the patient‐oriented
ity), for the target population (people with AD), and for SCORAD [24] and the self‐administered EASI [25].
the purpose of the measurement and when the question- However, SCORAD and EASI in their original forms
naire is considered to be comprehensive (from the per- were  identified as the two candidate instruments to
spective of both physicians and patients). Involving assess ­clinical signs.
patients (e.g. by means of focus group discussions) is The European Task Force on Atopic Dermatitis (ETFAD)
essential, but often neglected. developed the SCORAD (Severity Scoring of Atopic
A further example is internal consistency, which is Dermatitis) [8] instrument which combines objective crite-
assumed to be adequate if there is at least evidence for ria with subjective symptoms. The intensity of six clinical
unidimensionality or positive structural validity and signs (erythema, oedema/papulation, oozing/crusts,
Cronbach’s alpha(s) is ≥0.70 and ≤0.95 [18]. excoriation, lichenification, dryness) is assessed on a scale
216  Section 3 Atopic Dermatitis and Related Disorders

The Home roadmap to develop core sets of outcome measurements

Step 1: Define scope and applicability


Population (condition)
Intervention
Setting(e.g. trial,registry, clinical practice)
Geographical/regional scope*
Stakeholders

Step 2: Develop core set of outcome domains


ATOPIC DERMATITIS

Consensus study involving representatives of relevant


stakeholders.
SECTION 3:

Step 3: Develop core set of outcome measurements


Identification and recommendation of adequate measurement instrument(s)
for each core outcome domain by a 5 stage process

Stage 1 Stage 2 Stage 3 Stage 4 Stage 5


Identify all Establish the Determine which instruments are good enough quality meet the requirements of Carry out validation Finalise core
Task

instruments extent and the OMERACT filter and be shortlisted for further consideration. studies on shortlisted outcome(s) for
previously used quality of testing scales. domain.
to measure the of the identified
domain. instruments.

Systematic review of Systematic review Apply OMERACT filter: truth, discrimination, and feasibility: Consensus Re-apply the
outcome of validation studies discussion and voting OMERACT filter with
instruments used. of the long-list of Truth Discrimination Feasibility to determine what
the results of the
identified “Is the measure truthful, “Does the measure “Can the measure be validation studies will
instruments be conducted on completed validation
Methodology

does it measure what it discriminate between applied easily in its


short-listed studies.
intends to measure? Is the situation that are of intended setting, given
Highlight any gaps instruments. Gaps in
result unbiased and interest?” constraints of time,
in validation. testing were
relevant?” money and Consensus
highlighted in stage 2
interpretability?” discussion and voting
(systematic review).
on core outcome to
Consensus discussion and Consensus discussion Appropriate methods
Consensus discussion and voting on discrimination: and voting on feasibility: be recommended
used to fill the gaps in
voting on truth: validation.
1.Reliability 1.Time take
1.Face validity 2.Sensitivity to change 2.Cost
2.Content validity 3.Interpretability
3.Construct validity
4.Criterion validity

Long-list of all Summary of which Short-list of potential instruments that meet the requirements of the OMERACT Short-list of fully Recommended core
instruments instruments have filter. tested instruments.
Output

outcome instrument
previously used been tested and
for the domain.
to measure the the quality, extent
domain. and results of any
testing.

Step 4: Disseminate, prepare guidance


material, review, and possibly revise core set
of outcome measurements

*for trials the scope should generally be global


Fig. 16.1  HOME Roadmap. Source: Schmitt et al. 2015 [22]. Reproduced with permission of Elsevier.

from 0 (absence) to 3 (severe) for a representative body site. in less than 10 minutes [29,30]. Training is available freely
The resulting score is combined with a score for the extent on the SCORAD website (http://adserver.sante.univ‐
of the involved body area and an assessment of sleep loss nantes.fr/). There is also specific software designed to
and pruritus on a visual analogue scale, to yield an overall facilitate the assessment of AD, the ScoradCard © 2.0 [31].
SCORAD score with a maximum of 103 points. Based on The EASI was developed in 1998 based on the
the objective SCORAD (intensity of signs plus extent), it Psoriasis Area and Severity Index scoring system [6].
has been suggested that <15 points represents mild, 15–40 The intensity of four clinical signs (erythema, induration/
represents moderate and >40 points represents severe dis- papulation, excoriations, lichenification) is assessed at
ease [26–28]. After training, the SCORAD can be performed each of four defined body sites (Figs  16.2 and 16.3).
Chapter 16  Severity Scoring and Quality of Life Assessment in Atopic Dermatitis 217

Eczema Area and Severity Index (EASI) case report form – age <8 years

Area of Involvement: Each body region has potentially 100% involvement. Score 0 to 6 based on the following table:

% involvement 0 1–9% 10–29% 30–49% 50–69% 70–89% 90–100%


Region score 0 1 2 3 4 5 6

Severity of Signs: Grade the severity of each sign on a scale of 0 to 3:

0 None
1 Mild Take an average of the severity across the
involved area.
2 Moderate
Half points may be used e.g. 2.5.
3 Severe

ATOPIC DERMATITIS
Scoring table:

SECTION 3:
Erythema Edema/ Excoriation Lichenification Region score Multiplier Score per body
(0–3) Papulation (0–3) (0–3) (0–6) region
Body region (0–3)

Head/neck ( + + + ) X X 0.2

Trunk ( + + + ) X X 0.3

Upper extremities ( + + + ) X X 0.2

Lower extremities ( + + + ) X X 0.3

The final EASI score is the sum of the 4 region scores: ____________
(0–72)

Fig. 16.2  EASI (<8 years). Source: Reproduced with kind permission from the HOME group at the Centre of Evidence Based Dermatology at Nottingham, UK.

Eczema Area and Severity Index (EASI) case report form – age ≥8 years
Area of Involvement: Each body region has potentially100% involvement. Score 0 to 6 based on the following table:
% involvement 0 1–9% 10–29% 30–49% 50–69% 70–89% 90–100%
Region score 0 1 2 3 4 5 6

Severity of Signs: Grade the severity of each sign on a scale of 0 to 3:


0 None
1 Mild Take an average of the severity across the
involved area.
2 Moderate
Half points may be used e.g. 2.5.
3 Severe

Scoring table:

Erythema Edema/ Excoriation Lichenification Region score Multiplier Score per body
(0–3) Papulation (0–3) (0–3) (0–6) region
Body region (0–3)

Head/neck ( + + + ) X X 0.1

Trunk ( + + + ) X X 0.3

Upper extremities ( + + + ) X X 0.2

Lower extremities ( + + + ) X X 0.4

The final EASI score is the sum of the 4 region scores: ____________
(0–72)

Fig. 16.3  EASI (≥8 years). Source: Reproduced with kind permission from the HOME group at the Centre of Evidence Based Dermatology at Nottingham, UK.
218  Section 3 Atopic Dermatitis and Related Disorders

Additionally, d ­ isease extent is assessed for these sites. (without reference to another time point), some used
Multiplying severity by area by a constant weighted dynamic scales (with reference to a time point, e.g.
value yields a total score for each body region. Summing 50% improved compared to baseline). The number of
these scores yields the total EASI score with a maxi- categories ­varied between 4 and 7, with 6‐point scales
mum score of 72. As mentioned previously, feasibility, being the most frequently used. The most striking
including interpretability of the EASI, has been finding was the huge variation in the way in which the
assessed. The EASI is a unidimensional measure, mean- disease severity was defined. Only 24% of the trials
ing that it only assesses clinical signs and does not using an IGA provided instructions on how to imple-
incorporate symptoms. Unlike SCORAD, it measures ment the scales. All global assessment scales for which
the intensity of lesions at multiple body parts and does instructions were available included erythema and
not rely on a representative lesion. A user guide, a papulation/oedema with many adding oozing/weep-
training video, an EASI calculator and an EASI calcula- ing and crusting and only some considering excoria-
tor App for smartphone devices are all available tion, scaling and lichenification. Of note, only three of
ATOPIC DERMATITIS

through the website of the HOME initiative (www. the IGA measures included excoriation, which is the
homeforeczema.org). sign that shows the strongest correlation with patients’
SECTION 3:

At the third international HOME meeting (2013), assessments of disease severity [36]. For many scales
broad consensus was achieved that the preferred AD used, no instructions were reported or referenced at
clinical signs measurement instrument should capture all. These scales probably rely on a gestalt approach to
both intensity and extent of AD signs and that intensity assessing global severity, taking into account different
should be assessed for lichenification, oedema/papula- facets of the disease such as symptoms and QoL in
tion, excoriation and erythema [32]. Only the objective addition to clinical signs. Given their widespread use,
SCORAD and the EASI included those four essential primary validation research is urgently needed for
signs, and at the same time, these two instruments had IGA measures of AD disease severity, with the aim to
been identified as candidate instruments in the afore- enhance standardization. Until such research is done,
mentioned systematic review. In the final consensus recommendations cannot be made as to which global
voting, broad consensus was achieved that the EASI measurement instrument is most appropriate.
should be the core outcome measurement instrument Although a global measure of disease severity is not
to assess clinical signs in AD clinical trials [32,33]. included in the Core Outcome Set for AD [37,38], its
Perceived advantages of the EASI over the objective use is recommended by the US Food and Drug
SCORAD were that EASI only includes the four essen- Administration [39] for reasons that are unclear.
tial signs, that signs are assessed not at a single repre-
sentative site but at multiple body sites and that extent
of lesions is given sufficient weighting (50% of total
Other ways of measuring severity
score as opposed to 24% of total score in the objective
of atopic dermatitis
SCORAD).
The quest for objective serum markers to measure AD
severity reliably has had mixed fortunes, and is still far
from clear. A systematic review that included a meta‐
Global measurement instruments analysis of 108 articles suggested that serum thymus
of atopic dermatitis severity activation‐regulated chemokine (TARC) showed the
Although clinical scoring systems such as the EASI best correlation with clinical disease severity in a range
provide a comprehensive way of assessing disease of study designs [40]. A range of other biomarkers
severity as measured by an array of relevant clinical requiring more research such as serum cutaneous T‐
signs, global measures of disease severity are often felt cell attracting chemokine (CTACK) were also identi-
to be more readily accessible for patients and physi- fied. Other objective ways of measuring AD severity
cians as they can be applied in an easy and rapid way. such as measuring scratching at night by wearing
In these scales, disease severity is often rated on a accelerometers have not proven to be very useful to
nominal scale, for instance from 0 to 5 (clear, almost date because of the lack of correlation with other meas-
clear, mild, moderate, severe, very severe [34]. ures of disease severity and QoL, and incomplete
However, such a holistic judgement is subject to inter- and complex data that are difficult to interpret [41].
pretation and may be less precise c­ompared to a sever- Approaches such as infrared monitoring and video
ity measurement instrument with many items. A recording of nocturnal scratching are not practical for
systematic review investigated the use of Investigator most studies. Other indirect measures of AD severity
Global Assessments in randomized controlled trials such as topical corticosteroid and emollient use as
(RCTs) conducted in AD between 2000 and 2014 [35] rescue therapy [42] have been recorded in clinical
­
and found that about one third of the investigated 317 studies, but mainly as a potential confounder rather
trials used an IGA as outcome measure. There was than a surrogate of disease severity per se because
wide variation in the ways in which global severity many factors contribute to the decision for a patient to
was conceptualized. Some studies used static scales use and report treatment usage.
Chapter 16  Severity Scoring and Quality of Life Assessment in Atopic Dermatitis 219

Evidence‐based recommendation the patient [53]. Examples of PROs include illness


for the measurement of subjective ­ erceptions, satisfaction with care, and health‐related
p
severity of atopic dermatitis quality of life (further referred to as quality of life, QoL).
(symptoms) in clinical trials PROs are most commonly assessed by means of self‐
administered questionnaires, and are often termed as
Just as for the clinical signs domain, a systematic review PRO instruments or patient‐reported outcome measures
was also conducted to evaluate the measurement proper- (PROMs). One of the most commonly used PROs in
ties of existing AD symptom measurement instruments healthcare research and clinical practice is QoL [54].
[43]. Eighteen instruments were identified, five of which Following the definition of the World Health
were supported by sufficient evidence to consider them Organization, the construct QoL entails at least three
as candidate instruments to measure AD symptoms: the domains and reflects patients’ evaluation of the impact of
Patient‐Oriented Eczema Measure (POEM) [44,45], the disease and treatment on their physical, psychological and
paediatric Itch Severity Scale (ISS) [46], Patient‐Oriented social functioning and well‐being [55]. As it is well known

ATOPIC DERMATITIS
SCOring Atopic Dermatitis (PO‐SCORAD) [24,47], Self‐ that chronic skin diseases, such as AD, negatively affect the
Administered Eczema Area and Severity Index (SA‐EASI) lives of children [56,57], QoL has become an essential out-

SECTION 3:
[25] and adapted SA‐EASI [48]. come in clinical trials, clinical practice and healthcare man-
These and other instruments were discussed at the agement [58]. This is also reflected by the fact that the
HOME IV meeting and it was considered that irritated HOME initiative has identified QoL as a core outcome to be
skin, redness/inflamed skin, dryness, sleep loss and itch assessed in all AD clinical trials [40]. A previous study of
were all essential aspects of AD symptoms [49]. ISS was the National Eczema Association for Science and Education
not further considered because it only assesses itch. PO‐ surveyed 429 patients with AD or their parents and 303
SCORAD, SA‐EASI and POEM were voted on and POEM physicians about the effect of AD on QoL [59]. Results
was voted as the preferred core outcome measurement showed that children’s QoL is significantly affected by
instrument for AD symptoms. It is now recommended as their skin disease, particularly children with moderate to
a measure for all clinical trials in AD [50]. PO‐SCORAD severe AD, causing impairment of daily activities, school
and SA‐EASI were not favoured based on the argument performance, social and leisure activities and sleep. The
that these instruments largely represent a self‐assessment authors concluded that QoL issues are a key consideration
of clinical signs rather than true symptom measurement in management of AD. Another study reported on the
instruments. impact of skin disease on different aspects of adolescents’
POEM (Fig. 16.4) has seven items, assessing frequency QoL. The results of this study showed that adolescents’
of pruritus, sleep disturbance, bleeding, weeping or ooz- QoL is impacted by their skin diseases on several issues
ing, cracking, flaking, and dryness or roughness. It must that are specific to adolescents, such as psychological
be noted that the agreed essential symptom of redness is impact (91% of patients), physical impact (81%) and social
not included in POEM. It was deliberately excluded in the impact (81%). This also highlights the need for QoL assess-
development process because redness is difficult to detect ment [60]. Although the measurement of QoL has its ori-
in darker skin [44]. POEM is freely available in a range gins in the clinical trials setting, the use of QoL assessment
of languages (www.nottingham.ac.uk/research/groups/ in clinical dermatology practice is a more recent develop-
cebd/resources/poem.aspx) and takes less than 2 minutes ment and far from established. However, different efforts
for patients and/or carers to complete [43]. Its score ranges have been undertaken to highlight the relevance of meas-
from 0 to 28. A banding study has been conducted, linking uring QoL in clinical practice. An expert‐based opinion
categories to score ranges [45]: 0–2 denotes almost clear, statement of the Task Force on Quality of Life of the
3–7 mild, 8–16 moderate, 17–24 severe and 25–28 very European Academy of Dermatology and Venereology
severe disease. The responsiveness of the POEM has been (EADV) identified five broad categories in which using
investigated in young children [51]. It was found that the QoL information in practice is seen as useful [61]: aware-
minimal clinically important difference in relation to ness of skin disease burden, clinical service administration,
change recalled on a Parent Global Assessment (PGA) was informing consultation, clinician–patient communication,
around 3. Some validation gaps remain regarding struc- and informing clinical decisions. Another paper reported
tural and cross‐cultural validity which will be addressed on why QoL assessment in clinical practice is important, to
in the future by the HOME symptoms working group [50]. whom QoL assessment is particularly relevant and how
QoL can be assessed. The authors indicated that QoL data
can be used in clinical practice for five different purposes:
Measuring quality of life in children
(i) to increase patients’ self‐awareness and empowerment,
with atopic dermatitis
(ii) to increase patient‐centredness in healthcare, (iii) to
The importance of using patient‐reported outcomes make an optimal choice for treatment, (iv) to monitor treat-
(PROs) in research, clinical practice and healthcare man- ment over time and determine treatment effectiveness, and
agement is increasingly being recognized [52]. A PRO is (v) to improve treatment outcome [62].
a measurement of any aspect of a patient’s health status The relationship between QoL and disease severity is not
that is directly assessed by the patient without the inter- straightforward. A Dutch study administered the Infants’
pretation of the patient’s response by anyone other than Dermatitis Quality of Life Index (IDQOL) and SCORAD as
220  Section 3 Atopic Dermatitis and Related Disorders

well as the TIS as measures of disease severity to 66 children routine healthcare, measuring QoL in infants, children
at a mean age of 31 months [63]. Weak and moderate‐ and adolescents enables dermatologists to understand the
to‐strong correlations between IDQOL and disease sever- impact of the skin disease in an individual, and it high-
ity were found when investigators and parents assessed lights specific aspects of QoL that are affected the most. A
disease severity, respectively. When used in research, QoL position paper of the EADV provides a comprehensive
measurement instruments should thus be seen as comple- overview of the QoL measurement instruments that have
mentary to established disease severity instruments. In been developed, validated and used in AD [64].
ATOPIC DERMATITIS
SECTION 3:

POEM for proxy completion (e.g by parent)

Patient Details: _______________________


______________________________________
______________________________________
______________________________________
Date:_________________________________

Please circle one response for each of the seven questions below about your child’s eczema. If your child is
old enough to understand the questions then please fill in the questionnaire together. Please leave blank
any questions you feel unable to answer.

1. Over the last week, on how many days has your child’s skin been itchy because of their eczema?

No days 1-2 days 3-4 days 5-6 days Every day

2. Over the last week, on how many nights has your child’s sleep been disturbed because of their eczema?

No days 1-2 days 3-4 days 5-6 days Every day

3. Over the last week, on how many days has your child’s skin been bleeding because of their eczema?

No days 1-2 days 3-4 days 5-6 days Every day

4. Over the last week, on how many days has your child’s skin been weeping or oozing clear fluid because of
their eczema?

No days 1-2 days 3-4 days 5-6 days Every day

5. Over the last week, on how many days has your child’s skin been cracked because of their eczema?

No days 1-2 days 3-4 days 5-6 days Every day

6. Over the last week, on how many days has your child’s skin been flaking off because of their eczema?

No days 1-2 days 3-4 days 5-6 days Every day

7. Over the last week, on how many days has your child’s skin felt dry or rough because of their eczema?

No days 1-2 days 3-4 days 5-6 days Every day

Total POEM Score (Maximum 28):_________________

© The University of Nottingham

Fig. 16.4  POEM.Source: The University of Nottingham, UK.


Chapter 16  Severity Scoring and Quality of Life Assessment in Atopic Dermatitis 221

POEM for proxy completion (e.g by parent)

How is the scoring done? What does a poem score mean?

Each of the seven questions carries equal weight To help patients and clinicians to understand their
and is scored from 0 to 4 as follows: POEM scores, the following bandings have been
established (see references below):

ATOPIC DERMATITIS
No days =0

SECTION 3:
1-2 days =1 • 0 to 2 = Clear or almost clear
3-4 days =2 • 3 to 7 = Mild eczema
5-6 days =3 • 8 to 16 = Moderate eczema
• 17 to 24 = Severe eczema
Every day =4
• 25 to 28 = Very severe eczema

Note:
Do I need permission to use the scale?

• If one question is left unanswered this is scored 0 Whilst the POEM scale is protected by copyright, it
and the scores are summed and expressed as usual is freely available for use and can be downloaded
out of a maximum of 28 from: www.nottingham.ac.uk/dermatology
We do however ask that you register your use of
• If two or more questions are left unanswered the
the POEM by e-mailing cebd@nottingham.ac.uk
questionnaire is not scored
with details of how you would like to use the scale,
• If two or more response options are selected, the and which countries the scale will be used in.
response option with the highest score should be
recorded

References
Charman CR, Venn AJ, Williams HC. The Patient-Oriented Eczema Measure: Development and Initial Validation of
a New Tool for Measuring Atopic Eczema Severity From the Patients' Perspective.
Arch Dermatol. 2004;140:1513-1519

Charman CR, Venn AJ, Ravenscroft JC, Williams HC. Translating Patient-Oriented Eczema Measure (POEM) scores into clinical
practice by suggesting severity strata derived using anchor-based methods.
Br J Dermatol. Dec 2013; 169(6): 1326–1332.

© The University of Nottingham. The Patient Oriented Eczema Measure (POEM) scale is free to use. Permission is
granted to reproduce and/or redistribute this material in its entirety without modification. Any use which falls outside this
remit requires the express consent of the copyright owner.

Fig. 16.4  (Continued)

Types of instruments available Generic instruments


for measuring QoL Generic QoL instruments can be used for the measure-
A simple way of measuring QoL in children is asking ment of QoL in all kinds of diseases as well as in the gen-
patients or their proxy ‘How does your atopic dermatitis eral population. In theory, the use of generic instruments
affect your daily life/the life of your child?’. A single‐ such as the EuroQoL (EQ) – 5D [65] allows comparison of
item question, however, is often less reliable. In order to data across diseases in any specialty, making them par-
collect QoL data in a more comprehensive and system- ticularly useful for health economists to estimate and
atic way, reliable and valid multiple‐item QoL instru- compare the cost‐effectiveness of a treatment for one dis-
ments may be required. In general, QoL instruments can ease against another. Generic instruments, however, may
be distinguished into generic and specific QoL instru- not have been designed with reference to dermatology
ments that are self‐reported or proxy‐reported. and therefore may not capture all aspects that are important
222  Section 3 Atopic Dermatitis and Related Disorders

to patients with skin diseases. Multiple generic instru- diseases, such as cystic fibrosis, asthma, diabetes or
ments exist in paediatrics [66], however, data on their use epilepsy, although sample sizes were small [72]. The
in dermatology or AD are sparse. One German study results of this study also showed a reasonably good agree-
examined generic QoL in a population‐based sample of ment between the child’s and parents’ scores on the CLQI
6518 11‐ to 17‐year‐old children/adolescents [67], using a and the dermatology‐specific Children’s Dermatology
revised version of the KINDL (Kindliche Lebensqualität, Life Quality Index (CDLQI [73], see next section).
paediatric quality of life) questionnaire [68]. KINDL‐R
has 24 items and assesses QoL in six dimensions: physical
well‐being, emotional well‐being, self‐worth, well‐being
Evaluation of quality‐of‐life
in the family, well‐being related to friends/peers, and
instruments for children with
school‐related well‐being. A significant impact of AD in
atopic dermatitis
the past 4 weeks remained even when a number of covari- A systematic review of QoL measurement instruments
ates including atopic comorbidity and mental health were for infants, children and adolescents with AD reported
ATOPIC DERMATITIS

controlled for. on the results of 17 studies that evaluated the measure-


ment properties of different QoL instruments for chil-
SECTION 3:

Specific instruments dren with AD [74]. The aim of this systematic review
Specific instruments include dermatology‐specific was to identify the single best currently available instru-
and disease‐specific instruments. Dermatology‐specific ment to assess QoL in infants, the single best currently
instruments were designed for all kinds of skin diseases. available instrument to assess QoL in children, and the
They are generic to dermatology and, in theory, allow for single best currently available instrument to assess QoL
comparison across skin diseases. Disease‐specific instru- in adolescents with AD. Studies that were eligible for
ments were developed for, and at the same time limited inclusion were instrument development and validation
to, a specific skin disease. Disease‐specific instruments studies of dermatology‐ and disease‐specific (i.e. AD‐
may be most informative in clinical research as these specific) QoL instruments in which at least 50% of the
instruments are more sensitive to the patients’ health sta- patients were AD patients younger than 16 years of age,
tus under study. Disease‐specific instruments may also or subgroup analysis was done for AD patients. The
provide relevant and detailed information that can be review authors assessed the measurement properties of
used to inform patient management. five QoL instruments that fulfilled the strict eligibility
criteria: Childhood Atopic Dermatitis Impact Scale
Proxy‐reported instruments (CADIS) [1,75], Children’s Dermatology Life Quality
Proxy‐reported instruments (i.e. instruments that are com- Index (CDLQI) [73], Childhood Impact of Atopic
pleted by someone other than the child) are frequently Dermatitis (CIAD) [76], DISABKIDS Atopic Dermatitis
used as substitutes for the child’s rating when children are Module (DISABKIDS‐ADM) [77], and Infants’ Dermatitis
not able to complete the instrument by themselves or have Quality of Life Index (IDQoL) [56].
difficulty understanding the questions. The opinions of
parents, family members or caregivers may not adequately Dermatology‐specific instruments
reflect the impact of disease on patients’ QoL. There is no to measure QoL in children with atopic
clear consensus about the validity of using a proxy in the dermatitis [78]
assessment of childhood QoL. In the literature, differences The Children’s Dermatology Life Quality Index
between the responses of the child and parent(s) have (CDLQI)
been reported [69,70]. A previous systematic review deter- The CDLQI has been developed for children with skin
mined the relationship between ratings of children’s QoL diseases aged between 4 and 16 years. It is a self‐reported
made by parents and children [71]. Of the included stud- instrument, consisting of 10 items covering six domains.
ies, three studies assessed whether parents perceived the Answers can be given on a 4‐point Likert scale and sum
illness to have a greater impact on their child’s QoL than scores are calculated (range: 0–30), with higher scores
did the child, but no clear conclusions could be drawn having greater impact on QoL. It takes less than 2 minutes
[71]. The level of agreement between the parent’s rating for the child to complete and a cartoon version is availa-
and the child’s ratings may vary depending on the QoL ble [79]. The recall period is 1 week [73]. There are no
aspect assessed; in general, there was greater agreement charges for nonfunded studies. The CDLQI is available in
for observable constructs, such as physical functioning or translation in more than 50 language versions and differ-
symptoms, than with nonobservable constructs, such as ent language cartoon versions (available from: http://
emotional or social functioning [71]. sites.cardiff.ac.uk/dermatology/quality‐of‐life/
An example of a generic proxy‐reported instrument is childrens‐dermatology‐life‐quality‐index‐cdlqi/). The
the Children’s Life Quality Index (CLQI) [72]. The CLQI CDLQI is the most widely used dermatology‐specific
has been developed for school‐aged children. It consists instrument in AD clinical trials involving children [13].
of 12 questions that relate to the child’s health over the Olsen et  al. (2016) presented the results of a meta‐
previous 3 months. The CLQI is available in an English analysis of CDLQI data for different childhood skin
language version. ­diseases, including children with AD [80]. A total of 67
A cross‐sectional study using the CLQI demonstrated studies were included in the review, involving 7798
that AD has a greater impact on QoL than other chronic children with 20 different skin diseases. The authors
­
Chapter 16  Severity Scoring and Quality of Life Assessment in Atopic Dermatitis 223

c­oncluded that most skin diseases in children have a consisting of 10 items covering eight domains. Answers
‘small’ mean effect on QoL. However, the range of CDLQI can be given on a 4‐point Likert scale and sum scores
scores was wide, which means that a considerable num- are calculated (range: 0–30). The recall period is 1 week
ber of children may experience major impact on their QoL [56,82]. There is no charge for use in nonfunded studies
because of their skin disease. and routine clinical practice. For pharmaceutical com-
panies, $11.50 per patient is charged. The IDQoL is
available in translation in more than 20 language
Disease‐specific instruments to measure versions (available from: http://sites.cardiff.ac.uk/
QoL in children with atopic dermatitis dermatology/quality‐of‐life/the‐infants‐dermatitis‐
The Childhood Atopic Dermatitis Impact
quality‐of‐life‐index‐idqol/). The IDQoL is the most
Scale (CADIS)
widely used disease‐specific instrument in AD clinical
The CADIS has been developed for children with AD aged
trials involving infants [13].
between 0 and 6 years (and their parents). It assesses both

ATOPIC DERMATITIS
the QoL of the infant or child and the QoL of their parents.
It is a proxy‐reported instrument, consisting of 45/41/33 Quality of the included QoL instruments

SECTION 3:
items (i.e. 45 items in the original version, 41 items in the The results of the aforementioned systematic review
long Italian version, and 33 items in the short Italian ver- showed that of all instruments reviewed, only the US ver-
sion) covering five domains: child symptoms, child activity sion of the CADIS [75] has the potential to be recom-
limitation and behaviour, family and social function, par- mended to measure QoL in children, depending on the
ent sleep, parent emotions. The conceptual model under- results of further validation studies. The internal consist-
pinning the CADIS was developed based on published ency of the CADIS was found to be adequate (Cronbach’s
literature and interviews with both parents and expert cli- alpha values ranged from 0.76 to 0.93 for its domains). It is
nicians. Of note, with 17 of 45 items, the domain ‘parent a reliable instrument with adequate construct validity. The
emotions’ represents an important part of the CADIS. completion time of the 45‐item instrument was also found
Answers can be given on a 5‐point Likert scale. Domain to be acceptable. Measurement error and structural valid-
scores are calculated by summing up item scores of all ity of the CADIS have not yet been investigated. Future
items in one domain. A total score is calculated by sum- validation work should focus on content validity, struc-
ming up the scores of all items. The recall period is 4 weeks tural validity, cross‐cultural validity, measurement error,
[1,75]. There are no administration costs. The CADIS is responsiveness and interpretability of the CADIS [81].
available in US English, Italian and Japanese [74]. Use of the other instruments cannot be recommended
until additional validation studies are conducted [81]. It is of
Childhood Impact of Atopic Dermatitis (CIAD) concern that nearly all existing QoL instruments for infants,
CIAD has been developed for children with AD. It is a children and adolescents with AD are lacking validation
proxy‐reported instrument, consisting of 9/7 items (i.e. data. A major reason for this is that most studies of the meas-
9 items in the Dutch, English (UK), French and German urement properties were of poor methodological quality
versions; 7 items in the English (US) version). The num- (76%). The design and/or statistical methods that were used
ber of domains is not described. The response options to evaluate the instruments (i.e. its measurement properties)
are dichotomous (true/not true). The scoring algorithm were found to be inadequate, meaning that the results of the
is not described and a recall period is not applicable as quality of the instruments (i.e. its measurement properties)
questions are referring to ‘at the moment’. It has not cannot be trusted. In addition, data on the interpretability of
been described if administration costs are charged [76]. the included instruments is lacking [81].
CIAD is available in Dutch, English (UK), English (US), Proxy‐reported instruments, including the CADIS, may
French and German [81]. be particularly useful in infants and younger children;
however they are less suitable for older children and ado-
The DISABKIDS Atopic Dermatitis Module lescents. For these age groups, a self‐reported instrument
(DISABKIDS‐ADM) is recommended. However, the self‐reported instruments
The DISABKIDS‐ADM has been developed for children and that were included in the review (i.e. the CDLQI and the
adolescents with AD. It is a self‐ or proxy‐reported instru- DISABKIDS‐ADM) do not have the potential to be recom-
ment, consisting of 12 items covering two domains. Answers mended as they lack validation data. Further, these instru-
can be given on a 5‐point Likert scale (and ‘not applicable’). ments are intended for use in adolescents and not in
For each domain, a mean standardized score can be calcu- children. The review authors stated that factors influenc-
lated (range: 0–100). The recall period is not described. It is ing adolescents’ QoL are fundamentally different from
not clear if administration costs are charged [77]. The those observed in children [81].
DISABKIDS‐ADM is available in Brazilian, Portuguese and
other languages that were not further specified [81]. Conclusions of the systematic review
of QoL measurement instruments
The Infants’ Dermatitis Quality of Life Index for infants, children and adolescents
(IDQoL) with atopic dermatitis
The IDQoL has been developed for infants with AD The authors of the systematic review of QoL measure-
aged under 4 years. It is a proxy‐reported instrument, ment instruments for infants, children and adolescents
224  Section 3 Atopic Dermatitis and Related Disorders

with AD reported on the quality of the measurement first measurement instrument to assess the impact of AD
instruments. They concluded that at present there is no on family QoL [86] and the instrument most frequently
self‐reported QoL instrument for children with AD that used in clinical trials in AD [13]. It is a 10‐item AD‐specific
can confidently be recommended for use in AD clinical QoL questionnaire which was developed based on
trials without further research. The review authors sug- ­ethnographic focus groups and interviews that revealed
gested using the proxy‐reported CADIS for infants and the areas impacted by AD. Items cover the impact of
younger children with AD for the time being, depending helping with treatment on the main caregiver’s life, rela-
on the results of future validation studies. They recom- tionships, emotional distress, shopping, expenditure,
mended that future validation studies should focus on leisure activity, tiredness, sleep, housework and food
the CADIS, and that an evidence base for the validity and preparation. The total score of the DFI ranges from 0 to
interpretability of self‐reported instruments should be 30 with a higher score indicating greater impairment. A
established [81]. systematic review showed that there is evidence for the
construct validity, internal consistency and test–retest
ATOPIC DERMATITIS

reliability as well as responsiveness to change of the DFI,


Towards standardization of outcome but that evidence is missing regarding structural valid-
SECTION 3:

reporting for quality of life in atopic ity and interpretability [87]. In 2007, the Family
dermatitis Dermatology Life Quality Index (FDLQI) became avail-
The results of the meta‐analysis by Olsen et al. indicated able as a dermatology‐specific measurement instrument
that the CDLQI was the most frequently used instrument of the family impact of skin disease [88]. Evidence for its
to measure QoL in children with AD [80]. It seems to be validity is much more rigorous compared to the DFI
reliable, valid and responsive to change in skin diseases [89]. A small study demonstrated evidence for a correla-
[83]. Although the CDLQI is most commonly used, the tion between the FDLQI and the DFI of moderate
results of the systematic review by Heinl et al. suggested strength [90].
that the CDLQI cannot currently be recommended as an There are other instruments available to assess the
instrument of choice as it is lacking validation data in family impact of AD. The Childhood Atopic Dermatitis
children with AD. Impact Scale (CADIS) [75,78] has already been described.
HOME is currently working towards selecting the It puts more emphasis on emotional impact compared to
most appropriate instrument to measure QoL in children the IDLQI, CDLQI and DFI and is the only instrument
with AD in clinical trials. A future research agenda has that assesses the multidimensional effect of AD on both
been set to perform additional validation studies on parents and children. The development of the Parents’
instruments that have the potential to be included in the Index of Quality of Life in Atopic Dermatitis (PI‐QoL)
core outcome set. By using the single best QoL instru- [91,92] conceptually started from a needs‐based model,
ment in all clinical trials in children with AD, standardi- meaning that the instrument assesses how AD interferes
zation in QoL outcome measurement will be enhanced. with the fulfilment of needs rather than measuring
This allows for comparisons and pooling results in sys- impairments (symptoms) and disability (functioning). It
tematic reviews and meta‐analyses, which will lead to thus provides complementary information to existing
an improvement of evidence‐based healthcare decisions instruments. The PI‐QoL is unidimensional and has 28
in the treatment of children with AD. Further validation items. One advantage is that it has been developed and
studies should be undertaken when using instruments validated in several countries simultaneously (UK,
in other countries or ethnic groups differing from the Netherlands, Italy, Germany, France, US and Spain). The
original study group. When translating questionnaires Quality of Life in Primary Caregivers of Children with
into other languages, it is important to obtain at least AD (QPCAD) instrument was developed in Japan [93].
two backward and forward translations to avoid errors The rationale for its development was that instruments
of misunderstanding. such as the DFI and the PI‐QoL only include negative
items and lack positive items (such as pleasure in life)
and that the inclusion of positive items may help to
Instruments to measure family impact
assess QoL more accurately [94]. It has 19 items and
of atopic dermatitis
assesses health‐related QoL in the domains ‘worry about
The field of paediatric health‐related QoL is changing atopic dermatitis’, ‘achievement’, ‘family cooperation’
rapidly and can add much to our knowledge of the and ‘exhaustion’. Another questionnaire has been devel-
impact that skin disease has on children and their fami- oped and validated in Germany [95]. It is a 26‐item
lies. The impact of illness on family/carers is multifac- instrument that assesses health‐related QoL in five
eted, encompassing social activities, leisure time, domains: psychosomatic well‐being, impact on social
education and work, as well as financial, emotional and life, satisfaction with medical care, emotional coping
family relationships, with some of these themes such as with illness, and acceptance of illness. It is currently
social, financial and emotional impact being intercon- unclear which of these instruments best captures the
nected [84]. In dermatology, other family members impact of AD on primary caregivers. A systematic review
affected by a skin disease of one family member have is underway to compare the quality of these instruments
been referred to as the ‘greater patient’ [85]. The (www.crd.york.ac.uk/PROSPERO/display_record.
Dermatitis Family Impact (DFI) questionnaire was the asp?ID=CRD42015027481).
Chapter 16  Severity Scoring and Quality of Life Assessment in Atopic Dermatitis 225

Conclusion 3 Charman C, Chambers C, Williams H. Measuring atopic dermatitis


severity in randomized controlled clinical trials: what exactly are we
Capturing disease severity by clinical signs, patient measuring? J Invest Dermatol 2003;120:932–41.
4 Schmitt J, Langan S, Williams HC. What are the best outcome meas-
reports of symptoms and patient reports of QoL all pro- urements for atopic eczema? A systematic review. J Allergy Clin
vide important information on distinct aspects of AD. Immunol 2007;120:1389–98.
This is reflected by the fact that the HOME initiative has 5 Schmitt J, Langan S, Deckert S et al. Assessment of clinical signs of
reached consensus that these – and long‐term control of atopic dermatitis: a systematic review and recommendation.
J Allergy Clin Immunol 2013;132:1337–47.
flares – be part of the Core Outcome Set (COS) for AD 6 Hanifin JM, Thurston M, Omoto M et al. The eczema area and sever-
clinical trials, meaning that these domains should be ity index (EASI): assessment of reliability in atopic dermatitis. EASI
assessed as a minimum in all future AD clinical trials. Evaluator Group. Exp Dermatol 2001;10:11–18.
7 Berth‐Jones J. Six area, six sign atopic dermatitis (SASSAD) severity
However, substantial variation remains in the way dis- score: a simple system for monitoring disease activity in atopic der-
ease severity – both clinical signs and symptoms – and matitis. Br J Dermatol 1996;135(Suppl 48):25–30.
QoL are actually measured. Systematic reviews of meas- 8 Severity scoring of atopic dermatitis: the SCORAD index. Consensus

ATOPIC DERMATITIS
urement properties and consensus methodology are Report of the European Task Force on Atopic Dermatitis. Dermatology
1993;186:23–31.
suitable to identify appropriate measurement instru- 9 Costa C, Rilliet A, Nicolet M, Saurat JH. Scoring atopic dermatitis: the

SECTION 3:
ments. It is required that these instruments meet quality simpler the better? Acta Derm Venereol 1989;69:41–5.
standards based on validation studies of high methodo- 10 Rehal B, Armstrong AW. Health outcome measures in atopic dermati-
tis: a systematic review of trends in disease severity and quality‐of‐
logical quality. For measuring clinical signs in trials, the
life instruments 1985‐2010. PLoS One 2011;6:e17520.
HOME initiative has determined that the EASI is the 11 Hill MK, Kheirandish Pishkenari A et al. Recent trends in disease
best available instrument. The SCORAD index has also severity and quality of life instruments for patients with atopic
been extensively validated and is widely used. For dermatitis: A systematic review. J Am Acad Dermatol 2016;
75:906–17.
measuring symptoms in AD clinical trials, HOME 12 Gerbens LA, Chalmers JR, Rogers NK et al. Reporting of symptoms in
has identified POEM as the best available instrument, randomized controlled trials of atopic eczema treatments: a system-
although some measurement properties remain to be atic review. Br J Dermatol 2016;175:67886.
investigated. For quality of life, a variety of instruments 13 Heinl D, Chalmers J, Nankervis H, Apfelbacher CJ. Eczema trials:
quality of life instruments used and their relation to patient‐
exist with systematic reviews suggesting the CADIS as reported outcomes. A systematic review. Acta Derm Venereol 2016;
a promising proxy instrument to assess QoL in children 96:596–601.
with AD. Concerning the impact of children’s AD on the 14 Roekevisch E, Spuls PI, Kuester D et al. Efficacy and safety of systemic
treatments for moderate‐to‐severe atopic dermatitis: a systematic
QoL of their family, it is still unclear which instrument review. J Allergy Clin Immunol 2014;133:429–38.
should be used to measure this outcome. However, the 15 De Vet HC, Terwee CB, Mokkink LB, Knol DL. Measurement in
DFI questionnaire is the most widely used instrument. Medicine: A Practical Guide. Cambridge University Press, 2011.
It may be useful to include several different health‐ 16 Mokkink LB, Terwee CB, Patrick DL et al. The COSMIN study reached
international consensus on taxonomy, terminology, and definitions of
related QoL scales in order to capture the maximum measurement properties for health‐related patient‐reported out-
number of data for certain types of studies. Ideally, par- comes. J Clin Epidemiol 2010;63:737–45.
allel proxy and child data should be collected and, in 17 Boers M, Kirwan JR, Tugwell P et al. The OMERACT Handbook 2016.
Available at: https://omeract.org/resources (accessed 14 January
future, different scales should be devised for use in 2019).
infants, young children, preteens and teenagers. 18 Prinsen CA, Vohra S, Rose MR et al. How to select outcome measure-
As the recommendations made by the HOME initia- ment instruments for outcomes included in a “Core Outcome Set” – a
tive focus on clinical trials, they are particularly useful for practical guideline. Trials 2016;17:449.
19 Leshem YA, Hajar T, Hanifin JM, Simpson EL. What the Eczema
researchers. Even though the recommended ­measurement Area and Severity Index score tells us about the severity of atopic
instruments can also be used in clinical practice, dermatitis: an interpretability study. Br J Dermatol 2015; 172:1353–7.
­clinicians may prefer shorter and simpler instruments as 20 Terwee CB, Bot SD, de Boer MR et al. Quality criteria were proposed
for measurement properties of health status questionnaires. J Clin
feasibility aspects play a much more important role in
Epidemiol 2007;60:34–42.
routine clinical care than in clinical trials. Clear recom- 21 Schmitt J, Williams H. Harmonising Outcome Measures for Eczema
mendations for the measurement of disease severity and (HOME). Report from the First International Consensus Meeting
QoL in clinical practice are currently lacking. For the (HOME 1), 24 July 2010, Munich, Germany. Br J Dermatol 2010;
163:1166–8.
time being, we recommend that clinicians who seek to 22 Schmitt J, Apfelbacher C, Spuls PI et al. The Harmonizing Outcome
implement a measurement instrument for disease Measures for Eczema (HOME) roadmap: a methodological frame-
severity, symptoms or QoL in clinical practice use the work to develop core sets of outcome measurements in dermatol-
recommendations of the HOME initiative as guidance. ogy. J Invest Dermatol 2015;135:24–30.
23 Wolkerstorfer A, de Waard van der Spek FB, Glazenburg EJ et  al.
Nevertheless, other instruments may be used when Scoring the severity of atopic dermatitis: three item severity score as a
feasibility aspects are an obstacle to the use of the recom- rough system for daily practice and as a pre‐screening tool for studies.
mended instruments. Acta Derm Venereol 1999;79:356–9.
24 Vourc’h‐Jourdain M, Barbarot S, Taieb A et  al. Patient‐oriented
SCORAD: a self‐assessment score in atopic dermatitis. A preliminary
feasibility study. Dermatology 2009;218:246–51.
References 25 Housman TS, Patel MJ, Camacho F et al. Use of the Self‐Administered
1 Eichenfield LF, Tom WL, Chamlin SL et al. Guidelines of care for the Eczema Area and Severity Index by parent caregivers: results of a
management of atopic dermatitis: section  1. Diagnosis and assess- validation study. Br J Dermatol 2002;147:1192–8.
ment of atopic dermatitis. J Am Acad Dermatol 2014;70: 338–51. 26 Kunz B, Oranje AP, Labreze L et al. Clinical validation and guidelines
2 Deckert S, Apfelbacher C, Schmitt J. [Measurement of disease severity for the SCORAD index: consensus report of the European Task Force
in dermatology]. Hautarzt 2015;66:699–711. on Atopic Dermatitis. Dermatology 1997;195:10–19.
226  Section 3 Atopic Dermatitis and Related Disorders

27 Oranje AP, Glazenburg EJ, Wolkerstorfer A, de Waard‐van der Spek ures for atopic eczema/dermatitis clinical trials (HOME initiative). Br
FB. Practical issues on interpretation of scoring atopic dermatitis: the J Dermatol 2016;175:69–79.
SCORAD index, objective SCORAD and the three‐item severity score. 50 Spuls PI, Gerbens LA, Simpson E et  al. Patient‐Oriented Eczema
Br J Dermatol 2007;157:645–8. Measure (POEM), a core instrument to measure symptoms in clini-
28 Oranje AP. Practical issues on interpretation of scoring atopic derma- cal trials: a Harmonising Outcome Measures for Ecezema (HOME)
titis: SCORAD Index, objective SCORAD, patient‐oriented SCORAD statement. Br J Dermatol 2017;176:979–84.
and Three‐Item Severity score. Curr Probl Dermatol 2011;41:149–55. 51 Gaunt DM, Metcalfe C, Ridd M. The Patient‐Oriented Eczema
29 Schafer T, Dockery D, Kramer U et al. Experiences with the severity Measure in young children: responsiveness and minimal clinically
scoring of atopic dermatitis in a population of German pre‐school important difference. Allergy 2016;71:1620–5.
children. Br J Dermatol 1997;137:558–62. 52 Valderas JM, Kotzeva A, Espallargues M et al. The impact of measur-
30 Oranje AP, Stalder JF, Taieb A et  al. Scoring of atopic dermatitis by ing patient‐reported outcomes in clinical practice: a systematic review
SCORAD using a training atlas by investigators from different disci- of the literature. Qual Life Res 2008;17:179–93.
plines. ETAC Study Group. Early Treatment of the Atopic Child. 53 U.S. Department of Health and Human Services Food and Drug
Pediatr Allergy Immunol 1997;8:28–34. Administration: Food and Drug Administration guidance for indus-
31 Tripodi S, Panetta V, Pelosi S et al. Measurement of body surface area try on patient‐reported outcome measures: use in medical product
in atopic dermatitis using specific PC software (ScoradCard). Pediatr development to support labeling claims, 2009. Available from: http://
ATOPIC DERMATITIS

Allergy Immunol 2004;15:89–92. www.fda.gov/downloads/Drugs/Guidances/UCM193282.pdf


32 Chalmers JR, Schmitt J, Apfelbacher C et  al. Report from the third (accessed 22 October 2018).
international consensus meeting to harmonise core outcome meas- 54 Sprangers MA. Disregarding clinical trial‐based patient‐reported
SECTION 3:

ures for atopic eczema/dermatitis clinical trials (HOME). Br J outcomes is unwarranted: Five advances to substantiate the scientific
Dermatol 2014;171:1318–25. stringency of quality‐of‐life measurement. Acta Oncol 2010;49:155–63.
33 Schmitt J, Spuls PI, Thomas KS et al. The Harmonising Outcome 55 Study protocol for the World Health Organization project to develop
Measures for Eczema (HOME) statement to assess clinical signs of a Quality of Life assessment instrument (WHOQOL). Qual Life Res
atopic eczema in trials. J Allergy Clin Immunol 2014;134:800–7. 1993;2:153–9.
34 Eichenfield LF, Lucky AW, Boguniewicz M et al. Safety and efficacy of 56 Lewis‐Jones MS, Finlay AY, Dykes PJ. The Infants’ Dermatitis Quality
pimecrolimus (ASM 981) cream 1% in the treatment of mild and mod- of Life Index. Br J Dermatol 2001;144:104–10.
erate atopic dermatitis in children and adolescents. J Am Acad 57 Lewis‐Jones S. Quality of life and childhood atopic dermatitis: the
Dermatol 2002;46:495–504. misery of living with childhood eczema. Int J Clin Pract 2006;60:984–92.
35 Futamura M, Leshem YA, Thomas KS et al. A systematic review of 58 Valderas JM, Alonso J, Guyatt GH. Measuring patient‐reported out-
Investigator Global Assessment (IGA) in atopic dermatitis (AD) tri- comes: moving from clinical trials into clinical practice. Med J Aust
als: Many options, no standards. J Am Acad Dermatol 2016; 2008;189:93–4.
74:288–94. 59 Paller AS, McAlister RO, Doyle JJ, Jackson A. Perceptions of physi-
36 Charman CR, Venn AJ, Williams H. Measuring atopic eczema severity cians and pediatric patients about atopic dermatitis, its impact, and its
visually: which variables are most important to patients? Arch treatment. Clin Pediatr (Phila) 2002;41:323–32.
Dermatol 2005;141:1146–51; discussion 51. 60 Golics CJ, Basra MK, Finlay AY, Salek MS. Adolescents with skin dis-
37 Schmitt J, Langan S, Stamm T, Williams HC. Core outcome domains ease have specific quality of life issues. Dermatology 2009;218:357–66.
for controlled trials and clinical recordkeeping in eczema: interna- 61 Finlay AY, Salek MS, Abeni D et al. Why quality of life measurement
tional multiperspective Delphi consensus process. J Invest Dermatol is important in dermatology clinical practice: An expert‐based opin-
2011;131:623–30. ion statement by the EADV Task Force on Quality of Life. J Eur
38 Schmitt J, Spuls P, Boers M et al. Towards global consensus on out- Acad Dermatol Venereol 2017;31:424–31.
come measures for atopic eczema research: results of the HOME II 62 van Cranenburgh OD, Prinsen CA, Sprangers MA et  al. Health‐
meeting. Allergy 2012;67:1111–17. related quality‐of‐life assessment in dermatologic practice: relevance
39 Research USFaDACfDEa. Draft guidance on pimecrolimus. 2012. and application. Dermatol Clin 2012;30:323–32, x.
40 Thijs J, Krastev T, Weidinger S et al. Biomarkers for atopic dermati- 63 van Valburg RW, Willemsen MG, Dirven‐Meijer PC et al. Quality of
tis: a systematic review and meta‐analysis. Curr Opin Allergy Clin life measurement and its relationship to disease severity in children
Immunol 2015;15:453–60. with atopic dermatitis in general practice. Acta Derm Venereol
41 Wootton CI, Koller K, Lawton S et al. Are accelerometers a useful tool 2011;91:147–51.
for measuring disease activity in children with eczema? Validity, 64 Chernyshov PV, Tomas‐Aragones L, Manolache L et al. Quality of life
responsiveness to change, and acceptability of use in a clinical trial measurement in atopic dermatitis. Position paper of the European
setting. Br J Dermatol 2012;167:1131–7. Academy of Dermatology and Venereology (EADV) Task Force on
42 Lopes C, Soares J, Tavaria F et  al. Chitosan coated textiles may quality of life. J Eur Acad Dermatol Venereol 2017;31:576–93.
improve atopic dermatitis severity by modulating skin staphylococ- 65 Rabin R, de Charro F. EQ‐5D: a measure of health status from the
cal profile: a randomized controlled trial. PLoS One 2015;10:e0142844. EuroQol Group. Ann Med 2001;33:337–43.
43 Gerbens LA, Prinsen CA, Chalmers JR et al. Evaluation of the meas- 66 Davis E, Waters E, Mackinnon A et al. Paediatric quality of life instru-
urement properties of symptom measurement instruments for ments: a review of the impact of the conceptual framework on
atopic eczema: a systematic review. Allergy 2017;72:146–63. ­outcomes. Dev Med Child Neurol 2006;48:311–18.
44 Charman CR, Venn AJ, Williams HC. The patient‐oriented eczema 67 Matterne U, Schmitt J, Diepgen TL, Apfelbacher C. Children and ado-
measure: development and initial validation of a new tool for measur- lescents’ health‐related quality of life in relation to eczema, asthma
ing atopic eczema severity from the patients’ perspective. Arch and hay fever: results from a population‐based cross‐sectional study.
Dermatol 2004;140:1513–19. Qual Life Res 2011;20:1295–305.
45 Charman CR, Venn AJ, Ravenscroft JC, Williams HC. Translating 68 Bullinger M, Brutt AL, Erhart M, Ravens‐Sieberer U. Psychometric
Patient‐Oriented Eczema Measure (POEM) scores into clinical prac- properties of the KINDL‐R questionnaire: results of the BELLA study.
tice by suggesting severity strata derived using anchor‐based meth- Eur Child Adolesc Psychiatry 2008;17(Suppl 1):125–32.
ods. Br J Dermatol 2013;169:1326–32. 69 Varni JW, Limbers CA, Burwinkle TM. Parent proxy‐report of their
46 Dauden E, Sanchez‐Perez J, Prieto M, Roset M. [Validation of the children’s health‐related quality of life: an analysis of 13,878 parents’
Spanish Version of the Itch Severity Scale: the PSEDA study]. Actas reliability and validity across age subgroups using the PedsQL 4.0
Dermosifiliogr 2011;102:527–36. Generic Core Scales. Health Qual Life Outcomes 2007;5:2.
47 Stalder JF, Barbarot S, Wollenberg A et al. Patient‐Oriented SCORAD 70 Upton P, Lawford J, Eiser C. Parent‐child agreement across child
(PO‐SCORAD): a new self‐assessment scale in atopic dermatitis vali- health‐related quality of life instruments: a review of the literature.
dated in Europe. Allergy 2011;66:1114–21. Qual Life Res 2008;17:895–913.
48 Coutanceau C, Stalder JF. Analysis of correlations between patient‐ 71 Eiser C, Morse R. Can parents rate their child’s health‐related qual-
oriented SCORAD (PO‐SCORAD) and other assessment scores of ity of life? Results of a systematic review. Qual Life Res 2001;
atopic dermatitis severity and quality of life. Dermatology 2014; 10:347–57.
229:248–55. 72 Beattie PE, Lewis‐Jones MS. A comparative study of impairment of
49 Chalmers JR, Simpson E, Apfelbacher CJ et al. Report from the fourth quality of life in children with skin disease and children with other
international consensus meeting to harmonize core outcome meas- chronic childhood diseases. Br J Dermatol 2006;155:145–51.
Chapter 16  Severity Scoring and Quality of Life Assessment in Atopic Dermatitis 227

73 Lewis‐Jones MS, Finlay AY. The Children’s Dermatology Life Quality 84 Golics CJ, Basra MK, Finlay AY, Salek S. The impact of disease on
Index (CDLQI): initial validation and practical use. Br J Dermatol family members: a critical aspect of medical care. J R Soc Med
1995;132:942–9. 2013;106:399–407.
74 Heinl D, Prinsen CA, Sach T et  al. Measurement properties of 85 Basra MK, Finlay AY. The family impact of skin diseases: the Greater
quality‐of‐life measurement instruments for infants, children and adoles- Patient concept. Br J Dermatol 2007;156:929–37.
cents with eczema: a systematic review. Br J Dermatol 2017; 176:878–89. 86 Lawson V, Lewis‐Jones MS, Finlay AY et  al. The family impact of
75 Chamlin SL, Cella D, Frieden IJ et al. Development of the Childhood childhood atopic dermatitis: the Dermatitis Family Impact
Atopic Dermatitis Impact Scale: initial validation of a quality‐of‐life Questionnaire. Br J Dermatol 1998;138:107–13.
measure for young children with atopic dermatitis and their families. 87 Dodington SR, Basra MK, Finlay AY, Salek MS. The Dermatitis Family
J Invest Dermatol 2005;125:1106–11. Impact questionnaire: a review of its measurement properties and
76 McKenna SP, Doward LC, Meads DM et al. Quality of life in infants clinical application. Br J Dermatol 2013;169:31–46.
and children with atopic dermatitis: addressing issues of differential 88 Basra MK, Sue‐Ho R, Finlay AY. The Family Dermatology Life Quality
item functioning across countries in multinational clinical trials. Index: measuring the secondary impact of skin disease. Br J Dermatol
Health Qual Life Outcomes 2007;5:45. 2007;156:528–38.
77 Baars RM, Atherton CI, Koopman HM et al. The European DISABKIDS 89 Basra MK, Edmunds O, Salek MS, Finlay AY. Measurement of family
project: development of seven condition‐specific modules to measure impact of skin disease: further validation of the Family Dermatology

ATOPIC DERMATITIS
health related quality of life in children and adolescents. Health Qual Life Quality Index (FDLQI). J Eur Acad Dermatol Venereol
Life Outcomes 2005;3:70. 2008;22:813–21.
78 Chamlin SL, Lai JS, Cella Det al. Childhood Atopic Dermatitis Impact 90 Chernyshov PV, Kaliuzhna LD, Reznikova AA, Basra MK.

SECTION 3:
Scale: reliability, discriminative and concurrent validity, and respon- Comparison of the impairment of family quality of life assessed by
siveness. Arch Dermatol 2007;143:768–72. disease‐specific and dermatology‐specific instruments in children
79 Holme SA, Man I, Sharpe JL et al. The Children’s Dermatology Life with atopic dermatitis. J Eur Acad Dermatol Venereol 2015;
Quality Index: validation of the cartoon version. Br J Dermatol 29:1221–4.
2003;148:285–90. 91 McKenna SP, Whalley D, Dewar AL et al. International development
80 Olsen JR, Gallacher J, Finlay AY et al. Quality of life impact of child- of the Parents’ Index of Quality of Life in Atopic Dermatitis (PIQoL‐
hood skin conditions measured using the Children’s Dermatology AD). Qual Life Res 2005;14:231–41.
Life Quality Index (CDLQI): a meta‐analysis. Br J Dermatol 92 Meads DM, McKenna SP, Kahler K. The quality of life of parents of
2016;174:853–61. children with atopic dermatitis: interpretation of PIQoL‐AD scores.
81 Heinl D, Prinsen CA, Drucker AM et  al. Measurement properties of Qual Life Res 2005;14:2235–45.
quality of life measurement instruments for infants, children and adoles- 93 Kondo‐Endo K, Ohashi Y, Nakagawa H et al. Development and vali-
cents with eczema: protocol for a systematic review. Syst Rev 2016;5:25. dation of a questionnaire measuring quality of life in primary caregiv-
82 Beattie PE, Lewis‐Jones MS. An audit of the impact of a consultation ers of children with atopic dermatitis (QPCAD). Br J Dermatol
with a paediatric dermatology team on quality of life in infants with 2009;161:617–25.
atopic eczema and their families: further validation of the Infants’ 94 Fayers PM, Sprangers MA. Understanding self‐rated health. Lancet
Dermatitis Quality of Life Index and Dermatitis Family Impact score. 2002;359:187–8.
Br J Dermatol 2006;155:1249–55. 95 von Rüden U, Staab D, Kehrt R, Wahn U. Entwicklung und
83 Salek MS, Jung S, Brincat‐Ruffini LA et  al. Clinical experience and Validierung eines krankheitsspezifischen Fragebogens zur Erfassung
psychometric properties of the Children’s Dermatology Life Quality der Lebensqualität von Eltern neurodermitiskranker Kinder. Z f
Index (CDLQI), 1995‐2012. Br J Dermatol 2013;169:734–59. Gesundheitswiss 1999;7:335.
228 

CHA PTER  1 7

Atopic Dermatitis and Related Disorders:


Special Types of Presentation
Nawaf Almutairi
Department of Medicine, Faculty of Medicine, Kuwait University, Kuwait

Pityriasis alba, 228 Infantile seborrhoeic dermatitis, 237 Prurigo pigmentosa, 244
ATOPIC DERMATITIS

Pompholyx, 231 Lichen simplex chronicus, 240


Nummular dermatitis, 235 Prurigo nodularis, 242
SECTION 3:

Abstract may be followed by scaling and lichenification of the skin. Num-


mular eczema is characterized by formation of pruritic discoid
plaques commonly affecting the extensor surfaces of the limbs.
There are several dermatoses that are thought to be related to
Another type of dermatitis is seborrhoeic dermatitis that presents
atopic dermatitis. These include pityriasis alba, seborrhoeic der-
in areas of the skin that are rich in sebaceous glands. Lesions are
matitis, nummular eczema, pompholyx and prurigo. Pityriasis alba
erythematous, scaly plaques or take the form of cradle cap. Lichen
is characterized by hypopigmented patches that are covered with
simplex chronicus and prurigo are highly pruritic lesions that occur
fine scales mainly localized on the face. It could be considered as a
as a result of excessive itching of the skin. Corticosteroids in addi-
minor criterion for diagnosis of atopic dermatitis. Pompholyx usu-
tion to calcineurin inhibitors are the main treatment.
ally appears as deep‐seated vesicles or bullae, located mainly or
commonly on the edges of the fingers, toes, palms and soles. It

Key points • The incidence of pityriasis alba is significantly higher in those


with a history of AD.
• Pompholyx may be responsible for up to 20% of cases of hand
• Up to 10% of infants suffer from one or other form of eczema.
eczema seen in adults. However, it is less common in children.
• Atopic dermatitis (AD) is the most common chronic skin disorder
• Nummular dermatitis is characterized by coin‐shaped lesions
seen in children.
and is more commonly seen in adult males.
• AD is the most common form of eczema seen in children.
• Nummular lesions are the most common nontypical
• AD is not curable, but half of the children affected improve
morphological variants of AD.
between the ages of 5 and 15.
• Lichen simplex chronicus in children is more likely to occur in boys.
• About one quarter of children with AD may have associated
• Prurigo nodularis may be a primary cutaneous disease or a
food allergies.
secondary reaction to pruritus and scratching provoked by a
• Infantile seborrhoeic dermatitis usually clears on its own within
separate cause.
weeks to a few months.

­Pityriasis alba

Definition. Pityriasis alba (PA) is a chronic, benign, Epidemiology. PA is reported from all around the world,
inflammatory dermatosis resulting in disordered pig­ affecting about 1% of the general population. It most
mentation characterized by hypopigmented patches, and ­frequently affects children aged from 3 to 16 years [7,8].
thin plaques with fine scale [1,2]. It is usually localized on The incidence of PA in children ranges from 1.9% to
the face, trunk and extensor surfaces of arms and legs in 9.9%, but is higher in populations with darker skin pho­
children. PA is strongly associated with atopy [3]. It is totypes [9–11]. The role of sex in its incidence has not
considered a minor criterion for diagnosing atopic been established [9,12]. Seasonal variation does occur,
dermatitis (AD) [4]. In the literature, PA is referred to as with the lesions becoming more conspicuous in summer
pityriasis streptogenes, pityriasis simplex faciei, pseudo­ months when the affected areas do not tan like the sur­
leucoderma atopicum, erythema streptogenes, impetigo rounding skin [1,2]. The incidence of PA in patients with
furfuracea, impetigo pityroides, impetigo sicca and pityriasis a history of AD is significantly higher than in the general
maculata [5,6]. population [13].

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 17  Atopic Dermatitis and Related Disorders: Special Types of Presentation 229

microscopic examination of the lesions has revealed


Pathogenesis. The exact aetiology of PA is not known.
degenerative changes in some melanocytes, and a reduced
However, various factors have been implicated. Atopic
number of melanosomes in the keratinocytes [27]. Due to
diathesis has commonly been described as a pathogenic
the similarity of hypopigmentation in PA to postinflam­
factor in PA [14], and some authors believe that PA is a
matory hypopigmentation, some have considered PA to
healing AD, where there is regression of the inflamma­
be a residual postinflammatory manifestation of AD.
tion, thus leading to hypomelanosis [15]. A hypothesis
Carneiro et  al., in their study on dermal dendrocytes of
has been raised that PA and AD are different clinical forms
PA, rolled out this suggestion because, if this were the
within the spectrum of atopy, wherein a patient with a
case, a minimal amount of dendritic cells similar to the
history of atopic disease could develop either towards the
number found in normal skin would be present [16].
AD pole (more inflammatory) or the PA pole (less inflam­
matory) [16].
Clinical features. PA is a long‐term, multistage
Microorganisms such as Pityrosporum, Streptococcus,
c­ondition. The early lesions take the form of mildly

ATOPIC DERMATITIS
Aspergillus and Staphylococcus in addition to parasitic fac­
­erythematous, slightly scaly thin plaques with ill‐defined
tors (Ascaris) have been suggested as causal factors, but
margins; there are usually one to several lesions. PA

SECTION 3:
no relationship has been confirmed [17]. Other contribu­
lesions typically last for several weeks [3]. During the
tory factors, such as temperature variations, relative air
following stage, erythema subsides, leaving behind
humidity and altitude, have also been implicated. Also,
smooth scales. Clearly defined areas of hypopigmenta­
because of the location of lesions – typically in the areas
tion and scaling off at the edges are observed at this
exposed to sunlight  –  it has repeatedly been associated
stage [1,2,5]. The face, particularly the cheeks, is the
with overexposure to light [18,19].
most commonly involved site in children [1], although
In addition, a positive correlation between some per­
the lateral arms, shoulders, neck and anterior thighs
sonal hygiene habits and PA has been recorded. Long and
may also be affected [1–3]. The lesions of PA are typically
frequent baths, mechanical exfoliation and other similar
asymptomatic, although pruritus may accompany the
treatments may contribute to the development of lesions;
condition [2,28] (Figs  17.1 and 17.2). Generalized PA,
this is attributed to the reduction in the level of defensins
with widespread lesions, is usually seen in young adults,
and skin‐protecting factors [1,20]. In contrast, Inanir et al.
and has been reported by Zaynoun et al. [25].
reported an increased incidence of PA in schoolchildren
Previously, other defined clinical variants of PA included
with low parental income that is associated with poor
the extensive and the pigmenting types [25]. The exten­
housing conditions and poor hygiene including frequent
sive type is a less common disease variant. It affects
intestinal parasites [10].
­teenagers and young adults, more often females, and con­
Some nutritional deficiencies are thought to be con­
sists of single, symmetrical lesions that are larger than
nected to the disease. Low levels of serum copper may
those seen in the classical variants. It is not limited to the
account for PA because copper is important in activation
face but involves the neck, trunk and shoulders [25,29].
of tyrosinase in melanocytes, which is critical to melanin
The lesions of pigmenting PA are bluish and are sur­
production [21]. Azelaic acid, oxalic acid and other metab­
rounded by a halo of depigmented skin. There are usually
olites of Malassezia furfur yeasts that normally colonize
numerous lesions, located on the forehead and cheeks
healthy skin may also cause tyrosinase inhibition [19].
[23,30–32].

Pathology. Skin affected by PA shows a nonspecific pic­ Course and prognosis. If left untreated, PA tends to be a
ture consisting of spongiosis with exocytosis, hyperkera­ relatively chronic condition with frequent relapses and a
tosis and dermal perivascular lymphocytic infiltration variable course. The lesions usually persist for several
[3,20,22]. However, these findings may not be helpful in months to a few years [5,8], become more obvious in sum­
the diagnosis of PA. It is known that the main findings are mer due to tanning of surrounding skin, and tend to
in the pilar apparatus. The early stages usually show become dry, scaly and inflamed in winter months [3].
follicular plugging, follicular spongiosis and atrophic Although the disease most often resolves spontaneously
sebaceous glands [23], whereas late stages may show prior to adulthood, repigmentation is a slow process that
regression of keratotic aggregates in hair follicles, mild may take months to years to complete. Although PA is
acanthosis, slight spongiosis and sebaceous gland atro­ chronic and often a cosmetic burden, it is not associated
phy; in some cases the glands are almost completely with any complications or permanent disfigurement [2,3].
absent. The inflammatory infiltrate is predominantly
lymphohistiocytic [24].
Regarding the number of melanocytes in the lesions of Differential diagnosis. The diagnosis of PA is made clinically,
PA, some authors reported them to be fewer in number although certain laboratory tests (e.g. potassium hydroxide
than in the adjoining normal skin, and have suggested preparation) may be used to help differentiate it from
that this might be the possible cause of hypopigmentation other conditions. Biopsy is seldom indicated. Vitiligo
seen in PA [22,25]. On the other hand, some authors have lesions are more sharply demarcated than the lesions of
indicated the number of melanocytes to be normal [26] or PA and completely lack scale and pigment [5]. White
variable [22]. Melanophages have been rarely recorded, patches are accentuated by Wood’s lamp examination
most typically in the papillary dermis [22]. Electron due to complete depigmentation [8,33,34].
230 Section 3  Atopic Dermatitis and Related Disorders
ATOPIC DERMATITIS

Fig. 17.1  Pityriasis alba in a 9‐year‐old child characterized by facial annular hypopigmentation and scaling.
SECTION 3:

That is the reason why tacrolimus is effective when


applied to hypopigmented areas of PA [37].
Pimecrolimus, another calcineurin inhibitor, has a
comparable effect. Topical application of 1% cream
­
­significantly reduces itching, peeling, and extension of
hypopigmented areas [2]. Pimecrolimus therapy is pre­
ferred because of a rarely reported burning sensation
with tacrolimus application. However, both products are
safe and should be preferred over glucocorticoids, par­
ticularly in the case of longlasting lesions on the face.
Although there is no clear relationship between these
drugs and the occurrence of skin infections, a few cases of
molluscum contagiosum and herpes have been reported
during treatment (especially with tacrolimus) [38].
Calcitriol therapy also provides promising results. Its
efficacy is comparable to that of tacrolimus [28]. In addition
Fig. 17.2  Severe form of pityriasis alba on the face of an infant. to its regulatory effect on cell proliferation [39], topical
calcitriol is able to restore epidermal permeability and
the antimicrobial barrier by increasing epidermal and
antimicrobial peptides [40]. These features could account
Pityriasis versicolor can be differentiated from PA using
for its effect in treating PA lesions. Profound improve­
potassium hydroxide preparation, which reveals spores
ment of PA was also observed after 308‐nm excimer laser
and hyphae of mycelium. Wood’s lamp examination may
phototherapy [41].
also be helpful, and it shows yellow fluorescence. Disease
differentiation between psoriasis and PA is based on ­References
patient history and the presence of the typical scaly 1 Weber BM, Sponchiado de Avila LG, Albaneze R et al. Pityriasis
plaques manifested in psoriasis [33,35]. Naevus depig­ alba: a study of pathogenic factors. J Eur Acad Dermatol Venereol
2002; 16:463–8.
mentosus is manifested by a depigmented macule. It is 2 Fujita WH, McCormick CL, Parneix‐Spake A. An exploratory study to
often present at birth and usually becomes more visible at evaluate the efficacy of pimecrolimus cream 1 % for the treatment of
the age of 3 years. It may affect the whole body, and often pityriasis alba. Int J Dermatol 2007;46:700–5.
follows dermatomes [8]. Naevus anaemicus is a congeni­ 3 Vinod S, Singh G, Dash K, Grover S. Clinico epidemiological study
of pityriasis alba. Indian J Dermatol Venereol Leprol 2002;68:338–40.
tal nonpigmentary problem. It can be differentiated from 4 Hanifin JM, Rajka G. Diagnostic features of atopic dermatitis. Acta
PA using diascopy [24]. Derm Venereol Suppl (Stockh) 1980;92:44–7.
The differential diagnosis should also include leprosy, 5 Lin RL, Janniger CK. Pityriasis alba. Cutis 2005;76:21–4.
6 O’Farrell NM. Pityriasis alba. Cutis 1996;61:11–13.
scleroderma, hypopigmentary lesions formed in the 7 Wolf R, Wolf D, Trau H. Pityriasis alba in a psoriatic location. Acta
course of inflammatory skin conditions [8,33] and myco­ Derm Venereol 1992;872:360.
sis fungoides [5,22]. 8 Jadotte YT, Janniger CK. Pityriasis alba revisited: perspectives on
an enigmatic disorder of childhood. Cutis 2011;87:66–72.
9 Elfaituri SS. Pediatric dermatoses in Benghazi, Libya. Indian J Pediatr
Treatment. Many treatments for PA have been suggested. Dermatol 2015;16:64–71.
If inflammation is present, mild to potent topical corti­ 10 Inanir I, Sahin MT, Geundeuz K et al. Prevalence of skin conditions in
costeroids may be used [8]. Other authors suggest topical primary school children in Turkey: differences based on socioeco­
nomic factors. Pediatr Dermatol 2002;19:307–11.
tacrolimus 0.1%, a calcineurin inhibitor, to be effective. As well
11 Fung WK, Lo KK. Prevalence of skin disease among school children
as its anti‐inflammatory properties, it has an activating effect and adolescents in a student health service center in Hong Kong.
on tyrosinase and enhances melanin biosynthesis [36]. Pediatr Dermatol 2000;17:440–6.
Chapter 17  Atopic Dermatitis and Related Disorders: Special Types of Presentation 231

12 Dogra S, Kumar B. Epidemiology of skin disease among school chil­


dren: a study from northern India. Pediatr Dermatol 2003;20:470–3.
­Pompholyx
13 Brenninkmeijer EE, Spuls PI, Legierse CM et  al. Clinical differ-
ences between atopic and atopiform dermatitis. J Am Acad Dermatol Definition. Pompholyx, or dyshidrotic eczema, is a
2008; 58:407–14. chronic relapsing form of dermatitis characterized by the
14 Johnke H, Vach W, Norberg LA et al. A comparison between criteria for
diagnosing atopic eczema in infants. Br J Dermatol 2005;153:352–8.
formation of pruritic, small, deep‐seated, clear ‘sagolike’
15 Schmid‐Grendelmeier P, Simon D, Simon HU et al. Epidemiology, clini­ fluid‐filled vesicles or bullae that appear most commonly
cal features, and immunology of the “intrinsic” (non‐IgE‐mediated) type on the edges of the fingers, toes, palms and soles [1]. Later
of atopic dermatitis (constitutional dermatitis). Allergy 2001;56:841–9. on, scaling, fissures and sometimes lichenification of the
16 Carneiro F, Mendes M, Amaral G, Quaresma J. Tissue immunostain­
ing for factor XIIIa in dermal dendrocytes of pityriasis alba skin skin may occur [2,3]. It is also known as dyshidrosis or
lesions. An Bras Dermatol 2014;89:245–8. vesicular eczema of palms and soles. If pompholyx occurs
17 Zaki S, El Mansy M, El Shazly M. Bacteriological studies of pityriasis mainly on the palmar surface of the palms, the term chei­
alba. Int J Infect Dis 2010;14:S32–3.
18 Blessmann Weber M, Sponchiado de Avila LG, Albaneze R et  al.
ropompholyx may be applied, and if it occurs on the soles
and sides of the toes it is called podopompholyx. Some

ATOPIC DERMATITIS
Pityriasis alba: a study of pathogenetic factors. J Eur Acad Dermatol
Venereol 2002;16:463–8. authors believe that the terms pompholyx and dyshidro­
19 Burkhart CG, Burkhart CN. Pityriasis alba: a condition with possible sis are obsolete and favour a new term such as ‘acute and

SECTION 3:
multiple etiologies. Open Dermatol J 2009;3:7–8.
20 In SI, Yi SW, Kang HY et  al. Clinical and histopathological
recurrent vesicular hand dermatitis’ [4].
­characteristics of pityriasis alba. Clin Exp Dermatol 2009;34:591–7.
21 Galadari E, Helmy M, Ahmed M. Trace elements in serum of pity­
Epidemiology. Pompholyx occurs in 5–20% of patients
riasis alba patients. Int J Dermatol 1992;31:525–6.
22 Vargas‐Ocampo F. Pityriasis alba: a histologic study. Int J Dermatol with hand eczema [5]. It accounted for less than 1% of
1993;32:870–3. initial consultations in a 1‐year Swedish study [6] whereas
23 Landeck L, Visser M, Kezic S, John SM. Genotype‐phenotype associa­ Magina et al. found it to be the third most common type
tions in filaggrin loss‐of‐function mutation carriers. Contact
Dermatitis 2013;68:149–55.
of hand dermatitis (20.3%) in Portugal [7]. Dyshidrotic
24 Miazek N, Michalek I, Pawlowska‐Kisiel M et al. Pityriasis alba— eczema affects individuals aged 4–76 years; the mean age
common disease, enigmatic entity: up‐to‐date review of the litera- is 38 years. The peak incidence of the condition is in
ture. Pediatr Dermatol 2015;32:786–91. patients aged 20–40 years. After middle age, the frequency
25 Zaynoun ST, Aftimos BG, Tenekjian KK et  al. Extensive pityriasis
alba: a histological histochemical and ultrastructural study. Br J of dyshidrotic eczema episodes tends to decrease [8,9]. It
Dermatol 1983;108:83–90. is equally common in male and female individuals.
26 Weedon D. Disorders of Pigmentation, 2nd edn. London: Churchill Pompholyx can be affected by seasonal changes, hot and
Livingstone, 2002:321–41.
27 In S, Yi S, Kang H et al. Clinical and histopathological characteris-
cold temperatures, and is usually worse in the spring and
tics of pityriasis alba. Clin Exp Dermatol 2008;34:591–7. summer months (seasonal or summer pompholyx) [1].
28 Moreno‐Cruz B, Torres‐Álvarez B, Hernández‐Blanco D et al. Double‐
blind, placebo‐controlled, randomized study comparing 0.0003 % cal­
Pathogenesis. The exact cause of pompholyx is not yet
citriol with 0.1 % tacrolimus ointments for the treatment of endemic
pityriasis alba. Dermatol Res Pract 2012;2012:303275. identified. Various factors have been implicated in its
29 Di Lernia V, Ricci C. Progressive and extensive hypomelanosis and causation and/or aggravation [9,10]. The hypothesis of
extensive pityriasis alba: same disease, different names? J Eur Acad sweat gland dysfunction has been disputed because ves­
Dermatol Venereol 2005;19:370–2.
30 Hacker SM. Common disorders of pigmentation: when are more than icles have not been shown to be associated with sweat
cosmetic cover ups required? Postgrad Med 1996;99:177–86. ducts [11]. Thus, the term ‘dyshidrotic eczema’ is consid­
31 du Toit MJ, Jordaan HF. Pigmenting pityriasis alba. Pediatr Dermatol ered a misnomer. Dyshidrotic eczema may be associated
1993;10:1–5.
with atopy and familial atopy. Of all the patients with
32 Dhar S, Kanwar AJ, Dawn G. Pigmenting pityriasis alba. Pediatr
Dermatol 1995;12:197–8. dyshidrosis, 50% are found to have AD. The serum
33 Dıaz Uribe LH. Pitiriase alba: aspectos epidemiologicos, clinicos e immunoglobulin E (IgE) level frequently is increased,
terapeuticos. An Bras Dermatol 2000;75:359–67. even in patients who do not report a personal or familial
3 4 Khalifa ES, Adil AN, Haitham MS. Pityriasis alba versus viti­
ligo. J Saudi Soc Dermatol Dermatol Surg 2013;17:51–4.
history of atopy. Occasionally, dyshidrotic eczema is the
35 Aljabre SH, Alzayir AA, Abdulghani M et al. Pigmentary changes of first manifestation of an atopic diathesis [5].
tinea versicolor in dark‐skinned patients. Int J Dermatol 2001;40: Current knowledge suggests the role of a genetic factor
273–5. in pathogenesis of pompholyx. Monozygotic twins have
36 Kang HY, Choi YM. FK506 increases pigmentation and migration of
human melanocytes. Br J Dermatol 2006;155:1037–40. been found to be simultaneously affected. The pompho­
37 Rigopoulos D, Gregoriou S, Charissi C et  al. Tacrolimus ointment lyx gene has been mapped to band 18q22.1–18q22.3 in the
0.1% in pityriasis alba: an open‐label, randomized, placebo‐con­ autosomal dominant form of familial pompholyx [12].
trolled study. Br J Dermatol 2006;155:152–5.
38 Goksugur N, Ozbostanci B, Goksugur S. Molluscum contagiosum
Mutations on the filaggrin gene with loss of filaggrin
infection associated with pimecrolimus use in pityriasis alba. Pediatr leads to dyskeratinization, increased transepidermal
Dermatol 2007;24:E63–5. water loss and an increase in transepidermal antigen
39 Körver J, Vissers W, Van Rens D et  al. A double‐blind, randomized transfer [13]. Aquaporins, in particular aquaglyceropor­
quantitative comparison of calcitriol ointment and calcipotriol oint­
ment on epidermal cell populations, proliferation and differentiation. ins, are expressed in patients with AD and may be related
Br J Dermatol 2007;156:130–7. to exacerbation and chronicity of pompholyx through
40 Hong SP, Oh Y, Jung M et al. Topical calcitriol restores the impairment increasing transepidermal water loss [14].
of epidermal permeability and antimicrobial barriers induced by
Emotional stress [15] is a possible factor in dyshidrotic
corticosteroids. Br J Dermatol 2010;162:1251–60.
41 Al‐Mutairi N, Hadad AA. Efficacy of 308‐nm xenon chloride excimer eczema. Many patients report recurrences of pompholyx
laser in pityriasis alba. Dermatol Surg 2012;38:604–9. during stressful periods. Environmental factors (e.g. seasonal
232 Section 3  Atopic Dermatitis and Related Disorders

changes, hot or cold temperatures, humidity) have been frequent cause of pompholyx than that of nickel. However,
reported to exacerbate pompholyx. Pompholyx caused by simultaneous occurrence of both allergies is more
ultraviolet (UV)A exposure may possibly be considered a common [22].
variation of seasonal (summer) pompholyx [16]. Nalluri Other factors including aspirin ingestion, oral contra­
and Rhodes suggested the term photoaggravated pom­ ceptives and cigarette smoking have been reported as pos­
pholyx in 2016 [17]. sible causes of inducing or aggravating pompholyx [24].
Sometimes, a distant fungal or candidal infection can
cause palmar pompholyx as an id reaction. Pompholyx
Pathology. In acute lesions, the main features are spongi­
occasionally resolves when tinea pedis infection is treated,
osis and intraepidermal vesiculation. There is a mild
then relapses when the fungal infection recurs, support­
superficial perivascular lymphohistiocytic infiltrate with
ing the existence of this reaction pattern [18]. Bacterial
exocytosis of lymphocytes into spongiotic zones. However,
and viral infections, especially human immunodeficiency
eosinophils can also be present. The compact, thickened
virus (HIV), can also provoke pompholyx. Pompholyx
ATOPIC DERMATITIS

stratum corneum remains intact, and with normal epider­


has also been described as a manifestation of sympto­
mal thickness. As lesions progress to chronicity, signs of
matic HIV infection. Some patients do not respond to
SECTION 3:

active inflammation and spongiosis start to diminish,


topical and systemic treatments for pompholyx, and their
acanthosis and parakeratosis predominate, and serum
condition improves only with antiretroviral therapy [19].
may be identified within the stratum corneum [25].
Pompholyx‐like eczematous eruptions with the use of
Features of lichen simplex chronicus may be the hallmark
intravenous immunoglobulin (IVIG) infusions have been
of chronic lesions. A periodic acid–Schiff (PAS) stain
reported, and pompholyx is now identified as one of the
should be performed on vesicular lesions of the palms and
most important cutaneous adverse effects of IVIG, being
soles, because fungal infection may mimic pompholyx his­
present in 62.5% of the patients reported, with most of
tologically. The intraepidermal part of the eccrine sweat
them developing the lesions after their first IVIG treat­
duct (acrosyringium) is not altered even by spongiosis,
ment [20,21].
although histological study of pompholyx showed that
Exogenous factors (e.g. contact dermatitis to nickel, bal­
sweat ducts were often pushed aside by the tense vesicles
sam, cobalt; sensitivity to ingested metals) may trigger
or passed between them [26].
episodes. These antigens may act as haptens with a spe­
cific affinity for palmoplantar proteins of the stratum luci­
dum of the epidermis. The binding of these haptens to Clinical features. There is a sudden onset of symmetrical
tissue receptor sites may cause systemic allergic dermati­ crops of vesicles and/or bullae (filled with clear fluid) on
tis and pompholyx [22,23]. Nickel allergy has been con­ the palms and lateral aspects of the fingers. The feet, the
nected to pompholyx. Increased nickel excretion in the soles and the lateral aspects of toes also may be affected.
urine has been reported during exacerbations of pompho­ Burning pain or pruritus occasionally may be experienced
lyx. Low‐nickel diets have reportedly decreased the before vesicles appear. Palms and soles may be red and
frequency and severity of pompholyx flares. A high palmo­ wet with perspiration. Vesicles are deep‐seated and have
plantar perspiration rate has been suggested to result in a a tapioca pudding‐like appearance, without surrounding
local concentration of metal salts that may provoke erythema (Figs 17.3 and 17.4). The vesicles usually persist
the vesicular reaction. The oral ingestion of cobalt is a less for 3–4 weeks. They may become large, form bullae and

Fig. 17.3  Pompholyx affecting the palms of a young boy presenting with crops of tense clear vesicles.
Chapter 17  Atopic Dermatitis and Related Disorders: Special Types of Presentation 233

ATOPIC DERMATITIS
SECTION 3:
Fig. 17.4  Pompholyx affecting the distal parts of the fingers in a young boy.

The index was found to be a simple standardized method


for assessing the condition and can be used to assess disease
severity and treatment effectiveness [28].

Prognosis. The lesions appear abruptly and generally


persist for 2–3 weeks. Vesicle outbreaks may occur in
waves [29]. Episodes vary in frequency from once per
month to once per year. Each episode usually lasts for few
days to a few weeks, and subsides spontaneously even
without treatment. Secondary bacterial infection of vesi­
cles or bullae can rarely result in cellulitis, lymphangitis
and septicaemia. Dystrophic nail changes may develop,
characterized by transverse ridging, thickening, discol­
ouration and pitting. Pompholyx has no associated mor­
tality, although some severe cases can cause disability by
Fig. 17.5  Severe form of pompholyx presenting with dense bullae on the compromising the function of the hands due to extensive
palms of a young atopic child. bullae (e.g. in schoolchildren). Hsu et  al. in their study
determined that dyshidrosis is a risk factor in the devel­
opment of herpes zoster. Because of the strong association
become confluent (Fig.  17.5). Vesicles typically resolve between the two conditions, the authors have suggested
without rupturing, followed by desquamation. Patients that patients should be warned about the risk [30].
with pompholyx may report a variety of factors that pos­
sibly are related to eruptions such as emotional stress and Differential diagnosis. Palmoplantar pustulosis (PPP) is
hot weather, and more than 50% of individuals with the a chronic and intensely inflammatory skin disease with
pompholyx type of presentation had an atopic diathesis. pustules, erythema and scaling localized to the palms and
With longstanding disease, fingernails may reveal dys­ soles. Both PPP and pompholyx share similar clinical as
trophic changes (e.g. irregular, transverse ridging; pitting; well as histological features [2]. Therefore, the differential
thickening; discolouration). Interdigital maceration and diagnosis of these two diseases can be quite frustrating.
desquamation of the interdigital spaces often are present, Several histological features can differentiate between
despite the possible absence of a dermatophyte infection. PPP and pompholyx. The presence of confluent parakera­
Secondary infection may lead to pustular or haemor­ tosis, psoriasiform epidermal hyperplasia, clubbing and
rhagic fluid or crusts associated with the blistering. There anastomosing of the rete ridges, loss of the granular layer,
may be surrounding erythema of the hands with associated tortuous capillaries in the papillary dermis touching
pain rather than the more characteristic itch associated the undersurface of the epidermis and extravasated
with the acute blisters. Cellulitis and lymphangitis may erythrocytes in PPP patients could serve as useful ‘clues’
develop [1,27]. for ­differentiation [31].
The Dyshidrotic Eczema Area and Severity Index (DASI) Keratolysis exfoliativa is characterized clinically by
was developed based on severity grades for the number of intermittent air‐filled blisters with centrifugal collarette of
vesicles per square centimetre (V), erythema (E), desquama­ the palms and soles. It is histopathologically characterized
tion (S), itch (I), and the extent of affected areas (A) with by cleavage within the stratum corneum and partial degra­
defined score points (p) as: DASI = (pV + pE = pS + pI) × pA. dation of corneodesmosomes in the mid stratum corneum;
234 Section 3  Atopic Dermatitis and Related Disorders

normal expression of corneodesmosomal components is phosphodiesterase‐4 (PDE4) inhibitors, alitretinoin and


reported by immunofluorescence [32]. Dyshidrosiform phototherapy with high‐dose UVA‐1 and UV‐free photo­
pemphigoid may have vesicles (vesicular pemphigoid) or therapy have been introduced [3].
be evident as erythema and swelling with bullae on the
palmoplantar areas [33]. Histopathology and immunofluo­ ­References
rescence findings are of help in making the diagnosis. 1 Wollina U. Pompholyx: a review of clinical features, differential
diagnosis, and management. Am J Clin Dermatol 2010;11: 305–14.
Pompholyx may be differentiated from blistering fungus
2 Veien NK. Acute and recurrent vesicular hand dermatitis. Dermatol
infection by scraping of the skin surface scale to identify Clin 2009;3:337–53.
the causative organism under the microscope or by culture 3 Wollina U. Pompholyx: what’s new? Expert Opin Investig Drugs
in the laboratory. Infantile acropustulosis mainly affects 2008;6:897–904.
4 Storrs FJ. Acute and recurrent vesicular hand dermatitis not pompho­
infants at a younger age (first year of life) and the lesions lyx or dyshidrosis. Arch Dermatol 2007;143:1578–80.
are pustular rather than vesicular. Histologically, neutro­ 5 Lodi A, Betti R, Chiarelli G et al. Epidemiological, clinical and aller-
phils and eosinophils predominate [34]. gological observations on pompholyx. Contact Dermatitis 1992;
ATOPIC DERMATITIS

Other conditions to consider in the differential diagno­ 26:17–21.


6 Lofgren SM, Warshaw EM. Dyshidrosis: epidemiology, clinical

sis of pompholyx include palmoplantar pustular psoriasis,
SECTION 3:

characteristics, and therapy. Dermatitis 2006;17:165–81.


linear IgA disease with (haemorrhagic) pompholyx, 7 Magina S, Barros MA, Ferreira JA, Mesquita‐Guimaraes J. Atopy,
scabies, hand, foot and mouth disease, dermatitis herpeti­ nickel sensitivity, occupation, and clinical patterns in different types
of hand dermatitis. Am J Contact Dermat 2003;14:63–8.
formis, recurrent palmoplantar hidradenitis and papular– 8 Amini S. Dyshidrotic eczema. Available at: http://emedicine.

purpuric glove and socks syndrome [35]. medscape.com/article/1122527 (accessed 25 November 2018).
9 Wollina U, Karamfilov T. Adjuvant botulinum toxin A in dyshidrotic
Treatment. Management of pompholyx includes identifi­ hand eczema: a controlled prospective pilot study with left‐right com­
parison. J Eur Acad Dermatol Venereol 2002;16:40–2.
cation and treatment of underlying causes such as avoid­ 10 Pitche P, Boukari M, Tchangai‐Walla K. Factors associated with pal­
ing emotional stress and hot weather and treating moplantar or plantar pompholyx: a case‐control study. Ann Dermatol
dermatophyte infections. A low‐nickel diet or a low‐cobalt Venereol 2006;133:139–43.
11 Lee WJ, Lee DW, Kim CH et al. Pompholyx with bile‐coloured vesi-
diet should be considered for nickel‐sensitive or cobalt‐ cles in a patient with jaundice: are sweat ducts involved in the
sensitive patients respectively. Nickel chelators, such as development of pompholyx? J Eur Acad Dermatol Venereol 2010;24:
disulfiram (Antabuse), occasionally are used in nickel‐ 235–6.
sensitive patients who demonstrate a positive oral provo­ 12 Molin S, Vollmer S, Weiss EH et al. Filaggrin mutations may confer
susceptibility to chronic hand eczema characterized by combined
cation test [22,36]. allergic and irritant contact dermatitis. Br J Dermatol 2009;161:801–7.
In the acute stage, compresses with a 1 : 10 000 solution 13 Soler DC, Bai X, Ortega L et al. The key role of aquaporin 3 and aqua­
of potassium permanganate or 10% aluminium acetate porin 10 in the pathogenesis of pompholyx. Med Hypotheses
2015;84:498–503.
should be applied until resolution of bullae, which
14 Guillet MH, Wierzbicka E, Guillet S et al. A 3‐year causative study
may take several days. Drainage of large bullae with a of pompholyx in 120 patients. Arch Dermatol 2007;143:1504–8.
sterile syringe may be needed to relieve discomfort. 15 Shenefelt PD. Update on psychodermatological disorders. Expert Rev
Antibiotics are used for cases of pompholyx with second­ Dermatol 2010;5:95–107.
16 Tamer E, Ilhan MN, Polat M et al. Prevalence of skin diseases among
ary impetiginization. pediatric patients in Turkey. J Dermatol 2008;35:413–18.
Topical corticosteroids are the mainstay of treatment. 17 Nalluri R, Rhodes LE. Photoaggravated pompholyx. Photodermatol
Steroid potency choice is based on the patient’s response Photoimmunol Photomed 2016;12:1–3.
to treatment. However, the higher‐strength steroids are 18 Kaaman T, Torssander J. Dermatophytid—a misdiagnosed entity?
Acta Derm Venereol 1983;63:404–8.
usually necessary for disease control, as percutaneous 19 MacConnachie AA, Smith CC. Pompholyx eczema as a manifestation
absorption in palmoplantar areas is very limited. Systemic of HIV infection, response to antiretroviral therapy. Acta Derm
corticosteroids can also be used for severe episodes. Venereol 2007;87:378–9.
20 Gerstenblith MR, Antony AK, Junkins‐Hopkins JM et al. Pompholyx
Topical calcineurin inhibitors such as topical tacrolimus and eczematous reactions associated with intravenous immunoglob­
or pimecrolimus may be helpful [37]. Advantages of topi­ ulin therapy. J Am Acad Dermatol 2012;66:312–16.
cal calcineurin inhibitors over topical corticosteroids 21 Kotan D, Erdem T, Acar BA, Boluk A. Dyshidrotic eczema associated
include the lack of development of tachyphylaxis, telangi­ with the use of IVIg. BMJ Case Rep 2013;15:2013.
22 Stuckert J, Nedorost S. Low‐cobalt diet for dyshidrotic eczema

ectasias, and thinning and atrophy of the skin [3]. For patients. Contact Dermatitis 2008;59:361–5.
severe refractory pompholyx, azathioprine [38], metho­ 23 Thierse HJ, Gamerdinger K, Junkes C et al. T cell receptor (TCR) inter­
trexate [39], mycophenolate mofetil [40], ciclosporin [41] action with haptens: metal ions as non‐classical haptens. Toxicology
2005;209:101–7.
or etanercept [42] may be helpful. Paradoxically, although
24 Sehgal V, Srivastava G, Aggarwal A et al. Hand dermatitis/eczema:
sometimes linked as a possible cause, UVA or UVA‐1 Current management strategy. J Dermatol 2010;37:593–610.
alone or with oral or topical psoralen (PUVA) has been 25 Weedon D. Skin Pathology, 2nd edn. Edinburgh: Churchill‐

used in treatment [43,44]. Livingstone, 2002.
26 Kuntzer H, Wurzel RM, Wolff HH. Are acrosyringia involved in the
Botulinum toxin A injections may be helpful in some pathogenesis of ‘dyshidrosis’? Am J Dermatopathol 1986;8:109–16.
patients. Botulinum toxin A intradermal injections as an 27 Mittal M. Images in emergency medicine. Ann Emerg Med 2008;
adjuvant to topical corticosteroids cause reduction of pru­ 52:12.
ritus and vesiculation [9]. Tap water iontophoresis with 28 Vocks E, Plotz SG, Ring J. The Dyshidrotic Eczema Area and Severity
Index – A score developed for the assessment of dyshidrotic eczema.
pulsed direct current may be helpful as adjuvant treat­ Dermatology 1999;198:265–9.
ment [45]. New types of anti‐inflammatory oral drugs, 29 Man I, Ibbotson SH, Ferguson J. Photoinduced pompholyx: a report of
such as topical bexarotene, leukotriene inhibitors and 5 cases. J Am Acad Dermatol 2004;50:55–60.
Chapter 17  Atopic Dermatitis and Related Disorders: Special Types of Presentation 235

30 Hsu CY, Wang YC, Kao CH. Dyshidrosis is a risk factor for herpes increased susceptibility to allergic contact dermatitis to
zoster. J Eur Acad Dermatol Venereol 2015;27:2177–83.
materials such as metals, soaps and chemicals that may
31 Yoon S, Park HS, Lee JH, Cho S. Histological differentiation

between palmoplantar pustulosis and pompholyx. J Eur Acad exacerbate the condition [5].
Dermatol Venereol 2013;27:889–93. Some studies have underlined that bacterial allergens, in
32 Chang YY, van der Velden J, van der Wier G et al. Keratolysis exfolia­ particular staphylococcal allergens, released from a focus
tiva (dyshidrosis lamellosa sicca): a distinct peeling entity. Br J
Dermatol 2012;167:1076–84.
of infection (nose, teeth, tonsils, sinuses, bronchi) can cause
33 Yasuda M, Miyachi Y, Utani A. Two cases of dyshidrosiform pemphi­ allergic sensitization and activate the plaques [6]. Elevated
goid with different presentations. Clin Exp Dermatol 2009;34:e151–3. antistreptolysin and antistaphylolysin levels have been
34 Mazereeuw‐Hautier J. [Infantile acropustulosis]. Presse Med 2004; demonstrated in patients with nummular ­dermatitis [7].
33:1352–4.
35 Kluger N, Moguelet P, Khosrotehrani K et al. Idiopathic recurrent pal­ The relationship of nummular dermatitis to atopy has
moplantar hidradenitis: a case with late onset and long‐lasting been debated [8]. There are differing views on the possi­
course. Clin Exp Dermatol 2007;32:217–18. ble role of atopy in subjects with nummular dermatitis
36 Menne T, Kaaber K. Treatment of pompholyx due to nickel allergy
but recent studies have indicated that nummular lesions

ATOPIC DERMATITIS
with chelating agents. Contact Dermatitis 1978;4:289–90.
37 Schurmeyer‐Horst F, Luger TA, Bohm M. Long‐term efficacy of
are the most common nontypical morphological variants
occlusive therapy with topical pimecrolimus in severe dyshidrosi­ of AD in both children and adults [9]. In children, a vari­

SECTION 3:
form hand and foot eczema. Dermatology 2007;214:99–100. ant of AD may manifest as disseminated nummular
38 Scerri L. Azathioprine in dermatological practice. An overview with
special emphasis on its use in non‐bullous inflammatory dermatoses. lesions. Nummular dermatitis may or may not be associ­
Adv Exp Med Biol 1999;455:343–8. ated with high total IgE levels, suggesting no common
39 Egan CA, Ralis TM, Meadows KP et al. Low‐dose oral methotrexate role for IgE in both conditions [10].
treatment for recalcitrant palmoplantar pompholyx. J Am Acad
Contact dermatitis, either irritant or allergic, has been
Dermatol 1999;40:612–14.
40 Pickenacker A, Luger TA, Schwatz T. Dyshidrotic eczema treated with connected to nummular dermatitis. Irritant contact der­
mycophenolate mofetil. Arch Dermatol 1998;134:378–9. matitis may present with nummular lesions. Nummular
41 Petersen CS, Menne T. Cyclosporin A responsive chronic severe vesic­ lesions have been described in case reports of hypersensi­
ular hand eczema. Acta Derm Venereol 1992;72:436–7.
42 Ogden S, Clayton TH, Goodfield MJ. Recalcitrant hand pompholyx:
tivity to ethylenediamine [11] and nickel [12].
variable response to etanercept. Clin Exp Dermatol 2006;31:145–6.
43 Petering H, Breuer C, Herbst R et al. Comparison of localized high‐dose Pathology. Histopathological changes of subacute
UVA1 irradiation versus topical cream psoralen‐UVA for treatment spongiotic dermatitis are seen. There is mild to moderate
of chronic vesicular dyshidrotic eczema. J Am Acad Dermatol
2004;50:68–72. spongiosis, usually without vesiculation, together with
44 Polderman MC, Govaert JC, le Cessie S et al. A double‐blind placebo‐ irregular acanthosis with some exocytosis of inflamma­
controlled trial of UVA‐1 in the treatment of dyshidrotic eczema. Clin tory cells. There is a superficial perivascular infiltrate,
Exp Dermatol 2003;28:584–7.
45 Odia S, Vocks E, Rakoski J, Ring J. Successful treatment of dyshidrotic
predominantly of lymphocytes with some eosinophils
hand eczema using tap water iontophoresis with pulsed direct cur­ and occasional neutrophils. Chronic lesions demonstrate
rent. Acta Derm Venereol 1996;76:472–4. epidermal hyperplasia, hyperkeratosis and a pronounced
granular cell layer [13].

­Nummular dermatitis Clinical features. Nummular dermatitis is characterized


by coin‐shaped or discoid plaques of eczema that arise
Definition. Nummular (meaning round or ‘coin‐shaped’; quite rapidly from the confluence of tiny papules and
synonym: discoid) dermatitis or eczema is an inflammatory papulo‐vesicles measuring 1–3  cm, only occasionally
skin condition characterized by the presence of pruritic being larger. In the acute phase, the lesions are red, weep­
coin‐shaped plaques commonly situated on the extensor ing, crusted and highly irritable. They then progress to a
surfaces of the limbs. Other names used in the literature less vesicular and scalier stage, often with central clear­
have included discoid eczema and orbicular eczema. It was ing and peripheral extension, forming ring‐shaped or
first described by Devergie in 1857 [1]. Since then, it has annular lesions. The surface is usually weeping or
been reported in all age groups and in all body areas. crusted and the margins are flat. As they fade, they leave
dry and hyperpigmented areas that gradually lighten.
Epidemiology. Nummular lesions of eczema are not However, pigment may never completely fade, particu­
uncommon. The prevalence of nummular dermatitis larly when located below the knee. The itch is often
ranges from 0.1% to 9.1%. Males are affected slightly more severe with excoriations and lichenification secondary to
often and at a later age than females. It is less common in scratching [14]. Sites of predilection include the dorsum
children [2] and has no racial predilection. of the hands, the extensor surface of the forearms, the
lower part of the legs, the outer aspect of the thighs, and
Pathogenesis. The exact pathogenesis of nummular der­ the posterior aspect of the trunk [15] (Fig. 17.6). Face and
matitis is still obscure. It may represent a reaction pattern, scalp are usually spared [16]. Nummular dermatitis usu­
which can be induced and perpetuated by a variety of fac­ ally takes a very chronic course with remissions and
tors [3]. Nummular dermatitis is frequently accompanied exacerbations. Autoeczematization (i.e. lesional spread
by xerosis. Dryness of the skin results in dysfunction of from the initial focal site) may account for the presence of
the epidermal lipid barrier that allows environmental multiple plaques. A particularly therapy‐resistant variety
allergens to penetrate the skin and induce dermatitis [4]. has been termed ‘oid‐oid disease’ (Sulzberger–Garbe
Impairment of the cutaneous barrier may also result in syndrome) [17].
236 Section 3  Atopic Dermatitis and Related Disorders
ATOPIC DERMATITIS
SECTION 3:

Fig. 17.6  Nummular eczema affecting different


parts of the body.

inflammation and treat any associated infection. Lukewarm


Prognosis. Nummular dermatitis has usually a chronic
or cool baths can help in rehydration of the skin, and thus
relapsing course, and may persist for many months and
reduce itching. Application of the medicine to damp skin
require long‐term therapy. It often waxes and wanes with
allows more effective penetration and faster healing. Skin
the season, increasing in winter or in a cold and dry cli­
moisturizers should be then applied to wet skin, some­
mate. It may improve with sun exposure or humidity or
times under occlusion.
with moisturizer use [18].
Medium‐ to high‐potency topical corticosteroid oint­
ments and topical immune modulators (tacrolimus and
Differential diagnosis. Nummular dermatitis should be pimecrolimus) [21] represent the mainstay treatment
differentiated from tinea corporis. Tinea corporis may be options to reduce inflammation. Tar preparations have
excluded by scraping and microscopically analysing a also been used successfully. Systemic steroids may be
potassium hydroxide preparation of a lesion. Differentiating required in cases of severe, generalized eruptions.
between discoid eczema and childhood psoriasis on clini­ Phototherapy may be of help in generalized and pro­
cal grounds can be difficult. Often in discoid eczema, vesic­ longed eruptions [22]. Broadband or narrowband UVB is
ular lesions with less well‐defined margins to the plaques most commonly used. Immune suppressive medications
and a more severe degree of pruritus are found. Histological such as methotrexate have been found to be efficacious in
examination of skin biopsy may be needed. severely affected patients [23]. Oral antihistamines or
Nummular dermatitis must be distinguished from sedatives may help reduce itching and improve sleep.
nummular lesions of AD. A history of atopy, IgE and Topical antibiotics may help to avoid impetiginization but
radio‐allergosorbent test (RAST) determination and skin oral antibiotics should be used in cases of secondary
test may be diagnostically helpful [19]. Many studies have infection.
recommended patch testing in patients with nummular
dermatitis refractory to treatment to differentiate contact ­References
and nummular dermatitis [20]. Other conditions to be con­ 1 Devergie MGA. Traité Pratique des Maladies de la Peau. Paris: Librairie
de Victor Masson, 1857;2:238.
sidered in differential diagnosis are fixed drug eruption, 2 Ozkaya E. Adult‐onset atopic dermatitis. J Am Acad Dermatol

lichen simplex chronicus, pityriasis rosea, seborrhoeic der­ 2005;52:579–82.
matitis, Bowen disease and mycosis fungoides 3 Khurana S, Jain V, Aggarwal K et al. Patch testing in discoid eczema.
J Dermatol 2002;29:763–7.
4 Aoyama H, Tanaka M, Hara M et al. Nummular eczema: An addition
Treatment. The aim of treatment is to rehydrate the of senile xerosis and unique cutaneous reactivities to environmental
skin  with repair of the epidermal lipid barrier, reduce aeroallergens. Dermatology 1999;199:135–9.
Chapter 17  Atopic Dermatitis and Related Disorders: Special Types of Presentation 237

5 Bonamonte D, Foti C, Vestita M et al. Nummular eczema and contact


allergy: a retrospective study. Dermatitis 2012;23:153–7. Pathogenesis. The aetiology of SD is not completely
6 Kim W, Ko H, Kim M et al. Features of Staphylococcus aureus coloni­ understood. Its presence has been associated with over­
zation in patients with nummular eczema. Br J Dermatol 2013;
growth of yeast species such as Malassezia (previously
168:656–82.
7 Hellgren L, Mobacken H. Nummular eczema – clinical and statistical Pityrosporum) that normally inhabit sebaceous skin [8].
data. Acta Dermatol Venereol (Stockh) 1969;49:189–96. Various studies have been performed on the role of
8 Taieb A. Atopic dermatitis: definition, epidemiology, natural history, Malassezia and SD in infants and adults [9,10]. The genus
severity and scores. Ann Dermatol Venereol 2005;132:S35–43.
9 Pugliarello S, Cozzi A, Gisondi P et al. Phenotypes of atopic dermatitis.
Malassezia, initially thought to consist of only one species,
J Dtsch Dermatol Ges 2011;9:12–20. M. furfur, is now known to comprise seven species [11].
10 Kreuger GG, Kahn G, Weston WL et  al. IgE levels in nummular Six different species of Malassezia were recovered from
eczema and ichthyosis. Arch Dermatol 1973;107:56–8. different dermatosis cases and different anatomical sites
11 Caraffini S, Lisi P. Nummular dermatitis‐like eruption from ethylen­
ediamine hydrochloride in two children. Contact Dermatitis [12]. They are normal resident skin flora [12,13]. Under
1987;17:313–14. certain circumstances, M. furfur may behave as an oppor­

ATOPIC DERMATITIS
12 Patrizi A, Rizzoli L, Vincenzi C et  al. Sensitization to thimerosal in tunistic pathogen or cause or aggravate a spectrum of
atopic children. Contact Dermatitis 1999;40:94–7.
13 Wu H, Schapiro B, Harrist T. Noninfectious vesiculobullous and
skin diseases such as psoriasis, atopic dermatitis, tinea

SECTION 3:
vesiculopustular diseases. In: David E, Rosalie J, Bernett L (eds) versicolor or pityrosporum folliculitis [14].
Lever’s Histopathology of the Skin, 9th edn. Philadelphia: Several lines of evidence suggest a pathogenic role for
Lippincott Williams & Wilkins, 2005:244. yeasts of the genus Malassezia in SD [14–16]. Among the
14 Jiamton S, Tangjaturonrusamee C, Kulthanan K. Clinical features
and aggravating factors in nummular eczema in Thais. Asian Pac J multiple chemical entities that are effective in treating
Allergy Immunol 2013;31:36–42. seborrhoeic dermatitis, such as azoles, hydroxypyridones,
15 Weedon D. Disorders of Pigmentation, 2nd edn. London: Churchill allylamines, selenium and zinc, the sole common mecha­
Livingstone, 2002:321–41.
nism of action is antifungal activity [17,18]. Moreover, the
16 Krogh HK. Nummular eczema. Acta Derm Venereol 1960;40:114–26.
17 Sulzberger MB, Garbe W. Nine cases of distinctive exudative discoid number of yeast colonizations of skin was found to be
and lichenoid dermatosis. Arch Dermatol Venereol 1985;65:164–7. decreased with the use of antifungals and to rise again
18 Kulthanan K, Samutrapong P, Tuchina P. Adult‐onset atopic dermati­ when relapse occurred. Malassezia was shown to have
tis: a cross‐sectional study of natural history and clinical manifesta­
tion. Asian Pac J Allergy Immunol 2007;25:207–14.
lipase activity, which hydrolyses human sebum triglycer­
19 Agarwal U, Besarwal R, Gupta R et  al. Hand eczema. Indian J ides and releases unsaturated fatty acids such as oleic and
Dermatol 2014;59:213–24. arachidonic acid [19,20]. These metabolites cause disrup­
20 Bonamonte D, Foti C, Vestita M et al. Nummular eczema and con- tion of epidermal barrier function and induce keratino­
tact allergy: a retrospective study. Dermatitis 2012;23:153–7.
21 Bukhari IA. Successful treatment of chronic persistent vesicular cytes to produce pro‐inflammatory cytokines such as
hand dermatitis with topical pimecrolimus. Saudi Med J 2005; interleukin (IL)‐1α, IL‐6, IL‐8 and tumour necrosis factor
26:1989–91. (TNF)‐α, thus initiating an inflammatory response.
22 Gambichler T. Management of atopic dermatitis using photo (chemo)
therapy. Arch Dermatol Res 2009;301:197–203.
Furthermore, arachidonic acid can be a source of prosta­
23 Roberts H, Orchard D. Methotrexate is a safe and effective treatment glandins, which are pro‐inflammatory mediators that can
for paediatric discoid (nummular) eczema: a case series of 25 children. cause inflammation via neutrophil recruitment and vaso­
Australas J Dermatol 2010;51:128–30. dilation [21,22]. On the other hand, there is no simple rela­
tionship between yeast number and severity of SD, and
healthy skin may have a similar organism load to that of
­Infantile seborrhoeic dermatitis SD. Moreover, SD lesions that occur in HIV‐infected
patients do not harbour a higher Malassezia load than
Definition. Seborrhoeic dermatitis (SD) is a chronic, recur­ HIV‐infected patients without SD [23]. It seems that indi­
rent, inflammatory papulosquamous skin disorder that vidual predispositions and host interactions with
typically affects areas of the skin that are well supplied with Malassezia, rather than the mere presence of Malassezia,
sebaceous glands (face, scalp, chest, etc.). Its occurrence in contribute to the pathogenesis of SD.
early infancy may be related to hormone‐dependent sebum SD occurs predominantly in areas of the skin with
production, which decreases in childhood and increases active sebaceous glands, and is often associated with
again in adolescence and adulthood [1,2] (see Chapter 21). sebum overproduction. It also has a strong time correla­
tion with sebaceous gland activity and higher sebum
Epidemiology. The prevalence rate of SD is 3–5% of the secretion, with cradle cap after birth, increased incidence
total population. Dandruff, which is the mildest form throughout the teens, then a decrease between the third
of  this dermatitis, is probably more common, affecting and sixth decades [24]. However, SD patients may have
15–20% of the population [3]. SD has two peaks of normal sebum production, and people with excessive
occurrence, commonly appearing in both infancy (cradle sebum production are sometimes free from SD [25]. These
cap) and after puberty (dandruff) [4]. In a survey on findings suggest that although sebaceous gland activity
Australian children, the highest prevalence of SD was in strongly correlates with SD, the amount of sebum pro­
the first 3 months of life, decreasing rapidly by the age of duced per se does not appear to be the decisive risk
1 year [5]. Because of this onset and resolution, it has been factor.
suggested that transplacental hormones (the levels of Abnormalities of lipid composition may also play a role
which are elevated in infancy and decrease by the first in the development of SD. Triglycerides and squalene
year of life) may be a causative factor [6,7]. were found to be reduced in SD patients compared to
238 Section 3  Atopic Dermatitis and Related Disorders

healthy individuals. In addition, free fatty acids and cho­


lesterol were found to be elevated in sebum of SD patients
[16]. Triglyceride degradation by Malassezia’s lipase leads
to the inflammatory reaction [26]. A major constituent of
the resident microbial skin flora, P. acnes, has been found
to be greatly reduced in SD [27]. SD may thus be linked to
an imbalance of the microbial flora.
SD is usually associated with immune suppression,
particularly in HIV/AIDS patients. Because no clear dif­
ferences in Malassezia levels were found between indi­
viduals with and without SD, it is likely that an immune
or inflammatory reaction, not the yeast load, is the pre­
disposing cause [28]. Total serum IgA and IgG antibod­
ATOPIC DERMATITIS

ies were found to be elevated in SD patients [29].


However, no increase in the titres of antibodies against
SECTION 3:

Malassezia was detected [29,30], indicating a change in


cellular immune response instead of humoral response Fig. 17.7  Cradle cap on the scalp of an infant.
[31]. The specific role of lymphocyte activity remains
controversial [32].
The role of genetic factors in SD was underappreciated
until recently, when inherited dominant and recessive
forms of SD were identified, suggesting a role of genetic
factors in the pathogenesis [33]. Various medications have
been reported to flare or induce SD including chlorprom­
azine, cimetidine, ethionamide, fluorouracil, gold, griseof­
ulvin, interferon alfa, lithium, methoxsalen, methyldopa,
phenothiazines and psoralens [34].

Pathology. Histologically, the development of SD can be


divided into two stages. In the acute and subacute stages,
SD displays spongiosis with a superficial perivascular
and perifollicular lymphocytic infiltrate. Parakeratosis
around follicular openings (‘shoulder parakeratosis’) can
also be seen. Neutrophils can be found in the margins of
follicular ostia, but Munro microabscesses are absent. On
the other hand, marked psoriasiform hyperplasia and Fig. 17.8  Infantile seborrhoeic dermatitis affecting the groin area.
parakeratosis are the characteristics of chronic lesions.
Dilation of the venules of surface plexus, which resembles
SD may also be a cutaneous sign of the immune reconsti­
psoriasis, can also be present [24,35].
tution inflammatory syndrome in patients taking highly
active antiretroviral therapy (HAART) [38]. Therefore,
Clinical features. The infantile form usually begins about infants with generalized SD should be evaluated for more
1 week after birth and may persist for several months. serious disease.
Cradle cap is characterized by nonpruritic, thick, greasy
scales (white, off‐white or yellow) on the vertex and ante­ Differential diagnosis. The diagnosis of SD is usually
rior fontanelle regions (Fig.  17.7). Disseminated lesions, made on clinical grounds. The main differential diagnosis
usually of lesser intensity, can affect the centre of the face, is psoriasis. There is a common type of psoriasis known as
ears and forehead. seborrhoeic psoriasis. The areas affected and morphology
Lesions of the axillae and inguinal folds are acutely of lesions are almost identical to those typical of SD.
inflamed, oozing, sharply demarcated and surrounded by However, typical lesions in psoriasis are thicker and pre­
satellite lesions; occasionally they appear earlier than sent as sharply limited plaques with silvery white scales
those of the head and trunk (Fig. 17.8). Pruritus is not an [39]. Atopic dermatitis usually does not appear until after
obligatory feature, but it is often present, mainly in scalp 3 months of age, in contrast to SD which usually appears
involvement [36]. The main complication is secondary at an earlier age and rarely affects extensor areas. Tinea
bacterial infection, which increases the redness and exudate capitis typically shows scaly patches with scalp hair loss,
and local irritation [37]. Candidal overgrowth infantile ‘nap­ which is not a feature of SD [40]. Mycological examina­
kin dermatitis’ may occur. Generalized seborrhoeic eryth­ tion and fungal culture can be done to rule out tinea
roderma rarely occurs. Generalized SD (i.e. SD appearing capitis. Finally, skin lesions in systemic lupus erythema­
on the flexural folds) is uncommon in healthy children and tosus often follow a clear photodistribution, such as acute
is usually associated with immunodeficiencies (Fig. 17.9). flares of bilateral malar rash, and may be associated with
Chapter 17  Atopic Dermatitis and Related Disorders: Special Types of Presentation 239

ATOPIC DERMATITIS
SECTION 3:
(a) (b)
Fig. 17.9  Severe form of infantile seborrhoeic dermatitis affecting the face and napkin area.

extracutaneous abnormalities such as arthritis, mouth antifungal properties against Malassezia and can be used
ulcers, glomerulonephritis or cardiomyopathy [41]; SD as a shampoo [47].
does not have a photodistribution pattern, and does not Topical corticosteroids may hasten recurrences, may
affect organ systems other than the skin. foster dependence because of a rebound effect, and are
Other less common conditions that may resemble SD discouraged except for short‐term use. Short courses of
and should be considered in differential diagnosis are low‐potency topical corticosteroids can aid in resolution
pityriasis rosea, napkin dermatitis and cutaneous of erythema and itching. Topical calcineurin inhibitors
Langerhans cell histiocytosis [42]. The majority of these such as tacrolimus and pimecrolimus have been proposed
conditions can be differentiated by clinical presentation, as a steroid‐sparing therapy for SD. Antibiotics should be
although laboratory confirmation may at times be given in secondarily infected lesions.
required. Additionally, some drugs (griseofulvin, ethion­
amide, buspirone, haloperidol, chlorpromazine, IL‐2,
interferon alfa, methyldopa, psoralens) and nutritional ­References
1 Gupta AK, Bluhm R, Cooper EA et  al. Seborrheic dermatitis.
deficiencies (of pyridoxine, zinc, niacin and riboflavin) Dermatol Clin 2003;21:401–12.
may result in SD‐like dermatitis [43,44]. 2 Gee BC. Seborrhoeic dermatitis. Clin Evid 2004;2344–52.
3 Elish D, Silverberg NB. Infantile seborrheic dermatitis. Cutis 2006;
77:297–300.
Prognosis. For the majority of patients, treatment leads to 4 Singleton JK. Pediatric dermatoses: three common skin disruptions in
clearing of lesions. Infantile SD usually improves by the infancy. Nurse Pract 1997;22:32–33.
end of the first year of life. In adolescence, intermittent, 5 Foley P, Zuo Y, Plunkett A et al. The frequency of common skin condi­
active phases of SD can manifest with burning, scaling tions in preschool‐aged children in Australia: seborrheic dermatitis
and pityriasis capitis (cradle cap). Arch Dermatol 2003;139:318–22.
and itching, alternating with inactive periods. Activity is 6 Siegfried E, Hebert A. Diagnosis of atopic dermatitis: mimics, over-
increased in winter and early spring, with remissions laps and complications. J Clin Med 2015;4:884–917.
commonly occurring in summer. Active phases of SD 7 Palamaras I, Kyriakis KP, Stavrianeas NG. Seborrheic dermatitis: lifetime
detection rates. J Eur Acad Dermatol Venereol 2012;26:524–6.
may be complicated by secondary infection [24].
8 Gupta AK, Madzia SE, Batra R. Etiology and management of sebor-
rheic dermatitis. Dermatology 2004;208:89–93.
Treatment. The treatment of cradle cap is usually simple. 9 Ljubojevic S, Skerlev M, Lipozencic J, Basta‐Juzbasic A. The role of
Daily shampooing with a nonmedicated shampoo for 1 Malassezia furfur in dermatology. Clin Dermatol 2002;20:179–82.
10 Wananukul S, Chindamporn A, Yumyourn P et al. Malassezia furfur
week may be the only treatment needed in mild cases to in infantile seborrheic dermatitis. Asian Pac J Allergy Immunol
induce loosening and removal of scales. Emollients can be 2005;23:101–5.
applied to loosen scales in more resistant cases. Emollients 11 Gupta AK, Kohli Y, Summerbell C, Faergemann J. Quantitative cul-
ture of Malassezia species from different body sites of individuals
can be left on the scalp overnight and scales can be gently
with or without dermatoses. Med Mycology 2001;39:243–51.
removed with an infant hair brush following washing 12 Skinner RB Jr, Noah PW, Taylor RM, et al. Double‐blind treatment of
with a nonmedicated shampoo [45]. seborrheic dermatitis with 2% ketoconazole cream. J Am Acad
Antifungal creams, such as ketoconazole, and antifun­ Dermatol 1985;12:852–6.
13 Tatlican S, Eren C, Eskioglu F. Insight into pimecrolimus experience in
gal shampoos are part of first‐line pharmacological ther­ seborrheic dermatitis: close follow‐up with exact mean cure and
apy [46] and are used to decrease colonization by yeasts remission times and side‐effect profile. J Dermatolog Treat 2009;
and improve erythema. They are often effectively used 20:198–202.
for long‐term control. Other effective topical antifungals 14 Gaitanis G, Magiatis P, Hantschke M et al. The Malassezia genus in
skin and systemic diseases. Clin Microbiol Rev 2012;25:106–41.
include the azole group (miconazole, clotrimazole), 15 Hay RJ. Malassezia dandruff and seborrhoeic dermatitis: an over-
selenium sulfide and ciclopirox olamine. Tea tree oil has view. Br J Dermatol 2011;165 (Suppl 2):2–8.
240 Section 3  Atopic Dermatitis and Related Disorders

16 Ro BI, Dawson TL. The role of sebaceous gland activity and scalp
microfloral metabolism in the etiology of seborrheic dermatitis and
­Lichen simplex chronicus
dandruff. J Investig Dermatol Symp Proc 2005;10:194–7.
17 Berk T, Scheinfeld N. Seborrheic dermatitis. P T 2010;35:348–52. Definition. Lichen simplex chronicus (LSC), known also
18 DeAngelis YM, Gemmer CM, Kaczvinsky JR et al. Three etiologic fac­ as circumscribed neurodermatitis or neurodermatitis cir­
ets of dandruff and seborrheic dermatitis: Malassezia fungi, seba­
ceous lipids, and individual sensitivity. J Investig Dermatol Symp
cumscripta, is a skin disorder characterized by thickened,
Proc 2005;10:295–7. lichenified and often hyperpigmented plaque of the skin
19 Plotkin LI, Squiquera L, Mathov I et al. Characterization of the lipase as a result of excessive scratching [1]. It may appear de
activity of Malassezia furfur. J Med Vet Mycol 1996;34:43–8. novo or it may complicate other skin problems such as
20 DeAngelis YM, Saunders CW, Johnstone KR et  al. Isolation and

expression of a Malassezia globosa lipase gene, LIP1. J Invest allergic contact dermatitis [2].
Dermatol 2007;127:2138–46.
21 Faergemann J, Bergbrant IM, Dohse M et  al. Seborrhoeic dermatitis Epidemiology. The exact frequency in the general popula­
and Pityrosporum (Malassezia) folliculitis: characterization of inflam­
matory cells and mediators in the skin by immunohistochemistry. Br
tion is unknown. In different studies, it has been shown to
affect between 0.5% and 12% of the population [3]. It
ATOPIC DERMATITIS

J Dermatol 2001;144:549–56.
22 Schwartz JR, Messenger AG, Tosti A et  al. A comprehensive patho­ develops predominantly in mid to late adult life, with its
physiology of dandruff and seborrheic dermatitis – towards a more highest prevalence in persons aged 30–50 years, but it cer­
SECTION 3:

precise definition of scalp health. Acta Derm Venereol 2013;93:131–7.


23 Pechere M, Kirscher J, Remondat C et al. Malassezia spp carriage in tainly also occurs in children. When LSC develops in chil­
patients with seborrheic dermatitis. J Dermatol 1999;26:558–61. dren, it is slightly more likely to occur in boys, whereas in
24 Schwartz RA, Janusz CA, Janniger CK. Seborrheic dermatitis: an adults it is slightly more common in women. No differ­
overview. Am Fam Physician 2006;74:125–30.
25 Burton JL, Pye RJ. Seborrhoea is not a feature of seborrhoeic dermati­
ences are reported in frequency among races although
tis. Br Med J (Clin Res Ed) 1983;286:1169–70. secondary pigmentary alterations are more severe in indi­
26 Ostlere LS, Taylor CR, Harris DW et  al. Skin surface lipids in HIV‐ viduals with darker skin [1,4].
positive patients with and without seborrheic dermatitis. Int J
Dermatol 1996;35:276–9.
27 McGinley KJ, Leyden JJ, Marples RR, Kligman AM. Quantitative
Pathogenesis. Very little is known about the cause of
microbiology of the scalp in non‐dandruff, dandruff, and seborrheic LSC. It usually results from long‐term scratching and pru­
dermatitis. J Invest Dermatol 1975;64:401–5. ritus of skin. Excessive itching may be due to environ­
28 Dessinioti C, Katsambas A. Seborrheic dermatitis: etiology, risk
factors, and treatments: facts and controversies. Clin Dermatol 2013;
mental factors that are recognized to stimulate type C,
31:343–51. nonmyelinated sensory nerve endings, such as heat and
29 Bergbrant IM, Johansson S, Robbins D et al. An immunological study in sweat retention, rubbing of clothing, or excessive use of
patients with seborrhoeic dermatitis. Clin Exp Dermatol 1991;16:331–8. cleansers. Itching may be due to dermatological diseases,
30 Parry ME, Sharpe GR. Seborrhoeic dermatitis is not caused by an altered
immune response to Malassezia yeast. Br J Dermatol 1998;139:254–63. which presumably cause dysfunction of the barrier layer
31 Ashbee HR, Ingham E, Holland KT, Cunliffe WJ. Cell‐mediated
and allow peripheral nerve endings to be stimulated,
immune responses to Malassezia furfur serovars A, B and C in such as atopic dermatitis, allergic contact dermatitis, sta­
patients with pityriasis versicolor, seborrheic dermatitis and controls.
sis dermatitis and insect bites [4–6]. Psychogenic factors
Exp Dermatol 1994;3:106–12.
32 Bergbrant IM, Andersson B, Faergemann J. Cell‐mediated immunity may play a relevant role in LSC; psychological distress
to Malassezia furfur in patients with seborrhoeic dermatitis and pity­ might influence both the intensity and duration of the
riasis versicolor. Clin Exp Dermatol 1999;24:402–6. perception of itching [7].
33 Hoger H, Gialamas J, Adamiker D. Inherited seborrheic dermatitis—a
new mutant in mice. Lab Anim 1987;21:299–305.
Atopic dermatitis results in a higher probability of
34 Brodell EE, Smith E, Brodell RT. Exacerbation of seborrheic dermatitis developing LSC. The identification of LSC as a localized
by topical fluorouracil. Arch Dermatol 2011;147:245–6. form of atopic dermatitis is still debated [8,9]. Allergic
35 Sampaio AL, Mameri AC, Vargas TJ et al. Seborrheic dermatitis. An contact dermatitis to para‐phenylenediamine containing
Bras Dermatol 2011;86:1061–71.
36 Del Rosso JQ. Adult seborrheic dermatitis: a status report on practical hair dyes was shown to be linked to LSC, and improve­
topical management. J Clin Aesthet Dermatol 2011;4:32–8. ment in symptoms occurred after discontinuation of these
37 Ramos‐E‐Silva M, Sampaio AL, Carneiro S. Red face revisited:
dyes [2]. Similarly, lithium has been linked to LSC in one
Endogenous dermatitis in the form of atopic dermatitis and sebor­
rheic dermatitis. Clin Dermatol 2014;32:109–15.
reported case [10]. Scars (e.g. traumatic, postherpetic/
38 Osei‐Sekyere B, Karstaedt AS. Immune reconstitution inflammatory zoster [11]), acne keloidalis nuchae, xerosis [12], venous
syndrome involving the skin. Clin Exp Dermatol 2010;35:477–81. insufficiency and asteatotic eczema are common factors
39 Boehncke WH. Etiology and pathogenesis of psoriasis. Rheum Dis for development of LSC.
Clin North Am 2015;41:665–75.
40 Meadows‐Oliver M. Tinea capitis: diagnostic criteria and treatment
options. Pediatr Nurs 2009;35:53–7. Pathology. The histopathological features in LSC are
41 Naldi L, Rebora A. Clinical practice. Seborrheic dermatitis. N Engl hyperkeratosis, acanthosis, spongiosis and patches of
J Med 2009;360:387–96.
42 Clark GW, Pope SM, Jaboori KA. Diagnosis and treatment of sebor-
parakeratosis and hypergranulosis. Epidermal thickening
rheic dermatitis. Am Fam Physician 2015;91:185–90. of all layers is noted, with elongation of rete ridges and
43 Valia RG. Etiopathogenesis of seborrheic dermatitis. Indian J Dermatol with pseudoepitheliomatous hyperplasia. There is a
Venereol Leprol 2006;72:253–5. perivascular infiltrate of lymphocytes and occasionally of
44 Yamamoto T, Tsuboi R. Interleukin‐2‐induced seborrhoeic dermatitis‐
like eruption. J Eur Acad Dermatol Venereol 2008;22:244–5. macrophages [5]. Papillary dermal fibrosis with vertical
45 Johnson BA, Nunley JR. Treatment of seborrheic dermatitis. Am streaking of collagen bundles is characteristic. Silver
Fam Physician 2000;61:2703–10. impregnation techniques show proliferation of Schwann
46 Gupta AK, Nicol K, Batra R. Role of antifungal agents in the treatment
cells in cellular infiltrate [13]. By electron microscopy
of seborrheic dermatitis. Am J Clin Dermatol 2004;5:417–22.
47 Satchell AC, Saurajen A, Bell C. Treatment of dandruff with 5% tea frequent collagen fibres attached to and just above the
tree oil shampoo. J Am Acad Dermatol 2002;47:852–5. lamina basalis are usually seen.
Chapter 17  Atopic Dermatitis and Related Disorders: Special Types of Presentation 241

have proved to be effective in LSC for patients


Clinical features. LSC is characterized by the presence
unresponsive to topical corticosteroid or those with
of one or more erythematous, scaly, lichenified plaques
lesions on thin skin [18,19].
with varying degrees of overlying excoriation from
Other topical medications reported to decrease pruritus
habitual scratching and rubbing of the skin. In cases of
include doxepin cream [20] and capsaicin cream [21]. Oral
longstanding duration, areas of hyperpigmentation
antianxiety medications and sedation may be consid­
may also be present. Each plaque may have three
ered in certain patients according to individual need. For
zones: a wide peripheral thickened zone, a middle
infected lesions, a topical or oral antibiotic can be
zone with prurigo papules that may be excoriated, and
considered.
a central zone that has the greatest thickening and pig­
mentary alteration. There is a predilection for the
occipital, vulval and nuchal areas in women and the
perineum and scrotum in men. It is uncommon in chil­ ­References

ATOPIC DERMATITIS
dren. Other favoured areas include the wrists and 1 Rajalakshmi R, Thappa DM, Jaisankar TJ, Nath AK. Lichen simplex
chronicus of anogenital region: A clinico‐etiological study. Indian J
extensor surfaces of the forearms and lower legs. Dermatol Venereol Leprol 2011;77:28–36.

SECTION 3:
Pruritus is usually intermittent; the resultant scratch­ 2 Chey WY, Kim KL, Yoo TY, Lee AY. Allergic contact dermatitis from
ing provides temporary relief. Interestingly, patients hair dye and development of lichen simplex chronicus. Contact
Dermatitis 2004;51:5–8.
may start scratching while discussing the itch or 3 Ball SB, Wojnarowska F. Vulvar dermatoses: lichen sclerosus, lichen
describing the lesions [5,14]. planus, and vulval dermatitis/lichen simplex chronicus. Semin Cutan
Med Surg 1998:17:182–8.
4 Lynch PJ. Lichen simplex chronicus (atopic/neurodermatitis) of the
Prognosis. LSC is associated with a prolonged course. anogenital region. Dermatol Ther 2004;17:8–19.
If left untreated, the condition goes on indefinitely, 5 Lotti T, Buggiani G, Prignano F. Prurigo nodularis and lichen sim-
plex chronicus. Dermatol Ther 2008;21:42–6.
although it may show remissions and relapses. 6 Fischer G, Spurrett B, Fischer A. The chronically symptomatic vulva:
Unfortunately, even with highly effective therapy, the Aetiology and management. Br J Obstet Gynaecol 1995;102:773–9.
likelihood of recurrence is great, especially when 7 Tsintsadze N, Beridze L, Tsintsadze N et al. Psychosomatic aspects in
patients with dermatologic diseases. Georgian Med News 2015;243 :
the triggering factors are still present. LSC can be
70–5.
extremely uncomfortable, leading to vigorous scratch­ 8 Williams HC. Diagnostic criteria for atopic dermatitis. Where do we
ing. Superimposed infection, scarring and fibrosis can go from here? Arch Dermatol 1999;135:583–6.
occur [4]. 9 Williams HC. On the definition and epidemiology of atopic dermati­
tis. Dermatol Clin 1995;13:649–57.
10 Shukla S, Mukherjee S. Lichen simplex chronicus during lithium

treatment. Am J Psychiatry 1984;141:909–10.
Differential diagnosis. Clinical features are usually 11 Gerritsen MJ, Gruintjes FW, Andreissen MA et  al. Lichen simplex
sufficient for diagnosing this disorder. It is first impor­ chronicus as a complication of herpes zoster. Br J Dermatol
tant to determine whether the LSC is primary or sec­ 1998;138:921–2.
ondary to other underlying disease. Lichenified 12 Burkhart CG, Burkhart CN. Acne keloidalis is lichen simplex

chronicus with fibrotic keloidal scarring. J Am Acad Dermatol
plaques of atopic dermatitis may be difficult to distin­ 1998;39:661.
guish from LSC [4]. However, a history of atopic der­ 13 Thorstensen K, Birenbaum D. Recognition and management of vul­
matitis and the typical distribution can differentiate var dermatologic conditions: lichen sclerosus, lichen planus, and
lichen simplex chronicus. J Midwifery Womens Health
the two conditions. Lichenified psoriasis is another 2012;57:260–75.
condition that may be similar to LSC. The characteris­ 14 Gunasti S, Marakli SS, Tuncer I et  al. Clinical and histopatho-
tic psoriatic scales and histopathological changes logical findings of ‘psoriatic neurodermatitis’ and of typical
(hypogranulosis, neutrophilic microabscesses in the lichen simplex chronicus. J Eur Acad Dermatol Venereol
2007;21:811–17.
horny layer and thinning of the suprapapillary plates) 15 Brunner N, Yawalkar S. A double‐blind, multicenter, parallel‐group
are not usually seen in LSC [14]. trial with 0.05% halobetasol propionate ointment versus 0.1% diflu­
cortolone valerate ointment in patients with severe, chronic atopic
dermatitis or lichen simplex chronicus. J Am Acad Dermatol
Treatment. Treatment is aimed at reducing pruritus 1991;25:1160–3.
16 Datz B, Yawalkar S. A double‐blind, multicenter trial of 0.05% halobet­
and minimizing existing lesions because rubbing and
asol propionate ointment and 0.05% clobetasol 17‐propionate ointment
scratching cause LSC. The first step necessary in deal­ in the treatment of patients with chronic, localized atopic dermatitis or
ing with disturbed barrier layer function is the use of lichen simplex chronicus. J Am Acad Dermatol 1991;25:1157–60.
emollients. 17
Geraldez MC, Carreon‐Gavino M, Hoppe G, Costales A.
Diflucortolone valerate ointment with and without occlusion in
Topical steroids are the current treatment of choice lichen simplex chronicus. Int J Dermatol 1989;28:603–4.
because they decrease inflammation and itch while 18 Kelekci HK, Uncu HG, Yilmaz B et  al. Pimecrolimus 1% cream for
concurrently softening the hyperkeratosis [15–17]. pruritus in postmenopausal diabetic women with vulvar lichen
Potent topical glucocorticoid creams or ointments are simplex chronicus: A prospective non‐controlled case series.
J Dermatolog Treat 2008;11:1–5.
often successfully employed. Occlusion, with or with­ 19 Ashoff R, Wozel G. Topical tacrolimus for the treatment of lichen
out topical steroids, also provides a physical barrier to simplex chronicus. J Dermatolog Treat 2007;18:115–17.
the scratching. Addition of menthol or camphor to topi­ 20 Drake LA, Millikan LE. The antipruritic effect of 5% doxepin cream in
patients with eczematous dermatitis. Doxepin Study Group. Arch
cal steroid was used previously to alleviate itch [1]. Dermatol 1995;131:1403–8.
Intralesional corticosteroids may be used in resistant 21 Kantor GR, Resnik KS. Treatment of lichen simplex chronicus with
lesions in adults. Topical tacrolimus and pimecrolimus topical capsaicin cream. Acta Derm Venereol 1996;76:161.
242 Section 3  Atopic Dermatitis and Related Disorders

­Prurigo nodularis Psychosocial disorders are also significantly associated


with PN as a primary association or sometimes second­
Definition. Prurigo nodularis (PN; chronic circumscribed ary to the severe itch in PN [11]. Change in the density of
nodular lichenification, Picker’s nodules), is a chronic dermal nerve fibres and the interaction between nerve
inflammatory dermatosis characterized by the presence of fibres and neuropeptides/neurotrophins [12] may also
symmetrically distributed multiple, highly pruritic, hyper­ have a role in the pathogenesis of PN. Inflammation
keratotic, erosive or crusted nodules and papules usually caused by T lymphocytes, eosinophilic granulocytes
surrounded by an irregular hyperpigmented ring. PN is a and mast cells is involved in the development and
long‐term reaction to the chronic scratching of patients with chronification of PN. Pruritus can be caused by the
chronic pruritus. The extensor area of the limbs, face and release of mast cell products such as histamine, tryptase
trunk are usually affected by the lesions. It can occur at all and prostaglandins [13].
ages [1,2]. Hardaway was the first to describe PN as ‘multi­
ple tumors of the skin accompanied by intensive itching’ in Pathology. The histological characteristics of PN include
ATOPIC DERMATITIS

1880, and later it was called lichen obtusus corneus by Brocq thick, compact epidermal orthohyperkeratosis with acan­
[3]. Hyde, in 1909, defined the term PN clinically and distin­ thosis and parakeratosis or superficial epithelial necrosis.
SECTION 3:

guished it from the general term prurigo [4]. Rete ridges are elongated and irregular; there is fibrosis of
the papillary dermis with vertically arranged collagen
Epidemiology. PN can occur at any age, even in children, but fibres, and there are an increased number of fibroblasts
it most often occurs in middle‐aged and elderly people [5,6]. and capillaries with a dense dermal infiltrate consisting of
It can affect both sexes, with special predilection for women neutrophils, eosinophils, histiocytes and monocytes [14].
[7]. It may occur in conjunction with atopy; in that case, it has Also notable in the dermis are thickened nerve fibres and
an earlier age of onset and may be accompanied by cutane­ fibrosis with thickened collagen bundles [15,16]. Schwann
ous hypersensitivity to various environmental allergens [8]. cells show vacuolization and degeneration with no detect­
There is no known racial predilection for PN [9]. able mitochondria. Axons and Schwann cells both show
hyperproliferation [17]. There are increased numbers of
Pathogenesis. The cause of PN is still unknown. Repeated Merkel cells in the epidermis of PN nodules [18]. In addi­
mechanical trauma is the main factor. The vicious cycle of tion, mast cells are increased in number and show charac­
repeated itching and scratching leads to thickening with teristic morphological changes such as an enlarged cell
plaques or nodular lichenification of the skin. An under­ body size and a dendritic shape compared with the round
lying disease can be associated with PN in most of the or elongated shape seen in normal skin [19].
patients; atopic diathesis is the most common association.
An associated atopic diathesis may be responsible for ear­ Clinical features. Clinically, PN is characterized by firm,
lier age of onset (median age 19 years) compared to non­ pruritic, 0.5–3‐cm diameter, hyperpigmented or purpuric
atopic patients (median age 48 years) [9]. Other diseases nodules with a hyperkeratotic or excoriated surface,
that may be associated with PN include obstructive bil­ numbering from a few to hundreds (Fig.  17.10). The
iary diseases, mastocytosis, diabetes mellitus, drug reac­ lesions tend to be symmetrically distributed, with a predilection
tion, chronic renal failure, hepatitis B and C infection, and for the extensor area of the limbs, face and trunk. Palms are
some neoplasms such as Hodgkin lymphoma [10]. seldom affected. Postinflammatory hyper/hypopigmentation

Fig. 17.10  Prurigo nodularis in an adolescent


boy.
Chapter 17  Atopic Dermatitis and Related Disorders: Special Types of Presentation 243

with or without scarring may occur [19]. The skin between ­References
1 Spring P, Gschwind I, Gilliet M. Prurigo nodularis: retrospective study
the lesions is usually normal but can be xerotic, especially
of 13 cases managed with methotrexate. Clin Exp Dermatol 2014;
in cases with associated atopy [9]. There may be a butter­ 39:468–73.
fly‐shaped area of normal skin on the back where the 2 Errichetti E, Piccirillo A, Stinco G. Dermoscopy of prurigo nodularis.
patient is unable to reach (butterfly sign). J Dermatol 2015;42:632–4.
3 Brocq LAJ. Lichen obtusus corneus. Prat Dermatol 1900;3:201.
4 Hyde JN: Prurigo nodularis. In: Hyde JN, Montgomery FH (eds) A
Prognosis. PN is a skin disorder that has a chronic course. Practical Treatise on Diseases of the Skin for the Use of Students and
Severe pruritus can be frustrating for the patient and may Practitioners, 8th edn. Philadelphia: Lea and Febiger, 1909:174–5.
5 Iking A, Grundmann S, Chatzigeorgakidis E et  al. Prurigo as a
result in psychological problems [20]. symptom of atopic and non‐atopic diseases: aetiological survey in
a consecutive cohort of 108 patients. J Eur Acad Dermatol Venereol
2013; 27:550–7.
Differential diagnosis. PN mainly has to be differenti­
6 Schelnitz LS, Paller AS. Hodgkin’s disease manifesting as prurigo
ated from nodular dermatoses, particularly hypertrophic nodularis. Pediatr Dermatol 1990;7:136–9.

ATOPIC DERMATITIS
lichen planus, nodular scabies, lichen simplex 7 Amer A, Fischer H. Prurigo nodularis in a 9‐year‐old girl. Clin Pediatr
chronicus, multiple keratoacanthomas, atopic dermati­ 2009;48:93–5.
8 Tan W, Tey H. Extensive prurigo nodularis: characterization and etiol-

SECTION 3:
tis, allergic contact dermatitis, neurotic excoriations, ogy. Dermatology 2014;228:276–80.
reactive perforating collagenosis and pemphigoid 9 Tanaka M, Aiba S, Matsumura N et al. Prurigo nodularis consists of
­nodularis. Clinical features are usually sufficient for two distinct forms: early‐onset atopic and late‐onset non‐atopic.
Dermatology 1995;190:269–76.
diagnosis, however histopathological examination is
10 Jorizzo JL, Gatti S, Smith EB. Prurigo: a clinical review. Am Acad
sometimes needed to reach a definitive diagnosis. Dermatol 1981;4:723–8.
Dermoscopy has been shown to facilitate the clinical 11 Rowland Payne CM, Wilkinson JD, McKee PH et al. Nodular pru-
differentiation [21]. rigo  –  a clinicopathological study of 46 patients. Br J Dermatol
1985;113:431–9.
12 Stander S, Siepmann D, Herrgott I. Targeting the neurokinin receptor
Treatment. The goal of treatment for PN is to break the 1 with aprepitant: A novel antipruritic strategy. PLoS ONE 2010;
itch–scratch–itch cycle by reducing pruritus, rubbing, 5:e10968.
13 Zeidler C, Stander S. The pathogenesis of prurigo nodularis – ‘super‐itch’
picking and scratching. Underlying causes, if present, in exploration. Eur J Pain 2016;20:37–40.
should be controlled first. Simple general measures such 14 Feuerman EJ, Sandbank M. Prurigo nodularis. Histological and electron
as clipping the fingernails may be helpful. Habit reversal microscopical study. Arch Dermatol 1975;111:1472–7.
15 Weigelt N, Metze D, Steander S. Prurigo nodularis: Systematic analysis of
therapy for the itch–scratch cycle associated with PN may 58 histological criteria in 136 patients. J Cutan Pathol 2010; 37:578–86.
be helpful [22]. Applying emollients is also important as 16 Schuhknecht B, Marziniak M, Wissel A et al. Reduced intraepidermal
xerosis usually worsens the pruritus. Topical, oral and nerve fibre density in lesional and nonlesional prurigo nodularis skin
intralesional corticosteroids have all been used in PN in as a potential sign of subclinical cutaneous neuropathy. Br J Dermatol
2011;165:85–91.
an attempt to decrease the inflammation and sense of 17 Sandbank M. Cutaneous nerve lesions in prurigo nodularis. Electron
itching, and to soften and smoothen out firm nodules microscopic study of two patients. J Cutan Pathol 1976;3:125–32.
[23]. Occlusive bandages can also be used with or without 18 Nahass G, Penneys NS. Merkel cells and prurigo nodularis. J Am
Acad Dermatol 1994;31:86–8.
topical steroids. Treatment with DuoDerm or other
19 Lee M, Shumack S. Continuing Professional Development Program
occlusive therapies has been suggested to flatten lesions Prurigo nodularis: A review. Aust J Dermatol 2005;46:211–20.
while at the same time preventing patients from directly 20 Dazzi C, Erma D, Piccinno R et al. Psychological factors involved in
scratching nodules [24]. Topical antipruritics such as 1% prurigo nodularis: a pilot study. J Dermatolog Treat 2011;22:211–14.
21 Lallas A, Giacomel J, Argenziano G et al. Dermoscopy in general derma­
menthol or phenol in a creamy base may be used to reduce tology: practical tips for the clinician. Br J Dermatol 2014; 170:514–26.
the itch. Topical capsaicin may also be an effective 22 Grillo M, Long R, Long D. Habit reversal training for the itch‐scratch
treatment in some cases. Topical tacrolimus, pimecroli­ cycle associated with pruritic skin conditions. Dermatol Nurs
mus and vitamin D3 have also been reported to be 2007;19:243–8.
23 Richards RN. Update on intralesional steroid: focus on derma­
effective in the treatment of PN [25]. Antihistamines, toses. J Cutan Med Surg 2010;14:19–23.
anxiolytics, opiate receptor antagonists (nalfurafine) and 24 Meyers LN. Use of occlusive membrane in the treatment of prurigo
gabapentin have been reported to benefit PN in some nodularis. Int J Dermatol 1989;28:275–6.
25 Wong SS, Goh CL. Double‐blind, right/left comparison of calcipotriol
cases [26,27]. ointment and betamethasone ointment in the treatment of prurigo
Cryotherapy is a useful therapeutic modality for nodularis. Arch Dermatol 2000;136:807–8.
the treatment of PN [28]. It helps reduce pruritus and 26 Gencoglan G, Inanir I, Gunduz K. Therapeutic hotline: Treatment of
flattens lesions through destruction of sensory nerves prurigo nodularis and lichen simplex chronicus with gabapentin.
Dermatol Ther 2010;23:194–8.
and impairment of nerve regeneration [29]. UV light 27 Dereli T, Karaca N, Inanir I, Ozturk G. Gabapentin for the treatment of
exposure has been shown to lessen the pruritus, and recalcitrant chronic prurigo nodularis. Eur J Dermatol 2008;18:85–6.
can be beneficial in the treatment of PN; UVB or UVA 28 Waldinger TP, Wong RC, Taylor WB, Voorhees JJ. Cryotherapy

improves prurigo nodularis. Arch Dermatol 1984;120:1598–600.
plus psoralen may be used. The combination of UVB
29 Thai KE, Sinclair RD. Cryosurgery of benign skin lesions. Australas J
308‐nm excimer light and bath PUVA therapy may be Dermatol 1999;40:175–86.
effective in the treatment of PN [30]. UVA‐1 has also 30 Hammes S, Hermann J, Roos S, Ockenfels HM. UVB 308‐nm excimer light
been reported to benefit lichen simplex chronicus and and bath PUVA: combination therapy is very effective in the treatment of
prurigo nodularis. J Eur Acad Dermatol Venereol 2011;25:799–803.
PN. Pulsed dye laser therapy may also be of help 31 Rombold S, Lobisch K, Katzer K et al. Efficacy of UVA1 phototherapy
through reduction of the vascularity of individual in 230 patients with various skin diseases. Photodermatol Photoi­
lesions [31]. mmunol Photomed 2008;24:19–23.
244 Section 3  Atopic Dermatitis and Related Disorders

­Prurigo pigmentosa Although the individual lesions resolve quickly over


1 week, residual netlike hyperpigmentation remains. The
Prurigo pigmentosa (PP) is a rare cutaneous disorder of disease course is punctuated by frequent recurrences
unknown aetiology, first described by Nagashima et al. in followed by remissions lasting weeks to years [3,10,11].
1971. It is diagnosed most commonly in the Japanese pop­
ulation [1,2]. It presents with recurrent, pruritic erythe­ Differential diagnosis. Diseases to be considered in the
matous macules, papules and papulovesicles on the differential diagnosis clinically at an early stage of the
trunk, back, neck and chest that resolve, leaving behind a process are early manifestations of dermatitis herpetiformis,
netlike pigmentation. The disease is seen most commonly linear IgA dermatosis, pigmented contact dermatitis and
in young women, although it has been reported in adoles­ acute lupus erythematosus. At a resolving stage of the
cence [3,4]. process, PP may be confused with reticular pigmented
papules of confluent and reticulated papillomatosis of
Pathogenesis. The exact aetiology and pathogenesis of Gougerot and Carteaud but, in contrast to those lesions,
ATOPIC DERMATITIS

PP are still unknown. Due to its occurrence mainly in the pigmented macules of PP are not keratotic.
Japan, PP was thought to have a genetic background.
SECTION 3:

However, there are no reports of familial cases, and the Treatment. Several medications are available. Antihis­
number of PP cases discovered all over the world is tamines and topical or systemic corticosteroids are usu­
increasing. Various mechanisms have been suggested to ally ineffective in the treatment of PP. Oral minocycline
cause PP, such as friction, contact allergy, sensitivity to (200 mg) taken daily is usually considered as the first‐line
sunlight, endocrinological alterations such as diabetes therapy for PP [12,13]. Doxycycline, dapsone, potassium
mellitus and metabolic disorders such as ketosis [5,6], but iodide and macrolide antibiotics have also been reported
none of them has proved to be consistently identifiable. to be effective [14–16]. A favourable response of PP to
As the lesions are mainly restricted to covered areas of the isotretinoin has been reported [16]. Although the medica­
body, the occurrence of PP has been considered to be trig­ tions are quite effective in stopping the inflammatory
gered by rubbing. Clothing friction acting as a mechanical component, treatment of the postinflammatory hyperpig­
stimulus has also been suspected as a possible triggering mentation remains problematic.
factor [7].
­References
Pathology. Early reports of histological findings in PP 1 Nagashima M, Ohshiro A, Shimizu N. A peculiar pruriginous der-
were nonspecific and not diagnostic. However, Böer et al. matosis with gross reticular pigmentation. Jpn J Dermatol 1971;
81:38–9.
[8] proposed that the histopathological changes of PP 2 Nagashima M. Prurigo pigmentosa–clinical observations of our

may be specific. Early lesions of PP show spongiosis, bal­ 14 cases. J Dermatol 1978;5:61–7.
looning and necrotic keratocytes in the epidermis, and a 3 Böer A, Asgari M. Prurigo pigmentosa: an underdiagnosed disease?
perivascular neutrophilic infiltrate. Lymphocytes are Indian J Dermatol Venereol Leprol 2006;72:405–9.
4 Corley S, Mauro P. Erythematous papules evolving into reticulated
sprinkled along the dermoepidermal junction. In a fully hyperpigmentation on the trunk: A case of prurigo pigmentosa. JAAD
developed lesion, the infiltrate assumes a patchy lichenoid Case Rep 2015;1:60–2.
pattern, and there are more numerous lymphocytes in the 5 Kwon HJ, Kim MY, Kim HO, Park YM. Two cases of prurigo

­pigmentosa in atopic patients. J Dermatol 2006;33:579–82.
dermis. In the epidermis ballooning is more prominent
6 Kubota Y, Koga T, Nakayama J. Bullous prurigo pigmentosa and
than spongiosis, and lymphocytes are increased in num­ ­diabetes. Eur J Dermatol 1998;8:439–41.
ber in the lower part of the epidermis. In a resolving 7 Kim MH, Choi YW, Choi HY, Myung KB. Prurigo pigmentosa from
lesion, a sparse infiltrate of lymphocytes is present in the contact allergy to chrome in detergent. Contact Dermatitis 2001;44:
289–92.
upper part of the reticular dermis and the papillary der­ 8 Böer A, Ackerman AB. Prurigo pigmentosa is distinctive histo-
mis. Necrotic keratinocytes may be found in the basal pathologically. Int J Dermatol 2003;42:417–18.
layer. Immunofluorescence is negative [9]. 9 Kokol R, Miljković J, El Shabrawi‐Caelen L, Prurigo pigmentosa.
J Dtsch Dermatol Ges 2011;9:867–8.
10 Asgari M, Daneshpazhooh M, Chams Davatchi C, Boer A. Prurigo
Clinical features. The eruption of PP is distributed sym­ pigmentosa: An underdiagnosed disease in patients of Iranian
metrically on the trunk, with predilection for the upper descent? J Am Acad Dermatol 2006;55:131–6.
part of the back, the sacrum, abdomen and chest. 11 Oh YJ, Lee MH. Prurigo pigmentosa: A clinicopathologic study of
16 cases. J Eur Acad Dermatol Venereol 2012;26:1149–53.
Intermammary and submammary zones are involved 12 Matsumoto C, Kinoshita M, Baba S et al. Vesicular prurigo pigmentosa
more commonly. Rarely, the forehead, the arms or the cured by minocycline. J Eur Acad Dermatol Venereol 2001;15:354–6.
abdomen are affected. Individual lesions are symmetri­ 13 Aso M, Miyamoto T, Morimura T, Shimao S. Prurigo pigmentosa
successfully treated with minocycline. Br J Dermatol 1989;120:705–8.
cally distributed pruritic urticarial papules at the begin­ 14 Degavre B, Guilhou JJ, Guillot B. [Prurigo pigmentosa]. Ann Dermatol
ning of an eruption, evolving to short‐lived red papules Venereol 1994;121:46–9.
and papulovesicles, and coalescing into plaques. 15 Yazawa N, Ihn H, Yamane K et al. The successful treatment of prurigo
Resolving lesions are crusted and scaly red papules and pigmentosa with macrolide antibiotics. Dermatology 2001;202:67–9.
16 Requena Caballero C, Nagore E, Sanmartin O et al. Vesiculous pru­
smooth‐surfaced pigmented macules. Often, secondary rigo pigmentosa in a 13‐year‐old girl: Good response to isotretinoin.
changes of excoriation, scaling or crusting are observed. J Eur Acad Dermatol Venereol 2005;19:474–6.
245

C HA PTER 18

Atopic Dermatitis: Complications


Kevin B. Yarbrough1 & Eric L. Simpson2
 Department of Pediatric Dermatology, Phoenix Children’s Hospital, Phoenix, AZ, USA
1

 Department of Dermatology, Oregon Health and Science University, Portland, OR, USA
2

Bacterial infections, 245 Sleep disturbance, 248 Mental health disorders, 250
Viral infections, 247 Psychosocial complications, 249 Ocular complications, 251

ATOPIC DERMATITIS
Abstract

SECTION 3:
in children with AD. Extracutaneous complications of AD include
not only the classic atopic comorbidities like asthma and hay fever,
but also extend to ocular problems, sleep disturbance and mental
Complications of atopic dermatitis (AD) include both cutaneous and
health conditions. Recognizing the array of potential comorbid con-
extracutaneous conditions. Acute cutaneous complications include
ditions enables the clinician to work with consultants to treat the
both bacterial and viral infections that have unique presentations
whole patient.

Key points • Medical visits should include a discussion with the family regarding
a child’s sleep, mood, social functioning and school performance
• Caring for a child with atopic dermatitis (AD), especially one to help identify signs of mental health disorders such as anxiety,
with moderate‐to‐severe disease, encompasses much more than depression and attention‐deficit hyperactivity disorder.
treatment of the skin inflammation. • Recognizing the multidimensional impact AD may have on a
• Clinicians should identify Staphylococcus aureus infections of child aids the clinician in providing a comprehensive approach to
the skin and remain alert to the varied presentations of eczema the patient with the hopes of improving a child’s overall health
herpeticum. status.

­Bacterial infections Few studies address the rate of true infection (as com-
pared to colonization) in AD, possibly due to the difficulty
Epidemiology. Cutaneous infections with Staphylococcus in clinically differentiating infection from colonization.
aureus commonly complicate the course of atopic derma- One population‐based study from Japan found an almost
titis (AD), especially in patients with severe disease, and two‐fold increase of impetigo in children with a history of
are due to high Staph. aureus colonization rates. Lesional AD [7]. The risk of invasive Staph. aureus infections is also
colonization rates reported in the literature range between not well‐quantified in AD, but cases of endocarditis,
28% and 99%, with the first such study dating back to the osteomyelitis and septicaemia have all been reported in
1970s by Leyden et al. [1]. Totté et al., in a 2016 systematic this population [8–10].
review, identified 95 observational studies attempting to
quantify Staph. aureus colonization in AD [2]. Pooled esti-
mates for Staph. aureus carriage in AD were 70% on Pathogenesis. Microbiome studies confirm that AD
lesional skin, 39% on nonlesional skin and 62% in the flares are associated with heavy Staph. aureus growth and
nose. Studies showed significant variability in coloniza- correlate with a reduction in bacterial diversity on the
tion rates, probably due to differences in the ages of the skin [11]. Basic and translational research identifies sev-
patients and their underlying disease severity. This pro- eral potential pathogenic mechanisms underlying the
pensity for Staph. aureus colonization also leads to higher observed susceptibility to Staph. aureus colonization
colonization rates among family members [3]. Studies and infection. The elevated skin pH found in AD p­ romotes
conflict on whether patients with AD represent a signifi- Staph. aureus growth, as does a barrier disrupted by
cant reservoir of meticillin‐resistant Staph. aureus (MRSA) spongiosis and excoriation. Inflamed AD skin displays
­
colonization, with some studies showing increased colo- increased expression of fibronectin and fibrinogen‐binding
nization rates compared to the general population [4,5] sites for  Staph. aureus [12]. AD skin also exhibits an
and other studies failing to confirm these findings [6]. impaired ability to produce certain antimicrobial peptides

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
246 Section 3  Atopic Dermatitis and Related Disorders

(AMPs) that protect against Staph. aureus [13]. Underlying Hextall et  al. found no evidence that oral or topical
T‐helper type 2 (Th2) inflammation also probably plays antibiotics improved AD outcomes in clinically unin-
an important role in Staph. aureus susceptibility. Th2 fected patients [26]. An oral or topical antimicrobial can
cytokines inhibit AMP production by keratinocytes [14] be used depending on infection severity; there are no
and augment the destruction of keratinocytes by Staph. studies comparing various antimicrobial regimens in this
aureus alpha toxins [15]. AD patients colonized with Staph. population. Patients are at risk for developing antimicro-
aureus also display peripheral blood biomarkers consist- bial resistance with repeated or long‐term antibiotic
ent with Th2 skewing, such as elevated IgE levels and courses. Dicloxacillin or first‐generation cephalosporins
peripheral eosinophils [16]. The role of FLG mutations on are most commonly used as first‐line therapy. Antibiotics
Staph. aureus colonization/infection risk remains unclear such as clindamycin or trimethoprim–sulfamethoxazole
with some studies finding associations [17]. A study by may be needed in cases of MRSA. Dilute bleach baths
Lopes et al. suggests that different loss‐of‐function muta- have been shown to improve disease severity, and it was
tions in the FLG gene may have different effects on coloni- initially thought the effects were due to a reduction in
ATOPIC DERMATITIS

zation risk [18]. Staph. aureus colonization [27]. Wong et  al. measured a
Whereas several host factors allow Staph. aureus the reduction in Staph. aureus density with bleach bath use
SECTION 3:

opportunity to colonize, several Staph. aureus factors con- [28] but a recent trial found no additional benefit of bleach
tribute to AD severity, and in one study nasal colonization baths to standard topical corticosteroid treatment with
was found to actually precede AD onset in children [19]. respect to microbiome diversification [29]. Dilute bleach
Staph. aureus strains from AD patients, more so than from has, however, been shown to reduce inflammation by
normal controls, create exotoxins (e.g. toxic shock syndrome inhibiting expression of NF‐κB‐dependent genes [30].
toxin 1 and Staphylococcus enterotoxins) that act as superan- Further studies on the effects of bleach baths on Staph.
tigens leading to nonspecific activation of T cells and Th2 aureus colonization and infection rates are needed in AD.
cytokine production [20]. High levels of alpha toxin lead to
keratinocyte necrosis, whereas low levels promote inflam- ­References
mation [21]. Serine protease activity, both endogenous and 1 Leyden JJ, Marples RR, Kligman AM. Staphylococcus aureus in the
lesions of atopic dermatitis. Br J Dermatol 1974;90:525–30.
exogenous from Staph. aureus, probably adds to the deleteri- 2 Totté JE, van der Feltz WT, Hennekam M et al. Prevalence and odds
ous effects of Staph. aureus on the skin by degrading the skin of Staphylococcus aureus carriage in atopic dermatitis: a systematic
barrier and promoting inflammation [22,23]. review and meta‐analysis. Br J Dermatol 2016;175:687–95.
3 Bonness S, Szekat C, Novak N, Bierbaum G. Pulsed‐field gel electro-
phoresis of Staphylococcus aureus isolates from atopic patients
Clinical findings. Classic clinical findings of cutaneous revealing presence of similar strains in isolates from children and
Staph. aureus infection in children with AD include pus- their parents. J Clin Microbiol 2008;46:456–61.
tules and honey‐coloured crusts with erosion (Fig. 18.1). 4 Klein PA, Greene WH, Fuhrer J, Clark RA. Prevalence of methicillin‐
resistant Staphylococcus aureus in outpatients with psoriasis, atopic
Differentiating crusting and weeping found in acute AD dermatitis, or HIV infection. Arch Dermatol 1997;133:1463–5.
from Staph. aureus infection can be near impossible; how- 5 Chung HJ, Jeon HS, Sung H et al. Epidemiological characteristics of
ever, pain, erosion and pustulation are more frequently methicillin‐resistant Staphylococcus aureus isolates from children
found in infection. Overall AD severity and skin barrier with eczematous atopic dermatitis lesions. J Clin Microbiol 2008;
46:991–5.
function strongly associate with Staph. aureus coloniza- 6 Matiz C, Tom WL, Eichenfield LF et al. Children with atopic dermati-
tion [24,25]. tis appear less likely to be infected with community acquired methi-
cillin‐resistant Staphylococcus aureus: the San Diego experience.
Pediatr Dermatol 2011;28:6–11.
Treatment and  prevention. The use of antimicrobial 7 Hayashida S, Furusho N, Uchi H et  al. Are lifetime prevalence of
treatment in AD should be reserved for clinically appar- impetigo, molluscum and herpes infection really increased in chil-
ent infection. A systemic review of 26 studies by Bath‐ dren having atopic dermatitis? J Dermatol Sci 2010;60:173–8.
8 Benenson S, Zimhony O, Dahan D et al. Atopic dermatitis—a risk fac-
tor for invasive Staphylococcus aureus infections: two cases and
review. Am J Med 2005;118:1048–51. Review.
9 Hoeger PH, Ganschow R, Finger G. Staphylococcal septicemia in chil-
dren with atopic dermatitis. Pediatr Dermatol 2000;17:111–14.
10 Onoda K, Mizutan H, Komada T et al. Atopic dermatitis as a risk factor
for acute native valve endocarditis. J Heart Valve Dis 2000;9:469–71.
11 Kong HH, Oh J, Deming C et  al; NISC Comparative Sequence
Program, Murray PR, Turner ML, Segre JA. Temporal shifts in the
skin microbiome associated with disease flares and treatment in
children with atopic dermatitis. Genome Res 2012;22:850–9.
12 Cho SH, Strickland I, Boguniewicz M, Leung DY. Fibronectin and
fibrinogen contribute to the enhanced binding of Staphylococcus
aureus to atopic skin. J Allergy Clin Immunol 2001;108:269–74.
13 Ong PY, Ohtake T, Brandt C et al. Endogenous antimicrobial pep-
tides and skin infections in atopic dermatitis. N Engl J Med
2002;347:1151–60.
14 Howell MD, Boguniewicz M, Pastore S et al. Mechanism of HBD‐3
deficiency in atopic dermatitis. Clin Immunol 2006;121:332–8.
15 Brauweiler AM, Goleva E, Leung DY. Th2 cytokines increase

Staphylococcus aureus alpha toxin‐induced keratinocyte death
Fig. 18.1  Eroded plaques on the neck. Culture showed Staph. aureus and through the signal transducer and activator of transcription 6 (STAT6).
Group A Streptococcus. J Invest Dermatol 2014;134:2114–21.
Chapter 18  Atopic Dermatitis: Complications 247

16 Warner JA, McGirt LY, Beck LA. Biomarkers of Th2 polarity are pre- EH is much more common than EV and affects approxi-
dictive of staphylococcal colonization in subjects with atopic dermati-
mately 3% of AD patients [4]. Patients with more severe
tis. Br J Dermatol 2009;160:183–5.
17 Cai SC, Chen H, Koh WP et  al. Filaggrin mutations are associated skin disease and a history of other viral and bacterial
with recurrent skin infection in Singaporean Chinese patients with infections appear at greatest risk [5].
atopic dermatitis. Br J Dermatol 2012;166:200–3.
18 Lopes C, Rocha L, Sokhatska O et  al. Filaggrin polymorphism

Pro478Ser is associated with the severity of atopic dermatitis and Pathogenesis. The tendency towards exaggerated cuta-
colonization by Staphylococcal aureus. J Investig Allergol Clin neous viral infections observed in AD may be explained
Immunol 2016;26:70–2. by both underlying skin barrier and immune defects. FLG
19 Lebon A, Labout JA, Verbrugh HA et  al. Role of Staphylococcus
gene mutations confer a risk of EH in both European and
aureus nasal colonization in atopic dermatitis in infants: the
Generation R Study. Arch Pediatr Adolesc Med 2009;163:745–9. African populations [6], however it is unclear if the viral
Erratum in: Arch Pediatr Adolesc Med 2010;164:334. susceptibility is due to the barrier defect or the subse-
20 Akdis M, Simon HU, Weigl L et al. Skin homing (cutaneous lympho- quent immune alterations that occur downstream of a
cyte‐associated antigen‐positive) CD8+ T cells respond to superanti-
disrupted barrier. Observed immune defects include

ATOPIC DERMATITIS
gen and contribute to eosinophilia and IgE production in atopic
dermatitis. J Immunol 1999;163:466–75. peripheral blood Th2 skewing, expanded peripheral skin‐
21 Bantel H, Sinha B, Domschke W et  al. alpha‐Toxin is a mediator of homing T‐regulatory cells at the onset of the infection,

SECTION 3:
Staphylococcus aureus‐induced cell death and activates caspases via and reduced expression of interferon(IFN)‐γ and IFN‐γ
the intrinsic death pathway independently of death receptor signal-
ing. J Cell Biol 2001;155:637–48. receptor in peripheral blood mononuclear cells [5,7,8].
22 Williams MR, Nakatsuji T, Sanford JA et  al. Staphylococcus aureus Gao et  al. found loss‐of‐function variants in the IFN‐γ
induces increased serine protease activity in keratinocytes. J Invest receptor to be a predisposing factor for EH [9], partially
Dermatol 2017;137:377–84.
23 Hirasawa Y, Takai T, Nakamura T et al. Staphylococcus aureus extra-
explaining some of the immune abnormalities observed.
cellular protease causes epidermal barrier dysfunction. J Invest
Dermatol 2010;130:614–17. Clinical features. EH can start as a simple herpes labialis
24 Hon KL, Tsang YC, Pong NH et al. Clinical features and Staphylococcus infection with subsequent spreading to the head and neck
aureus colonization/infection in childhood atopic dermatitis. J
Dermatolog Treat 2016;27:235–40. or it may occur de novo in any area of the body. It is most
25 Simpson EL, Babineau D, Villarreal M et al. S. aureus colonization is often characterized by vesicles spreading beyond the
associated with impaired skin barrier function and greater Th2 activa- original focus of infection accompanied by fever and lym-
tion in adult subjects with AD. J Invest Dermatol 2016;136:S56.
26 Bath‐Hextall FJ, Birnie AJ, Ravenscroft JC, Williams HC. Interventions
phadenopathy. Often vesicles are not witnessed and the
to reduce Staphylococcus aureus in the management of atopic eczema: condition can be recognized by the presence of multiple
an updated Cochrane review. Br J Dermatol 2010;163:12–26. 2–3‐mm monomorphous punched‐out erosions (Fig. 18.2).
27 Huang JT, Abrams M, Tlougan B et al. Treatment of Staphylococcus A scalloped border created from coalesced erosions is
aureus colonization in atopic dermatitis decreases disease severity.
Pediatrics 2009;123:e808–14.
28 Wong SM, Ng TG, Baba R. Efficacy and safety of sodium hypochlorite
(bleach) baths in patients with moderate to severe atopic dermatitis in
Malaysia. J Dermatol 2013;40:874–80.
29 Gonzalez ME, Schaffer JV, Orlow SJ et  al. Cutaneous microbiome
effects of fluticasone propionate cream and adjunctive bleach baths in
childhood atopic dermatitis. J Am Acad Dermatol 2016;75:481–93.e8.
30 Leung TH, Zhang LF, Wang J et al. Topical hypochlorite ameliorates
NF‐kB‐mediated skin diseases in mice. J Clinc Invest 2013;123:
5361–70.

­Viral infections
Epidemiology. It is not clear whether patients with AD are
more susceptible to routine cutaneous viral infections such
as warts or molluscum. Studies are sparse and conflicting.
The tendency for patients with AD to develop exaggerated
viral infections such as to molluscum, vaccinia or cox-
sackie exposures, however, has been well documented.
Two exaggerated infections that are particularly
important to recognize in the AD patient are eczema vac-
cinatum (EV) and eczema herpeticum (EH), as these con-
ditions can cause acute and severe morbidity and
possibly death. EV is exceedingly rare and mainly occurs
when military recruits receiving the smallpox vaccine
inadvertently expose susceptible family members with
AD at home. Having AD or someone in the household
with AD is a strict contraindication to receiving the
smallpox vaccine. The ability of vaccinees to report these
contraindications is poor, however, and new cases of EV Fig. 18.2  Grouped crusted erosions consistent with eczema herpeticum in
are emerging [1–3]. a young adult.
248 Section 3  Atopic Dermatitis and Related Disorders

Treatment. Antiviral therapy should be initiated imme-


diately for EH, either oral or intravenously depending
on disease severity. Age less than 1 year, male sex, fever
and systemic symptoms were all important factors lead-
ing to admission in one large retrospective study of EH
paediatric patients [11]. Treatment of EV includes vac-
cinia immune globulin, cidofovir or tecovirimat  –  an
antiviral with anti‐orthopox activity [12]. Treatment
options for widespread warts or molluscum mirror
those in children without AD. Topical steroids do not
appear to complicate viral infections of the skin and may
be used. Topical calcineurin inhibitors are contraindi-
ATOPIC DERMATITIS

cated in active infection, although there are no rigorous


data regarding their role in predisposing to infection or
SECTION 3:

in disease exacerbation.

­References
1 Naleway AL, Belongia EA, Greenlee RT et al. Eczematous skin disease
and recall of past diagnoses: implications for smallpox vaccination.
Ann Intern Med 2003;139:1–7.
2 Moses AE, Cohen‐Poradosu R. Images in clinical medicine. Eczema
vaccinatum—a timely reminder. N Engl J Med 2002;346:1287.
3 Vora S, Damon I, Fulginiti V et  al. Severe eczema vaccinatum in a
household contact of a smallpox vaccinee. Clin Infect Dis 2008;46:
1555–61.
Fig. 18.3  Verrucae vulgaris in a child with atopic dermatitis. 4 Tay YK, Khoo BP, Goh CL. The epidemiology of atopic dermatitis at a
tertiary referral skin center in Singapore. Asian Pac J Allergy Immunol
1999;17:137–41.
5 Beck LA, Boguniewicz M, Hata T et al. Phenotype of atopic derma-
another morphological clue to the diagnosis. Atypical titis subjects with a history of eczema herpeticum. J Allergy Clin
presentations often occur and can include pustules, haem- Immunol 2009;124:260–9, 9.e1–7.
6 Gao PS, Rafaels NM, Hand T et  al. Filaggrin mutations that confer
orrhagic papules or crusts. Pain, fussiness and fever are risk  of atopic dermatitis confer greater risk for eczema herpeticum.
additional clues that EH should be considered. Herpes J Allergy Clin Immunol 2009;124:507–13, 513.e1–7.
keratitis may be a complication of EH, and viral dissemi- 7 Leung DY, Gao PS, Grigoryev DN et al. Human atopic dermatitis
complicated by eczema herpeticum is associated with abnormali-
nation may occur leading to death. ties in IFN‐γ response. J Allergy Clin Immunol 2011;127:965–73.
Other viral infections of note in children with AD e1–5. Erratum in: J Allergy Clin Immunol 2011;128:833.
include widespread wart or molluscum infections 8 Takahashi R, Sato Y, Kurata M et al. Pathological role of regulatory T
(Fig. 18.3). Recently, a particularly virulent strain of cox- cells in the initiation and maintenance of eczema herpeticum lesions.
J Immunol 2014;192:969–78.
sackie A6 has emerged leading to a cutaneous eruption 9 Gao L, Bin L, Rafaels NM et al. Targeted deep sequencing identifies
that may mimic EH, termed eczema coxsackium [10]. The rare loss‐of‐function variants in IFNGR1 for risk of atopic dermatitis
exanthem affects hands and feet like a typical coxsackie complicated by eczema herpeticum. J Allergy Clin Immunol 2015;
136:1591–600.
infection, but commonly affects perioral areas and trunk
10 Mathes EF, Oza V, Frieden IJ et al. “Eczema coxsackium” and unusual
with accentuation in areas of AD lesions. cutaneous findings in an enterovirus outbreak. Pediatrics 2013;132:
e149–57.
Laboratory testing. Samples of blister fluid should be 11 Luca NJ, Lara‐Corrales I, Pope E. Eczema herpeticum in children:
clinical features and factors predictive of hospitalization. J Pediatr
sent for polymerase chain reaction (PCR) testing for 2012;161:671–5.
viral DNA or for culture. Electron microscopy and 12 Jordan R, Leeds JM, Tyavanagimatt S, Hruby DE. Development

immunofluorescence testing are alternative methods for of  ST‐246® for treatment of poxvirus infections. Viruses 2010;2:
2409–35.
confirming the presence of HSV. Serological testing has
no value as many children have been exposed to HSV‐1
and will test positive. If EV is suspected, local health
­Sleep disturbance
authorities and, in the USA, the Centers for Disease
Control should be contacted for immune globulin ther- Sleep disturbance is a common issue faced by patients
apy consideration. with AD as well as their families. It affects up to 60% of
Coxsackie virus testing can be performed using culture children with AD, with closer to 80% experiencing
or PCR from blister fluid, oropharynx or stool sample, but sleep disturbance during flares [1]. Patients with AD
the diagnosis is most often made on clinical grounds. have difficulty falling asleep as well as staying asleep,
Testing of the stool or a swab from the rectal area may leading to difficulty awakening, daytime tiredness and
provide greater sensitivity (and perhaps less specificity) irritability [1–5]. Nocturnal pruritus and scratching
for detection since shedding of the virus in the gastroin- lead to difficulty falling asleep, as well as frequent
testinal tract may continue even after clinical disease is no movements during sleep and prolonged awakenings.
longer evident. Some sleep changes are even seen when the AD is in
Chapter 18  Atopic Dermatitis: Complications 249

clinical remission [3]. Interestingly, while prolonged 8 Chamlin SL, Mattson CL, Frieden IJ et al. The price of pruritus: Sleep
disturbance and cosleeping in atopic dermatitis. Arch Pediatr Adolesc
nighttime arousals are common in patients with AD,
Med 2005;159:745–50.
total sleep time and overall sleep architecture appear to 9 Romanos M, Gerlach M, Warnke A, Schmitt J. Association of
be similar to healthy children [1,3,6,7], perhaps point- attention‐deficit/hyperactivity disorder and atopic eczema modified
ing to compensatory mechanisms whereby children by sleep disturbance in a large population‐based sample. J Epidemiol
Community Health 2010;64:269–73.
modify their bed and rise times [1]. Parents also suffer 10 Stewart AC, Thomas SE. Hypnotherapy as a treatment for atopic der-
from fragmented, disrupted sleep as they care for their matitis in adults and children. Br J Dermatol 1995;132:778–83.
children with AD. This familial sleep disruption can 11 Klein PA, Clark RF. An evidence‐based review of the efficacy of anti-
negatively impact the reported happiness of parents histamines in relieving pruritus in atopic dermatitis. Arch Dermatol
1999;135:1522–5.
and children struggling with AD [8]. Similar to children 12 Sidbury R, Tom WL, Bergman JN et al. Guidelines of care for the man-
without AD, sleep disturbance has also been linked to agement of atopic dermatitis: Section 4. Prevention of disease flares
impaired neurocognitive functioning and behavioural and use of adjunctive therapies and approaches. J Am Acad Dermatol
2014;71:1218–33.
issues [5]. Lack of sleep may be one component contrib-

ATOPIC DERMATITIS
13 Endo K, Sano H, Fukuzumi T et al. Objective scratch monitor evalua-
uting to the association of AD with attention deficit tion of the effect of an antihistamine on nocturnal scratching in atopic
hyperactivity disorder (ADHD) and other mental dermatitis. J Dermatol Sci 1999;22:54–61.

SECTION 3:
health disorders [9] (see Mental health disorders). 14 Schad CA, Skoner DP. Antihistamines in the pediatric population:
Achieving optimal outcomes when treating seasonal allergic rhinitis
and chronic urticaria. Allergy Asthma Proc 2008;29:7–13.
Treatment. Few studies have evaluated specific treat- 15 Chang Y‐S, Lin M‐H, Lee J‐H et  al. Melatonin supplementation for
children with atopic dermatitis and sleep disturbance: a randomized
ments for improving sleep in patients with AD. One clinical trial. JAMA Pediatr 2016;170:35.
study showed improvement in sleep with hypnother- 16 Sowden JM, Allen BR, Berth‐Jones J et  al. Double‐blind, controlled,
apy as reported by parents using a 10‐point visual ana- crossover study of cyclosporin in adults with severe refractory atopic
logue scale [10]. Antihistamines have long been used in dermatitis. The Lancet 1991;338:137–40.
17 Leo HL, Bender BG, Leung SB et al. Effect of pimecrolimus cream 1%
the treatment of AD for their soporific effects, with a on skin condition and sleep disturbance in children with atopic der-
goal of decreasing sleep latency as well as nighttime matitis. J Allergy Clin Immunol 2004;114:691–3.
scratching and awakenings; however, most of the evi- 18 Bieber T, Vick K, Fölster‐Holst R et al. Efficacy and safety of methyl-
prednisolone aceponate ointment 0.1% compared to tacrolimus 0.03%
dence for this is anecdotal [11]. Short‐term intermittent in children and adolescents with an acute flare of severe atopic der-
use of sedating antihistamines may be helpful in com- matitis. Allergy 2007;62:184–9.
bating sleep disturbance, but more research in this area
is needed [12,13]. The sedative effect of these drugs may
help with sleep but care is needed in prescribing to chil- ­Psychosocial complications
dren because sedating antihistamines have been shown
to affect school performance [14]. Melatonin supple- AD imposes a tremendous financial, social, emotional and
mentation has also been investigated, and in one rand- personal toll on patients and their families. These burdens
omized controlled trial of 48 children it decreased are not always immediately evident but nonetheless
sleep‐onset latency and led to an improvement in dis- important to consider when treating children with AD
ease severity. No significant difference was seen, how- and in interacting with caregivers. The interplay between
ever, in patient self‐reported sleep parameters [15]. children’s psychosocial environment and their skin dis-
Sleep can also improve with management of the under- ease is complex and multidirectional. Increased personal
lying AD. Studies evaluating the changes in sleep dur- stress and family tension can increase the severity of AD
ing treatment of AD, with both topical and systemic [1], while worsening AD can lead to increased stress.
treatments, have shown some improvement in sleep Chronic AD can also lead to high rates of psychological
parameters [1,16–18]. disturbance in children, causing behavioural difficulties
and an impaired capacity to cope with stress [2,3].
­References AD most often begins in early infancy or childhood,
1 Camfferman D, Kennedy JD, Gold M et al. Eczema and sleep and its coinciding with crucial periods of psychosocial develop-
relationship to daytime functioning in children. Sleep Med Rev ment. Children with AD have behavioural difficulties,
2010;14:359–69.
increased dependency, fearfulness and clinginess [4].
2 Dahl RE, Bernhisel‐Broadbent J, Scanlon‐Holdford S et al. Sleep dis-
turbances in children with atopic dermatitis. Arch Pediatr Adolesc Noticeable skin disease can lead to difficulties in social
Med 1995;149:856–60. development with poor self‐esteem, teasing and isolation
3 Reuveni H, Chapnick G, Tal A, Tarasiuk A. Sleep fragmentation in from peers. Other children and adults may avoid contact
children with atopic dermatitis. Arch Pediatr Adolesc Med 1999;153:
249–53.
with children with AD, due to fear of a communicable
4 Bender BG, Leung SB, Leung DY. Actigraphy assessment of sleep dis- condition [5]. This can lead to poor self‐image and low
turbance in patients with atopic dermatitis: an objective life quality self‐esteem. In school‐age children academic performance
measure. J Allergy Clin Immunol 2003;111:598–602. may be negatively affected by missed days, as well as
5 Camfferman D, Kennedy JD, Gold M et al. Sleep and neurocognitive
functioning in children with eczema. Int J Psychophysiol 2013;89: poor attention related to lack of sleep and possibly ADHD
265–72. (see Mental health disorders).
6 Stores G, Burrows A, Crawford C. Physiological sleep disturbance in The psychosocial burden is felt by caregivers as well. As
children with atopic dermatitis: a case control study. Pediatr Dermatol
1998;15:264–8.
with most chronic childhood illness, AD has been shown to
7 Monti JM, Vignale R, Monti D. Sleep and nighttime pruritus in chil- lead to increased family stress. The lack of sleep and paren-
dren with atopic dermatitis. Sleep 1989;12:309–14. tal anxiety can lead to higher levels of depression. Parents
250 Section 3  Atopic Dermatitis and Related Disorders

can feel tremendous amounts of guilt related to their child’s 7 Fivenson D. The effect of atopic dermatitis on total burden of illness
and quality of life on adults and children in a large managed care
AD. Mothers caring for children with AD report feelings of
organization. J Manag Care Pharm 2002;8:333–42.
social isolation due to their child’s illness and they are less 8 Ersser SJ, Cowdell F, Latter S et al. Psychological and educational
likely to be employed outside the home [4]. Strangers and interventions for atopic eczema in childrenCochrane Database Syst
family members may offer conflicting advice as well as Rev 2014;CD004054.
9 Staab D. Age related, structured educational programmes for the
judgement regarding care of the child’s AD. This can be dif- management of atopic dermatitis in children and adolescents: mul-
ficult to manage as a parent or primary caregiver [5]. ticentre, randomised controlled trial. BMJ 2006;332:933–8.
Management of AD can be equally demanding of parents’ 10 Chida Y, Steptoe A, Hirakawa N et al. The effects of psychological inter-
economic resources, with missed days of work, frequent vention on atopic dermatitis. Int Arch Allergy Immunol 2007;144:1–9.
specialist visits and prescription and non‐prescription
treatments. This is especially true in our modern health- ­Mental health disorders
care system with increasing patient financial responsibil-
ity. Costs can be divided into direct and indirect costs. Since the early 1900s, a link between one’s mental state
ATOPIC DERMATITIS

Direct costs include funds used to provide care for AD, and AD has been recognized, leading to the term ‘neuro-
including physician visits and medications. Indirect costs dermatitis’ [1]. Psychological disturbances affect children
SECTION 3:

include loss of productivity resulting from missed work/ with AD more so than their peers, with the severity of
school, as well as poor work performance and changes in disturbance related to the severity of the skin disease
quality of life. Annual direct costs in the USA have been [2–4]. Parents of children with AD experience higher lev-
estimated at US$1–4 billion [6]. Indirect costs are more els of stress and difficulty with child discipline [5].
difficult to quantify but one study found loss of work to Several population‐based studies revealed that children
account for at least 50% of the total economic burden [7]. with AD, especially those with severe disease or with
sleep disturbance, often carry a diagnosis of ADHD [6].
Treatment. AD has many effects on children’s lives More detailed study of this population is needed to
beyond the skin. Correspondingly, our treatment determine whether these children have true ADHD
should aim to address the psychological and emotional upon careful diagnostic scrutiny, or whether they have
needs of our patients and their families. Psychosocial been misclassified based on the symptoms of severe AD.
interventions can be primarily educational or more Other neuropsychiatric disorders have been reported to
structured psychological therapy, depending on need. be increased in children with AD including anxiety,
Educational programmes have been studied in a ­variety depression and autism [7–10]. These studies have been
of settings including multidisciplinary teams, nurse‐led cross‐sectional in nature and higher levels of evidence
groups, educational videos and parent‐ or patient‐ are needed to confirm these associations and establish
directed education [8]. The largest study of this kind causal relationships.
evaluated the ­effectiveness of age‐specific education
for 992 families. After 6 weeks of weekly sessions, sig- Pathogenesis. The causal relationships between AD and
nificant reductions in disease severity as well as mental disorders are not understood. Chronic sleep dis-
improvement in quality of life measures were seen [9]. turbance from pruritus may be an important causal link.
Psychological treatments can include cognitive behav- It is well‐known that disrupted sleep patterns negatively
ioural therapy, hypnotherapy, individual and family affect the mental state of normal children, affecting behav-
therapy and stress management. These interventions iour, mood and school functioning [11–13]. Disrupted sleep
probably have significant benefit but are less well stud- patterns are features of childhood depression, anxiety,
ied. The need for additional high‐quality studies in this ADHD and autism [14,15]. Inflammatory mediators may
area was highlighted by a Cochrane review as well as a also play a role. Pro‐inflammatory cytokines may not
meta‐analysis [8,10]. only affect skin, but could affect the developing brain
[16]. Pro‐inflammatory cytokines have been found in
­References patients with depression, autism and ADHD [17–19].
1 King RM, Wilson GV. Use of a diary technique to investigate psychoso-
matic relations in atopic dermatitis. J Psychosom Res 1991;35:697–706.
2 Absolon CM, Cottrell D, Eldridge SM, Glover MT. Psychological dis- Treatment. A good medical history and exploration of a
turbance in atopic eczema: the extent of the problem in school‐aged child’s mood, behaviour, social life and school perfor-
children. Br J Dermatol 1997;137:241–5. mance with the family helps identify concerning pat-
3 Saunes M, Smidesang I, Holmen TL, Johnsen R. Atopic dermatitis in
adolescent boys is associated with greater psychological morbidity
terns. Referral to a childhood psychologist or other
compared with girls of the same age: the Young‐HUNT study. Br J mental health professional can yield significant benefits
Dermatol 2007;156:283–8. for both child and family and potentially improve skin
4 Daud LR, Garralda ME, David TJ. Psychosocial adjustment in pre- outcomes.
school children with atopic eczema. Arch Dis Child 1993;69:670–6.
5 Chamlin SL, Frieden IJ, Williams ML, Chren M‐M. Effects of atopic
dermatitis on young American children and their families. Pediatrics ­References
2004;114:607–11. 1 Brunsting LA. Atopic dermatitis (disseminated neurodermatitis) of
6 Mancini AJ, Kaulback K, Chamlin SL. The socioeconomic impact of young adults: analysis of precipitating factors in one hundred and
atopic dermatitis in the United States: a systematic review. Pediatr one cases and report of ten cases with associated juvenile cataract.
Dermatol 2008;25:1–6. Arch Derm Syphilol 1936;34:935–57.
Chapter 18  Atopic Dermatitis: Complications 251

2 Faulstich ME, Williamson DA, Duchmann EG et al. Psychophysiological reveal giant cobblestone‐like papillae of the upper tarsal
analysis of atopic dermatitis. Jo Psychosom Res 1985;29:415–17.
conjunctiva or at the limbus. Although vernal keratocon-
3 Absolon CM, Cottrell D, Eldridge SM, Glover MT. Psychological dis-
turbance in atopic eczema: the extent of the problem in school‐aged junctivitis has a favourable prognosis, with resolution in
children. Br J Dermatol 1997;137:241–5. the majority of patients around puberty, it can be associ-
4 Chamlin SL. The psychosocial burden of childhood atopic dermati- ated with permanent reduction in visual acuity due to cor-
tis. Dermatol Ther 2006;19:104–7.
5 Daud LR, Garralda ME, David TJ. Psychosocial adjustment in pre-
neal damage in about 6% of patients [5]. Atopic
school children with atopic eczema. Arch Dis Child 1993;69:670–6. keratoconjunctivitis is bilateral and symmetrical, usually
6 Schmitt J, Buske‐Kirschbaum A, Roessner V. Is atopic disease a risk with associated eyelid swelling, erythema and scaling
factor for attention‐deficit/hyperactivity disorder? A systematic (atopic blepharoconjunctivitis) and is more prone to scar-
review. Allergy 2010;65:1506–24.
7 Yaghmaie P, Koudelka CW, Simpson EL. Psychiatric comorbidity in ring than vernal keratoconjunctivitis. Symptoms of itch-
pediatric eczema. J Invest Dermatol 2011;131(suppl 1):S41. Abstract #246. ing, discharge and conjunctival injection are similar to
8 Magalhaes ES, Pinto‐Mariz F, Bastos‐Pinto S et  al. Immune allergic vernal keratoconjunctivitis [2]. Patients with AD are also
response in Asperger syndrome. J Neuroimmunol 2009;216(1‐2):108–12.
prone to developing both irritant and allergic contact der-

ATOPIC DERMATITIS
9 Mostofa GA, Hamza RT, El‐Shahawi HH. Allergic manifestations in
autistic children: Relation to disease severity. J Pediatr Neurol 2008; matitis and blepharitis affecting the eyelids [4].
6:115–23. Other ocular complications including cataracts, kerato-

SECTION 3:
10 Sacco R, Curatolo P, Manzi B et al. Principal pathogenetic components conus and retinal detachment are uncommon in patients
and biological endophenotypes in autism spectrum disorders. Autism
Res 2010;3:237–52. with AD but can result in severe impairment [3,6,7]. Both
11 Yokomaku A, Misao K, Omoto F et  al. A study of the association posterior and anterior subcapsular cataracts have been
between sleep habits and problematic behaviors in preschool chil- described in AD [4,6]. Anterior subcapsular cataracts are
dren. Chronobiol Int 2008;25:549–64.
less commonly seen but are more specific for AD, as they
12 Hiscock H, Canterford L, Ukoumunne OC, Wake M. Adverse associa-
tions of sleep problems in Australian preschoolers: national popula- are not seen frequently in patients without AD [6]. The
tion study. Pediatrics 2007;119:86–93. aetiology of cataracts in patients with AD is not com-
13 Gregory AM, Van der Ende J, Willis TA, Verhulst FC. Parent‐reported pletely understood. It does not seem to correlate with
sleep problems during development and self‐reported anxiety/
depression, attention problems, and aggressive behavior later in life.
severity of AD, or corticosteroid therapy [4,6,8–10]. Some
Arch Pediatr Adolesc Med 2008;162:330–5. studies have implicated oxidative stress to the lens result-
14 Owens JA, Maxim R, Nobile C et al. Parental and self‐report of sleep ing from chronic inflammation [6,8].
in children with attention‐deficit/hyperactivity disorder. Arch Keratoconus is a noninflammatory progressive ectasia
Pediatr Adolesc Med 2000;154:549–55.
15 Ivanenko A, Johnson K. Sleep disturbances in children with psychiat- of the cornea. It is characterized by thinning and protru-
ric disorders. Semin Pediatr Neurol 2008;15:70–8. sion of the central cornea [11,12]. Classically it presents at
16 Buehler MR. A proposed mechanism for autism: an aberrant neuroim- puberty and progresses until the third or fourth decade
mune response manifested as a psychiatric disorder. Med Hypotheses
2011;76:863–70.
[13]. Evidence suggests that keratoconus is related more
17 Kiecolt‐Glaser JK, Belury MA, Andridge R et al. Omega‐3 supplemen- to the chronic rubbing of the eyelid commonly seen in
tation lowers inflammation and anxiety in medical students: a rand- patients with AD, than to the presence of AD alone [12].
omized controlled trial. Brain Behav Immun 2011;25:1725–34. Retinal detachment is a rare ocular complication of AD
18 Pollak Y, Yirmiya R. Cytokine‐induced changes in mood and behav-
iour: implications for ’depression due to a general medical condi- that is primarily seen in Japanese patients with facial
tion’, immunotherapy and antidepressive treatment. Int J involvement [14–16]. The aetiology seems to be related to
Neuropsychopharmacol 2002;5:389–99. trauma caused by either rubbing or tapping the eyes or
19 Zimmerman AW, Jyonouchi H, Comi AM et  al. Cerebrospinal fluid
eyelids but it can also occur as a complication following
and serum markers of inflammation in autism. Pediatr Neurol
2005;33:195–201. surgical correction of cataracts [14,16,17].
These ocular complications require prompt treatment
and evaluation, usually with the help of an ophthalmolo-
­Ocular complications gist. Periodic ocular examination and questioning regard-
ing ocular symptoms should be a part of routine
Atopic eye disease encompasses similar but distinguisha- dermatological care for a patient with AD.
ble conditions including seasonal allergic conjunctivitis,
vernal keratoconjunctivitis, atopic keratoconjunctivitis
­References
and atopic blepharoconjunctivitis. Atopic keratoconjunc- 1 Chen JJ, Applebaum DS, Sun GS, Pflugfelder SC. Atopic kerato-
tivitis is a chronic condition characterized by noninfec- conjunctivitis: A review. J Am Acad Dermatol 2014;70:569–75.
tious inflammation and is one of the most severe and 2 Guglielmetti S, Dart JK, Calder V. Atopic keratoconjunctivitis and
atopic dermatitis. Curr Opin Allergy Clin Immunol 2010;10:478–85.
potentially damaging ocular complications of AD [1]. 3 Carmi E, Defossez‐Tribout C, Ganry O et  al. Ocular complications
Atopic keratoconjunctivitis is most often seen in adults of atopic dermatitis in children. Acta Derm Venereol 2006;86:515–17.
with onset ranging from 20 to 50 years of age but it has 4 Rich LF, Hanifin JM. Ocular complications of atopic dermatitis and
been reported in children as young as 7 years old [1], other eczemas. Int Ophthalmol Clin 1985;25:61–76.
5 Bonini S, Bonini S, Lambiase A et  al. Vernal keratoconjunctivitis
while vernal keratoconjunctivitis is more common in chil- revisited: A case series of 195 patients with long‐term followup.
dren, typically appearing between 5 and 15 years of age Ophthalmology 2000;107:1157–63.
[2–4]. Vernal keratoconjunctivitis is seen more often in 6 Bair B, Dodd J, Heidelberg K, Krach K. Cataracts in atopic dermati-
tis: A case presentation and review of the literature. Arch Dermatol
boys and is characterized by intense itching, mucous dis- 2011;147:585–8.
charge, conjunctival injection and photophobia that is not 7 Hirano S, Katoh N, Kishimoto S et  al. Cataract in atopic dermatitis
always bilateral [2,5]. Ophthalmological examination can with facial involvement. J Dermatol Sci 1993;6:94.
252 Section 3  Atopic Dermatitis and Related Disorders

8 Niwa Y, Iizawa O. Abnormalities in serum lipids and leukocyte 14 Hida T, Tano Y, Okinami S et  al. Multicenter retrospective study of
superoxide dismutase and associated cataract formation in patients retinal detachment associated with atopic dermatitis. Jpn J
with atopic dermatitis. Arch Dermatol 1994;130:1387–92. Ophthalmol 2000;44:407–18.
9 Garrity JA, Liesegang TJ. Ocular complications of atopic dermatitis. 15 Yoneda K, Okamoto H, Wada Y et  al. Atopic retinal detachment.
Can J Ophthalmol 1984;19:21–4. Report of four cases and a review of the literature. Br J Dermatol
10 Tm B, Ja B, Jm S. Atopic dermatitis: a case report and current clinical 1995;133:586–91.
review of systemic and ocular manifestations. Optom St Louis Mo 16 Taniguchi H, Ohki O, Yokozeki H et al. Cataract and retinal detach-
2001;72:94–102. ment in patients with severe atopic dermatitis who were with-
11 Krachmer JH, Feder RS, Belin MW. Keratoconus and related nonin- drawn from the use of topical corticosteroid. J Dermatol 1999;26:
flammatory corneal thinning disorders. Surv Ophthalmol 1984;28: 658–65.
293–322. 17 Oka C, Ideta H, Nagasaki H et al. Retinal detachment with atopic der-
12 Bawazeer AM, Hodge WG, Lorimer B. Atopy and keratoconus: a matitis similar to traumatic retinal detachment. Ophthalmology 1994;
multivariate analysis. Br J Ophthalmol 2000;84:834–6. 101:1050–4.
13 Rabinowitz YS. Keratoconus. Surv Ophthalmol 1998;42:297–319.
ATOPIC DERMATITIS
SECTION 3:
 253

C HA PTER   19

Management of Atopic Dermatitis


Lea Solman & Mary Glover
Department of Paediatric Dermatology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK

Introduction, 253 Systemic therapy, 257 Psychological impact of atopic


Education and general advice, 253 Biologics in atopic dermatitis, 259 dermatitis, 261
Initial therapy, 254 Allergy in atopic dermatitis, 260 Conclusion, 261

ATOPIC DERMATITIS
Unresponsive disease, 256 Prevention of atopic dermatitis, 261

SECTION 3:
Abstract of emollients to improve the integrity of the skin barrier, topical
application of corticosteroids and calcineurin inhibitors, avoidance
Atopic dermatitis is a chronic inflammatory disease, affecting more of apparent exacerbating factors, and multidisciplinary patient
than 20% of children in some developed countries. The main aims education. In patients with severe atopic dermatitis that is nonre-
of treatment are to control symptoms, prevent exacerbations and sponsive to topical treatment, phototherapy or systemic therapy
minimize therapeutic risk. Initial therapy consists of the application may be required.

Key points • Systemic immunomodulating medications are indicated for


the management of severe atopic dermatitis that has failed
to respond to appropriate topical treatment and attention to
• The management of atopic dermatitis is complex and will
aggravating factors.
depend on the age of the child, the extent, severity, location
• There is relatively little experience with biological response
and chronicity of the condition; it must take into consideration
modifiers in the management of childhood atopic dermatitis,
social and environmental factors.
but they can be expected to play an increasing role.
• Education, emollients and topical corticosteroids are the
mainstay in the treatment of childhood atopic dermatitis.

Introduction so need the opportunity to understand when and why a


particular treatment or intervention is needed.
The management of atopic dermatitis (AD) is complex, and Attention to education of families of children with
will depend on the age of the child and the extent, severity, atopic dermatitis (AD) has been shown to reduce disease
location and chronicity of the condition (Fig. 19.1); it must severity and improve quality of life over a period of at
take into consideration social and environmental factors. least 1 year [1]. Discussion time with families is important
Although prevention remains the ideal, early effective to dispel misconceptions, bring clarity to areas of confu­
treatment offers the best hope of reducing the likelihood sion, limit exposure of the child to irritants, and prevent
of progression to chronic disease and the risk of develop­ inappropriate or harmful treatments and unnecessary
ment of allergen sensitization. dietary intervention. It is particularly important to dis­
Families of children with AD are often subjected to cuss the role of factors that may impair the epidermal
conflicting advice, particularly in relation to the safety of barrier, including low‐humidity environments and expo­
topical corticosteroids and the role of diet, leading to sure to soaps and detergents. Parents should be reminded
confusion, anxiety, adoption of unhelpful interventions that AD may worsen during common childhood illnesses
and failure to use effective treatment. Effective, safe and teething. Family members should be discouraged
management depends on listening to and addressing from smoking in the house.
concerns, full explanation of the rationale for specific Many parents have been led to believe that food
treatments and discussion of risk and benefit. allergies are the primary driving force of AD, leading
to children being on exclusion diets despite lack of evi­
dence of a clinically relevant allergy, or of benefit from
Education and general advice
the exclusion.
The management of AD is complicated, time‐consuming, Although swimming pool water can lead to drying of
messy and often resisted by the children concerned. the skin, because swimming is a vitally important life skill
Parents are frequently subjected to conflicting advice and and an enjoyable family activity, patients with AD should

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
254 Section 3  Atopic Dermatitis and Related Disorders

Biologic therapy

T Systemic therapy

r
Phototherapy
e
a
Calcineurin inhibitors Calcineurin inhibitors
t
m
Mild-potency TCS Moderate-potency TCS Potent TCS
e
n
Emollient Emollient Emollient
t
ATOPIC DERMATITIS

Mild eczema Moderate eczema Severe eczema


SECTION 3:

Fig. 19.1  Treatment options in atopic dermatitis.


Eczema severity
TCS, topical corticosteroids.

not be excluded from swimming lesions but should be of symptoms [5]. Emollients should be applied at least
encouraged to shower and moisturize thoroughly after twice a day and immediately after bathing and hand
swimming. washing. It can be useful to allow patients to try a range
Parents should be advised that live vaccines should be of emollients. To reduce the risk of folliculitis, emollients
avoided in children on systemic immunosuppressive should be applied in the direction of hair growth. In warm
medication, but the vaccination schedule should be conditions, less greasy emollients will be required. It is
followed as normal in other children with AD. important to prescribe sufficient quantities of emollients
(250–500 g weekly).
Initial therapy
Topical corticosteroids
Bathing TCS have been the mainstay of therapy for AD since they
Parents frequently receive conflicting advice regarding were first introduced in 1952. The anti‐inflammatory
the benefits of bathing, probably because hard water, action is thought to arise mainly from regulation of gene
detergents and soaps can aggravate AD. However, bath­ transcription through the ligand‐activated glucocorti­
ing with moisturizing cleansers hydrates the skin and coid receptor and posttranslational regulation of gene
the majority of patients find it soothing. The skin must expression [6].
be cleansed gently, but thoroughly, to remove skin scales, Age, anatomical site and vehicle, as well as severity
crusts and exudate. Nonirritant, moisturizing soap and type of AD, should be taken into consideration when
substitutes with or without antiseptics should be used. prescribing TCS treatment. TCS are available in a variety
The addition of bath oil provides the skin with a thin of potencies and preparations. The European classifi­
lipid layer. Caution needs to be taken when adding oils cation uses four potency groups from mild (group I) to
to the bath, as the bathtub can become very slippery. super‐potent (group IV). The American classification uses
Bath time should be kept to around 7–10 minutes. The seven groups, with group 1 being the most potent and
water temperature should be just warm enough for group 7 the least potent.
the child to be comfortable, but not hot. The skin should For dry skin, application of TCS in ointment base is
be patted dry rather than rubbed, and an emollient most effective, providing better lubrication and occlu­
applied while the skin is still slightly damp. sion than a cream base, though the greasy quality of
ointments can be unappealing, particularly in warm
Emollients conditions, and may result in poor compliance. Creams
Poor skin barrier function is a key risk factor for develop­ are more cosmetically acceptable but contain preserva­
ment of AD [2,3]. Skin barrier dysfunction enables aller­ tives, which can cause stinging, irritation and contact
gen penetration into the skin, leading to irritation and dermatitis. Gels and lotions are the least greasy and so
inflammation. Application of emollients improves the are convenient to use on the scalp.
integrity of the skin barrier. In a randomized controlled Absorption of TCS has been demonstrated to vary
trial (RCT) of infants with AD requiring moderate‐ or not only between individuals but also with anatomical
high‐potency topical corticosteroids (TCS), infants treated location [7]. Anatomical regions with thinner epidermis
with emollients had significantly decreased requirements are significantly more permeable than areas with thicker
for TCS compared with a control group of infants who skin. While absorption on the forearm is relatively low
were not treated with emollients [4]. A more recent RCT (1%), the scalp absorbs around 4% and the scrotum up to
demonstrated that the regular use of emollients alone in 35% of applied TCS [7]. Penetration varies between eyelid
children with mild‐to‐moderate AD reduces the severity and plantar skin about 300‐fold [7]. Corticosteroids are
Chapter 19  Management of Atopic Dermatitis 255

better absorbed through areas of inflammation and Wet wraps


desquamation than normal skin, and more readily Wet wraps can be useful for severe AD, especially in
through the thin stratum corneum of infants [8]. Hydration young children. A widely used technique involves the
promotes steroid penetration, for example, after a bath or application to the torso and limbs of tubular bandage
shower. To limit the risk of adverse effects, eyelid and soaked in diluted TCS cream, changed twice a day, for
genital AD should be treated with low‐potency topical 3–5 days. Another approach is to apply TCS directly to
steroid. Low‐ to medium‐strength preparations should the areas of inflammation, followed by dampened tubular
be considered when large areas are treated, to limit sys­ bandage, then a dry layer of tubular bandage. Wet wraps
temic absorption, but medium‐ to high‐potency TCS serve as an occlusive dressing to prevent itching and
may be considered for limited areas of severe nonfacial/ improve retention of the TCS on the skin. Wet wraps can
nonintertriginous AD. be administered in hospital, but may be used at home if
Most studies on TCS use in AD have involved twice‐ the parents are educated and closely supervised by a
daily application, but it has been suggested that once‐ specialist nurse. Emollient‐only wet wraps are less effec­

ATOPIC DERMATITIS
daily application may not result in loss of efficacy and tive than wet wraps using diluted TCS [13], but can be
could lead to fewer local side‐effects. Application of TCS soothing and may help to reduce nocturnal pruritus.

SECTION 3:
just once a day may be more convenient for patients and
may save costs if established preparations are used [9]. Silk therapeutic clothing
Application of TCS of appropriate potency in sufficient Preliminary results of an RCT to evaluate the effective­
quantities early after the onset of AD, and promptly to ness of silk therapeutic clothing when used in addition
flares, offers the best chance of bringing the inflamma­ to usual care over a period of 6 months in children aged
tory process under rapid control, allowing restoration of 1–15 years with AD suggest that that there is unlikely to
skin barrier function and so keeping TCS requirement to be additional benefit [14].
a minimum. The TCS must be sufficiently potent and
applied in sufficient quantities to induce remission over Topical calcineurin inhibitors
a matter of days, so that control can then be maintained Topical calcineurin inhibitors (TCI) are macrolactam deriv­
primarily with emollients and avoidance of aggravating atives with immune‐modulating and anti‐inflammatory
factors. properties. Their anti‐inflammatory effects result from
In order to reassure parents about TCS, clear informa­ inhibition of pro‐inflammatory cytokine production from
tion must be provided and sufficient time allocated to T cells, whereas their antipruritic effects are attributed to
listen and respond fully to their concerns. Education of an inhibition of mast cell degranulation [15]. Pimecrolimus
parents, carers and patients is an essential aspect of TCS cream and tacrolimus ointment have been licensed for the
prescription, with particular attention to the significance treatment of AD in the UK since 2001, subject to National
of different potencies, and how much, for how long, and Institute for Health and Care Excellence (NICE) guidance.
to what site each TCS should be applied. Demonstration Both topical pimecrolimus and tacrolimus (0.03%) are
of exactly how to apply the preparation, which areas second‐line treatments for children in whom AD is not
should be covered and how thick the layer of ointment or adequately controlled with TCS, or when prolonged
cream should be is extremely helpful. Although the fin­ use of TCS is not advisable. They can be used in non‐
gertip unit has been used widely as a simple guide to how immunocompromised patients as steroid‐sparing agents
much TCS should be applied to a particular area [10], this for the treatment of mild‐to‐moderate AD involving the
method can lead to dabbing of inadequate amounts of the face, including the eyelids, neck and skin folds. Three
TCS, resulting in undertreatment. studies on pimecrolimus noted greater improvement on
Anxiety among both the general public and some fam­ the face and neck compared to other body sites [16–18].
ily doctors about potential adverse effects of TCS has led TCI can be used as a proactive, intermittent treatment two
to widespread inadequate treatment [11]. A survey of 200 or three times a week to prevent relapses [19]. TCS may be
dermatology outpatients with AD found that 72.5% were required initially, to induce remission.
worried about use of TCS on their own or their child’s TCI applied once or twice a day have been shown to be
skin, with 24% admitting noncompliance with therapy as significantly more effective than vehicle in decreasing
a result of these concerns [11]. signs of inflammation [20]. A systematic review compar­
In practice, clinically significant adverse effects of TCS ing TCI and TCS has shown similar rates of improvement
are rarely encountered. Excessive application of the more of AD and treatment success [21]. TCI were associated
potent TCS may lead to atrophy, telangiectases, purpura, with higher costs and more adverse events, such as burn­
striae, focal hypertrichosis and acneiform eruptions. ing and pruritus [21]. A meta‐analysis of 25 RCTs found
Cutaneous atrophy is most likely to occur with extended tacrolimus 0.1% to be as effective as the mid‐potency TCS
application to thin skin, such as the flexures, and use of hydrocortisone butyrate 0.1%, while tacrolimus 0.03% is
higher‐potency steroids, particularly under occlusion. If a less effective than hydrocortisone butyrate 0.1% but
potent or super‐potent steroid is used, sites of treatment more effective than the low‐potency TCS hydrocortisone
should be closely monitored and the skin regularly acetate 1% [22].
reviewed for signs of atrophy. Because young children The most frequently observed adverse effect is transient
have a higher surface‐to‐weight ratio than adults, systemic burning at the application site during the first few days
absorption represents a proportionately greater risk [12]. [23], usually from about 5 minutes after application and
256 Section 3  Atopic Dermatitis and Related Disorders

lasting up to 1 hour. The intensity and duration of the family and encourage a positive approach to treatment,
sensation typically decrease within 7 days if twice‐daily and to re‐explore the role of aggravating factors.
application is continued [24]. This should be explained to
patients and parents before the start of treatment, to avoid Management of infection
premature discontinuation. A few days of treatment with Skin infections are very common in patients with AD,
TCS before switching to TCI may reduce the burning arising as a result of a disrupted skin barrier and abnor­
sensation, and an overlap period of 1 week may be advis­ malities in both innate and adaptive immune responses.
able [25]. Generalized viral infections, such as eczema Staphylococcus aureus is a frequent skin colonizer in
herpeticum [26] and widespread molluscum contagio­ patients with AD. It has an important role in disease
sum [27], have been reported in patients treated with TCI, pathogenesis as it can trigger recurrent flares by stimu­
and although a number of studies have failed to replicate lating the release of the highly pruritogenic cytokine,
these findings [15], it is advised that TCI should not be IL‐3. Staph. aureus can also directly disrupt the skin
applied in the context of cutaneous infection. barrier through production of bacterial proteases, which
ATOPIC DERMATITIS

Rare cases of malignancy (skin cancer and lymphoma) break down epidermal proteins such as filaggrin [35].
have been reported in patients treated with TCI, although Staph. aureus can be isolated from skin lesions in 76–100%
SECTION 3:

a causal relationship has not been confirmed. Long‐term of patients with AD compared to 2–25% of unaffected
studies have not shown increased risk of malignancy with individuals [36]. Clinical signs of infection include
TCI in a paediatric population [28]. However, parents and weeping, honey‐coloured crusts, worsening of AD and
patients should be informed of the concern about a theo­ poor response to treatment. Nasal and skin swabs should
retical increased risk of skin malignancy, based on data be taken from patients with recurrent infection that fails
from long‐term oral calcineurin inhibitor therapy in trans­ to respond to treatment. For patients with localized skin
plant patients, and from animal studies using exposures infection, short‐term use of topical antibiotics may be
25 to nearly 50‐fold the maximum recommended human beneficial. In patients with more extensive infection, oral
dose [29]. antibiotics are recommended.
Patients with recurrent infection may benefit from anti­
Phosphodiesterase 4 inhibitors septic baths, although the evidence is limited [37,38].
Phosphodiesterase 4 (PDE4) is involved in the regulation Eczema herpeticum is a serious complication of AD
of pro‐inflammatory cytokines and is increased in the associated with high morbidity. Affected children are often
inflammatory cells of patients with AD. Targeting PDE4 unwell and in pain, with widespread vesicles, erosions
reduces the production of these pro‐inflammatory media­ and crusted punched‐out ulcers. Secondary complications
tors in AD. Both topical and oral PDE4 inhibitors have a such as meningitis, keratoconjunctivitis and encephalitis
favourable safety profile. Crisaborole 2% ointment, a can occur. Immediate treatment with aciclovir is required,
topical PDE4, is now US Food and Drug Administration‐ which must be given intravenously in severe cases.
approved for children older than 2 years for the treatment Eczema coxsackium is a newer entity caused by
of AD. Crisaborole 2% ointment shows early and sus­ ­coxsackievirus A6. In children with AD, lesions tend to
tained improvement in disease severity and pruritus and concentrate in areas previously or currently affected by
other AD symptoms, with burning and/or stinging upon the AD, similar to eczema herpeticum [39]. Treatment of
application as the only related adverse event. Other PDE4 eczema coxsackium is supportive and affected children
inhibitors are currently in trials with promising efficacy recover fully without permanent scarring.
and safety [30,31].
Contact allergy
Janus kinase (JAK) inhibitors Contact allergy should be suspected when compliance
Janus kinases (JAKs) are required for several inflamma­ has been good, no infection is evident and the AD is
tory cytokine signalling pathways and have been impli­ failing to respond. The most common allergens are nickel,
cated in the pathogenesis of AD [32]. Although the study cobalt, thimerosal and fragrance [40]. A retrospective
of JAK inhibitors for AD is still in its early stage, evidence study of 2614 children showed similar rates of positive
has identified topical JAK inhibitors as a potential new patch test reactions in children with and without AD, but
treatment for AD [33]. Large safety and efficacy trials are children with AD were more likely to have positive patch
needed before widespread use of JAK inhibitors can be test reactions to potassium dichromate, Compositae mix
advocated for AD [34]. and disperse blue [41]. Patients with isolated hand and/
or foot eczema should always have patch testing.
Unresponsive disease Phototherapy
Failure to respond to initial treatment should not auto­ Narrowband UVB is the preferred form of phototherapy
matically lead to provision of more potent TCS. The most for AD in children. The mechanism of action in AD is still
common causes of failure to respond to treatment are not fully understood, although narrowband UVB is known
inadequate application and persistence of aggravating to induce T‐cell apoptosis and anti‐inflammatory and
factors. Clinicians should enquire about the quantities of immunosuppressive cytokines [42] and has been shown
emollients and TCS used between visits. The follow‐up to reduce levels of cutaneous Staph. aureus and suppress
appointment is an excellent opportunity to educate the production of superantigen in children with AD [43].
Chapter 19  Management of Atopic Dermatitis 257

Older children and adolescents with severe AD gener­ Systemic therapy


ally tolerate narrowband UVB phototherapy well, but it
is time consuming and the frequency of hospital visits Systemic immunomodulating medications are indicated
for treatment can be disruptive of schooling. Adverse for the management of severe AD that has failed to
effects may include transient erythema, blistering, herpes respond to appropriate topical treatment and attention to
simplex reactivation and anxiety [44]. aggravating factors (Table 19.1).
Data on the efficacy of narrowband UVB in paediatric
patients with AD are limited. In a study of 77 patients Systemic corticosteroids
aged from 4 to 16 years with psoriasis or AD treated Although systemic corticosteroids can offer short‐term
with narrowband UVB phototherapy, 17 of the 25 relief of symptoms, severe rebound of AD occurs when
patients with AD had minimal residual disease after a attempts are made to withdraw treatment. Long‐term
median of 24 treatments [44]. A retrospective study of treatment with systemic corticosteroids is not recom­
paediatric patients with severe AD who completed mended for AD, owing to the likelihood of significant

ATOPIC DERMATITIS
more than 10 narrowband UVB treatments showed adverse effects including weight gain, adrenal suppres­
complete clearance or good improvement in 30 of the sion, decreased linear growth, hypertension, glucose

SECTION 3:
50 patients [45]. A cohort study comparing patients aged intolerance and decreased bone density [48]. Occasionally
3–16 years who received narrowband UVB phototherapy systemic steroids may be considered for relatively short‐
with unexposed children reported a 61% reduction in term use while another slower‐acting systemic treatment
mean six‐area, six‐sign atopic dermatitis (SASSAD) is being initiated.
score in the phototherapy cohort at 12 weeks of treat­
ment compared with an increase of 6% in the unexposed Methotrexate
cohort [46]. Methotrexate inhibits DNA synthesis by substrate compe­
There are no studies evaluating the risk of cancer in tition with dihydrofolate reductase. It is also thought to
children with AD treated with phototherapy. A systematic negatively affect T‐cell function. Gastrointestinal distur­
review of four studies assessing the risk of skin cancer bance, anorexia, fatigue, stomatitis and alopecia are the
among adults and children with psoriasis treated with most common adverse effects. Hepatotoxicity and bone
narrowband UVB did not find an increased risk of skin marrow suppression are the most serious adverse effects.
cancer [47]. An RCT of 40 patients aged 8–14 showed very similar

Table 19.1  Systemic treatments for AD

Systemic Starting dose Baseline testing Monitoring Adverse effects


agent

Methotrexate 10–15 mg/m2 weekly FBC, U&E, LFT, VZV status FBC, U&E, LFT every 1–2 weeks Hepatotoxicity, bone marrow
Consider HIV, TB, hepatitis for the first month, then suppression, GI disturbance, nausea,
screen and pregnancy test monthly for the first 3 fatigue, headache
months, then every 3 months
Ciclosporin 2.5–5 mg/kg/day Blood pressure measurement Blood pressure measurement, Nephrotoxicity, hypertension, nausea,
FBC, U&E, LFT FBC and U&E every 2 weeks paraesthesia, hypertrichosis,
VZV status for the first 1–2 months, then swollen gums, headache, rhinitis,
Consider HIV, TB, hepatitis 4–6‐weekly upper respiratory tract infection,
screen abdominal pain, folliculitis and
hyperuricaemia
Azathioprine Dependent on TPMT TPMT activity, FBC, U&E, LFT, FBC and LFT weeks 1, 3, 7, then Viral cutaneous infections, nausea,
activity VZV status every 3 months headaches, bone marrow
TMPT normal: Consider HIV, TB, hepatitis suppression, malignancy,
2–3 mg/kg/day screen and pregnancy test hepatotoxicity

TPMT reduced:
1–1.5 mg/kg/day

TPMT absent – do not


give azathioprine
Mycophenolate 40–50 mg/kg/day FBC, U&E, LFT FBC and LFT every 1–2 weeks Nausea, vomiting, diarrhoea, bone
mofetil young children VZV status for the first month then every marrow suppression, hypertension,
30–40 mg/kg/day Consider HIV, TB, hepatitis 3 months nephrotoxicity
adolescents screen and pregnancy test

FBC, full blood count; GI, gastrointestinal; HIV, human immunodeficiency virus; LFT, liver function tests; TB, tuberculosis; TPMT, thiopurine methyl‐transferase;
U&E, urea and electrolytes; VZV, varicella zoster virus.
258 Section 3  Atopic Dermatitis and Related Disorders

efficacy between methotrexate and ciclosporin, with a recurrent neutropenia (1), persistently raised alanine
mean SCORing Atopic Dermatitis (SCORAD) reduction aminotransferase (1), headaches (1), recurrent chest infec­
of 45% in both groups [49]. Time to response was margin­ tions (1), and recurrent herpes labialis (1) [55].
ally longer for patients in the methotrexate arm, but better Malignancy has been reported in patients treated with
sustained on discontinuation of treatment. Adverse azathioprine for inflammatory bowel disease, and
effects of methotrexate reported in this study included although malignancy has not been reported in children
anaemia (30%), fatigue (30%), abnormal liver function on azathioprine for AD, it is advisable to explain these
(25%), nausea and vomiting (20%) and glossitis with oral reports to parents and to limit treatment to no more than
ulceration (20%). No patient had significant liver toxicity 2 years’ duration if possible.
or other adverse effects that required stopping or adjust­ Parents and patients should be advised that azathioprine
ment in treatment. Another multicentre retrospective has a slow onset of action, with clinical improvement
study reviewing the medical records of 26 paediatric sometimes not evident until 8–12 weeks after starting.
patients with AD confirmed that methotrexate is a toler­ The dose of azathioprine required depends on the
ATOPIC DERMATITIS

able and effective agent for treating refractory childhood activity of the enzyme thiopurine methyl‐transferase
AD [50]. Multiple studies in paediatric patients with (TPMT). Patients with reduced or absent TPMT activity
SECTION 3:

psoriasis have shown methotrexate to be a safe and (10% and 0.3%) are at risk of profound immunosuppres­
well‐tolerated treatment [51]. sion. If TPMT activity is within the normal range the start­
Baseline blood tests should include full blood count ing dose should be 2–3 mg/kg/day. If TPMT activity
(FBC), urea and electrolytes (U&E), liver function tests is reduced, the starting dose should be no more than
(LFT) and varicella zoster immune status. In selected 1–1.5 mg/kg/day. In patients with absent TPMT activity,
cases tuberculosis (TB), human immunodeficiency virus azathioprine should not be administered. Children with
(HIV), hepatitis B and C serology, and pregnancy testing higher TPMT levels may respond less well to treatment
should be considered. If the child is not varicella immune, but may have a greater risk of hepatotoxicity [56]. In addi­
vaccination should be considered before starting treat­ tion to TPMT, baseline blood tests should include FBC,
ment. Dosing for the paediatric population is 10–15 mg/m2 U&E, LFT and varicella immune status. In selected cases
once a week. Dividing the dosing results in fewer gastro­ HIV, TB, hepatitis B and C serology and pregnancy testing
intestinal adverse effects [49]. Treatment with methotrexate should be considered. Monitoring of FBC and LFT should
must be supplemented with folic acid (1 mg/day). be done at weeks 1, 3, and 7 after starting treatment, and
Recommendations for monitoring vary. A commonly three 3‐monthly thereafter, if there is no breach in blood
used schedule consists of FBC, U&E and LFT every parameters [55]. Photoprotection should be advised.
1–2 weeks for the first month, then monthly for 3 months, Live vaccines are contraindicated during treatment
and then every 3 months thereafter if the dose is stable with azathioprine and should be given at least 2 weeks
and results have been normal. Amino terminal type III (preferably 4 weeks) prior to starting treatment with
procollagen peptide (P3NP) measurement is not useful as azathioprine and at least 3 months after stopping. Varicella
an indicator of liver fibrosis in children as levels are influ­ vaccine should be considered in children without
enced by linear growth rate. immunity.
Live vaccines must not be given 2 weeks (preferably
4 weeks) before starting the treatment with methotrexate Ciclosporin
and at least 3 months after stopping. Inactivated vaccines Ciclosporin (cyclosporine) is an effective medication that
can be given, however methotrexate may lower the level is useful when a rapid response is required; however,
of immunity achieved. Varicella vaccination should be patients can experience similarly rapid relapse after
considered in children without immunity before starting discontinuation.
the treatment. A multicentre study involving 27 children with AD
found 22 patients to have marked improvement or com­
Azathioprine plete clearing at 6 weeks of treatment [57]. Most of the
Azathioprine is converted into 6‐mercaptopurine in the patients relapsed within weeks of stopping treatment, but
body where it blocks purine metabolism and DNA three were in remission 6 months after ceasing. Headache
synthesis. It selectively inhibits lymphocytes, especially and abdominal pain were the most commonly reported
T lymphocytes [52]. Three retrospective studies, including adverse effects. A trial of 40 children aged 2–16 years
17, 28 and 82 patients respectively, and one prospective with severe AD comparing ciclosporin 5 mg/kg per day
study including 12 patients, showed that azathioprine is for multiple periods of 3 months or continuously for
an effective and well‐tolerated medium‐term treatment 12 months showed no difference between the two groups
for paediatric AD [52–55]. In the largest study, clinical in overall disease improvement at 1 year [58]. There is
adverse effects were seen in 16 of 82 patients (20%). Most limited evidence that a lower dose of ciclosporin for
common were cutaneous viral infections (molluscum longer periods may be associated with prolonged remis­
contagiosum and viral warts) in 12%, with single cases sion after discontinuation [59].
of nausea, lethargy, indigestion, asthma exacerbation, Adverse effects of ciclosporin include nephrotoxicity,
unconfirmed possible myopathy, headache, and recurrent hypertension, hypertrichosis, tremor, infection, headache,
chest infections [55]. In total, five patients (6%) had to gingival hyperplasia and increased risk of skin cancer
stop azathioprine because of adverse effects including and lymphoma. Nephrotoxicity and hypertension are the
Chapter 19  Management of Atopic Dermatitis 259

most concerning but no evidence of these adverse effects It inhibits downstream signalling of IL‐4 and IL‐13,
was seen at 1‐year follow‐up in a study of 40 patients [58]. cytokines of Th2 lymphocytes that are believed to play a
Long‐term safety studies in children are lacking. key role in atopic diseases. A series of four RCTs were
In paediatric patients the recommended dose is recently published in which dupilumab was evaluated in
2.5–5 mg/kg/day, divided into two doses. Treatment can 207 adult patients with moderate‐to‐severe AD [62].
be started at 2.5 mg/kg/day and gradually increased or Dupilumab treatment in adults with moderate‐to‐
at 5 mg/kg/day and tapered. FBC, U&E, LFT, varicella severe AD resulted in marked reductions in signs,
serology and blood pressure should be checked before symptoms and associated biomarker levels in two
starting. In selected cases HIV, TB, hepatitis B and C serology 4‐week monotherapy trials, one 12‐week monotherapy
should be considered. Monitoring should include blood trial and a 4‐week combination study with topical
pressure measurement, FBC, urea and creatinine every gluco­corticoids [62].
2 weeks for the first 1–2 months, then 4‐ to 6‐weekly. If Two double‐blinded, randomized phase III trials
creatinine increases more than 30% from baseline, ciclo­ (SOLO 1 and SOLO 2) enrolled adults with moderate‐to‐

ATOPIC DERMATITIS
sporin should be discontinued. severe AD whose disease was inadequately controlled by
Live vaccines are contraindicated during treatment topical treatment [63]. Patients were randomly assigned

SECTION 3:
with ciclosporin and should be given at least 2 weeks to receive, for 16 weeks, subcutaneous dupilumab
(preferably 4 weeks) before starting the treatment, including (300 mg) or placebo weekly or the same dose of dupilumab
varicella vaccine in children without immunity and at every other week alternating with placebo. A total of 1379
least 3 months after stopping the treatment. patients were enrolled in both studies. In SOLO1, 38% of
the patients who received dupilumab every other week
Mycophenolate mofetil and 37% who received dupilumab weekly reached
Mycophenolate mofetil (MMF) blocks the purine biosyn­ primary outcome (investigator’s global assessment clear
thesis pathway via the inhibition of inosine monophos­ or almost clear) as compared with 10% of patients who
phate dehydrogenase [48]. It selectively affects B cells and received placebo. The results were very similar in the
T cells, as other cells have compensatory purine scaven­ SOLO 2 study. Off‐label use of dupilumab in children has
ger mechanisms [48]. A paediatric case series involving 14 shown it to be effective and well tolerated [64]. There are
patients with AD treated with mycophenolate mofetil currently studies under way in the use of dupilumab in
showed a very good response with minimal adverse children [65].
effects [60]. A case series [53] of 12 patients with severe The significant impact on disease severity achieved
AD reported significant improvement in eight patients with dupilumab probably reflects the role of IL‐4 and
who had failed to improve with azathioprine [48]. IL‐13 in the Th2 pathway. Pruritus was not previously
Adverse effects include nausea, vomiting, diarrhoea, thought to be related to these mediators, although the
leucopenia, hypertension and nephrotoxicity. extent of improvement with dupilumab suggests a rela­
The suggested dose of MMF in children is 600–1200 mg/ tionship [66].
m2 [48] equating to 40–50 mg/kg/day in young children
and 30–40 mg/kg/day in adolescents [48]. FBC, U&E, Rituximab
LFT and varicella serology should be checked before Rituximab is a chimeric monoclonal antibody against
starting treatment. Pregnancy test, tuberculosis testing, CD20 leading to circulating B‐cell depletion and inter­
HIV, hepatitis B and C serology should be considered ference with antigen presentation. A study of six adult
before starting the treatment. FBC and LFT should be patients showed rituximab to be very effective in clear­
checked every 1–2 weeks for the first month of treatment, ing recalcitrant AD [67]. This finding was not repeated
and every 3 months thereafter. in two other patients [68]. There are no reports of pae­
Live vaccines are contraindicated during treatment diatric patients treated for AD with rituximab in the
with MMF and at least 2 weeks (preferably 4 weeks) literature.
before starting and at least 3 months after stopping the
treatment. Varicella vaccine should be considered in chil­ Omalizumab
dren without varicella immunity. Omalizumab is a humanized monoclonal antibody selec­
tively directed against circulating IgE, although the
extent to which IgE plays a role in the pathogenesis of
Biologics in atopic dermatitis
AD is unclear [69]. A randomized, double‐blind, placebo‐
There is relatively little experience with biological response controlled study of eight patients between the ages of
modifiers in the management of childhood AD, but they 4 and 22 with severe AD treated with omalizumab every
can be expected to play an increasing role [61]. 2–4 weeks for a total of 24 weeks [70] showed a signifi­
cantly greater decrease in serum IgE levels and in several
Dupilumab cytokines involved in Th2 polarization in the omalizumab
Dupilumab has recently been approved by the US Food group compared to the placebo group [70]. This was not
and Drug Administration (FDA) and licensed in the UK reflected in clinical benefit based on SCORAD. No severe
as the first biologic treatment for adults with moderate adverse events were reported. The ADAPT clinical study,
and severe eczema. Dupilumab is a human monoclonal comparing omalizumab with placebo in paediatric
antibody that binds to the alpha subunit of the IL‐4 receptor. patients with AD, will provide valuable data [71].
260 Section 3  Atopic Dermatitis and Related Disorders

Ustekinumab of EASI [81]. Two patients stopped the treatment early


Ustekinumab is a human monoclonal antibody against due to worsening of the disease.
IL‐12 and IL‐23. Although high IL‐17 and IL‐23 levels
have been observed in children with AD, their role in the Mepolizumab
AD inflammatory process remains poorly understood. Mepolizumab is a monoclonal antibody against IL‐5,
Improvement has been reported in an adolescent with which is essential for eosinophil growth, differentiation
refractory AD after 4 months of treatment with usteki­ and migration [82]. In an RCT involving 18 adults with
numab [72]. Another adolescent experienced initial moderate‐to‐severe AD [82], no significant clinical improve­
improvement, but the benefit diminished with continued ment was noted in the group receiving mepolizumab
treatment [73]. compared with placebo, despite a significant decrease of
eosinophil count in peripheral blood.
Infliximab and etanercept
Infliximab is a chimeric monoclonal tumour necrosis Allergy in atopic dermatitis
ATOPIC DERMATITIS

factor (TNF)‐α inhibitor. Nine adult patients are reported


to have experienced initial improvement of severe AD, Food allergy
SECTION 3:

but responses were not sustained through the mainte­ There is increasing evidence that sensitization to food
nance phase [74]. There are no reports to date of using antigens arises through environmental exposure early in
infliximab for AD in children. infancy through eczematous skin [83], and that early oral
Etanercept is a recombinant dimeric fusion protein of intake may help produce immune tolerance [84].
the human 75 kDa TNF‐α receptor (TNFR) with the Fc The European Academy of Allergy and Clinical Immu­
fragment of human immunoglobulin IgG1 [75]. Etanercept nology’s (EAACI) Taskforce on Prevention of Food Allergy
has been reported to be unhelpful in two children with Guidance advises all mothers to have a normal diet
recalcitrant AD [76]. without restrictions during pregnancy and lactation [85].
For all infants, exclusive breastfeeding is recommended
Tralokinumab and lebrikizumab for at least the first 4–6 months of life. If breastfeeding is
Tralokinumab and lebrikizumab are humanized IgG4 insufficient or not possible, infants at high risk can be rec­
monoclonal anti‐IL‐13 antibodies. Because IL‐13 is ommended a hypoallergenic formula. There is no evidence
responsible for many of the pathogenic Th2 effects in AD, of benefit from delaying the introduction of complemen­
tralokinumab and lebrikizumab may hold promise for tary foods beyond 4 months, or from withholding or
AD treatment [77]. Lebrikizumab has been evaluated in a encouraging exposure to potentially allergenic foods after
phase II RCT (TREBLE) in adults with moderate‐to‐severe 4 months once weaning has commenced, irrespective of
AD, showing significant improvement when combined atopic heredity. There is no evidence to support the use of
with the use of topical steroids [78]. prebiotics or probiotics for food allergy prevention [85].
For infants and children with AD who already have
symptoms suggestive of food allergy, diagnosis and
Nemolizumab
management should follow evidence‐based recommen­
Nemolizumab is IL‐31 targeting agent, currently in phase
dations [85]. In unselected patients with AD there does
II clinical trials. IL‐31 is a Th2 cytokine, also known as the
not appear to be any benefit in cow’s milk avoidance, or
‘pruritus cytokine’ that has been shown to compromise
in the use of elemental or few foods diets [86].
epidermal terminal differentiation and inhibit lipid
synthesis, perpetuating the ‘itch–scratch’ cycle of AD [35].
Airborne allergens and pollution
Many children experience exacerbation of AD on expo­
Tocilizumab sure to airborne allergens, pollens, house dust mite and
Tocilizumab is a monoclonal IL‐6 receptor antibody. animal dander being the most commonly cited. Avoidance
Treatment of three adult patients with AD with tocilizumab is problematic and of variable benefit. There is limited
resulted in improvement in Eczema Area and Severity understanding of how and when airborne allergies
Index (EASI) score by more than 50% after 3 months, com­ develop, with some evidence that very early exposure
pared to baseline. During treatment one patient developed may be relevant to prevention [87].
uncomplicated conjunctivitis and another developed Neonatal domestic dog exposure has been shown to be
streptococcal bursitis of the heel [79]. associated with a strongly reduced risk of AD in two
independent birth cohorts and in a dose‐dependent man­
Alefacept ner. While the mechanisms involved are unclear, the find­
Alefacept is a fusion protein that binds to CD2. It inhibits ings raise the question whether in utero exposures may
T‐cell activation, and selectively reduces memory T cells. affect the risk of AD [88]. Specific immunotherapy, mainly
One uncontrolled pilot study of ten adult patients with to house dust mite and pollens, has been reported to
moderate‐to‐severe AD, treated for 12 weeks, reported a improve symptoms in a small number of controlled stud­
78% reduction in AD severity [80]. In a study of nine adult ies, but the evidence is limited [89].
patients, two had at least a 50% reduction of EASI score There is limited evidence that traffic‐related air pollu­
after 18 weeks of treatment, four had a less than 50% tion is associated with AD. Better long‐term studies are
reduction of baseline EASI score, and one had worsening needed [90].
Chapter 19  Management of Atopic Dermatitis 261

Management of pruritus A recent systematic review [101] of 37 RCTs did not find
Pruritus is an integral part of AD and it is the most intense a significant difference in risk of developing AD between
and distressing symptom, to the extent that AD has been infants fed hydrolysed formula and those fed standard
called the ‘itch that rashes’. Scratching can worsen AD, as cow’s milk formula, leading to the need to revise previous
it causes breaching of the skin, increasing the risk of infec­ guidelines recommending the use of hydrolysed milk
tion. As patients become more pruritic, they scratch more, formula to prevent allergic disease in high‐risk patients
which then worsens their dermatitis and leads to more who cannot be exclusively breastfed [102,103].
itch [91]. Nocturnal itching and scratching are very com­
mon, frequently leading to sleep disturbance. AD‐related Psychological impact of atopic
pruritus is the most common cause of chronic sleep loss in dermatitis
young people [92]. The stress and sleep disturbance
derived from the itch–scratch cycle profoundly disrupt Management of AD should include attention to the
the well‐being of children and the overall quality of their impact of the disease on education, social life and emo­

ATOPIC DERMATITIS
adolescent development [93]. The whole family can be tional well‐being of the child and family.
adversely affected by the child’s scratching. Children with AD rate itching and sleep loss as the most

SECTION 3:
The only really effective way to control pruritus is to important aspects of their disease [104]. Sleep disturbance
adequately treat the AD that is causing the pruritus. In the due to the itching leads to tiredness, mood changes and
short term pruritus may be relieved temporarily by the impaired psychosocial functioning of the child and the par­
application of emollients and wet dressings. Although his­ ents. Pharmacological treatments of pruritus, usually in the
tamine has been recognized as an important mediator of form of sedating antihistamines, are of limited benefit and
pruritus in certain conditions, it does not seem to have a may leave the child tired and unable to concentrate.
major role in AD, accounting for the limited efficacy of The ability to play or do sports, particularly swimming,
oral antihistamines in controlling the pruritus of AD. may be limited because of embarrassment, discomfort and
Sedating antihistamines may have a role as a result of their exacerbation of disease [104]. Embarrassment, unkind
soporific effect [94]. Nonsedating antihistamines such as comments, teasing and bullying frequently cause social
cetirizine or loratadine may occasionally be useful when isolation and may lead to depression or school avoidance
there is a concurrent urticarial element, food allergic reac­ [105]. In older age groups, when physical appearance
tions or allergic rhinoconjunctivitis. Distraction techniques becomes increasingly important, chronically inflamed skin
focusing on manual or mental activities such as games, is seen as unattractive and can cause anxiety and depres­
storytelling and music may be helpful in some cases. sion. Rates of psychological disturbance have been shown
The severity of pruritus can be limited by avoidance of to be twice as high in children with moderate‐to‐severe AD
aggravating factors such as overheating and sweating as in children without AD [106]. Rapid improvement in
[95]. At night, bedroom temperatures should be kept AD has been observed following management of the dys­
below 20 °C. Bedding and night clothes should be light functional parent–child relationships that have been found
and made of cotton. Contact with wool should be avoided. to occur in many cases of intractable childhood AD [107].
Cognitive behavioural therapy, as an adjunct to phar­
macological treatment, is effective in reducing disease
Prevention of atopic dermatitis severity and psychological sequelae in patients with
Skin barrier enhancement AD [108]. Interventions such as multidisciplinary group
There is evidence from two RCTs that daily emollient patient education and relaxation methods have been
application could reduce the incidence of AD in infants shown to lead to improvements in AD severity and
with a family history of atopy, with relative risk reduc­ quality of life for both children and families [109].
tion of 50% [96] and 26% [97]. Whether eczema can
be  prevented by daily application of emollient for
Conclusion
12 months in newborns with a family history of AD is
being assessed in The Barrier Enhancement for Eczema Although there have been advances in the understanding
Prevention (BEEP) study, currently running (www. of aetiopathogenesis, AD, particularly if severe, remains a
isrctn.com/ISRCTN21528841). condition that can be very difficult to manage.
To improve management we require better understand­
Probiotics and nutritional intervention ing of many issues, including whether restoration of
A systematic review of 21 studies showed that supple­ barrier function and/or increase in filaggrin expression
mentation with probiotics in infants and pregnant moth­ early in the course of AD can prevent disease progression
ers can prevent development and reduce severity of AD and the atopic march, the role of genetic factors other than
[98]. A meta‐analysis of 16 randomized trials including filaggrin, and whether specific genetic factors could be
approximately 3500 participants found that probiotics used as a guide towards particular treatments [110].
given in the prenatal and postnatal period reduced the Currently success depends on detailed assessment of
risk of AD in the first years of life in both children at high the clinical features, an understanding of the percep­
risk of AD and in those from the general population [99]. tions and needs of the child and the family, thorough
However, subsequent RCTs have not provided evidence explanation of the rationale for, and risks and benefits of,
that probiotics either prevent AD or reduce its severity [100]. each intervention, and ongoing support.
262 Section 3  Atopic Dermatitis and Related Disorders

References 25 Wollenberg A, Oranje A, Deleuran M et al. ETFAD/EADV Eczema


1 Ersser SJ, Latter S, Sibley A et al. Psychological and educational task force 2015 position paper on diagnosis and treatment of atopic
interventions for atopic eczema in children. Cochrane Database dermatitis in adult and paediatric patients. J Eur Acad Dermatol
Syst Rev 2007(3):CD004054. Venereol 2016;30:729–47.
2 Boguniewicz M, Leung DY. Atopic dermatitis: a disease of altered 26 Lubbe J, Pournaras CC, Saurat JH. Eczema herpeticum during treat­
skin barrier and immune dysregulation. Immunol Rev 2011;242: ment of atopic dermatitis with 0.1% tacrolimus ointment. Dermatology
233–46. 2000;201:249–51.
3 Brown SJ, McLean WH. One remarkable molecule: filaggrin. J Invest 27 Wetzel S, Wollenberg A. Eczema molluscatum in tacrolimus treated
Dermatol 2012;132:751–62. atopic dermatitis. Eur J Dermatol 2004;14:73–4.
4 Grimalt R, Mengeaud V, Cambazard F; Study Investigators’ Group. 28 Koo JY, Fleischer AB Jr, Abramovits Wet  al. Tacrolimus ointment is
The steroid‐sparing effect of an emollient therapy in infants with safe and effective in the treatment of atopic dermatitis: results in 8000
atopic dermatitis: a randomized controlled study. Dermatology patients. J Am Acad Dermatol 2005;53(2 Suppl 2):S195–205.
2007;214:61–7. 29 Tennis P, Gelfand JM, Rothman KJ. Evaluation of cancer risk related to
5 Tiplica GS, Boralevi F, Konno P et al. The regular use of an emollient atopic dermatitis and use of topical calcineurin inhibitors. Br J
improves symptoms of atopic dermatitis in children: a randomized Dermatol 2011;165:465–73.
controlled study. J Eur Acad Dermatol Venereol 2018;32:1180–7. 30 Eichenfield LF, Call RS, Forsha DW et al. Long‐term safety of crisab­
ATOPIC DERMATITIS

6 Ishmael FT, Stellato C. Post‐transcriptional regulation of glucocorti­ orole ointment 2% in children and adults with mild to moderate
coid function. In: Menon K, Goldstrohm A (eds) Post‐transcriptional atopic dermatitis. J Am Acad Dermatol 2017;77:641–9.
Mechanisms in Endocrine Regulation. Springer, 2015:277–313. 31 Zebda R, Paller AS. Phosphodiesterase 4 inhibitors. J Am Acad
SECTION 3:

7 Feldmann RJ, Maibach HI. Regional variation in percutaneous Dermatol 2018;78(3 suppl 1):S43–52.
penetration of 14C cortisol in man. J Invest Dermatol 1967;48:181–3. 32 Tanimoto A, Shinozaki Y, Yamamoto Y et  al. A novel JAK inhibitor
8 Feiwel M, James VH, Barnett ES. Effect of potent topical steroids on JTE‐052 reduces skin inflammation and ameliorates chronic dermati­
plasma‐cortisol levels of infants and children with eczema. Lancet. tis in rodent models: Comparison with conventional therapeutic
1969;1:485–7. agents. Exp Dermatol 2018;27:22–9.
9 Williams HC. Established corticosteroid creams should be applied 33 Bissonnette R, Papp KA, Poulin Y et al. Topical tofacitinib for atopic
only once daily in patients with atopic eczema. BMJ 2007;334:1272. dermatitis: a phase IIa randomized trial. Br J Dermatol 2016;175:
10 Long CC, Finlay AY. The finger‐tip unit—a new practical measure. 902–11.
Clin Exp Dermatol 1991;16:444–7. 34 Cotter DG, Schairer D, Eichenfield L. Emerging therapies for atopic der­
11 Charman CR, Morris AD, Williams HC. Topical corticosteroid phobia matitis: JAK inhibitors. J Am Acad Dermatol 2018;78(3 suppl 1):S53–62.
in patients with atopic eczema. Br J Dermatol 2000;142:931–6. 35 Sonkoly E, Muller A, Lauerma AI et al. IL‐31: a new link between
12 Hengge UR, Ruzicka T, Schwartz RA, Cork MJ. Adverse effects of T  cells and pruritus in atopic skin inflammation. J Allergy Clin
topical glucocorticosteroids. J Am Acad Dermatol 2006;54:1–15; Immunol 2006;117:411–17.
quiz 6–8. 36 Huang JT, Abrams M, Tlougan B et al. Treatment of Staphylococcus
13 Devillers AC, Oranje AP. Efficacy and safety of ’wet‐wrap’ dressings aureus colonization in atopic dermatitis decreases disease severity.
as an intervention treatment in children with severe and/or refractory Pediatrics 2009;123:e808–14.
atopic dermatitis: a critical review of the literature. Br J Dermatol 37 Lee M, Van Bever H. The role of antiseptic agents in atopic dermatitis.
2006;154:579–85. Asia Pac Allergy 2014;4:230–40.
14 Thomas K, Bradshaw LE, Batchelor JM et al. Preliminary results of 38 Bath‐Hextall FJ, Birnie AJ, Ravenscroft JC, Williams HC. Interventions
a randomized controlled trial of silk therapeutic clothing for the to reduce Staphylococcus aureus in the management of atopic eczema:
management of eczema in children (CLOTHES Trial). Br J Dermatol an updated Cochrane review. Br J Dermatol 2011;164:228.
2016;175:11–12. 39 Mathes EF, Oza V, Frieden IJ et al. “Eczema coxsackium” and unusual
15 Bornhovd E, Burgdorf WH, Wollenberg A. Macrolactam immu­ cutaneous findings in an enterovirus outbreak. Pediatrics 2013;132:
nomodulators for topical treatment of inflammatory skin diseases. e149–57.
J Am Acad Dermatol 2001;45:736–43. 40 Simonsen AB, Deleuran M, Johansen JD, Sommerlund M. Contact
16 Murrell DF, Calvieri S, Ortonne JP et al. A randomized controlled trial allergy and allergic contact dermatitis in children  –  a review of
of pimecrolimus cream 1% in adolescents and adults with head and current data. Contact Dermatitis 2011;65:254–65.
neck atopic dermatitis and intolerant of, or dependent on, topical 41 Belloni Fortina A, Fontana E, Peserico A. Contact sensitization in
corticosteroids. Br J Dermatol2007;157:954–9. children: a retrospective study of 2,614 children from a single center.
17 Draelos ZD. Use of topical corticosteroids and topical calcineurin Pediatr Dermatol 2016;33:399–404.
inhibitors for the treatment of atopic dermatitis in thin and sensitive 42 Ibbotson SH, Bilsland D, Cox NH et al. An update and guidance on
skin areas. Curr Med Res Opin 2008;24:985–94. narrowband ultraviolet B phototherapy: a British Photodermatology
18 Lubbe J, Friedlander SF, Cribier B et al. Safety, efficacy, and dosage of Group Workshop Report. Br J Dermatol 2004;151:283–97.
1% pimecrolimus cream for the treatment of atopic dermatitis in daily 43 Silva SH, Guedes AC, Gontijo B et al. Influence of narrow‐band UVB
practice. Am J Clin Dermatol 2006;7:121–31. phototherapy on cutaneous microbiota of children with atopic
19 Eichenfield LF, Tom WL, Berger TG et al. Guidelines of care for the dermatitis. J Eur Acad Dermatol Venereol 2006;20:1114–20.
management of atopic dermatitis: section 2. Management and treat­ 44 Jury CS, McHenry P, Burden AD et  al. Narrowband ultraviolet B
ment of atopic dermatitis with topical therapies. J Am Acad Dermatol (UVB) phototherapy in children. Clin Exp Dermatol 2006;31:196–9.
2014;71:116–32. 45 Clayton TH, Clark SM, Turner D, Goulden V. The treatment of severe
20 Reitamo S, Harper J, Bos JD et al. 0.03% Tacrolimus ointment applied atopic dermatitis in childhood with narrowband ultraviolet B photo­
once or twice daily is more efficacious than 1% hydrocortisone acetate therapy. Clin Exp Dermatol 2007;32:28–33.
in children with moderate to severe atopic dermatitis: results of a ran­ 46 Darne S, Leech SN, Taylor AE. Narrowband ultraviolet B photother­
domized double‐blind controlled trial. Br J Dermatol 2004;150:554–62. apy in children with moderate‐to‐severe eczema: a comparative
21 Broeders JA, Ahmed Ali U, Fischer G. Systematic review and cohort study. Br J Dermatol 2014;170:150–6.
meta‐analysis of randomized clinical trials (RCTs) comparing topical 47 Hearn RM, Kerr AC, Rahim KF et al. Incidence of skin cancers in 3867
calcineurin inhibitors with topical corticosteroids for atopic derma- patients treated with narrow‐band ultraviolet B phototherapy. Br J
titis: A 15‐year experience. J Am Acad Dermatol 2016;75:410–9.e3. Dermatol 2008;159:931–5.
22 Ashcroft DM, Dimmock P, Garside R et al. Efficacy and tolerability 48 Sidbury R, Davis DM, Cohen DEet  al. Guidelines of care for the
of topical pimecrolimus and tacrolimus in the treatment of atopic management of atopic dermatitis: section 3. Management and treat-
dermatitis: meta‐analysis of randomised controlled trials. BMJ ment with phototherapy and systemic agents. J Am Acad Dermatol
2005;330:516. 2014;71:327–49.
23 Ruzicka T, Bieber T, Schopf E et  al. A short‐term trial of tacrolimus 49 El‐Khalawany MA, Hassan H, Shaaban D et al. Methotrexate v ­ ersus
ointment for atopic dermatitis. European Tacrolimus Multicenter cyclosporine in the treatment of severe atopic dermatitis in children: a
Atopic Dermatitis Study Group. N Engl J Med 1997;337:816–21. multicenter experience from Egypt. Eur J Pediatr 2013;172:351–6.
24 Bornhovd EC, Burgdorf WH, Wollenberg A. Immunomodulatory 50 Taieb Y, Baum S, Ben Amitai D et  al. The use of methotrexate for
macrolactams for topical treatment of inflammatory skin diseases. treating childhood atopic dermatitis: a multicenter retrospective
Curr Opin Investig Drugs 2002;3:708–12. study. J Dermatolog Treat 2018:1–5.
Chapter 19  Management of Atopic Dermatitis 263

51 Dadlani C, Orlow SJ. Treatment of children and adolescents with meth­ 77 Kagami S, Saeki H, Komine M et al. Interleukin‐4 and interleukin‐13
otrexate, cyclosporine, and etanercept: review of the dermatologic and enhance CCL26 production in a human keratinocyte cell line, HaCaT
rheumatologic literature. J Am Acad Dermatol 2005;52:316–40. cells. Clin Exp Immunol 2005;141:459–66.
52 Caufield M, Tom WL. Oral azathioprine for recalcitrant pediatric 78 Simpson EL, Flohr C, Eichenfield LF et  al. Efficacy and safety of
atopic dermatitis: clinical response and thiopurine monitoring. J Am lebrikizumab (an anti‐IL‐13 monoclonal antibody) in adults with
Acad Dermatol 2013;68(1):29–35. moderate‐to‐severe atopic dermatitis inadequately controlled by
53 Waxweiler WT, Agans R, Morrell DS. Systemic treatment of pediatric topical corticosteroids: A randomized, placebo‐controlled phase II
atopic dermatitis with azathioprine and mycophenolate mofetil. trial (TREBLE). J Am Acad Dermatol 2018;78:863–71.
Pediatr Dermatol 2011;28:689–94. 79 Navarini AA, French LE, Hofbauer GF. Interrupting IL‐6‐receptor
54 Hon KL, Ching GK, Leung TF et al. Efficacy and tolerability at 3 and signaling improves atopic dermatitis but associates with bacterial
6 months following use of azathioprine for recalcitrant atopic dermatitis superinfection. J Allergy Clin Immunol 2011;128:1128–30.
in children and young adults. J Dermatolog Treat 2009;20:141–5. 80 Simon D, Wittwer J, Kostylina G et al. Alefacept (lymphocyte func­
55 Fuggle NR, Bragoli W, Mahto A et al. The adverse effect profile of tion‐associated molecule 3/IgG fusion protein) treatment for atopic
oral azathioprine in pediatric atopic dermatitis, and recommendations eczema. J Allergy Clin Immunol 2008;122:423–4.
for monitoring. J Am Acad Dermatol 2015;72:108–14. 81 Moul DK, Routhouska SB, Robinson MR, Korman NJ. Alefacept for
56 Murphy LA, Atherton D. A retrospective evaluation of azathioprine moderate to severe atopic dermatitis: a pilot study in adults. J Am

ATOPIC DERMATITIS
in severe childhood atopic eczema, using thiopurine methyltrans­ Acad Dermatol 2008;58:984–9.
ferase levels to exclude patients at high risk of myelosuppression. Br J 82 Oldhoff JM, Darsow U, Werfel T et  al. Anti‐IL‐5 recombinant
Dermatol 2002;147:308–15. humanized monoclonal antibody (mepolizumab) for the treatment

SECTION 3:
57 Berth‐Jones J, Finlay AY, Zaki I et al. Cyclosporine in severe childhood of atopic dermatitis. Allergy 2005;60:693–6.
atopic dermatitis: a multicenter study. J Am Acad Dermatol 1996; 83 Du Toit G, Katz Y, Sasieni P et al. Early consumption of peanuts
34:1016–21. in infancy is associated with a low prevalence of peanut allergy.
58 Harper JI, Ahmed I, Barclay G et al. Cyclosporin for severe childhood J Allergy Clin Immunol 2008;122:984–91.
atopic dermatitis: short course versus continuous therapy. Br J 84 Du Toit G, Roberts G, Sayre PH et al. Randomized trial of peanut
Dermatol 2000;142:52–8. consumption in infants at risk for peanut allergy. N Engl J Med
59 Sibbald C, Pope E, Ho N, Weinstein M. Retrospective review of 2015;372:803–13.
relapse after systemic cyclosporine in children with atopic dermatitis. 85 Muraro A, Halken S, Arshad SH et  al. EAACI food allergy and
Pediatr Dermatol 2015;32:36–40. anaphylaxis guidelines. Primary prevention of food allergy.
60 Heller M, Shin HT, Orlow SJ, Schaffer JV. Mycophenolate mofetil for Allergy 2014;69:590–601.
severe childhood atopic dermatitis: experience in 14 patients. Br J 86 Bath‐Hextall F, Delamere FM, Williams HC. Dietary exclusions for
Dermatol 2007;157:127–32. improving established atopic eczema in adults and children: system­
61 Fabbrocini G, Napolitano M, Megna M et al. Treatment of atopic der­ atic review. Allergy 2009;64:258–64.
matitis with biologic drugs. Dermatol Ther (Heidelb) 2018;8:527–38. 87 Harris JM, Williams HC, White C et al. Early allergen exposure and
62 Beck LA, Thaci D, Hamilton JD et al. Dupilumab treatment in adults atopic eczema. Br J Dermatol 2007;156:698–704.
with moderate‐to‐severe atopic dermatitis. N Engl J Med 2014;371:130–9. 88 Thorsteinsdottir S, Thyssen JP, Stokholm J et al. Domestic dog expo­
63 Simpson EL, Bieber T, Guttman‐Yassky E et al. Two Phase 3 Trials of sure at birth reduces the incidence of atopic dermatitis. Allergy
Dupilumab versus Placebo in Atopic Dermatitis. N Engl J Med 2016;71:1736–44.
2016;375:2335–48. 89 Tam HH, Calderon MA, Manikam L et al. Specific allergen immuno­
64 Treister AD, Lio PA. Long‐term off‐label dupilumab in pediatric therapy for the treatment of atopic eczema: a systematic review.
atopic dermatitis: A case series. Pediatr Dermatol 2019;36(1):85–8. Allergy 2016;71:1345–56.
65 National Institute for Health Research. NIHR Innovation Observatory 90 Bowatte G, Lodge C, Lowe AJ et  al. The influence of childhood
Evidence Briefing: May 2018: http://www.io.nihr.ac.uk/wp‐content/ traffic‐related air pollution exposure on asthma, allergy and sensi­
uploads/2018/06/20586‐Dupilumab‐for‐12‐17‐years‐old‐children‐ tization: a systematic review and a meta‐analysis of birth cohort
with‐atopic‐dermatitis‐V1.0‐MAY‐2018‐NON‐CONF.pdf [accessed 13 studies. Allergy 2015;70:245–56.
January 2019]. 91 Homey B, Steinhoff M, Ruzicka T, Leung DY. Cytokines and
66 Notaro ER, Sidbury R. Systemic agents for severe atopic dermatitis in chemokines orchestrate atopic skin inflammation. J Allergy Clin
children. Paediatr Drugs 2015;17:449–57. Immunol 2006;118:178–89.
67 Simon D, Hosli S, Kostylina G et al. Anti‐CD20 (rituximab) treatment 92 Torrelo A, Ortiz J, Alomar A et al. Atopic dermatitis: impact on qual­
improves atopic eczema. J Allergy Clin Immunol 2008;121:122–8. ity of life and patients’ attitudes toward its management. Eur J
68 Sediva A, Kayserova J, Vernerova E et al. Anti‐CD20 (rituximab) treat­ Dermatol 2012;22:97–105.
ment for atopic eczema. J Allergy Clin Immunol 2008;121:1515–16; 93 Blume‐Peytavi U, Metz M. Atopic dermatitis in children: management
author reply 6–7. of pruritus. J Eur Acad Dermatol Venereol 2012;26(Suppl 6):2–8.
69 Heil PM, Maurer D, Klein B et al. Omalizumab therapy in atopic der­ 94 Eichenfield LF, Hanifin JM, Luger TA et al. Consensus conference on
matitis: depletion of IgE does not improve the clinical course – a ran­ pediatric atopic dermatitis. J Am Acad Dermatol 2003;49:1088–95.
domized, placebo‐controlled and double blind pilot study. J Dtsch 95 Hanifin JM. Basic and clinical aspects of atopic dermatitis. Ann
Dermatol Ges 2010;8:990–8. Allergy 1984;52:386–95.
70 Iyengar SR, Hoyte EG, Loza A et  al. Immunologic effects of omali­ 96 Simpson EL, Chalmers JR, Hanifin JM et al. Emollient enhancement
zumab in children with severe refractory atopic dermatitis: a rand­ of the skin barrier from birth offers effective atopic dermatitis pre­
omized, placebo‐controlled clinical trial. Int Arch Allergy Immunol vention. J Allergy Clin Immunol 2014;134:818–23.
2013;162:89–93. 97 Horimukai K, Morita K, Narita M et al. Application of moisturizer to
71 Chan S, Cornelius V, Cro S et al. A randomised controlled clinical trial neonates prevents development of atopic dermatitis. J Allergy Clin
of omalizumab for severe pediatric atopic dermatitis. JAMA Pediatr Immunol 2014;134:824–30.e6.
2019 (in press). 98 Foolad N, Brezinski EA, Chase EP, Armstrong AW. Effect of nutrient
72 Agusti‐Mejias A, Messeguer F, Garcia R, Febrer I. Severe refractory supplementation on atopic dermatitis in children: a systematic
atopic dermatitis in an adolescent patient successfully treated with review of probiotics, prebiotics, formula, and fatty acids. JAMA
ustekinumab. Ann Dermatol 2013;25:368–70. Dermatol 2013;149:350–5.
73 Wlodek C, Hewitt H, Kennedy CT. Use of ustekinumab for severe 99 Panduru M, Panduru NM, Salavastru CM, Tiplica GS. Probiotics
refractory atopic dermatitis in a young teenager. Clin Exp Dermatol and primary prevention of atopic dermatitis: a meta‐analysis of
2016;41:625–7. randomized controlled studies. J Eur Acad Dermatol Venereol
74 Jacobi A, Antoni C, Manger B et  al. Infliximab in the treatment of 2015;29:232–42.
moderate to severe atopic dermatitis. J Am Acad Dermatol 2005; 100 Allen SJ, Jordan S, Storey M et  al. Probiotics in the prevention of
52:522–6. eczema: a randomised controlled trial. Arch Dis Child 2014;99:
75 Goffe B, Cather JC. Etanercept: An overview. J Am Acad Dermatol 1014–19.
2003;49(2 Suppl):S105–11. 101 Boyle RJ, Ierodiakonou D, Khan T et al. Hydrolysed formula and
76 Buka RL, Resh B, Roberts B et al. Etanercept is minimally effective in 2 risk of allergic or autoimmune disease: systematic review and
children with atopic dermatitis. J Am Acad Dermatol 2005;53(2):358–9. meta‐analysis. BMJ 2016;352:i974.
264 Section 3  Atopic Dermatitis and Related Disorders

102 Fleischer DM, Spergel JM, Assa’ad AH, Pongracic JA. Primary 107 Koblenzer CS, Koblenzer PJ. Chronic intractable atopic eczema. Its
prevention of allergic disease through nutritional interventions. occurrence as a physical sign of impaired parent‐child relationships
J Allergy Clin Immunol Pract 2013;1:29–36. and psychologic developmental arrest: improvement through par­
103 Thygarajan A, Burks AW. American Academy of Pediatrics recom­ ent insight and education. Arch Dermatol 1988;124:1673–7.
mendations on the effects of early nutritional interventions on the 108 Ehlers A, Stangier U, Gieler U. Treatment of atopic dermatitis: a com­
development of atopic disease. Curr Opin Pediatr 2008;20:698–702. parison of psychological and dermatological approaches to relapse
104 Beattie PE, Lewis‐Jones MS. A comparative study of impairment of prevention. J Consult Clin Psychol 1995;63:624–35.
quality of life in children with skin disease and children with other 109 Ersser SJ, Cowdell F, Latter S et  al. Psychological and educational
chronic childhood diseases. Br J Dermatol 2006;155:145–51. interventions for atopic eczema in children. Cochrane Database Syst
105 Lewis‐Jones S. Quality of life and childhood atopic dermatitis: the mis­ Rev 2014;1:CD004054.
ery of living with childhood eczema. Int J Clin Pract 2006;60:984–92. 110 Healy E, Brown SJ, Langan SM et al. Identification of translational
106 Absolon CM, Cottrell D, Eldridge SM, Glover MT. Psychological dermatology research priorities in the U.K.: results of an electronic
disturbance in atopic eczema: the extent of the problem in school‐ Delphi exercise. Br J Dermatol 2015;173:1191–8.
aged children. Br J Dermatol 1997;137:241–5.
ATOPIC DERMATITIS
SECTION 3:
 265

C HA PTER   20

Napkin Dermatitis
Arnold P. Oranje1, Ernesto Bonifazi2, Paul J. Honig3,4 & Albert C. Yan4,5,6
Kinderhuid.nl, Rotterdam, Hair Clinic, Breda and Dermicis Skin Clinic, Alkmaar, The Netherlands
1 

Dermatologia Pediatrica Association Bari, Italy


2 

Division of Dermatology, Denver Children’s Hospital, Denver, CO, USA


3 

Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
4 

Section of Dermatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA


5 

Department of Dermatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
6 

Introduction, 265 Granuloma gluteale infantum, 271 Neoplastic napkin conditions, 276
Aetiology, 266 Infectious napkin dermatitis, 271 Metabolic napkin conditions, 277
Clinical and differential diagnosis, 268 Primary and secondary inflammatory Other important napkin conditions, 278

SECTION 4: OTHER TYPES


Contact napkin dermatitis, 269 conditions, 274
Simple intertrigo, 270

OF DERMATITIS
Abstract ­ apkin dermatitis. Allergic forms of contact dermatitis in the
n
napkin area may be difficult to distinguish from irritant napkin
dermatitis, although clinically allergic contact dermatitis is often
Napkin dermatitis refers to a collection of common disorders
pruritic rather than sore, and unusual patterns corresponding
­affecting the napkin area. Napkin dermatitis includes a range of
to specific components of the napkin may indicate an aetiology.
conditions including irritant dermatitis, Candida napkin dermati-
Napkin psoriasis may be the presenting sign of psoriasis during
tis, erosive napkin dermatitis and allergic contact dermatitis. Nap-
infancy, and in older children is often the initial manifestation of
kin area involvement may also be the presenting sign for napkin
Kawasaki disease.
psoriasis or Kawasaki disease, and may rarely be an indicator of
This chapter focuses on a description of the clinical features,
child abuse.
­aetiology and treatment of napkin dermatitis.
Primary irritant napkin or diaper dermatitis may over time
evolve into secondary Candida napkin dermatitis or erosive

Key points • Treatment of napkin dermatitis if persisting longer than 1


week should include anticandidal and antibacterial creams or
ointments.
• Napkin dermatitis is only present in the napkin area when a child
• Napkin psoriasis is sometimes the initial presenting sign of
or adult uses napkins.
psoriasis.
• Candida albicans as secondary invader plays a moderate role.
• Napkin dermatitis and napkin psoriasis in an ill infant may be an
• Primary candidiasis is relatively rare, except in the early neonatal
early characteristic feature of Kawasaki disease.
period.
• Unexplained severe napkin dermatitis may be a part of child abuse.

Introduction and urine as a secondary factor. Most cases of napkin der-


matitis are mild and it is estimated that only 5% of cases
Definition. Primary irritant napkin dermatitis (nappy are severe [2].
rash, diaper dermatitis) is a dermatitis typically localized,
at least initially, to the area covered by napkins. It is only History. At the beginning of the twentieth century,
observed when napkins are used. It is also observed in i­nvestigators assumed that ammonia released from the
adults using large napkins because of urinary and faecal urine was the primary irritant in napkin dermatitis [2,3].
incontinence. In 1921, Cooke claimed that Bacterium ammoniagenes was
responsible for napkin dermatitis, resulting in the term
Aetiology and  clinical presentation. The aetiology of ammoniacal dermatitis [4]. This theory was widely
napkin dermatitis involves multiple factors [1] (Fig. 20.1). accepted for decades. Subsequent studies showed, how-
Irritant napkin dermatitis can be considered as a toxic ever, the same incidence of urea‐splitting bacteria in the
contact dermatitis that occurs in the napkin area because napkins of babies with and without rash. In 1977, Leyden
of prolonged contact with faeces as the primary factor et  al. demonstrated that ammonia did not cause skin

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
266 Section 4  Other Types of Dermatitis

Intact skin forms are now infrequently observed. Variants of napkin


dermatitis characterized by severe erythema, large ­pustules
Activating factors Caregiver intervention
and ulcers have become rare.
• skin hydration • increased napkin
• mixing faeces change Napkins or diapers
and urine • medical treatment The role played by napkins is supported by all causative
Induction
considerations. The decreased frequency and severity of
Compromised skin Napkin dermatitis napkin dermatitis in developed countries can be attrib-
Napkin dermatitis uted to the improvement of napkin materials. The intro-
• Irritation, friction duction of disposable napkins and, more importantly,
• enzyme activity
• microbial infection
newer disposable napkins with superabsorbent gels that
are able to absorb 50 times their own weight of water, has
Fig. 20.1  The aetiology of napkin dermatitis, in a model according to Berg
probably been responsible for significant improvement in
and Buckingham, adapted from Oranje and de Waard‐van der Spekk 1995
and Oranje 1995.
the prevalence of napkin dermatitis. Despite the term
napkin dermatitis, napkins are not themselves frequent
causes of the rash. In fact, allergic or irritant contact der-
SECTION 4: OTHER TYPES

irritation when patch tested on the skin of adults and chil- matitis caused by napkins has been reported but remains
dren [5]. They observed no irritation even when the con- an uncommon cause of napkin rashes in children [1].
OF DERMATITIS

centrations of ammonia were much higher than those Modern disposable napkins mainly consist of an inner
attributed to ‘ammoniacal diapers’. Investigations in the filtering layer, an intermediate layer able to absorb liq-
1980s and 1990s showed that the aetiology of napkin der- uids, and an outer layer that is waterproof. The water-
matitis was more complex and different from that thought proof layer prevents perspiration and therefore increases
previously [6,7]. the temperature and humidity of the environment. This
Activated faecal lipases and proteases primarily dam- layer plays a fundamental role in maintaining the imper-
age the skin. These enzymes are activated in alkaline and meability of the napkin. From this point of view, the mod-
hydrated conditions. Therefore, the key approach to pre- ern disposable napkin is much better than the traditional
venting napkin dermatitis is to keep the skin in the nap- cotton napkin. The latter is characterized by significantly
kin area protected from faeces and urine. lower absorbency and thus is associated with higher
The specific role of Candida albicans in the pathogenesis humidity in the napkin area. In addition, cotton napkins
of napkin dermatitis remains debated though its presence must always be worn together with outer plastic water-
is well established, especially in prolonged napkin der- proof pants. Compared with the traditional cotton nap-
matitis [8]. kins, modern disposable napkins reduce the contact time
of the skin with urine. Tight elastic around the waist and
References legs of disposable napkins is useful, especially in nurser-
1 Atherton D. Maintaining healthy skin in infancy using prevention of ies and daycare centres, to prevent the spread of gastroin-
irritant napkin dermatitis as a model. Community Pract
2005;78:255–7.
testinal infection. Allergic contact dermatitis has been
2 Jordan WE, Lawson KD, Berg RW et al. Diaper dermatitis: frequency occasionally reported and attributed to dyes used in older
and severity among a general infant population. Pediatr Dermatol napkin products as well as to fragrances that are common
1986;3:198–207. to many napkin brands. Most modern products now use
3 Zahorsky J. The ammoniacal diaper in infants and young children. Am
J Dis Child 1915;10:437–44. newer plant‐based pigments that are less likely to trigger
4 Cooke JV. The etiology and treatment of ammonia dermatitis of the contact dermatitis.
gluteal region of infants. Am J Dis Child 1921;22:481–92. The ideal napkin should be able to contain water
5 Leyden JL, Katz S, Stewart R et  al. Urinary ammonia and ammonia
­without preventing air flow. As such, the ideal napkin
producing microorganism in infants with and without diaper dermati-
tis. Arch Dermatol 1977;113:1678–80. does not yet exist, although significant improvements
6 Berg RW, Miligan MC, Sarbaugh FC. Association of skin wetness and in  technology now allow for rapid absorption of waste
pH with diaper dermatitis. Pediatr Dermatol 1994;11:18–20. products and can inform caregivers with colour indica-
7 Buckingham KW, Berg RW. Etiologic factors in diaper dermatitis: the
role of feces. Pediatr Dermatol 1986;3:107–12.
tors when the baby urinates.
8 Stamatas GN, Tierney NK. Diaper dermatitis. Pediatr Dermatol
2014;31:1–7. Faeces
Prolonged contact with faeces is the most irritant toxic
factor on the skin. Even babies who have never suffered
Aetiology
from napkin dermatitis will develop it when they have
Prolonged contact with urine, faeces, friction, hydration, diarrhoea, especially that due to infections [2,3].
temperature, chemical irritants and the napkin itself are Diarrhoea results in prolonged contact of faeces with
considered among the causative factors for napkin der- the skin and excessive hydration of the skin. Napkin
matitis [1]. Napkin dermatitis only occurs in countries or dermatitis improves immediately after diarrhoea stops.
settings where conventional napkins are used. In the Many factors have been suggested to be responsible for
1990s, the frequency and severity of napkin dermatitis the irritant capacity of faeces, ranging from acid stools
significantly decreased in westernized countries com- burning the skin to alkaline stools [3]. Berg et al. [4] and
pared with previous rates. Above all, the most severe Buckingham and Berg [5] suggested that faecal enzymes,
Chapter 20  Napkin Dermatitis 267

including proteases and lipases, could be responsible Chemical irritants


for the irritant potential of faeces. However, Yamamoto Chemical compounds have a direct toxic effect on the
[6] was not able to show increased levels of proteases in skin. Among these chemical irritants are deodorants, pre-
children with napkin dermatitis. Kuwayama et al. [7,8] servatives, creams and oils, in particular when used fre-
showed that faecal proteases and lipases play a minor quently throughout the day. Many antimycotics are not
role in the pathogenesis of napkin dermatitis. No sig- well tolerated [14].
nificant relationship between faecal enzyme activity
and severity of napkin dermatitis could be found. Microorganisms
Moreover, lipase and protease activities were destroyed Candida albicans is now considered the most important
by heating faeces at 100 °C for 5 min without a signifi- causative microorganism contributing to napkin dermati-
cant decrease in the irritation potential of the faeces. tis. Previously, Bacterium ammoniagenes was seen as
Kuwayama et  al. concluded that other faecal factors responsible for urea splitting and free ammonia release,
are  responsible for such toxic activities. The toxic leading to the term ‘ammoniacal dermatitis’ [14,15].
substances are probably present in the fraction of
­ Besides C. albicans and B. ammoniagenes, other microor-
substances of the water‐soluble extract that exceed
­ ganisms have been implicated, such as Proteus,
5000 kDa. The interaction between faeces and other Pseudomonas, Escherichia coli, Streptococcus, Staphylococcus

SECTION 4: OTHER TYPES


­factors is complex. However, the occurrence of napkin and Enterococcus [15]. Often, Staph. aureus and C. albicans
­dermatitis around the normal, healthy anus shows that were isolated simultaneously. The role played by anaero-

OF DERMATITIS
faeces alone can induce napkin dermatitis [5,9]. bic bacteria has been emphasized [5].
The frequency with which C. albicans occurs in the
Urine ­various clinical forms of napkin dermatitis is not signifi-
Urine may contribute to the pathogenesis of napkin cantly different [14]. However, the severity and duration
dermatitis through skin hydration [10,11]. Urine and
­ of napkin dermatitis are more important in determining
ammonia as single factors fail to induce erythema. The colonization by C. albicans than the type of napkin derma-
interaction of ammonia, bacteria, faeces, friction and titis [16–18]. Three infantile disorders are definitively asso-
other factors contributes to the development of dermatitis ciated with C. albicans: oral thrush, chronic mucocutaneous
[12]. However, urine, especially after incubation at 37 °C candidiasis and congenital candidiasis [17]. An association
(body temperature), may contain as yet unidentified between oral thrush and napkin dermatitis has not been
factors that could be responsible for irritation after
­ clearly demonstrated [17–19]. Meneghini and Bonifazi [20]
­prolonged exposure [13]. prospectively studied 112 healthy children aged 5–15
months. They studied the clinical aspects of the napkin
area, and combined their data with bacterial and fungal
Friction
cultures. The mycological examination proved positive for
Rubbing of the napkin on the skin or skin‐to‐skin contact
C. albicans in 19 of 1001 cases, not a particularly low pro-
(friction) due to movement of the infant can be responsi-
portion considering that the authors studied healthy chil-
ble for chafing dermatitis [13]. The causative role of fric-
dren. Conversely, the study of a healthy population
tion is stressed by the predilection of napkin dermatitis
allowed the observation of the initial lesions of napkin
for the convex surfaces of the genitalia, the buttocks and
dermatitis. These investigations were repeated consecu-
the waistline. This type of napkin dermatitis spares the
tively for many weeks, thus establishing that C.  albicans
depth of the folds. The latter variant is also called W‐
can be isolated from the most intensely erythematous
shaped napkin dermatitis in female infants [13].
lesions independent of the site involved and the type of
lesions. C. albicans was rarely isolated before the appear-
Hydration ance of napkin dermatitis. In this study, oral thrush was
Excessive hydration of the skin in the napkin area can be never associated with a positive culture for C. albicans
caused by napkins that prevent evaporation of moisture from the napkin area. These findings seem to suggest that
from the skin [13]. Napkins may facilitate hydration C. albicans is a secondary invader of skin that is already
induced by fever and sweating (causing miliaria). Urea damaged. Most authors favour this hypothesis [17–23].
also facilitates hydration of the skin. Excessive hydration is However, other authors believe that C. albicans can be a
responsible for maceration of the skin, leading to compro- primary factor in the pathogenesis of napkin dermatitis
mised barrier properties of the epidermis and providing [17,23]. The quantity of organisms might be more impor-
an ideal environment for the proliferation of microorgan- tant than the simple presence of C. albicans in establishing
isms [12]. Excessive hydration also makes the skin more its aetiological role. According to some authors [17], under
susceptible to frictional trauma [12,13]. occlusive conditions, 10 000 organisms of C. albicans per
square centimetre could induce primary skin infection.
Temperature
Increased temperature in the napkin area is caused by Allergy
napkins that prevent perspiration, reducing heat loss. The term napkin dermatitis is often interpreted as a ­contact
Napkins may also aggravate the effect of fevers. The allergy to the napkin. This misunderstanding leads some
increased temperature is responsible for vasodilation and caregivers to choose cotton in preference to disposable nap-
promotion of inflammation. kins, with consequent worsening of the napkin dermatitis.
268 Section 4  Other Types of Dermatitis

Allergic contact dermatitis in the n


­ apkin area is a possibility, 13 Leyden JL, Katz S, Stewart R et al. Urinary ammonia and ammonia
producing microorganisms in infants with and without diaper der-
with possible sensitizers including rubber, detergents, lano-
matitis. Arch Dermatol 1977;113:1678–80.
lin, preservatives, fragrances, neomycin and mercurial 14 Dixon PN, Warin RP, English MP. Role of Candida albicans infec-
compounds [22,23]. In addition, disperse dye has recently tion in napkin rashes. Br Med J 1969;5:23–7.
been implicated as a potential contact sensitizer in some 15 Weston WL, Lane AT, Weston JA. Diaper dermatitis: current concepts.
Pediatrics 1980;66:532–6.
children with napkin dermatitis, as confirmed by epicuta- 16 Meneghini CL, Barile F, Bonifazi E. Ruolo della Candida albicans nella
neous patch testing [23]. However, allergic contact dermati- dermatite da pannolino. Arch Argent Dermat 1988;38:287–9.
tis is uncommon, especially in the first year [21]. Some 17 Caputo RV. Fungal infections in children. Dermatol Clin 1986;4:
authors consider napkin dermatitis as contact hypersensi- 137–49.
18 Jacobs AH. Eruptions in the diaper area. Pediatr Clin N Am 1978;25:
tivity to products associated with the presence of C. albicans 209–24.
[20]. However, no evidence of acquired hypersensitivity to 19 Katz R. Treatment of diaper dermatitis. Postgrad Med 1972; Nov:153–6.
C. albicans could be demonstrated in subjects with multiple 20 Meneghini CL, Bonifazi E. Diaper rash. Pediatr Dermatol News
1982;1:69–75.
consecutive infections of C. albicans [21]. 21 Koblenzer PJ. Diaper dermatitis – an overview emphasis on rational ther-
apy based on etiology and pathodynamics. Clin Pediatr 1973;12:386–92.
22 Roul S, Ducombs G, Leaute‐Labreze C et  al. Lucky Luke contact
References dermatitis due to rubber components of diapers. Contact Dermatitis
1 Oranje AP, de Waard‐van der Spek FB. Comparison of cloth and 1998;38:363–4.
SECTION 4: OTHER TYPES

superabsorbent paper diapers for preventing diaper dermatitis. Eur 23 Alberta L, Sweeney SM, Wiss K. Diaper dye dermatitis. Pediatrics
J Pediatr Dermatol 1991;1:225–32. 2005;116:e450–2.
2 Austin AP, Miligan MC, Pennington K et al. A survey of factors associ-
OF DERMATITIS

ated with diaper dermatitis in 36 pediatric practices. J Pediatr Health


Care 1988;2:295–9.
3 Jordan WE, Lawson KD, Berg RW et al. Diaper dermatitis: frequency Clinical and differential diagnosis
and severity among a general infant population. Pediatr Dermatol
1986;3:198–207. Napkin dermatitis represents a descriptive phenotype for
4 Berg RW, Miligan MC, Sarbaugh FC. Association of skin wetness and multiple dermatoses that may affect the anogenital skin
pH with diaper dermatitis. Pediatr Dermatol 1994;11:18–20. of napkin‐wearing infants. These range from common
5 Buckingham KW, Berg RW. Etiologic factors in diaper dermatitis: the
role of feces. Pediatr Dermatol 1986;3:107–12. problems such as a primary irritant dermatitis directly
6 Yamamoto K. Hygienic care of the diaper area in childhood. Pediatr caused by occlusion and friction, to rare diseases such as
Dermatol News 1987;6:239–41. Langerhans cell histiocytosis, unrelated to the wearing of
7 Kuwayama M, Kon Y, Yamamoto K. New etiologic factors in diaper
dermatitis: skin irritability and focal irritancy as major causes of dia-
a napkin. Thus, napkin dermatitis should be viewed not
per dermatitis. J Pediatr Dermatol 1992;11:93–102. as a specific diagnostic entity but rather as a regional
8 Kuwayama M, Kon Y, Yamamoto K. New etiologic factors in diaper diagnosis, like hand dermatitis, that encompasses various
dermatitis: the confirmation of irritant in feces. J Pediatr Dermatol skin diseases of multifactorial aetiology.
1992;11:103–11.
9 Bonifazi E. Diaper rash in practical pediatric dermatology. Eur J There can be significant overlap in the clinical presenta-
Pediatr Dermatol 1993;3:57–62. tion of the conditions that cause napkin dermatitis, and
10 Leyden JJ. Diaper dermatitis. Dermatol Clin 1986;4:23–8. specific diagnosis may be difficult. Although history and
11 Cooke JV. The etiology and treatment of ammonia dermatitis of the
correlating physical signs are often helpful, the differen-
gluteal region of infants. Am J Dis Child 1921;22:481–92.
12 Brook J. Microbiology of secondarily infected dermatitis. Int J tial diagnosis is based primarily on the morphology and
Dermatol 1992;31:700–2. location of the rash (Table 20.1, Box 20.1).

Table 20.1  Common types of napkin rash

Morphology Location History Diagnostic


tests

Primary Glazed, confluent erythema or wrinkled Convex surfaces (buttocks, thighs, Rash may wax and wane; recent None
irritant parchment‐like skin, scale, papules abdomen, mons pubis, labia majora, diarrhoea; infrequent napkin
scrotum) changes; use of plastic pants
Candidal Bright‐red, scaly plaques; sharply Skinfolds Antibiotic therapy; diarrhoea; rule KOH
marginated satellite papules and out oral thrush
pustules; papulosquamous id (rare)
Atopic Same as primary irritant, excoriations, Convex surfaces; eczema at other sites Family history of atopy; pruritus; None
lichenification, crusting (e.g. cheeks, antecubital and popliteal chronicity
fossae)
Seborrhoeic Salmon‐coloured, scaly plaques; sharply Skinfolds with subsequent spread over Asymptomatic; presents early in None
marginated, greasy, yellow scale convex surfaces; involvement of scalp, infancy; good response to
postauricular folds, axilla, neck treatment
Intertrigo Sharply demarcated erythema; skin Skinfolds Often seen in overweight infants None
maceration; little scale; no satellite lesions
Bullous Flaccid bullae; honey‐coloured crust Usually convex surfaces (thighs, buttocks, Common in newborns Gram stain;
impetigo lower abdomen); may spread rapidly to (Staphylococcus colonization of bacterial
other areas of the body umbilicus) culture

KOH, potassium hydroxide.


Chapter 20  Napkin Dermatitis 269

Box 20.1  Differential diagnosis of napkin rash*

Eczematous/papulosquamous

• Primary irritant contact dermatitis


• Allergic contact dermatitis
• Candidiasis
• Intertrigo
• Seborrhoeic dermatitis
• Atopic dermatitis
• Psoriasis
• Kawasaki disease
• Langerhans cell histiocytosis (Letterer–Siwe)
• Perianal streptococcal disease

Nodular

SECTION 4: OTHER TYPES


• Granuloma gluteale infantum
• Extrafacial periorificial granulomatous dermatitis
• Metastatic Crohn disease

OF DERMATITIS
Vesiculobullous/erosive Fig. 20.2  Irritant contact napkin dermatitis. Erythema primarily involves
• Acrodermatitis enteropathica convexities of the napkin area.
• Biotin‐responsive multiple carboxylase deficiency
• Cystic fibrosis compromised skin [3]. In some cases, irritant contact
• Bullous impetigo ­ ermatitis may arise from topically applied medicaments
d
• Miliaria designed to protect the napkin area.
• Scabies Clinically, irritant contact napkin dermatitis is charac-
• Other vesiculobullous disease (varicella, herpes simplex, hand, foot terized by a glazed, confluent erythema, sometimes
and mouth disease, chronic bullous disease of childhood, bullous resembling a burn. There may be erythematous papules,
mastocytosis, incontinentia pigmenti, epidermolysis bullosa) oedema and scaling of the involved skin (Fig. 20.2). When
• Child abuse (burns) the eruption begins to resolve, a wrinkled parchment‐like
• Laxative‐induced dermatitis
appearance may be noted. The rash, however, may wax
• Human immunodeficiency virus
and wane. This form of napkin rash primarily involves
the skin where contact with the napkin is greatest, i.e. the
Verrucous
convexities of the buttocks, medial thighs, mons pubis
• Congenital syphilis (condylomata lata) and scrotum or labia majora. The intertriginous areas are
• Perianal pseudoverrucous papules and nodules generally spared. An early manifestation of this type of
napkin dermatitis, especially in infants less than 4 months
* There is significant overlap with regard to the morphological grouping of age, is mild perianal erythema [4].
of these dermatoses. This list is intended to provide general guidelines. There are two less common morphological subtypes of
irritant contact napkin dermatitis. One is the so‐called
‘tide water’ mark dermatitis in which an erythematous
macular eruption is band‐like and confined to the mar-
Contact napkin dermatitis gins of the napkin area on the thighs (Fig. 20.3) or abdo-
Primary irritant contact napkin dermatitis men. This characteristic rash results from the exaggerated
Irritant contact dermatitis is by far the most prevalent chafing that may occur at the edges of the napkin. Skin
form of napkin dermatitis. In fact, this ‘chafing’ napkin integrity is compromised by the frequent cycles of wet-
rash is probably the most common skin problem of ting and drying [5] combined with the traumatic friction
infancy. It is precipitated by the constant moisture, due to the plastic border of a disposable napkin. There is
­occlusion and frictional forces that occur under a napkin also a severe form of irritant contact napkin dermatitis
and which compromise skin integrity. With maceration known as Jacquet dermatitis or erosive napkin dermatitis.
of  the stratum corneum, the skin barrier function is This presents with papuloerosive lesions that have a
impaired, predisposing it to secondary irritants. These punched‐out or crater‐like appearance (Fig.  20.4). These
secondary factors include urinary ammonia, increased ulcers have also been referred to as ‘ammoniacal ulcers’.
urine pH (due to urea‐splitting bacteria), faecal proteases This eruption, when it occurs, tends to affect older
and lipases, C. albicans, bacterial overgrowth and frequent napkin‐wearing children. In male infants, ulceration
washing, especially with detergent soaps [1,2]. Urinary involving the glans penis and urinary meatus may cause
ammonia was for many years mistakenly believed to be discomfort or difficulty urinating.
the primary instigator of irritant napkin dermatitis. It is The differential diagnosis in atypical cases (wherein
now thought to increase inflammation only in already an  antecedent history of napkin dermatitis is absent)
270 Section 4  Other Types of Dermatitis
SECTION 4: OTHER TYPES

Fig. 20.3  Irritant contact napkin dermatitis: the ‘tide water’ mark.
OF DERMATITIS

Fig. 20.5  Toxic napkin dermatitis (induced by bleaching solution). Source:


Courtesy of Professor Arnold Oranje.

Fig. 20.4  Jacquet erosive napkin dermatitis.

would include herpes simplex virus infection, which Fig. 20.6  Allergic contact dermatitis secondary to rubber components in
the napkin: the ‘holster sign’. Source: Courtesy of Dr. Neil Prose.
may  be associated with a history of abuse or vertical
transmission.
an eczematous eruption. The vesicular phase may not be
Allergic contact napkin dermatitis apparent after the first few days of the rash. Allergic con-
A true allergic contact dermatitis may complicate another tact napkin rash typically complicates a primary irritant
type of napkin rash or present de novo. This is an uncom- napkin rash and therefore follows the same distribution,
mon occurrence, particularly in children under 2 years of i.e. the convex surfaces of the skin under occlusion. There
age. Allergic contact dermatitis should be considered, may, however, be prominent flexural involvement, par-
however, if a child does not respond appropriately to ticularly if the sensitivity is to a topical preparation that
treatment. It should also be suspected if the application of concentrates within the skinfolds. The characteristic pat-
a potential allergen causes the napkin rash to become tern of contact dermatitis to rubber components around
more intense or to spread. Contact allergy may develop to the proximal thighs and the waistline associated with dis-
certain medicaments applied to the skin such as those posable napkins has been dubbed the ‘holster sign’ [8,9]
containing paraben, lanolin or neomycin. Allergic sensi- (Fig. 20.6).
tivity may also occur to chemicals contained within dis-
posable napkins or to napkin covers (as with disperse
Simple intertrigo
dyes used to colour older napkins) [6] or fragrances, or an
irritant reaction may develop to laundry detergents used Intertrigo is an inflammatory process that occurs in areas
with cloth napkins (Fig. 20.5) [7]. where skin rubs against skin, as in the folds of the groin,
Morphologically, allergic contact dermatitis begins posterior thighs and intergluteal cleft. Heat, moisture and
with erythema and small vesicles that rupture, leading to sweat retention, coupled with friction, cause maceration,
Chapter 20  Napkin Dermatitis 271

inflammation and sometimes skin erosion. Intertrigo may factor, but Candida is frequently cultured from common
present with sharply demarcated moist erythema con- irritant napkin rash and the development of granuloma
fined to the skinfolds. In contrast to a candidal napkin gluteale infantum in these patients is rare. Finally, fluori-
rash, there is little associated scale and no satellite lesions. nated topical steroids have also been implicated in the
As intertrigo is essentially due to friction and retained development of these nodules because, in most reported
moisture, it may be considered a subtype of irritant con- cases, relatively potent topical steroids were used [11].
tact dermatitis. The tropical climate of the napkin area The role of topical corticosteroids is supported by the lack
makes the napkin‐wearing child susceptible to the devel- of cases of granuloma gluteale infantum prior to the intro-
opment of simple intertrigo; however, it is most often duction of corticosteroids. Its lower prevalence today
seen in overweight infants. might be attributable to parental corticosteroid phobia.
In cases of intertrigo, there may be secondary infection
with C. albicans or bacteria. Also, the clinical presentation References
of intertrigo may be indistinguishable from other disor- 1 Berg RW. Etiology and pathophysiology of diaper dermatitis. Adv
Dermatol 1988;3:75–98.
ders in which the role of C. albicans is controversial, such
2 Weston WL, Lane AT, Weston JA. Diaper dermatitis: current concepts.
as infantile seborrhoeic dermatitis or inverse napkin pso- Pediatrics 1980;66:532–6.
riasis. This overlap in clinical features may make defini- 3 Leyden JJ, Katz S, Stewart R et  al. Urinary ammonia and ammonia

SECTION 4: OTHER TYPES


tive diagnosis difficult. producing microorganisms in infants with and without diaper der-
matitis. Arch Dermatol 1977;113:1678–80.
4 Rasmussen JE. Diaper dermatitis. Pediatr Rev 1984;6:77–82.

OF DERMATITIS
Granuloma gluteale infantum 5 Koblenzer PJ. Diaper dermatitis: an overview. Clin Pediatr
1973;12:386–92.
Granuloma gluteale infantum is a rare nodular eruption 6 Alberta L, Sweeney SM, Wiss K. Diaper dye dermatitis. Pediatrics
2005;116:e450–2.
that may arise within an area of pre‐existing irritant 7 Jacobs AH. Eruptions in the diaper area. Pediatr Clin North Am
­napkin dermatitis. It is characterized by firm, painless, 1978;25:209–24.
reddish‐brown to purple nodules varying in size from 0.5 8 Larralde M, Raspa ML, Silvia H et al. Diaper dermatitis: a new clinical
feature. Pediatr Dermatol 2001;18:167–8.
to 4 cm (Fig.  20.7). These nodules usually appear on the 9 Roul S, Ducombs G, Leaute‐Labreze C et al. ‘Lucky Luke’ contact der-
buttocks and inner thighs and occasionally on the lower matitis to the rubber components of diapers. Contact Dermatitis
abdomen. Lesions have also been reported outside the 1998;38:363–4.
napkin area involving the folds of the axilla and neck. 10 Sweiden NA, Salman SM, Kibbi AG et  al. Skin nodules over the
­diaper area: granuloma gluteale infantum. Arch Dermatol 1989;
These angioma‐like swellings can be ominous in appear- 125:1706–7.
ance, resembling lesions of Kaposi sarcoma or lymphoma. 11 Uyeda K, Nakayasu K, Takaisski Y et al. Kaposi sarcoma‐like granu-
The nodules may persist for weeks to months but they loma on diaper dermatitis. Arch Dermatol 1973;107:605–7.
eventually regress spontaneously. When the lesions
resolve, atrophic scars may remain.
Infectious napkin dermatitis
The aetiology of granuloma gluteale infantum is
unclear. Several factors, however, have been suspected. Candidal (monilial) napkin dermatitis
Because this condition invariably arises in an area of pre‐ Provided there is sufficient warmth and moisture, as
existing napkin dermatitis, it may represent a localized occurs under an occlusive napkin, C. albicans can prolifer-
cutaneous response to longstanding inflammation. There ate on the skin surface and penetrate the stratum cor-
is no correlation, however, between the severity of the neum. It can then activate complement via the alternative
napkin rash and the incidence of this nodular eruption pathway and induce inflammation [1].
[10]. Lesions may appear even when the napkin rash is Morphologically, candidal napkin dermatitis is perhaps
resolving. C. albicans has been considered an aetiological the most characteristic of the napkin rashes. It presents
with sharply marginated, intensely red, scaly plaques
with satellite papules and pustules (Fig. 20.8). These satel-
lite lesions may have peripheral scale. In some severe
cases of candidiasis, there may be widespread skin ero-
sion. When candidiasis involves the genitalia, there is
usually confluent erythema involving the entire scrotum
or labia. This is unlike psoriasis, in which genital involve-
ment is usually more localized and in which the lesions
are more sharply demarcated.
The diagnosis of candidal napkin rash is primarily
based on its characteristic morphology. Potassium
hydroxide (KOH) scrapings may reveal pseudohyphae
and help to confirm the diagnosis. Scrapings may be neg-
ative, however, if they are taken from inflamed areas or in
longstanding cases in which the inflammatory response
has led to the death of the Candida organism. Yield on
KOH is greatest from scrapings of the periphery of the
Fig. 20.7  Granuloma gluteale infantum. rash or a fresh papular or pustular lesion.
272 Section 4  Other Types of Dermatitis

trunk, usually sparing the extremities. This eruption may


last for days to weeks. Generally, C. albicans cannot be
cultured from these plaques. However, active candidiasis
of skinfolds in the neck and axilla, in addition to the
inguinal area, may be seen. The pathogenesis of this
allergic skin eruption is not well understood; it is pre-
sumed to be a response to an antigenic stimulus.
Although it was once thought that this reaction sug-
gested an underlying psoriatic or atopic diathesis, most
affected infants do not go on to develop psoriasis or
atopic dermatitis [2]. Confusion exists over the entity
‘napkin psoriasis’ [3]. Balasubramanian et al. [3] recently
described napkin psoriasis as diaper dermatitis with pso-
riasiform id eruptions and stressed that it is nowadays
rare. Psoriasis‐like eruptions were described in patients
with Kawasaki disease by Haddock et al. [4]; the overall
SECTION 4: OTHER TYPES

Fig. 20.8  Candida napkin dermatitis with satellite lesions. clinical and histopathological findings were identical to
conventional psoriasis. The last two reports illustrate
OF DERMATITIS

that a psoriatic clinical presentation includes an aspecific


id reaction and does not predict psoriasis in later life.
The rare congenital onset of candidal napkin dermatitis
may represent a manifestation of congenital cutaneous
candidiasis. Neonates with congenital cutaneous candidi-
asis may present with characteristic macerated areas of
erythema in the anogenital region; however, infected
infants may also present with papulopustules, widespread
erosions or a burn‐like dermatitis [5]. In term infants,
congenital cutaneous candidiasis is typically a benign,
self‐limited eruption attributed to vertical transmission
from the colonized mother. The condition responds to
topical antifungal agents. However, preterm neonates,
especially those manifesting signs of respiratory distress,
are at risk for associated systemic candidiasis, which war-
rants systemic antifungal therapy.

Perianal streptococcal disease


and streptococcal intertrigo
Perianal streptococcal disease is an often unsuspected
condition, the signs of which may be quite subtle. It may
be mistaken for an irritant napkin dermatitis, a perianal
candidiasis, psoriasis or pinworm infestation. When
blood‐streaked stools are present, inflammatory bowel
disease may also be a consideration. Perianal cellulitis is
Fig. 20.9  Psoriasiform skin reaction to Candida napkin dermatitis. caused by group A β‐haemolytic Streptococcus. Despite its
name, it is not a true cellulitis but rather a superficial skin
infection.
Children presenting with candidal napkin dermatitis This entity typically presents as sharply demarcated,
frequently have a recent history of treatment with broad‐ bright erythema in the perianal area. There may be peri-
spectrum antibiotics. Diarrhoea also makes an infant rectal fissuring. Other intertriginous areas, such as the
more susceptible to candidiasis. The oral cavity should be neck, axillae and inguinal folds, may also become affected.
examined to rule out concomitant thrush. When the rash The condition is usually associated with itching and pain.
is chronic or recurrent, seeding from the gastrointestinal The pain occurs particularly during defaecation and can
tract or from a maternal candidal vaginitis should be con- lead to faecal hoarding. Often, there is a history of
sidered. Other potential sources include maternal masti- repeated streptococcal pharyngitis in family members.
tis, if the mother is breastfeeding, or a contaminated Diagnosis can be made by cotton swab bacterial culture of
dummy (pacifier) or nipple. the affected perianal skin.
On rare occasions, a psoriasiform skin reaction Streptococcal intertrigo involves bacterial infection of
(Fig. 20.9) may occur as a complication of severe candidal the inguinal creases (Fig. 20.10) and other intertriginous
napkin rash. Shortly after therapy is initiated, scaly pso- areas, including the axillae, popliteal spaces and neck
riasiform papules and plaques rapidly develop on the folds of infants [6]. Coexistent perianal disease may or
Chapter 20  Napkin Dermatitis 273

and lower abdomen [7]. Rapid spread to other areas of the


body also occurs.
This infection is caused by coagulase‐positive, usually
phage type II, Staph. aureus. This organism produces an
epidermolytic toxin that separates the upper layers of the
epidermis. Diagnosis can be confirmed with a Gram stain
and bacterial culture.
Nonbullous (encrusted) impetigo due to other types of
Staph. aureus and streptococci may occur in the napkin
area. Nonbullous impetigo is characterized by vesicles
and pustules that develop quickly into scabs with thick,
often honey‐coloured crusts.

Scabies
Scabies should be considered in the differential diagnosis
of napkin rash when pruritic papules, vesicles and vesic-

SECTION 4: OTHER TYPES


Fig. 20.10  Streptococcal intertrigo. Intertriginous, foul‐smelling, ulopustules are seen. Typically, these primary lesions
macerated erythema is a typical presentation. are intermingled with eczematization, excoriation and

OF DERMATITIS
crusting. Linear burrows are pathognomonic of scabies.
However, because burrows are extremely difficult to visu-
alize, this clinical sign is not of great clinical value. Sites of
predilection for scabies are the finger webs, wrists, ante-
cubital fossae, axilla, areolae and areas around the umbili-
cus, lower abdomen, genitals and buttocks. In infants and
young children, the palms, soles, head, neck and face may
also be affected. A high percentage of children with
­scabies may develop nodular lesions. These persistent,
reddish‐brown, infiltrated nodules are more likely to
occur in covered parts of the body, such as the groin, but-
tocks and genitalia. The nodules often persist for months,
despite therapy, and may be mistaken for mastocytosis,
histiocytosis or cutaneous lymphoma.
In addition to the presence of lesions in a characteristic
distribution, the diagnosis of scabies is aided by a history
Fig. 20.11  Impetigo bullosa. Source: Courtesy of Professor Arnold Oranje. of intense itching and pruritus in family members or car-
egivers. Definitive diagnosis is made by microscopic
may not be present. The infection is characterized by a examination of scrapings of suspicious lesions with the
foul‐smelling, macerated erythema of intertriginous finding of the mite, ova or faecal matter. In many cases of
areas. Occasionally, mixed infections with Staphylococcus scabies infestation a scabies scraping may be negative,
aureus, Pseudomonas or Proteus species have been observed. and therapy may be prescribed empirically.
Frequently misdiagnosed as candidal intertrigo, cases of
streptococcal intertrigo are often resistant to topical anti- Congenital syphilis
fungal medications. Therapy with oral penicillin deriva- Until the early years of the twentieth century, syphilis was
tives for streptococcal intertrigo or appropriate coverage believed to be the major cause of napkin rash [8]. Although
for mixed infections is curative. Topical anti‐inflamma- we are now more enlightened as to the causes of napkin
tory medications, such as topical corticosteroids, may dermatitis, because of the recent increase in the incidence
assist in reducing associated inflammation more quickly of syphilis, congenital syphilis remains within the differ-
to provide more rapid symptomatic relief. ential diagnosis.
Clinical signs of early congenital syphilis may present
Bullous impetigo at birth or develop within the first 3 months of life. In the
The constant moisture and warmth of the napkin environ- anogenital region raised, moist, wart‐like lesions (condy-
ment are predisposing factors to the development of bul- lomata lata) may be seen in addition to moist eroded
lous impetigo, which is relatively common in the napkin areas. There may be a generalized erythematous papulos-
area. It is especially common in newborns as a result of quamous eruption similar to that seen in secondary syph-
colonization of the umbilicus with Staphylococcus aureus. ilis. Erythema, oedema, vesiculobullous lesions, fissuring
Bullous impetigo is characterized by the formation of and desquamation may occur on the palms and soles, and
large flaccid bullae arising from apparently normal skin. pale, slightly raised mucous membrane patches may be
The bullae rupture easily, leaving red, moist, denuded found in the mouth and on the lips. Other clinical features
areas and honey‐coloured crusts (Fig.  20.11). There are include anaemia, fever, wasting, hepatosplenomegaly,
usually multiple lesions involving the thighs, buttocks rhinitis (‘snuffles’), osteochondritis or periostitis, and
274 Section 4  Other Types of Dermatitis

pseudoparalysis. Dark‐field examination of moist skin


lesions, especially the condylomata lata, should reveal
spirochaetes, and serological testing for syphilis will con-
firm the diagnosis.

Human immunodeficiency virus (HIV)


Severe napkin dermatitis can be a manifestation of paedi-
atric HIV infection. Leibovitz et  al. [9], in a study of
Romanian children with HIV, noted napkin dermatitis, in
some cases very severe, in eight out of 23 infants. Unusual
erosive napkin dermatitis with deep gluteal cleft ulcera-
tion has been reported as a presenting sign of HIV infec-
tion [10]. In addition, ulcerative eruptions in the anogenital
area of HIV‐infected infants can be caused by, or compli-
cated by, infection with herpes simplex, cytomegalovirus
[11] and other opportunistic agents. Therefore, HIV infec-
SECTION 4: OTHER TYPES

tion should be considered in infants with unusually


severe or erosive napkin dermatitis.
OF DERMATITIS

References
1 Leyden JJ. Diaper dermatitis. Dermatol Clin 1986;4:23–8.
2 Singalavanija S, Frieden IJ. Diaper dermatitis. Pediatr Rev
1995;16:142–7.
Fig. 20.12  Infantile seborrhoeic dermatitis with involvement of the
3 Balasubramanian P, Jagadeesan S, Thomas J et al. Diaper dermatitis
with psoriasiform id eruptions. Indian J Dermatol 2015;81:435–7. inguinal folds.
4 Haddock ES, Calame A, Shimizu C et al. Psoriasiform eruptions dur-
ing Kawasaki disease (KD): a distinct phenotype. J Am Acad Dermatol
2016;75:69–76.e2.
5 Darmstadt GL, Dinulos JG, Miller Z. Congenital cutaneous can- spread to involve the forehead or the entire face. Other
didiasis, clinical presentation, pathogenesis, and management typical areas of skin involvement, primarily flexural
guidelines. Pediatrics 2000;105:438–44.
areas, include the axilla, neck, postauricular folds and
6 Honig PJ, Frieden IJ, Kim HJ et al. Streptococcal intertrigo: an under-
recognized disease. Pediatrics 2003;112(6):1427–9. umbilicus.
7 Rasmussen JE. Diaper dermatitis. Pediatr Rev 1984;6:77–82. The lesions of infantile seborrhoeic dermatitis are gen-
8 Leyden JJ, Kligman AM. The role of microorganisms in diaper derma- erally asymptomatic; affected children appear to be com-
titis. Arch Dermatol 1978;114:56–9.
9 Leibovitz E, Orlow S, Lawrence R et  al. Children of Romania: the
fortable and do not exhibit signs of significant pruritus.
AIDS legacy. Children’s Hosp Q 1991;3:99–108. Infantile seborrhoeic dermatitis typically presents early in
10 Sweiden NA, Salman SM, Kibbi AG et  al. Skin nodules over the infancy, generally within the first 3 months of life, most
­diaper area: granuloma gluteale infantum. Arch Dermatol 1989; commonly at 3–6 weeks of age. The prognosis is good
125:1706–7.
11 Thiboutot DM, Beckford A, Mart CR et  al. Cytomegalovirus diaper and, in most cases, the condition resolves spontaneously
dermatitis. Arch Dermatol 1991;127:396–8. by 3–6 months of age.

Atopic dermatitis
Primary and secondary inflammatory Atopic dermatitis usually spares the napkin area. This is
conditions somewhat surprising given that atopic individuals are
Infantile seborrhoeic dermatitis more susceptible to irritant reactions and do poorly with
Infantile seborrhoeic dermatitis is an inflammatory skin frequent exposure of the skin to water. Nevertheless,
disease which can commonly affect the napkin area of when atopic dermatitis presents in the napkin area, its
infants. It is characterized by salmon‐coloured, scaly appearance is similar to that of primary irritant napkin
plaques that involve the inguinal folds (Fig.  20.12). In dermatitis. The rash, however, tends to be more chronic
severe cases, there can be spread of the eruption from the and refractory to treatment. There may be oozing or crust-
skinfolds to convex skin surfaces of the napkin area. ing due to secondary infection with Staph. aureus. There
Typically, the lesions are sharply demarcated. The scales may also be lichenification and excoriations. In most cases
of infantile seborrhoeic dermatitis tend to be greasy and of atopic napkin dermatitis there are more typical eczem-
yellowish in colour. There may be associated fissuring, atous lesions elsewhere on the body, involving particu-
crusting and weeping. Unlike candidiasis, which also larly the cheeks and antecubital and popliteal fossae.
involves the skinfolds of the napkin area, there are no sat- Pruritus is a cardinal feature of this disease, and the atopic
ellite lesions. infant may be irritable and uncomfortable, constantly
A major clue to the diagnosis of this type of napkin rash rubbing or scratching the affected areas. One must bear in
is the presence of the eruption in characteristic areas away mind, however, that the signs of pruritus usually do not
from the anogenital region. Infantile seborrhoeic dermati- occur until the child is 2 months of age and is able to rub
tis often begins with diffuse erythema and thick, adher- or scratch. A family history of atopy is often obtained and
ent, greasy scales involving the scalp. The eruption may may also lend support to the diagnosis.
Chapter 20  Napkin Dermatitis 275

cases of simple intertrigo or seborrhoeic dermatitis


because of the primary flexural involvement.
There is also a risk for secondary bacterial infection
of  these types of napkin dermatitis. This is particularly
important in cases of atopic dermatitis. If crusting
or  blistering occurs, Staph. aureus infection should be
considered.
Psoriasis can be precipitated by pre‐existing napkin
dermatitis such as an irritant contact dermatitis or sebor-
rhoeic dermatitis, but a psoriasis id reaction pattern is
also possible. This is due to the isomorphic response to
skin injury known as the Koebner phenomenon. Thus,
because psoriasis may appear secondarily, it may present
with a distribution more characteristic of the primary
condition. Diagnosis may be suggested by the relatively
rapid response of the primary condition to specific

SECTION 4: OTHER TYPES


­therapy and the recalcitrance of the secondary psoriatic
eruption.

OF DERMATITIS
Fig. 20.13  Psoriasis napkin change. Note sharply marginated border of
plaque. Source: Courtesy of Professor Arnold Oranje. Miliaria
Miliaria (prickly heat) is commonly concentrated in the
napkin area because of the tropical conditions under
Psoriasis
the  napkin. Other typical areas of involvement include
Psoriasis is rare in infancy. When it does occur in the first
the face, neck and axilla. Miliaria may be precipitated
year of life, it is likely to appear in the napkin area because
by  fever, high ambient temperature or use of an occlu-
of the Koebner phenomenon caused by more common
sive  ointment or plastic napkin pants [2]. Increased
types of napkin rash. It typically presents as sharply
heat  and humidity cause blockage of the eccrine sweat
demarcated, erythematous plaques of variable size, with
glands. Different clinical forms of miliaria result based
involvement of the convex surfaces of the buttocks as well
on the level at which the eccrine duct is obstructed.
as the inguinal folds (Fig. 20.13). Unlike psoriatic plaques
Miliaria crystallina is more commonly seen in the new-
elsewhere on the body, there may be little or no scale
born and appears as clear, small, superficial vesicles with-
due to the constant hydration under the napkin. Diagnosis
out associated erythema. The eccrine duct obstruction in
may be difficult and become clear only after long‐term
this form of miliaria occurs within the stratum corneum.
observation. Psoriasis‐like eruptions were described in
In older infants, miliaria rubra (or pustulosa) is more
patients with Kawasaki disease by Haddock et al. [1]. As
common. This form occurs when the sweat duct is
opposed to irritant contact napkin dermatitis, or sebor-
obstructed within the deeper layers of the epidermis.
rhoeic dermatitis, psoriasis in the napkin area tends to be
Miliaria rubra is characterized by small, uniform erythe-
chronic and more resistant to treatment with low‐potency
matous grouped papules and pustules which are nonfol-
topical steroids. Topical corticosteroids that are slightly
licular. The pustules are sterile, and Gram stain will reveal
more potent than 1% hydrocortisone, such as desonide
polymorphonuclear leucocytes but no bacteria or yeast.
0.05% or alclometasone dipropionate 0.05%, may be nec-
In cases of miliaria pustulosa, it is important to rule out a
essary. A positive family history of psoriasis may be elic-
staphylococcal pustulosis.
ited. Often, when psoriasis involves the groin, it is also
present in other flexural areas such as the axillae and
neck. Certainly, the development of more classic psoriatic ‘Perianal pseudoverrucous papules
lesions with silvery micaceous (mica‐like) scale elsewhere and nodules’ entity
on the body and the presence of nail pitting can help in ‘Perianal pseudoverrucous papules and nodules’ entity is
making the diagnosis. a rare condition that was first described in patients with
urostomies [3]. It was attributed to chronic irritation from
Mixed napkin dermatitis leakage of urine. This eruption has more recently been
Sometimes several of the previously described types of reported also to occur in the perianal area of children in
napkin dermatitis occur together, making diagnosis very association with chronic faecal soiling. In the reported
difficult. Irritant contact napkin dermatitis is commonly cases, stool leakage was due to severe constipation with
secondarily infected with C. albicans, and this may alter secondary encopresis or followed surgical colonic re‐
the appearance of the rash. If the rash is severe or present anastomosis in patients with Hirschsprung disease [4].
for more than 2–3 days, anticandidal agents such as nys- This entity is characterized by 2–8 mm, red, moist, flat‐
tatin, clotrimazole, econazole, ketoconazole or mupirocin topped round papules and papulonodules (Fig. 20.14). It
should be used in addition to low‐potency topical ster- is believed that the pathogenesis of this entity is similar to
oids, barrier creams and frequent napkin changes. that of Jacquet dermatitis in that it is a local response to
Additionally, it is often difficult to rule out candidiasis in chronic irritation. Pseudoverrucous papules and nodules
276 Section 4  Other Types of Dermatitis

Fig. 20.14  Perianal pseudoverrucous papules and nodules. Source:


Courtesy of Dr Neil Prose.
SECTION 4: OTHER TYPES

may be mistaken for cutaneous Crohn disease, granu-


loma gluteale infantum, condylomata acuminata or
OF DERMATITIS

Langerhans cell histiocytosis.

Perioral dermatitis with extrafacial


genital manifestations and metastatic
Crohn disease
Perioral dermatitis classically presents with noncome- Fig. 20.15  Metastatic Crohn disease of the vulvar area.
donal, acneiform papules in a perioral and periocular dis-
tribution. Histologically, these lesions resemble the first week of the onset of symptoms. The rash was charac-
granulomatous lesions of adult rosacea. The condition terized by confluent, sometimes tender, macular to
appears to be associated in some cases with a history of plaque‐type erythema involving part or all of the perineal
topical steroid use. Similar granulomatous lesions may region. The erythema was followed shortly by desquama-
appear on the genital surfaces [5]+. Patients with such tion. In fact, the authors found that this rash appeared
findings should also be screened for the possibility of more frequently than ‘strawberry’ tongue or reddened
inflammatory bowel disease because granulomatous oropharyngeal mucosa. They concluded that this perineal
lesions in the perioral, oral and genital areas, which may eruption, which may be mistaken for napkin rash, was a
be associated with labial or genital swelling, may r­ epresent valuable early clinical finding of Kawasaki disease, facili-
metastatic areas of Crohn disease (Fig. 20.15) [6]. tating more rapid diagnosis and treatment.

Kawasaki disease References


An erythematous, desquamating perineal eruption has 1 Haddock ES, Calame A, Shimizu C et al. Psoriasiform eruptions dur-
ing Kawasaki disease (KD): a distinct phenotype. J Am Acad
been noted as an early sign of Kawasaki disease, but a pso-
Dermatol 2016;75:69–76.e2.
riasis id reaction pattern is also possible [1]. This disease, 2 Uyeda K, Nakayasu K, Takaisski Y et al. Kaposi sarcoma‐like granu-
also known as mucocutaneous lymph node syndrome, is loma on diaper dermatitis. Arch Dermatol 1973;107:605–7.
an acute multisystemic illness of unknown aetiology that 3 Borglund E, Nordstrom G, Nyman CR. Classification of peristomal
skin changes in patients with urostomy. J Am Acad Dermatol
affects infants and young children. The principal clinical 1988;19:623–8.
signs include: (i) fever for more than 5 days; (ii) nonsup- 4 Goldberg NS, Esterly NB, Rothman KF et  al. Perianal pseudoverru-
purative cervical lymphadenopathy; (iii) conjunctival cous papules and nodules in children. Arch Dermatol 1992;128:240–2.
infection; (iv) reddened, cracked lips, ‘strawberry’ tongue 5 Urbatsch AJ, Frieden I, Williams ML et  al. Extrafacial and
­generalized  granulomatous periorificial dermatitis. Arch Dermatol
and erythema of the oropharyngeal mucosa; (v) oedema, 2002;138:1354–8.
erythema and desquamation of the hands and feet; and 6 Schrodt BJ, Callen JP. Metastatic Crohn’s disease presenting as chronic
(vi) a polymorphous exanthem. Although usually a self‐ perivulvar and perirectal ulcerations in an adolescent patient.
Pediatrics 1999;103:500–2.
limiting illness, cardiovascular abnormalities, particularly 7 Friter BS, Lucky AW. The perineal eruption of Kawasaki syndrome.
coronary artery aneurysm, may occur. This may lead to Arch Dermatol 1988;124:1805–10.
coronary thrombosis, stenosis and ultimately myocardial
infarction. Early treatment with intravenous gamma glob-
ulin and high‐dose aspirin may prevent these potentially
Neoplastic napkin conditions
fatal cardiovascular complications. Thus, early diagnosis Langerhans cell histiocytosis (formerly
of Kawasaki disease is crucial. Letterer–Siwe disease)
Friter and Lucky [7] studied 58 patients with Kawasaki This acute disseminative, potentially fatal form of
disease and found that 39 (67%) presented with a distinc- Langerhans cell histiocytosis (LCH) can present with
tive perineal eruption that usually occurred within the skin findings in the napkin area. Although this is a rare
Chapter 20  Napkin Dermatitis 277

which appears to be mediated in some affected patients


by SLC39A4 (ZIP4), an intestinal zinc/iron transporter
protein [2], or there may be a defective zinc transporter,
SLC30A2 (ZnT2) family, that affects the movement of zinc
into breast milk in affected mothers. [3]. Manifestations of
the disease typically present when the affected infant is
weaned from breastfeeding; breast milk is felt to be pro-
tective because it contains a zinc ligand‐binding protein
[4]. The transient acquired form is due to nutritional zinc
deficiency. It is seen in infants with severe malabsorption,
in premature infants who receive prolonged parenteral
alimentation [5] and in breastfed infants when the zinc
level in the mother’s milk is abnormally low [4].
Acrodermatitis enteropathica is characterized by a
periorificial and acral vesiculobullous eczematoid erup-
tion. Scaly, sharply demarcated, crusted plaques are

SECTION 4: OTHER TYPES


seen  in the folds of the napkin area, around the mouth,
nose and eyes and on the distal extremities. In chronic,

OF DERMATITIS
untreated cases, the lesions become lichenified and psori-
asiform in appearance. Secondary infection with bacteria
and C. albicans is frequent. Other clinical characteristics
include failure to thrive, photophobia, diarrhoea, alope-
cia, paronychia, nail dystrophy, irritability or listlessness.
Diagnosis is confirmed by a plasma zinc level lower than
Fig. 20.16  Langerhans cell histiocytosis. Note haemorrhagic satellite 50 μg/mL (normal values: 70–110 μg/mL). The alkaline
papules and intertriginous dermatitis. phosphatase, a zinc‐dependent metalloenzyme, is also
low in prolonged cases of acrodermatitis enteropathica.
The disease responds rapidly and quite dramatically to
treatment with zinc supplementation. The irritability or
disease, it should be considered in cases of intractable apathy usually disappears within 24 h after initiating
napkin dermatitis. This infantile form of LCH usually therapy.
occurs during the first year of life but may occur in chil- Biotin‐responsive multiple carboxylase deficiency is a
dren up to the age of 3 years. It often starts with an rare, recessively inherited, metabolic disorder that can
inguinal rash resembling seborrhoeic dermatitis. The present in infancy with a periorificial eruption similar to
inguinal eruption of infantile LCH, however, is often that of acrodermatitis enteropathica. Likewise, superin-
more erosive (Fig.  20.16) and fails to respond to local fection with C. albicans is common. Other manifestations
therapy. The infant then develops postauricular erosions of biotin deficiency include alopecia, seizures, ataxia and
and a scaly rash of the scalp and trunk. This eruption intermittent metabolic acidosis.
consists of yellowish to reddish‐brown infiltrated pap- Cystic fibrosis is yet another disease to keep in mind
ules, often with a purpuric component. Lesions may when one sees acrodermatitis enteropathica‐like skin
show haemorrhagic crusting. Petechiae and epidermal lesions. The main clinical features of cystic fibrosis are
atrophy may also be seen. The development of purpuric chronic lung disease, exocrine pancreatic insufficiency
nodules of the palms and soles is associated with a poor with malabsorption, retarded growth and hepatic dis-
prognosis. ease. In this condition, secreted mucus has an increased
Systemic manifestations of the disease include fever, viscosity and there is an abnormally high sodium concen-
anaemia, diarrhoea, thrombocytopenia, hepatospleno- tration in the sweat. The periorificial eruption is consid-
megaly, lymphadenopathy and skeletal tumours. A skin ered to be a manifestation of essential fatty acid and zinc
biopsy should be performed in order to confirm the diag- deficiency which is due to gastrointestinal malabsorption.
nosis in suspected cases. Other metabolic disorders associated with acrodermatitis
enteropathica‐like presentations include ornithine tran-
scarbamylase deficiency [6], nonketotic hyperglycinae-
Metabolic napkin conditions
mia [7], methylmalonic acidaemia [8] and maple syrup
Acrodermatitis enteropathica urine disease [9], as well as other organic acidurias. In
Acrodermatitis enteropathica is another rare disease that these cases, low‐protein diets to minimize excess patho-
should be considered in cases of atypical or persistent logical organic acids may result in secondary zinc defi-
napkin rash. This entity is caused by zinc deficiency. ciency and acrodermatitis enteropathica‐like eruptions.
There is an inherited form of the disease as well as an However, in some patients with maple syrup urine dis-
acquired, transient form. In the autosomal recessive ease and acrodermatitis enteropathica, zinc levels are nor-
inherited form of acrodermatitis enteropathica, there is an mal and the clinical course of the skin eruption appears to
inborn defect in the gastrointestinal absorption of zinc [1] be correlated with serum isoleucine levels [9].
278 Section 4  Other Types of Dermatitis

References
1 Van Wouwe JP. Clinical and laboratory diagnosis of acrodermatitis
enteropathica. Eur J Pediatr 1989;149:2–8.
2 Kury S, Dreno B, Bezieau S et  al. Identification of SLC39A4, a gene
involved in acrodermatitis enteropathica. Nat Genet 2002;31:239–40.
3 Michalczyk A, Varigos G, Catto‐Smith A et al. Analysis of zinc trans-
porter hZnT4(Slc30a4), gene expression in a mammary gland disor-
der leading to reduced zinc secretion into milk. Hum Genet 2003;
113:202–10.
4 Zimmerman AW, Hambridge M, Lepow ML et  al. Acrodermatitis in
breast‐fed premature infants, evidence for a defect of mammary zinc
secretion. Pediatrics 1982;69:176–83.
5 Arlette JP. Zinc and the skin. Pediatr Clin North Am 1983;30:583–96.
6 Lee JY, Chang SE, Suh CW et al. A case of acrodermatitis enteropathica
like dermatosis caused by ornithine transcarbamylase deficiency. J Am
Acad Dermatol 2002;46:965–7.
7 Samady JA, Schwartz RA, Shih LY. Acrodermatitis enteropathica‐like
eruption in an infant with nonketotic hyperglycinemia. J Dermatol
2000;27:604–8.
8 Howard R, Frieden IJ, Crawford D et  al. Methylmalonic acidemia,
cobalamin C type, presenting with cutaneous manifestations. Arch
SECTION 4: OTHER TYPES

Dermatol 1997;133:1563–6.
9 Giacoia GP, Berry GT. Acrodermatitis enteropathica‐like syndrome sec-
OF DERMATITIS

ondary to isoleucine deficiency during treatment of maple syrup urine


disease. Am J Dis Child 1993;147:954–6.

Other important napkin conditions Fig. 20.17  Herpes simplex virus infection. Source: Courtesy of Professor
Arnold Oranje.
Other vesiculobullous disorders
The maculopapular and vesicular lesions of varicella can
sometimes be more florid in areas of pre‐existing skin feet are affected, then child abuse by lowering a child into
inflammation. Thus, patients with a napkin dermatitis scalding hot water must be seriously considered. The
have a predilection for the development of varicella burned skin is usually sharply demarcated and involves
lesions in the napkin area. the convexities of the buttocks with sparing of the peria-
Uncommon systemic blistering diseases may present nal area. A thorough history should exclude the inciden-
with lesions in the anogenital region of napkin‐wearing tal ingestion of caustic laxatives by children, which may
children. These include neonatal herpes simplex present with findings suggestive of abusive immersion
(Fig.  20.17), hand, foot and mouth disease, chronic bul- injury because of the perianal erosive erythema that can
lous dermatosis of childhood, bullous mastocytosis, result [2,3].
incontinentia pigmenti, epidermolytic hyperkeratosis
and epidermolysis bullosa. Diagnosis is based on the References
1 Schanzer MC, Wilkin JK. Diaper dermatitis. Am Fam Physician
morphology of the lesions, their distribution outside the 1982;25:127–32.
napkin area and other diagnostic criteria. 2 Giacoia GP, Berry GT. Acrodermatitis enteropathica‐like syndrome
­secondary to isoleucine deficiency during treatment of maple syrup
urine disease. Am J Dis Child 1993;147:954–6.
Child abuse 3 Leventhal JM, Griffin D, Duncan KO et  al. Laxative‐induced
If the skin of the buttocks is red and blistered in a ‘dunk- ­dermatitis of the buttocks incorrectly suspected to be abusive burns.
ing’ pattern [1], and especially if the lateral aspects of the Pediatrics 2001;107:178–9.
 279

C HA PTER   21

Adolescent Seborrhoeic Dermatitis


Roselyn Kellen & Nanette Silverberg
Icahn School of Medicine at Mount Sinai, New York, NY, USA

Abstract probably a combination of individual predisposition, the presence


of Malassezia yeast on the skin and inflammatory response to the
yeast species. Physical examination reveals mildly erythematous
While seborrhoeic dermatitis is a common inflammatory skin
patches with yellowish‐white, greasy scales, typically affecting
disorder often observed in infants 0–3 months of age, it is also
­areas of the skin with increased sebum production such as the
seen in adolescents with onset at the age of puberty. Occasion-
scalp, eyebrows and nasolabial folds. The prognosis is favourable
ally, the condition is found in toddlers and school‐aged children
and treatment options include medications with antifungal, anti‐
as  well. Although the exact pathophysiology is unknown, it is

SECTION 4: OTHER TYPES


inflammatory and keratolytic properties.

OF DERMATITIS
Key points • The scalp, ears, eyebrows and nasolabial folds are most
commonly involved and l show erythematous patches with
yellowish‐white greasy scales.
• Paediatric seborrhoeic dermatitis is a biphasic illness with clinical
• Treatment can involve the use of agents with antifungal, anti‐
peaks in infancy and adolescence.
inflammatory and keratolytic properties.
• There is strong evidence for the involvement of Malassezia
• The prognosis is favourable; however, inflammatory
yeast.
symptoms may be chronic and require intermittent or ongoing
• There also appears to be individual predisposition towards an
intervention.
inflammatory response.

M. restricta [18–21] although there may be geographical


Introduction/history. Seborrhoeic dermatitis (SD)
variation [22].
(verna­
­ cular: dandruff or cradle cap) is a chronic
It is important to note the detrimental effects of SD on
­infla­mmatory skin disease caused by Malassezia yeasts,
quality of life. In infancy, SD can be generalized and asso-
which are commensal eukaryotic organisms found in
ciated pruritus can interfere with sleep and daily activi-
the normal skin flora of the superficial sebaceous folli-
ties if severe. Children and adolescents can suffer from
cle [1]. Knowledge of the association of yeast with skin
social stigma and feelings of low self‐esteem. In turn, all
diseases goes back as far as the nineteenth century
these factors also place a tremendous amount of stress on
when yeast cells and filaments were found in the scales
their caregivers. As there is no cure for dermatitis, it is
of patients with pityriasis versicolor (tinea versicolor)
essential to establish a management plan to keep the dis-
[2] and M. furfur was identified as the organism associ-
ease under control and address flares in a timely manner.
ated with dandruff [3,4].
In the early 1900s, Sabouraud separated Malassezia
­furfur, which forms filaments, and Pityrosporum malassezii, Epidemiology. SD is a relatively common skin disorder,
which does not form filaments and is found in dandruff estimated to have a prevalence of approximately 11.6%
and scales from patients with SD, into separate genera [5]. in  the general population (with males more commonly
Pityrosporum malassezii was then re‐named Pityrosporum affected than females) and 3–5% in the USA [23,24].
ovale [6], which was then confirmed to be identical to Disease activity peaks at two time points during child-
Pityrosporum orbiculare [7]. The common denominator of hood: the first 3 months of life (when it is known as infan-
successful treatments for SD was antifungal activity, and tile SD or cradle cap) and during puberty, time periods of
in 1984 it was shown that P. ovale was the causative agent increased sebaceous gland activity [1].
of SD [8,9]. The genus Malassezia includes 14 species, of In contrast to infantile SD, which is self‐limiting and
which the majority are lipophilic [1]. The species most resolves by 1 year of age [25], adolescent SD tends to be
commonly associated with SD are M. globosa [10–17] and more chronic [26]. In an Australian study of over 1000

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
280 Section 4  Other Types of Dermatitis

children ≤5 years of age, the prevalence was 10.4% in boys be of benefit [36,45]. Interestingly, M. furfur appears to
and 9.5% in girls [27]. Prevalence was highest during the produce pityriacitrin, which acts as a UV filter [46].
first 3 months of life and rapidly decreased after age 1 Nutrients in sweat such as amino acids might also play a
[27]. Of those with disease, 72% had minimal to mild role as one study showed that the addition of glycine
severity [27]. Berk and Scheinfeld reported that up to 70% stimulated the growth of M. furfur [47]. The virulence fac-
of infants have SD in the first 3 months of life [23]. tor might be production of phospholipases, a known vir-
Aside from the two peaks during childhood, SD is also ulence factor of  Candida albicans [48], which results in
noted in school‐aged children and later jumps in fre- disruption of epidermal lipids and therefore the barrier
quency among adults in their third and fourth decades in protection of the epidermis [1]. Other contributors could
whom sebum production is decreased [1,28–30]. SD is also be emotional stress, sleep deprivation and nutritional
more common in those with Parkinson disease, cranial deficiencies [33]. DeAngelis et  al. suggested that fungal
nerve palsies [31] and drug‐induced parkinsonism [32], metabolites such as free fatty acids released by sebaceous
and in patients taking haloperidol decanoate, lithium, triglycerides trigger the clinical findings of dandruff [41].
buspirone and chlorpromazine [23], calling into question They found that a Malassezia metabolite called oleic acid
neurogenic factors [33]. SD is one of the most common was able to induce scaling similar to that of dandruff but
dermatological disorders in patients with human immu- only in patients known to have dandruff [41]. The authors
SECTION 4: OTHER TYPES

nodeficiency virus (HIV), with incidence rates as high as attributed this to underlying defects in the permeability
85% [31], and is often severe, especially in patients with barrier function, which brings into play the idea of indi-
OF DERMATITIS

low CD4 counts [34]. SD is also associated with depres- vidual susceptibility to developing disease [41]. Further
sion, acute traumatic spinal cord injuries [35,36], familial research can help us understand why certain species are
amyloidotic polyneuropathy [37] and children with pathogenic or virulent, what the virulence factors are, and
Down syndrome [38] and Leiner’s phenotype [39]. clarify the mechanism behind the inflammatory process
generated in the skin [1].
Pathogenesis. The role of Malassezia in SD has been
c­ ontroversial; as reviewed by Gupta et al., there are con- Clinical features. The name of this skin condition is
flicting data on the number of yeasts in affected versus actually a misnomer because seborrhoea, or increased
­
nonaffected skin [36]. Yet the efficacy of antifungal treat- sebum production from sebaceous glands, is not a feature
ments is strong proof that these yeasts play a central role of seborrhoeic dermatitis [49]. Rather, a more appropriate
in triggering inflammation at the centre of disease patho- name would be ‘dermatitis of the sebaceous areas’ [49].
genesis [40]. That being said, these are commensal yeasts Since most species of Malassezia depend on lipids for
found in normal flora [31], so why do only certain people growth [50], they have a preference for seborrhoeic
develop disease? There are probably multiple compo- regions with increased sebum production [1,36].
nents involved in SD: the presence of Malassezia yeast, The classic presentation consists of symmetrical,
the role of sebum production and inflammation, and an poorly‐demarcated erythematous patches with white to
individual predisposition, the latter mechanisms not yellow greasy scales [23], but there is considerable varia-
being mutually exclusive [41]. tion in the degree of erythema and scaling [36]. Dandruff
The clinical manifestations of SD are probably the result is probably a mild form of SD characterized by dry flakes
of an inflammatory reaction to the yeast [1]. As reviewed (Fig.  21.1) confined to the scalp with minimal to no
by Gaitanis et  al., Malassezia yeast cells interact with
keratinocytes [42] and immune cells including dendritic
cells, macrophages, eosinophils and neutrophils, with
various effects on levels of cytokines, chemokines and
adhesion molecules [1]. The presence or absence of the
lipid capsule of Malassezia spp. is associated with an
increase or decrease in the production of interleukins
(IL)‐1α, IL‐6, IL‐8 and IL‐10 [43]. Skin biopsies from
patients with SD showed higher levels of markers for
CD4, human leucocyte antigen (HLA)‐DR, natural killer
(NK)1, CD16 and CD54 (ICAM‐1) cells compared to
healthy volunteers [8]. The same was true for markers for
C1q, IL‐1α, IL‐1β, TNF‐α, IFN‐γ, IL‐2, IL‐4, IL‐6, IL‐10,
IL‐12 and IL‐4 [8]. Moreover, the various Malassezia spp.
can stimulate different profiles of these inflammatory
cytokines, which might explain the numerous clinical
presentations of Malassezia‐associated skin conditions
[1,42]. In addition, certain species might be preferentially
found in specific body regions [44].
Although sebum production on the face is lower in
autumn compared to summer, flares are often seen dur- Fig. 21.1  Teenage female with seborrhoeic dermatitis of the scalp in
ing the autumn and winter, suggesting that sunlight may association with androgenic alopecia and polycystic ovaries.
Chapter 21  Adolescent Seborrhoeic Dermatitis 281

Differential diagnosis. Patients with simultaneous fea-


tures of seborrhoeic ­dermatitis and psoriasis, known as
sebopsoriasis, can be difficult to diagnose [1]. The same is
true for severe SD that presents as erythematous plaques
with thicker scale [31]. Clinicians should look for extracu-
taneous signs of psoriasis, such as nail pitting or other
nail involvement as well as joint disease. Yellow, greasy
scales are also more suggestive of SD, whereas sharply
demarcated erythematous plaques with a silvery scale
are more consistent with psoriasis. The distribution of
lesions can also be helpful given that psoriasis tends to
favour the extensor surfaces.
Atopic dermatitis can be difficult to discern from SD,
particularly in infancy; the two conditions often coexist
and might be associated [53–55]. Reports document infants
with infantile seborrhoeic dermatitis (ISD) who go on to

SECTION 4: OTHER TYPES


develop atopic dermatitis 4 months later [55]. This overlap
Fig. 21.2  African‐American male with nummular hypopigmented
can be seen in adolescents and adults as well on occasion.

OF DERMATITIS
seborrhoeic dermatitis, sometimes termed petaloid seborrhoeic dermatitis
due to the flower‐like shape in some cases. Treatment is similar in all cases, The distinction between the two is primarily clinical: one
but pigmentation can take months to return. can consider the age of the patient, the presence or absence
of pruritus, oozing and weeping, response to topical medi-
cations, family history and distribution of the lesions [55].
In atopic dermatitis, skin lesions are pruritic and diffuse
inflammation [1]. Adolescents with SD may report mild and involve the face, trunk and extensor surfaces in
pruritus [26] and paediatric cases have been associated infants, and flexural surfaces in children [53]. On the other
with posterior neck adenopathy, although the latter is not hand, involvement of the axilla and anterior neck is more
specific for SD and should prompt screening for active suggestive of ISD [55]. Chronic atopic dermatitis can lead
infections of the head and neck such as tinea capitis and/or to lichenified skin with associated pigment changes [53].
upper respiratory infections [28]. Patients with atopic dermatitis usually have a parent or
In adolescents, the most commonly affected areas are sibling with atopic disorders including asthma and aller-
the scalp, external ears, eyelids, eyebrows, moustache gic rhinitis [53,55]. They may also have high levels of
and beard areas, nasolabial folds, and retroauricular immunoglobulin E (IgE) and eosinophils [53]. Interestingly,
folds, and occasionally nuchal and neck with patches and Malassezia might also play a role in atopic dermatitis, espe-
plaques that are thinner than their adult counterparts [26]. cially of the head and neck; such patients are more likely
Occasionally, the upper/mid chest and back are involved, to have Malassezia‐specific IgE compared to patients with
and less commonly the axillae and inframammary and atopy but no head and neck dermatitis [56,57].
perineal folds are affected [26]. Patients who are dark‐ Williams et al. conducted a cross‐sectional study of 300
skinned, especially African Americans, may manifest children to determine the most common causes of scalp
with petaloid seborrhoeic dermatitis which is hypopig- scaling [28]. Overall, the most common aetiologies were
mented and can be concentrated on the cheeks and fore- seborrhoeic dermatitis (54.5%), undetermined (30.3%) and
head areas. This may mimic pityriasis alba, vitiligo and atopic dermatitis (24.2%). The most common diagnoses
mycosis fungoides, but is typically located in seborrhoeic in  children <2 years of age were seborrhoeic dermatitis
areas and has typical histology (Fig. 21.2). and atopic dermatitis. For children between the ages of 2
There are several conditions that can present alongside and  10, the most common causes were unknown aetiol-
SD that clinicians should be aware of. Malassezia folliculi- ogy, seborrhoeic dermatitis and atopic dermatitis/eczema.
tis is characterized by erythematous papules or (less com- Tinea capitis is also a common concern in the differential
monly) pustules on the back, chest and upper arms, which diagnosis although the incidence of tinea capitis begins to
can be pruritic or asymptomatic [51,52]. Biopsy results wane during adolescence. Potassium hydroxide (KOH)
would illustrate the presence of abundant yeast growth in preparation of skin scrapings from these patients would
hair follicles but there is also an accompanying inflamma- reveal hyphae (whereas pseudohyphae would be indica-
tory infiltrate composed of lymphocytes, histiocytes and tive of candidiasis) [23,58,59]. A retrospective chart review
neutrophils [51]. Similar to SD, Malassezia folliculitis is to determine the causes of scalp hyperkeratosis and/or
treated with topical antifungals, with systemic therapy alopecia in inner‐city children [58] identified the records of
reserved for severe, refractory cases  [51,52]. Aside from 164 children aged 0–17, of whom 75 were black and 56
folliculitis, patients may have blepharitis leading to mei- were Hispanic/Latino. Of the children with scalp hyper-
bomian gland blockage and abscess, otitis externa and keratosis, 60% had tinea capitis. Of the children with
simultaneous acne vulgaris [33]. Patients should also be hyperkeratosis and alopecia, 82.1% had tinea capitis.
examined for signs of secondary infection, most com- Interestingly, another cross‐sectional study also found that
monly due to Staphylococcus aureus. only 10.5% of children who had scalp scaling combined
282 Section 4  Other Types of Dermatitis

with head and neck adenopathy had tinea capitis in their


Treatment and prevention. As shown in Box 21.1, there
population, which was mostly Caucasian [28]. Three per-
are many treatment options for adolescents and adults
cent of patients had cultures positive for a dermatophyte;
with seborrhoeic dermatitis, ranging from topical to sys-
all these cultures grew Trichophyton tonsurans and all were
temic agents, which can be divided into four groups: anti-
from black patients.
fungals (azoles, selenium sulfide), anti‐inflammatory
In one retrospective chart review where treatment out-
agents (topical corticosteroids, topical calcineurin inhibi-
comes for paediatric patients with skin of colour were
tors), keratolytics (salicylic acid, urea, ammonium lactate)
evaluated, 60.6% of patients were African‐American and
and others (tar, tea tree oil) [23,40]. Table  21.1 lists the
28.2% were Hispanic [59]. Eighty patients (of 84 identified
therapies that are approved by the US Food and Drug
cases of tinea capitis) were started on griseofulvin sus-
Administration (FDA) for use in the paediatric popula-
pension and/or crushed tablets and 61 had complete
tion [26]. As many therapies are used off‐label in children,
responses. Of the 19 griseofulvin failures, 15 were treated
both sets of agents are herein reviewed.
and cleared in response to fluconazole suspension, terbi-
Several agents work through more than one mechanism,
nafine sprinkles or oral itraconazole.
which makes them especially useful for compliance in
Erythema of the central face can be misdiagnosed as
adolescents. Ketoconazole has antibacterial, anti‐
rosacea, systemic lupus erythematosus or discoid lupus
inflammatory, sebostatic and antiproliferative effects [61].
SECTION 4: OTHER TYPES

[23]. The differential also includes drug eruptions, contact


Tar is anti‐inflammatory and keratolytic and is available
dermatitis, erythrasma and, less commonly, dermatomy-
as a shampoo or in precombined topical preparation [62].
OF DERMATITIS

ositis, cutaneous lymphoma, Langerhans cell histiocyto-


Sulphur is antifungal, antibacterial and keratolytic, and
sis and vitamin B or zinc deficiency [23,33].
can be combined with sodium sulfacetamide or salicylic
Hypopigmented seborrhoeic dermatitis can mimic
acid [63].
mycosis fungoides, pityriasis alba and vitiligo.
The most commonly used agents for SD are antifungal
Children can present with a generalized eczematous
agents, available in both topical and systemic form,
eruption of SD similar to that found in Wiskott–Aldrich
which come in two classes: imidazoles (e.g. ketoconazole,
syndrome, however purpura and petechiae, as well as
other systemic symptoms, will be absent [23]. Individuals
with rapid onset of widespread lesions in adulthood
should be tested for HIV [23]. Box 21.1  Different classes of medications used for the
treatment of adolescent and adult seborrhoeic dermatitis
Histological findings. SD is characterized by changes in Antifungal Anti‐ Keratolytic Other
the stratum corneum that can be seen throughout the inflammatory
scalp, even in areas without flaking [60]. Findings include
parakeratosis, the presence of lipids within corneocytes, Azoles Corticosteroids Salicylic acid Tea tree oil
fewer desmosomes and excess of disorganized lipids in Terbinafine Tacrolimus Coal tar Crude honey
the cells [60]. Ciclopirox Pimecrolimus Urea Hormonal
therapies that
Biopsy is rarely performed unless there is suspicion for
block
a disease such as Langerhans cell histiocytosis, but find- androgens
ings include neutrophils in the scale crust near the follicu- Zinc Nonsteroidal Ammonium Borage oil
lar ostia [33]. Individuals with acquired immune pyrithione creams (e.g. lactate
deficiency syndrome (AIDS)‐induced SD can have par- Promiseb®)
akeratosis, necrotic keratinocytes, and plasma cells visible Selenium More
sulfide frequent
in the dermis [33]. Special stains can be used to identify
washing
the yeast cells associated with keratinocytes [33]. Hyphae Sulfacetamide
are an indicator of dermatomycosis whereas short hyphae
and spores, so‐called ‘meatballs and spaghetti’, are con- Source: Adapted from Poindexter et al. 2009 [26].
sistent with tinea versicolor [33].

Table 21.1  FDA‐approved therapies for paediatric seborrhoeic dermatitis

Therapy Brand name Frequency of use Age group

Ciclopirox 0.77% gel Loprox® Twice a day for 4 weeks ≥16 years of age
Ciclopirox 1% shampoo Twice a week for 4 weeks ≥16 years of age
Ketoconazole 2% foam Exina® Twice a day for 4 weeks ≥12 years of age
Ketoconazole 2% gel Xelogel® Daily for 2 weeks ≥12 years of age
Selenium sulfide 1% or 2.5% shampoo Selsun Twice a week for 2 weeks ≥2 years of age
Sulfacetamide 10% lotion, cream, gel, wash, foam Carmol Scalp Treatment®, Klaron®, Ovace® Daily for 8–10 days ≥12 years of age
Sulfacetamide/sulphur 10%/5% wash, cream, or gel Plexion®, Rosac®, Rosula® Daily or twice daily ≥12 years of age
Chapter 21  Adolescent Seborrhoeic Dermatitis 283

econazole, clotrimazole, miconazole) and triazoles (flu- adverse effect, irritative dermatitis, was thought to be
conazole) [40]. Azoles inhibit the fungal P450 enzyme, treatment related [77]. In 1996, Zeharia et al. reported the
thereby blocking the formation of ergosterol from lanos- results of evaluating the use of bifonazole shampoo for
terol, which is required for cell membrane synthesis [40]. scalp seborrhoeic dermatitis in 36 patients between the
The end result is a decrease in growth yet many antifun- ages of 1 month and 10 years [73]. Ninety seven percent of
gals have also been shown to have anti‐inflammatory the patients improved with 70.6% showing complete
properties [24,40]. One of the best studied, and most fre- response. Although two patients reported burning of the
quently used agents, is ketoconazole. Currently, there is a eyes when using the shampoo and one patient had a tem-
2% ketoconazole gel approved for children ≥12 years of porary generalized rash (that resolved after a few hours),
age with seborrhoeic dermatitis [64]. The double‐blind there were no major side‐effects. Van Esso et al. found that
trial enlisted 459 patients over the age of 12 with moderate 2% sertaconazole cream was well tolerated in 16 children
to severe seborrhoeic dermatitis who applied the gel once (aged 2–16) being treated for cutaneous mycoses with no
a day for 14 days [64]. The proportion of patients that were local or systemic adverse effects observed [78].
effectively treated was 25.3% in the group that received Anti‐inflammatory agents include topical corticosteroids
the gel compared to 13.9% in the placebo group [64]. and nonsteroidal creams. Corticosteroids are available
Collectively, in the three trials that were carried out, 7% of in  numerous strengths and vehicles, such as ointments,

SECTION 4: OTHER TYPES


patients reported at least one adverse event with the most creams, lotions, solutions, shampoos, gels and foam
common being burning upon application (4%) [64]. [26,33]. Although off‐label, the topical calcineurin inhibi-

OF DERMATITIS
A large, double‐blind, placebo‐controlled study com- tors tacrolimus [79] and pimecrolimus have been used for
pared the use of 2% ketoconazole shampoo to placebo in SD in adults, especially for facial SD [33], and are espe-
575 adult patients with SD [65]. Results showed that topi- cially helpful for patients of colour with petaloid sebor-
cal application of 2% ketoconazole shampoo was more rhoeic dermatitis of the mid‐face. Side‐effects include
effective in clearing scalp SD and dandruff and was well local pruritus and burning [79,80]. Off‐label usage should
tolerated. Furthermore, the 312 patients who responded be weighed against the FDA‐issued black‐box warning for
were instructed to use the shampoo as prophylaxis for topical calcineurin inhibitors concerning a risk of increased
6  months and fewer of these patients suffered relapses malignancies and age restriction to 2 years and over [81].
(19%) compared to the placebo group (47%) [65]. Safety A nonsteroidal anti‐inflammatory cream known as
data in the paediatric population are more limited and Promiseb® was extremely well tolerated in one double‐
often extrapolated from use in other paediatric disorders. blind, placebo‐controlled study of 42 paediatric patients
The use of 2% ketoconazole shampoo for tinea capitis was with cradle cap, with no adverse events related to either
evaluated in 16 black children (ages 3–6), and no patient treatment group [69].
or parent reported adverse effects [66]. No major safety Keratolytic agents can help remove scale to improve
concerns were noted in a retrospective analysis evaluat- absorption of other topical medications such as topical
ing 2% ketoconazole shampoo prophylaxis for children corticosteroids [26]. Salicylic acid, the agent most com-
(aged 1–21) at high risk for tinea capitis [67]. Wannanukul monly used, can be found as a shampoo or as part of a
et  al. compared the efficacy of 2% ketoconazole cream topical mixture [26]. A combination of keratolytics and
and 1% hydrocortisone cream for infants with sebor- corticosteroids has been found to be very effective for
rhoeic dermatitis [68]. Both therapies were comparable, treatment of SD [36]. The efficacy of tar had been noted as
and again, no major safety concerns were noted with early as the nineteenth century [82] and it is available in
ketoconazole. Of note, keratolytic shampoos (e.g. zinc many preparations, either alone or mixed with other topi-
pyrithione, salicylic acid) are not recommended in infants cal compounds [31]. However, its use is limited by con-
because of the potential for systemic absorption [69]. cerns regarding its safety, notably a possible association
Other topical therapies that have been used in adults with squamous cell carcinoma [83] and side‐effects such
include flutrimazole gel [70], topical clotrimazole, topical as irritation to the skin [23]. However, these agents are
miconazole [71], 2% sertaconazole cream [72], bifonazole typically reserved for severe cases and adolescent or adult
shampoo [73], ciclopirox olamine shampoo [74], zinc patients with thick scale.
pyrithione (which also has keratolytic activity) [33] and The last category comprises other agents that do not fall
selenium sulfide. Unfortunately, scant safety data are into the above three mechanisms. These include MAS04D
available in children and infants to support these thera- (Sebclair®), a steroid‐free herbal cream found to be effec-
pies. A randomized, double‐blinded study evaluated the tive for mild to moderate SD of the face [84], crude honey
use of 1% selenium sulfide shampoo and 1% ciclopirox [85], 5% tea tree (Melaleuca) oil shampoo for dandruff [86],
shampoo, in combination with griseofulvin, for children borage oil containing 24% gamma linolenic acid [87], lith-
(aged 1–11) with tinea capitis [75]. No major safety con- ium gluconate ointment [88], and conflicting evidence
cerns were noted and no doses were missed because of regarding the efficacy of topical metronidazole [89,90].
adverse effects. A randomized trial comparing 1% and However, none of these therapies has been studied exten-
2.5% selenium sulfide in children with tinea capitis (aged sively in the paediatric population. One agent that was
1–15) did not report major safety concerns [76]. A small studied in 48 children with ISD was borage oil, on the
phase II study in 21 patients (aged 3 months to 9 years) basis that ISD is associated with impaired function of an
found that ciclopirox cream 1% was effective for der- enzyme that desaturates linoleic acid into a product that
matomycosis but also had a good safety profile: only one is found in borage oil [87]. All the children were treated
284 Section 4  Other Types of Dermatitis

successfully with twice‐daily applications of the oil for 9 Shuster S. The aetiology of dandruff and the mode of action of
­therapeutic agents. Br J Dermatol 1984;111:235–42.
10–12 days. They were maintained on a prophylactic regi-
10 Gupta AK, Kohli Y, Summerbell RC, Faergemann J. Quantitative
men of oil application 2–3 times/week for 6–7 months, ­culture of Malassezia species from different body sites of individuals
after which no child suffered relapses. Most importantly, with or without dermatoses. Med Mycol 2001;39:243–51.
no side‐effects occurred. There are limited in vitro data 11 Nakabayashi A, Sei Y, Guillot J. Identification of Malassezia species
isolated from patients with seborrhoeic dermatitis, atopic dermatitis,
that cinnamic acid inhibits cell growth of M. ovalis [91]. pityriasis versicolor and normal subjects. Med Mycol 2000;38:337–41.
Interestingly, although topical oils (such as olive oil) may 12 Tajima M, Sugita T, Nishikawa A, Tsuboi R. Molecular analysis of
not be physically harmful, Siegfried and Glenn point out Malassezia microflora in seborrheic dermatitis patients: comparison
that the use of oils is not advisable in children because with other diseases and healthy subjects. J Invest Dermatol 2008;
128:345–51.
saturated fatty acids might precipitate Malassezia over- 13 Gaitanis G, Velegraki A, Alexopoulos EC et  al. Distribution of
growth and excess unsaturated fatty acids could trigger Malassezia species in pityriasis versicolor and seborrhoeic dermatitis
inflammation and scaling [92]. in Greece. Typing of the major pityriasis versicolor isolate M. globosa.
Br J Dermatol 2006;154:854–9.
Dandruff is usually treated with medicated shampoos 14 Arsic Arsenijevic VS, Milobratovic D, Barac AM et al. A laboratory‐
such as 1% or 2% ketoconazole (≥12 years of age), ciclopirox based study on patients with Parkinson’s disease and seborrheic der-
shampoo (≥16 years of age) and 2.5% selenium sulfide matitis: the presence and density of Malassezia yeasts, their different
(≥2 years of age) [26,36]. Interestingly, clinical improvement species and enzymes production. BMC Dermatol 2014;14:5.
15 Hedayati MT, Hajheydari Z, Hajjar F et al. Identification of Malassezia
SECTION 4: OTHER TYPES

is associated with a reduction in yeast levels on the scalp species isolated from Iranian seborrhoeic dermatitis patients. Eur Rev
but the absolute number of organisms does not appear to Med Pharmacol Sci 2010;14:63–8.
OF DERMATITIS

correlate with disease severity [36,93]. According to one 16 Zarei‐Mahmoudabadi A, Zarrin M, Mehdinezhad F. Seborrheic der-
matitis due to Malassezia species in Ahvaz, Iran. Iran J Microbiol
study, treatment with zinc pyrithione shampoo corrected
2013;5:268–71.
the histological abnormalities in the stratum corneum [60]. 17 Sosa Mde L, Rojas F, Mangiaterra M, Giusiano G. [Prevalence of
For refractory disease in severely affected adolescents, Malassezia species associated with seborrheic dermatitis lesions in
isotretinoin might rarely be required to suppress sebaceous patients in Argentina]. Rev Iberoam Micol 2013;30:239–42.
18 Crespo Erchiga V, Ojeda Martos AA, Vera Casano Aet  al. [Isolation
gland activity [31]. Of course it is crucial to keep in mind and identification of Malassezia spp. in pityriasis versicolor, sebor-
the teratogenicity of this medication, mucocutaneous side‐ rheic dermatitis and healthy skin.]. Rev Iberoam Micol1999;
effects (alopecia, xerosis, cheilitis, conjunctivitis, urethritis) 16(S):S16–21.
and other systemic side‐effects [31]. In females with excess 19 Prohic A. Distribution of Malassezia species in seborrhoeic dermatitis:
correlation with patients’ cellular immune status. Mycoses 2010;
male hormone, addressing the underlying endocrinopathy 53:344–9.
can reduce seborrhoeic dermatitis severity. 20 Lee YW, Byun HJ, Kim BJ et al. Distribution of malassezia species on
Prevention strategies include adequate hygiene (e.g. the scalp in korean seborrheic dermatitis patients. Ann Dermatol
2011;23:156–61.
increased washing) and cleansing of high‐risk regions to 21 Gemmer CM, DeAngelis YM, Theelen B et  al. Fast, noninvasive
remove excess oil [31]. Sunlight appears to improve SD method for molecular detection and differentiation of Malassezia
but caution against too much sun exposure is necessary yeast species on human skin and application of the method to dan-
[31]. As described previously, the use of 2% ketoconazole druff microbiology. J Clin Microbiol 2002;40:3350–7.
22 Prohic A, Jovovic Sadikovic T, Krupalija‐Fazlic M, Kuskunovic‐
shampoo was also shown to reduce relapses [65]. There is Vlahovljak S. Malassezia species in healthy skin and in dermatologi-
also evidence that 2% miconazole can be applied twice a cal conditions. Int J Dermatol 2016;55:494–504.
month for prophylaxis against recurrence [94]. 23 Berk T, Scheinfeld N. Seborrheic dermatitis. P T 2010;35:348–52.
24 Wolff K, Goldsmith L, Katz SI et  al. Fitzpatrick’ s Dermatology in
General Medicine, 7th edn. New York: McGraw‐Hill, 2008:219–25.
Conclusions. Seborrhoeic dermatitis is a benign, self‐ 25 Singleton JK. Pediatric dermatoses: three common skin disruptions in
limited skin condition of the scalp and seborrhoeic infancy. Nurse Pract 1997;22:32–3, 7, 43–4 passim.
gland‐containing areas of the skin. Biphasic infantile and 26 Poindexter GB, Burkhart CN, Morrell DS. Therapies for pediatric
adolescent peaks exist. A variety of topical agents seborrheic dermatitis. Pediatr Ann 2009;38:333–8.
27 Foley P, Zuo Y, Plunkett A et al. The frequency of common skin condi-
including antifungals have been used to improve clinical tions in preschool‐aged children in Australia: seborrheic dermatitis
appearance of this chronic condition. and pityriasis capitis (cradle cap). Arch Dermatol 2003;139:318–22.
28 Williams JV, Eichenfield LF, Burke BL et al. Prevalence of scalp scaling
References in prepubertal children. Pediatrics 2005;115:e1–6.
1 Gaitanis G, Magiatis P, Hantschke M et al. The Malassezia genus in 29 Cotterill JA, Cunliffe WJ, Williamson B, Bulusu L. Age and sex
skin and systemic diseases. Clin Microbiol Rev 2012;25:106–41. ­variation in skin surface lipid composition and sebum excretion rate.
2 Robin C. Histoire naturelle des végétaux parasites qui croissent sur Br J Dermatol 1972;87:333–40.
l’homme et les animaux vivants. Paris: Baillière,1853. 30 Johnson MLT. Skin Conditions and Related Need for Medical Care
3 Malassez L. Notes sur le champignon de la pilade. Arch Physiol Norm among Persons 1–74 Years US, 1971–1974. Series 11, Data from the
Pathol 1874;1:203–12. National Health Survey November, No. 212, DHEW Pub No. (PHS)
4 Hay RJ. Malassezia, dandruff and seborrhoeic dermatitis: an overview. 79–1660. Hyattsville, MD: U.S. Department of Health, Education,
Br J Dermatol 2011;165(suppl 2):2–8. and  Welfare, Public Health Service, National Center for Health
5 Sabouraud R. Maladies du cuir chevelu. II. Les maladies desquamatives. Statistics, 1978.
Paris: Masson & Cie, 1904. 31 Johnson BA, Nunley JR. Treatment of seborrheic dermatitis. Am Fam
6 Castellani A, Chalmers AJ. A Manual of Tropical medicine, 2nd edn. Physician 2000;61:2703–10, 2713–14.
London: Baillière, Tindall and Cox, 1913. 32 Binder RL, Jonelis FJ. Seborrheic dermatitis in neuroleptic‐induced
7 Faergemann J. Pityrosporum infections. In: Elewski BE (ed.) Cutaneous parkinsonism. Arch Dermatol 1983;119:473–5.
Fungal Infections. New York: Igaku‐Shoin, 1992. 33 Schwartz RA, Janusz CA, Janniger CK. Seborrheic dermatitis:
8 Faergemann J, Bergbrant IM, Dohse M et  al. Seborrhoeic dermatitis an overview. Am Fam Physician 2006;74:125–30.
and Pityrosporum (Malassezia) folliculitis: characterization of inflam- 34 Munoz‐Perez MA, Rodriguez‐Pichardo A, Camacho F, Colmenero
matory cells and mediators in the skin by immunohistochemistry. MA. Dermatological findings correlated with CD4 lymphocyte
Br J Dermatol 2001;144:549–56. counts  in a prospective 3 year study of 1161 patients with human
Chapter 21  Adolescent Seborrhoeic Dermatitis 285

immunodeficiency virus disease predominantly acquired through 63 Gupta AK, Nicol K. The use of sulfur in dermatology. J Drugs
intravenous drug abuse. Br J Dermatol 1998;139:33–9. Dermatol 2004;3:427–31.
35 Rubin‐Asher D, Zeilig G, Klieger M et al. Dermatological findings fol- 64 XOLEGEL [package insert]. Research Triangle Park, NC: Stiefel
lowing acute traumatic spinal cord injury. Spinal Cord 2005;43:175–8. Laboratories, 1981.
36 Gupta AK, Batra R, Bluhm R et  al. Skin diseases associated with 65 Peter RU, Richarz‐Barthauer U. Successful treatment and prophylaxis
Malassezia species. J Am Acad Dermatol2004;51:785–98. of scalp seborrhoeic dermatitis and dandruff with 2% ketoconazole
37 Rocha N, Velho G, Horta M et al. Cutaneous manifestations of familial shampoo: results of a multicentre, double‐blind, placebo‐controlled
amyloidotic polyneuropathy. J Eur Acad Dermatol Venereol trial. Br J Dermatol 1995;132:441–5.
2005;19:605–7. 66 Greer DL. Successful treatment of tinea capitis with 2% ketoconazole
38 Ercis M, Balci S, Atakan N. Dermatological manifestations of 71 Down shampoo. Int J Dermatol 2000;39:302–4.
syndrome children admitted to a clinical genetics unit. Clin Genet 67 Bookstaver PB, Watson HJ, Winters SD et al. Prophylactic ketocona-
1996;50:317–20. zole shampoo for tinea capitis in a high‐risk pediatric population.
39 Weston WL. Cutaneous manifestations of defective host defenses. J Pediatr Pharmacol Ther 2011;16:199–203.
Pediatr Clin North Am 1977;24:395–407. 68 Wannanukul S, Chiabunkana J. Comparative study of 2% ketocona-
40 Gupta AK, Nicol K, Batra R. Role of antifungal agents in the treatment zole cream and 1% hydrocortisone cream in the treatment of infantile
of seborrheic dermatitis. Am J Clin Dermatol 2004;5:417–22. seborrheic dermatitis. J Med Assoc Thai 2004;87(suppl 2):S68–71.
41 DeAngelis YM, Gemmer CM, Kaczvinsky JR et al. Three etiologic fac- 69 David E, Tanuos H, Sullivan T et  al. A double‐blind, placebo‐con-
ets of dandruff and seborrheic dermatitis: Malassezia fungi, seba- trolled pilot study to estimate the efficacy and tolerability of a non-
ceous lipids, and individual sensitivity. J Investig Dermatol Symp steroidal cream for the treatment of cradle cap (seborrheic dermatitis).
Proc 2005;10:295–7. J Drugs Dermatol 2013;12:448–52.
42 Watanabe S, Kano R, Sato H et al. The effects of Malassezia yeasts on 70 Noguera J, Leris E, Alguero M et al. [Review of the clinical efficacy of

SECTION 4: OTHER TYPES


cytokine production by human keratinocytes. J Invest Dermatol flutrimazole gel in the treatment of dandruff and/or seborrheic der-
2001;116:769–73. matitis.]. Rev Iberoam Micol 1998;15:28–32.

OF DERMATITIS
43 Thomas DS, Ingham E, Bojar RA, Holland KT. In vitro modulation of 71 Okokon EO, Verbeek JH, Ruotsalainen JH et al. Topical antifungals for
human keratinocyte pro‐ and anti‐inflammatory cytokine production seborrhoeic dermatitis. Cochrane Database Syst Rev 2015;5:Cd008138.
by the capsule of Malassezia species. FEMS Immunol Med Microbiol 72 Goldust M, Rezaee E, Raghifar R. Treatment of seborrheic dermatitis:
2008;54:203–14. comparison of sertaconazole 2 % cream versus pimecrolimus 1 %
44 Gupta AK, Kohli Y, Summerbell RC, Faergemann J. Quantitative cul- cream. Ir J Med Sci 2013;182:703–6.
ture of Malassezia species from different body sites of individuals 73 Zeharia A, Mimouni M, Fogel D. Treatment with bifonazole shampoo
with or without dermatoses. Med Mycol 2001;39:243–51. for scalp seborrhea in infants and young children. Pediatr Dermatol
45 Youn SW, Na JI, Choi SY et  al. Regional and seasonal variations in 1996;13=:151–3.
facial sebum secretions: a proposal for the definition of combination 74 Ratnavel RC, Squire RA, Boorman GC. Clinical efficacies of shampoos
skin type. Skin Res Technol 2005;11:189–95. containing ciclopirox olamine (1.5%) and ketoconazole (2.0%) in the
46 Mayser P, Schafer U, Kramer HJ et  al. Pityriacitrin  –  an ultraviolet‐ treatment of seborrhoeic dermatitis. J Dermatolog Treat 2007;
absorbing indole alkaloid from the yeast Malassezia furfur. Arch 18:88–96.
Dermatol Res 2002;294:131–4. 75 Chen C, Koch LH, Dice JE et al. A randomized, double‐blind study
47 Barchmann T, Hort W, Kramer HJ, Mayser P. Glycine as a regulator of comparing the efficacy of selenium sulfide shampoo 1% and ciclopirox
tryptophan‐dependent pigment synthesis in Malassezia furfur. Myco shampoo 1% as adjunctive treatments for tinea capitis in children.
ses2011;54:17–22. Pediatr Dermatol 2010;27:459–62.
48 Kumar CP, Kumar SS, Menon T. Phospholipase and proteinase activi- 76 Givens TG, Murray MM, Baker RC. Comparison of 1% and 2.5% sele-
ties of clinical isolates of Candida from immunocompromised nium sulfide in the treatment of tinea capitis. Arch Pediatr Adolesc
patients. Mycopathologia 2006;161:213–18. Med 1995;149:808–11.
49 Burton JL, Pye RJ. Seborrhoea is not a feature of seborrhoeic dermati- 77 Gomez‐Moyano E, Hiraldo Gamero A, Vera Casano A et al. [Phase III
tis. Br Med J (Clin Res Ed) 1983;286:1169–70. study of the efficacy and safety of ciclopirox olamine cream in small
50 Gordon MA. The lipophilic mycoflora of the skin. 1. In vitro culture of children with dermatomycosis]. Rev Iberoam Micol 2015;32:164–9.
Pityrosporum orbiculare n. sp. Mycologia 1951;43:524–35. 78 Van Esso D, Fajo G, Losada I et al. Sertaconazole in the treatment of
51 Harada K, Saito M, Sugita T, Tsuboi R. Malassezia species and their pediatric patients with cutaneous dermatophyte infections. Clin Ther
associated skin diseases. J Dermatol 2015;42:250–7. 1995;17:264–9.
52 Back O, Faergemann J, Hornqvist R. Pityrosporum folliculitis: a com- 79 Braza TJ, DiCarlo JB, Soon SL, McCall CO. Tacrolimus 0.1% ointment
mon disease of the young and middle‐aged. J Am Acad Dermatol for seborrhoeic dermatitis: an open‐label pilot study. Br J Dermatol
1985;12(1 Pt 1):56–61. 2003;148:1242–4.
53 Siegfried EC, Hebert AA. Diagnosis of atopic dermatitis: mimics, 80 Meshkinpour A, Sun J, Weinstein G. An open pilot study using tac-
overlaps, and complications. J Clin Med 2015;4:884–917. rolimus ointment in the treatment of seborrheic dermatitis. J Am Acad
54 Alexopoulos A, Kakourou T, Orfanou I et al. Retrospective analysis of Dermatol 2003;49:145–7.
the relationship between infantile seborrheic dermatitis and atopic 81 Munzenberger PJ, Montejo JM. Safety of topical calcineurin inhibitors
dermatitis. Pediatr Dermatol 2014;31:125–30. for the treatment of atopic dermatitis. Pharmacotherapy 2007;
55 Elish D, Silverberg NB. Infantile seborrheic dermatitis. Cutis 27:1020–8.
2006;77:297–300. 82 Kaposi M. Pathology and Treatment of Diseases of the Skin for
56 Darabi K, Hostetler SG, Bechtel MA, Zirwas M. The role of Malassezia Practitioners and Students [translation from the German under the
in atopic dermatitis affecting the head and neck of adults. J Am Acad supervision of Johnston James C, MD, translator and editor.]. New
Dermatol 2009;60:125–36. York: William Wood, 1895.
57 Glatz M, Bosshard PP, Hoetzenecker W, Schmid‐Grendelmeier P. The 83 Lin AN, Moses K. Tar revisited. Int J Dermatol 1985;24:216–18.
role of Malassezia spp. in atopic dermatitis. J Clin Med 2015;4:1217–28. 84 Veraldi S, Menter A, Innocenti M. Treatment of mild to moderate seb-
58 Coley MK, Bhanusali DG, Silverberg JI et al. Scalp hyperkeratosis and orrhoeic dermatitis with MAS064D (Sebclair), a novel topical medical
alopecia in children of color. J Drugs Dermatol 2011;10:511–16. device: results of a pilot, randomized, double‐blind, controlled trial. J
59 Bhanusali D, Coley M, Silverberg JI et al. Treatment outcomes for tinea Eur Acad Dermatol Venereol 2008;22:290–6.
capitis in a skin of color population. J Drugs Dermatol 2012;11:852–6. 85 Al‐Waili NS. Therapeutic and prophylactic effects of crude honey on
60 Warner RR, Schwartz JR, Boissy Y, Dawson TL Jr. Dandruff has an chronic seborrheic dermatitis and dandruff. Eur J Med Res 2001;
altered stratum corneum ultrastructure that is improved with zinc 6:306–8.
pyrithione shampoo. J Am Acad Dermatol 2001;45:897–903. 86 Satchell AC, Saurajen A, Bell C, Barnetson RS. Treatment of dandruff
61 Faergemann J, Borgers M, Degreef H. A new ketoconazole topical gel with 5% tea tree oil shampoo. J Am Acad Dermatol 2002;47:852–5.
formulation in seborrhoeic dermatitis: an updated review of the 87 Tollesson A, Frithz A. Borage oil, an effective new treatment for infan-
mechanism. Expert Opin Pharmacother 2007;8:1365–71. tile seborrhoeic dermatitis. Br J Dermatol 1993;129:95.
62 Schwartz JR, Cardin CW, Dawson TL. Dandruff and seborrheic 88 Dreno B, Moyse D. Lithium gluconate in the treatment of seborrhoeic
dermatitis. In: Baron R, Maibach HI (eds) Textbook of Cosmetic
­ dermatitis: a multicenter, randomised, double‐blind study versus pla-
Dermatology, 3rd edn. New York: Taylor & Francis, 2004:259–72. cebo. Eur J Dermatol 2002;12:549–52.
286 Section 4  Other Types of Dermatitis

89 Koca R, Altinyazar HC, Esturk E. Is topical metronidazole effec- 92 Siegfried E, Glenn E. Use of olive oil for the treatment of seborrheic
tive  in  seborrheic dermatitis? A double‐blind study. Int J Dermatol dermatitis in children. Arch Pediatr Adolesc Med 2012;166:967.
2003;42(8):632–5. 93 Pierard GE, Arrese JE, Pierard‐Franchimont C, DE Doncker P.
90 Ozcan H, Seyhan M, Yologlu S. Is metronidazole 0.75% gel effective in Prolonged effects of antidandruff shampoos ‐ time to recurrence of
the treatment of seborrhoeic dermatitis? A double‐blind, placebo con- Malassezia ovalis colonization of skin. Int J Cosmet Sci 1997;19:111–17.
trolled study. Eur J Dermatol 2007;17(4):313–16. 94 Faergemann J. Seborrhoeic dermatitis and Pityrosporum orbiculare:
91 Baroni A, De Rosa R, De Rosa A et al. New strategies in dandruff treat- treatment of seborrhoeic dermatitis of the scalp with miconazole‐
ment: growth control of Malassezia ovalis. Dermatology (Basel, hydrocortisone (Daktacort), miconazole and hydrocortisone. Br J
Switzerland) 2000;201:332–6. Dermatol 1986;114:695–700.
SECTION 4: OTHER TYPES
OF DERMATITIS
 287

C HA PTER   22

Irritant Contact Dermatitis


David Luk1,2
 United Christian Hospital, Hong Kong
1

 The Chinese University of Hong Kong, The University of Hong Kong, Hong Kong
2

Abstract and include biological agents, household products, medical agents,


skin care products, hobby‐ and recreation‐related equipment,
food, physical irritants and even personal electronic devices. Both
Irritant contact dermatitis (ICD) is common but is under‐researched
genetic and environmental factors may contribute to the develop-
in children. It is a nonspecific local inflammatory dermatosis caused
ment of ICD.
by single or repeated skin contact with irritants. The exposure to
ICD is mainly a diagnosis of exclusion based on a thorough
irritants is specific to the developmental stages of children and

SECTION 4: OTHER TYPES


history, negative or irrelevant patch tests, proven exposure to
­
can occur from neonates to adolescents. The clinical presentation
­irritants and a typical course of disease. There may be many clinical
of ICD is highly variable and can be classified into acute, delayed
differential diagnoses for ICD and the clinical distinction between

OF DERMATITIS
acute and slowly developing forms. Most ICDs are cumulative ICD,
ICD and allergic contact dermatitis can be vexing. Stopping and
in which exposure to one or more chemical irritants occurs over
avoiding further contact with irritants is the mainstay of manage-
weeks to months and face, hands, feet and perineum are com-
ment of ICD. Short‐period application of topical steroid is generally
monly affected sites. Irritants reported to cause ICD are numerous
helpful, and monitoring for complications may be necessary.

Key points • The irritants cause direct damage to the skin resulting in a
nonspecific inflammatory reaction.
• ICD occurs on the area of skin exposed to the irritant and is a
• Irritant contact dermatitis (ICD) is common in children.
clinical diagnosis supported by a negative patch test.
• ICD can occur in any child when the irritant is strong enough and
• Avoidance of irritants and skin protection remain the mainstay
the exposure is long enough.
of management.
• The exposure to irritants is related to the age and the
developmental environment of the child.

and bubble baths [1,6]. However, while there is no lack of


Background. Contact dermatitis is common in children
research on adult ICD, large‐scale studies on  paediatric
and is an under‐recognized burdensome paediatric
ICD have been rare.
disease that leads to significant health and economic
adversities [1]. It is usually divided into irritant and
allergic forms [2]. Irritant contact dermatitis (ICD) is Definition. ICD is a nonspecific local inflammatory
more common and has less serious potential conse- dermatosis caused by single or repeated skin contact
­
quences than allergic reactions such as allergic contact with chemical, physical or biological agents (the irritants)
dermatitis (ACD) and allergic ­contact urticaria [34]. in the environment p ­ roducing a direct toxic insult to
The circumstances leading to ICD in children are ­cutaneous cells [2,7–9]. There is no specific antigen‐primed
­different from those in adult ICD. ICD in adults is often immune response [1] and the nonimmunological reaction
occupational or related to housework [5]. On the other develops after sufficient contact with the substance.
hand, paediatric ICD has been reported from extremely Everyone exposed to the same irritating agent is poten-
premature neonates in neonatal intensive care units to tially capable of responding in a stereotyped manner,
adolescents with various hobbies, behaviours and topical although factors such as the presence of ongoing dermati-
medications. Thus, some contact irritants are specific tis (atopic dermatitis), environmental factors or physical
to developmental stage. For example, the most common traumas may facilitate onset [2].
form of ICD in early infancy is nappy rash, which becomes ICD is distinct from ACD, in which the inflammatory
less common after children have established continence [2]. skin disease is initiated by specific cell‐mediated (type IV)
Other common causes of ICD in the paediatric age group hypersensitivity reactions to a hapten and requires an
include drooling (toddlers, cerebral palsy), o­ verbathing initial sensitization phase followed by the elicitation

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
288 Section 4  Other Types of Dermatitis

phase that causes the skin lesions in predisposed subjects r­eaction [7,12,22,23]. In this process, keratinocytes have
[2]. It is also distinct from photodermatitis, in which light a major role, and they release cytokines on the disruption
exposure is required to cause skin inflammation. of the skin barrier, upregulate their major histocompati-
bility class II antigens, and upregulate cell adhesion mole-
Epidemiology cules [24]. The pro‐inflammatory cytokines interleukin‐1α,
Age interleukin‐1β and TNF‐α are upregulated [25]. The
Contact dermatitis is an extremely common occurrence in chemokine CCL21, produced by dermal lymphatic
the paediatric age group. It accounts for a large number of endothelial cells, is also upregulated [26]. These mediators
paediatric cases of dermatitis [1], representing approxi- facilitate the migration of naive T lymphocytes, resulting in
mately 20% of all dermatitis seen in childhood [6]. a skin inflammatory response [23]. Interestingly, many of
Although the exact incidence is unknown, ICD is the most the same cytokines that are found in ACD are also detected
common form of contact dermatitis, comprising approxi- in ICD [12]. Indeed, Smith and colleagues reported that
mately 80% of cases  [1,2,10]. Susceptibility to ICD is ICD may promote or enhance ACD because some of these
inversely proportional to age [11,12] and the threshold for mediators play a role in both ICD and ACD [27,28].
skin irritation is lower in younger children [7]. Children
under 8 years old are more vulnerable to irritation Clinical features. The clinical presentation of ICD is
SECTION 4: OTHER TYPES

whereas skin reactivity becomes normal above this age highly variable and depends on multiple factors includ-
[11]. ICD may develop in newborns and infants as the ing the nature and intrinsic properties of the irritant and
OF DERMATITIS

percutaneous absorption of substances is greater with specific individual‐ and environment‐related variables
their thinner skin and higher skin surface area to body- [7]. Ten clinical types of ICD have been recognized and
weight ratio [2], and their high microcirculatory efficiency can be categorized into acute, delayed acute, and slowly
also facilitates inflammatory cell recruitment to the site of developing forms [21,29].
inflammation [7,11]. Whereas ICD may develop from
birth, ACD generally starts to develop at the age of Clinical types of ICD
2–3 years (sometimes even as early as 6 months) because Acute ICD
of progressive exposure to sensitizing agents and the Acute ICD results from skin exposure to a potent irritant
immaturity of the cell‐mediated immune system during or caustic chemical such as acids or alkaline solutions [21]
the first two years of life [2]. and the onset of symptoms is within minutes to hours of
exposure [7,18]. There could be immediate burning, itch-
Gender ing or stinging [21]. It usually occurs in a private environ-
Researches on gender differences in ICD have not been ment, for example, when disinfection solutions were used
conclusive [11,13]. There have been conflicting data on incorrectly [5]. It is difficult to differentiate very severe
whether males or females have a greater tendency irritant contact dermatitis from a chemical burn when
towards ICD [11,14]. erythema, oedema and vesicles may rapidly progress to
necrosis and ulceration [30] [Table 22.1].
Race
There have been few studies on racial differences of skin Delayed acute ICD
irritation responsiveness in adults [15,16] and an increased A delayed acute ICD may be initiated 8–24 hours follow-
reactivity was noted for Asians versus Caucasians [14]. ing acute exposure to chemicals such as anthralin
(dithranol), benzalkonium chloride and hydrofluoric acid
Genetic factors [31]. Contact dermatitis related to surgical exposures is
Genetic factors certainly have a role in chronic ICD [7,17]. also typified by delayed onset of symptoms and signs
Atopic dermatitis has been shown to predispose to ICD [32]. Otherwise, the symptoms and signs of delayed acute
[18] and skin atopy at least doubles the risk of developing ICD are similar to those of acute ICD [21].
ICD in adults [19]. Loss‐of‐function polymorphisms in
Cumulative ICD
the filaggrin gene were also found to be associated with
Most ICDs are cumulative ICD, in which exposure to one
increased susceptibility to chronic ICD [17]. A nonatopic
or more chemical irritants [7,23,33] occurs over weeks
genetic marker for irritant susceptibility, TNF‐α polymor-
to months [21] (arbitrarily defined as 6 weeks [34]) after
phism in normal individuals, has also been reported [20].
multiple subthreshold skin insults from milder irritants [18].
In cumulative ICD, the skin barrier function does not
Pathogenesis. As ICD does not elicit an antigen‐specific
immune response to irritants, the pathogenesis of ICD was Table 22.1  Clinical severity of ICD
thought to be nonimmunological in the past. It is now
known that pro‐inflammatory cytokines and the innate Grade Severity Signs
immune system play a key role in its pathogenesis [21].
Grade 1 Mild Erythema
In  ICD, chemical or physical insults lead to a cascade of
Grade 2 Moderate Erythema, oedema, small vesicles
innate immune responses: (i) skin barrier disruption; Grade 3 Severe Blisters, bullae
(ii) direct cytotoxic effects on keratinocytes; (iii) induction Grade 4 Most severe Epidermo‐dermal necrosis, ulceration
of cytokine  cascades; and (iv) a clinical inflammatory
Chapter 22  Irritant Contact Dermatitis 289

have time to heal properly, resulting in the associated


itch,  pain, dryness, erythema and sharply demarcated
lesions [21]. The chronic exposure leads to lichenification
while vesiculation may be less apparent or absent [6].

In addition to the above three major forms, other forms


reported include irritant reactions, subjective irritations,
nonerythematous (suberythematous) irritations, traumit-
erative dermatitis, traumatic irritant dermatitis, pustular
and acneiform dermatitis [21].

Patient characteristics
Age
Age is an important factor for ICD in children as different
types of ICD tend to occur at various stages of develop-
ment of childhood with different exposure to irritants.

SECTION 4: OTHER TYPES


The most common form of ICD in early infancy is nappy
Fig. 22.2  Irritant contact dermatitis due to thumb sucking in an infant.
rash [2], and this topic is covered in Chapter 20. In the first

OF DERMATITIS
few years of life, perioral dermatitis and irritant cheilitis
caused by contact with a pacifier, habitual lip biting or as to increased transepidermal water loss on the chin and
a result of excessive saliva production during erupting nasolabial areas in younger people [40,41]. Perioral
dentition are common [2,35]. Irritation from oral secre- ICD may be aggravated by the use of tooth whiteners/
tions (saliva or food) is frequently noted in toddlers [6]. fluoride with clinical signs accentuated at the commissure
The repeated wet and dry cycles from saliva disrupt the of the lips [36]. In addition to the face, intertriginous
epithelial barrier and cause perioral fissuring [10,36] and thigh areas are also vulnerable areas for ICD [22].
(Fig. 22.1). The common habit of thumb and finger suck- ICD of hands in children may be related to inappropri-
ing in young children can result in ICD on their fingers ate use of cleansers, exposure to harsh soaps, sanitizing
[37] (Fig. 22.2). Lip licker dermatitis is more often found solutions [1], hair care products, hobbies, activities
in older children who lick their lips frequently to soothe that  involve contact with water (e.g. washing clothes,
lips irritated by cold dry weather [6]. Children who play cookery) and even prolonged wearing of protective
outdoors may develop phytodermatitis by means of a gloves [42]. On the feet, chemicals used in shoe manu-
traumatic, chemical or toxic mechanism (airborne or the facturing have been reported to cause severe acute ICD
result of direct contact), which may also be favoured or and ACD in adults and contact urticaria in children
worsened by exposure to the sun (contact phytophoto- [43]. Barefoot individuals in India have been reported
dermatitis) [2]. Adolescents may have ICD due to irrita- to develop ICD with continuous exposure to water and
tion by topical acne treatment, cosmetics [23], henna detergents [44].
tattooing and hair highlighting [38]. In this age group, Perineal ICD can occur at any age [6]. Poor hygiene,
hobbies and regular activities may result in ICD by repeti- such as inadequate wiping after urinating, wiping poste-
tive contact with the equipment. riorly to anteriorly, or where a young child showers with-
out adult supervision, may result in perineal ICD [45].
Body sites The use of perfumed creams or powders and feminine
There is regional variation in ICD on the body. The face hygiene sprays can lead to either an irritant reaction from
is a common site for ICD by virtue of its permeability the propellant or an allergic contact reaction from per-
[39] due to a relatively thin epidermis and a tendency fumes in the spray. Bubble bath is a common cause of
ICD  in children and can cause contact dermatitis in the
vulvar area [6].

Coexisting medical conditions


Children with coexisting medical conditions may have
additional causes of ICD [22]. Licking of the lips is found
frequently among developmentally delayed patients. The
tongues of such patients are commonly enlarged, and
salivation is often excessive. The habit involves sweeping
the tongue about the perioral region, keeping the part wet
with saliva, which may result in perioral ICD [37].
Obsessive compulsive behaviours involving handwash-
ing may result in ICD of the hands [46,47]. In the intensive
care unit, the barrier function of the skin of critically ill
children is compromised by its hypoperfusion with a
Fig. 22.1  Perioral dermatitis with perioral fissuring in an infant. resultant increased susceptibility to ICD [22].
290 Section 4  Other Types of Dermatitis

History
Children are exposed to irritants in the environment
where they live, learn and play, which can be both indoor
or outdoor [8]. Details including plants that they have
plucked or touched [48], hygiene products, clothing, foot-
wear, jewellery, cosmetics, recent travel, new products,
medicaments, herbal supplements, hobbies, extracurricu-
lar activities and environments may be helpful. As ICD
may occur after only brief exposures or intermittent expo-
sures over a prolonged period of time [48], a thorough
history is important. Sometimes the offending substance
may not be apparent, but it is also possible that multiple Fig. 22.4  Sharp ‘cut‐off’ borders of irritant contact dermatitis skin lesions.

irritants are acting at the same time [2]. In addition, clari-


fying the details of exposure including time, duration, provide diagnostic information regarding the aetiology of
concentration, chemical ingredients, environmental fac- the reaction [6].
tors, protection employed and subsequent treatment Although the clinical presentation of ICD is usually
SECTION 4: OTHER TYPES

received would be very helpful. It is often useful to ask confined to sites of direct contact, fomites can secondarily
also about parents’ or siblings’ products because of the transfer irritants from sites of primary contact to other
locations on the body. There have been reports of a leather
OF DERMATITIS

potential for indirect or even direct exposure from their


products [10,49]. Furthermore, one must also consider the watchband serving as a fomite, causing cutaneous irrita-
time course necessary for an ICD reaction which can tion for up to 3 months after initial contact with spurge
range from minutes to hours for acute ICD and weeks to latex [52]. The secondary eruptions that occur by fomite
months for cumulative ICD [30]. transfer can manifest themselves anywhere but frequently
Family, drug or atopy history including atopic derma- present on sensitive areas such as the axilla, eyes, but-
titis, a history of previous contact dermatitis and coex- tocks and genitalia [27]. A general examination of the skin
isting medical problems may be helpful [50]. Patients is also helpful to identify autoeczematization of ICD [19],
with ICD usually do not have fever or other systemic concomitant dermatoses such as atopic dermatitis and
symptoms [50]. xerosis and other differential diagnoses of ICD.
Apart from examining the patient, the examination of
Physical examination the suspected irritants and understanding the environ-
ICD is generally characterized by erythema and vesicula- ment by reviewing relevant photos is also helpful.
tion in the acute phase, and by xerosis, fissuring and
lichenification in the chronic phase [51]. Eczematous, ery- Irritants
themato‐desquamative lesions [2], pruriginous pustular A substance is an irritant when it has the potential to irri-
lesions, pustule remnants and thin scales over an ery- tate skin [5] and virtually every agent can function as an
thema have all been described [50]. More severe cases irritant when there is excessive exposure, including even
with bullae or necrosis accompanied by intense pain may paper and water [5–7,33]. The irritation potential of the
suggest a severe ICD reaction [29,48] (Fig. 22.3). agent is affected by its concentration, pH [53,54], molec-
Rashes localized to one area (e.g. hands) or those that ular size, ionization state and fat solubility which deter-
have discrete shapes and sharp ‘cut‐off’ borders raise a mine its skin penetration [7,22,23,55]. The nature of the
suspicion for contact dermatitis [6] (Fig. 22.4). The distri- contact such as the time duration [7,55], the frequency of
bution of lesions corresponding to the contact areas may contact [5], the amount [7] and the solvent [7] may also
affect the degree of ICD. Occlusion (e.g. by gloves and
clothing) may increase the signs of skin inflammation
by  altering the pH, transepidermal water loss, stratum
corneum hydration, skin surface temperature and skin
permeability [12].
Environmental temperature and humidity are exoge-
nous factors contributing to the development of ICD
[12,23]. Cold temperatures increase transepidermal water
loss and susceptibility to irritants [56] but warm ambient
temperatures are also damaging [57]. A low environmental
humidity may enhance irritability of skin [23]. The pres-
ence of sweat and friction [58] may further aggravate ICD
as mechanical irritation supports chemical irritation [5].
Irritants reported to cause ICD are numerous and can
be grouped into eight categories: (i) biological agents; (ii)
household products; (iii) medical‐related agents; (iv) skin
care products; (v) hobbies and recreations; (vi) food; (vii)
Fig. 22.3  Severe irritant contact dermatitis with blisters. physical irritants; and (viii) others.
Chapter 22  Irritant Contact Dermatitis 291

Biological agents greenhouse‐cultivated plants, would cause ICD [61]. In


Body fluids. Children’s own body fluids are an important North America, the bright red foliage of E. pulcherrima
source of irritants. Saliva (toddlers, cerebral palsy), urine, and child‐accessible placement in households resulted in
faeces, and the body fluid leaking around a stoma are the majority of exposures in children, especially for those
some of the common causes of ICD in children [1]. younger than 2 years [62]. On the other hand, blisters
developing after children played in the fields were also
Plants. Phytodermatitis can be classified into ICD, ACD, reported [48].
photoallergic contact dermatitis and phototoxic reaction
(phytophotodermatitis) [59]. ICD accounts for the major- Insects. Paederus dermatitis is a self‐limiting form of acute
ity of plant dermatoses [27] (Fig.  22.5). Potential irritant ICD caused by pederin released by rove beetles when
plants are ubiquitous in our environment: home, garden, accidentally brushed or crushed on the skin [63,64]. Itch,
workplace and recreational settings [27]. Calcium oxalate, stinging or burning sensation on exposed parts of the
protoanemonin, isothiocyanates, bromelain, diterpene body may be accompanied by vesicles and pustules on an
esters, alkaloids and naphthoquinone are some of the erythematous base in a linear configuration [63]. In severe
known plant chemicals causing ICD [27]. The clinical cases, pederin can cause skin necrosis [65] and constitu-
presentation may range from mild chronic eczematous tional symptoms [66]. Seasonal outbreaks have been

SECTION 4: OTHER TYPES


dermatitis due to mechanical irritation by hairs on some reported worldwide including Africa, South America,
plants to severe acute toxic reactions with necrosis due to Australia, the Middle East and Asia [63,64,66–69] and the

OF DERMATITIS
potent alkaloids from plant sap [27]. Juglone, the active signs of periorbital dermatitis and linear lesions may hint
ingredient of green shells of walnuts, is a strong irritant at the diagnosis [63,64].
and circumscribed hyperpigmented and combustiform Topical envenomation from caterpillars is another
skin lesions have been reported to appear suddenly after insect‐associated ICD [1]. Caterpillars can cause reactions
two children played in a garden with green walnut husks through mechanical, chemical and allergic means [70].
[60]. The spurge (Euphorbiaceae family) is also ubiqui- The barbed hairs adhere to the skin and mucous mem-
tous and toxic, and is a significant cause of ICD world- branes, causing clinical symptoms due to mechanical
wide [41]. Children often unwittingly encounter the plant irritation and the presence of a foreign body [71].
through play [41]. The active irritants in the spurge sap Envenomation by caterpillars with thaumetopoein elicits
are diterpene esters, which are also present in the plant’s a toxic response by direct non‐IgE‐mediated mediator
stem, petals, roots and leaves and can cause both ICD release [71]. Type 1 hypersensitivity reactions have also
and  ACD [41]. It has been suggested that wild, but not been reported after contact with caterpillars [71,72].

Household products
Common household items, such as soap and detergents,
can be a cause of ICD in children. Detergents may con-
tain highly irritating compounds such as quaternary
ammonium compounds, phenol and formaldehyde [53].
When detergent is left on a toilet seat, bilateral symmetri-
cally distributed ICD on the posterior thighs and but-
tocks can result [1] and toilet‐seat dermatitis continues to
be a common condition in children around the world [53]
(Fig. 22.6). Residues of detergents or fabric softeners in
laundered clothes can also cause ICD.

Medical‐related agents
Medications, antiseptics and medical devices have been
reported to result in ICD in both hospital and non‐hospi-
tal settings.

Medications.  In the paediatric age group, topical acne


drugs are notorious for irritation because of their intrinsic
mechanisms of action [73]. Benzoyl peroxide is known to
be a strong irritant [74], resulting mainly in ICD rather
than ACD [73,74]. Formulations of less than 5% cause less
irritation, and cream and lotion vehicles are better toler-
ated than gel preparations [73]. In the topical retinoid
group, tretinoin 0.05–0.1% (all‐trans retinoic acid) and
tazarotene 0.05–0.1% are most irritating whereas isotreti-
noin 0.05% has a lower irritating potential [73]. Adapalene,
a third‐generation retinoid, at 0.1%, is less irritating [73].
Fig. 22.5  Configuration of lesion providing important clinical clues. Ultraviolet light protection is recommended to reduce
292 Section 4  Other Types of Dermatitis

Fig. 22.6  Toilet‐seat dermatitis.


SECTION 4: OTHER TYPES

irritation due to topical retinoids [73]. In addition, s­ alicylic


OF DERMATITIS

acid 2% is commonly associated with scaling and burning


[73], and azelaic acid is commonly associated with itching, Fig. 22.7  Irritant contact dermatitis due to topical acne drugs.
burning and dysaesthesia. Although topical antibiotics
(tetracyclines, erythromycin, clindamycin) in the treatment as medical adhesives [89], pulse oximetry [90], immobili-
of acne have low irritating potential, their prolonged zation boards [90] and casts [91], orthotics [90], silicone
application may induce antibiotic resistance [73] (Fig. 22.7). gel sheets [92], ear prostheses [93], dental materials [94]
Topical medications for other common dermatoses are and continuous positive airway pressure devices [90], all
also reported to cause ICD. Topical calcipotriol [75], of which have the potential to induce mechanical or
dithranol [76] and tar [77] for the treatment of psoriasis chemical ICD. Surgery‐related ICD [32] and peristomal
can cause both ICD and ACD [75]. Scabies medications ICD can be caused by the use of dressings, adhesive tapes,
known to be irritating include lindane, 1% gamma ben- antiseptics, medicaments, ostomy pouch systems, sutures,
zene hexachloride [78], benzyl benzoate and precipitated barrier films and adhesive paste and removers [90,95,96].
sulphur. Applying the same preparation to the whole
family may result in ICD in young children without par- Skin care products
ents being affected. It is common for skin care products to cause ICD [97] and
Topical analgesic creams (lidocaine and prilocaine there is a long list of potential irritants in these products
cream under occlusion) have been reported to cause both (Tables  22.2 and 22.3). Modern cleansers are primarily
ICD and ACD [79–81]. The methylphenidate transdermal made up of surfactants that act by decreasing the surface
system used in children with attention deficit hyperactiv- tension between water and oil. Surfactants create lather
ity disorder also commonly causes ICD [4]. and allow fat‐soluble impurities to be removed from the
skin surface [98]. However, surfactants are also irritants
Antiseptics.  Chlorhexidine is a widely used broad‐ as they weaken the skin barrier and increase transepi-
spectrum topical antiseptic agent, and chlorhexidine dermal water loss [98]. Aqueous creams or emollients
baths are among the methods employed to reduce containing sodium lauryl sulphate (a detergent and sur-
­meticillin‐resistant Staphylococcus aureus (MRSA) infection factant) should not be used as emollients or as leave‐on
[82]. Although ICD due to chlorhexidine is rare [22,83], skin products due to their irritating properties  [108].
infants and young children are the most susceptible age Bath oil has also been reported to cause ICD whether
groups [22]. The Centers for Disease Control and with inadequate dilution [109], prolonged contact [101]
Prevention recommend its use as a skin cleanser prior to or  even with appropriate use [102] (Fig.  22.8). Active
insertion of central venous catheters in children but not ingredients of sunscreens may be allergens or irritants.
in  infants less than 2 months of age [84]. Premature Dexpanthenol is widely used in topical medical products
infants, immunosuppressed or critically ill patients are and cosmetics [50] due to its moisturizing and skin‐
particularly vulnerable and cases of severe ICD to softening properties, but ICD after accidental use of a
chlorhexidine have been reported [85,86]. Although
­ parental moisturizer on a child’s skin has been reported
povidone–iodine has low toxicity, its postoperative use [50]. Bandages, hair dye, deodorants and antiperspirants
on skin for 8 hours has also resulted in ICD [87]. are also reported to contain irritants [32].
Antiseptics containing cetrimide are also widely used as
disinfectants and sanitizers but improper (undiluted) Hobbies and recreations
application of the antiseptic liquids has led to ICD [88]. As children become actively involved in various recrea-
tions and hobbies, the use of equipment may result in
Medical devices.  Children requiring long‐term medical ICD. Face paints mixed with dishwashing liquid were
support are exposed to a variety of medical devices such reported to cause severe ICD in children in 1980 [110].
Chapter 22  Irritant Contact Dermatitis 293

Table 22.2  Irritancy potential of common surfactants In sports activities, children have been reported to have
ICD secondary to soccer shin guards [57,111], kendo prac-
Relative irritancy Surfactant tice [112] and baseball pitching [113]. Accidental contact
with jellyfish during swims has also resulted in ICD [114].
High Benzalkonium chloride
In a recent survey of instrument‐related skin disorders in
Bromide
Dodecyl trimethyl ammonium
musicians, various patterns of ICD were found [115]. Lip
Linear alkyl benzene sulphate ICD caused by a saxophone causing and fiddler’s neck due
Sodium lauryl sulphate to ICD or ACD have been reported in children [116–118].
Sodium dodecyl sulphate
Sodium alkyl sulphate Food
Sodium or potassium cocoate Irritants in food usually result in cumulative chronic ICD
Sodium or potassium tallowate
affecting the hands. The erythematous scaly eruptions can
Sodium palmitate
Sodium or potassium stearate be limited to the finger tips but other distributions are also
Sodium olefin sulphonate possible: scattered, generalized, arms and even face [119].
Triethanolamine laurate ICD is more common among food handlers and children
Moderate Sodium ethoxylates with atopic dermatitis [120]. Non‐occupational exposure

SECTION 4: OTHER TYPES


Sodium laureth sulphate to food in children is usually sporadic and may occur at
Ammonium laureth sulphate
home or at school [120]. Occupational hand dermatitis
Low Sodium cocoyl isethionate

OF DERMATITIS
Sodium alkyl glycerol ether sulphonate
in food industry apprentices has been reported [121].
Sodium cocoyl sulphosuccinate Much of the food we eat is capable of causing one or
Disodium stearyl sulphosuccinate more types of dermatitis [122]. For example, garlic causes
ACD and ICD as well as photoallergic contact dermatitis
Source: Kuller [98]. Reproduced with permission of Wolters Kluwer [122]. In the Cross‐Sectional Analysis of North American
Health, Inc. Contact Dermatitis Group Data, 2001–2004, ICD is com-
monly due to general food products, bakery products,
Table 22.3  Skin care products with potential to cause ICD fruits, nuts, vegetables, meat and poultry [119] while spices,
garlic, onion, citrus fruits, corn, radish, mustard, potato,
Skin care products Irritants pineapple and carrot are also common [120,122]. Pineapple
contains bromelin, a proteolytic enzyme that can cause a
Cleansers and See Table 22.2 [98,99]
separation in the epidermis along with an increase in capil-
soaps [98]
Shampoo [100] Sodium lauryl sulphate, benzalkonium chloride
lary permeability [122,123]. Capsicum peppers such as cay-
Emollients Sodium lauryl sulphate enne and jalapeno contain the oleoresin capsaicin which
Bath additives Benzalkonium chloride can irritate the mucous membranes of the eyes, nose and
[101,102] mouth in addition to causing ICD to skin.
Sunscreen [103] PABA, benzophenones, benzotriazole derivatives Food additives are also a source of irritants which
Dressing [32,104] Rubber products, adhesives (methacrylate, include acetic acid, ascorbinic acid, calcium acetate, cal-
epoxy diacrylate)
cium sulphate, lactic acid, potassium bicarbonate, potas-
Cosmetics [50,100] Dexpanthenol
Hair dye [105] Hydrogen peroxide, persulphates sium iodide, potassium bromate and yeast [122].
Deodorant [106] Aluminium sulphate
Antiperspirant [107] Aluminium chloride Physical irritants
Apart from chemical irritants, ICD can also be caused by
physical irritants but this area is relatively under‐
researched [33]. Physical irritants include friction [12],
pressure [12], vibration [12], heat [12] and sweat [12,57]
and these can result in ICD via mechanical trauma or
microtrauma [27].
Mechanical ICD caused by labels from clothing
may lead to ‘label dermatitis’ over the upper back [124].
Although textile contact dermatitis is more often an
immune‐mediated reaction, rough‐textured clothing,
woollen clothing and clothes contaminated by fibreglass
can lead to mechanical ICD [125]. Fibreglass with fibres of
diameter 5.3 μm or more may cause mechanical irritation,
and a fibreglass‐reinforced plastic chair has been reported
to cause ICD at school [126].
There can be overlap between chemical ICD and
mechanical ICD. For example, in plant‐induced ICD
caused by Narcissus bulbs, the physical microtrauma from
handling the bulbs may enhance the cutaneous penetra-
Fig. 22.8  Irritant contact dermatitis due to bath additives. tion and effects of chemical irritant calcium oxalate
294 Section 4  Other Types of Dermatitis

Fig. 22.9  Irritant contact dermatitis due to traditional remedies for joint
pain.
Fig. 22.10  Skin lesion localized to the contact area – bandage.
SECTION 4: OTHER TYPES

crystals [27,127]. Physical irritation by friction on skin has


also been shown to potentiate the chemical irritation on the duration and concentration of the exposure)
caused by sodium lauryl sulphate [128].
OF DERMATITIS

and  a  monomorphic rather than polymorphic clinical


In the wake of the use of personal electronic devices in ­picture  [149]. All potential exposures to irritants should
the modern era, new groups of cutaneous pathologies be explored [1]. Careful physical examination should be
have been reported. Games console/cellphone thumb performed to identify features of ICD and the possible
due to friction was first described in 2004 [129] and started differential diagnoses [51]. Typically, ICD remains local-
as a new epidemic in teenagers. Physical ICD can cause ized to the area of skin contact (Fig. 22.10). This is unlike
blisters on the exact site of fingers under recurrent pres- ACD, which can spread to distant parts of the body [19].
sure and friction with the controller of a games console
[130]. Chemical ICD may result from exposure to certain Differential diagnoses
chemicals in keyboards, mice and wrist pads [131] but There is a long list of differential diagnoses for contact
ACD from nickel is also possible from the use of laptop dermatitis, and diagnosis depends on the clinical appear-
computers and pads [131–133]. With repeated and pro- ance of individual lesions. ICD should be differentiated
longed use of laptop computers, erythema ab igne may be from ACD, photodermatitis and contact urticaria [122].
found on the thighs of children [131,134,135]. This was Xerosis [88], infections (e.g. tinea [88], herpes zoster, her-
first described in 2004 and was due to long‐term exposure pes simplex [63], impetigo), inflammatory dermatoses
to mild heat in the range 43– 47 °C [131,136,137]. This is (e.g. periorificial dermatitis), structural skin lesions (e.g.
now an emerging condition among adolescents [138] with lymphangioma circumscriptum [150]) and infiltrative
the youngest reported child being 9 years old [135]. skin diseases (e.g. histiocytosis [6]) have been included as
differential diagnoses in appropriate clinical settings.
Others Cases of contact dermatitis mimicking child abuse have
The list of irritants causing ICD is long but studies on also been reported [151,152] with a 12‐year‐old girl hav-
local products, e.g. traditional remedies [139–141] and ing skin lesions due to contact dermatitis which were ini-
traditional medicines [48,142–144], may be lacking tially suspected to be caused by nonaccidental injury or
(Fig. 22.9). Airborne ICD is mainly an occupational haz- self‐harm, thus causing great distress and concern to the
ard in adults and is rarely reported in children. Airborne family [153].
irritants implicated include phosphates, metal dusts, ACD should be differentiated from ICD because treat-
cleaning products, animal dander, epoxy resin, isothia- ment and prognosis are different. On clinical features
zolinones, ammonia, anhydrous calcium sulphate and alone, ICD and ACD may be very similar and difficult to
formaldehyde [145,146]. Fibre‐related ICD has been distinguish. Like ICD, ACD can present in many different
reported in an apprentice fitter in an aircraft factory (car- forms, the most common of which is an erythematous
bon‐fibre composite wing components) [147] and an out- oedematous eruption at sites of allergen contact [27].
break has been reported in an intensive care unit due to Acute ICD tends to develop on the day of contact with the
fibres from an air‐conditioning filter [148]. In airborne irritant [27]. ACD generally intensifies between 24 and 48
ICD, the skin lesions are usually localized to uncovered hours after exposure, but persons who have been previ-
areas of skin initially. ously sensitized to an allergen may develop symptoms
more rapidly [27]. Pruritus is also not a distinguishing
Diagnosis feature as both types of reactions have been documented
Diagnosis to be extremely pruritic [27].
ICD is an exclusion diagnosis based on a thorough his- In fact, many chemical agents are capable of producing
tory, negative or irrelevant patch tests, proven exposure more than one pathological effect on contacting skin. For
to irritants, typical course of disease (healing and new example, colourants used in cosmetics, foods and feeds
outbreak within a short time of one another, depending may cause ICD, ACD and phototoxic reactions [154].
Chapter 22  Irritant Contact Dermatitis 295

Chemicals such as benzoyl peroxide [74], formaldehyde Ultimately, removing the suspected offending agent is
[1], benzalkonium chloride [155], dexpanthenol [50,156], a test that can also provide treatment if there is symptom
henna tattoo [157] and dimethyl sulphate [158] are known relief [1]. In case of outbreaks of ICD, multidisciplinary
to cause both ICD and ACD. In addition, contact dermati- investigations including field evaluation may also be
tis may have concurrent allergic and irritant components needed to identify and remediate the causes.
[159] and it is not uncommon for more than one condition
to be present in the same patient [119]. In fact, ICD is a risk Management. Stopping and avoiding further contact
factor for the development of ACD [5] and skin irritation with irritants is the mainstay of management of ICD.
predisposes the skin to develop sensitization so that a
clinical exacerbation of ICD may reflect development of
ACD. The clinical distinction between the two types of Acute care
contact dermatitis can be vexing and may not be always In acute ICD, cool compresses of water or physiological
possible [6]. In the cross‐sectional analyses of North saline not only provide symptomatic relief but can also
American Contact Dermatitis Group Data, 1994–2004, have a profound effect on the healing of experimentally‐
one third of patients with hand ACD had identifiable rel- induced ICD [165]. Water has a known hygroscopic effect
evant irritants [159]. that may increase the capacity for intracellular moisture
retention [166]. Saline’s osmotic properties may allow

SECTION 4: OTHER TYPES


Phytophotodermatitis is a phototoxic reaction resulting
from the interaction of plant psoralens with ultraviolet fluid to be drawn from the oedematous lesions in some
patients [166]. Finally, cool compresses may decrease both

OF DERMATITIS
radiation and can result from exposure to plant fruit rind,
juices (most commonly limes), leaves (figs) or, less com- the inflammation and surface temperature associated
monly, stems. An example might be a child at the beach with the ICD [21].
under the sun in contact with plant psoralens from his Rarely, when the acute ICD is very severe with
parents’ drinks such as lemon, lime, celery, dill, parsnip intense pain, bullae and necrosis, hospitalization may
and carrot juices [6]. Severe phytophotodermatitis in a 10‐ be needed [30].
year‐old child has been reported to be caused by contact
with giant hogweed [160]. Phytophotodermatitis is dis- Minimizing contact with irritants
cussed in more detail in Chapter 80. Preferably, contact with irritants should be completely
removed. For example, prevention of ‘label dermatitis’
Investigations from clothes requires the complete removal of labels
There is a lack of confirmatory diagnostic tests for ICD including the threads [124]. If use of the irritant is
[18] and abnormal results on blood tests such as complete ­essential, replacement with a less irritating agent may
blood count, routine biochemistry including hepatic and help. ICD due to chlorhexidine‐impregnated dressings
renal function testing, thyroid function tests and erythro- may improve on changing to povidone–iodine dressings
cyte sedimentation rate are not expected [50]. Laboratory [22,55]. In situations when the irritant cannot be removed,
studies may be helpful to rule out some of the differential a reduction in exposure may also be helpful [5]. Lip ICD
diagnoses. caused by a saxophone in a 12‐year‐old boy improved
The use of patch testing to differentiate ICD from ACD with less frequent playing [116]. Clear instructions may
is important [23] and is indicated when there is a clinical help reduce irritation of topical acne drugs by specifying
suspicion of contact dermatitis or when a child fails to the quantity, frequency and duration of application
benefit from the recommended treatment for a dermato- of  the drugs. Topical acne drugs applied on the face
logical disease such as atopic dermatitis. In addition, it is should be limited to 0.6 g each time, usually used once
helpful for dermatitis on areas at risk of contact dermati- daily, and be rinsed off after 8–12 hours. The use of
tis: face, eyelids, genitals, hands and feet [2]. Although lower  concentration preparations may also be helpful.
patch testing in children using reagents at the same con- Conversely, cleansers and masks containing salicylic
centrations as those used for adults has generally been acid, benzoyl peroxide, sulphur or sodium sulphaceta-
recommended [2], a reduction of allergen concentration mide should be rinsed off after few minutes. Alternating
has been suggested for children below 8 years old [161]. the site of application may improve ICD in chil-
Some patch tests may show irritant reactions at 48 hours, dren  requiring regular transdermal methylphenidate
especially when using higher adult concentrations, but ­treatment [4].
these have often resolved by the 96‐hour reading whereas When irritants such as urine, saliva and wound dis-
allergic contact reactions are often more manifest at this charges are unavoidable, using exogenous barrier pro-
later time. The details of patch testing as well as repeated tectants becomes important [1]. Barrier creams may
open application tests are discussed in Chapter 23. inhibit and delay cutaneous penetration of irritants
Histology is generally thought to be unhelpful in estab- [167]. Zinc paste, petrolatum or special dressings are
lishing the diagnosis but may be required in atypical pres- helpful and emollients may provide additional barrier
entations when alternative dermatological diagnoses are protection [10,104]. Protective gloves, clothing, dress-
suspected [23,50]. Newer technologies such as confocal ings and equipment reducing contact with irritants are
microscopy [162,163] and high‐definition optical coher- also important. However, protective equipment may
ence tomography [164] may have a role in facilitating the also be a cause of ICD so care must also be taken when
differentiation of acute ACD and ICD. wearing it [5,168].
296 Section 4  Other Types of Dermatitis

Skin care to enhance recovery Phototherapy


Moisturizing creams Phototherapy is mainly reserved for patients with refrac-
Humectants in moisturizers increase skin hydration and tory ACD and chronic ICD that is unresponsive to topical
their lipid components act as emulsifiers to accelerate or oral corticosteroids or for patients who cannot avoid all
skin barrier recovery [168] so applying moisturizers gen- provoking factors in their daily environment. Ultraviolet
erously and frequently may be helpful [23]. However, the light has intrinsic immunosuppressive properties  [51]
effect of moisturizers on transepidermal water loss and and both PUVA and UVB light therapy have been used to
susceptibility to irritants depends on the composition of treat chronic ICD in adults [176,177]. However, research
the moisturizer [169]; care must be taken to choose an on the role of phototherapy in children is lacking.
appropriate one [170]. It is prudent to recommend using
moisturizers which have been used on the child previ- Monitoring for complications
ously without problems. ICD has also been reported to have short‐term complica-
tions such as superimposed infection [6,22] and long‐term
Avoiding factors that exacerbate irritation complications such as residual hypopigmentation [157],
Exogenous factors aggravating ICD should be minimized. scar [23] and keloid formation [114].
Mechanical friction on the skin should be avoided and
SECTION 4: OTHER TYPES

washing clothes with fabric softeners may be beneficial Prevention of recurrences


[168,171]. The risk of ICD to topical acne drugs is increased Healing in ICD is described as a decrescendo phenomenon,
OF DERMATITIS

if there is repeated daily washing or rubbing by sponges implying that this process starts immediately after removal
of the face [73]. Skin should be washed with mild cleanser of the offending agent, in contrast to ACD when there is a
in cool or tepid rather than hot water, and baths and transient worsening before healing occurs (crescendo phe-
showers should be limited to 5–10 minutes [23]. Adhesive nomenon) [23]. The prognosis for ICD is usually good if
tape on skin should be avoided [104] and oil‐based prod- contact with irritants can be aborted. Complete healing
ucts (e.g. petroleum jelly or bland oils) can be used to after removal of the irritants may take up to 4 weeks.
loosen any residual adhesive instead of nonmedical Although lesions of acute ICD may resolve within a
removers or acetone‐based products. week with appropriate management [48], it may take
more than 4 months for barrier function to normalize in
Specific medical treatment adults [23,178]. Therefore, protection measures need to be
Topical corticosteroids [50,172] continued after the skin lesion heals and regular moistur-
ICD is generally treated with topical corticosteroids [172] izing [23] is useful. Continuation of avoidance of irritants
due to their anti‐inflammatory effects and inhibitory is important as the symptoms of ICD may not reappear
effects on T‐cell activation and leucocyte migration [51]. until 1–2 weeks after re‐exposure.
Theoretically, their antiproliferative properties may pre- For ICD related to medical procedures and devices,
vent proper healing of the stratum corneum barrier [21] planning to reduce the risk of ICD is helpful [89]. To moti-
but short‐term use of a low‐ or mid‐potency topical corti- vate and educate patients for self‐care to prevent recur-
costeroid is usually helpful. Stronger halogenated topical rence of ICD, training programmes and even behavioural
corticosteroids such as fluocinonide or mometasone furo- modification [168,179] may be effective. Career counsel-
ate can be used to treat contact dermatitis reactions for a ling may be relevant to atopic adolescents or those already
short period (<2 weeks) in areas other than the face, axil- exhibiting features of contact dermatitis who are contem-
lae and groin. Occasionally, severe contact dermatitis plating high‐risk occupations for ICD [19,149].
requires systemic corticosteroid therapy for 10–14 days as
early discontinuation may lead to recurrence of the erup-
Summary. In summary, ICD in children is common and
tion [6]. However, studies on the effects of corticosteroids
the clinical problem and its management are age specific.
on ICD are far from unanimous [51] and their role still
The diagnosis of ICD requires a thorough history, detailed
warrants further studies [51].
clinical examination and a negative patch test. Avoidance
Topical immunomodulators of irritants and skin protection are the mainstay of treat-
Calcineurin inhibitors block signal transduction path- ment. More large‐scale studies are required to understand
ways of T cells and inhibit inflammatory cytokine pro- the full scope of ICD in children.
duction [51]. Both topical tacrolimus and pimecrolimus
References
were found to be effective in the treatment of ACD 1 Pelletier JL, Perez C, Jacob SE. Contact dermatitis in pediatrics.
[51,173]. Pimecrolimus cream 1% provides an option for Pediatr Ann 2016;45:e287–92.
treatment of ICD of the periocular region, head and neck 2 Pigatto P, Martelli A, Marsili C, Fiocchi A. Contact dermatitis in chil-
in adults, and marked improvement within 2–3 days of dren. Ital J Pediatr 2010;36:2.
3 Beltrani VS, Beltrani VP. Contact dermatitis. Ann Allergy Asthma
treatment with disease clearance in 2 weeks has been Immunol 1997;78:160–73; quiz 74–6.
reported [174]. Topical tacrolimus has been shown to 4 Warshaw EM, Paller AS, Fowler JF, Zirwas MJ. Practical management
improve susceptibility to skin irritation in 7 days [175] but of cutaneous reactions to the methylphenidate transdermal system:
recommendations from a dermatology expert panel consensus meet-
its intrinsic stinging properties may be a concern. The role ing. Clin Ther 2008;30:326–37.
of immunomodulators in the treatment of paediatric ICD 5 Loffler H, Effendy I. Prevention of irritant contact dermatitis. Eur J
needs to be further studied. Dermatol 2002;12:4–9.
Chapter 22  Irritant Contact Dermatitis 297

6 Friedlander SF. Consultation with the specialist: Contact dermatitis. 35 Schena D, Fantuzzi F, Girolomoni G. Contact allergy in chronic
Pediatr Rev 1998;19:166–71. eczematous lip dermatitis. Eur J Dermatol 2008;18:688–92.
7 Zhai H, Meier‐Davis SR, Cayme B et  al. Irritant contact dermatitis: 36 Nedorost ST. Medical Pearl: the evaluation of perioral dermatitis:
effect of age. Cutan Ocul Toxicol 2012;31:138–43. Use  of an extended patch test series. J Am Acad Dermatol
8 Le Maitre M, Crickx B, Lacour JP et al. Validation of a clinical evalua- 2007;56:S100–2.
tion score for irritative dermatitis: SCOREPI. J Eur Acad Dermatol 37 Butterworth T, Strean LP. Behavior disorders of interest to dermatolo-
Venereol 2013;27:1138–42. gists. Arch Dermatol 1963;88:859–67.
9 Fonacier L, Bernstein DI, Pacheco K et  al. Contact dermatitis: 38 Lund JJ, Unwala R, Xia L, Gottlieb V. Chemical scalp burns secondary
a  ­
practice parameter‐update 2015. J Allergy Clin Immunol Pract to the hair highlighting process: clinical and histopathologic features.
2015;3:S1–39. Pediatr Dermatol 2010;27:74–8.
10 Nijhawan RI, Matiz C, Jacob SE. Contact dermatitis: from basics to 39 Rougier A, Dupuis D, Lotte C et al. Regional variation in percutane-
allergodromes. Pediatr Ann 2009;38:99–108. ous absorption in man: measurement by the stripping method. Arch
11 Patil S, Maibach HI. Effect of age and sex on the elicitation of irritant Dermatol Res 1986;278:465–9.
contact dermatitis. Contact Dermatitis 1994;30:257–64. 40 Marrakchi S, Maibach HI. Sodium lauryl sulfate‐induced irritation in
12 Seyfarth F, Schliemann S, Antonov D, Elsner P. Dry skin, barrier func- the human face: regional and age‐related differences. Skin Pharmacol
tion, and irritant contact dermatitis in the elderly. Clin Dermatol Physiol 2006;19:177–80.
2011;29:31–6. 41 Huerth KA, Hawkes JE, Meyer LJ, Powell DL. The scourge of
13 Darlenski R, Fluhr JW. Influence of skin type, race, sex, and ana- the  spurge family–an imitator of Rhus dermatitis. Dermatitis
tomic  location on epidermal barrier function. Clin Dermatol 2012; 2016;27:372–81.
30:269–73. 42 Tiedemann D, Clausen ML, John SM et al. Effect of glove occlusion on
14 Robinson MK. Population differences in acute skin irritation the skin barrier. Contact Dermatitis 2016;74:2–10.

SECTION 4: OTHER TYPES


responses. Race, sex, age, sensitive skin and repeat subject compari- 43 Gimenez‐Arnau A, Silvestre JF, Mercader P et al. Shoe contact derma-
sons. Contact Dermatitis 2002;46:86–93. titis from dimethyl fumarate: clinical manifestations, patch test

OF DERMATITIS
15 Robinson MK. Population differences in skin structure and physiol- results, chemical analysis, and source of exposure. Contact Dermatitis
ogy and the susceptibility to irritant and allergic contact dermatitis: 2009;61:249–60.
implications for skin safety testing and risk assessment. Contact 44 Chougule A, Thappa DM. Patterns of lower leg and foot eczema in
Dermatitis 1999;41:65–79. South India. Indian J Dermatol Venereol Leprol 2008;74:458–61.
16 Robinson MK. Racial differences in acute and cumulative skin irrita- 45 Fischer G, Rogers M. Vulvar disease in children: a clinical audit of 130
tion responses between Caucasian and Asian populations. Contact cases. Pediatr Dermatol 2000;17:1–6.
Dermatitis 2000;42: 134–43. 46 Shab A, Matterne U, Diepgen TL, Weisshaar E. Are obsessive‐compul-
17 de Jongh CM, Khrenova L, Verberk MM et al. Loss‐of‐function pol- sive disorders and personality disorders sufficiently considered in
ymorphisms in the filaggrin gene are associated with an increased occupational dermatoses? A discussion based on three case reports.
susceptibility to chronic irritant contact dermatitis: a case‐control J Dtsch Dermatol Ges 2008;6:947–51.
study. Br J Dermatol 2008;159:621–7. 47 Koblenzer CS. Psychiatric syndromes of interest to dermatologists.
18 Spiewak R. Contact dermatitis in atopic individuals. Curr Opin Int J Dermatol 1993;32:82–8.
Allergy Clin Immunol 2012;12:491–7. 48 Almis H, Bucak IH, Tekin M et  al. Acute irritant bullous contact
19 Williams J, Cahill J, Nixon R. Occupational autoeczematization or dermatitis caused by Euphorbia helioscopia. Contact Dermatitis
­
atopic eczema precipitated by occupational contact dermatitis? 2015;72:184–5.
Contact Dermatitis 2007;56:21–6. 49 Nijhawan RI, Jacob SE. Connubial dermatitis revisited: mother‐to‐
20 Allen MH, Wakelin SH, Holloway D et al. Association of TNFA gene child contact dermatitis. Dermatitis 2009;20:55–6.
polymorphism at position ‐308 with susceptibility to irritant contact 50 Gulec AI, Albayrak H, Uslu E et  al. Pustular irritant contact
dermatitis. Immunogenetics 2000;51:201–5. ­dermatitis caused by dexpanthenol in a child. Cutan Ocul Toxicol
21 Levin CY, Maibach HI. Irritant contact dermatitis: is there an immu- 2015;34:75–6.
nologic component? Int Immunopharmacol 2002;2:183–9. 51 Cohen DE, Heidary N. Treatment of irritant and allergic contact
22 Weitz NA, Lauren CT, Weiser JA et  al. Chlorhexidine gluconate‐ ­dermatitis. Dermatol Ther 2004;17:334–40.
impregnated central access catheter dressings as a cause of erosive 52 D’Arcy WG. Letter: Severe contact dermatitis from poinsettia. Arch
contact dermatitis: a report of 7 cases. JAMA Dermatol 2013;149:195–9. Dermatol 1974;109:909–10.
23 Slodownik D, Lee A, Nixon R. Irritant contact dermatitis: a review. 53 Litvinov IV, Sugathan P, Cohen BA. Recognizing and treating toilet‐
Australas J Dermatol 2008;49:1–9; quiz 10–11. seat contact dermatitis in children. Pediatrics 2010;125:e419–22.
24 Effendy I, Loffler H, Maibach HI. Epidermal cytokines in murine 54 Lakshmi C, Srinivas CR, Anand CV, Mathew AC. Irritancy ranking of
cutaneous irritant responses. J Appl Toxicol. 2000;20:335–41. 31 cleansers in the Indian market in a 24‐h patch test. Int J Cosmet Sci
25 Hunziker T, Brand CU, Kapp A et al. Increased levels of inflammatory 2008;30:277–83.
cytokines in human skin lymph derived from sodium lauryl sulphate‐ 55 Garland JS, Alex CP, Mueller CD et al. A randomized trial comparing
induced contact dermatitis. Br J Dermatol 1992;127:254–7. povidone‐iodine to a chlorhexidine gluconate‐impregnated dressing
26 Eberhard Y, Ortiz S, Ruiz Lascano A et  al. Up‐regulation of the for prevention of central venous catheter infections in neonates.
chemokine CCL21 in the skin of subjects exposed to irritants. BMC Pediatrics 2001;107:1431–6.
Immunol 2004;5:7. 56 Uter W, Gefeller O, Schwanitz HJ. An epidemiological study of the
27 Modi GM, Doherty CB, Katta R, Orengo IF. Irritant contact dermatitis influence of season (cold and dry air) on the occurrence of irritant skin
from plants. Dermatitis 2009;20:63–78. changes of the hands. Br J Dermatol 1998;138:266–72.
28 Smith HR, Basketter DA, McFadden JP. Irritant dermatitis, irritancy 57 Hu CH. Sweat‐related dermatoses: old concept and new scenario.
and its role in allergic contact dermatitis. Clin Exp Dermatol 2002; Dermatologica 1991;182:73–6.
27:138–46. 58 Powell D, Ahmed S. Soccer shin guard reactions: allergic and irritant
29 Chew A, Maibach H. Irritant Dermatitis. Berlin: Springer, 2006. reactions. Dermatitis 2010;21:162–6.
30 Poppe H, Poppe LM, Brocker EB, Trautmann A. Do‐it‐yourself cement 59 Lenkovic M, Cabrijan L, Gruber F et  al. Phytophotodermatitis in
work: the main cause of severe irritant contact dermatitis requiring Rijeka region, Croatia. Coll Antropol 2008;32(suppl 2):203–5.
hospitalization. Contact Dermatitis 2013;68:111–15. 60 Neri I, Bianchi F, Giacomini F, Patrizi A. Acute irritant contact
31 Malten KE, den Arend JA, Wiggers RE. Delayed irritation: hexanediol ­dermatitis due to Juglans regia. Contact Dermatitis 2006;55:62–3.
diacrylate and butanediol diacrylate. Contact Dermatitis 61 Massmanian A. Contact dermatitis due to Euphorbia pulcher-
1979;5:178–84. rima  Willd, simulating a phototoxic reaction. Contact Dermatitis
32 Cook KA, Kelso JM. Surgery‐related contact dermatitis: a review of 1998;38:113–14.
potential irritants and allergens. J Allergy Clin Immunol Pract 62 Krenzelok EP, Jacobsen TD, Aronis JM. Poinsettia exposures
2017;5:1234–40. have  good outcomes … just as we thought. Am J Emerg Med
33 Morris‐Jones R, Robertson SJ, Ross JS et  al. Dermatitis caused by 1996;14:671–4.
physical irritants. Br J Dermatol 2002;147:270–5. 63 Assaf M, Nofal E, Nofal A et al. Paederus dermatitis in Egypt: a clin-
34 Yokota M, Maibach HI. Moisturizer effect on irritant dermatitis: an icopathological and ultrastructural study. J Eur Acad Dermatol
overview. Contact Dermatitis 2006;55:65–72. Venereol 2010;24:1197–201.
298 Section 4  Other Types of Dermatitis

64 Rahmah E, Norjaiza MJ. An outbreak of Paederus dermatitis in a pri- 94 Hensten‐Pettersen A, Jacobsen N. Toxic effects of dental materials.
mary school, Terengganu, Malaysia. Malays J Pathol 2008;30:53–6. Int Dent J 1991;41:265–73.
65 Qadir SN, Raza N, Rahman SB. Paederus dermatitis in Sierra Leone. 95 Lyon CC, Smith AJ, Griffiths CE, Beck MH. The spectrum of skin dis-
Dermatol Online J 2006;12:9. orders in abdominal stoma patients. Br J Dermatol 2000;143:1248–60.
66 Banney LA, Wood DJ, Francis GD. Whiplash rove beetle dermatitis in 96 Cressey BD, Belum VR, Scheinman P et al. Stoma care products rep-
central Queensland. Australas J Dermatol 2000;41:162–7. resent a common and previously underreported source of peristomal
67 Zargari O, Kimyai‐Asadi A, Fathalikhani F, Panahi M. Paederus contact dermatitis. Contact Dermatitis 2017;76:27–33.
­dermatitis in northern Iran: a report of 156 cases. Int J Dermatol 97 Sportiello L, Cammarota S, de Portu S, Sautebin L. Notification of
2003;42:608–12. undesirable effects of cosmetics and toiletries. Pharmacol Res
68 Sendur N, Savk E, Karaman G. Paederus dermatitis: a report of 46 2009;59:101–6.
cases in Aydin, Turkey. Dermatology 1999;199:353–5. 98 Kuller JM. Infant skin care products: what are the issues? Adv
69 Huang C, Liu Y, Yang J et  al. An outbreak of 268 cases of Paederus Neonatal Care 2016;16(suppl 5S):S3–12.
dermatitis in a toy‐building factory in central China. Int J Dermatol 99 Cork M, Murphy R, Carr J et al. Allergens: The rising prevalence of
2009;48:128–31. atopic eczema and environmental trauma to the skin. Dermatol
70 Vega ML, Vega J, Vega JM et al. Cutaneous reactions to pine proces- Pract 2002;10:22–7.
sionary caterpillar (Thaumetopoea pityocampa) in pediatric popula- 100 Tan CH, Rasool S, Johnston GA. Contact dermatitis: allergic and
tion. Pediatr Allergy Immunol 2003;14:482–6. irritant. Clin Dermatol 2014;32:116–24.
71 Bonamonte D, Foti C, Vestita M, Angelini G. Skin reactions to pine 101 Loo WJ. Irritant dermatitis due to prolonged contact with Oilatum
processionary caterpillar Thaumetopoea pityocampa Schiff. Scientific Plus. Br J Dermatol 2003;148:171–2.
World Journal 2013;2013:867431. 102 Ling TC, Highet AS. Irritant reactions to an antiseptic bath emollient.
72 Hossler EW. Caterpillars and moths: Part II. Dermatologic manifesta- J Dermatolog Treat 2000;11:263–7.
SECTION 4: OTHER TYPES

tions of encounters with Lepidoptera. J Am Acad Dermatol 103 Heurung AR, Raju SI, Warshaw EM. Adverse reactions to sunscreen
2010;62:13–28; quiz 9–30. agents: epidemiology, responsible irritants and allergens, clinical
OF DERMATITIS

73 Foti C, Romita P, Borghi A et  al. Contact dermatitis to topical acne characteristics, and management. Dermatitis 2014;25:289–326.
drugs: a review of the literature. Dermatol Ther 2015;28:323–9. 104 Alavi A, Sibbald RG, Ladizinski B et al. Wound‐related allergic/irritant
74 Ockenfels HM, Uter W, Lessmann H et al. Patch testing with benzoyl contact dermatitis. Adv Skin Wound Care 2016;29:278–86.
peroxide: reaction profile and interpretation of positive patch test 105 Chan HP, Maibach HI. Hair highlights and severe acute irritant
reactions. Contact Dermatitis 2009;61:209–16. ­dermatitis (“burn”) of the scalp. Cutan Ocul Toxicol 2010;29:229–33.
75 Krayenbuhl BH, Elsner P. Allergic and irritant contact dermatitis to 106 Gallego H, Lewis EJ, Crutchfield CE 3rd. Crystal deodorant
calcipotriol. Am J Contact Dermat 1999;10:78–80. ­dermatitis: irritant dermatitis to alum‐containing deodorant. Cutis
76 Loffler H, Effendy I, Happle R. Skin susceptibility to dithranol: con- 1999;64:65–6.
tact allergy or irritation? Eur J Dermatol 1999;9:32–4. 107 Swaile DF, Elstun LT, Benzing KW. Clinical studies of sweat rate
77 Paghdal KV, Schwartz RA. Topical tar: back to the future. J Am Acad reduction by an over‐the‐counter soft‐solid antiperspirant and com-
Dermatol 2009;61:294–302. parison with a prescription antiperspirant product in male panelists.
78 Yu KJ, Chen HH, Chang YC et al. Ulcerative irritant contact dermatitis Br J Dermatol 2012;166(suppl 1):22–6.
from lindane. Contact Dermatitis 2005;52:118–19. 108 Tsang M, Guy RH. Effect of Aqueous Cream BP on human stratum
79 Neri I, Savoia F, Guareschi E et al. Purpura after application of EMLA corneum in vivo. Br J Dermatol 2010;163:954–8.
cream in two children. Pediatr Dermatol 2005;22:566–8. 109 Storer E, Koh KJ, Warren L. Severe contact dermatitis as a result of an
80 Dong H, Kerl H, Cerroni L. EMLA cream‐induced irritant contact der- antiseptic bath oil. Australas J Dermatol 2004;45:73–5.
matitis. J Cutan Pathol 2002;29:190–2. 110 Mathias CG. Contact dermatitis in children due to face paints. Cutis
81 Ismail F, Goldsmith PC. Emla cream‐induced allergic contact derma- 1980;26:584–5.
titis in a child with thalassaemia major. Contact Dermatitis 2005;52:111. 111 Weston WL, Morelli JG. Dermatitis under soccer shin guards: allergy
82 Nelson MU, Bizzarro MJ, Dembry LM et al. One size does not fit all: or contact irritant reaction? Pediatr Dermatol 2006;23:19–20.
why universal decolonization strategies to prevent methicillin‐resist- 112 Yoshida M, Oiso N, Kawada A. Hyperkeratosis and frictional der-
ant Staphylococcus aureus colonization and infection in adult inten- matitis from practicing kendo. Case Rep Dermatol 2010;2:65–8.
sive care units may be inappropriate for neonatal intensive care units. 113 Inui S, Yamamoto S, Ikegami R et al. Baseball pitcher’s friction der-
J Perinatol 2014;34:653–5. matitis. Contact Dermatitis 2002;47:176–7.
83 Liippo J, Kousa P, Lammintausta K. The relevance of chlorhexidine 114 Peca G, Rafanelli S, Galassi G et al. Contact reactions to the jellyfish
contact allergy. Contact Dermatitis 2011;64:229–34. Carybdea marsupialis: observation of 40 cases. Contact Dermatitis
84 O’Grady NP, Alexander M, Burns LA et al. Summary of recommenda- 1997;36:124–6.
tions: Guidelines for the Prevention of Intravascular Catheter‐related 115 Patruno C, Napolitano M, La Bella S et al. Instrument‐related skin
Infections. Clin Infect Dis 2011;52:1087–99. disorders in musicians. Dermatitis 2016;27:26–9.
85 Chapman AK, Aucott SW, Milstone AM. Safety of chlorhexidine glu- 116 Miller S, Helms A, Brodell RT. Occlusive irritant dermatitis: when is
conate used for skin antisepsis in the preterm infant. J Perinatol “allergic” contact dermatitis not allergic? Skinmed 2007;6:97–8.
2012;32:4–9. 117 Caero JE, Cohen PR. Fiddler’s neck: Chin rest‐associated irritant
86 Lashkari HP, Chow P, Godambe S. Aqueous 2% chlorhexidine‐ contact dermatitis and allergic contact dermatitis in a violin player.
induced chemical burns in an extremely premature infant. Arch Dis Dermatol Online J 2012;18:10.
Child Fetal Neonatal Ed 2012;97:F64. 118 Kaufman BH, Hoffman AD, Zimmerman D. Fiddler’s neck in a
87 Okano M. Irritant contact dermatitis caused by povidone‐iodine. child. J Pediatr 1988;113:89–90.
J Am Acad Dermatol 1989;20:860. 119 Warshaw EM, Botto NC, Zug KA et al. Contact dermatitis associated
88 Lee JY, Wang BJ. Contact dermatitis caused by cetrimide in antisep- with food: retrospective cross‐sectional analysis of North American
tics. Contact Dermatitis 1995;33:168–71. Contact Dermatitis Group data, 2001‐2004. Dermatitis 2008;19:252–60.
89 Hitchcock J, Savine L. Medical adhesive‐related skin injuries associ- 120 Bahna SL. Adverse food reactions by skin contact. Allergy
ated with vascular access. Br J Nurs 2017;26:S4–12. 2004;59(suppl 78):66–70.
90 Noonan C, Quigley S, Curley MA. Skin integrity in hospitalized 121 Bauer A, Bartsch R, Hersmann C et al. Occupational hand dermatitis
infants and children: a prevalence survey. J Pediatr Nurs 2006; in food industry apprentices: results of a 3‐year follow‐up cohort
21:445–53. study. Int Arch Occup Environ Health 2001;74:437–42.
91 Mak MF, Li W, Mahadev A. Calamine lotion to reduce skin irritation 122 Brancaccio RR, Alvarez MS. Contact allergy to food. Dermatol Ther
in children with cast immobilisation. J Orthop Surg (Hong Kong) 2004;17:302–13.
2013;21:221–5. 123 Polunin I. Pineapple dermatosis. Br J Dermatol 1951;63:441–55.
92 O’Brien L, Jones DJ. Silicone gel sheeting for preventing and treating 124 Veien NK, Hattel T, Laurberg G. Can ’label dermatitis’ become
hypertrophic and keloid scars. Cochrane Database Syst Rev ’creeping neurotic excoriations’? Contact Dermatitis 1992;27:272–3.
2013:Cd003826. 125 Lazarov A, Cordoba M, Plosk N, Abraham D. Atypical and unusual
93 Goiato MC, dos Santos DM, Haddad MF, Moreno A. Rehabilitation clinical manifestations of contact dermatitis to clothing (textile con-
with ear prosthesis linked to osseointegrated implants. Gerodontology tact dermatitis): case presentation and review of the literature.
2012;29:150–4. Dermatol Online J 2003;9:1.
Chapter 22  Irritant Contact Dermatitis 299

126 Eby CS, Jetton RL. School desk dermatitis. Primary irritant contact 155 Oiso N, Fukai K, Ishii M. Irritant contact dermatitis from benza-
dermatitis to fiberglass. Arch Dermatol 1972;105:890–1. lkonium chloride in shampoo. Contact Dermatitis 2005;52:54.
127 Lovell CR. Phytodermatitis. Clin Dermatol 1997;15:607–13. 156 Stables GI, Wilkinson SM. Allergic contact dermatitis due to pan-
128 Fluhr JW, Akengin A, Bornkessel A et al. Additive impairment of the thenol. Contact Dermatitis 1998;38:236–7.
barrier function by mechanical irritation, occlusion and sodium lau- 157 Kind F, Hofmeier KS, Bircher AJ. Irritant contact dermatitis from a
ryl sulphate in vivo. Br J Dermatol 2005;153:125–31. black henna tattoo without sensitization to para‐phenylendiamine.
129 Karim SA. From ’playstation thumb’ to ’cellphone thumb’: the new Pediatrics 2013;131:e1974–6.
epidemic in teenagers. S Afr Med J 2009;99:161–2. 158 Yagami A, Kawai N, Kosai N et  al. Occupational allergic contact
130 Vaidya HJ. Playstation thumb. Lancet 2004;363:1080. ­dermatitis due to dimethyl sulfate following sensitization from a
131 Corazza M, Minghetti S, Bertoldi AM et  al. Modern electronic severe acute irritant reaction to the reagent. Contact Dermatitis
devices: an increasingly common cause of skin disorders in con- 2009;60:183–4.
sumers. Dermatitis 2016;27:82–9. 159 Warshaw EM, Ahmed RL, Belsito DV et  al. Contact dermatitis
132 Jacob SE. Xbox—a source of nickel exposure in children. Pediatr of  the  hands: cross‐sectional analyses of North American Contact
Dermatol 2014;31:115–6. Dermatitis Group Data, 1994‐2004. J Am Acad Dermatol 2007;
133 Jacob SE, Admani S. iPad—increasing nickel exposure in children. 57:301–14.
Pediatrics 2014;134:e580–2. 160 Chan JC, Sullivan PJ, O’Sullivan MJ, Eadie PA. Full thickness burn
134 Arnold AW, Itin PH. Laptop computer‐induced erythema ab igne in caused by exposure to giant hogweed: delayed presentation, histo-
a child and review of the literature. Pediatrics 2010;126:e1227–30. logical features and surgical management. J Plast Reconstr Aesthet
135 Karlsson M, Linde YW. [Erythema ab igne—classical dermatological Surg. 2011;64:128–30.
diagnosis hot again. Laptop computer dermatitis most common in 161 Goldenberg A, Mousdicas N, Silverberg N et  al. Pediatric contact
young people]. Lakartidningen 2010;107:1844. dermatitis registry inaugural case data. Dermatitis 2016;27:293–302.

SECTION 4: OTHER TYPES


136 Bilic M, Adams BB. Erythema ab igne induced by a laptop computer. 162 Astner S, Gonzalez S, Gonzalez E. Noninvasive evaluation of
J Am Acad Dermatol 2004;50:973–4. allergic  and irritant contact dermatitis by in vivo reflectance
­

OF DERMATITIS
137 Jagtman BA. Erythema ab igne due to a laptop computer. Contact ­confocal microscopy. Dermatitis 2006;17:182–91.
Dermatitis 2004;50:105. 163 Slodownik D, Levi A, Lapidoth M et al. Noninvasive in vivo confocal
138 Brazzelli V, Grassi S, Barruscotti S et al. Erythema ab igne induced by laser scanning microscopy is effective in differentiating allergic
laptop computer: an emerging disease among adolescents? G Ital from  nonallergic equivocal patch test reactions. Lasers Med Sci
Dermatol Venereol 2015. [Epub ahead of print]. 2015;30:1081–7.
139 Djerrou Z, Djaalab H, Riachi F et  al. Irritantcy potential and sub 164 Boone MA, Jemec GB, Del Marmol V. Differentiating allergic and
acute dermal toxicity study of Pistacia lentiscus fatty oil as a irritant contact dermatitis by high‐definition optical coherence
­topical  traditional remedy. Afr J Tradit Complement Altern Med tomography: a pilot study. Arch Dermatol Res 2015;307:11–22.
2013;10:480–9. 165 Levin CY, Maibach HI. Do cool water or physiologic saline com-
140 Teshome K, Gebre‐Mariam T, Asres K, Engidawork E. Toxicity presses enhance resolution of experimentally‐induced irritant con-
­studies on dermal application of plant extract of Dodonaea viscosa tact dermatitis? Contact Dermatitis 2001;45:146–50.
used in Ethiopian traditional medicine. Phytother Res 2010;24:60–9. 166 Levin CY, Maibach HI. Irritant contact dermatitis: is there an immu-
141 Lee TY, Lam TH. Contact dermatitis due to topical treatment with nologic component? Int Immunopharmacol 2002;2:183–9.
garlic in Hong Kong. Contact Dermatitis 1991;24:193–6. 167 Mostosi C, Simonart T. Effectiveness of barrier creams against irri-
142 Cappelletti EM, Trevisan R, Caniato R. External antirheumatic and tant contact dermatitis. Dermatology 2016;232:353–62.
antineuralgic herbal remedies in the traditional medicine of north‐ 168 Saary J, Qureshi R, Palda V et al. A systematic review of contact der-
eastern Italy. J Ethnopharmacol 1982;6:161–90. matitis treatment and prevention. J Am Acad Dermatol 2005;53:845.
143 Lee TY, Lam TH. Irritant contact dermatitis due to a Chinese herbal 169 Buraczewska I, Berne B, Lindberg M et al. Changes in skin barrier
medicine lu‐shen‐wan. Contact Dermatitis 1988;18:213–18. function following long‐term treatment with moisturizers, a rand-
144 Tsai KS, Lin TC, Wu MT et al. Irritant contact dermatitis risk of com- omized controlled trial. Br J Dermatol 2007;156:492–8.
mon topical traditional chinese medicines used for skin‐lightening: a 170 Held E, Agner T. Effect of moisturizers on skin susceptibility to irri-
pilot clinical trial with 30 volunteers. Evid Based Complement tants. Acta Derm Venereol 2001;81:104–7.
Alternat Med 2014;2014:609064. 171 Pierard GE, Arrese JE, Rodriguez C, Daskaleros PA. Effects of sof-
145 Schloemer JA, Zirwas MJ, Burkhart CG. Airborne contact dermatitis: tened and unsoftened fabrics on sensitive skin. Contact Dermatitis
common causes in the USA. Int J Dermatol 2015;54:271–4. 1994;30:286–91.
146 Handa S, De D, Mahajan R. Airborne contact dermatitis  –  current 172 English JS. Current concepts of irritant contact dermatitis. Occup
perspectives in etiopathogenesis and management. Indian J Dermatol Environ Med 2004;61:722–6, 674.
2011;56:700–6. 173 Han JS, Won KH, Chang SE, Kim JE. Tacrolimus 0.1% ointment in the
147 Eedy DJ. Carbon‐fibre‐induced airborne irritant contact dermatitis. treatment of allergic contact dermatitis: a new approach. Int J
Contact Dermatitis 1996;35:362–3. Dermatol 2014;53:e470–1.
148 Patiwael JA, Wintzen M, Rustemeyer T, Bruynzeel DP. Airborne irri- 174 Mensing CO, Mensing CH, Mensing H. Treatment with pimecroli-
tant contact dermatitis due to synthetic fibres from an air‐conditioning mus cream 1% clears irritant dermatitis of the periocular region, face
filter. Contact Dermatitis 2005;52:126–9. and neck. Int J Dermatol 2008;47:960–4.
149 Dickel H, Bruckner TM, Schmidt A, Diepgen TL. Impact of atopic 175 Jungersted JM, Hogh JK, Hellegren LI et al. Effects of topical corti-
skin diathesis on occupational skin disease incidence in a working costeroid and tacrolimus on ceramides and irritancy to sodium
population. J Invest Dermatol 2003;121:37–40. ­lauryl sulphate in healthy skin. Acta Derm Venereol 2011;91:290–4.
150 Mendiratta V, Sarkar R, Sharma RC. Lymphangioma circumscrip- 176 Rosen K, Mobacken H, Swanbeck G. Chronic eczematous dermatitis
tum masquerading as irritant contact dermatitis. J Dermatol of the hands: a comparison of PUVA and UVB treatment. Acta Derm
1999;26:474–5. Venereol 1987;67:48–54.
151 Barradell R, Addo A, McDonagh AJ et  al. Phytophotodermatitis 177 Simons JR, Bohnen IJ, van der Valk PG. A left‐right comparison of
mimicking child abuse. Eur J Pediatr 1993;152:291–2. UVB phototherapy and topical photochemotherapy in bilateral
152 MacDuff S, Sun J, Bell DE et  al. Accidental burn by intentional chronic hand dermatitis after 6 weeks’ treatment. Clin Exp Dermatol
­laxative use. Pediatr Emerg Care 2016;32:541–3. 1997;22:7–10.
153 Mehta AJ, Statham BN. Phytophotodermatitis mimicking non‐ 178 Choi JM, Lee JY, Cho BK. Chronic irritant contact dermatitis: recov-
accidental injury or self‐harm. Eur J Paediatr 2007;166:751–2. ery time in man. Contact Dermatitis 2000;42:264–9.
154 Tomankova K, Kejlova K, Binder S et  al. In vitro cytotoxicity 179 Andersen F, Andersen KE. Management of irritant contact dermati-
and  phototoxicity study of cosmetics colorants. Toxicol In Vitro tis: continuously a problem for patients and dermatologists. G Ital
2011;25:1242–50. Dermatol Venereol 2008;143:207–12.
300 

CHA PTER  2 3

Allergic Contact Dermatitis


Sharon E. Jacob1, Hannah Hill2 & Alina Goldenberg3
1
 Loma Linda University, Loma Linda, CA, USA
2
 Department of Dermatology, Mayo Clinic, Scottsdale, AZ, USA
3
 Department of Dermatology, University of California, San Diego, CA, USA

Abstract (AD), even more so when they are concurrent. Furthermore, the
barrier disruption inherent to AD appears to increase the risk for
developing ACD.
In the last decade, allergic contact dermatitis (ACD) has become
The gold standard diagnostic tool for ACD is the epicutaneous
an increasingly recognized diagnosis in children presenting with
patch test. Confirmation of clinically relevant contact allergens is
eczematous skin. Although the immunological pathway of a
­
critical in order to direct avoidance and appropriately manage the
SECTION 4: OTHER TYPES

­delayed hypersensitivity reaction is specifically distinct, it can be


disease. This chapter discusses the epidemiology, ­pathophysiology
phenotypically difficult to distinguish from the clinical presentations
and clinical features of ACD in the context of the available ­evidence
OF DERMATITIS

of chronic irritant contact dermatitis (ICD) and atopic dermatitis


in the literature.

Key points • The most common allergens in children share some overlap with
those seen in adults and include: metals (such as nickel, cobalt),
antibiotics (neomycin, bacitracin), fragrances and other plant‐
• Allergic contact dermatitis is increasingly being recognized as a
related exposures, preservatives, surfactants and emulsifiers, lano-
common problem among children presenting with eczematous
lin, rubber‐ and leather‐related agents, corticosteroids and dyes.
skin.
• In some cases, systemic exposure to contact allergens can also
• Comprehensive epicutaneous patch testing, while not approved
result in widespread dermatitis, as can be seen with nickel and
for children by the US Food and Drug Administration (FDA), can
propylene glycol.
be performed on children by experienced clinicians. However,
• Once allergens are identified, patient education to help provide
the selection of allergens for testing and the concentrations of
guidance for prevention of re‐exposures to allergens is especially
some allergens may need to be titrated for use in children.
important.

Epidemiology. In 1931, Straus found that neonates have Norwegian schoolchildren aged 7–12 years screened with
the potential for developing contact sensitization to 20 contact allergens found that 23% had at least one PPTR,
Rhus – and thus proved that even immature skin has the most frequently to nickel, and in one third of cases,
capacity to develop delayed‐type hypersensitivity sensitization to nickel was associated with contact
­
­reactions [1]. In 1986, Weston et al. patch‐tested 314 well ­dermatitis [5].
children, of whom 20% experienced at least one positive A review of more recent patch test studies (interna­
patch test reaction. With this same study, the authors con­ tional and national) shows prevalence rates of at least one
cluded the safety and efficacy of the commercially avail­ positive patch test between 25.1% and 95.6% in children
able American Academy of Dermatology (AAD) patch referred for suspected ACD (Table  23.1). Extrapolating
test kit in infants, children and adolescents [2]. To date, from both the domestic and international prevalence rates
this is the only general paediatric population‐based study in both well and afflicted children, the conservative
to enroll more than 100 children for patch testing in the ­estimate is that between 1.5% and 5.4% of the general
USA. That said, comparative analysis suggests that aller­ population has ACD. Applying these rates to the current
gens causing significant morbidities and cost burden are US population of children of 72 million, it is estimated
similar in American, Canadian and European children, that there are at least one million incident cases of ACD in
where investigators have examined the frequencies of children each year [16].
contact allergy in unselected populations [3]. For exam­ The prevalence of ACD represents a considerable cost
ple, a Danish cross‐sectional analysis of 1501 Odense burden, both in direct and indirect patient morbidity and
schoolchildren aged 12–16 years demonstrated that 15% to the general healthcare system. An estimated 250 000 US
had at least one positive patch test reaction (PPTR) (8% to children are affected by nickel contact dermatitis each
nickel and 2% to fragrance mix) and 7% had associated year, which represents one quarter of at least one million
past or present ACD [4]. Likewise, a similar survey of 424 children affected with ACD each year. Denmark reported

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 23  Allergic Contact Dermatitis 301

Table 23.1  Review of 2010–2015 paediatric patch test studies

Author/date/ Study n Patient population/series tested Findings


country period

Milingou et al. 1994–2007 255 Children <15 years old • 60% displayed 1+ PPT
Greece 2010 Modified European basic standard series • Most frequent allergens nickel, thiomersal (thimerosal),
[6] (Trolab, Hermal, Germany), and with cobalt, potassium dichromate
additional series as indicated
Fortina et al. 2002–2008 321 Children <3 years old • 62.3% displayed 1+ PPT
2010 Italy [7] Paediatric standard series of 30 allergens • Most frequent allergens nickel, potassium dichromate,
CAPB, cobalt, neomycin, MCI/MI
• Rates of contact sensitization similar between children
with and without AD
Sarma et al. 2005–2008 70 Children 5–15 years old • 80% displayed 1+ PPT
2010 Indian standard battery • Top allergens parabens, potassium dichromate,
India [8] fragrance mix, cobalt, PPD, neomycin
Jacob et al. 2004–2006 45 Children 6 months to 18 years old • 95.6% displayed 1+ PPT

SECTION 4: OTHER TYPES


2010 Custom‐tailored patch test series • 76.7% of children in this study had history of AD
USA [9] • Those tested with history of AD were more likely to
react to CAPB and disperse dyes

OF DERMATITIS
Toledo et al. 2005–2008 111 Children 15 years and younger • 46.8% displayed 1+ PPT
2011 Spanish baseline series • Top allergens nickel sulphate, MCI/MI, fragrance mix,
Spain [10] balsam of Peru
Kuljanac et al. 1994–2009 412 Children <18 ears old • 26% displayed 1+ PPT
2011 Standard series of allergens manufactured • Most frequent allergens nickel, thiomersal, cobalt,
Croatia [11] in Croatia fragrance, white mercury, formaldehyde
Jacob et al. 2008–2009 102 Children 6–18 years • 76.2% PPT
2011 T.R.U.E. test ® • Most frequent allergens nickel, p‐tert‐butylphenol
USA [12] formaldehyde resin, wool alcohols, fragrance and cobalt
• Demonstrated T.R.U.E. test® is safe and efficacious in
paediatric population
Schena et al. 2004–2011 349 Children 0–15 years old • 69.3% PPT
2012 Standard series of allergens of the SIDAPA • Younger children showed higher sensitization rates
Italy [13] • Most frequent allergens nickel, cobalt, MCI/MI,
potassium dichromate, fragrance, neomycin
• Patients with AD had more widespread dermatitis than
those without AD
Silny et al. 2008–2011 155 Children 1–20 years old (104 with AD, 15 • 45.2% of patients with AD had PPT
2013 with seborrhoeic dermatitis, 36 healthy) • 13.9% of healthy patients had PPT
Poland [14] 1–5 years: paediatric set (established in our • Highest frequency of PPT was in youngest age group
department) consisting of 12 haptens 1–5 years old
6–20 years old: European standard series • Highest‐frequency allergens were nickel, cobalt and
chromium
Zug et al. 2005–2012 883 Children 18 years and younger suspected • 62.3% 1+ PPT
2014 of ACD • Most frequent allergens nickel, cobalt, neomycin,
USA, Canada [15] Tested with variants of screening series from balsam of Peru and lanolin alcohol 50% petrolatum
NACDG and/or supplemental allergens

ACD, allergic contact dermatitis; AD, atopic dermatitis; CAPB, cocamidopropyl betaine; MCI/MI, methylchloroisothiazolinone/methylisothiazolinone;
NACDG, North American Contact Dermatitis Group; PPD, para‐phenylenediamine; PPT, positive patch test; SIDAPA, Società Italiana di Dermatologia
Allergologica Professionale e Ambientale; T.R.U.E., thin‐layer rapid use epicutaneous.

cost savings of $2 billion over the past 20‐year period occurs when patients are incorrectly assigned into ACD
since adoption of legislation that limited the level of ‘affected’ and ‘unaffected’ groups. In particular, in the
nickel that could be released from items intended for sus­ context of AD, many study participants have been reflex­
tained contact with the skin [17]. As the US population ively classified into a presumed AD‐affected category
(318 881 992) is 56 times that of Denmark (5 627 235), US without full diagnostic work‐up and exclusion of other
adoption of a similar nickel directive has been estimated diagnoses, such as ACD. This ultimately leads to null‐
to project savings of $5.7 billion per year, ultimately favouring results and further complicates final diagnosis.
­reaching $113 billion in savings over the next 20‐year Finally, of considerable note, unlike the global evaluation
period [16]. of comorbidities prevalent to the study of psoriasis, the
When it comes to correctly reporting the epidemiology incidence of contact allergy and ACD in relation to envi­
of ACD and identifying associated risk factors, the litera­ ronmental exposures, concomitant prevalence of AD and
ture may be studded with misclassification bias which risk factors for sensitization are yet to be elucidated.
302 Section 4  Other Types of Dermatitis

­ eterinarians and athletes [22]. Persons with underlying


v
Pathogenesis. There are two phases in the mechanism of
atopy‐related skin diseases or other chronic skin barrier
ACD: sensitization/induction and elicitation. Both phases defects or inflammation are also at higher risk. In particu­
require a complex interaction to occur between the cells lar, children with AD have a greater likelihood of having
typically residing in the skin and cells of the immune a relative positive patch test than those without AD
­system that are recruited to the epidermis in contact with [23,24]. Furthermore, the standard treatment for AD
the allergen. For ‘sensitization’ to occur, lipophilic hap­ requires application of multiple topical preparations
tens (low‐molecular‐weight chemicals) must penetrate potentially containing sensitizing chemicals to dermatitic
the skin. Although this process is not dependent on the skin. Exposure of the skin to entrapped haptens under
epidermal barrier being impaired, it occurs more readily occlusion or in moist environments is also believed to
if this is the case. Once the hapten has penetrated the increase the risk of developing contact allergy to a chemi­
­epidermis, it combines with endogenous protein, forming cal [25–28]. An example of this is development of allergy
the hapten–protein complex. When a dendritic cell to textile dyes from clothing or cosmetics that are in
encounters the hapten–protein complex, it releases inter­ ­prolonged contact with skin. In a similar manner, irritant
leukin (IL)‐1β, resulting in nearby mast cell, keratinocyte contact dermatitis has been associated with the develop­
and endothelial cell release of tumour necrosis factor ment of ACD, because the epidermal barrier is disrupted
(TNF)‐α, IL‐1 and IL‐18. Eosinophils and IL‐5 have also
SECTION 4: OTHER TYPES

as a result of irritation. One reason hairdressers have a


been shown to play a modulator role in this induction higher rate of developing ACD is their increased expo­
phase [18]. Ultimately, these interleukins and cytokines
OF DERMATITIS

sure to skin irritants such as water and detergents.


stimulate the migration of the dendritic cell to the local In the past it was questioned whether or not the
lymph node where antigen presentation takes place. ­developing immune system of very young children had
Specifically, antigen presentation involves interaction the capability of producing the delayed‐type hypersensi­
between the hapten–protein complex and a naive, CD4+ tivity response seen in ACD. It has since been shown that
or CD8+ T cell, via the major histocompatibility complex the immature dermal–epidermal barrier of children in
(MHC), type II or I respectively. IL‐12 and IL‐1β, impor­ their first few years of life, with its thinner epidermis,
tant chemokines in the antigen presentation process in the lower lipid content and thus lower quantity of natural
lymph node, play a role in T‐cell maturation and subse­ moisturizing components, and higher pH may actually
quent formation of a memory T‐cell population [19,20]. increase the ability of haptens to penetrate and encounter
The sensitization phase is considered complete when cells of the immune system [29–31]. This confirms that
memory T cells are released into circulation. prevention, when possible, and recognition of the correct
Elicitation occurs when the skin is re‐exposed to the diagnosis and treatment of ACD in even the youngest
same hapten. Immunologically, the hapten–protein paediatric patients, is critical.
­complex gets presented once again to the immune system There are also genetic factors that are known to predis­
via an antigen‐presenting cell. A number of cells are capa­ pose individuals to developing ACD. Loss‐of‐function
ble of acting as antigen presenters, most importantly mutations in the filaggrin gene, commonly associated
Langerhans cells but also keratinocytes, mast cells, with AD and other forms of atopy, have been associated
endothelial cells, and macrophages [18]. Current data with predisposition to ACD [32,33]. One specific German
suggest that both Th1 and Th2 subtypes are involved in study connected deficits in filaggrin to higher incidence
the response to re‐challenge [18,19]. The release of of contact allergy to nickel, particularly in fashion jewel­
­numerous chemical mediators, including IFN‐γ, leads to lery, which explains the stronger association that was
neutrophil recruitment to the site of hapten exposure and noted in women [34]. Two small case‐control studies have
the maturation of T cells (specifically under the influence investigated the relationship between hand eczema and
of IL‐1), resulting in the secondary inflammatory response filaggrin mutations, the first noting that loss‐of‐function
and the clinical signs of contact allergy [19,21]. In most mutations may be implicated in hand eczema when in
individuals repeated exposure to a chemical is required to conjunction with AD, and the second showing significant
incite an immune response and subsequent clinical ACD. association between chronic irritant hand dermatitis and
It is important also to note that the majority of people in filaggrin mutations [35,36]. More evidence to associate
regular contact with known sensitizers may not develop filaggrin deficiency to hand dermatitis is necessary,
allergy. That said, there are certain known strong sensitiz­ although filaggrin may play less of a role in the thick‐
ers, including paraphenylenediamine (PPD) and formal­ skinned palmar area than in areas of thinner skin, such as
dehyde, which have been top allergens for over 70 years, flexural areas. Patients with diffuse exposure to certain
that play a significant clinical role in both adult and chemicals have developed contact dermatitis only in the
­paediatric contact dermatitis. flexural areas, suggesting that the thinner epidermis and
Any alteration of an intact skin barrier can disrupt the lower level of protective function may increase the sus­
protective role the epidermis plays against transepider­ ceptibility of flexural surfaces to developing ACD, and
mal water loss, infectious agents and other chemicals, the potential for misdiagnosis as AD [28].
thus allowing a higher degree of contact between causa­ The development of chronic allergic dermatitis has also
tive haptens and cells of the immune system. Particular been linked to deficiency of cathelicidin LL‐37, an antimi­
populations are at higher risk of developing ACD, for crobial peptide (AMP) that is located in the skin’s ­cornified
example certain occupations such as hairdressers, envelope and functions as part of the skin’s innate
Chapter 23  Allergic Contact Dermatitis 303

immune system, with antiviral, antibacterial and Table 23.2  Regional dermatitis more prevalent in children
­antifungal properties. Cathelicidin LL‐37 also plays an
anti‐inflammatory role, having been found to have a sup­ Eyelids Shampoos, cleansers – cocamidopropyl betaine;
pressive function on dendritic cell activation via ­inhibition metals, e.g. nickel from hand to face – toys, coins, keys

of toll‐like receptor 4. One study showed that the addition


Face Cell phones – nickel and chromate; connubial – PPD,
of exogenous cathelicidin was able to inhibit ACD in vivo fragrance
[37]. Polymorphisms in the IL‐16 gene and the gene region Perioral Musical instruments – nickel; flavoured lip balms;
encoding N‐acetyltransferase, the enzyme responsible for chewing gum – flavourings; pacifiers – rubber
metabolism of the strong sensitizer PPD, have also been Neck Cell phones – nickel; atomizer sign – fragrance
associated with development of ACD [38,39]. Ears Costume jewellery, wireless headsets – nickel
Although the most well‐known exposure to chemicals Diaper Baby wipes – preservatives;
diapers – mercaptobenzothiazole, PTBFR
leading to the development of allergy is through the
Thighs Toilet seat – wood, plastic, cleaners (irritant);
integument, it is also recognized that oral, inhalation and chairs – nickel bolts
intravenous exposure to haptens all have the potential to Extremities Shin guards, splints – urea formaldehyde resin/PTBFR,
lead to dermatitis [18,40]. In the context of ‘oral exposure’, thioureas
chemicals used as food additives (e.g. propylene glycol Scalp Car seats

SECTION 4: OTHER TYPES


and propolis), pesticides and flavouring agents found in Trunk Nickel clothing snaps on infant clothing, breast and
both natural and artificial products have been indicated nipple dermatitis from PTBFR in padded training bras
Feet Shoes – PTBFR, rubber accelerants in children’s shoes,

OF DERMATITIS
as causative agents [41]. Once absorbed from the gastroin­
e.g. Crocs™, potassium dichromate (leather sandals
testinal tract, these haptens can distribute through the and shoes)
blood and initiate a widespread inflammatory reaction
[42]. Of interest, oral exposure has been linked to cutane­ Source: Brod et al. 2015 [46]. Reproduced with permission of Elsevier.
ous tolerance of certain haptens in some individuals, PPD, para‐phenylenediamine; PTBFR, para‐tertiary butylphenol‐
including nickel and chromate, especially if the oral expo­ formaldehyde resin.
sure predates the cutaneous one. For example, it has been
shown among Danish schoolchildren that exposure to
nickel from orthodontic braces through the mucocutane­ allergen and may cause difficulty in establishing r­ elevance
ous surfaces resulted in a 93% reduction in the risk of and connecting the culprit allergen, especially if consider­
sensitization to nickel [43]. Another study comparing
­ able time has passed since the prior exposure [47,48].
populations of Russian and Norwegian women with the Systemic or idiopathic reactions are also possible, pre­
same rates of ear piercing credited the lower rates of senting as nonspecific dermatitis in locations not in direct
nickel sensitization in the Russian population to higher contact with the allergen [47]. Because of the varied pres­
levels of nickel in the Russian cohort’s drinking water entation of ACD, an indication for patch testing children
[44]. In a similar way, it is postulated that the common is uncontrollable or worsening chronic dermatitis that has
practice of consuming chrysanthemum (a plant in the been present for 2 or more months, or failure to improve
Compositae plant family) tea in Singapore led to the following the standard protocols [47,49].
lower rates of ACD to Compositae in that area [40,45]. Dermatitis located in the flexural surfaces, including
the skin crease area in the neck, antecubital and popliteal
Clinical features fossae and the anterior ankle, may not necessarily confer
Everyone knows how eczema looks like, yet no one the diagnosis of AD [25,50]. Due to the specific combina­
knows what eczema is. tion of moisture, warmth and friction in the flexural areas,
Heinrich Adolf Gottron they are a common place to experience skin barrier dis­
ruption and potentially increased percutaneous chemical
The variable clinical features of ACD in children are akin absorption, predisposing to the development of ACD
to the clinical presentations seen in adult populations, [26,28]. Other factors contributing to this disruption may
and include acute eczematous reactions, (localized, be particular types of microorganisms and antimicrobial
­idiopathic and systematized), oedema and/or erythema, peptides that reside preferentially in the flexural areas as
lichenoid, granulomatous and microvesicular or bullous‐ opposed to the environment of the skin in other parts of
type reactions. Phenotype association is related both to the body [25]. It should be noted that patients presenting
the nature of the allergen, regional distribution of the con­ with flexural eczema should have a complete work‐up
tact and duration of the hypersensitivity (with chronicity referring to stringent diagnostic criteria for AD prior to
conferring greater lichenification), in addition to the associating the skin findings to atopy, and should be con­
inherent barrier function of the individual [22]. In its most sidered for patch testing if the diagnosis is uncertain. Of
commonly recognized form, eczematous ACD is known note, a recent study review examined 1142 patch test cases
to occur in the location specifically exposed to the aller­ of children younger than 18 years and found that patients
gen, with a propensity to extend beyond the direct expo­ with AD had different reaction profiles than those with­
sure site. See Table 23.2 for location‐based ACD, adapted out AD, specifically increased frequency of reactions to
from Brod et al. [46]. cocamidopropyl betaine, wool alcohol and lanolin, and
‘Recall reactions’ are possible; these result in the devel­ lower frequency of reactions to methylisothiazolinone,
opment of dermatitis at a site of previous exposure to the cobalt and potassium dichromate [51].
304 Section 4  Other Types of Dermatitis

As with adults, patch testing is the gold standard method


Patch testing in children. A multicentre retrospective study
for confirming a clinically relevant contact allergy in chil­
reviewed the results of paediatric patch testing in North
dren. Guidelines established by the International Contact
America [52]. The study reported patch testing results from
Dermatitis Research Group (ICDRG) have standardized
456 affected children who were tested over an average of
patch test procedures, allergen concentrations and screen­
5.3 years, indicating that an average of only 86 children in
ing panels in the evaluation of adults that have provided
total were being tested and reported on per year [52]. A
an opportunity to draw comparisons across international
limitation of the study was that the majority of the data
databases. The US Food and Drug Administration (FDA)
were being reported by large tertiary care centres, which
has granted a paediatric indication for patch testing with
confer a risk of referral bias and may not be generalizable to
the thin‐layer rapid use epicutaneous test (T.R.U.E. Test®,
the paediatric population as a whole. In 2014, the Loma
Smartpractice, Phoenix, AZ). That said, contact dermatitis
Linda University Pediatric Registry Initiative administered
experts advocate for comprehensive patch testing in chil­
a cross‐sectional electronic demographic survey to provid­
dren with strong clinical indication. The general consen­
ers patch testing children across the USA [53]. In the first
sus is that children 12 years of age and over may be patch
year of enrollment, over 250 providers confirmed that they
tested equivalently to adults. This is supported by global
patch test children across all 50 states, as well as Washington,
paediatric patch test studies that have demonstrated
DC. The data reported a range of 1700–4700 afflicted
SECTION 4: OTHER TYPES

safety and efficacy utilizing the adult patch test allergen


­children patch tested per year, as determined by provider‐
concentrations, with no increased rates of false‐positive
stated ranges for testing (1–10, 11–25, 26–50, 51–99, and over
OF DERMATITIS

reactions [52]. Some authorities extend these parameters


100 cases per year). Within one year of data collection 1142
down to include 6‐ to 12‐year‐olds, while other experts
cases from 34 US states, entered by 84 providers, were ana­
such as the German Patch Test Research Group and
lysed. The study found that 65% of paediatric cases had one
authors of the North American Pediatric Standard
or more positive patch test (PPT), and 48% of cases had one
­advocate a separate limited patch test series for children
or more relevant positive patch test (RPPT). The most com­
6–12 years of age, with some allergens specifically not
mon PPT allergens were nickel (22%), fragrance mix I (11%),
tested without indication (e.g. paraphenylenediamine,
cobalt (9.1%), balsam of Peru (8.4%), neomycin (7.2%), pro­
epoxy, gold); see Table 23.3 for recommendations for pae­
pylene glycol (6.8%), cocamidopropyl betaine (6.4%),
diatric screening series, adapted from Admani and Jacob.
­bacitracin (6.2%), formaldehyde (5.7%) and gold (5.7%) [54].

Table 23.3  Recommendations for paediatric screening series

Allergen Concentration

1 Bacitracin 20% pet.


2 Budesonide 0.1% pet.
3 Carba mix 3% pet.
4 Cobalt chloride 1% pet.
5 Cocamidopropyl betaine 1% aq.
6 Colophonium (colophony) 20% pet.
7 Compositae mix/dandelion extract 6% pet/2.5% pet.
8 Disperse blue 124/106a 1% pet./1% pet.
9 Formaldehyde 1% aq.
10 Fragrance mix I 8% pet.
11 Fragrance mix II (Lyral) 14% pet. (5% pet.)
12 Lanolin alcohol 50% pet.
13 Methylchloroisothiazolinone/methylisothiazolinone (MCI/MI)b (3 : 1) 0.01% aq.
14 Myroxylon pereirae (balsam of Peru) 25% pet.
15 Neomycin sulfate 20% pet.
16 Nickel sulphate 2.5% pet.
17 Potassium dichromate 0.25% pet.
18 Propylene glycol 30% aq.
19 Quaternium 15 2% pet.
20 Tixocortol‐21‐pivalate 1% pet.

Source: Adapted from Admani and Jacob [47]. Reproduced with permission of Springer Nature.
pet., petrolatum; aq, aqueous.
a
 Mix two components in equal amounts.
b
 MI can be tested alone as well.
Additional allergens to consider screening (if indicated by associated history): Amerchol L‐10125 (exposure to emollients, lotions), black rubber mix
(tyres, playground, toys), clobetasol 17‐propionate (topical steroid, chronic dermatitis patient), mixed dialkyl thioureas (rubber‐containing sportswear, bras,
footwear), mercaptobenzothiazole (rubber cements, gardening hose, rubber in sports equipment, toys), methylisothiazolinone (preservative), para‐
phenylenediamine (hair dye, black henna tattoo), para‐tertiary butylphenol‐formaldehyde resin (rubber‐containing bras, footwear, sports gear), sorbitan
sesquioleate (emulsifier, emollients, lotions), thiuram (rubber‐containing footwear, athletic gear, bras, toys).
Chapter 23  Allergic Contact Dermatitis 305

[47]. It has been suggested that for this younger age group coatings attached to the back as a single polyester film
(6–12) allergens could be applied initially for only with paper tape. With the limited number of allergens
24 hours and readings be performed at 48 and 72 hours that can be tested with the commercially available series,
[55], though the standard 48‐hour initial application cycle there is a higher likelihood that the diagnosis of ACD
has been shown to be both safe and efficacious in this would be missed than when custom‐tailored comprehen­
same age group [12,15]. There are also recommendations sive testing is utilized (especially in children). A recent
by the German Contact Dermatitis group that children systematic review of the latest paediatric patch‐test results
under 6 only be patch tested when there is a high degree published in US‐based medical journals identified the
of clinical ­suspicion for allergen exposure [55]. most common allergens in the paediatric population [29].
Several of these top allergens, including propylene glycol
Patch procedural considerations and cocamidopropyl betaine (CAPB), do not appear on
Several different types of chambers can be used to apply the T.R.U.E. test®, neither do other allergens coming
allergens to be tested to the skin, from small circular discs under increasing scrutiny in the paediatric population,
made of aluminium (commonly Finn® chambers) to plas­ such as benzoates [56]. Notably, the T.R.U.E. Test® was
tic chambers. Aluminium chambers should not be used to shown to be safe and efficacious for use in children over
test patients with known or suspected metal allergy, or 6 years of age in the PREA‐1 study [12], with the original

SECTION 4: OTHER TYPES


those being tested for suspected allergy to vaccine compo­ set of 28 allergens in 2011. In 2017, the FDA approved the
nents. Finn® chambers (SmartPractice, Canada) are also T.R.U.E test® for individuals 6 years of age and older [57].

OF DERMATITIS
available with a polypropylene coating, which should be Special considerations and limitations for patch testing
used in cases of suspected aluminium allergy or when children include their small back size, limiting the number
testing mercury solutions. Plastic options include aller­ of allergens that can be tested, particularly in very small
gEAZE® and allergEAZEclear® (SmartPractice, Canada) children. The limited space for testing further underlines
made of polyethylene terephthalate, additive‐free poly­ the need for detailed history taking of potential allergen
ethylene chambers from IQ UltraTM (Chemotechnique exposures, utilizing this information to guide choosing
Diagnostics, Vellinge, Sweden), and polypropylene cham­ allergens, and testing patients to their own personal care
bers from van der Bend (van der Bend, Brielle, the products, which may carry a particularly high yield. Other
Netherlands). Curatest® and Curatest® F (Lohmann and important pieces of the patient’s history to gather include
Raucher, Rengsdorf, Germany) provide disc frames with personal history of eczema or atopy, family history of
a polyester film (Table 23.4). Loadable chambers allow for atopy, known allergies, hobbies, leisure and after‐school
customization of the allergen series to be tested, and or work activities, topical and systemic medications used,
smaller chambers can be used when space is limited, and use of or exposure to cosmetics and skin care products
though this leads to volume variances, and therefore [58]. A thorough history should also include information
­differences in the amount of allergen applied. The com­ on all caregivers and those coming into ­contact with the
mercially available limited screening panel (T.R.U.E. patient. The age of the child should also be taken into con­
[thin‐layer rapid use epicutaneous] Test®, Smartpractice, sideration when suggesting more likely allergens.
Phoenix, AZ) may be utilized in instances where compre­ There are certain allergens that have been recom­
hensive testing is not available. mended to be tested in lower concentration in very young
The current T.R.U.E. test® contains 35 allergens/mixes children (less than 6 years old), including nickel, formal­
and a negative control in the form of uniform dried gel dehyde, and rubber additives, such as carbamates [49,59],

Table 23.4  Available patch test chambers

Type of chamber Material composing Contents Notes


chamber

Aluminium Finn® chambers Aluminium Flexible sizing of aluminium Epitest Ltd, Oy, Tuusula, Finlandare
Polypropylene‐coated Aluminium chambers (8, 12 and 18 mm) Use polypropylene‐coated when
Finn® chamber without pre‐fixed filter paper suspected aluminium allergy or
when testing mercury solutions
Plastic allergEAZE® Polyethylene terephthalate 40 μL with pre‐fixed filter paper SmartPractice, Canada
allergEAZEclear® Polyethylene terephthalate and raised edges for occlusion
IQ Ultra™ Additive‐free polyethylene 65 μL with pre‐fixed filter paper Chemotechnique Diagnostics, Vellinge,
Sweden
Van der Bend Polypropylene 40 μL with pre‐fixed filter paper van der Bend, Brielle, the Netherlands
Curatest® Polyester film disc frame Pre‐fixed filter paper Lohmann and Raucher, Rengsdorf,
Curatest® F Polyester film disc frame Germany
Commercially T.R.U.E. Test® Single polyester film Pre‐measured hydrophilic gels Limited to 35 allergens/mixes and a
available limited (dried to film) on negative control; higher likelihood
screening panel impermeable backing for that the diagnosis of allergic contact
maximum contact and dermatitis would be missed
penetration
306 Section 4  Other Types of Dermatitis

which presents another limitation of the commercially Later visits can and should also be planned for, to confirm
available screening tool. When patch tests are not that the dermatitis has cleared upon avoidance of the
­customizable, testing with nonspecific preloaded aller­ indicated allergen.
gens may lead to children being tested for chemicals that Clinical relevance is then designated as definite, prob­
they have not previously been routinely exposed to and able, possible, past or unlikely by the practitioner at the
unnecessary exposures, for example epoxy and paraphe­ follow‐up visit, once avoidance has been initiated to the
nylenediamine. Furthermore, utilizing the preconstructed allergen associated with the positive patch test reaction
panel also means potential exposure to chemicals that are (PPT). Definite relevance is determined only if the derma­
no longer routinely tested (unless there is specific titis is improved with avoidance of the inciting allergen,
­indication) at patch test centres nationwide due to their and then re‐occurs after re‐exposure to the responsible
clinical ­ irrelevance, such as the mercuric derivative allergen.
­preservative ­thiomersal (thimerosal) and gold. If the diagnosis is uncertain due to highly suspicious
As with adults, activity should be limited during patch allergens yielding negative results on patch testing, or the
testing to prevent movement of the patches. Adhesive relevance of a positive test is in question, further tests can
tape can be used to more firmly secure the patch test to be confirmed to guarantee a more certain diagnosis of
the child’s skin. Whenever possible, patch tests should be allergy, such as repeated open application tests (ROAT).
SECTION 4: OTHER TYPES

applied to unaffected skin on the upper back or inner In a ROAT, the allergic substance is applied to the flexor
arms, noting that in children with AD even seemingly aspect of the forearm twice daily for 10–14 days, unless
OF DERMATITIS

unaffected skin can be more reactive or prone to irritant dermatitis appears earlier. A positive test is the develop­
reactions [60]. In addition, because approximately 90% of ment of clinical dermatitis [58].
patients with AD are colonized with Staphylococcus aureus
[61], the environment created by patch testing has the Allergens of  particular importance in  children. It has
potential to result in bacterial overgrowth and secondary been demonstrated that although there are similar rates of
infection because of the inherent occlusive nature of the contact sensitization, there are some variances in allergen
patch testing procedure. This can further complicate the frequency between children and adults [15]. For example,
reading of patch tests. For this reason, particularly in children are less likely to suffer from ACD due to occupa­
patients with AD, it is important to note carefully areas of tional exposures (e.g. to adhesives, epoxy resin and
baseline dermatitis prior to patch testing. acrylates), but are more likely to become sensitized to
It has been recommended that patients with a history of ­personal care products and cosmetics [65]. A review of
bacterial superinfection be evaluated for, and if necessary many of the top paediatric allergens by location can be
treated for, nasal carriage of Staph. aureus, or perform found in Table 23.2.
bleach baths for 10 minutes, three times a day, for 1 week
prior to patch testing, stopping 4 days prior to patch Nickel
testing. Furthermore, it is paramount in this population to Nickel is the number one cutaneous allergen detected in
perform delayed reads (day 5–7) in order to record irritant US children through patch testing [15,66,67]. It is esti­
reactions as false positives [62]. Topical steroids should mated that nickel allergy affects 250 000 children each year,
not be used in the area of the patch test application for and the rates of nickel sensitization in the US paediatric
1  week prior to testing, and oral corticosteroids should population have been on the rise [16,47,68]. Sensitization
also be avoided in the week surrounding testing, if to nickel is not uncommon in asymptomatic children, as
possible. Topical corticosteroids can, however, be used in the first test proving the safety of patch testing children in
other areas of dermatitis on the body during the patch test 1986 found that 7.6% of 314 healthy, asymptomatic chil­
process in order to suppress further flares that could dren were already sensitized to nickel [2]. Sensitization is
occur during the testing process. Administration of oral caused by exposure to metal objects that release a signifi­
antihistamines to control pruritus during the testing cant amount of nickel, for example as a result of ear pierc­
process is also appropriate. In extreme cases, it may be ing, which is believed to be the reason why sensitization to
necessary to maintain a minimal level of systemic nickel is more common in girls [43,69]. Ear piercing is
immunosuppressant therapy during patch testing [63,64]. highly associated with sensitization due to the disruption
Allergens are typically applied on day 1 and then kept of the skin barrier through piercing of the skin. Although
on for 24–48 hours. Between application and the first read no study has identified a specific point of highest likeli­
the patients are required to keep their back dry and to not hood of developing nickel sensitization by comparing
rub the area to disrupt the position of the allergens. At the early or late ear‐piercing practices, it stands to reason that
first read (day 3), the patches are removed and sites are the earlier the piercing, the greater the time for cumulative
evaluated. On day 5–7, the second read is performed. The direct skin‐piercing exposure. One study demonstrated
second reading is critical because it allows for the differ­ that women who had ear piercing performed before the
entiation of irritant reactions, which are delineated by a age of 20 were more likely to become sensitized to nickel
more intense reaction on the first read than on the second. compared to those who had ear piercing after the age of 20
Usually, a true allergic reaction tends to increase in inten­ [70]. This may be related to a greater tendency in youths to
sity between the first and second reads, whereas irritant wear cheaper metal jewellery [71]. Of interest, a study
reactions dissipate. Certain reactions require a third read found that adolescent girls pierced after wearing dental
(day 7 or later) because of a particularly delayed response. braces had a decreased risk of nickel sensitization
Chapter 23  Allergic Contact Dermatitis 307

c­ ompared to girls who had ears pierced without ­previously rates of sensitization in Danish children, from 24.8% in
wearing dental braces. It is believed that the mucosal 1992 to 9.2% in 1998 [79,80]. The European Union (EU)
exposure to nickel‐releasing metals led to development of emulated this in 1994 by passing similar legislation, called
immunological tolerance prior to the opportunity for cuta­ the EU Nickel Directive. Not only have sensitization rates
neous sensitization [43]. decreased in countries belonging to the EU, but Denmark
Nickel is inexpensive and is used in many industries, reported an estimated savings of 2 billion US dollars over
making avoidance difficult for those suffering from nickel the 20‐year period since the enactment of related legisla­
allergy. More important than the amount of nickel in an tion [17]. The cost‐savings potential seen in Denmark
item is the rate at which nickel is released from it. For when projected to the much larger US population is
example, although stainless steel is a metal alloy that ­striking [16]. Nevertheless, such regulation is not cur­
often contains nickel, contact with stainless steel rarely rently present in the USA, however the alarming rise in
incites dermatitis in allergic patients because the rate of sensitization rates has gained the attention of national
nickel release is low. Certain items made of alloys includ­ groups such as the American Academy of Dermatology.
ing nickel may initially release a low amount of nickel,
but over time, as the outer layer receives more wear, more Cobalt and gold
nickel may be released. One alloy known to do this is Cobalt results in many positive patch test reactions in

SECTION 4: OTHER TYPES


nitinol, a nickel–titanium alloy with shape memory used children, often second only to nickel, yet its clinical rele­
for eyeglass frames, underwired bras, medical devices vance can be difficult to determine. To explain this, cobalt

OF DERMATITIS
and novelty items [16,72]. Nickel often incites dermatitis sensitization is a well‐known ‘co‐reactor’ to nickel, due to
in classic locations, including the ear lobes from jewellery simultaneous exposures to both metals in metal alloys.
(most often costume jewellery), at the umbilicus from but­ Gold is another metal that should probably not be
tons or belt buckles containing nickel, and on the waist­ tested on children unless their history warrants it. The
line from nickel‐containing studs on clothing. Idiopathic North American Contact Dermatitis Group data have
reactions have also been reported from nickel, including a determined that there is very little clinical relevance to
paediatric case resulting from electronic tablet use [73,74]. sensitization to gold in children under 12, and for this rea­
Other sources of nickel exposure in children include eye­ son it has been removed from their standard patch test
glass frames, zips, clothing snaps, orthodontic braces, screening tray [52]. Gold currently remains an allergen on
school chairs, mobile phones and toys [13]. the T.R.U.E. test®.
Systemic contact dermatitis to nickel, or dermatitis
­following systemic exposure to a previously sensitized Neomycin and other topical antibiotics (e.g. bacitracin)
allergen (e.g. by inhalation, ingestion or implantation), Neomycin, a topical aminoglycoside antibiotic, has a long
has been reported [75]. A meta‐analysis of 17 studies on history of being one of the most prevalent contact
oral nickel exposure noted that 1% of nickel‐sensitized ­allergens in adults in the USA – over three decades [81]. It
patients suffered adverse systemic symptoms when is believed that a week or more of consistent use of neo­
exposed to significant nickel in their diet [76]. Certain mycin has greater potential to cause sensitization than
foods are known to be consistently higher in nickel, occasional application to small cuts or wounds [81,82].
including wholegrain flour, oats, soybeans, shellfish, Bacitracin is commonly seen in combination topical
nuts and legumes [77], and a dose–response relationship ­antibiotic preparations, often with neomycin and/or pol­
has been noted between nickel‐rich foods and dermatitis ymyxin B sulfate. Due to their popularity, the use of these
flares [78]. The real‐life effectiveness of prescribing a over‐the‐counter combination medicaments can result in
low‐nickel diet for nickel‐sensitive patients has been co‐sensitization or simultaneous sensitization to both of
controversial, particularly because compliance with a these allergens, leading to polysensitization of patients
low‐nickel diet can be difficult, as avoidance and elimi­ [9]. Of note, more than 25 cases of anaphylaxis have
nation of nickel from the diet can seem impossible with resulted from application of topical bacitracin to skin
nickel ­levels in foods varying with the level of nickel in abrasions, indicating the potential for type I hypersensi­
soil and water, potentially even from season to season. tivity reaction to the compound as well as delayed type
Despite this, a trial of a low‐nickel diet is a potential [83]. Similar to what is seen with corticosteroids, atopic
treatment option to consider in patients highly sensi­ patients may suffer from higher rates of sensitization to
tized to nickel, as evidence suggests the potential for topical antibiotics such as neomycin because of their
clinical improvement in symptoms in some cases [75]. A increased rate of use to treat superficial bacterial infec­
points‐based system utilizing mean concentration of tions imposed on atopic skin [84].
nickel in foods (as reported by the FDA), limiting nickel
consumption for the most nickel‐sensitized patients to Fragrance (including fragrance mixes I and II, cinnamic
less than 150 μg per day (aka 15 points) [77] can be uti­ aldehyde, cinnamic alcohol, propolis, Compositae/
lized to improve compliance. dandelion colophony, balsam of Peru)
In response to a rising trend of nickel sensitization in Many allergens are clumped into the ‘fragrance’ category,
Denmark, legislation was passed in 1992 that limited the including balsam of Peru, fragrance mixes I and II,
amount of nickel that can be released from products benzoates, cinnamic aldehyde and cinnamic alcohol,
­
coming into prolonged contact with skin (limited to
­ propolis (beeswax), Compositae/dandelion and colophony.
0.5 μg/cm2 per week), resulting in significantly lower Fragrance mix includes geraniol, cinnamaldehyde,
308 Section 4  Other Types of Dermatitis

hydroxycitronellal, cinnamyl alcohol, eugenol, ­isoeugenol, sesquiolate is used in screening trays to test for fragrance‐
α‐amylcinnamaldehyde and oak moss. Although cases of related allergy, as it is an emulsifier and is found in many
systemic dermatitis have been connected to fragrance fragrance cosmetics.
allergy, typical sites of dermatitis resulting from fragrance
allergy are the face, neck and axilla, where the greatest Potassium dichromate
contact with the chemicals occurs [85]. Apart from the Tanned leather products such as shoes and belts are the
obvious sources such as ­perfumes, fragrances are used in most common sources of positive patch tests to the
many products such as cosmetics, sunscreens, cleaning ­allergen potassium dichromate. In fact, potassium dichro­
products and other hygiene products. In children, fra­ mate is a known allergen responsible for inducing foot
grance is commonly the cause of ‘connubial’ dermatitis, or dermatitis in children [13,96,97]. Several studies have
facial eczema in a child being carried or cheek‐to‐cheek demonstrated a higher rate of sensitization to potassium
with an adult wearing cosmetics or other fragrances. dichromate in children with AD than those without,
Fragrance, consistently one of the top allergens in both which has been explained by atopic children having a
adult and paediatric populations, is used even in products higher incidence of juvenile plantar dermatosis, suggest­
marked ‘unscented’ if the fragrance is used to mask ing that the impaired epidermal barrier may enable pen­
another fragrance. For this reason, patients with fragrance etration of haptens across the skin of the foot [98]. An
SECTION 4: OTHER TYPES

allergy are encouraged to seek products that are marked alternative for affected children is footwear made from
specifically ‘fragrance‐free’. Fragrances have also been vegetable‐tanned leather. Vitamins, paints and anti‐rust
OF DERMATITIS

used in products marked ‘hypoallergenic’ if their purpose products, and matches are other important sources of
in the products is for preservative properties instead of exposure to potassium dichromate [99].
fragrance properties  [86,87]. Clinical significance of fra­
grance allergy is extremely high, with over 90% of positive Lanolin and other wool alcohols (including Amerchol
patch tests reported in adults with definite, probable or L‐101™)
possible relevance [88]. Lanolin is a product of alcohol extraction of sebum from
Balsam of Peru, an extract from a tree known as wool, and consists of long‐chain waxy esters. Utilized for
Myroxylon pereirae, is a mixture of many fragrant chemi­ its emulsifying and hydrating properties, lanolin is com­
cals and is related to fragrances such as benzoates, monly incorporated into cosmetic and hygiene products
­cinnamic aldehyde and alcohol. It is tested in screening (including shaving cream, soap and shampoos) and heal­
trays as a general marker of fragrance allergy, along with ing salves. Rates of contact sensitization to lanolin in the
the fragrance mixes, and is used in many products, paediatric population have been reported as recently
including toothpaste, mouthwash, topical medications increasing [100,101]. However, patch testing patients to
and healing salves [89,90]. Related chemicals are used as lanolin has been notoriously difficult. In part this is due to
artificial flavouring in soft drinks and other food items, the ‘lanolin paradox’: essentially that contact sensitization
including flavours such as vanilla, curry, cloves and cin­ to the allergen is less likely to be apparent on intact skin
namon. Because of the known potential for oral exposure than expression on disrupted skin. Variability in lanolin
to related chemicals, there is some evidence that dietary composition can result in low yield from testing patients
intervention and avoidance of related flavours may if just one lanolin‐containing agent or single concentra­
benefit patients with particularly difficult‐to‐treat fra­
­ tion is tested [102]. Amerchol L‐101™ (50%) has been rec­
grance allergy [91,92]. By testing both to balsam of Peru ommended as the testing substrate and is now currently
and fragrance mix, it is estimated that more than 80% of included in the American Contact Dermatitis Society core
fragrance allergy can be detected [93,94]. allergen series because in at least one study it was shown
Compositae, also known as Asteraceae, is a plant fam­ to be a better determinant of allergy to wool alcohol than
ily representing 10% of the world’s flowering plants, the standard lanolin 30% [103]. Also, due to potential var­
including chamomile, dandelion and ragweed. Both iability of lanolin used in products, patients should be
localized and diffuse‐airborne patterns of ACD to these tested with their own lanolin‐containing personal items if
compounds have been reported, at times mimicking pho­ allergy is suspected.
todermatoses. Use and exposure to these allergens in teas,
herbal supplements and personal care products leads to Carba mix/carbamates (and other rubber materials:
sensitization. The sesquiterpene lactones are the chemi­ thiuram mix, thioureas, mercaptobenzothiazole,
cals responsible for most of the Compositae plant family’s mercapto mix)
allergenicity [48,95]. Colophonium, balsam of Peru and Carbamates, thiuram, mercaptobenzothiazole and mer­
propolis have all been known to cross‐react in individuals capto mix are included in all standard patch test series.
allergic to this family of plants [92]. Children may become These chemicals are rubber accelerators, used in rubber
exposed to colophony, a member of the Pinaceae family, production to catalyse transformation from a liquid to
in preparations used for treating verrucae, soaps, glues, solid state. The mercapto compounds are primarily found
varnishes, violinist’s rosin and gripping powder used by in heavier rubber items, such as the soles of shoes, while
gymnasts. Propolis (beeswax) is advertised and widely thiurams and carbamates are a significant component of
used for its immune‐stimulating, antioxidant and wound‐ gloves. Exposure and resulting allergy in children can be
healing properties, resulting in strong association with traced to gloves and shoes, elastic waistbands in pants and
self‐medication with ‘natural’ substances [13]. Sorbitan elastic in socks, sports gear, bras, swimwear, pacifiers and
Chapter 23  Allergic Contact Dermatitis 309

other toys [99]. Napkin dermatitis has been ­historically Tixocortol‐21‐pivalate and budesonide are the corticos­
associated with mercaptobenzothiazole allergy, as it is teroids on the recommended paediatric screening tray.
used in the elastic waistband [104]. In addition, the Identification of this allergy is particularly important for
­environment in which a person is exposed to mercapto paediatric atopic patients or others with chronic inflam­
compounds is believed to affect sensitization. For exam­ matory skin diseases who require frequent application of
ple, the warm, moist environment in shoes and gloves topical medicaments as part of their treatment regimen.
may increase the rate of release of rubber additives from There is an increased risk that these patients develop sen­
these compounds, leading to sensitization [105]. sitization to the medicament ingredients because of their
disrupted skin barrier. A newer classification of steroids
Cocamidopropyl betaine (CAPB) and related allergens was suggested by Baeck et al. in 2011, which differenti­
(3‐dimethylaminopropylamine, amidoamine) ated steroids based on their allergic potential: ‘Group 1’
Cocamidopropyl betaine is used as a surfactant, foaming representing non‐methylated ­molecules (traditional clas­
and thickening agent in many soaps, shampoos, body sification Group A, D2 and budesonide) which carry the
washes and toothpastes. It was initially seen in Johnson & most allergic potential, ‘Group 2’ being the halogenated
Johnson’s ‘No More Tears’ shampoo in the 1950s, as it is C16/17 cis‐ketal/diol structure and Group B steroids,
less irritating to eyes than alternative surfactants due to and ‘Group 3’ (Group C and D1) carrying the least aller­

SECTION 4: OTHER TYPES


its zwitterion chemical structure, with both positively and gic potential [113].
negatively charged elements [106,107]. While increas­ As for ingredients, propylene glycol has been identi­

OF DERMATITIS
ingly recognized as causing ACD, particularly in paediat­ fied as the top allergen found in many topical steroids
ric populations, CAPB is not an allergen included in the [114,115]. The vehicle used for patch testing patients to
commercially available patch test. Positive patch tests to corticosteroids may also affect the sensitivity of the test,
3‐dimethylaminopropylamine (DMAPA) and amidoam­ relating to the penetration of the allergen into the skin.
ine (AA), chemical byproducts/impurities generated in Patients not responding as expected to treatment for
the manufacturing of CAPB, have been reported, some chronic dermatitis in particular should be patch tested
investigators even suggesting that these chemicals may for allergy to both steroids and individual components of
be the true sensitizers leading to CAPB allergy [107]. steroids, in addition to the patient’s current personal top­
ical formulation. These patients may also benefit from
Formaldehyde and formaldehyde releasers (p‐tert‐ use of the ‘Group 3’ steroids with the least allergenic
butyl‐phenol formaldehyde resin, diazolidinyl urea, potential. Examples of steroids in this group include
imidazolidinyl urea, quaternium‐15, DMDM alclometasone dipropionate, betamethasone valerate,
hydantoin, bronopol, etc.) mometasone furoate, desoximetasone and clobetasol
Formaldehyde and related chemicals have been tested as propionate (listed in increasing order of potency). If a
part of standard screening series since the first screening reaction to a topical steroid is observed, it is important to
series was recommended in 1939 [108]. These products determine the range of hypersensitivity by patch testing
are ubiquitous, as they are used in personal hygiene prod­ with other steroids as well.
ucts and cosmetics as antimicrobial preservatives to
maintain shelf‐life and to maintain wrinkle‐free fabrics, to Para‐phenylenediamine
name a few. The FDA notes that formaldehyde‐releasing Para‐phenylenediamine (PPD) is a common hair dye
preservatives are found in an estimated 20% of personal chemical that is related to printer ink and photography
hygiene products [109]. Cases of persistent dermatitis products and is used to enhance and darken the colour of
have been traced to systemic exposure to formaldehyde henna tattoos (black henna). The permitted concentration
in cigarette smoke inhalation and products in foods that of PPD in permanent hair dyes is 6%, but the concentra­
metabolize into formaldehyde (e.g. aspartame, smoked tion in black henna tattoos has been found to be up to
meats and maple syrup), suggesting that there may be 15.7% [116]. Reactions to PPD in temporary and black
benefit from dietary avoidance in patients with difficult‐ henna tattoos have caused permanent scarring and hypo­
to‐treat formaldehyde allergy [110,111]. P‐tert‐butyl‐phe­ pigmentation as a result of severe hypersensitivity reac­
nol formaldehyde resin (PTBFR), a compound chemically tions. Known as one of the five allergens recognized as
related to formaldehyde, is found in adhesives used to ‘strong sensitizers’, PPD has been responsible for leading
make shoes, watch straps or sports gear such as shin to extreme, even life‐threatening reactions in children and
guards. One study with 102 patients found a surprising adolescents [117–119]. Testing is warranted only if a rele­
number of children with positive patch tests to PTBFR vant history is elicited, due to the potential harm from
associated with sneakers and sports equipment, where possibly sensitizing children to this allergen or eliciting a
the moist environment due to sweat combined with hypersensitivity reaction. Almost 25% of those sensitized
mechanical irritation probably increases the chance of to PPD may also demonstrate allergy to related semi‐­
sensitization [12,112]. permanent dyes in dark synthetic clothing [120,121].

Corticosteroids Thiomersal (thimerosal)


ACD from topical corticosteroids is not rare, yet can be Sensitization commonly occurs during routine vaccina­
complex to detect because allergy can be to both the active tion, where thiomersal serves as a preservative [122].
steroid components and the ingredients of the vehicle. Other less common sources of exposure and sensitization
310 Section 4  Other Types of Dermatitis

include contact lens solution and other ophthalmic or otic of sensitization to propylene glycol [114,115]. Historically,
preparations, which have resulted in periorbital d­ ermatitis propylene glycol has created difficulty in identifying
[123]. Although positive patch test reactions to thiomersal allergy through patch testing, as it has been known to
are common, relevance is often not established and may cause irritant reactions that may be difficult to distinguish
only represent prior sensitization from past exposure. from mild allergic reactions [137].
Routine patch testing for this chemical is more recently
considered unnecessary, and one study suggested that Benzalkonium chloride
rates of sensitization decrease with age [124]. Thiomersal Benzalkonium chloride is another preservative utilized in
has been removed from all US vaccines routinely recom­ topical medications, cleaning products and personal care
mended for children, with the exception of inactivated products. A significant amount of sensitization and expo­
influenza vaccine, tetanus toxoid vaccine and diphtheria, sure is believed to occur from its use in disinfectants and
tetanus and activated pertussis, but is still used in vac­ sanitizers as an alternative to alcohol‐based products
cines given in European countries [125]. [138]. Known recently as a contact allergen from its use in
household cleaning wipes, it has been known for longer
Methylchloroisothiazolinone/methylisothiazolinone as a skin irritant, thus necessitating the exclusion of irri­
(MCI/MI) tant reactions when testing for contact allergy to this
SECTION 4: OTHER TYPES

The combination preservative methylchloroisothiazol­ chemical.


inone/methylisothiazolinone, or MCI/MI (also known
OF DERMATITIS

as Kathon CG and Euxyl K100), is found in many p ­ ersonal Disperse dyes


care products specifically for use in children, including Disperse dyes are used in polyester‐blended textiles, as
wipes, body washes, bubble baths and moisturizers. It seen in children’s gym clothes and school uniforms [139].
also has the potential of causing airborne (systematized) Allergy to disperse dyes has also been attributed to dyes
ACD from freshly painted sources (since these agents are used in napkin materials [140]. Although disperse blue is
frequently used as preservative agents in common house the most common colour of dye allergen, the colour of
paints), body mist [126], air fresheners [127] and cleaning fabric or material causing allergy may not be blue, as it
supplies [128–130]. In 2013, Aerts et al. reported the first should be considered that multiple colours are often com­
case of a 4‐year‐old girl who was sensitized specifically to bined to create colour blends. While disperse dyes are
MI in her baby wipes, but subsequently had airborne included in many paediatric screening series, it is also an
(partly systemic) contact dermatitis (presumably by option to apply a suspect garment directly to the skin to
­inhalation of vapour from house paint), which notably test for development of hypersensitivity reaction [99].
mimicked atopic dermatitis [131].
MCI/MI allergy has been increasingly reported [132], Rhus tree (Toxicodendron)
leading to such great concern in the EU that MCI/MI is There are a number of Toxicodendron species, and the most
now prohibited from use in ‘leave‐on’ products [114,115]. common ones to cause dermatological problems are poi­
The particular concern for this chemical being used in son ivy (Toxicodendron radicans) and poison oak
paediatric personal care products is due to its significant (Toxicodendron diversilobum), as they contain a potent aller­
sensitization rates [133]. Most standard screening panels gen present in the oleoresin. Rhus dermatitis occurs
test for allergy to MCI/MI mix (3 : 1) 0.01% aqueous solu­ ­particularly in the USA. The acute dermatitis develops
tion, yet it is estimated that testing with the mix alone after direct contact with the plant but may occur from
may miss approximately 40% of allergy [134]. Instead, MI indirect exposure such as from burning plant material.
(0.2% aqueous solution) can be tested alone in addition to The eruption is characteristically a papular/vesicobul­
the mix. MI alone is also not currently included in the lous event with a streaky distribution. Cross‐reactions
T.R.U.E. test®, which makes testing for sensitization to may occur with the oleoresins of other plants including
this common allergen less accessible. mango, cashew and the Japanese lacquer tree. In other
circumstances, phototoxic reactions to plant materials
­
Propylene glycol such as rue may result in a similar rash.
Propylene glycol has been implicated as the causative
agent in several reported cases of generalized dermatitis Primary interventions  –  prevention, avoidance, educa-
[7,66,135]. Ingestion of propylene glycol, used as an addi­ tion, regulation. Once patch testing is completed and an
tive in food as a solvent for flavouring agents and dyes, allergen has been identified, relevance can be assigned to
has been shown to carry the potential to flare propylene guide avoidance strategy. Patients can be directed in their
glycol dermatitis, suggesting that patients with dermati­ avoidance of pertinent allergens and seeking alternative
tis related to cutaneous allergy to propylene glycol may products through provider‐registered electronic manage­
also benefit from an avoidance diet [136]. The most likely ment databases such as the Contact Allergen Management
sources of exposure of paediatric patients to propylene database (CAMP) and Contact Allergen Replacement
glycol include personal care products such as leave‐on Database (CARD), which is now known as ‘Skin Safe’. The
moisturizers, deodorants and cosmetics, while it is also Dermatitis Academy offers free open‐access, online, patient‐
used in varnishes, lacquers, lubricants and cutting fluids, centred resources including a ‘Simple and Free’ guideline
serving as an excellent solvent, vehicle, emulsifier or highlighting products devoid of the top 10 paediatric aller­
humectant. Topical steroids are another common source gens. It is important that sufficient post‐patch test patient
Chapter 23  Allergic Contact Dermatitis 311

education be performed to assure understanding of the At the same time, not all eczema is associated with a
need for avoidance of the identified allergens, such as where filaggrin mutation, and not all with the mutation develop
the allergen is found, what are the most likely exposures, AD [146]. It is possible that filaggrin and other epidermal
and alternative practices, strategies or products for its defects may contribute to enhanced penetration of
avoidance. Patients should be encouraged to bring to the ­allergens through the damaged epidermis, leading to a
visit their routinely used personal products for review with polarized immune response that is more Th2 heavy [147].
the patch test education coordinator to eliminate source The Th2‐dominant immune response common to atopics
products containing the contact allergen. may also interact differently with epidermal proteins, and
For easy identification of items releasing significant while not completely understood, this immune dysregu­
amount of nickel, available to both patients and provid­ lation may affect both the integrity of the epidermal bar­
ers, the dimethylglyoxime/ammonia (DMG‐A) nickel rier and the response of the innate immune system [148].
release spot test allows for detection of the level of nickel The Th2‐polarized response (high in IL‐4 and IL‐13), com­
release sufficient to cause sensitization. Upon rubbing a mon in atopics, along with the reduction of antimicrobial
white cotton swab coated with DMG on a metal item, for­ peptides (e.g. cathelicidin LL‐37 and HBD‐3) and dimin­
mation of a pink precipitate indicates a concentration of ished recruitment of cells such as neutrophils, also prob­
0.44 μg/cm2 nickel release (0.05 μg/cm2 has been shown to ably leads to the high incidence of colonization of atopic

SECTION 4: OTHER TYPES


elicit nickel sensitization) [141,142]. A colour scale can be patients by Staph. aureus [148]. Downstream changes in
utilized to determine the level of nickel being released inflammatory and regulatory innate immune system

OF DERMATITIS
from an item [143]. Patients should re‐test their items over responses, and a decreased response of the innate immune
time as the top layer of items may contain less nickel than system, could explain the prevalence of growth of the
the alloys underneath that will be exposed for further bacteria in these patients [145,148].
wear and time. Investigation into the realm of possibilities for treating
Strict avoidance regimens may need to be adhered to for ACD with biologic agents, comparable to the likes of what
2–3 months in more extensive cases of dermatitis [49], we have for psoriasis, has lately been under way and
requiring diligence and patience on the part of the patient, sheds light on the immune mechanisms at play in the
patient’s family and providers. For the many patients development of ACD. Phosphodiesterase inhibitors
without access to patch testing, the concept of ‘virtual (­particularly PDE‐4), in the form of topical agents, have
patch testing’ has arisen, utilizing trends from national been known to inhibit TNF‐α, an important inflammatory
patch test data sets to estimate which allergens are proba­ cytokine in the development of ACD, and have been
bly causing the majority of ACD, which patients can pre‐ investigated for the treatment of ACD [149,150]. They
emptively avoid and observe if their symptoms abate ­represent an additional nonsteroidal option to topical tac­
[144]. A review of the largest five studies published by US‐ rolimus and pimecrolimus. Other anti‐TNF‐α medica­
based medical journals reporting results of patch testing tions, such as etanercept and infliximab, have been
children after the year 2000 indicated a high frequency of suggested as holding potential roles in the treatment of
known sensitizers appearing in paediatric personal care contact dermatitis, although this is not as well documented
products. By utilizing products devoid of these a­ llergens, as their role in treating other inflammatory ­diseases such
pre‐emptive avoidance strategy (PEAS), a significant as psoriasis. Additionally, there is evidence that topical
number of children could potentially improve their calcineurin inhibitors could aid in the repair of barrier
­dermatitis. Likewise, a similar PEAS has been suggested defects common to atopic dermatitis, and that antibiotics
for children with chronic dermatitis, recommending such as gentamicin may be able to influence production of
avoidance of highly prevalent high‐frequency sensitizing filaggrin chains by impacting the interaction between resi­
allergens found in paediatric‐marked personal care prod­ dent skin flora and the innate immune system [151].
ucts. The authors suggested that it may be possible to Further evaluation of the role of racial differences in the
improve approximately one third of paediatric ACD with development of ACD is necessary. Despite their signifi­
strategies pre‐patch test avoidance utilizing personal care cant populations in the USA, both Hispanics and blacks
products devoid of these allergens [29]. have been historically underrepresented in the dermato­
logical literature. Data from the Pediatric Contact
Emerging concepts. From its pivotal discovery by Dermatitis Registry showed that black children were
Jadassohn in 1895, the immunology of contact sensitiza­ noted to experience dermatitis 1.2 years longer than
tion and subsequent allergic presentation has intrigued whites and 1.6 years longer than Hispanics [152].
dermatologists and allergists alike. The complex inter­ Additionally, patch‐tested patients designated as Asian or
play between ACD and atopic skin disease is poorly African American were more likely to have concurrent
understood, confounded by conflicting evidence in the atopic dermatitis (odds ratio [OR], 1.92; 95% confidence
literature. An estimated 20–40% of atopic infants are interval [CI], 1.20–3.10; P = .008; and OR, 4.09; 95% CI,
known to have an innate genetic filaggrin mutation 2.70–6.20; P  < 
.001, respectively). Because minorities
resulting in an impaired skin barrier and increased tran­ account for a growing proportion of the American
sepidermal water loss. Also related to filaggrin mutations ­population, the need for more data to better understand
are an increased pH and alteration of inherent antimicro­ disease manifestation in skin of colour is imperative.
bial peptides, all which could potentially contribute to an Understanding the intrinsic and extrinsic co‐morbidi­
increased risk for allergen sensitization [145]. ties that put patients at risk for developing ACD is an area
312 Section 4  Other Types of Dermatitis

in need of further research, especially given the reported 7 Fortina AB, Romano I, Peserico A, Eichenfield LE. Contact sensitiza­
tion in very young children. J Am Acad Dermatol 2010;65:772–8.
rising rates in the paediatric population. Rates of both AD
8 Sarma N, Ghosh S. Clinico‐allergological pattern of allergic contact
and ACD in the USA are notably higher than that of dermatitis among 70 Indian children. Indian J Dermatol Venereol
developing nations. Interestingly, vitamin D deficiency Leprol 2010;76:38–44.
has been connected to the development of allergic 9 Jacob SE, Yang A, Herro E, Zhang C. Contact allergens in a pediatric
population: association with atopic dermatitis and comparison with
­illnesses, and furthermore vitamin D may play an impor­ other North American referral centers. J Clin Aesthet Dermatol
tant role in regulating antimicrobial peptides in 2010;3:29.
keratinocytes [153,154]. Other environmental factors have 10 Toledo F, García‐Bravo B, Fernández‐Redondo V et al. Patch testing in
been postulated to play roles, including the abundant use children with hand eczema. A 5‐year multicentre study in Spain.
Contact Dermatitis 2011;65:213–19.
of cleansing antibacterial products aimed at removing 11 Kuljanac I, Kneževic ́ E, Cvitanovic ́ H. Epicutaneous patch test results in
bacteria from the skin, including the protective normal children and adults with allergic contact dermatitis in Karlovac county:
skin flora such as Staphylococcus epidermidis. These resi­ a retrospective survey. Acta Dermatovenerol Croat 2011;19:91–7.
12 Jacob SE, Herro EM, Sullivan K, Matiz C et  al. Safety and efficacy
dent flora have been shown to perform many ‘mutualis­ evaluation of TRUE Test Panels 1.1, 2.1, 3.1, in children and adoles­
tic’ functions contributing to effective skin barrier cents. Dermatitis 2011;22:204–10.
function, including producing antibacterial peptides that 13 Schena D, Papagrigoraki A, Tessari G et al. Allergic contact dermatitis
aid the skin’s innate immune system and assist in main­ in children with and without atopic dermatitis. Dermatitis
2012;23:275–80.
SECTION 4: OTHER TYPES

taining the skin’s barrier function through enhancing 14 Silny W, Bartoszak L, Jenerowicz D et al. Prevalence of contact allergy
tight junction expression and decreasing pro‐inflamma­ in children suffering from atopic dermatitis, seborrhoeic dermatitis
OF DERMATITIS

tory cytokines [145,155,156]. Colonization of the skin by and in healthy controls. Ann Agric Environ Med 2013;20:55–60.
15 Zug KA, Pha AK, Belsito DV et al. Patch testing in children from 2005
Staph. epidermidis is also known to impair the establish­
to 2012: Results from the North American Contact Dermatitis Group.
ment of colonization with Staph. aureus, an organism often Dermatitis 2014;25:345–55.
responsible for superimposed infection on atopic or other 16 Jacob S, Goldenberg A, Pelletier J et  al. Nickel allergy and our
chronically inflamed skin [145]. Researchers recently ­children’s health: a review of indexed cases and a view of future
­prevention. Pediatric Dermatology. 2015;32(6):779–85.
found increased Staph. aureus infections in atopic patients 17 Thyssen J, Hald M, Avenstorp C, et  al. Characteristics of nickel‐­
with nickel allergy in comparison to atopic patients and allergic dermatitis patients seen in private dermatology clinics in
healthy volunteers without nickel allergy. Furthermore, Denmark: a questionnaire study. Acta Derm Venereol 2009;89:384–8.
elevated secretion of IL‐2 under nickel sulphate stimula­ 18 Elsaie M, Olasz E, Jacob S. Cytokines and Langerhans cells in allergic
contact dermatitis. G Ital Dermatol Venereol 2008;143:195–205.
tion in vitro was found exclusively in atopic patients with 19 Fuhlbridge R, Kieffer J, Amerding D, Kupper T. Cutaneous lympho­
nickel allergy infected by Staph. aureus, suggesting a link cyte antigen is specialized form of PSGL‐1 expressed on skin‐homing
between nickel allergy and staphylococcal infection in T cells. Nature 1997;389:978–81.
20 Friedmann P. Contact sensitization and allergic contact dermatitis:
AD [157]. immunobiological mechanisms. Toxicol Lett 2006;162:49–54.
As ACD in children continues to be better recognized, 21 Rietschel R, Fowler JJ. The pathogenesis of allergic contact hypersen­
further investigation is needed. Collaboration between sitivity. In: Fisher’s Contact Dermatitis, 5th edn. Philadelphia:
patients, medical practitioners, industry and policy Lippincott Williams and Wilkins.
22 So J, Hamstra A, Calame A et  al. Another great imitator: allergic
makers is necessary to promote safety directives that
­ ­contact dermatitis: differential diagnosis, clues to diagnosis, histopa­
appropriately regulate known chemical sensitizers result­ thology, and treatment. Curr Treat Options Allergy 2015;2:333–48.
ing in preventable sensitizations and a reduction of the 23 Czarnobilska E, Obtulowicz K, Dyga W et  al. The most important
­contact sensitizers in Polish children and adolescents with atopy and
burden of ACD.
chronic recurrent eczema as detected with the extended European
Baseline Series. Pediatr Allergy Immunol 2011;22:252–6.
24 Goldenberg A, Jacob S. Pediatric Patch Test Registry: Abstract
Disclosure Presentation. Paper presented at the ACDS Annual Conference, 2016,
Washington, DC.
Sharon E. Jacob is the Founder and Chief Executive 25 Jacob S, Goldenberg A, Nedorost S,et  al. Flexural eczema versus
Officer of the Dermatitis Academy, a free online resource atopic dermatitis. Dermatitis. 2015;26:109–15.
on contact dermatitis. 26 Winnicki M, Shear N. A systematic approach to systematic contact
dermatitis and symmetric drug‐related intertriginous and flexural
exanthema (SDRIFE): a closer look at these conditions and an
References approach to intertriginous eruptions. Am J Clin Dermatol.
1 Straus H. Artificial sensitization of infants to Poison Ivy. J Allergy 2011;12:171–80.
1931;2:137–44. 27 Allen H, Vaze N, Choi C et al. The presence and impact of biofilm‐
2 Weston WL, Weston JA, Kinoshita J et al. Prevalence of positive epicu­ producing staphylococci in atopic dermatitis. JAMA Derm
taneous tests among infants, children, and adolescents. Pediatrics 2014;150:260–5.
1986;78:1070–4. 28 Hann S, Hughes T, Stone N. Flexural allergic contact dermatitis to
3 Thyssen J, Linneberg A, Menné T, et al. The epidemiology of contact benzalkonium chloride in antiseptic bath oil. Br J Dermatol
allergy in the general population  –  prevalence and main findings. 2007;157:795–8.
Contact Dermatitis 2007;57:287–99. 29 Hill H, Goldenberg A, Golkar L et  al. Pre‐Emptive Avoidance
4 Mortz C, Lauritsen J, Blindslev‐Jensen C et al. Prevalence of atopic der­ Strategy (P.E.A.S.) – addressing allergic contact dermatitis in pediat-
matitis, asthma, allergic rhinitis and hand and contact dermatitis in ric populations. Expert Rev Clin Immunol 2016;12:551–61.
adolescents. Br J Dermatol 2001;144:523–32. 30 Thyssen JP, Menné T. Metal allergy – a review on exposures, penetra­
5 Dotterud L, Falk E. Contact allergy in relation to hand eczema and tion, genetics, prevalence, and clinical implications. Chem Res Toxicol
atopic diseases in north Norwegian schoolchildren. Acta Paediatr 2009;23:309–18.
1995;84:402–6. 31 Carlsen BC, Johansen JD, Menné T et al. Filaggrin null mutations
6 Milingou M, Tagka A, Armenaka M et  al. Patch tests in children: a and association with contact allergy and allergic contact dermatitis:
review of 13  years of experience in comparison with previous data. results from a tertiary dermatology clinic. Contact Dermatitis
Pediatr Dermatol 2010;27:255–9. 2010;63:89–95.
Chapter 23  Allergic Contact Dermatitis 313

32 Nijhawen R, Matiz C, Jacob SE. Contact dermatitis: from basics to 59 Fisher A. Childhood allergic contact dermatitis. Cutis 1975;15:635–42.
allergodromes. Pediatr Ann 2009;38:99–108. 60 Darlenski R, Kazandjieva J, Tsankov N et al. Acute irritant threshold
33 Hanifin J. Filaggrin mutations and allergic contact sensitization. correlates with barrier function, skin hydration and contact hypersen­
J Invest Dermatol 2008;128:1362–4. sitivity in atopic dermatitis and rosacea. Exp Dermatol 2013;22:752–3.
34 Novak N, Baurecht H, Shäfer T et  al. Loss‐of‐function mutations in 61 Hoeger P. Antimicrobial susceptibility of skin‐colonizing S. auerus
the filaggrin gene and allergic contact sensitization to nickel. J Invest strains in children with atopic dermatitis. Pediatr Allergy Immunol
Dermatol 2008;128:1430–5. 2004;15:474–7.
35 Lerbaek A, Bisgaard H, Agner T et al. Filaggrin null mutations are not 62 Admani S, Matiz C, Jacob SE. Countering Staphylococcus overgrowth
associated with hand eczema or contact allergy. Br J Dermatol during patch testing in children with moderate to severe atopic der­
2007;157:1199–204. matitis. Pediatr Dermatol 2016;33:56–61.
36 De Jongh C, Khrenova L, Verbeck M et al. Loss‐of‐function polymor­ 63 Wee JS, White JM, McFadden JP, White IR. Patch testing in patients
phisms in the filaggrin gene are associated with an increased suscepti­ treated with systemic immunosuppression and cytokine inhibitors.
bility to chronic irritant dermatitis: a case‐control study. Br J Dermatol Contact Dermatitis 2010;62:165–9.
2008;159:621–7. 64 Olupona T, Scheinman P. Successful patch testing despite concomi­
37 Di Nardo A, Braff M, Taylor K et  al. Cathelicidin antimicrobial tant low‐dose prednisone use. Dermatitis 2008;19:117–18.
­peptides block dendritic cell TLR‐4 activation and allergic contact 65 Kohl L, Blondeel A, Song M. Allergic contact dermatitis from cosmet­
sensitization. J Immunol 2007;178:1829–34. ics. Dermatology 2002;204:334–7.
38 Nacak M, Erbgci Z, Aynacioglu A. Human argylamine N‐acetyl‐ 66 Jacob SE, Brod B, Crawford GH. Clinically relevant patch test reac­
transferase‐2 polymorphism and susceptibility to allergic contact der­ tions in children—a United States based study. Pediatr Dermatol
matitis. Int J Dermatol 2006;45:323–6. 2008;25:520–7.
39 Reich K, Westphal G, Konig I et al. Association of allergic contact der­ 67 Hogeling M, Pratt M. Allergic contact dermatitis in children: the

SECTION 4: OTHER TYPES


matitis with a promoter polymorphism in the IL‐16 gene. J Allergy Ottawa hospital patch‐testing clinic experience, 1996‐2006. Dermatitis
Clin Immunol 2003;112:1191–4. 2008;19:86–9.

OF DERMATITIS
40 White J, Goon A, Jowsey I et al. Oral tolerance to contact allergens: a 68 Rietschel RL, Fowler JF, Warshaw EM et al. Detection of nickel sensi­
common occurrence? A review. Contact Dermatitis 2007;56:247–54. tivity has increased in North America patch‐test patients. Dermatitis
41 Spiewak R. Food‐provoked eczema: a hypothesis on the possible role 2008;19:16–19.
of systemic contact allergy to haptens present in both cosmetics and 69 Fors R, Persson M, Berström E et al. Nickel allergy—prevalence in a
foods. Estetol Med Kosmetol 2011;1:35–40. population of Swedish youths from patch test and questionnaire data.
42 Moed H, Boorsma D, Tensen C et al. Increased CCL27‐CCR10 expres­ Contact Dermatitis 2008;58:80–7.
sion in allergic contact dermatitis: implications for local skin memory. 70 Rystedt I, Fischer T. Relationship between nickel and cobalt sensitiza­
J Pathol 2004;204:39–46. tion in hard metal workers. Contact Dermatitis 1983;9:195–200.
43 Mortz CG, Lauritsen JM, Bindslev‐Jensen C, Andersen KE. Nickel 71 Silverberg NB. The “Jewelry Addict”: allergic contact dermatitis from
sensitization in adolescents and association with ear piercing, use of repetitive multiple children’s jewelry exposures. Pediatr Dermatol
dental braces and hand eczema. Acta Derm Venereol 2002;82:6. 2016;33:e103–5.
44 Smith‐Silversten T, Tchachtchine V, Lund E. Environmental nickel 72 Pelton A, Russell S, DiCello J. The physical metallurgy of nitinol for
pollution: Does it protect against nickel allergy? J Am Acad Dermatol medical applications. JOM 2003;55:33–7.
2002;36:460–2. 73 Jacob SE, Admani S. Ipad  –  increasing nickel exposure in children.
45 Mak R, White I, White J et al. Lower incidence of sesquiterpene lac­ Pediatrics 2014;134:580–2.
tone sensitivity in a population in Asia versus a population in Europe: 74 Silverberg N, Licht J, Friedler S et al. Nickel contact hypersensitivity
an effect of Crysanthemum tea? Contact Dermatitis 2007;57:163–4. in children. Pediatr Dermatol 2002;19:110–13.
46 Brod B, Treat J, Rothe M, Jacob S. Allergic contact dermatitis: kids 75 Matiz C, Jacob S. Systemic contact dermatitis in children: how an avoid­
are not just little people. Clin Dermatol 2015;33:605–12. ance diet can make a difference. Pediatr Dermatol 2011;28:368–74.
47 Admani S, Jacob SE. Allergic contact dermatitis in children: review of 76 Jensen CS, Menné T, Duus Johansen J. Systemic contact dermatitis
the past decade. Curr Allergy Asthma Rep 2014;14:1–11. after oral exposure to nickel: a review with a modified meta‐analysis.
48 Jacob S, Herro E. Practical patch testing and chemical allergens in Contact Dermatitis 2006;54:79–86.
contact dermatitis: Compositae Mix. Springer Science & Business
­ 77 Bergman D, Goldenberg A, Rundle C, Jacob S. Low nickel diet: a
Media, 2013. patient‐centered review. J Clin Exp Dermatol Res 2016;7:355.
49 Jacob S, Burk C, Connelly E. Patch testing: another steroid‐sparing 78 Jensen C, Menné T, Lisby S et al. Experimental systemic contact der­
agent to consider in children. Pediatr Dermatol 2008;25:81–7. matitis from nickel: a dose‐response study. Contact Dermatitis
50 Flohr C, Weiland S, Weinmayr G et al. The role of atopic sensitization 2003;49:124–32.
in flexural eczema: findings from the International Study of Asthma 79 Johansen J, Menne T, Christophersen J et al. Changes in the pattern of
and Allergies in Childhood Phase Two. J Allergy Clin Immunol sensitization to common contact allergens in Denmark between
2008;121:141–7. 1985‐86 and 1997‐98 with a special view to the effect of preventive
51 Jacob SE, McGowan M, Silverberg NB, Pelletier JL, Fonacier L, strategies. Br J Dermatol 2000;142:490–5.
Mousdicas N, Powell D, Scheman A, Goldenberg A. Pediatric Contact 80 Thyssen J, Uter W, McFadden J et al. The EU Nickel Directive revis­
Dermatitis Registry Data on Contact Allergy in Atopic Children. ited‐future steps toward better protection against nickel allergy.
JAMA Derm 2017;153:765–70. Contact Dermatitis 2011;64:121–5.
52 Zug KA, McGinley‐Smith D, Warshaw EM et al. Contact allergy in 81 Gehrig K, Warshaw E. Allergic contact dermatitis to topical antibiot­
children referred for patch testing: North American Contact ics: Epidemiology, responsible allergens, and management. J Am
Dermatitis Group data, 2001‐2004. Arch Dermatol 2008;144:1329–36. Acad Dermatol 2008;58:1–21.
53 Goldenberg A, Jacob S. Demographics of US Pediatric Contact 82 Leyden J, Kligman A. Rationale for topical antibiotics. Cutis
Dermatitis Registry Providers. Dermatitis 2015;26:184–8. 1978;22:515–27.
54 Goldenberg A, Mousdicas N, Silverberg N et  al. Pediatric Contact 83 Jacob S, James W. From road rash to top allergen in a flash: bacitracin.
Dermatitis Registry Inaugural Case Data. Dermatitis J 2016;27:293–302. Dermatol Surg 2004;30:521–4.
55 Worm M, Aberer W, Agathos M et al. Patch testing in children – rec­ 84 Mailhol C, Lauwers‐Cances V, Rance F et  al. Prevalence and risk
ommendations of the German Contact Dermatitis Research Group ­factors for allergic contact dermatitis to topical treatment in atopic
(DKG). J Dtsch Dermatol Ges 2007;5:107–9. dermatitis: a study in 641 children. Allergy 2009;64:801–6.
56 Jacob S, Hill H, Lucero H, Nedorost S. Benzoate allergy in children— 85 Lerbaek A, Kyvik K, Menne T et al. Retesting with the T.R.U.E. Test in
from foods to personal hygiene products. Pediatr Dermatol a population‐based twin cohort with hand eczema: allergies and
2016;33:213–15. persistence in an 8‐year old follow up study. Contact Dermatitis
­
57 Package Insert ‐ TRUE TEST ‐ ucm294327.pdf. 2018; Available at: 2007;57:248–52.
https://www.fda.gov/downloads/biologicsbloodvaccines/533 86 Hamann C, Bernard S, Hamann D et al. Is there a risk using hypoal­
allergenics/ucm294327.pdf (accessed 26 November 2018). lergenic cosmetic pediatric products in the United States? J Allergy
58 Waard‐van der Spek FB, Darsow U, Mortz CG et al. EAACI position Clin Immunol 2014;135:4.
paper for practical patch testing in allergic contact dermatitis in chil­ 87 Scheinman P. Exposing covert fragrance chemicals. Am J Contact
dren. Pediatr Allergy Immunol 2015;26:598–606. Dermat 2001;12:225–8.
314 Section 4  Other Types of Dermatitis

88 Warshaw E, Maibach H, Taylor J. North American Contact Dermatitis 120 Arroyo M. Black henna tattoo reaction in a person with sulfonamide
Group Patch Test Results: 2011‐2012. Dermatitis 2015;26:49–59. and benzocaine allergy. J Am Acad Dermatol 2003;48:301–2.
89 Jacob S. Allergen Focus: Myroxylon Pereirae (Balsam of Peru). The 121 Leysat S, Boone M, Blondeel A et al. Two cases of cross‐sensitivity in
Dermatologist 2014(March):20–22. subjects allergic to paraphenylenediamine following ingestion of
90 Hill H, Jacob S. Peri‐anal ulcerations in a patient with essential pru­ polaronil. Dermatology 2003;206:379–80.
ritus. Dermatitis 2015;26:292–3. 122 Schafer T, Enders F, Przybilla B. Sensitization to thimerosal and pre­
91 Scheman A, Cha C, Jacob S. Food avoidance diets for systemic, lip vious vaccination. Contact Dermatitis 1995;32:114–16.
and oral contact allergy. Dermatitis 2012;23:248–57. 123 Breithaupt A, Jacob S. Thimerosal and the relevance of patch‐test
92 Scheman A, Rakowski E‐M, Chou V et al. Balsam of Peru: past and reactions in children. Dermatitis 2008;19:275–7.
future. Dermatitis 2013;24:153–60. 124 Wohrl S, Hemmer W, Focke M et al. Patch testing in children, adults,
93 Marks J, Alsner P, DeLeo V. Contact and Occupational Dermatology: and the elderly: influence of age and sex on sensitization patterns.
Standard Allergens, 3rd edn. St. Louis, MO: Mosby, 2001. Pediatr Dermatol 2003;20:119–23.
94 Larsen W. How to instruct patients sensitive to fragrances. J Am 125 Belsito D. Thimerosal: contact (non) allergen of the year. Am J Contact
Acad Dermatol 1989;21:880–4. Dermat 2002;13:1–2.
95 Paulsen E, Otkjaer A, Andersen K. Sesquiterpene lactone dermatitis 126 Center for Food Safety and Applied Nutrition’s (CFSAN’s) Adverse
in the young: is atopy a risk factor? Contact Dermatitis 2008;59:1–6. Event Reporting System (CAERS) MAUDE report on 8/20/2015
96 Warshaw E, Schram S, Belsito D et al. Shoe allergens: retrospective Body Mist by Kim Kardashian. Data available at: https://www.fda.
analysis of cross‐sectional data from the North American Contact gov/food/complianceenforcement/ucm494015.htm#files (accessed
Dermatitis group, 2001‐2004. Dermatitis 2008;58:1–21. 28 November, 2018).
97 Holden C, Gawkrodger D. 10 years experience of patch testing with 127 Wright AM, Cahill JL. Airborne exposure to methylisothiazolinone
a shoe series in 230 patients: which allergens are important? Contact in paint causing allergic contact dermatitis: An Australian perspec­
SECTION 4: OTHER TYPES

Dermatitis 2005;53:37–9. tive. Australas J Dermatol 2016;57:294–5.


98 Darling M, Horn H, McCormack S et al. Sole dermatitis in children: 128 Hunter K, Shelley J, Haworth A. Airborne allergic contact dermatitis
OF DERMATITIS

patch testing revisited. Pediatr Dermatol 2012;29:254–7. to methylchloroisothiazolinone/methylisothiazolinone in ironing


99 Lee PW, Elsaie ML, Jacob SE. Allergic contact dermatitis in children: water. Contact Dermatitis 2008;58:183–4.
common allergens and treatment: a review. Curr Opin Pediatr 128 Lundov MD, Menné T. Airborne exposure to methylchloroisothia­
2009;21:491–8. zolinone and methylisothiazolinone from a toilet cleaner. Contact
100 Jacob S. The lanolin‐wool wax alcohol update. The Dermatologist Dermatitis 2013;68:252–3.
2014;22:2. 130 US Department of Health and Human Services. Household Products
101 Lee B, Warshaw E. lanolin allergy: history, epidemiology, responsible Database; Chemical Name: Methylisothiazolinone. 2015. Available
allergens, and management. Dermatitis 2008;19:63–72. at: https://householdproducts.nlm.nih.gov/cgi‐bin/household/
102 Wolf R. The lanolin paradox. Dermatology 1996;192:198–202. brands?tbl=chem&id=72 (accessed 22 November, 2018).
103 Matthieu L, Dockx P. Discrepancy in patch test results with wool wax 131 Aerts O, Cattaert N, Lambert J, Goossens A. Airborne and systemic
alcohols and Amerchol L‐101. Contact Dermatitis 1997;36:150–1. dermatitis, mimicking atopic dermatitis, caused by methylisothia­
104 Onken AT, Baumstark J, Belloni B et al. Atypical diaper dermatitis: zolinone in a young child. Contact Dermatitis 2013;68:250–1.
contact allergy to mercapto compounds. Pediatr Dermatol 2011; 132 Lundov M, Opstrup M, Johansen J. Methylisothiazolinone contact
28:739–41. allergy – a growing epidemic. Contact Dermatitis 2013;69:271–5.
105 Adams A, Warshaw E. Allergic contact dermatitis from Mercapto 133 Lipp M, Bertolino M, Goldenberg A, Jacob S. Allergen Focus: Update
Compounds. Dermatitis 2006;17:56–70. on Isothiazolinones. The Dermatologist 2016;May:43–8.
106 Jacob S, Amani S. Cocamidopropyl betaine. Dermatitis 2008; 134 Castanedo‐Tardana M, Zug K. Methylisothiazolinone. Dermatitis
19:157–60. 2013;24:2–6.
107 Parsons L. Cocamidopropyl betaine. Dermatitis 2008;19:E49–50. 135 Zug K, Rietschel R, Warshaw E et  al. The value of patch testing
108 Pontén A, Bruze M. Formaldehyde. Dermatitis 2015;26:3–6. patients with a scattered generalized distribution of dermatitis:
109 de Groot A, Flyvholm M, Lensen G et al. Formaldehyde releasers: rela­ ­retrospective cross‐sectional analyses of North American Contact
tionship to contact allergy. Contact allergy to formaldehyde and inven­ Dermatitis Group data, 2001‐2004. J Am Acad Dermatol 2008;
tory of formaldehyde releasers. Contact Dermatitis 2009;61:63–85. 59:426–31.
110 Hill A, Belsito D. Systemic contact dermatitis of the eyelids caused 136 Hannuksela M, Forstrom L. Reactions to perioral propylene glycol.
by formaldehyde derived from aspartame? Contact Dermatitis Contact Dermatitis 1978;4:41.
2003;49:258–9. 137 Pootongkam S, Nedorost S. Focus on propylene glycol. The
111 Jacob S, Stechschulte S. Formaldehyde, aspartame and migraines: a Dermatologist 2013;21.
possible connection. Dermatitis 2008;19:E10–11. 138 Dao H, Fricker C, Nedorost S. Sensitization prevalence for benzalko­
112 Weston W, Morelli J. Dermatitis under soccer shin guards: allergy or nium chloride and bezethonium chloride. Dermatitis 2012;23:162–6.
contact irritant reactions? Pediatr Dermatol 2006;23:19–20. 139 Giusti F, Massone F, Bertoni L et al. Contact sensitization to disperse
113 Baeck M, Chemelle J, Goossens A et al. Corticosteroid cross‐reactiv­ dyes in children. Pediatr Dermatol 2003;20:393–7.
ity: clinical and molecular modelling tools. Allergy 2011;66:1367–74. 140 Alberta L, Sweeney S, Wiss K. Diaper dye dermatitis. Pediatr
114 Warshaw E, Botto N, Maibach H et al. Positive patch‐test reactions to Dermatol 2005;116:E450–2.
propylene glycol: a retrospective cross‐sectional analysis from the 141 Fischer L, Menné T, Johansen J. Dose per unit area  –  a study of
North American Contact Dermatitis Group, 1996 to 2006. Dermatitis elicitation of nickel allergy. Contact Dermatitis 2007;56:7.
2009;20:14–20. 142 Fischer T, Fregert S, Gruvberger B, Rystedt I. Nickel release from ear
115 Coloe J, Zirwas M. Allergens in corticosteroid vehicles. Dermatitis piercing kits and earrings. Contact Dermatitis 1984;10:39–41.
2008;19:38–42. 143 Herro EM, Jacob SE. A nickel for your thoughts: determining relative
116 Jacob S, Zapolanski T, Chayavichitsilp P et al. P‐phenylenediamine nickel content using an analog color scale. Dermatitis 2012;23:2.
in black henna tattoos: a practice in need of policy in children. Arch 144 Yiannias J. Common Contact Allergens. Paper presented at the
Pediatr Adolesc Med 2008;162:790–2. Annual Meeting of the American College of Allergy, Asthma and
117 United States Consumer Product Safety Commission (CPSC). Immunology, Houston, 2015.
Hazardous substances and articles; supplemental definition of 145 Baviera G, Leoni MC, Capra L et al. Microbiota in healthy skin and
“strong sensitizer”. 2013. Available at: https://www.federalregister. in atopic eczema. BioMed Res Int 2014;2014.
gov/documents/2013/03/12/2013‐05577/hazardous‐substances‐ 146 O’Regan G, Sandilands A, McLean W et  al. Filaggrin in atopic
and‐articles‐supplemental‐definition‐of‐strong‐sensitizer (accessed ­dermatitis. J Allergy Clin Immunol 2008;122:689–93.
28 November, 2018). 147 Gao P, Leung D, Rafaels N et  al. Genetic variants in TSLP and its
118 Sosted H, Johansen J, Andersen K et al. Severe allergic hair dye reac­ receptor, IL7R, contribute to an increased risk for atopic dermatitis
tions in 8 children. Contact Dermatitis 2006;54:87–91. and eczema herpeticum in two American populations. J Allergy Clin
119 Spornraft‐Ragaller P, Kämmerer E, Gillitzier C et al. Severe allergic Immunol 2009;123:S70.
reactions to para‐phenylenediamine in children and adolescents: 148 Boguniewicz M, Leung DY. Recent insights into atopic dermatitis
should the past test concentration of PPD be changed? J Dtsch and implications for management of infectious complications.
Dermatol Ges 2012;10:258–64. J Allergy Clin Immunol 2010;125:4–13.
Chapter 23  Allergic Contact Dermatitis 315

149 Cohen D, Heidary N. Treatment of irritant and allergic contact der­ 154 Schauber J, Gallo R. Antimicrobial peptides and the skin immune
matitis. Dermatol Ther 2004;17:334–40. defense system. J Allergy Clin Immunol 2008;122:261–6.
150 Kucharekova M, Hornix M, Ashikaga T et al. The effect of the PDE‐4 155 Lai Y, Di Nardo A, Nakatsuji T et al. Commensal bacteria upregulate
inhibitor (cipamfylline) in two human models of irritant contact der­ toll‐like receptor 3‐dependent inflammation after skin injury. Nat
matitis. Arch Dermatol Res 2003;295:29–32. Med 2009;15:1377–82.
151 Jung T, Stingl G. Atopic dermatitis: Therapeutic concepts evolving 156 Yuki I, Yoshida H, Akazawa Y et  al. Activation of TLR2 enhances
from new pathophysiologic insights. J Allergy Clin Immunol tight junction barrier in epidermal keratinocytes. J Immunol
2008;122:1074–81. 2011;187:3230–7.
152 McGowan MA, Goldenberg A, Jacob SE. Contact dermatitis in 157 Anna BM, Grazyna A, Wojciech D et al. Nickel allergy and relation­
underrepresented minority. Dermatitis 2018;29:97–9. ship with Staphylococcus aureus in atopic dermatitis. J Trace Elem
153 Oren E, Banerji A, Camargo CJ. Vitamin D and atopic disorders in an Med Biol 2016;33:1–7.
obese population screened for vitamin D deficiency. J Allergy Clin
Immunol 2008;121:533–4.

SECTION 4: OTHER TYPES


OF DERMATITIS
316 

CHA PTER 2 4

Hypereosinophilic Disorders
Eirini E. Merika & Nerys Roberts
Chelsea & Westminster Hospital, London, UK

Introduction, 316 Hypereosinophilic syndrome, 324 Eosinophilic panniculitis, 333


Eosinophilic pustular folliculitis in infancy Eosinophilic cellulitis (Wells syndrome), 328
and childhood (syn. Ofuji disease), 318 Eosinophilic fasciitis, 331

Abstract ­ utlines the clinical features, investigation and treatment of those


o
hypereosinophilic disorders where the eosinophil is thought to be
SECTION 4: OTHER TYPES

central to the pathogenesis (i.e. eosinophilic cellulitis, panniculitis,


Eosinophils are well established as inflammatory cells and are now
fasciitis and pustular folliculitis and hypereosinophilic syndrome).
recognized to have an important role in host protection, immune
OF DERMATITIS

Childhood rashes where eosinophils are present (such as atopic


regulation and inflammation. This chapter discusses the common-
dermatitis, drug rashes and immunobullous disorders) are not
est context in which peripheral blood eosinophilia is a ­ ssociated
­discussed as they are covered elsewhere.
with a skin rash in neonates, infants and older children and

Key points with high mortality in view of potential cardiac complications.


Symptomatic patients are usually steroid responsive.
• Eosinophilic cellulitis is a rare but important diagnosis.
• Eosinophils are multifunctional cells that contribute to both
Infection, drug reaction, trauma, connective tissue disease or
innate and adaptive immunity. They are involved in the
haematological malignancy may underlie it. It is characterized
initiation, propagation and resolution of immune responses,
by abrupt onset of tender lesions that rapidly spread in a well
including tissue repair.
child. Flame figures on histology are the most common feature.
• Eosinophilic pustular folliculitis of infancy most frequently
If treatment is required, it should be anti‐inflammatory rather
appears in the first months of life and resolves by the age of 3.
than antibiotic treatment.
All patients have scalp lesions, tissue eosinophilia and recurrent
• Eosinophilic fasciitis is characterized by eosinophil‐rich, symmetrical
outbreaks. Blood eosinophilia is present during flares but may
sclerodermatous skin changes in the extremities. It is associated with
normalize in between. It is usually benign and self‐limiting.
connective tissue disorders, underlying malignancy, haematological
• A hypereosinophilic syndrome (HES) diagnosis requires blood
disease and drug reactions. Peripheral blood eosinophilia is seen in
eosinophilia and tissue damage due to eosinophil accumulation.
70% of cases. It is usually steroid responsive.
Persistent eosinophilia is a feature. End‐organ damage must be
• Eosinophilic panniculitis presents with subcutaneous nodular
present for the diagnosis to be made. Patients should be investigated
swelling. It is associated with underlying disease. It is a lobular
for an underlying haematopoietic neoplasm. Reactive causes must
and septal eosinophil‐rich panniculitis without vasculitis and is
be excluded for a diagnosis of idiopathic HES. HES is associated
usually steroid responsive.

Introduction
also dependent on transcription factors – GATA‐binding
Eosinophils have been easily recognizable in tissues for protein 1, enhancer binding protein alpha, box binding
over a century because of the characteristic staining of protein and interferon consensus sequence binding pro-
their granules with eosin on haematoxylin, yet their pre- tein. Surface immunoglobulin‐like transcripts 2 and 5
cise role in health and disease is only gradually being receptors are also emerging as important controllers of
elucidated. development and apoptosis.
Eosinophils are multifunctional cells that contribute to Eosinophils are involved in the initiation, propagation
both innate and adaptive immunity. They are bone mar- and resolution of immune responses, including tissue
row derived and develop from progenitor cells when repair. Eosinophils are able to sense pathogens and pro-
stimulated by interleukin (IL)‐3, granulocyte‐macrophage mote innate immune responses via expression of comple-
colony‐stimulating factor and IL‐5, the last being the ment receptors, Fc receptors and pattern recognition
major regulator of eosinophil accumulation in tissues, receptors including multiple toll‐like receptors.
which can modulate eosinophil behaviour at every stage Once activated, eosinophils promote host protection
from maturation to survival. Eosinophil development is via direct effects on pathogens, immune responses by

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 24  Hypereosinophilic Disorders 317

modulation of lymphocyte and dendritic cell function Familial or hereditary eosinophilia has been described,
and inflammation via tissue damage, remodelling and with multiple family members showing eosinophilia, and
mast cell activation. an autosomal dominant inheritance was proposed. Initial
Eosinophils release soluble mediators such as reports, however, were not followed by other familial
cytokines, chemokines, growth factors and bioactive cases so it is difficult to be certain if this is a distinct entity
lipids. Their cytokines can induce both T‐helper 1 and [10,11]. Furthermore, Ota et  al. reported three siblings
T‐helper 2 type inflammation as well as fibrosis (trans- with features of familial eosinophilia but who clinically
forming growth factor beta). IL‐4 production by eosino- had lung disease  –  two had a Churg–Strauss syndrome
phils is thought to be important not only in regulating (eosinophilic granulomatosis with polyangiitis [EGPA])
immune, but also metabolic, processes including beige phenotype and the third had eosinophilic pruritus. Thus
fat development. In addition, eosinophils produce cyto- there seems to be an overlap with hypereosinophilic syn-
toxic proteins: major basic protein, eosinophil peroxi- drome (HES) [12].
dase, eosinophil cationic protein and eosinophil‐derived In Down syndrome, eosinophil numbers are often nor-
neurotoxin. mal but there is a higher percentage of eosinophils with
Eosinophil transit time in the blood is short (about multilobed nuclei in the bone marrow. Clonal eosino-
1  day) but they accumulate in tissues for several days. philia in Down syndrome has recently been shown to be

SECTION 4: OTHER TYPES


Extravasation is chemokine receptor 3 dependent. Thymic linked to the N‐terminal truncating mutation of GATA‐1
stromal lymphopoietin and IL‐33 are important recruiting (which has also been linked to transient leukaemia in

OF DERMATITIS
factors. Down syndrome neonates) [13].
Eosinophils undergo apoptosis in the absence of the A number of genetic syndromes may have eosinophilia
proper cytokine milieu but when activated induce as a feature, including Netherton syndrome, incontinentia
increased vascular permeability, recruit leucocytes (espe- pigmenti, Dorfman–Chanarin syndrome, trichothiodys-
cially T‐helper T cells but also prime B cells to produce trophy, hyper‐IgE syndrome, Omenn syndrome, IPEX
immunoglobulin [Ig]M) to sites of inflammation, interact (immune dysregulation, polyendocrinopathy, enteropathy,
with dendritic cells (activation, maturation and migration X‐linked) syndrome and peeling skin syndrome; also
to lymph nodes) and IgA and IgE antibodies and produce hypereosinophilic syndrome in a child mosaic for a congen-
or store mediators which are pro‐inflammatory, profibro- ital triplication of the short arm of chromosome 8 has been
genic, proangiogenic and cytotoxic (some are bacterio- described. Congenital Langerhans histiocytosis, congenital
static). In addition, neurotoxin release may account for Wells syndrome, congenital Ofuji and congenital toxoplas-
the intense itch (and sometimes pain) associated with mosis also may be associated with hypereosinophilia.
eosinophilic dermatoses. Recent studies suggest that
eosinophils may also have a role in antitumoural immune Neonatal eosinophilia
surveillance and have tumouricidal potential [1–3]. In neonates, tissue eosinophilia is seen in erythema toxi-
Furthermore eosinophils are now thought to be key par- cum neonatorum, infantile acropustulosis and transient
ticipants in innate immunity and tissue remodelling neonatal pustular melanosis.
events [4] given their ability to both store and synthesize Peripheral blood eosinophilia may be a feature of
a range of immunomodulatory factors that can be rapidly incontinentia pigmenti, but marked peripheral blood
released upon activation [5]. eosinophilia in a baby with erythroderma should arouse
In the clinical setting, in developed countries, a circulat- suspicion of an immunodeficiency, namely Omenn
ing eosinophilia most often signals an atopic condition syndrome.
(eczema, asthma, hay fever) or other allergic state (e.g. In a sick neonate, sepsis should be considered, as eosin-
drug reaction); in the East or Africa, an underlying inter- ophilia in this age group may be an indicator of sepsis
nal parasitosis is the cause until proven otherwise. and thus infection should be in the differential diagnosis
International air travel can thus spring occasional clinical of a sick neonate with eosinophilia >700/mm3.
surprises. Transient hypereosinophilia has been described in the
infant of a mother with HES.
Congenital eosinophilia
In the newborn, the blood eosinophil count is usually Infantile eosinophilia
<0.7 × 109/L, although counts of 1–2 × 109/L are seen occa- Eosinophilia is most common in food allergy and atopic
sionally in otherwise normal infants [6,7]. Cord blood dermatitis, in which tissue eosinophilia is characteristic.
contains more eosinophils and precursor cells than adult Tissue eosinophilia may also be a feature of Langerhans
peripheral blood. cell histiocytosis and annular erythema of infancy. A high
Congenital eosinophilia following intrauterine and to moderate eosinophilia is a feature of eosinophilic pus-
exchange blood transfusions is not uncommon and tular folliculitis.
occurred in 60% of neonates in one series [8]. This is Very marked peripheral blood eosinophilia in infants
thought to be the result of a hypersensitivity reaction as it must be taken seriously, as it may herald either idiopathic
can be associated with a rash, thrombocytopenia and HES or leukaemia. WT1 reverse transcription polymerase
lymphopenia. A case has recently been reported of con- chain reaction (RT‐PCR) is a useful diagnostic tool to
genital blaschkoid angiolymphoid hyperplasia with distinguish these as WT1 gene expression is increased in
eosinophilia of the anogenital region [9]. virtually all patients with acute leukaemias [14,15].
318 Section 4  Other Types of Dermatitis

Eosinophilia in older children 17 Salomon J, Białynicki‐Birula R, Woźniak Z, Baran E. Pachydermatous


eosinophilic dermatitis. Acta Dermatovenerol Croat 2011;19:31–5.
Marked eosinophilia is seen in idiopathic HES, Kimura 18 Dierksmeier U, Frosch PJ, Czarnetzki BM. Eosinophil chemotactic
disease/angiolymphoid hyperplasia with eosinophilia factor (ECF) in blister fluid of dermatological diseases. Br J Dermatol
and hyper‐IgE syndrome (the last two are described in 1980;102:43–8.
Chapter 113).
Moderate peripheral blood eosinophilia is a feature of
Wells syndrome, eosinophilic fasciitis and Ofuji syn-
Eosinophilic pustular folliculitis
drome. The few cases of pachydermatous eosinophilic
in infancy and childhood (syn.
dermatitis are characterized by peripheral blood eosino-
Ofuji disease)
philia and pruritic papular lesions on a pachydermatous
base [16,17]. Clearly, the possibility of infestations and Brief introduction/history. Eosinophilic pustular folliculitis is
infections such as scabies and coccidioidomycosis must an uncommon disorder first described in adults by Ise and
be considered. Ofuji in 1965 [1], although the term eosinophilic pustular
Tissue eosinophilia is a feature of numerous derma- folliculitis was ascribed by Ofuji et  al. in 1970 [2]. There
toses discussed elsewhere. A low‐molecular‐weight have been fewer than 70 reports of this entity in children.
chemotactic factor for eosinophils has been found in the Broadly, there are three variants: (i) classic, (ii) immu-
nosuppression‐associated, which is subdivided into
SECTION 4: OTHER TYPES

skin of patients with bullous pemphigoid, atopic dermati-


tis/eczema, drug reaction and contact sensitivity [18]. human immunodeficiency virus (HIV) and non‐HIV
associated, and (iii) infancy‐associated.
OF DERMATITIS

Other dermatoses include urticaria (Chapter 63), mycosis


fungoides (Chapter  88), graft‐versus‐host disease The classic adult disease is a rare inflammatory disease
(Chapter  157), mastocytosis (Chapter  92), vasculitides of unknown aetiology characterized by recurrent plaques
including polyarteritis nodosa and EGPA (Chapter 148), with clusters of sterile follicular pustules and papules
systemic sclerosis (Chapter 100), dermatitis herpetiformis affecting the face, trunk and limbs, mainly in Japanese
(Chapter 75), pemphigus (Chapter 8) and Langerhans cell patients. The use of the term ‘folliculitis’ has been chal-
histiocytosis (Chapter 90). lenged because hair follicles are not seen on the palms or
the soles, which may be affected.
References
There is a variation of the classic type that has been
1 Neves JS, Weller PF. Functional extracellular eosinophil granules: reported as ‘episodic eosinophilic dermatosis of the face’
novel implications in eosinophil immunobiology. Curr Opin which affects seborrhoeic areas of the face but lacks typi-
Immunol 2009;21:694–9. cal features of classic EPF such as pustule formation and
2 Minai‐Fleminger Y, Levi‐Schaffer F. Mast cells and eosinophils: the
two key effector cells in allergic inflammation. Inflamm Res peripheral extension of the lesions; it responds well to
2009;58:631–8. indometacin.
3 Blanchard C, Rothenberg ME. Biology of the eosinophil. Adv In 1984, Lucky et al. described a variant in infants that
Immunol 2009;101:81–121.
4 ShamriR, Xenakis JJ, Spencer LA. Eosinophils in innate immunity:
had its own set of characteristics, which distinguish it
an evolving story. Cell Tissue Res 2011;343:57–83. from the adult and childhood forms [3]. There is debate
5 Davoine F, Lacy P. Eosinophil cytokines, chemokines, and growth over whether the infantile form is truly a variant of the
factors: emerging roles in immunity. Front Immunol 2014;5:570. adult form or a completely separate disorder [3–15], and
6 Lawrence R Jr, Church JA, Richards W,Lipsey AI. Eosinophilia in
the hospitalized neonate. Ann Allergy 1980;44:349–52. the name eosinophilic pustulosis of the scalp has been
7 Kien CL, Chusid MJ. Eosinophilia in children receiving parenteral proposed for infants in whom the scalp is primarily
nutrition support. JPEN J Parenter Enteral Nutr 1979;3:468–9. affected [16] and there is no true follicular involvement
8 Chudwin DS, Ammann AJ, Wara DW et al. Posttransfusion syndrome.
[17,18]. In adult patients, an association with HIV infec-
Rash, eosinophilia, and thrombocytopenia following intrauterine and
exchange transfusions. Am J Dis Child 1982;136:612–14. tion and haematological malignancy or its treatment is
9 Su HH, Shan SJ, Elston DM et  al. Congenital blaschkoid angiolym- well established [19–22]. There has been a single case
phoid hyperplasia with eosinophilia of the anogenital region. Am J report of HIV‐associated eosinophilic pustular folliculitis
Dermatopathol 2016;38:305–6.
10 Rioux JD, Stone VA, Daly MJ et al. Familial eosinophilia maps to
in an infant [23]. In children, the condition has not been
the cytokine gene cluster on human chromosomal region 5q31‐q33. associated with any underlying disease before or after its
Am J Hum Genet 1998;63:1086–94. diagnosis.
11 Klion AD Law MA, Riemenschneider W et  al. Familial eosino-
philia: a benign disorder? Blood 2004;103:4050–5.
12 Ota M, Takenaka K, Takahashi M, Nagasaka K. Eosinophilia with Epidemiology and  pathogenesis. Both the classic and
organ involvement in 3 siblings. Nihon Rinsho Meneki Gakkai Kaishi infantile forms exhibit a strong male predilection
2012;35:533–8.
13 Maroz, A, Stachorski L, Emmrich S et al. GATA1s induces hyperpro- (male : female ratio 4 : 1); however, in the 4‐ to 9‐year age
liferation of eosinophil precursors in Down syndrome transient group, which represents by far the smallest cohort, more
leukemia. Leukemia 2014;28:1259–70. cases have been reported in girls. The disease tends to
14 Menssen HD, Renkl HJ, Rieder H et al. Distinction of eosinophilic leu-
present before 14 months of life and spontaneously
kaemia from idiopathic hypereosinophilic syndrome by analysis of
Wilms’ tumour gene expression. Br J Haematol 1998;101:325–34. resolves by 3 years of age in 80% of cases. Presence at
15 Cilloni D, Messa F, Martinelli G et al. WT1 transcript amount discrimi- birth has been recorded [24]. It has a higher incidence in
nates secondary or reactive eosinophilia from idiopathic hypereo- Caucasians and is rare in Japan. Although there does not
sinophilic syndrome or chronic eosinophilic leukemia. Leukemia
2007;21:1442–50.
seem to be a familial predisposition, eosinophilic pustular
16 Jacyk WK, Simson IW, Slater DN, Leiferman KM. Pachydermatous folliculitis has been reported in two brothers, born at differ-
eosinophilic dermatitis. Br J Dermatol 1996;134:469–74. ent times, both with onset in the neonatal period [25].
Chapter 24  Hypereosinophilic Disorders 319

The origin of this dermatosis is unknown. Currently, The study of indometacin, a potent cyclo‐oxygenase
many theories exist, particularly for the classic adult inhibitor, has provided valuable insight and potential
form, none of which has been validated. Most theories into the pathogenesis and treatment of the disease
evoke immunological mechanisms in the initiation of respectively (Fig.  24.1). It acts as a potent agonist of
lesions, particularly because of the association with atopy, CRTH2 (chemoattractant receptor‐homologous mole-
diabetes, infection (HIV, hepatitis C and Giardia), stem cell cule expressed on Th2 cells), which is a receptor for
transplantation and drugs (minocycline, allopurinol, PGD2 (prostaglandin D2). PGD2 mediates its biological
anticonvulsant therapy, timepidium bromide and activities through CTH2 receptors, including eosinophil
indeloxazine hydrochloride). Resolution of the lesions of chemokinesis and chemotaxis and degranulation
eosinophilic pustular folliculitis in a patient with (Fig.  24.1). Indometacin is thought to both desensitize
Gorlin syndrome when her jaw cyst was excised was the CRTH2 receptor and downmodulate cell surface
thought to reflect the removal of an immune dysregula- expression of both CRTH2 and CCR3 (eotaxin receptor
tory stimulus [26]. on eosinophils) and therefore reduce eosinophil responses
The presence of an eosinophilic infiltrate around to PGD2. Importantly, Th2 cells and basophils are also
and into the dermal hair follicles is hypothesized to known to express CRTH2 [29] and a basophilic infiltrate
suggest involvement of a Th2‐type response in the has also been reported in skin lesions of eosinophilic

SECTION 4: OTHER TYPES


pathogenetic mechanism and resultant IL‐4 and IL‐13 pustular folliculitis [30]. Reduction of eosinophil CRTH2
mediated eotaxin synthesis and thus eosinophil expression in association with improvement of skin

OF DERMATITIS
induction and migration. Eotaxin‐1, a highly specific lesions and reduction in blood eosinophilia has been
chemokine, is also thought to play a crucial role in observed in the context of oral indometacin treatment,
eosinophil recruitment, inflammation and tissue dam- further supporting the hypothesis of PGD2–CRTH2
age, and its autocrine production has been postulated interaction in the pathogenesis [30,31].
to account for the chronicity seen in eosinophilic pus- The presence of PGD2‐producing enzyme in hair
tular folliculitis [27,28]. follicle epithelium, dermal eccrine glands, eccrine ostia in

PPAR-γ

PGD2 Sebocyte
CRTH2
in hair
PGD2
follicle
Th2
L-PDGS
Th0
INDOM

IL-4
CCL26
IL-5
TGF-β Eotaxin-3
Dendritic IL-4
cell
TLR INDOM
PRR
Produces
HPGDS PGD2
Fc
Basophil PGD2
is
ax
ot

R
R
CRTH2 activation em
Ca2+mobilisation Ch
cell migration
CCR3
TSLP degranulation

IFN-γ Eotaxin
IL-12 CD11b

INDOM

Th1
Proinflammation
Host
protection Immunoregulation
Fig. 24.1  Eosinophil function. CCL, chemokine (C‐C motif) ligand; CRTH, chemoattractant receptor‐homologous molecule; HPGDS,
­haematopoietic prostaglandin D synthase; IFN, interferon; IL, interleukin; INDOM, indometacin; L‐PDGS, lipocalin‐type prostaglandin D2
synthase protein; PGHDE, prostaglandin; PPAR, peroxisome proliferator‐activated receptor; PRR, pattern recognition receptor; TGF,
­transforming growth factor; Th, T helper; TLR, toll‐like receptor; TSLP, thymic stromal lymphopoietin.
320 Section 4  Other Types of Dermatitis

stratum corneum and epidermal keratinocytes but also face, trunk and extremities. There is a tendency for central
the acrosyringium [32] (where hair follicles are lacking) healing with peripheral extension. Scalp involvement is
around eosinophilic pustules is suggestive of local syn- present in only 6% of adult patients [38]. This classic form
thesis of PGD2 and accumulation of CRTH2‐expressing has rarely been reported in older children and adolescents
cells in the disease pathogenesis [31]. [5,36,38–48].
PGD2 also stimulates sebocyte production of eotaxin‐3 Eosinophilic pustular folliculitis in infancy appears to
(CCL26) leading to accumulation of eosinophils in seba- be a variant of this disorder and principally involves the
ceous hair follicles [33]. It is postulated that upstream scalp [3–15,25,49]. The lesions, similar to those in the
PGD2 signalling as well as downstream CCR3 signalling adult form, occur as grouped, pruritic firm papules
is implicated in the underlying accumulation of eosino- (occasionally vesicles), which evolve into follicular
phils, and indometacin mediates its effects through pustules, measuring pinpoint to 3 mm, on an erythema-
desensitization of the CRTH2 signals in eosinophils as tous base. The lesions can present as a solitary plaque or
well as through inhibition of PGD2 synthesis locally [31]. as scattered pustules throughout the scalp [23]. Unlike
An exaggerated response to skin saprophytes or der- the adult and childhood forms, annular or serpiginous
matophytes, or autoantibodies directed against the inter- plaques are not observed in infants and small children.
cellular substance of the lower epidermis [34] or the They resolve spontaneously without scarring after
SECTION 4: OTHER TYPES

cytoplasm of basal cells of the epidermis and the outer evolving through a yellow, crusted phase in approximately
sheath of hair follicles [35], has also been proposed. In 5–10 days, often leaving a 3–4 mm hyperpigmented
OF DERMATITIS

adult eosinophilic pustular folliculitis, autoimmunity macule. New crops of lesions appear approximately
against sebaceous glands has been postulated as an aetio- every 2–8 weeks. In addition to the scalp, the majority of
logical factor, especially for the classic and HIV‐related infants have involvement of the extremities, hands, feet
forms [19]; however, it is presumed that this is probably and trunk. The face [5,16,40], groin [6] and genitals [5]
not the case for the paediatric variants. may be affected. Mucosal involvement was noted in one
A case series of 15 paediatric cases published in 2013 7‐year‐old child [43]. In the infantile variant, onset is
identified a possible association with atopy [18]. Boone most likely in the first 6 months of life, with approxi-
et  al. [36] described three children, two with childhood mately one quarter presenting at or just after birth [9,15].
onset and one with onset in infancy, who tested positive Spontaneous resolution occurs mostly after a total dura-
with specific immunoglobulin E (sIgE) radio‐allergosorbent tion of 3 months to 5 years. It appears that the earlier the
testing to the dust mite Dermatophagoides pteronyssinus disorder appears, the less chronic it becomes. The clini-
(DPT), suggesting a hypersensitivity reaction in the cal characteristics of eosinophilic pustular folliculitis in
pathogenesis of eosinophilic pustular folliculitis. Two of the 4‐ to 9‐year age group fall between the other groups
the three patients had neither personal nor family history in that scalp involvement is unusual, seborrhoeic areas
of atopy. are less involved than in the classic form and there are
Nitric oxide has been implicated in the pathogenesis of fewer annular and more diffuse lesions as seen in the
eosinophilic pustular folliculitis. Nitric oxide is a simple infantile form [38].
inorganic molecule that can regulate vascular tone, plate- The term necrotizing eosinophilic folliculitis has been
let aggregation and leucocyte adhesion and is implicated coined for a variant of eosinophilic folliculitis presenting
in neurotransmission, immunomodulation and with ulcerating nodules. Magro and Crowson [50]
inflammation. Neuronal nitric oxide synthase (nNOS) is described 10 patients, one of whom was an 11‐year‐old
expressed by neurons but was also found in the eosino- girl, who had in common a history of atopy and evidence
phils that infiltrate the dermis and hair follicles of patients of follicular and dermal necrosis with an eosinophilic
with eosinophilic pustular folliculitis. Similar immuno- vasculitis on biopsy of their ulcerated nodules.
histochemical findings have been described in other
eosinophilic dermatoses such as Kimura disease, leading
the authors to speculate that eosinophil‐derived nitric Differential diagnosis. The differential diagnosis for
oxide may be pathogenic in this inflammatory condition. eosinophilic pustular folliculitis in infancy should
It is postulated that nNOS can catalyse the formation of include most pustular eruptions in infancy, in particular
ONOO–, which leads to cellular and tissue injury. scalp pyoderma or infectious folliculitis caused by
Furthermore, nitric oxide and its other isoforms have Staphylococcus aureus, dermatophyte infections, sca-
been shown to reduce eosinophil locomotion and prolong bies, erythema toxicum neonatorum, transient neonatal
eosinophil survival, hence aiding their accumulation [37]. pustular melanosis, herpes simplex infection, infantile
Eosinophilic pustular folliculitis shares some histo- acropustulosis, Langerhans cell histiocytosis, incontinen-
pathological characteristics with erythema toxicum neo- tia pigmenti, hypereosinophilic syndrome and drug
natorum and some hypothesize that it may represent a eruptions. The generalized form of eosinophilic pustular
more persistent form [16]. folliculitis in adults tends to be drug induced, most often
in association with allopurinol, but carbamazepine and
Clinical features. Eosinophilic pustular folliculitis (Fig. 24.2) minocycline have also been implicated, whereas there
is characterized by recurrent crops of intensely pruritic have been no such reports in children.
annular or polycyclic plaques, composed of coalescing, When eosinophilic pustular folliculitis presents in older
sterile papulopustules on the seborrhoeic areas of the children, the differential diagnosis should include fungal,
Chapter 24  Hypereosinophilic Disorders 321

(a) (b)

SECTION 4: OTHER TYPES


OF DERMATITIS
(c)

(d)
Fig. 24.2  Eosinophilic pustular folliculitis on the (a) scalp, (b) face, (c) trunk and (d) limbs. Note the sparing of the skin creases.

parasitic and herpetic infections, eosinophilic cellulitis, mid dermis, composed predominantly of eosinophils
insect bites, eczema, impetigo, lymphoma and drug together with neutrophils and mononuclear cells
eruptions. (Fig.  24.3). Mast cells of the tryptase‐positive chymase‐
negative subtype are moderately increased [39]. The infil-
Laboratory findings. In all age groups there is mild to trate may extend to the deep dermis [25,39,40] and
moderate leucocytosis and in 50–83% of cases eosino- subcutaneous fat [5,16]. Most cases demonstrate intersti-
philia; this is usually associated with exacerbations and tial eosinophilic flame figures between collagen bundles,
an absence of systemic symptoms [18,51]. IgE is usually representing degranulated eosinophils [5,52]. Eosinophil,
normal or marginally elevated. neutrophil and mononuclear cell exocytosis into the
A Wright’s‐stained smear of pustular contents demon- pilosebaceous unit, with spongiosis, subsequent abscess
strating abundant eosinophils may be helpful in making formation and ultimate destruction of the hair follicle, is
the diagnosis. The diagnosis is based on the clinical pres- frequently demonstrated in the adult and older paediatric
entation, associated haematological abnormalities and cases [49,53], whereas most, but not all, of the infantile
histological evidence of eosinophilic folliculitis. cases present with simply perifollicular mixed, but pre-
dominantly eosinophilic, inflammation [21]. Sebaceous
Histopathology. Histopathology of the infantile scalp glands may be infiltrated and, with follicular involve-
lesion displays a moderate to dense perifollicular and ment, the infundibulum is most affected [40]. The epidermis
periappendiceal inflammatory infiltrate in the upper and may also exhibit exocytosis, spongiosis and occasionally
322 Section 4  Other Types of Dermatitis
SECTION 4: OTHER TYPES

(a) (b)
Fig. 24.3  Histopathology of skin biopsy taken from the child illustrated in Fig. 24.2. Source: Courtesy of Dr Nick Francis.
OF DERMATITIS

eosinophilic subcorneal pustules, parakeratosis or crust Responses to oral antibiotics (penicillin G, erythromycin,
[5,9]. The scalp is the preferred biopsy site in infants, as it cephalexin) have been variable [3,13,16,40].
is invariably affected in this age group and consistently In adults, it has been suggested that nonsteroidal
yields a specific histological picture [16,40]. Follicular anti‐inflammatory drugs such as indometacin and nap-
mucinosis, mycosis fungoides and other cutaneous T‐cell roxen are the agents of choice [16,57]. Both topical and
lymphomas are important to consider in the histopatho- oral (0.4  mg/kg) indometacin have been successfully
logical differential diagnosis. used for refractory infantile cases [58,59]. It is not advisa-
ble to use indometacin in children; it is unlicensed in the
Treatment. Because of the rarity of eosinophilic pustular UK, has a high incidence of side‐effects including head-
folliculitis, there have been no controlled clinical trials for ache, dizziness, and gastrointestinal disturbances, and
its treatment in adults or children. In addition, its recur- children may be more sensitive to serious liver problems.
rent and self‐limited features, especially in infants, make In patients with HIV‐associated disease, there is generally
evaluation of anecdotal treatments difficult. a good response to antiretroviral therapy, but eosinophilic
Antihistamines are indicated for associated pruritus, pustular folliculitis can occur with immune reconsti-
and there may be a role for the eosinophil antimigration tution. Other treatment options include topical and
drugs such as cetirizine dihydrochloride [6,54] and systemic steroids, isotretinoin, dimethyl diphenyl
ketotifen. Dose‐dependent response to hydroxyzine has sulfone, sulfamethoxazole, oxyphenbutazone, minocycline,
also been reported [24], but concerns regarding prolon- doxycycline, metronidazole, itraconazole, cyprohepta-
gation of the QT interval with this sedating antihista- dine, colchicine, glycyrrhizin, interferon‐α2b, interferon‐γ
mine should be heeded in infants and young children. and ciclosporin. Topical permethrin 5% cream, transdermal
The H2 antagonist cimetidine was used with success in a nicotine patches and, for the acquired immunodefi-
9‐month‐old girl [55]. ciency syndrome (AIDS)‐associated variety, phototherapy
A large comprehensive analysis of therapeutic (PUVA [psoralens and ultraviolet A] and narrow‐band)
responses by Nomura et al. [56] reported on 115 regimens may be successful [40,60]. Radiation was helpful in a
used in infantile eosinophilic pustular folliculitis. The recalcitrant case [61].
review showed that topical steroids were highly effica- Monitoring of children with infantile eosinophilic
cious in the majority of cases (n = 50, efficacy 82%). pustular folliculitis is advised as it may rarely be the
Tacrolimus was associated with the highest clearance rate presenting complaint of hyper‐IgE syndrome. Some
in a small number of children. Carbon light and topical children went on to develop hyper‐IgE syndrome an
tacalcitol have also been tried without reports on efficacy. average of 18 months later [62,63], but these children had
Combination therapies, such as antihistamines with topi- associated systemic features.
cal indometacin or topical steroids, or systemic antimicro-
bials with topical steroids, have also been used in infants.
Dapsone, at a dose of 2 mg/kg per day, was effective in References
1 Ise S, Ofuji S. Subcorneal pustular dermatosis. A follicular variant?
one infant who was unresponsive to topical steroids; Arch Dermatol 1965;92:169–71.
however, it was discontinued secondary to haematological 2 Ofuji S, Ogino A, Horio T et  al. Eosinophilic pustular folliculitis.
side‐effects. The eruption recurred after the dapsone was Acta Derm Venereol 1970;50:195–203.
3 Lucky AW, Esterly NB, Heskel N et al. Eosinophilic pustular follicu-
stopped [25]. Dapsone has been further reported in other
litis in infancy. Pediatr Dermatol 1984;1:202–6.
paediatric cases with overall 67% efficacy in clearing the 4 Taieb A. Infantile eosinophilic pustular “folliculitis” in infancy: a
lesions [56]. nonfollicular disease. Pediatr Dermatol 1994;11:186.
Chapter 24  Hypereosinophilic Disorders 323

5 Taieb A, Bassan‐Andrieu L, Maleville J. Eosinophilic pustulosis of the 33 Nakahigashi K, Doi H, Otsuka A et  al. PGD2 induces eotaxin‐3 via
scalp in childhood. J Am Acad Dermatol 1992;27:55–60. PPARgamma from sebocytes: a possible pathogenesis of eosinophilic
6 Larralde M, Morales S, Santos Muñoz A et al. Eosinophilic pustular pustular folliculitis. J Allergy Clin Immunol 2012;129:536–43.
folliculitis in infancy: report of two new cases. Pediatr Dermatol 34 Vakilzadeh F, Suter L, Knop J, Macher E. Eosinophilic pustulosis with
1999;16:118–20. pemphigus‐like antibody. Dermatologica 1981;162:265–72.
7 Vicente J, España A, Idoate M et al. Are eosinophilic pustular fol- 35 Nunzi E, Parodi A, Rebora A. Ofuji’s disease: high circulating titers of
liculitis of infancy and infantile acropustulosis the same entity? IgG and IgM directed to basal cell cytoplasm. J Am Acad Dermatol
Br J Dermatol 1996;135:807–9. 1985;12:268–73.
8 Coulson IH, Ling TC, Stringfellow HF. Case 2. Infantile eosinophilic 36 Boone M, Dangoisse C, André J et al. Eosinophilic pustular folliculitis
pustular folliculitis (IEPF). Clin Exp Dermatol 2002;27:80–1. in three atopic children with hypersensitivity to Dermatophagoides
9 Buckley DA, Munn SE, Higgins EM. Neonatal eosinophilic pustular pteronyssinus. Dermatology 1995;190:164–8.
folliculitis. Clin Exp Dermatol 2001;26:251–5. 37 Maruo K, Kayashima KI, Ono T. Expression of neuronal nitric oxide
10 Ziemer M, Boer A. Eosinophilic pustular folliculitis in infancy: not synthase in dermal infiltrated eosinophils in eosinophilic pustular fol-
a distinctive inflammatory disease of the skin. Am J Dermatopathol liculitis. Br J Dermatol 1999;140:417–20.
2005;27:443–55. 38 Dekio S, Jidoi J, Kawasaki Y. Eosinophil‐infiltrating folliculitis in
11 Luelmo Aguilar J, Saez Artacho A. [Eosinophilic pustular folliculitis childhood–report of a case. J Dermatol 1989;16:388–91.
in childhood]. An Esp Pediatr 2001;55:154–8. 39 Ishiguro N, Shishido E, Okamoto R et al. Ofuji’s disease: a report on
12 Rybojad M, Guibal F, Vignon‐Pennamen MD et al. [Eosinophilic pus- 20 patients with clinical and histopathologic analysis. J Am Acad
tulosis in an infant accompanied by immune deficit]. Ann Dermatol Dermatol 2002;46:827–33.
Venereol 1999;126:29–31. 40 Giard F, Marcoux D, McCuaig C et al. Eosinophilic pustular folliculitis
13 Garcia‐Patos V, Pujol RM, de Moragas JM. Infantile eosinophilic (Ofuji disease) in childhood: a review of four cases. Pediatr Dermatol

SECTION 4: OTHER TYPES


­pustular folliculitis. Dermatology 1994;189:133–8. 1991;8:189–93.
14 Khorshid SM, Glover M, Cerio R. Infantile eosinophilic pustular 41 Takematsu H, Nakamura K, Igarashi M, Tagami H. Eosinophilic pus-

OF DERMATITIS
folliculitis: report of the first British case. Eur J Dermatol 1997;7:385–7. tular folliculitis. Report of two cases with a review of the Japanese
15 Picone Z, Madile B. Eosinophilic pustular folliculitis (Ofuji’s disease) literature. Arch Dermatol 1985;121:917–20.
in a newborn. Pediatr Dermatol 1992;9:178. 42 Hsu PJ, Huang CJ, Wu MT. Pathergy in atypical eosinophilic pustular
16 Duarte AM, Kramer J, Yusk JW et al. Eosinophilic pustular follicu- folliculitis. Int J Dermatol 2005;44:203–5.
litis in infancy and childhood. Am J Dis Child 1993;147:197–200. 43 Boudaya S, Turki H, Bouassida S et al. [Eosinophilic pustular folliculitis
17 Patel NP, Laguda B, Roberts NM et al. Treatment of eosinophilic pus- in infancy: an unusual case]. Ann Dermatol Venereol 2003;130:451–4.
tulosis of infancy with topical tacrolimus. Br J Dermatol 2012; 44 Kawaguchi M, Mitsuhashi Y, Kondo S. Successful treatment of eosin-
167:1189–91. ophilic pustular folliculitis with topical tacrolimus. Int J Dermatol
18 Hernandez‐Martin A, Nuño‐González A, Colmenero I, Torrelo A. 2004;43:608–10.
Eosinophilic pustular folliculitis of infancy: a series of 15 cases and 45 Lazarov A, Wolach B, Cordoba M et al. Eosinophilic pustular follicu-
review of the literature. J Am Acad Dermatol 2013;8:150–5. litis (Ofuji disease) in a child. Cutis 1996;58:135–8.
19 Fearfield LA, Rowe A, Francis N et al. Itchy folliculitis and human 46 Jang KA, Chung ST, Choi JH et  al. Eosinophilic pustular folliculitis
immunodeficiency virus infection: clinicopathological and immu- (Ofuji’s disease) in myelodysplastic syndrome. J Dermatol 1998;25:742–6.
nological features, pathogenesis and treatment. Br J Dermatol 1999; 47 Souissi A, Fenniche S, Benmously R et  al. [Eosinophilic pustular
141:3–11. ­folliculitis in a child]. Tunis Med 2008;86:190–2.
20 Bull RH, Harland CA, Fallowfield ME, Mortimer PS. Eosinophilic fol- 48 De Dulanto F, Armijo Moreno M, Díaz Flores L, et al. [Eosinophilic
liculitis: a self‐limiting illness in patients being treated for haemato- pustular folliculitis (Ofuji syndrome)]. Med Cutan Ibero Lat Am
logical malignancy. Br J Dermatol 1993;129:178–82. 1977;5:323–9.
21 Otley CC, Avram MR, Johnson RA. Isotretinoin treatment of human 49 Darmstadt GL, Tunnessen WW Jr, Swerer RJ. Eosinophilic pustular
immunodeficiency virus‐associated eosinophilic folliculitis. Results folliculitis. Pediatrics 1992;89:1095–8.
of an open, pilot trial. Arch Dermatol 1995;131:1047–50. 50 Magro CM, Crowson AN. Necrotizing eosinophilic folliculitis as a
22 Takamura S, Teraki Y. Eosinophilic pustular folliculitis associated manifestation of the atopic diathesis. Int J Dermatol 2000;39:672–7.
with hematological disorders: A report of two cases and review of 51 Alonso‐Castro L, Pérez‐García B, González‐García C, Jaén‐Olasolo P.
Japanese literature. J Dermatol 2016;43:432–5. Eosinophilic pustular folliculitis in infancy: report of a new case.
23 Ramdial PK, Morar N, Dlova NC, Aboobaker J. HIV‐associated eosin- Dermatol Online J 2012;18:6.
ophilic folliculitis in an infant. Am J Dermatopathol 1999;21:241–6. 52 Onorato J, Heilman ER, Laude TA. Pruritic pustular eruption in an
24 Lee JH, Kang JH, Cho BK, Park HJ. Generalized eosinophilic pustular infant. Pediatr Dermatol 1993;10:292–4.
folliculitis of infancy responding to hydroxyzine. Ann Dermatol 53 Lee JY, Tsai YM, Sheu HM. Ofuji’s disease with follicular mucinosis
2015;27:458–60. and its differential diagnosis from alopecia mucinosa. J Cutan Pathol
25 Dupond AS, Aubin F, Bourezane Y et  al. Eosinophilic pustular 2003;30:307–13.
folliculitis in infancy: report of two affected brothers. Br J Dermatol 54 Fadel R, Herpin‐Richard N, Rihoux JP, Henocq E. Inhibitory effect of
1995;132:296–9. cetirizine 2HCl on eosinophil migration in vivo. Clin Allergy 1987;17:373–9.
26 Kishimoto S, Yamamoto M, Nomiyama T et al. Eosinophilic pustular 55 Rogers M. Successful treatment of eosinophilic pustulosis with oral
folliculitis in association with nevoid basal cell carcinoma syndrome. cimetidine. Pediatr Dermatol 1999;16:335–6.
Acta Derm Venereol 2001;81:202–4. 56 Nomura T, Katoh M, Yamamoto Y. Eosinophilic pustular folliculi-
27 Amerio P, Frezzolini A, Feliciani C et al. Eotaxins and CCR3 receptor tis: A published work‐based comprehensive analysis of therapeutic
in inflammatory and allergic skin diseases: therapeutical implica- responsiveness. J Dermatol 2016;43:919–27.
tions. Curr Drug Targets Inflamm Allergy 2003;2:81–94. 57 Youn CS, Cho KH. Eosinophilic pustular folliculitis treated with nap-
28 Amerio, P, Verdolini R, Proietto G et  al. Role of Th2 cytokines, roxen. Br J Dermatol 2001;145:514–15.
RANTES and eotaxin in AIDS‐associated eosinophilic folliculitis. 58 Hashizume S, Ansai S, Kosaka M, Kawana S. Infantile case of eosino-
Acta Derm Venereol 2001;81:92–5. philic pustular folliculitis successfully treated with topical indometh-
29 Hirai H, Tanaka K, Yoshie O et  al. Prostaglandin D2 selectively acin. J Dermatol 2014;41:196–7.
induces chemotaxis in T helper type 2 cells, eosinophils, and basophils 59 Toyonaga E, Abe R, Moriuchi R. Indomethacin for refractory infan-
via seven‐transmembrane receptor CRTH2. J Exp Med 2001;193:255–61. tile eosinophilic pustular folliculitis. JAMA Dermatol 2013;149:36728.
30 Satoh T, Ito Y, Miyagishi C, Yokozeki H. Basophils infiltrate skin 60 Ellis E, Scheinfeld N. Eosinophilic pustular folliculitis: a compre-
lesions of eosinophilic pustular folliculitis (Ofuji’s disease). Acta hensive review of treatment options. Am J Clin Dermatol 2004;5:189–97.
Derm Venereol 2011;91:371–2. 61 Wilson BD, Kucera JC, Shin PJ. The role of radiation treatment in the
31 Kataoka N, Satoh T, Hirai A et  al. Indomethacin inhibits eosinophil management of eosinophilic pustular folliculitis. J Med 2002;33:111–13.
migration to prostaglandin D2: therapeutic potential of CRTH2 62 Chamlin SL, McCalmont TH, Cunningham BB. Cutaneous manifesta-
desensitization for eosinophilic pustular folliculitis. Immunology tions of hyper‐IgE syndrome in infants and children. J Pediatr
2013;140:78–86. 2002;141:572–5.
32 Satoh T, Ikeda H, Yokozeki H. Acrosyringeal involvement of palmo- 63 Lo CS, Yang CY, Ko JH. Hyperimmunoglobulin E syndrome present-
plantar lesions of eosinophilic pustular folliculitis. Acta Derm ing as eosinophilic pustular folliculitis: a case report. Int J Dermatol
Venereol 2013;93:99. 2015;54:211–4.
324 Section 4  Other Types of Dermatitis

Hypereosinophilic syndrome [5–7] and lungs [8]. The muscles [9], gastrointestinal
tract [10–13], eyes, nasopharynx [14], bone marrow and
Brief introduction. The normal eosinophil count in the central nervous system [15–18] may be involved.
peripheral blood ranges from 50 to 500  ×  109/L. Rarely, the kidneys [19] and bladder [20] are involved.
Eosinophilia can be divided into mild eosinophilia (up to 3 Exclusion of known primary and secondary causes of
1500 × 109/L) and marked eosinophilia (>1500 × 109/L). eosinophilia.
Recent advances in cytogenetics, immunology and
molecular biology have led to the re‐classification of Epidemiology and  pathogenesis. Although HES is rare
hypereosinophilia (HE) as follows [1–3]: in childhood (mean age of onset 37 years), in the series
1 Familial (hereditary); reported by Alfaham 16 of the 18 children had cardiac
2 Of undetermined significance (unexplained, persistent, involvement; untreated, this carries a high mortality [21].
asymptomatic eosinophilia) (in lieu of the previous term There is a male to female ratio of 1.47.
idiopathic hypereosinophilia); HES is an important syndrome to recognize because
3 Primary (clonal/neoplastic, where eosinophils are con- eosinophilic infiltration of the cardiac tissue induces
sidered neoplastic); and endomyocardial thrombosis and fibrosis and resultant
4 Secondary (reactive, to an underlying condition with restrictive cardiomyopathy, valvular dysfunction and
SECTION 4: OTHER TYPES

cytokine‐driven eosinophilia, such as parasitic or viral increased thrombotic tendency as well as aneurysmal
infections, allergic diseases, drug‐ or chemical‐induced disease [22–24].
OF DERMATITIS

eosinophilia, hypocortisolism and neoplasms). It is still not clear whether eosinophils themselves or
‘Hypereosinophilic syndrome’ (HES) is a term reserved for their precursors are abnormal in HES, or whether there is
any variant with blood eosinophilia and clear evidence of a problem in the regulatory mechanism of eosinophil
end‐organ damage and is a multisystem disease with a high production.
mortality rate. It is usually considered a provisional diagno- The Splendore–Hoeppli phenomenon (presence of
sis until a primary or secondary cause for eosinophilia is asteroid bodies) may occur in HES. This phenomenon is
identified. An absolute eosinophil count of >1500/mm3 well recognized as a pathological response to infection
must be present on two occasions 1 month apart and some and has been described with fungi (including sporotri-
authorities suggest that the eosinophilia should persist for chosis, Pityrosporum folliculitis, zygomycosis, candidiasis,
at least 6 months before this diagnosis is established. aspergillosis and blastomycosis), bacteria (botryomyco-
Similar to HE, HES is classified as follows: sis, nocardiosis and actinomycosis) and parasites (includ-
1 Idiopathic HES, a diagnosis of exclusion, where there is ing orbital pythiosis, strongyloidiasis, schistosomiasis
end‐organ damage attributable to HE but no underly- and cutaneous larva migrans). It can also occur around
ing cause for HE, including a reactive or neoplastic biologically inert substances (in HES and allergic conjunc-
condition. tival granulomas).
2 Primary (neoplastic), where there is an underlying stem It is thought to represent the in vivo formation of
cell, myeloid or eosinophilic neoplasm classified intensely eosinophilic material which produces radiating,
according to WHO guidelines and end‐organ damage star‐like, asteroid or club‐shaped configurations (around
attributable to HE. In this case eosinophils are consid- microorganisms or inert material). The Splendore–
ered neoplastic. Hoeppli reaction material is made up of antigen–antibody
3 Secondary (reactive), where there is an underlying non‐ complexes, tissue debris and fibrin and is thought to be a
neoplastic or paraneoplastic condition responsible for localized immunological response to an antigen–antibody
the production of nonclonal eosinophils. Eosinophils in precipitate related to fungi, parasites, bacteria or inert
this group are usually cytokine (most often IL‐5) driven materials. It is thought that this reaction prevents phago-
and end‐organ damage is attributable to HE. The lym- cytosis and intracellular killing of the insulting agent,
phoid variant (L‐HES) is a form of reactive HES, but the leading to chronicity of infection [25].
lymphocytes show an aberrant phenotype (CD3‐CD4+),
and sometimes associated with monoclonality of T cells. Clinical features. Presentation may be with an asympto-
Therefore, if there is no end‐organ damage and no under- matic eosinophilia in as many as 10% of cases, but angi-
lying cause is identified, ‘eosinophilia of undetermined oedema or muscle pains was the presenting feature in 14%
significance’ is the preferred diagnosis. If there is end‐ of the National Institutes of Health (NIH) series, whereas
organ damage, ‘idiopathic hyper‐eosinophilic syndrome’ a rash or fever was the main problem in another 12% [26].
is a diagnosis of exclusion. Tiredness, excessive sweating, cough, breathlessness and
The most recent diagnostic criteria for idiopathic HES retinal lesions are other common presentations.
are as follows [4]: The most common cutaneous features are urticarial and
1 Peripheral blood eosinophilia more than 1.5 × 109/L on include erythematous, pruritic maculopapules and
at least two occasions 1 month apart, and/or tissue nodules [27], angioedema [28,29] or urticarial plaques
hypereosinophilia. (Fig. 24.4). Immediate‐pressure urticaria has been observed
2 Organ damage and/or dysfunction attributable to tis- in some patients. Dermographism may be marked but is
sue hypereosinophilia. Symptoms and signs of end‐ not correlated with other features of the disease.
organ involvement with eosinophil tissue infiltration/ Eczematous changes and erythroderma can be seen,
injury of the skin and principally the heart (40–70%) but there are no features distinguishing them from other
Chapter 24  Hypereosinophilic Disorders 325

(a) (b)

SECTION 4: OTHER TYPES


Fig. 24.4  Idiopathic hypereosinophilic syndrome extensive papular eruption with perifollicular accentuation affecting the torso and arms with postinflam-
matory hyperpigmentation.

OF DERMATITIS
(a) (b)
Fig. 24.5  Haematoxylin–eosin low and high power. Nonspecific chronic dermatitis with tissue eosinophilia and pigmentary incontinence. Source: Merika
et al. [22]. Reproduced with permission of John Wiley & Sons.

types of eczema. However, some case reports have indi- youngest child was 5, and three other patients were aged
cated that eosinophilic cellulitis [20,30], cutaneous 18 or under.
necrotizing eosinophilic vasculitis [31], Raynaud phe-
nomenon [31,32], superficial thrombophlebitis, cutaneous Differential diagnosis. Eosinophilia may be due to parasitic
ulceration as a result of dermal arteriole thrombosis [33], infection, tumours and hypersensitivity. These some-
and digital gangrene [34,35] may also occur as cutaneous times only become apparent at a later date.
features of HES. A recent case report suggested that juve- Investigations to exclude clonality and lymphoprolif-
nile temporal arteritis of unknown cause may also be a erative disorders are also required as HES is a diagno-
manifestation of HES. Juvenile temporal arteritis is char- sis of exclusion.
acterized by an asymptomatic nodule in the temporal A child with an eosinophilia of >1500/mm3 on at least
artery area in young adults. Histologically, the lesion is two occasions 1 month apart and/or persisting for
characterized by a significant intimal thickening with 6  months and/or with tissue eosinophilia should be
moderate eosinophilic infiltrates, constriction or occlu- investigated (Fig. 24.6).
sion of the vascular lumen and absence of giant cells [32]
(Fig. 24.5). Secondary (reactive causes)
In a review of the lymphoid variant of HES in the These patients have an underlying inflammatory, neo-
French population [36], 81% had skin manifestations, plastic or other condition causing hypereosinophilia.
with associated lymphadenopathy (62%), rheumatologi- Nevertheless an underlying haematopoietic neoplasm
cal (29%), gastrointestinal (24%), pulmonary (19%), neu- producing clonal eosinophils must be excluded by histo-
rological (10%) and cardiovascular (5%) involvement. The pathological, cytogenetic and molecular analysis.
326 Section 4  Other Types of Dermatitis

Screen for reactive causes


Atopic or allergic disease Neoplasia
Total and specific lgEs Immunoglobulins
as appropriate Protein electrophoresis
Blood film
Other infections
Syphilis serology Parasitosis
TB Elispot/mantoux Stool OCP
Borrelia serology Strongyloides
EBV Medications
EBV lgG/lgM and PCR

If negative
SECTION 4: OTHER TYPES

Screen for: Screen for:


FIP1L1-PDGFRA by FISH or RT-PCR Abnormal T-cell immunophenotype
OF DERMATITIS

Bone marrow aspirate and biopsy with and/or in vitro Th2 cytokine production Fig. 24.6  Algorithm for investigating a child with
cytogenetic analysis (lymphocyte variant) hypereosinophilia of >1500/mm3 on at least two
Reciprocal translocations Other clonal or molecular abnormality, occasions 1 month apart. EBV, Epstein–Barr virus;
clonal eosinophils and,or increased FISH, fluorescence in situ hybridization; Ig,
4q12(PDGFRA), 5q31-q33 (PDGFRB)
marrow blasts (0.5–20%) in chronic immunoglobulin; OCP, ova, cysts and parasites;
or 8p11-12 (FGFR1) in myeloid and
eo leukaemia or WHO defined myeloid PCR, polymerase chain reaction; RT‐PCR, reverse
lymphoid neoplasms
neoplasm with associated eosinophilia transcription polymerase chain reaction; TB,
tuberculosis.

Reactive causes include: lymphomyeloproliferative disease. One study has identi-


• Common atopic allergic diseases. fied immunoglobulin heavy chain rearrangement as a fre-
• Parasites/helminth infections. quent molecular feature of paediatric HES and an
• Epstein–Barr virus (EBV) infection: EBV has recently association with B‐cell clonal expansion and subsequent
been implicated as a cause of lymphocyte‐mediated HE B‐cell malignancies in children [38].
(see Section 8).
• Drug‐induced eosinophilia (allergic or toxic).
• Other infectious agents: syphilis, tuberculosis, coccidi- Primary (clonal) eosinophilia (HES‐N)
oidomycosis, Borrelia burgdorferi, allergic bronchopul- Haematopoietic neoplasms producing HE are usually,
monary aspergillosis and even neuroborreliosis [37]. but not always, associated with a molecular or cytoge-
• Chronic graft‐versus‐host disease. netic marker (as proof of clonality).
• Indolent systemic mastocytosis. • Myeloid, lymphoid and haematopoietic stem cell
• Chronic inflammatory disorders such as irritable bowel neoplasms:
disease. • PDGFRA‐rearranged neoplasms (4q12 translocation)
• Autoimmune diseases. • PDGFRB‐rearranged neoplasms (5q31–q33 translocation)
• Hypocortisolism – salivary cortisol is a useful measure • FGFR1 (8p11–12 translocation)
but clues may be present with hyperkalaemia, • Others: JAK2 fusion gene, FLT3 fusion gene
hyponatraemia, hypoglycaemia, hypercalcaemia and a without chromosomal or gene defect
leucocytosis (due to eosinophilia, lymphocytosis and with a nonspecific chromosomal/gene defect.
sometimes accompanied by neutropenia). In some • Eosinophilic leukaemia
cases hypothyroidism and hyperprolactinaemia may • WHO‐defined myeloid neoplasm with HE:
occur. BCR/ABL1+ chronic myeloid leukaemia
• Neoplasia: JAK2 V617F+ myeloproliferative neoplasms
Hodgkin disease KIT D816V+ systemic mastocytosis
B‐ or T‐cell lymphoma/leukaemia CBFB fusion gene‐related acute myeloid leukaemia
solid tumours/malignancy Myelodysplastic syndrome with HE
Langerhans cell histiocytosis. Other WHO‐defined myeloid neoplasms with HE.
In certain haematopoietic neoplasms, such as Hodgkin All patients in whom a neoplastic HES is being consid-
lymphoma, T‐cell lymphoma and B‐lymphoblastic leu- ered should receive histopathological, cytogenetic and
kaemia/lymphoma (with certain molecular defects, such molecular analysis by fluorescence in situ hybridization
as the translocation t(5;14)(q31;q32) that induces hypere- (FISH) or reverse transcription polymerase chain reaction
osinophilia), HE may be reactive. (RT‐PCR), bone marrow aspirate and biopsy with cytoge-
Early identification of HES in the paediatric population netic analysis and evaluation for reciprocal translocations
is of paramount importance as it may evolve towards a or other gene defects.
Chapter 24  Hypereosinophilic Disorders 327

If the above investigations are negative, proceed to When treatment is necessary, then glucocorticosteroids
screening for the lymphoid variant of HES (L‐HES). This are the first‐line treatment [41] although the skin manifes-
includes abnormal T‐cell immunophenotype and/or in tations may be responsive to montelukast and ketotifen.
vitro Th2 cytokine production. This results in IL‐5 over- The response to corticosteroids is variable, but a rapid
production, which is key in eosinophil activation. The response to prednisolone, especially with a rapid reduc-
expansion of the abnormal T‐cell subset is usually asymp- tion in peripheral blood eosinophilia, is a good prognostic
tomatic but a few cases evolve to a T‐cell lymphoma. indicator, along with a high IgE level and a history of
Importantly, Lefevre et al. observed that nearly half of the angioedema. In adults, the usual starting dose is 1 mg/kg
patients were atopic before CD3‐CD4+ L‐HES diagnosis prednisolone with tapering to the lowest dose maintain-
[36]; their report highlights the fact that L‐HES should be ing remission. For isolated cutaneous involvement lower
considered in atopic patients who have HE. Changes in starting doses can be used (e.g. 0.5 mg/kg).
skin lesions, appearance of new symptoms (i.e. gastroin- Although in general patients with HES do not respond
testinal, rheumatological, pulmonary symptoms) and/or to the tyrosine kinase (TK) inhibitor imatinib mesylate at
appearance of marked HE in atopic patients should lead doses used in chronic eosinophilic leukaemia (CEL), there
to consideration of HES, and notably L‐HES. The majority have been almost 50 adult case reports of its successful
of these patients have a persistent CD3‐CD4+ T‐cell sub- use in FIP1L1–PDGFR α‐negative HES. Adults have also

SECTION 4: OTHER TYPES


set on blood T‐cell phenotype. Seventy six percent had a been treated with other TK inhibitors, and dasatinib, nilo-
clonal TCRγδ gene rearrangement [36]. Two other immu- tinib, sorafenib and etoposide proved helpful when other

OF DERMATITIS
nophenotypes are more frequent: CD3+CD4+CD7‐ and treatments, including imatinib, had failed.
CD3+CD4+CD8‐TCRαβ+. It is, however, notable that imatinib mesylate has been
Progression to T‐cell lymphoma is rare but L‐HES may associated with a variety of adverse cutaneous reactions,
evolve to: including urticaria, maculopapular exanthem, acute gen-
• Hodgkin lymphoma. eralized exanthematous pustulosis (AGEP), exfoliative
• Cutaneous T‐cell lymphoma. dermatitis and Stevens–Johnson syndrome.
• Angioimmunoblastic T‐cell lymphoma. In patients with steroid resistance, or if a steroid‐sparing
• Mediastinal T‐cell anaplastic lymphoma kinase (ALK) agent is required (e.g. if requiring more than 8 mg predni-
negative systemic anaplastic large‐cell lymphoma has solone/day maintenance), options include hydroxycarba-
been reported in a 2.5‐year‐old boy with HES [39]. mide [42–45], ciclosporin [36,46,47], interferon‐α [42] and
• B‐lymphoblastic lymphoma. Overproduction of IL‐3 mepolizumab [48], dapsone, sodium cromoglicate,
may also be an important factor in the induction of suplatast tosilate [49], cyclophosphamide, vincristine
eosinophilia. Bomken et al, describe a child with eosin- [50–53], L‐asparaginase [52] and 6‐mercaptopurine [51].
ophilia and Loeffler endocarditis who subsequently Infliximab or the humanized monoclonal antibodies to
developed a B‐lymphoblastic lymphoma and was IL‐5 and CD52 (alemtuzumab, reslizumab) have recently
found to have an IL3/IgH@ fusion in the cutaneous lym- shown efficacy as steroid‐sparing agents in HES
phoma cells [40]. [36,54–56].
Immunoglobulin heavy chain (IGH) clonality can In newly diagnosed symptomatic children with evi-
precede a B‐cell lymphoma in children with eosino- dence of acute life‐threatening complications involving
philia. Rapanotti et  al. reported five children who vital organs, corticosteroids are the recommended first‐
showed IGH clonality at presentation: of these, two line option followed by vincristine [57]. In adults with
developed a B‐cell non‐Hodgkin lymphoma and a severe disease or refractory to all other treatments, both
B‐lineage acute lymphocytic leukaemia at 6 and bone marrow transplantation and stem cell transplanta-
12 months respectively, two spontaneously reverted tion have been successful.
to a polyclonal IGH profile during the follow‐up,
and the last one persisted with HES without B‐clonal References
evolution after 19 months. One patient had a 1 Valent P, Gleich GJ, Reiter A. Pathogenesis and classification of
PDGFRA/FIP1L1 fusion [38]. eosinophil disorders: a review of recent developments in the field.
Expert Rev Hematol 2012;5:157–76.
• Carcinoembryonic antigen was noted in one case with- 2 Valent P, Horny HP, Bochner BS. Controversies and open questions
out a linked malignancy and the levels returned to nor- in the definitions and classification of the hypereosinophilic syn-
mal with corticosteroid treatment. dromes and eosinophilic leukemias. Semin Hematol 2012;49:171–81.
3 Valent P, Klion AD, Horny HP. Contemporary consensus proposal on
criteria and classification of eosinophilic disorders and related syn-
Treatment. Treatment of HES is generally aimed at long‐ dromes. J Allergy Clin Immunol 2012;130:607–12.e9.
term reduction in blood and tissue levels of eosinophils to 4 Klion AD, Mejia R, Cowen EW et al. Chronic active Epstein‐Barr virus
avoid end‐organ damage. Treatment should be guided by infection: a novel cause of lymphocytic variant hypereosinophilic syn-
drome. Blood 2013;121:2364–6.
the urgency of lowering the eosinophil level depending 5 Olson TA, Virmani R, Ansinelli RA et  al. Cardiomyopathy in a child
on the degree of end‐organ damage, preventing the devel- with hypereosinophilic syndrome. Pediatr Cardiol 1982;3:161–9.
opment of complications and balancing the advantages 6 Aoki Y, Nata M, Hashiyada M, Sagisaka K. Sudden unexpected death
and disadvantages of treatment. in childhood due to eosinophilic myocarditis. Int J Legal Med
1996;108:221–4.
Screening for occult cardiac disease every 6 months 7 Kocaturk H, Yilmaz M. Idiopathic hypereosinophilic syndrome associ-
with electrocardiography and echocardiography is highly ated with multiple intracardiac thrombi. Echocardiography 2005;
recommended. 22:675–6.
328 Section 4  Other Types of Dermatitis

8 Chusid MJ, Dale DC, West BC, Wolff SM. The hypereosinophilic 36 Lefevre G, Copin MC, Staumont‐Sallé D et al. The lymphoid variant
syndrome: analysis of fourteen cases with review of the literature. of hypereosinophilic syndrome: study of 21 patients with CD3‐CD4+
Medicine (Baltimore) 1975;54:1–27. aberrant T‐cell phenotype. Medicine (Baltimore) 2014;93:255–66.
9 Pena Segura JL, Adrados I, Jiménez Bustos JM et  al. [Eosinophilic 37 Buda P, Zawadka K, Wadowska‐Kłopotek W et al. Cerebrospinal fluid
myositis in a 9 year old boy]. Rev Neurol 2001;33:960–3. eosinophilia in a child with neuroborreliosis. Wiad Lek 2015;68:9224.
10 de Carpi JM, Rives S, Prada F, Varea V. Eosinophilic ascites as the first 38 Rapanotti MC, Caruso R, Ammatuna E et al. Molecular characteriza-
sign of idiopathic hypereosinophilic syndrome in childhood. J Clin tion of paediatric idiopathic hypereosinophilia. Br J Haematol 2010;
Gastroenterol 2007;41:864–5. 151:440–6.
11 Cay A, Imamoglu M, Cobanoglu U. Eosinophilic pancreatitis mimick- 39 Tekkesin F, Pınarlı FG, Kaya Z et al. High‐risk ALK negative anaplas-
ing pancreatic neoplasia. Can J Gastroenterol 2006;20:361–4. tic large‐cell lymphoma presenting with hypereosinophilic syndrome
12 Katz HT, Haque SJ, Hsieh FH. Pediatric hypereosinophilic syndrome in a 2.5‐year‐old child. Pediatr Hematol Oncol 2012;29:686–90.
(HES) differs from adult HES. J Pediatr 2005;146:134–6. 40 Bomken S, Haigh S, Bown N et al. Cutaneous B‐lymphoblastic lym-
13 Falade AG, Darbyshire PJ, Raafat F, Booth IW. Hypereosinophilic syn- phoma with IL3/IgH translocation presenting with hypereosino-
drome in childhood appearing as inflammatory bowel disease. philia and acute endocarditis. Pediatr Blood Cancer, 2015;62:1055–7.
J Pediatr Gastroenterol Nutr 1991;12:276–9. 41 Rizzari C, Conter V, D’Angelo P et al. Efficacy of prolonged low‐dose
14 Spiegel R, Zalman L, Gavriel H, Horovitz Y. Hypereosinophilic syn- steroid treatment in a child with idiopathic hypereosinophilic syn-
drome in a child presenting as eosinophilic pharyngitis. J Pediatr drome: a case report. Pediatr Hematol Oncol 1995;12:209–12.
Hematol Oncol 2003;25:747–9. 42 Butterfield JH. Treatment of hypereosinophilic syndromes with pred-
15 Noureen N, Rana MT. Hypereosinophilic syndrome causing acute nisone, hydroxyurea, and interferon. Immunol Allergy Clin North
hemiplegia in childhood. J Coll Physicians Surg Pak 2008;18:384–5. Am 2007;27:493–518.
16 Sakuta R, Tomita Y, Ohashi M et al. Idiopathic hypereosinophilic syn- 43 Srinivasan A, Lavanya R, Sankar J. Steroid‐unresponsive hypereosin-
SECTION 4: OTHER TYPES

drome complicated by central sinovenous thrombosis. Brain Dev ophilic syndrome. Ann Trop Paediatr 2011;31:273–7.
2007;29:182–4. 44 Kanthila J, Bhaskaranand N. Idiopathic hypereosinophilic syndrome in
OF DERMATITIS

17 Schulman H, Hertzog L, Zirkin H, Hertzanu Y. Cerebral sinovenous children: 3 cases with review of literature. Indian J Pediatr 2013;80:124–7.
thrombosis in the idiopathic hypereosinophilic syndrome in child- 45 Parrillo JE, Fauci AS, Wolff SM. Therapy of the hypereosinophilic
hood. Pediatr Radiol 1999;29:595–7. syndrome. Ann Intern Med 1978;89:167–72.
18 Kumar KA, Anjaneyulu A, Murthy JM. Idiopathic hypereosinophilic syn- 46 Nadarajah S, Krafchik B, Roifman C, Horgan‐Bell C. Treatment of
drome presenting as childhood hemiplegia. Postgrad Med J 1992;68:831–3. hypereosinophilic syndrome in a child using cyclosporine: implica-
19 Nakahara C, Kamoda T, Kinugasa H et  al. Simultaneous onset of tion for a primary T‐cell abnormality. Pediatrics 1997;99:630–3.
nephrotic syndrome and insulin‐dependent diabetes mellitus in a 47 Hosoki K, Nagao M, Iguchi K et al. An 8‐year‐old boy with hypereo-
case with hypereosinophilia syndrome. Clin Nephrol 2000;53:312–14. sinophilic syndrome. Int Arch Allergy Immunol 2011;155:117–22.
20 Nofal R. [Eosinophilic cystitis as symptom of an idiopathic hypereo- 48 Mehr S, Rego S, Kakakios A et  al., Treatment of a case of pediatric
sinophilic syndrome]. Aktuelle Urol 2007;38:148–51. hypereosinophilic syndrome with anti‐interleukin‐5. J Pediatr 2009;
21 Alfaham MA, Ferguson SD, Sihra B, Davies J. The idiopathic hypere- 155:289–91.
osinophilic syndrome. Arch Dis Child 1987;62:601–13. 49 Yamada Y, Yoshihara S, Arisaka O. Successful treatment of pediatric
22 Merika EE, Lefroy D, Milojkovic D, Wakelin SH. Hypereosinophilic hypereosinophilic syndrome with suplatast tosilate. Ann Allergy
syndrome: an indolent rash with a serious cardiac complication. Asthma Immunol 2007;99:380–1.
Clin Exp Dermatol 2016;41:170–4. 50 Van Slyck EJ, Adamson TC 3rd. Acute hypereosinophilic syndrome.
23 Kleinfeldt T, Nienaber CA, Kische S et al. Cardiac manifestation of Successful treatment with vincristine, cytarabine, and prednisone.
the hypereosinophilic syndrome: new insights. Clin Res Cardiol JAMA 1979;242:175–6.
2010;99:419–27. 51 Marshall GM, White L. Effective therapy for a severe case of the idio-
24 Ogbogu PU, Rosing DR, Horne MK 3rd. Cardiovascular manifesta- pathic hypereosinophilic syndrome. Am J Pediatr Hematol Oncol
tions of hypereosinophilic syndromes. Immunol Allergy Clin 1989;11:178–83.
North Am 2007;27:457–75. 52 Chilcote RR, Pergament E, Kretschmer R, Mikuta JC. The hypereosin-
25 Hussein MR. Mucocutaneous Splendore‐Hoeppli phenomenon. ophilic syndrome and lymphoblastic leukemia with extra C‐group
J Cutan Pathol 2008;35:979–88. chromosome and q14+ marker. J Pediatr 1982;101:57–60.
26 Fauci AS, Harley JB, Roberts WC et al. NIH conference. The idio- 53 Sakamoto K, Erdreich‐Epstein A, deClerck Y, Coates T. Prolonged
pathic hypereosinophilic syndrome. Clinical, pathophysiologic, clinical response to vincristine treatment in two patients with idio-
and therapeutic considerations. Ann Intern Med 1982;97:78–92. pathic hypereosinophilic syndrome. Am J Pediatr Hematol Oncol
27 Kazmierowski JA, Chusid MJ, Parrillo JE et al. Dermatologic mani- 1992;14:348–51.
festations of the hypereosinophilic syndrome. Arch Dermatol 54 Yamada Y, Rothenberg ME, Lee AW et al. The FIP1L1‐PDGFRA fusion
1978;114:531–5. gene cooperates with IL‐5 to induce murine hypereosinophilic syn-
28 Armstrong JL, Lantz AB, Jerath RS, Meyer CF. Urticaria, angioedema, drome (HES)/chronic eosinophilic leukemia (CEL)‐like disease.
and an elevated eosinophil count in an adolescent. Ann Allergy Blood 2006;107:4071–9.
Asthma Immunol 2001;87:457–60. 55 Quintas‐Cardama A, Cortes J. Therapeutic options for patients with
29 Garcia Bravo P, Martín Mateos MA, Giner MT et al. [Recurrent angi- clonal and idiopathic hypereosinophia. Expert Opin Investig Drugs
oedema and hypereosinophilia]. Allergol Immunopathol (Madr) 2008;17:1039–50.
2005;33:169–71. 56 Legrand F, Klion AD. Biologic therapies targeting eosinophils: cur-
30 Bogenrieder T, Griese DP, Schiffner R et al. Wells’ syndrome associ- rent status and future prospects. J Allergy Clin Immunol Pract
ated with idiopathic hypereosinophilic syndrome. Br J Dermatol 2015;3:167–74.
1997;137:978–82. 57 Cogan E, Roufosse F Clinical management of the hypereosinophilic
31 Jang KA, Lim YS, Choi JH et al. Hypereosinophilic syndrome present- syndromes. Expert Rev Hematol 2012;5:275–89.
ing as cutaneous necrotizing eosinophilic vasculitis and Raynaud’s
phenomenon complicated by digital gangrene. Br J Dermatol
2000;143:641–4.
Eosinophilic cellulitis (Wells syndrome)
32 Ito K, Hara H, Okada T, Terui T. Hypereosinophilic syndrome with
various skin lesions and juvenile temporal arteritis. Clin Exp Dermatol Brief history/introduction. This syndrome was first
2009;34:e192–5. described by Wells [1] in 1971 as ‘a changing symptom
33 Oppliger R, Gay‐Crosier F, Dayer E, Hauser C. Digital necrosis in a
patient with hypereosinophilic syndrome in the absence of cutaneous
complex with a phase of the disease characterized by
eosinophilic vasculitis. Br J Dermatol 2001;144:1087–90. ­episodes of acute eosinophilic cellulitis followed by granu-
34 Funahashi S, Masaki I, Furuyama T. Hypereosinophilic syndrome lomatous dermatitis with its distinctive histopathology’.
accompanying gangrene of the toes with peripheral arterial occlu-
sion—a case report. Angiology 2006;57:231–4.
Epidemiology and pathogenesis. This is a rare condition,
35 Hamada T, Kimura Y, Hayashi S et al. Hypereosinophilic syndrome
with peripheral circulatory insufficiency and cutaneous micro- with some 100 cases reported in the literature, and fewer
thrombi. Arch Dermatol 2007;143:812–13. than 50 paediatric cases [2–9]. Both sexes are equally
Chapter 24  Hypereosinophilic Disorders 329

affected. Two cases of congenital eosinophilic cellulitis


have been documented [10,11] and familial eosinophilic
cellulitis associated with short stature and mental retar-
dation has been recorded [12].
It is postulated that different triggers, such as insect
[6,8,13] and spider [13] bites, viral infections (particularly
mumps, molluscum contagiosum and varicella) [14], bac-
terial, fungal and parasitic (onchocerciasis) infection,
rheumatoid arthritis and Raynaud disease, as well as
drug eruptions (in particular antibiotics [2], cholesterol‐
lowering drugs, local anaesthetics) and vaccinations
[15,16], surgery and cryosurgery [17] may induce a hyper-
sensitivity reaction with eosinophil‐induced cytotoxicity
of collagen and histiocytic phagocytosis as a response to
the damage. A recent study reported high prevalence of Fig. 24.7  A child with Wells syndrome. Note the urticaria‐like plaques
atopy in affected children serving as a predisposing envi- resembling cellulitis with some blistering.

SECTION 4: OTHER TYPES


ronment for an eosinophil‐rich inflammatory response to
these triggers [18]. In adults, there is a recognized associa-

OF DERMATITIS
tion with haematological malignancy, and there have Milder cases have multiple annular or circinate erythe-
been three case reports of eosinophilic cellulitis associated matous plaques with infiltrated borders persisting and
with non‐haematological malignancy: a 42‐year‐old recurring over a few years. Erythematous papulonodular
patient with a squamous cell carcinoma, a 79‐year‐old lesions with a faint punctate erythema at another skin site
woman with a squamous cell carcinoma of the bronchus, were noted in one patient and, in another, scaling (mycol-
a 58‐year‐old with renal cell carcinoma and a 17‐year‐old ogy negative) of the feet was associated with scaly, itchy
girl with nasopharyngeal carcinoma [19,20]. Histological annular lesions resembling granuloma annulare, which
features of Wells syndrome within a mastocytoma have showed the histology of eosinophilic cellulitis.
been reported [21].
IL‐5 and eosinophil cationic protein (ECP) have been Differential diagnosis. Annular erythema of infancy is a
implicated in the pathogenesis. Levels of IL‐5 and ECP rare entity first described by Peterson and Jarratt in 1981
are elevated in blister fluid and the elevation in the blood [24]. Kunz et  al. [25] demonstrated a predominantly
and marrow of patients with eosinophilic cellulitis corre- perivascular eosinophilic infiltrate but no peripheral
lates with disease activity [12,22]. Furthermore, Plotz eosinophilia. Clinically the lesions start as nonpruritic
et al. [23] reported evidence of spontaneous overproduc- erythematous papules and progress within weeks to
tion of IL‐5 by lymphocytes (predominantly of the gyrate and annular erythematous, often urticated, lesions.
CD3+CD4+ T‐cell phenotype) in a patient with eosino- The condition shows a relapsing course and may remit for
philic cellulitis. several months. Spontaneous resolution occurs; response
to systemic steroids, antimalarial therapy [26], dapsone,
Clinical features. Patients are generally well but may be nicotinamide, indometacin and UVB [27] has been
febrile, and occasionally there is general malaise and reported.
arthralgia. Presentation can be abrupt, especially in Peripheral blood eosinophilia is a feature of eosino-
severe cases with single or multiple lesions that itch or philic cellulitis, but is not usually sufficiently severe or
cause a tender burning sensation. Lesions spread rap- persistent to cause confusion with HES. Furthermore, the
idly and evolve over a few weeks, and often recur over lack of multisystemic involvement and the presence of
many months. Lesions begin as red plaques or, less flame figures (which are not seen in the cutaneous lesions
commonly, nodules that clinically resemble urticaria of HES, although perivascular eosinophil and monocyte
and cellulitis, often with some blistering, and are haem- infiltration can be marked) help to differentiate these
orrhagic in some cases (Fig. 24.7). As the lesions extend conditions.
rapidly, they often demonstrate blue central discoloura- Serological tests for Toxocara should be carried out to
tion and a peripheral red rim, and are sometimes pain- exclude this infection. Heiner and Kevy [28] described a
ful at the outset. The skin is not generally hot to the child with itchy papular erythema due to Toxocara canis
touch. Lesions undergo gradual resolution over about larva migrans infection and the biopsy showed dermal
4–8 weeks and often develop a greenish hue and firm infiltration of eosinophils and histiocytes with areas of
induration before resolving. Generally there is no scar- ‘fibrinoid degeneration of collagen’. Rook and Staughton
ring, although postinflammatory hyperpigmentation is [29] similarly observed perivascular and fat infiltration by
common, but atrophic alopecia was observed following eosinophils and histiocytes in an adult patient with
scalp involvement in an 11‐year‐old boy [3]. Any site toxocariasis and infiltrated skin lesions. The resolving
can be involved. Mucosal involvement with tongue lesions may resemble morphoea. EGPA is differentiated
swelling and throat swelling was a feature in Wells’ clinically by the systemic onset, the less favourable out-
original cases. Lesions following Blaschko’s lines have come and histologically by the presence of a true vasculi-
been described. tis. Clinically, the lesions tend to be purpuric, with tender
330 Section 4  Other Types of Dermatitis
SECTION 4: OTHER TYPES

(a) (b)
OF DERMATITIS

(c)
Fig. 24.8  Histology of Wells syndrome.

subcutaneous nodules and cutaneous infarcts rather than In cases subjected to direct immunofluorescence (IMF),
urticarial/cellulitic lesions. However progression from perivascular C3, IgM and IgA have been observed.
Wells syndrome to EGPA has been reported in the litera- Dermal fibrin may also stain positively. Electron micros-
ture [20,30]. copy reveals eosinophil granule‐coating collagen fibres in
the flame figures.
Laboratory findings/histopathology. Where there is blis-
tering, it is subepidermal and crowded with eosinophils. Treatment. Often, no treatment is necessary. Cetirizine [33],
There is marked dermal oedema and infiltration with low‐dose corticosteroids [2,7,34], antimalarials [35], dap-
eosinophils and phagocytic histiocytes together with scat- sone [36] or ciclosporin can be helpful if needed for resistant
tered ‘flame figures’ in the mid to deep dermis, which cases. Steroid‐recalcitrant bullous eosinophilic cellulitis in a
consist of a core of collagen and eosinophilic debris sur- child was successfully treated with dapsone [37].
rounded by eosinophils, palisading histiocytes and, in
established lesions, foreign body‐type giant cells
(Fig.  24.8). The collagen appears to be displaced rather References
than destroyed. 1 Wells GC. Recurrent granulomatous dermatitis with eosinophilia.
Extracellular eosinophil granules may be seen free in Trans St Johns Hosp Dermatol Soc 1971;57:46–56.
2 Wells GC, Smith NP. Eosinophilic cellulitis. Br J Dermatol
the dermis, within phagocytes and in the flame figures. 1979;100:101–9.
ECP [31] and major basic protein (MBP) are abundant 3 Nielsen T, Schmidt H, Sogaard H. Eosinophilic cellulitis. (Well’s
within the eosinophils and in the flame figures [32]. syndrome) in a child. Arch Dermatol 1981;117:427–9.
4 Saulsbury FT, Cooper PH, Bracikowski A, Kennaugh JM. Eosinophilic
Eosinophils and other cells can be seen transiting through cellulitis in a child. J Pediatr 1983;102:26629.
oedematous small venules, but there is no structural 5 Fisher GB, Greer KE, Cooper PH. Eosinophilic cellulitis (Wells’ syn-
vasculitis. drome). Int J Dermatol 1985;24:101–7.
Chapter 24  Hypereosinophilic Disorders 331

6 Wood C, Miller AC, Jacobs A et al. Eosinophilic infiltration with flame 35 Dereure O, Guilhou JJ. [Eosinophilic‐like erythema: a clinical subset
figures. A distinctive tissue reaction seen in Wells’ syndrome and of Wells’ eosinophilic cellulitis responding to antimalarial drugs?].
other diseases. Am J Dermatopathol 1986;8:186–93. Ann Dermatol Venereol 2002;129:720–3.
7 Lindskov R, Illum N, Weismann K, Thomsen OF. Eosinophilic celluli- 36 Consigny S, Courville P, Young P et  al. [Histological and clinical
tis: five cases. Acta Derm Venereol 1988;68:325–30. forms of the eosinophilic cellulitis]. Ann Dermatol Venereol 2001;
8 Anderson CR, Jenkins D, Tron V, Prendiville JS. Wells’ syndrome in 128:213–16.
childhood: case report and review of the literature. J Am Acad 37 Moon SH, Shin MK. Bullous eosinophilic cellulitis in a child
Dermatol 1995;33:857–64. treated with dapsone. Pediatr Dermatol 2013;30:e46–7.
9 Van der Straaten S, Wojciechowski M, Salgado R et  al. Eosinophilic
cellulitis or Wells’ syndrome in a 6‐year‐old child. Eur J Pediatr
2006;165:197–8. Eosinophilic fasciitis
10 Garty BZ, Feinmesser M, David M et al. Congenital Wells syndrome.
Pediatr Dermatol 1997;14:312–15.
Brief introduction/history. Also known as Schulman syn-
11 Kuwahara RT, Randall MB, Eisner MG. Eosinophilic cellulitis in a
newborn. Pediatr Dermatol 2001;18:89–90. drome [1], eosinophilic fasciitis (EF) is a rare connective
12 Davis MD, Brown AC, Blackston RD et al. Familial eosinophilic cel- tissue disorder characterized clinically by symmetrical,
lulitis, dysmorphic habitus, and mental retardation. J Am Acad progressive induration then sclerodermatous skin
Dermatol 1998;38:919–28.
13 Schorr WF, Tauscheck AL, Dickson KB, Melski JW. Eosinophilic
changes primarily affecting the extremities and histologi-
­cellulitis (Wells’ syndrome): histologic and clinical features in arthropod cally by thickening of the fascia with a chronic inflamma-
bite reactions. J Am Acad Dermatol 1984;11:1043–9. tory, eosinophil‐rich infiltrate.

SECTION 4: OTHER TYPES


14 Hamamoto Y, Ichimiya M, Yoshikawa Y, Muto M. Eosinophilic cellu-
litis associated with molluscum contagiosum. Br J Dermatol 2004;
Epidemiology and  pathogenesis. Eosinophilic fasciitis

OF DERMATITIS
151:1279–81.
15 Koh KJ, Warren L, Moore L et al. Wells’ syndrome following thiomer- appears to be a distinct entity, although it may be an early
sal‐containing vaccinations. Australas J Dermatol 2003;44:199–202. phase of systemic sclerosis or a variant of linear scleroderma.
16 Simpson JK, Patalay R, Francis N, Roberts N. Influenza vaccination
as a novel trigger of Wells syndrome in a child. Pediatr Dermatol
Its aetiology is unknown but infectious and immunological
2015;32:e171–2. theories have been proposed. Cases have been described in
17 Stavropoulos PG, Kostakis PG, Panagiotopoulos AK et al. Molluscum which the Borrelia spirochaete was isolated from skin and
contagiosum and cryosurgery: triggering factors for Wells’ syn- fascia but the prevalence is low compared to infection rates
drome? Acta Derm Venereol 2003;83:380–1.
18 Brun J, Chiaverini C, Bessis D et al. [Wells Syndrome in children so that a genetic predisposition to fascial inflammation as a
and atopy: Retrospective study of 11 cases and review of the litera- reaction has been postulated. One adult case report sug-
ture]. Ann Dermatol Venereol 2015;142:320–31. gested that Mycoplasma arginine may have a pathogenic role.
19 Farrar CW, Guerin DM, Wilson NJ. Eosinophilic cellulitis associated
with squamous cell carcinoma of the bronchus. Br J Dermatol
Eosinophilic fasciitis can occur in association with con-
2001;145:678–9. nective tissue disorders, particularly Sjögren syndrome
20 Rajpara A, Liolios A, Fraga G, Blackmon J. Recurrent paraneoplastic and Hashimoto thyroiditis but also primary biliary cir-
wells syndrome in a patient with metastatic renal cell cancer. Dermatol rhosis, vitiligo, lupus, morphoea, systemic sclerosis and
Online J 2014;20.
21 Hunt SJ, Santa Cruz DJ. Eosinophilic cellulitis: histologic features in a lichen sclerosus. Underlying malignancy may exist, par-
cutaneous mastocytoma. Dermatologica 1991;182:132–4. ticularly haematological malignancies, multiple mye-
22 Espana A, Sanz ML, Sola J, Gil P. Wells’ syndrome (eosinophilic loma, Hodgkin disease and, rarely, T‐cell lymphoma [2].
cellulitis): correlation between clinical activity, eosinophil levels,
In adults, paraneoplastic eosinophilic fasciitis has also
eosinophil cation protein and interleukin‐5. Br J Dermatol 1999;
140:127–30. been noted in a patient with metastatic skeletal melanoma
23 Plotz SG, Abeck D, Behrendt H et  al. [Eosinophilic cellulitis (Wells from a uveal primary melanoma and with colorectal,
syndrome)]. Hautarzt 2000;51:182–6. breast and prostatic carcinoma. Recently, an association
24 Peterson AO Jr, Jarratt M. Annular erythema of infancy. Arch
Dermatol 1981;117:145–8.
was suggested with a toxic thyroid adenoma. A Japanese
25 Kunz M, Hamm K, Bröcker EB, Hamm H. [Annular erythema in case was reported where eosinophilic fasciitis preceded
childhood—a new eosinophilic dermatosis]. Hautarzt 1998;49:131–4. multiple myeloma by 3 years.
26 Kahofer P, Grabmaier E, Aberer E. Treatment of eosinophilic annular In other cases, haematological abnormalities have been
erythema with chloroquine. Acta Derm Venereol 2000;80:70–1.
27 Thomas L, Fatah S, Nagarajan S, Natarajan S. Eosinophilic annular noted, including congenital asplenia, aplastic anaemia,
erythema: successful response to ultraviolet B therapy. Clin Exp haemolytic anaemia and thrombocytopenia.
Dermatol 2015;40:883–6. Preceding trauma, insect bites, strenuous exertion or
28 Heiner DC, Kevy SV. Visceral larva migrans; report of the syndrome
in three siblings. N Engl J Med 1956;254:629–36.
exposure to trichloroethylene have been implicated in
29 Rook A, Staughton R. The cutaneous manifestations of toxocariasis. some cases.
Dermatologica 1972;144:129–43. Drugs may induce the disease, including lipid‐lowering
30 Ratzinger G, Zankl J, Eisendle K, Zelger B. Eosinophilic leukocyto- agents, ramipril, antibiotics, nonsteroidal anti‐inflammatory
clastic vasculitis ‐ a spectrum ranging from Wells’ syndrome to
Churg‐Strauss syndrome? Eur J Dermatol 2014;24:603–10. drugs, antineoplastic medicines (including the PD‐1
31 Selvaag E, Roald B. Eosinophil activation in Wells’ syndrome demon- inhibitor pembrolizumab for metastatic melanoma), pheny-
strated immunohistochemically with antibodies against eosinophilic toin, carbidopa, natalizumab (a selective adhesion molecule
cationic protein. Acta Derm Venereol 2000;80:63–4.
inhibitor used for multiple sclerosis), vaccines and over‐the‐
32 Peters MS, Schroeter AL, Gleich GJ. Immunofluorescence identifi-
cation of eosinophil granule major basic protein in the flame fig- counter essential amino acids such as L‐tryptophan (and its
ures of Wells’ syndrome. Br J Dermatol 1983;109:141–8. metabolite quinolinic acid).
33 Aroni K, Aivaliotis M, Liossi A, Davaris P. Eosinophilic cellulitis in a child Case reports have emerged of eosinophilic fasciitis
successfully treated with cetirizine. Acta Derm Venereol 1999;79:332.
34 Gilliam AE, Bruckner AL, Howard RM et  al. Bullous “cellulitis”
occurring as a rare complication of haemodialysis, radio-
with eosinophilia: case report and review of Wells’ syndrome in therapy, stem cell transplantation and allogeneic bone
childhood. Pediatrics 2005;116:e149–55. marrow transplantation.
332 Section 4  Other Types of Dermatitis

Familial cases have emerged [3,4] and a purported role Bone marrow biopsy may reveal plasmocytosis and
for HLA‐A2 suggested. eosinophilia and, rarely, aplastic anaemia and thrombocy-
There is no apparent racial predisposition. There is a topenia occur.
female preponderance in children. Over 100 cases have A deep biopsy including muscle fascia is necessary for
now been reported, the youngest patient being only diagnosis. Magnetic resonance imaging and fluorodeoxy-
1 year old [2,5–16]. glucose positron emission tomography have proved use-
ful both in diagnosis and in monitoring progress of the
Clinical features. Painful swelling of the distal part of the disease [19,20].
limbs, progressing to symmetrical indurated lesions that On histology, there is dermal sclerosis with inflamma-
limit movement although the hands and feet are usually tion and fibrosis of the fat and deep fascia. The fascia
spared, is the usual presentation. Occasionally the face, becomes thickened, with collagen hypertrophy and infil-
abdomen and hands and feet are affected [3]. There may tration by lymphocytes, plasma cells, histiocytes and
be superficial blistering and haemorrhage. Peau d’orange eosinophils. Lack of new collagen deposition in early
surface changes and erythema are seen and the ‘groove’ lesions distinguishes this from scleroderma.
sign with depression of the veins is a useful clue [16]. Immunofluorescence demonstrates IgG and C3 in
Rarely, pitting oedema symmetrically affecting the arms the deep fascia. The serum of these patients contains a
SECTION 4: OTHER TYPES

as well as the legs can be the first sign. serum factor that suppresses in vitro myeloid and
Fever, myalgia and weight gain may be a feature. erythrocyte precursors and factors with eosinophil
OF DERMATITIS

Raynaud phenomenon and nailfold capillary defects are chemotactic activity [21]. Serum levels of the tissue
not usually a feature, although Quintero‐Del‐Rio et  al. inhibitor of metalloproteinase 1 (TIMP‐1) are elevated
[17] argued that Raynaud phenomenon and hepatosple- and positive staining for TIMP‐1 is observed in the fas-
nomegaly can occur in the spectrum of childhood eosino- cia of affected patients. Furthermore, serum TIMP‐1
philic fasciitis. Myalgia and arthralgia may be experienced levels in patients with eosinophilic fasciitis were sig-
but polyarthritis is rarely associated. Established cases nificantly correlated with serum γ‐globulin and IgG
may have joint contractures [10]. Development of aplastic levels, suggesting that TIMP‐1 is involved in the patho-
anaemia is reported more commonly than by chance and genesis, and that it may be a useful marker for disease
isolated chronic severe neutropenia unresponsive to activity as well as serum γ‐globulin or IgG levels [22].
granulocyte colony‐stimulating factor has occurred. Pro‐inflammatory and fibrogenic cytokine responses
Internal organs are not usually involved although peri- including IL‐5, CD‐40 ligand, transforming growth fac-
carditis, pulmonary fibrosis and reduced pulmonary dif- tor β1 and interferon‐γ are postulated to result in
fusion capacity, primary biliary cirrhosis, oesophageal inflammatory cell infiltration that dysregulates pro-
hypokinesia, eosinophilic colitis and intestinal pseudo‐ duction of extracellular matrix proteins and increases
obstruction, nephritis [13], myositis and haemorrhagic collagen production by fibroblasts which ultimately
stroke with cerebral vasculitis have been reported in asso- leads to progressive fibrosis of affected tissues [23].
ciation with eosinophilic fasciitis.
Treatment. There is no generally accepted effective treat-
Differential diagnosis. Early disease can mimic angi- ment. Many patients will respond to systemic steroids
oedema [3]. The differential diagnosis of more established (prednisolone or pulsed methylprednisolone) [5,10,12,15].
disease includes morphoea profunda, disabling panscle- Spontaneous remission can occur so that more aggressive
rotic morphoea of children, amyotrophic lateral sclerosis, treatments should only be used in recalcitrant cases.
sclerodermatous graft‐versus‐host disease, nephrogenic Individual patients have responded to antihistamines,
systemic fibrosis (nephrogenic fibrosing dermopathy), including cetirizine, ketotifen, hydroxyzine [14], cimeti-
scleromyxoedema, scleroedema, fasciitis panniculitis dine, isotretinoin, chloroquine and hydroxychloroquine,
syndrome, eosinophilia myalgia syndrome and cutane- sulfasalazine, dapsone, d‐penicillamine, cyclophospha-
ous amyloidosis. mide, methotrexate [3,5], ciclosporin, tacrolimus, azathio-
prine, mycophenolate mofetil [24], colchicine, infliximab
Laboratory and  histology findings. Peripheral blood [3,6] and rituximab. Phototherapy (bath PUVA and
eosinophilia, up to 30%, is observed in the majority (65– UVA‐1) and extracorporeal photochemotherapy can be
80% of cases) but is transient and thus not necessary for beneficial.
diagnosis. The sedimentation rate is raised and there is In adults, Mendoza et al. [25] concluded that the early
hypergammaglobulinaemia. Serum aldolase may be ele- introduction of antifibrotic treatment offered better out-
vated, but creatinine phosphokinase is normal. comes following a prospective but non‐randomized trial
Cytoplasmic antineutrophil cytoplasmic antibodies of d‐penicillamine in conjunction with corticosteroids.
(c‐ANCA) and anti‐nuclear and thyroid antibodies may Residual fibrosis is more common in childhood‐
be positive. Hypercalcaemia for which no other cause was onset eosinophilic fasciitis, particularly where there
found, but which resolved with treatment of the eosino- are morphoea‐like skin changes and truncal
philic fasciitis, has been reported in one adolescent [18]. involvement [9,26].
Chapter 24  Hypereosinophilic Disorders 333

References
1 Schulman H, Hertzog L, Zirkin H, Hertzanu Y. Cerebral sinovenous
Eosinophilic panniculitis
thrombosis in the idiopathic hypereosinophilic syndrome in child-
hood. Pediatr Radiol 1999;29:595–7. Brief introduction. There is some controversy as to
2 Doyle JA, Connolly SM, Hoagland HC. Hematologic disease in scle- whether eosinophilic panniculitis is a distinct entity or
roderma syndromes. Acta Derm Venereol 1985;65:521–5.
3 Poliak N, Orange JS, Pawel BR, Weiss PF. Eosinophilic fasciitis mim-
represents a reaction pattern to an underlying systemic
icking angioedema and treatment response to infliximab in a pediat- condition. The original patient reported by Burket and
ric patient. Ann Allergy Asthma Immunol 2011;106:444–5. Burket [1] presented with asymptomatic subcutaneous
4 Sullivan C, Coughlan R. Eosinophilic fasciitis in siblings. J Rheumatol swellings that histologically showed eosinophilic lobular
2013;40:105.
5 Ortega‐Loayza AG, Merritt BG, Groben PA, Morrell DS. Eosinophilic and septal panniculitis with flame figures. The lesions
fasciitis in a female child. J Am Acad Dermatol 2008;58:S72–4. underwent spontaneous resolution within days.
6 Tzaribachev N, Holzer U, Schedel J et al. Infliximab effective in ster- Subsequent cases have had a similar presentation, includ-
oid‐dependent juvenile eosinophilic fasciitis. Rheumatology (Oxford)
ing a 6‐year‐old boy reported by Samlaska et  al. [2] in
2008;47:930–2.
7 Antic M, Lautenschlager S, Itin PH. Eosinophilic fasciitis 30 years whom other causes of eosinophils in the skin and subcutis
after ‐ what do we really know? Report of 11 patients and review of were excluded.
the literature. Dermatology 2006;213:93–101.
8 Pillen S, van Engelen B, van den Hoogen F et al. Eosinophilic fasciitis in
a child mimicking a myopathy. Neuromuscul Disord 2006;16:144–8. Epidemiology and pathogenesis. The exact pathogenesis

SECTION 4: OTHER TYPES


9 Farrington ML, Haas JE, Nazar‐Stewart V, Mellins ED. Eosinophilic remains unclear but the entity is thought to be associated
fasciitis in children frequently progresses to scleroderma‐like cuta- with the atopic diathesis, heightened allergic reactivity or

OF DERMATITIS
neous fibrosis. J Rheumatol 1993;20:128–32.
infection of the gastrointestinal tract. It has also been
10 Huppke P, Wilken B, Brockmann K et al. Eosinophilic fasciitis leading
to painless contractures. Eur J Pediatr 2002;161:528–30. reported in advanced HIV disease. An antigenic trigger in
11 Huemer M, Seeber A, Huemer C. Scleroderma‐like syndrome in a the setting of underlying immunodeficiency may trigger
child: eosinophilic fasciitis or scleredema adultorum? Eur J Pediatr an altered immune response with excess IL‐4 and IL‐5
2000;159:520–2.
12 Balat A, Akinci A, Turgut M et al. Eosinophilic fasciitis—progression
production and eosinophil drive. It is seen more fre-
to linear scleroderma: a case report. Turk J Pediatr 1999;41:381–5. quently in females (3 : 1) and is more common in the third
13 Kirschstein M, Helmchen U, Jensen R et al. Kidney involvement in a and sixth decade. Two patients aged 6 are thought to be
17‐year‐old boy with eosinophilic fasciitis. Clin Nephrol 1999; the youngest reported cases: a boy with scalp nodules [2],
52:183–7.
14 Uckun A, Sipahi T, Akgün D, Oksai A. Eosinophilic fasciitis success- the other a girl with an insidiously enlarging swelling
fully treated with oral hydroxyzine: a new therapeutic use of an old over the back of the left knee [3].
drug? Eur J Pediatr 2002;161:118–19.
15 Britt WJ, Duray PH, Dahl MV, Goltz RW. Diffuse fasciitis with
eosinophilia: a steroid‐responsive variant of scleroderma. J Pediatr
Clinical features and differential diagnosis. Eosinophilic
1980;97:432–4. panniculitis most often presents as subcutaneous nodules
16 Pinal‐Fernandez I, Callejas‐Moraga EL, Roade‐Tato ML, Simeon‐ on the legs, arms, trunk and face. Urticarial papules, pur-
Aznar CP. Groove sign in eosinophilic fasciitis. Lancet 2014;15:1774. ple plaques or even purpura, pustules and ulcerative
17 Quintero‐Del‐Rio AI, Punaro M, Pascual V. Faces of eosinophilic
fasciitis in childhood. J Clin Rheumatol 2002;8:99–103. lesions have been reported. There will invariably be a
18 Rutter MM, Prahalad S, Passo M, Backeljauw PF. Idiopathic hypercal- nodular subcutaneous component.
cemia and eosinophilic fasciitis: a novel association. J Pediatr There is a wide differential diagnosis, including B‐ and
Endocrinol Metab 2004;17:1251–4.
19 Kirchgesner T, Dallaudière B, Omoumi P et al., Eosinophilic fasciitis:
T‐cell lymphoma, arthropod bites, gnathostomiasis (nod-
typical abnormalities, variants and differential diagnosis of fasciae ular migratory eosinophilic panniculitis due to foodborne
abnormalities using MR imaging. Diagn Interv Imaging 2015; parasitic zoonosis, usually from contaminated raw fish),
96:341–8. zygomycosis (e.g. Basidiobolus ranarum [4]), streptococcal
20 Marie I, Sauvetre G. Fluorodeoxyglucose positron emission tomogra-
phy in eosinophilic fasciitis. Joint Bone Spine 2014;81:541. and other bacterial infections, toxocariasis, Fasciola hepat-
21 Wasserman SI, Seibold JR, Medsger TA Jr, Rodnan GP. Serum eosino- ica infection, leucocytoclastic vasculitis, polyarteritis
philotactic activity in eosinophilic fasciitis. Arthritis Rheum nodosa, erythema nodosum, atopic and contact dermati-
1982;25:1352–6.
22 Jinnin M, Ihn H, Yamane K et al. Serum levels of tissue inhibitor of
tis, eosinophilic cellulitis, angiolymphoid hyperplasia
metalloproteinase‐1 and 2 in patients with eosinophilic fasciitis. Br J with eosinophilia, Ofuji disease, granuloma faciale,
Dermatol 2004;151:407–12. EGPA, Langerhans cell histiocytosis [5], HES, drug erup-
23 Dziadzio M, Chee R, McNamara C et  al. EBV‐driven diffuse tions, injection granuloma [6] and refractory anaemia
large B‐cell lymphoma confined to the liver in a patient with a
history of idiopathic CD4 lymphocytopenia. BMJ Case Rep 2013; with blasts.
21:2013.
24 Loupasakis K, Derk CT. Eosinophilic fasciitis in a pediatric patient. Histology findings. The condition is characterized by
J Clin Rheumatol 2010;16:129–31.
prominent eosinophil infiltration of subcutaneous fat
25 Mendoza FA, Bai R, Kebede AG, Jimenez SA. Severe eosinophilic
fasciitis: comparison of treatment with d‐penicillamine plus corti- septa and lobules with occasional fat necrosis and flame
costeroids vs. corticosteroids alone. Scand J Rheumatol 2016; figures but absence of vasculitis. Changes may extend up
45:129–34. to the reticular tissue of fascia. Although eosinophils are
26 Endo Y, Tamura A, Matsushima Y et  al. Eosinophilic fasciitis:
report of two cases and a systematic review of the literature deal-
seen in the panniculitis of both lupus and morphoea pro-
ing with clinical variables that predict outcome. Clin Rheumatol funda, they were found in the minority of cases (24% and
2007;26:1445–51. 25% respectively) in the series of Peters and Su [7].
334 Section 4  Other Types of Dermatitis

4 Mahamaytakit N, Singalavanija S, Limpongsanurak W. Subcutaneous


Treatment and  prevention. Most patients respond to zygomycosis in children: 2 case reports. J Med Assoc Thai 2014;97:
prednisolone or supersaturated potassium iodide but the S248–53.
5 Ahmed H, Rafindadi AH, Pindiga UH, Opara WE. Suppurative granu-
lesions will also resolve spontaneously. Success with dap-
lomatous eosinophilic panniculitis: case report. East Afr Med J 1999;76:
sone and ciclosporin has also been reported. 709–12.
6 Gomez Rodriguez N, Ortiz‐Rey JA, de la Fuente Buceta A, Ibáñez
References Ruán J. [Auto‐induced eosinophilic panniculitis: a diagnostic
1 Burket JM, Burket BJ. Eosinophilic panniculitis. J Am Acad Dermatol dilemma]. An Med Interna 2001;18:635–7.
1985;12:161–4. 7 Peters MS, Su WP. Eosinophils in lupus panniculitis and morphea
2 Samlaska CP, de Lorimier AJ, Heldman LS. Eosinophilic panniculi- profunda. J Cutan Pathol 1991;18:189–92.
tis. Pediatr Dermatol 1995;12:35–8.
3 Jain S, Jain P, Jakhar P, Shivkumar VB. Eosinophilic panniculitis in a
female child: An unusual presentation. Indian Dermatol Online J
2015;6:34–6.
SECTION 4: OTHER TYPES
OF DERMATITIS
 335

C HA PTER   25

Juvenile Plantar Dermatosis


John C. Browning1,2 & Margaret Brown3
1
 Baylor College of Medicine, Houston, TX, USA
2
 Children’s Hospital of San Antonio, San Antonio, TX, USA
3
 Division of Dermatology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA

Abstract The differential diagnosis of JPD includes atopic dermatitis, a ­ llergic


contact dermatitis, tinea pedis and psoriasis. The diagnosis is
clinical, but patch testing may be helpful in identifying relevant
Juvenile plantar dermatosis (JPD) is a chronic condition affecting
­allergens if contact sensitivity is suspected. Biopsy is rarely, if ever,
children and young adolescents and characterized by symmetri-
indicated for diagnosis but often shows characteristic inflammation
cal, shiny erythema, superficial desquamation and fissuring of the

SECTION 4: OTHER TYPES


of the acrosyringium or eccrine duct. Treatment and prevention are
weightbearing surfaces of the feet. Moisture, friction and the tran-
aimed at moisture control via the use of absorbent cotton socks and
sition from ‘wet to dry’ probably play a role in its development,
breathable footwear or sandals. Topical barrier creams, emollients,

OF DERMATITIS
but the exact aetiology and pathophysiology remain unknown.
antiperspirants and topical steroids may also be effective.

Key points • The diagnosis of JPD is clinical; the differential diagnosis


includes atopic dermatitis, allergic contact dermatitis, tinea
pedis and psoriasis.
• Juvenile plantar dermatosis (JPD) affects children and young
• Patch testing may be helpful in identifying relevant allergens if
adolescents.
a sensitivity is suspected.
• JPD is characterized by symmetrical, erythematous, shiny skin on
• Biopsy is rarely, if ever, indicated for diagnosis but often shows
the weightbearing surfaces of the feet, especially at the great
characteristic inflammation of the acrosyringium or eccrine duct.
toe and forefoot, sparing the interdigital web spaces.
• Treatment and prevention are aimed at moisture control and
• Moisture, friction and the transition from ‘wet to dry’ probably
include the use of absorbent cotton socks, breathable footwear,
play a role in the development of JPD.
sandals, topical barrier creams, emollients, antiperspirants or
topical steroids for acute flares.

Brief introduction/history. Juvenile plantar dermatosis Epidemiology and pathogenesis. The prevalence of JPD
(JPD) is a chronic condition affecting children and is not known, although it may be quite common [5]. JPD
young adolescents and characterized by symmetrical, occurs in children aged 3–15 and is more common in boys
shiny erythema, superficial desquamation and fissuring aged 4–8 [6]. In one South Indian study of 200 patients
of the weightbearing surfaces of the feet. with various types of lower leg and foot eczemas, juvenile
JPD was first recognized as a distinct entity in 1976 plantar dermatosis was the most common type of lower
when Mackie and Husain described a group of school‐ leg and foot eczema in patients younger than 15 years of
aged children with scaling, fissuring and itching or age. In the same study, JPD was the most common type of
burning of the weightbearing areas of the feet [1]. Prior lower leg and foot eczema in students, presumably due to
to that, ‘atopic winter feet’ had been described by Moller their regular use of shoes and socks [7]. JPD has also been
in 1972 [2] and ‘recurrent juvenile eczema’ was also described in adults [8,9]. In a study of 64 patients, JPD
reported by Schultz and Zachairiae in 1972 [3]. Before was found more commonly in Chinese than Malays or
then, JPD was probably thought to be eczema, contact Indians [8]. It seems that some individuals have a family
dermatitis or tinea pedis. In the medical literature, JPD predisposition to developing JPD, as it has been described
has been described under a variety of names including in twins [10]. A personal or family history of atopy is com-
forefoot eczema, juvenile plantar dermatitis, forefoot mon in affected children, and this may implicate an
dermatitis, dermatitis plantaris sicca, peridigital derma- impaired skin barrier in its development.
titis, wet and dry syndrome, sneaker feet and sweating Occlusive footwear, moisture and friction play an
sock dermatitis [4]. important role in the development of JPD [11]. It has been

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
336 Section 4  Other Types of Dermatitis

hypothesized that transition from ‘wet to dry’ is probably


the culprit in precipitating the condition [12,13]. Occlusion,
especially by shoes with rubber soles or synthetic socks,
can lead to perspiration. Many affected individuals also
complain of plantar hyperhidrosis. It has been posited that
persistent, occluded moisture disrupts the epidermal bar-
rier which leads to maceration and permits the excessive
evaporation of subcutaneous moisture. This results in des-
iccation, shrinkage and separation of corneocytes which is
clinically manifested as fissuring [5]. JPD often becomes
clinically evident during the summer months, which is
thought to be related to perspiration leading to aggrava-
tion of the condition [14]. However, worsening of the con-
Fig. 25.1  Erythematous, shiny skin with superficial peeling on the
dition during the winter months has also been reported, weightbearing surfaces of the forefoot.
perhaps due to the use of warmer shoes and thicker socks,
and one study found no seasonal variation [15,16].
SECTION 4: OTHER TYPES

Clinical features and  differential diagnosis. JPD is


OF DERMATITIS

c­ haracterized by symmetrical, erythematous, shiny skin


on the weightbearing surfaces of the feet, especially at
the great toe and forefoot [17] (Figs  25.1 and 25.2). Less
commonly, it may extend to the dorsal foot as well. There
is notable sparing of the interdigital web spaces and
arches. Some children experience discomfort with peeling,
redness and fissuring [15]. Pruritus is less common and is
noted to be a variable associated symptom [4]. JPD is typi-
cally seen in school‐aged children and often recurs over
many years. JPD is self‐limiting and usually resolves by
12–16 years of age [6,12].
The differential diagnosis of JPD includes tinea pedis,
atopic dermatitis, allergic contact dermatitis (ACD) and
psoriasis. Tinea pedis may be considered in the differen-
tial diagnosis but it would be unusual without interdigi-
tal involvement. JPD may be misdiagnosed as atopic
dermatitis of the foot, given its prevalence in children [5].
ACD may also be considered but it is typically character-
ized by more intense pruritus, along with involvement of
the dorsal foot and lichenification in longstanding
cases  [18]. Psoriasis may also affect the feet but would
probably be accompanied by thick, silvery scale and Fig. 25.2  Erythema with a glazed appearance on the plantar foot with
fissuring which is accentuated at the great toe.
often involves the non‐weightbearing areas of the plantar
foot.
Interestingly, JPD has not been seen as frequently in the impaired epidermal barrier from JPD. Of note, rubber
paediatric dermatology clinic in recent years, according to components were found to be the principal allergens in
one set of authors who suggest a decreasing frequency of one study of ACD of the feet, followed by chromated
the condition [15]. It may also be that JPD is now more leather and adhesives [20].
commonly recognized by paediatricians and other primary Biopsy is rarely, if ever, indicated but may help guide
care providers and not referred as often to dermatologists. the clinical diagnosis [21]. Histologically, JPD is character-
ized by subacute or chronic spongiotic dermatitis with
Laboratory/histology findings. JPD is a clinical diagno- inflammation at the junctions of the acrosyringium or
sis. Mycological studies and a potassium hydroxide prep- eccrine sweat duct at the epidermis starting edge [16,22].
aration can help rule out tinea pedis if it is clinically Eccrine duct involvement has not been described in other
suspected. Patch testing should be considered to investi- forms of eczema and may be a clue to the diagnosis of
gate an allergic component, particularly in recalcitrant JPD, but its absence does not rule out the diagnosis [22].
cases [19]. In contrast to earlier studies that did not iden-
tify a significant correlation between ACD and JPD, a Treatment and  prevention. The use of cotton socks,
2012 study showed a high prevalence of contact allergy in breathable footwear and sandals has been recommended
children with sole dermatoses [1,19]. While this finding to decrease the perspiration and friction which can serve
could represent cases of ACD that are clinically similar to as aggravating factors in JPD. Lubrication with petrola-
JPD, it could also suggest sensitization secondary to an tum or use of dimethicone barrier creams has been
Chapter 25  Juvenile Plantar Dermatosis 337

s­ uggested for acute flares and maintenance [6]. Mid‐ to 6 Kalia S, Adams SP. Dermacase. Juvenile plantar dermatosis. Can Fam
Physician 2005;51:1203, 1213.
high‐potency topical steroids may also be used in cases
7 Chougule A, Thappa DM. Patterns of lower leg and foot eczema in
where pruritus is a significant factor, but recurrence is south India. Indian J Dermatol Venereol Leprol 2008;74:458–61.
common after discontinuation and their use may pre- 8 Moorthy TT, Rajan VS. Juvenile plantar dermatosis in Singapore. Int J
cipitate secondary tinea pedis [5]. Tacrolimus ointment Dermatol 1984;23:476–9.
9 Czarnecki DB, Cowen PS, Connors TJ. Juvenile plantar dermatosis in
0.1% has been reported to be beneficial, when used in an adult. Br J Dermatol 1981;104:599–600.
conjunction with an emollient [15]. Topical antiperspi- 10 Stankler L. Juvenile plantar dermatosis in identical twins. Br J

rants such as aluminium chloride can be helpful if Dermatol 1978;99:585.
hyperhidrosis is a significant associated symptom. 11 Lachapelle JM, Tennstedt D. Juvenile plantar dermatosis: a report of
80 cases. Am J Ind Med 1985;8:291–5.
Avoidance of topical allergens is advisable. 12 Jones SK, English JS, Forsyth A et al. Juvenile plantar dermatosis: an
8‐year follow up of 102 patients. Clin Exp Dermatol 1987;12:5–7.
­Acknowledgements 13 Steck WD. Juvenile plantar dermatosis: the ‘wet and dry foot syn-
drome’. Cleve Clin Q 1983;50:145–9.
The authors would like to acknowledge Dr Alanna Bree 14 Kint A, Hecke EV, Leys G. Dermatitis plantaris sicca. Dermatologica
for her authorship in the Third Edition of this chapter 1982;165:500–1.
(Harper’s Textbook of Pediatric Dermatology, 3rd edition: 15 Shipley DR, Kennedy CTC. Juvenile plantar dermatosis responding
to topical tacrolimus treatment. Clin Exp Dermatol 2006;31:453–4.
Chapter 43, Juvenile Plantar Dermatosis).
16 Ashton RE, Jones RR, Griffiths A. Juvenile plantar dermatosis. A clin-

SECTION 4: OTHER TYPES


icopathologic study. Arch Dermatol 1985;121:225–8.
­References 17 Neering H, van Dijk E. Juvenile plantar dermatosis. Acta Derm

OF DERMATITIS
1 Mackie RM, Husain SL. Juvenile plantar dermatosis: a new entity? Clin Venereol 1978;58:531–4.
Exp Dermatol 1976;1:253–60. 18 Weston JA, Hawkins K, Weston WL. Foot dermatitis in children.
2 Moller H. Atopic winter feet in children. Acta Derm Venereol
Pediatrics 1983;72:824–7.
1972;52:401–5. 19 Darling MI, Horn HM, McCormack SK et al. Sole dermatitis in chil-
3 Schultz H, Zachairiae H. The trafuril test in recurrent juvenile eczema dren: patch testing revisited. Pediatr Dermatol 2012;29:254–7.
of hand and feet. Acta Derm Venereol 1972;52:398–400. 20 Shackelford KE, Belsito DV. The etiology of allergic‐appearing foot
4 Brar KJ, Shenoi SD, Balachandran C, Mehta VR. Clinical profile of fore- dermatitis: a 5‐year retrospective study. J Am Acad Dermatol
foot eczema: a study of 42 cases. Indian J Dermatol Venereol Leprol 2002;47:715–21.
2005;71:179–81. 21 Zagne V, Fernandes NC, Cuzzi T. Histopathological aspects of juve-
5 Bikowski J. Barrier disease beyond eczema: management of juvenile nile plantar dermatosis. Am J Dermatopathol 2014;36:359–61.
plantar dermatosis. Practical Dermatology for Pediatrics 2010;July/ 22 Van Diggelen MW, van Dijk E, Hausman R. The enigma of juvenile
August:28–31. plantar dermatosis. Am J Dermatopathol 1986;8:336–40.
338 

CHA PTER  2 6

Perioral Dermatitis
Marius Rademaker1,2
1
 Dermatology Department, Waikato, District Health Board, Hamilton, New Zealand
2
 Waikato Clinical Campus, Faculty of Medical and Health Sciences, The University of Auckland, Hamilton, New Zealand

Abstract variant has been described as childhood granulomatous periorificial


dermatitis.
Several factors have been implicated in its pathogenesis, includ-
Perioral, or periorificial, dermatitis is an inflammatory skin disease
ing decreased barrier function, infections (Demodex, Fusobacte-
that affects predominantly older girls and women. In prepubescent
rium), cosmetics and topical glucocorticosteroids.
children, perioral dermatitis affects boys and girls equally. Perioral
It is generally self‐limiting, but responds well to avoidance of
dermatitis is characterized by a papulosquamous inflammatory erup-
SECTION 4: OTHER TYPES

topical preparations and/or sub‐antimicrobial doses of systemic


tion around the mouth (generally sparing the vermillion), nose and
tetracyclines/macrolides.
eyes, with an absence of flushing or comedones. A g ­ ranulomatous
OF DERMATITIS

Key points • Several factors have been implicated in its pathogenesis,


including decreased barrier function, infections (Demodex,
Fusobacterium), cosmetics and topical glucocorticosteroids.
• Perioral, or periorificial, dermatitis is an inflammatory skin
• It is generally self‐limiting, but responds well to avoidance of
disease that affects predominantly older girls and women.
topical preparations and/or sub‐antimicrobial doses of systemic
• In prepubescent children, perioral dermatitis affects boys and
tetracyclines/macrolides.
girls equally.
• A granulomatous variant has been described as childhood
• It is characterized by a papulosquamous inflammatory eruption
granulomatous periorificial dermatitis.
around the mouth (generally sparing the vermillion), nose and
eyes, with an absence of flushing or comedones.

History. The term ‘perioral dermatitis’ was first used in Epidemiology and  pathogenesis. Childhood perioral
1964 [1], although the dermatosis had been described as dermatitis is relatively uncommon, with no apparent gen-
a  ‘light sensitive seborrheid’ in a series of 92 patients, der predominance [11,12]. In a large study of 222 children,
including children from 11 years of age, several years average age of presentation was 6.6 years, 45% were
­earlier [2]. boys, 62.2% were Caucasian, and a third reported a past
In 1970, five cases of perioral dermatitis were reported history of atopic dermatitis [12]. In contrast, adult perio-
specifically in children [3]. Several years later an unusual ral dermatitis is more common, affecting predominantly
papular and acneiform facial eruption was reported in 22 young women. The granulomatous variant of perioral
black children aged 9 months to 12 years [4]. In a 12‐year dermatitis occurs mostly in children with skin phototypes
review of perioral dermatitis, 15 out of 173 cases (8.6%) 4 and 5 [4,7,9,13].
were in children [5]. In a series of 14 children aged 9 The aetiology of perioral dermatitis remains unclear; it is
months to 6 years with perioral dermatitis, most, but not probably an idiosyncratic response to a variety of exoge-
all, had been using fluorinated topical corticosteroids to nous factors (Box 26.1). Traditionally, intentional or inad-
treat pre‐existing mild facial dermatoses [6]. vertent application of topical corticosteroids [5–7,11,12,14]
Gianotti and colleagues were the first to characterize a (including inhaled and/or intranasal steroids [15–17]) has
granulomatous variant of perioral dermatitis in children, been implicated in the pathogenesis of perioral dermatitis.
both clinically and histologically [3]. Several subsequent However, in over half of patients there is no history of prior
reports have further delineated childhood granuloma- topical corticosteroid use, thus other factors have been
tous periorificial dermatitis [4,7–9], including some cases reported in association with perioral dermatitis, including
that had been previously diagnosed as sarcoidosis [7,10]. sunscreens, moisturizers, toothpastes, antiseptics, etc.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 26  Perioral Dermatitis 339

Box 26.1  Triggers of perioral dermatitis in children

Cosmetics

• Skin care ointments and creams


• Sunscreens
• Fluorinated toothpaste

Microbiological factors

• Fusobacterium spp.
• Candida spp.
• Demodex folliculorum

Physical factors

• Ultraviolet light
• Heat

SECTION 4: OTHER TYPES


• Wind

OF DERMATITIS
Pharmaceuticals

• Topical corticosteroids (fluorinated and nonfluorinated)


• Inhaled/intranasal prescription steroid sprays/solutions

Skin disease

• Barrier dysfunction/atopic dermatitis


Fig. 26.1  Perioral and periocular dermatitis. Source: Courtesy of Dr
Amanda Oakley, DermNet, New Zealand.
[18–21]. This has led one author to suggest separating
perioral dermatitis into ‘corticosteroid‐induced rosacea‐
­
like dermatitis’ (CIRD) and perioral dermatitis not
associated with topical corticosteroid use (idiopathic
­
­perioral dermatitis) [22].
Various microbial organisms have also been linked
with perioral dermatitis [23], including Fusobacterium
spp. [24,25] and Candida spp. Demodex folliculorum has
been implicated in the development of perioral dermati-
tis, but whether it plays a primary or secondary role
remains undetermined [26]. Impaired barrier dysfunction
with significantly increased transepidermal water loss
compared to controls has been reported in adults with
perioral dermatitis [20,27]. Topical corticosteroids may
alter the epidermal structure, increase transepidermal
water loss and cause vascular instability in some children
[14]. Abrupt discontinuation of corticosteroids often
results in flaring of perioral dermatitis with intense
­erythema and stinging, burning and itching. Fig. 26.2  Perioral dermatitis. Source: Courtesy of Associate Professor
David Orchard, Royal Children’s Hospital, Melbourne, Australia.

Clinical features. The term ‘perioral dermatitis’ is an inac-


curate appellation, as both perinasal and periocular In childhood granulomatous periorificial dermatitis,
lesions (Fig. 26.1) are common, occasionally even without the lesions are often discrete, small, firm, dome‐shaped,
perioral involvement [5,6,12,13,28]. Periorificial dermati- flesh‐coloured papules without perceptible erythema or
tis may be a more appropriate term. In perioral disease, scale. It occurs predominantly in children with skin pho-
the vermillion border of the lip is usually spared totypes 4 and 5, aged 9 months to 13 years of age. Diascopy
(Fig. 26.2). In most cases, small erythematous papules are of individual lesions may reveal an apple‐jelly colour.
present, superimposed on a background of erythema and Extrafacial involvement (e.g. trunk, extremities and geni-
scaling. In severe cases, pinpoint vesiculopustular lesions tals) may be extensive [30,31].
may be evident (Fig.  26.3). Rarely, other sites including
the glabella [12], chest [29], perianal skin and vulva [28] Differential diagnosis. The differential diagnosis of peri-
may be involved. Pruritus is variable and mild, but older oral dermatitis includes contact dermatitis (both irritant
patients may complain of a burning itch [1,2]. and allergic), lip licking, atopic and seborrhoeic dermatitis,
340 Section 4  Other Types of Dermatitis

Fig. 26.4  Noncaseating granuloma in a patient with granulomatous


perioral dermatitis.
SECTION 4: OTHER TYPES

Fig. 26.3  Severe perioral dermatitis. Note vesiculopustular lesions.

Table 26.1  Key therapeutic steps in treating perioral dermatitis in children


OF DERMATITIS

Demodex folliculitis [26,32] and, very rarely, diverse meta-


Step 1 ‘Zero/null’ treatment. Discontinue any potential triggers
bolic disorders such as acrodermatitis enteropathica and
(moisturizers, sunscreens, topical steroids, toothpastes,
biotin deficiency [33]. There is an overlap between perioral other cosmetic substances)
dermatitis and rosacea. Step 2 Avoid lipophilic agents
A clue to possible allergic contact dermatitis may be Step 3 If the perioral dermatitis is mild, and not responding to
involvement of the vermillion of the lips, such as is steps 1/2: metronidazole gel, twice daily, for 3–4 weeks
described to colophony (rosin) [34]. Step 4 If not controlled by step 3: topical calcineurin inhibitors, or
Taken in isolation, individual lesions of perioral derma- mild topical steroids (class VII, but only if no history of
antecedent use)
titis may be difficult to distinguish from lesions of rosa-
Step 5 Sub‐antimicrobial doses of oral antibiotics (>8 years of
cea; however, perioral dermatitis lacks the associated age: tetracyclines; <8 years: macrolides) for 6–8 weeks
telangiectasia, flushing, periorbital swelling and blephar- Step 6 In more persistent cases, consider low‐dose isotretinoin
itis of rosacea. Steroid‐induced rosacea in prepubertal (5–10 mg /day) for 3–6 months (non‐FDA‐approved)
children is nearly indistinguishable in appearance from
perioral dermatitis [35].
The differential diagnosis of childhood granulomatous inflammatory infiltrate may be mixed lymphohistiocytic.
periorificial dermatitis includes granulomatous rosacea Noncaseating granulomas are found in childhood gran-
(including lupus miliaris disseminatus faciei), acne agmi- ulomatous periorificial dermatitis (Fig.  26.4). The dis-
nata, sarcoidosis, benign cephalic histiocytosis, granulo- tinction between childhood granulomatous periorificial
sis rubra nasi and Demodex folliculitis [7,26,36,37]. dermatitis and sarcoidosis may be difficult [3,4,7].
Demodex folliculitis in particular can have marked clinical Patch testing [42], bacteriology, mycology for Candida,
overlap [26]. It most commonly occurs in patients receiv- and a search for Demodex (e.g. dermoscopy, which can
ing chemotherapy for malignancy and/or in immunode- demonstrate follicular openings containing Demodex
ficiency [32]. ‘tails’ [43]), may be helpful in the investigation of the dif-
There is overlap between childhood granulomatous peri- ferential diagnosis of perioral dermatitis.
orificial dermatitis and granulomatous rosacea. Potential
distinguishing features of rosacea include skin lesions pre- Treatment and  prevention. The response to treatment
dominantly involving the cheeks, telangiectases and facial (Table 26.1) depends on the severity, extent and duration
flushing [38]. The coexistence of ocular signs or symptoms of the disease. In severe, longstanding disease, the
of rosacea such as blepharitis can also suggest rosacea, response to therapy is one of slow improvement, with
although these are quite uncommon in children [39]. occasional relapses over many months, whereas mild dis-
ease often clears within a few weeks [11,44–49]. Children
Laboratory/histology findings. The histopathology of whose perioral dermatitis develops from a pre‐existing
perioral dermatitis is essentially indistinguishable from perinasal erythema (Fig. 26.5) tend to have a relapsing/
that of rosacea including both granulomatous and non- remitting course, particularly if moisturizers and/or sun-
granulomatous forms [40,41]. Histologically there may be screens continue to be used. Patients with steroid‐induced
mild epidermal acanthosis, focal spongiosis, hyperkera- perioral dermatitis frequently rebound (often with
tosis with parakeratosis and a mild perivascular and peri- increasing severity) after stopping topical corticosteroids.
adnexal lymphohistiocytic infiltrate centred around hair Childhood granulomatous periorificial dermatitis tends
follicles and in the upper dermis. Focal follicular rupture to resolve over a few months to several years, but may
with microabscess formation is present in some cases. The leave behind milia and pinpoint scarring [31,37].
Chapter 26  Perioral Dermatitis 341

References
1 Mihan R, Ayres S. Perioral dermatitis. Arch Dermatol 1964;89:803–5.
2 Frumess GM, Lewis HM. Light‐sensitive seborrheid. AMA Arch
Derm 1957;75:245–8.
3 Gianotti F, Ermacora E, Benelli MG, Caputo R. Particulière dermatite
périorale infantile. Observations sur 5 cas. Bull Soc Fr Dermatol Syph
1970;77:341–4.
4 Marten RH, Presbury DGC, Adamson J, Cardell BS. An unusual pap-
ular and acneiform facial eruption in the negro child. Br J Dermatol
1974;91:435–8.
5 Wilkinson DS, Kirton V, Wilkinson JD. Perioral dermatitis: a 12‐year
review. Br J Dermatol 1979;101:245–57.
6 Manders SM, Lucky AW. Perioral dermatitis in childhood. J Am Acad
Dermatol 1992;27:688–92.
7 Frieden IJ, Prose NS, Fletcher V, Turner ML. Granulomatous perioral
dermatitis in children. Arch Dermatol 1989;125:369–73.
8 Georgouras K, Kocsard E. Micropapular sarcoidal facial eruption in a
child: Gianotti‐type perioral dermatitis. Acta Dermatol Venereol
Fig. 26.5  Perinasal scaling, with erythematous micropapules, of early 1978;58:433–6.
9 Williams HC, Ashworth J, Pembroke AC, Breathnach SM.
perioral dermatitis. Source: Courtesy of Dr Anthony Yung, Waikato
FACE – facial Afro‐Caribbean childhood eruption. Clin Exp Dermatol

SECTION 4: OTHER TYPES


Hospital, New Zealand. 1990;15:163–6.
10 Gupta AK, Goldfarb MT, Rasmussen JE. Papular midfacial eruption

OF DERMATITIS
in a child. Arch Dermatol 1989;125:1704–5.
11 Hafeez ZH. Perioral dermatitis: an update. Int J Dermatol 2003;
The use of topical skin care products should be discour- 42:514–17.
12 Goel NS, Burkhart CN, Morrell DS. Pediatric periorificial dermati-
aged (‘zero’ or ‘null’ therapy), in particular lipophilic tis: clinical course and treatment outcomes in 222 patients. Pediatr
ointments. It is very important to stop any topical corti- Dermatol 2015;32:333–6.
costeroids as soon as possible, although the potency may 13 Andry P, Bodemer C, Teillac‐Hamel D et al. Granulomatous perioral
dermatitis in childhood: eight cases. Pediatr Dermatol 1995;12:76.
need to be stepped down slowly (over weeks/months) to 14 Mooney E, Rademaker M, Dailey R et al. Adverse effects of topical
avoid significant rebound. corticosteroids in paediatric eczema: Australasian consensus state-
In less severe cases, metronidazole gel applied twice ment. Australas J Dermatol 2015;56:241–51.
daily may be helpful [47,50,51]. Only in cases with a 15 Henningsen E, Bygum A. Budesonide‐induced periorificial dermatitis
presenting as chalazion and blepharitis. Pediatr Dermatol 2011;
prominent eczematous component should low‐potency 28:596–7.
topical corticosteroids (class VII only) be considered, but 16 Dubus J‐C, Marguet C, Deschildre A et al. Local side effects of inhaled
care must be taken not to perpetuate and/or exacerbate corticosteroids in asthmatic children: influence of drug, dose, age, and
device. Allergy 2001;56:944–8.
the disease [6,52].
17 Peralta L, Morais P. Perioral dermatitis  –  the role of nasal steroids.
The topical calcineurin inhibitors, tacrolimus ointment Cutan Ocul Toxicol 2012;31:160–3.
and pimecrolimus cream, have been reported as effective 18 El‐Rifaie ME. Perioral dermatitis with epithelioid cell granulomas in a
treatments for steroid‐induced cases [53–55] as they may woman: a possible new etiology. Acta Dermatol Venereol 1979;
60:359–60.
help reduce the rebound flares associated with stopping 19 Beacham BE, Kurgansky D, Gould WM. Circumoral dermatitis and
topical steroids. cheilitis caused by tartar control dentifrices. J Am Acad Dermatol
Anecdotally, other topical therapies such as clindamy- 1990;22:1029–32.
cin, erythromycin [56], azelaic acid [57], ivermectin, keto- 20 Dirschka T, Tronnier H, Fölster‐Holst R. Epithelial barrier function
and atopic diathesis in rosacea and perioral dermatitis. Br J Dermatol
conazole, praziquantel [58], sulphur and sulfacetamide 2004;150:1136–41.
[59] have been found useful [45,48], although most have 21 Abeck D, Geisenfelder B, Brandt O. Physical sunscreens with high
only been studied in adults. sun protection factor may cause perioral dermatitis in children.
J Dtsch Dermatol Ges 2009;7:701–3.
Sub‐antimicrobial doses of oral tetracyclines, continued 22 Rosso JQ. Management of papulopustular rosacea and perioral der-
for 6–12 weeks, are usually effective in treating more matitis with emphasis on iatrogenic causation or exacerbation of
recalcitrant cases [12,44–49] but should only be prescribed inflammatory facial dermatoses: use of doxycycline‐modified release
to children older than 8 years of age, according to the 40mg capsule once daily in combination with properly selected skin
care as an effective therapeutic approach. J Clin Aesthet Dermatol
World Health Organization (WHO) and to guidelines in 2011;4:20–30.
the USA; however in the UK tetracyclines are contraindi- 23 Fritsch P, Pichler E, Linser I. Perioral dermatitis. Hautarzt 1989;

cated under the age of 12 years. Oral macrolides (e.g. 40:475–9.
24 Takiwaki H, Tsuda H, Arase S, Takeichi H. Differences between intra-
erythromycin, azithromycin) may also be helpful, par-
follicular microorganism profiles in perioral and seborrhoeic dermati-
ticularly when tetracyclines are contraindicated [12]. Oral tis. Clin Exp Dermatol 2003;28:531–4.
antibiotics may be particularly helpful in preventing/ 25 Maeda A, Ishiguro N, Kawashima M. The pathogenetic role of rod‐
reducing the flare of perioral dermatitis when topical cor- shaped bacteria containing intracellular granules in the vellus hairs of
a patient with perioral dermatitis: A comparison with perioral corti-
ticosteroids are discontinued. costeroid‐induced rosacea. Australas J Dermatol 2016;57:225–8.
Clinical experience shows that very‐low‐dose isotreti- 26 Chen W, Plewig G. Human demodicosis: revisit and a proposed

noin (5–10 mg/day, decreasing to three times per week ­classification. Br J Dermatol 2014;170:1219–25.
for 6 months) is very effective in more persistent disease, 27 Dirschka T, Weber K, Tronnier H. Topical cosmetics and perioral
­dermatitis. J Dtsch Dermatol Ges 2004;2:194–9.
but it is not FDA approved and has not been subject to 28 Nguyen V, Eichenfield LF. Periorificial dermatitis in children and
formal clinical trials [60]. ­adolescents. J Am Acad Dermatol 2006;55:781–5.
342 Section 4  Other Types of Dermatitis

29 Fisher AA. Sarcoid‐like periocular dermatitis due to strong topical 46 Caputo R, Barbareschi M. Current and future treatment options for
corticosteroids: prompt response to treatment with tetracycline. Cutis perioral dermatitis. Expert Rev Dermatol 2007;2:351–5.
1987;40:95–6. 47 Laude TA, Salvemini JN. Perioral dermatitis in children. Semin Cutan
30 Hansen KK, McTigue K, Esterly NB. Multiple facial, neck, and upper Med Surg 1999;18:206–9.
trunk papules in a black child. Childhood granulomatous perioral 48 Tempark T, Shwayder TA. Perioral dermatitis: a review of the
dermatitis with involvement of the neck and upper trunk. Arch ­condition with special attention to treatment options. Am J Clin
Dermatol 1992;128:1396–7. Dermatol 2014;15:101–13.
31 Urbatsch AJ, Frieden I, Williams ML et al. Extrafacial and generalized 49 Zeba HH. Perioral dermatitis: an update. Int J Dermatol 2003;
granulomatous periorificial dermatitis. Arch Dermatol 2002;138:1354–8. 42:514–17.
32 Morrás PG, Santos SP, Imedio IL et al. Rosacea‐like demodicidosis in 50 Veien NK, Munkvad JM, Nielsen AO et al. Topical metronidazole in
an immunocompromised child. Pediatr Dermatol 2003;20:28–30. the treatment of perioral dermatitis. J Am Acad Dermatol 1991;
33 Williams ML, Packman S, Cowan MJ. Alopecia and perioral dermati- 24:258–60.
tis in biotin‐responsive multiple carboxylase deficiency. J Am Acad 51 Miller SR, Shalita AR. Topical metronidazole gel (0.75%) for the treat-
Dermatol 1983;9:97–103. ment of perioral dermatitis in children. J Am Acad Dermatol 1994;
34 Satyawan I, Oranje AP, van Joost T. Perioral dermatitis in a child due 31:847–8.
to rosin in chewing gum. Contact Dermatitis 1990;22:182–3. 52 Sneddon I. A trial of hydrocortisone butyrate in the treatment of rosa-
35 Weston WL, Morelli JG. Steroid rosacea in prepubertal children. Arch cea and perioral dermatitis. Br J Dermatol 1973;89:505–8.
Pediatr Adolesc Med 2000;154:62–4. 53 Goldman D. Tacrolimus ointment for the treatment of steroid induced
36 Misago N, Nakafusa J, Narisawa Y. Childhood granulomatous perior- rosacea: a preliminary report. J Am Acad Dermatol 2001;44:995–8.
ificial dermatitis: lupus miliaris disseminatus faciei in children? J Eur 54 Oppel T, Pavicic T, Kamann S et al. Pimecrolimus cream (1%) efficacy
Acad Dermatol Venereol 2005;19:470–3. in perioral dermatitis – results of a randomized, double‐blind, vehi-
SECTION 4: OTHER TYPES

37 Gianotti F. Cutaneous benign histiocytoses of childhood. Mod Prob cle‐controlled study in 40 patients. J Eur Acad Dermatol Venereol
Paediatr 1975;17:193–203. 2007;21:1175–80.
OF DERMATITIS

38 Drolet B, Paller AS. Childhood rosacea. Pediatr Dermatol 1992;9:22–6. 55 Rodriguez‐Martin M, Saez‐Rondriguez M, Carnerero‐Rodriguez A
39 Chamaillard M, Mortemousque B, Boralevi F et  al. Cutaneous and et  al. Treatment of perioral dermatitis with topical pimecrolimus.
ocular signs of childhood rosacea. Arch Dermatol 2008;144:167–71. J Am Acad Dermatol 2007;56:529–30.
40 In: Calonje JE, Brenn T, Lazar AJ, McKee PH (eds). McKee’s Pathology 56 Weber K, Thurmayr R, Meisinger A. A topical erythromycin prepara-
of the Skin, 4th edn. Saunders/Elsevier 2011. tion and oral tetracycline for the treatment of perioral dermatitis: a
41 Marks R, Black MM. Perioral dermatitis. A histopathological study of placebo‐controlled trial. J Dermatol Treat 1993;4:57–9.
26 cases. Br J Dermatol 1971;84:242–7. 57 Jansen T. Azelaic acid as a new treatment for perioral dermatitis:
42 Nedorost ST. Medical Pearl: the evaluation of perioral dermatitis: results from an open study. Br J Dermatol 2004;151:933–4.
use  of an extended patch test series. J Am Acad Dermatol 2007; 58 Bribeche MR, Fedotov VP, Jillella A et  al. Topical praziquantel as
56:S100–2. a  new treatment for perioral dermatitis: results of a randomized
43 Segal R, Mimouni D, Feuerman H et al. Dermoscopy as a diagnostic ­vehicle‐controlled pilot study. Clin Exp Dermatol 2014;39:448–53.
tool in demodicidosis. Int J Dermatol 2010;49:1018–23. 59 Olansky S. Old drug  –  in a new system  –  revisited. Cutis 1977;
44 Ellis CN, Stawiski MA. The treatment of perioral dermatitis, acne rosa- 19:852–4.
cea, and seborrheic dermatitis. Med Clin North Am 1982;66:819–30. 60 Smith KW. Perioral dermatitis with histopathologic features of granu-
45 Bikowski JB. Topical therapy for perioral dermatitis. Cutis lomatous rosacea: successful treatment with isotretinoin. Cutis
1983;31:678–82. 1990;46:413–5.
 343

C HA PTER   27

Psoriasis: Epidemiology
Matthias Augustin & Marc Alexander Radtke
Institute for Health Services Research in Dermatology and Nursing (IVDP), University Medical Center Hamburg‐Eppendorf (UKE), Hamburg, Germany

Epidemiology of childhood psoriasis, 343 Epidemiology of different clinical Epidemiology of comorbidity in childhood
Prevalence of childhood psoriasis, 344 presentations, 345 psoriasis, 346
Incidence of childhood psoriasis, 345 Epidemiology of nail psoriasis in
Gender distribution, 345 childhood, 346

Abstract and North America. The reported percentage of children developing


guttate psoriasis ranges from 6.4% to 44%. Although guttate pso­
riasis is mostly self‐limiting, resolving within 3–4 months of onset,
Psoriasis vulgaris can occur at any age, and prevalence in child­
the long‐term prognosis is unknown. About one third of children
hood ranges between 0.1% and 1.5%, showing a nearly linear
with guttate psoriasis develop classical plaque‐type ­disease. Nail
increase from birth to the age of 18 years. Early onset is more
involvement is reported in between 2% and 39.3% of children.
frequently associated with a positive family history and heralds
Increasing evidence suggests an association between juvenile
a graver prognosis. The median age at onset of psoriasis was

SECTION 5: PSORIASIS
psoriasis and other morbidities including hyperlipidaemia, obesity,
found to be between 7 and 10 years. Data from the UK suggest
hypertension, diabetes mellitus and rheumatoid arthritis. Disease
a prevalence of 0.55% in the age group 0–9 years and 1.37% in
patterns of the metabolic syndrome may occur independently of
the age group 10–19 years, which is consistent with data from the
patient age and disease ­duration. A high proportion of children
Netherlands, Germany and the USA. Estimates of the proportion
and adolescents will ­continue to have psoriatic disease and co­
of psoriatic children with a positive family history in any family
morbidity throughout their lives, suggesting the need for careful
member range between 4.5% and 88%. Psoriasis may represent
work‐up and specialized care and information.
4.1% of all dermatological diseases under the age of 16 in Europe

• Disease burden and quality of life impairment can be considerable


Key points and there is marked risk of cumulative life course impairment.
• Clinical pictures show large variations but a higher proportion of
• Psoriasis can occur in all age groups; prevalence increases in an guttate psoriasis in children compared to adults.
almost linear fashion from early childhood to adolescence. • Nail disease is common.
• In most children and adolescents, onset of psoriasis predicts • Children with psoriasis show significantly increased rates of comorbi­
lifelong disease. dity, including obesity, cardiovascular disease, arthritis and diabetes.

Epidemiology of childhood psoriasis leading to some uncertainty on the age of onset in child-
hood psoriasis. It is estimated that psoriasis begins in
Childhood psoriasis differs in many ways from adult‐ childhood in almost one third of the cases [1–3]. Although
onset psoriasis. It is crucial that patients and their parents it is well accepted that two incidence peaks may exist, one
understand the natural history of the disease [1] as the in early adolescence and the other in adulthood, there are
stigma associated with visible skin lesions creates a strong few data with regard to the early peak. Thirty percent of
psychological burden affecting different facets of patients’ all psoriasis patients develop first symptoms before the
everyday lives. Difficulties in developing coping strate- age of 15 years, which underlines the epidemiological
gies may lead to cumulative life course impairments, dimension in the paediatric age group [4–6]. Furthermore
especially when disease onset is in early childhood. there is some evidence that early onset is more frequently
Although psoriasis is common in childhood, data on associated with a positive family history and heralds a
epidemiology are sparse despite a growing number of graver prognosis [7]. Familial occurrence is more com-
observational studies during the last decade. Furthermore mon in childhood‐onset than in adult‐onset psoriasis
there is inconsistency in epidemiological research on [8–12]. Estimates of the proportion of psoriatic children
subtype definition (e.g. napkin psoriasis) and a notable with a positive family history in any family member
variation in how psoriasis is ascertained (secondary data, range between 4.5% and 88%. In a literature review by
physician examination, self‐reported, diagnostic codes), Burden‐Teh et al. [13], the proportion of children with a

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
344 Section 5  Psoriasis

positive history in a first‐degree relative varied from 6.2% prevalence of children with atopic dermatitis/eczema
to 54.7%. In a study by Farber et al. [14], adolescents were decreased from 17.1% at the age of 0–2 years to 7.3% at
most likely to have a positive family history compared to the  age of 14–18 years. These data were comparable
other age groups during childhood. Gene–environment with a study by De Jager et al. [2] from the Netherlands
interactions might play a role when trying to explain who found a prevalence of 0.37% in the age group from
major differences between latitudes and countries. 0 to 10 years and a prevalence of 1.09% in the age group
from 11 to 19  years old. Gelfand et  al. [18] reported a
­prevalence of 0.55% in the age group 0–9 years and 1.37%
Prevalence of childhood psoriasis in the age group 10–19 years in the UK. A study of 2194
Population‐based German statutory health insurance children in Egypt postulated that the prevalence of pso-
data from 1.3 million persons, including 33 981 patients riasis among people 18 years of age and younger was
with psoriasis, revealed a total prevalence rate of 0.71% in 0.05% [19] (Table  27.1). However, there are few studies
children younger than 18 years [15,16]. The prevalence and those that do exist reveal variations between almost
rates increased linearly from 0.2% at the age of 1 year to absence of juvenile psoriasis in Taiwan, China, and 2.15%
1.2% at the age of 18 years (Fig.  27.1). Another data set lifetime prevalence in children in Italy [26–28].
from Germany comprised 1.64 million persons nation- Psoriasis both in children and adults is supposed to
wide, including 293 181 children and young adults aged affect 2.0–3.5% of the global population [8]. Depending on
up to 18 years (Fig. 27.1). Approximately 1313 children the study population the prevalence ranges up to 8.5%
(0.45%) carried the diagnosis of psoriasis. The data con- [9]. Psoriasis may represent 4.1% of all dermatological
firmed that the prevalence in children correlates linearly diseases under the age of 16 in Europe and North America
with age and increases from 0.13% at the age of 0–2 years [10]. Taking all available data together, the average age of
SECTION 5: PSORIASIS

to 0.67% at the age of 14–18 years [17]. In contrast, the onset varies from 2.1 months to 10.6 years [13].

1.40

1.22 1.24
1.20 1.17
1.12
1.04
1.00
Prevalence (in %)

1.00

0.88
0.79
0.80
0.72

0.60 0.56 0.55 0.56

0.40 0.41
0.40
0.30
0.22 0.24 0.20
0.20
0.12

0.00
<1 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18
Age (in years)
Fig. 27.1  Prevalence of juvenile psoriasis according to age group (n = 306 020 persons). Physician‐recorded ICD‐10 codes were computed from a database
comprising 1.3 million persons. Persons identified as psoriasis patients <18 years (n = 2549) were studied. There is an almost linear increase in prevalence
rates with age.

Table 27.1  Summary of studies on the prevalence of psoriasis in children

Country Years of study Sample size (n) Estimate Female Male Age

China, Taiwan [3] 2004 4067 0.00% 6–11


China, Taiwan [20] 2005 3273 0.00% 6–11
Egypt [19] 2008–2009 2194 0.05% <18
Germany [15] 2005 306 020 0.71% 0.76% 0.66% <18
Germany [21] 2009 293 181 0.45% <18
Germany [22] 2008 16 500 1.37% 0–17
Italy [23] 2006 145 233 0.09% 0–14
Italy [23] 2012 145 233 0.20% 0–14
Sweden [24] 1975–1976 8298 0.30% 0.50% 0.10% 12–17

Source: Adapted from WHO 2016 [25]. Reproduced with permission of the World Health Organization.
Chapter 27  Psoriasis: Epidemiology 345

Among children (0–18 years), the median age at onset Morris et al. [6] reported no difference in gender distribu-
of psoriasis was found to be between 7 and 10 years tion whereas Fan et al. reported that 46.9% of the patients
[18,29–31]. It has been estimated that in 10% of psoriasis were male and 53.1% female [28]. Accordingly, in the
patients disease onset is before the age of 10 years and in larger studies, Matusiewicz et al. [21] and Augustin et al.
2% before 2 years [32], although data regarding onset [15] concluded that the prevalence of psoriasis in child-
vary among investigators. In a study on 5600 patients hood was lower in boys than in girls: the ratio was 0.35%
with psoriasis, Farber and Carlsen stated an onset before versus 0.45% in the paper by Matusiewicz and 0.42% ver-
the age of 20 years in 35% of cases  [11], whereas sus 0.48% in the study by Augustin et  al. The latter
Raychaudhuri and Gross [1] reported an onset before the revealed a relative ratio of 47.7% boys versus 52.3% girls
age of 20 years in 46.3%. Furthermore, different studies in the psoriasis population. The sex difference is most
demonstrated that 2–12% of adult patients had psoriasis obvious in children under the age of 10 years where the
before the age of 10 years [10,33]. manifestation of psoriasis is more common in girls than in
boys. In contrast, when considering all epidemiological
studies focusing on gender and age (including adults),
Incidence of childhood psoriasis the vast majority indicate a male predominance [42].
Few studies elaborated on the incidence of psoriasis in Looking at the lifetime course independent of age, psoriasis
childhood. Tollefson et al. [34] assembled a population‐ is considered almost equally prevalent in both sexes [43].
based incidence cohort of children aged <18 years first
diagnosed with psoriasis between 1970 and 1999. The
overall age‐ and sex‐adjusted annual incidence of Epidemiology of different clinical
paediatric psoriasis was 40.8 per 100 000. When psoria- presentations

SECTION 5: PSORIASIS
sis diagnosis was restricted to cases confirmed by a The clinical presentation of psoriasis in childhood can
dermatologist, the incidence was 32.2 per 100 000 with be very heterogeneous with guttate lesions, napkin
a significant increase over time from 29.6 per 100 000 in (diaper) distribution and facial involvement being more
1970 to 1974 to 62.7 per 100 000 in 1995–1997 [35]. Seyhan typical in children. In children, lesions are commonly
et  al. [29] revealed an incidence of psoriasis among a triggered by upper respiratory infections, tonsillitis,
group of dermatological patients in childhood and skin injury (Koebner phenomenon) and emotional stress.
adolescence of 3.8%. The development of plaque psoriasis in children is often
There is some evidence that the incidence and preva- preceded by guttate psoriasis [37,44]. Guttate psoriasis
lence of psoriasis in general have increased over the last (with papules less than 1 cm in diameter presenting in a
decades. Data from the USA derived from the National rash‐like distribution mainly on the trunk) is one of the
Health and Nutrition Examination Survey in 2004 indi- main manifestations in childhood. It typically presents
cated an increase in prevalence from 1.62% in 2004 to following infections caused by β‐haemolytic streptococci
3.10% in 2010 although limitations in the methodology such as tonsillitis or pharyngitis, but also subsequent to
of this study do not allow for a precise conclusion [35,36]. viral infections [45]. The reported percentage of children
In China the prevalence of psoriasis was estimated to be developing guttate psoriasis ranges from 6.4% to 44%
0.17% in 1984, whereas 25 years later another study [46]. Although guttate psoriasis is mostly self‐limiting,
found it to be 0.59% [37,38]. The prevalence in Spain in resolving within 3–4 months of onset, the long‐term
1998 was 1.43%, whereas 15 years later it was reported prognosis is unknown. There is evidence that about one
as 2.31% [39,40]. A study in Norway (Tromsø) analysed third of children with guttate psoriasis develop classical
the trend between 1979 and 2008 and revealed an plaque‐type disease [44]. Horton et al. [47] reported gut-
increased self‐reported lifetime prevalence from 4.8% to tate psoriasis to be the most common variant in 18.9% of
11.4% [41]. Tollefson et  al. identified an increase in the the cases. Fan et al. [28] revealed a proportion of 28.9%
overall annual incidence when the incidence cohort was displaying guttate psoriasis and 68.9% of the patients
restricted to dermatologist‐confirmed subjects from 25.6 displaying plaque‐type psoriasis. Kumar et  al. [30]
per 100 000 (1970–1974) to 49.4 per 100 000 (1995–1999) found classical plaque‐type psoriasis to be the most fre-
[34]. Taken together, the published studies indicate an quent type of psoriasis at the time of presentation
increase in psoriasis incidence in the past decades. (60.6%), followed by plantar psoriasis (12.8%). Data
However, due to methodological limitations, the data from China and India have revealed that in juvenile pso-
need further confirmation. riasis 28.9% of the patients exhibited guttate psoriasis
[28]. Kwon et  al. [48] analysed the clinical data of 358
patients under 18 years of age referred to a tertiary refer-
Gender distribution ral psoriasis clinic. The most prevalent phenotype was
Different studies on the gender distribution revealed plaque‐type psoriasis (67.3%) and the most frequently
inconsistent results, some of them reporting that female affected body part was the trunk (69.5%), followed by
patients are primarily affected by childhood psoriasis the legs (65.3%).
[1,28,31], while others claim similar prevalences for male Studies on the clinical presentation of childhood psori-
and female patients. Seyhan et al. [29] reported a gender asis collectively showed that the most commonly affected
distribution of 37.7% boys and 62.3% girls for 61 derma- body sites are the scalp (17.9–64.8%), limbs (9.5–90%), and
tological patients in childhood and adolescent groups. trunk (7.8–93.3%) respectively [13]. The face is involved
346 Section 5  Psoriasis

Table 27.2  Mean age at onset and clinical distribution at onset

Country Sample size (n) Ages (years) Mean age at onset (years) Clinical distribution at onset

Turkey [29] 61 <18 Girls: 9.2 Scalp (60.7%), face (18.0%), trunk (73.8%), extremities (78.7%),
Boys: 6.8 nail (21.3%)
• Plaque psoriasis: 54.1%
• Guttate psoriasis: 35.9%
• Inverse psoriasis: 6.6%
• Palmoplantar: 1.6%
• Pustular psoriasis: 13.1%
• Erythrodermic psoriasis: 3.3%
Korea [48] 358 ≤18 Childhood: 7.1 Scalp (57.8 %), face (46.3%), trunk (69.5%), arms (47.5%), legs
(0–12 years) (65.3%)
Adolescence: 12.4 • Plaque psoriasis: 52.0% (childhood); 76.0% (adolescence)
(13–18 years) • Guttate psoriasis: 27.6% (childhood); 13.5% (adolescence)
• Erythrodermic psoriasis: 1.6% (childhood); 0.9% (adolescence)
Malaysia 315 <16 7.7 Scalp (64.8%), face (19.7%), trunk (30.8%), extensor limbs
[50] (37.1%), palms and soles (5.1%), nails (7.6%)
• Plaque psoriasis: 54.3%
• Guttate psoriasis: 1.6%
• Inverse psoriasis: 1.6%
• Pustular psoriasis: 1.3%
• Palmoplantar pustulosis: 1.0%
SECTION 5: PSORIASIS

China [28] 277 <16 Median age at onset: 10 Scalp (46.5%), face (19.7%), trunk (42.9%), arms (51.4%), legs
(65.5%), knees (23.9%), palms and soles (39.2%), nail (4.4%)
• Plaque psoriasis: 68.6%
• Guttate psoriasis: 28.9%
• Pustular psoriasis: 1.1%
• Erythrodermic psoriasis: 1.4%
India [30] 419 <14 Girls: 9.3 Scalp (20.7%), face (4.7%), trunk (7.8%), arms (10%), legs
Boys: 8.1 (25%), palms (3.1%), nails (31%)
• Plaque psoriasis: 60.6%
• Guttate psoriasis: 9.7%
• Inverse psoriasis: 0.4%
• Pustular psoriasis: 0.4%
• Palmoplantar psoriasis: 5.7%
Kuwait [31] 305 ≤12 5.7 Scalp (30%), face (3.6%), trunk (10%), legs (22%), arms (9.5%),
soles (12%)
• Plaque psoriasis: 89.2%
• Guttate psoriasis: 10.5%
• Palmoplantar pustulosis: 0.3%

in 3.5–56.7 % of all cases, being a more common sign in


Epidemiology of comorbidity
children compared to adults [49] (Table 27.2).
in childhood psoriasis
Large‐scale studies show that the overall rate of comor-
Epidemiology of nail psoriasis bidity in juvenile psoriatic patients has markedly
in childhood increased. Augustin et al. revealed that the overall rate of
Nail involvement is reported in between 2% and 39.3% of comorbidity in psoriatic patients younger than 20 years
children suffering from psoriasis. De Jager et  al. [2] was two‐fold compared with persons without psoriasis
reported nail deformities in 5.3–7.4% of juvenile psoriasis (14.4% vs. 7.2%) (Table 27.3). After adjusting for age and
patients whereas Kumar et  al. [30] found nail involve- sex, patients with psoriasis have a significantly higher
ment in 31% of cases. All types of nail changes were seen risk of developing metabolic disease. Juvenile psoriasis
in these patients, pitting being the most frequent one was associated with increased rates of hyperlipidaemia,
followed by ridging and discolouration. In a survey obesity, hypertension, diabetes mellitus, rheumatoid
reported from Kuwait, nail involvement was observed in arthritis and Crohn disease [15].
36% of children, with pitting being the most common fea- In a consecutive analysis of German claims data on
ture (87%) [51]. Mercy et al. [52] found that nail psoriasis 293 181 children, prevalence data were derived from the
was more frequent in boys compared to girls whereas database of a German statutory health insurance com-
scalp psoriasis was more frequent in girls than in boys. pany (Gmünder Ersatzkasse) according to ICD‐10 codes
They explained this with the Koebner phenomenon but L40 for psoriasis and L20 for atopic eczema. Data extrac-
there are insufficient data to confirm this observation, tion involved all data sets of persons younger than
especially in children. 18 years insured in 2009. In addition, prevalence rates of
Chapter 27  Psoriasis: Epidemiology 347

Table 27.3  Prevalence of comorbidities (%) in persons in the age range 0–20 years with psoriasis (n = 2549) and without psoriasis (n = 331 758 persons)

Diagnosis ICD‐10 code(s) % without psoriasis % with psoriasis Prevalence rate


(95% confidence interval)

Crohn disease K50 0.14 0.51 3.69 (2.15–6.35)


Hyperlipidaemia E78 0.99 2.12 2.15 (1.65–2.80)
Diabetes mellitus E11, E13, E14 0.43 0.86 2.01 (1.32–3.04)
Arterial hypertension I10– I13 0.83 1.65 1.89 (1.47–2.67)
Rheumatoid arthritis M05 4.90 8.40 5.21 (1.40–19.44)
Obesity E66 4.90 8.40 1.70 (1.49–1.93)
Ischaemic heart disease I20– I25 0.49 0.75 1.52 (0.97–2.38)
Ulcerative colitis K51 0.10 0.12 1.13 (0.38–3.33)
All comorbidities (at least one) 7.20 14.40 2.00 (1.82–2.20)

comorbidity were evaluated by ICD‐10 diagnoses (1313 psoriasis [53–57]. Although children with psoriasis display
diagnosed with psoriasis and 30  354 diagnosed with a higher risk of developing diseases such as diabetes
atopic eczema). Obesity, hyperlipidaemia, arterial hyper- and hypertension, clinical studies are so far lacking to
tension and diabetes were more often diagnosed in further characterize the causal association of psoriasis
children with psoriasis in comparison to children without with comorbidity.
psoriasis and those with atopic eczema (Table 27.4) [17]. The study conducted by Augustin et al. [17] confirmed
However, these studies do not prove a causal relation- previous descriptions of specific comorbidity patterns in

SECTION 5: PSORIASIS
ship. Nevertheless, they suggest that disease patterns of juvenile psoriasis and atopic eczema [15,58]. Comorbidities
chronic inflammation associated with metabolic syndrome known to be associated with adult psoriasis are also seen
may start independently of patient age and disease dura- in children, indicating the need for screening and inter-
tion. Rheumatoid arthritis, ulcerative colitis and Crohn disciplinary management.
disease have also been suggested as being caused by Hazard ratios for psychiatric disorders have signifi-
underlying inflammatory immune mechanisms similar to cantly increased [59]. Kimball et al. compared incidences

Table 27.4  Prevalence of comorbidity (%) in persons in the age range 0–18 years with psoriasis (n = 1313), without psoriasis (n = 291 868 persons),
with atopic eczema (n = 30 354) and without atopic eczema (n = 262 827)

Diagnosis Children Children with Prevalence of Prevalence of Children with Prevalence of Prevalence of
(n) psoriasis and diagnoses in diagnoses of atopic eczema diagnoses in diagnoses of children
diagnosis psoriasis (%) children without and diagnosis atopic without atopic
of (n) psoriasis (%) of (n) eczema (%) eczema (%)

Hyperlipidaemia 1883 15 1.14 0.64 216 0.71 0.55


Obesity 10 630 93 7.08 3.61 1248 4.11 3.10
Arterial hypertension 1289 12 0.91 0.44 120 0.40 0.39
Diabetes mellitus 900 8 0.61 0.31 103 0.34 0.26
Keratitis 953 7 0.53 0.32 101 0.33 0.28
Arthritis 17 2 0.15 0.01 2 0.01 0.005
Crohn disease 213 0 0 0.07 23 0.08 0.06
Ulcerative colitis 151 2 0.15 0.05 21 0.07 0.04
Ischaemic heart 183 1 0.08 0.06 24 0.08 0.05
disease
Depression 2274 17 1.29 0.77 251 0.83 0.67
ADHD 20 950 106 8.07 7.14 2462 8.11 6.10
Vitiligo 328 3 0.23 0.11 80 0.26 0.08
Allergic rhinitis 28 505 199 15.16 9.70 5963 19.64 7.44
Chronic bronchitis 6153 29 2.21 2.10 1178 3.88 1.64
Bronchial asthma 27 484 160 12.19 9.36 5780 19.04 7.16
Contact dermatitis 5097 48 3.66 1.73 1096 3.61 1.32
Herpes infection 6217 36 2.74 2.12 1035 3.41 1.71
Urticaria 5941 30 2.28 2.03 1096 3.61 1.60
Nail disorders 2756 38 2.89 0.93 391 1.29 0.78
Alopecia areata 349 6 0.46 0.12 65 0.21 0.09
Impetigo 4338 44 3.35 1.47 1084 3.57 1.07
Viral warts 21 449 159 12.11 7.29 2963 9.76 6.10
Iridocyclitis 120 5 0.38 0.04 19 0.06 0.03
Atopic eczema 30 354 322 24.52 10.29 30,354 100 0

ADHD, attention deficit hyperactivity disorder.


348 Section 5  Psoriasis

of psychiatric disorders between paediatric patients 16 Radtke MA, Fölster‐Holst R, Beikert F et al. Juvenile psoriasis: reward-
ing endeavours in contemporary dermatology and pediatrics. G Ital
with psoriasis and control subjects. Approximately 7404
Dermatol Venereol 2011;146:31–45.
patients with juvenile psoriasis were matched 1 : 5 for age 17 Augustin M, Radtke MA, Glaeske G et al. Epidemiology and comor-
and sex to psoriasis‐free control subjects (n = 37 020). bidity in children with psoriasis and atopic eczema. Dermatology
Psoriasis proved to be an independent risk factor for 2015;231:35–40.
18 Gelfand JM, Weinstein R, Porter SB et al. Prevalence and treatment of
­psychiatric disorders, in particular anxiety and depres- psoriasis in the United Kingdom: a population‐based study. Arch
sion, and patients with juvenile psoriasis were more likely Dermatol 2005;141:1537–41.
to receive antidepressant and anxiolytic medications [59]. 19 Yamamah GA, Emam HM, Abdelhamid MF et  al. Epidemiologic
It is widely accepted that psoriasis and depression share study of dermatologic disorders among children in South Sinai,
Egypt. Int J Dermatol 2012;51:1180–5.
aetiopathogenic mechanisms [60]. These may translate 20 Chen GY, Cheng YW, Wang CY et  al. Prevalence of skin diseases
into higher levels of pro‐inflammatory cytokines such as among schoolchildren in Magong, Penghu, Taiwan: a community‐
interleukin (IL)‐6 and tumour necrosis factor (TNF)‐α based clinical survey. J Formos Med Assoc 2008;107:21–9.
21 Matusiewicz D, Koerber A, Schadendorf D et al. Childhood psoriasis:
which are shared by psoriasis and depression, within a an analysis of German health insurance data. Pediatr Dermatol
vicious circle. These cytokines modify the metabolism of 2014;31:8–13.
serotonin, norepinephrine and dopamine in the limbic 22 Kämpfe S, Augustin M, Schäfer I et al. Prevalence and health care situ-
system and basal ganglia, leading to symptoms of ation of juvenile psoriasis in Germany. Exp Dermatol 2012;21(3):e21.
23 Cantarutti A, Donà D, Visentin F et al. Epidemiology of frequently
depression. Pro‐inflammatory cytokines such as IL‐6
­ occurring skin diseases in Italian children from 2006 to 2012: a retro-
drive maturation of naive T cells to produce T‐helper spective population‐based study. Pediatr Dermatol 2015;32:668–78.
(Th)17 cells, which have a role in psoriatic lesions. 24 Larsson PA, Lidén S. Prevalence of skin diseases among adolescents
12–16 years of age. Acta Derm Venereol 1980;60:415–23.
In summary, juvenile psoriasis is a common disease in
25 World Health Organization (WHO). Global report on psoriasis. 2016.
western countries showing distinct clinical patterns dif- http://apps.who.int/iris/bitstream/10665/204417/1/9789241565189_
SECTION 5: PSORIASIS

ferent from those in adults and a markedly increased level eng.pdf (accessed 19 November 2018).
of comorbidity compared to healthy individuals. A high 26 Naldi L, Parazzini F, Gallus S; GISED Study Centres. Prevalence of
atopic dermatitis in Italian schoolchildren: factors affecting its varia-
proportion of children and adolescents will have psoriatic tion. Acta Derm Venereol 2009;89:122–5.
disease and comorbidity throughout their lives, suggest- 27 Schmitt J, Apfelbacher C. Epidemiology of pediatric psoriasis:
ing the need for careful work‐up, specific care and concise a  representative German cross‐sectional study. Exp Dermatol
information for patients, family members and healthcare 2010;19:219.
28 Fan X, Xiao FL, Yang S et  al. Childhood psoriasis: a study of 277
providers within an interdisciplinary setting. patients from China. J Eur Acad Dermatol Venereol 2007;21:762–5.
29 Seyhan M, Coskun BK, Saglam H et  al. Psoriasis in childhood and
References adolescence: evaluation of demographic and clinical features. Pediatr
1 Raychaudhuri SP, Gross J. A comparative study of pediatric onset Int 2006;48:525–30.
psoriasis with adult onset psoriasis. Pediatr Dermatol 2000;17:174–8. 30 Kumar B, Jain R, Sandhu K et al. Epidemiology of childhood psoria-
2 De Jager MEA, van de Kerkhof PCM, De Jong EM, Seyger MM. sis: a study of 419 patients from northern India. Int J Dermatol
Epidemiology and prescribed treatments in childhood psoriasis: 2004;43:654–8.
A  survey among medical professionals. J Dermatolog Treat 31 Nanda A, Kaur S, Kaur I, Kumar B. Childhood psoriasis: an epide-
2009;20:254–8. miologic survey of 112 patients. Pediatr Dermatol 1990;7:19–21.
3 Yang YC, Cheng YW, Lai CS, Chen W. Prevalence of childhood acne, 32 Farber EM, Jacobs AH. Infantile psoriasis. Am J Clin Dis Child
ephelides, warts, atopic dermatitis, psoriasis, alopecia areata and 1977;131:1266–9.
keloid in Kaohsiung County, Taiwan: a community‐based clinical 33 Henseler T, Christophers E. Psoriasis of early and late onset: charac-
survey. J Eur Acad Dermatol Venereol 2007;21: 643–9. terization of two types of psoriasis vulgaris. J Am Acad Dermatol
4 Rogers M. Childhood psoriasis. Curr Opin Pediatr 2002;14:404–9. 1985;13:450–6.
5 Fond L, Michel JL, Gentil‐Perret A et al. Psoriasis in childhood. Arch 34 Tollefson MM, Crowson CS, McEvoy MT, Kremers HM. Incidence
Pediatr 1999;6:669–74. of psoriasis in children: A population‐based study. J Am Acad
6 Morris A, Rogers M, Fischer G, Williams K. Childhood psoriasis: a Dermatol 2010;62:979–87.
clinical review of 1262 cases. Pediatr Dermatol 2001;18:188–98. 35 Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence
7 Stefanaki C, Lagogianni E, Kontochristopoulos G et  al. Psoriasis in among adults in the United States. J Am Acad Dermatol 2014;70:
children: a retrospective analysis. J Eur Acad Dermatol Venereol 512–6.
2011;25:417–21. 36 Kurd SK, Gelfand JM. The prevalence of previously diagnosed and
8 Bronckers IMGJ, Paller AS, van Geel MJ. Psoriasis in children and undiagnosed psoriasis in US adults: results from NHANES 2003–
adolescents: diagnosis, management and comorbidities. Paediatr 2004. J Am Acad Dermatol 2009;60:218–24.
Drugs 2015;17:373–84. 37 Shao CG, Zhang GW, Wang GC. Distribution of psoriasis in China: a
9 Parisi R, Symmons DP, Griffiths CE, Ashcroft DM. Identification, nationwide screening. Proc Chin Acad Med Sci Peking Union Med
management of psoriasis and associated comorbidity project. Global Coll 1987;2:59–65.
epidemiology of psoriasis: a systematic review of incidence and 38 Ding X, Wang T, Shen Y et al. Prevalence of psoriasis in China: a popu-
prevalence. J Invest Dermatol 2013;133:377–85. lation‐based study in six cities. Eur J Dermatol 2012;22:663–7.
10 Marcoux D, de Prost Y. Pediatric psoriasis revisited. J Cutan Med Surg 39 Ferrándiz C, Bordas X, García‐Patos V et al. Prevalence of psoriasis in
2002;6(3 suppl):22–8. Spain (Epiderma Project: phase I). J Eur Acad Dermatol Venereol
11 Farber EM, Carlsen RA. Psoriasis in childhood. Calif Med 1966;105: 2001;15:20–3.
415–20. 40 Ferrándiz C, Carrascosa JM, Toro M. Prevalence of psoriasis in Spain
12 Kaur I, Handa S, Kumar B. Natural history of psoriasis: a study from in the age of biologics. Actas Dermo‐Sifiliográficas 2014;105:504–9.
the Indian subcontinent. J Dermatol 1997;24:230–4. 41 Danielsen K, Olsen AO, Wilsgaard T, Furberg AS. Is the prevalence of
13 Burden‐Teh E, Thomas KS, Ratib S et al. The epidemiology of childhood psoriasis increasing? A 30–year follow‐up of a population–based
psoriasis: a scoping review. Br J Dermatol 2016;174:1242–57. cohort. Br J Dermatol 2013;168:1303–10.
14 Farber EM, Mullen RH, Jakobs AH, Nall L. Infantile psoriasis: a follow‐ 42 Hägg D, Eriksson M, Sundström A, Schmitt‐Egenolf M. The higher
up study. Pediatr Dermatol 1986;3:237–43. proportion of men with psoriasis treated with biologics may be
15 Augustin M, Glaeske G, Radtke MA et al. Epidemiology and comor- explained by more severe disease in men. PLoS ONE 2013;8:e63619.
bidity of psoriasis in children. Br J Dermatol 2010;162:633–6. 43 Boehncke W‐H, Schön MP. Psoriasis. Lancet 2015;386:983–94.
Chapter 27  Psoriasis: Epidemiology 349

44 Martin BA, Chalmers RJ, Telfer NR. How great is the risk of further 53 Cohen R, Robinson D Jr, Paramore C et al. Autoimmune disease con-
psoriasis following a single episode of acute guttate psoriasis? Arch comitance among inflammatory bowel disease patients in the United
Dermatol 1996;132:717–8. States, 2001–2002. Inflamm Bowel Dis 2008;14:738–43.
45 Griffiths CE, Barker JN. Pathogenesis and clinical features of psoria- 54 Wolf N, Quaranta M, Prescott NJ et  al. Psoriasis is associated with
sis. Lancet 2007;370:263–71. pleiotropic susceptibility loci identified in type II diabetes and Crohn
46 Nyfors A, Lemholt K. Psoriasis in children. A short review and a sur- disease. J Med Genet 2008;45:114–6.
vey of 245 cases. Br J Dermatol 1975;92:437–42. 55 Piruzian ES, Ishkin AA, Nikolskaia TA et al. The comparative analysis
47 Horton DB, Scott FI, Haynes K et  al. Antibiotic exposure, infection, of psoriasis and Crohn disease molecular‐genetical processes under
and the development of pediatric psoriasis: a nested case‐control pathological conditions. Mol Biol (Mosk) 2009;43:175–9.
study. JAMA Dermatol 2016;152:191–9. 56 Li Y, Begovich AB. Unraveling the genetics of complex diseases: sus-
48 Kwon HH, Na SJ, Jo SJ, Youn JI. Epidemiology and clinical features of ceptibility genes for rheumatoid arthritis and psoriasis. Semin
pediatric psoriasis in tertiary referral psoriasis clinic. J Dermatol Immunol 2009;21:318–27.
2012;39:260–4. 57 Annunziato F, Cosmi L, Liotta F et al. Type 17 T helper cells‐origins,
49 Kim SK, Kang HY, Kim YC, Lee ES. Clinical comparison of psoriasis in features and possible roles in rheumatic disease. Nat Rev Rheumatol
Korean adults and children: correlation with serum anti‐streptolysin O 2009;5:325–31.
titers. Arch Dermatol Res 2010;302:295–9. 58 Silverberg JI, Simpson EL. Association between severe eczema in
50 Chandran NS, Gao F, Goon AT et al. Clinical characteristics of child- children and multiple comorbid conditions and increased health care
hood psoriasis in a multi‐ethnic Asian population. J Dermatol utilization. Pediatr Allergy Immunol 2013;24:476–86.
2012;39:278–9. 59 Kimball AB, Wu EQ, Guerin A et al. Risks of developing psychiatric
51 Al‐Fouzan AS, Nanda A. A survey of childhood psoriasis in Kuwait. disorders in pediatric patients with psoriasis. J Am Acad Dermatol
Pediatr Dermatol 1994;11:116–9. 2012;67:651–7.e1–2.
52 Mercy K, Kwasny M, Cordoro KM et  al. Clinical manifestations of 60 Ferreira BI, Abreu JL, Reis JP, Figueiredo AM. Psoriasis and associated
pediatric psoriasis: results of a multicenter study in the United States. psychiatric disorders: a systematic review on etiopathogenesis and
Pediatr Dermatol 2013;30:424–8. clinical correlation. J Clin Aesthet Dermatol 2016;9:36–43.

SECTION 5: PSORIASIS
350 

CHA PTER  2 8

Psoriasis: Aetiology and Pathogenesis


Jonathan Barker
St John’s Institute of Dermatology (King’s College), Guy’s Hospital, London, UK

Introduction, 350 Environmental risk factors, 351 Pathogenetic model of psoriasis, 352
Genetic epidemiology, 350 Pathogenetic mechanisms, 351 Pustular psoriasis, 353

Abstract epidermal barrier and innate immune mechanisms leading to ac-


tivation of adaptive T‐cell immune pathways which in turn lead
There have been very significant advances made over the past 20 years to changes in skin homeostasis and inflammation. Specific signal-
in our understanding of the pathogenesis of psoriasis underpinned ling (e.g. NF‐κB) and T‐cell differentiation pathways (Th IL‐17) have
by a combination of genetic discovery and immunological func- been identified which in turn have led to therapeutic advances.
tional studies. Key to pathogenesis is the interplay between the Pustular forms of the disease have a distinct genetic background.

Key points • Genetic studies reveal multiple loci associated with psoriasis.
SECTION 5: PSORIASIS

However a full genetic map remains to be established.


• The primary genetic determinant for psoriasis is within the
• Genetic and immunological studies indicate that the skin barrier
major histocompatibility complex on chromosome 6.
and innate and adaptive immunity play a key role in psoriasis
• There is evidence that pustular forms of psoriasis have a different
vulgaris pathogenesis.
pathogenetic basis when compared to plaque forms.
• Current evidence strongly supports a pivotal role of Th17 T cells
in psoriasis pathogenesis.

Introduction second‐degree relatives of sufferers than unaffected


Psoriasis vulgaris (PV) is a common, complex chronic c­ ontrol subjects. Swedish studies support these data and
inflammatory disorder often referred to as an immune‐ reveal the prevalence of psoriasis to be 7.8% in first‐degree
mediated inflammatory disease in common with other relatives compared with a prevalence of 3.14% in matched
organ‐specific immune‐mediated diseases such as rheu- controls and 1.97% in the overall population. Based on
matoid arthritis and inflammatory bowel disease. Disease population data, the risk of psoriasis to a child has been
pathogenesis is multifactorial and in great part involves calculated to be 14% if one parent was affected, 41% if
interplay between genetic mechanisms and environmen- both parents were affected, and 6% if one sibling was
tal agents [1]. Rarer forms of psoriasis including general- affected, compared to 2% when no parent or sibling was
ized pustular psoriasis (GPP) may have a different affected [4]. Henseler and Christophers [5] demonstrated
pathogenetic basis and potentially more closely follow a bimodal peak in disease onset. Thus, type I is hereditary,
Mendelian patterns of inheritance. Furthermore, their strongly HLA associated (particularly HLA‐C:06:02),
onset at a very early age makes them important in paedi- early onset and more likely to be severe. Type II is spo-
atric dermatological practice. radic, HLA unrelated, of late onset and usually mild.
Thus when disease onset is in childhood it tends to be
more severe and a family history is more likely, as is asso-
Genetic epidemiology ciation with HLA‐C:06:02. Interestingly, the development
There is overwhelming evidence that psoriasis has an and severity of psoriasis may be influenced by the sex of
important genetic component (reviewed in [2, 3]). In the contributing parent. Thus several studies have noted
Lomholt’s classic epidemiological study of psoriasis in evidence for a preferential paternal effect, while one
the Faroe Islands (1963), in which he examined more than Scottish study further showed earlier age of onset when
10 000 inhabitants, he made the key observation that the the disease was inherited from the father, consistent with
incidence of psoriasis was much greater among first‐ and ‘genetic anticipation’ [6].

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 28  Psoriasis: Aetiology and Pathogenesis 351

Support for these population studies comes from Historically, psoriasis was classified as a disorder of
a­ nalysis of various family pedigrees in which psoriasis keratinization but a combination of serendipitous obser-
appears throughout multiple generations. Perhaps the vations and science has revealed a primary role for
most robust data supporting a genetic basis to psoriasis immune mechanisms in disease pathogenesis (reviewed
come from studies examining concordance for the ­disease in [3]). There is considerable evidence that T lymphocytes
in twins (reviewed in [2]). Examination of individuals play an important role in the development of plaques of
from the Danish Twin Registry has shown concordance psoriasis. This evidence includes:
for psoriasis in 64% of monozygotic (identical) twins • early influx of T cells into expanding lesions
compared to 15% for dizygotic twins, corresponding to • strong association with the major histocompatibility
an estimated heritability of 91%. Very similar figures complex (MHC), particularly HLA‐C:06:02
(73% and 20% respectively) were found in a retrospective • ablative effect of anti‐T‐cell therapy
study in the USA, although in an Australian study lower • anecdotal evidence of development of psoriasis after
concordance rates were observed: 35% in monozygotic syngeneic bone marrow transplant
twins compared to 12% in dizygotic twins. Of interest, • induction of lesional phenotype in nonlesional psori-
when monozygotic twins are concordant for psoriasis, atic skin transplanted on to severe combined immuno-
the age of onset, distribution of the disease and severity deficient mice after injection of autologous T cells.
are very similar, suggesting that genetic factors play a More recently it has emerged that the innate immune
role in these parameters. However, concordance rates do system, which provides an early response against harm
not reach 100%, even when older twins are examined, to the host, is dysregulated in psoriasis. Further, in
indicating that other factors including the environment psoriatic stratum corneum, there is an abundance of
­
play a key role in disease expression. Variable extent, ­antimicrobial peptides such as defensins and cathelici-

SECTION 5: PSORIASIS
clinical features and pattern of inheritance suggest that dins, which have the capacity to activate innate immune
significant disease heterogeneity exists. Thus rare fami- responses. These in turn lead to antigen‐driven T‐cell
lies exist in which changes in a single gene may be suffi- expansion and activation. The key question remains the
cient to cause the disease while in more common forms it nature of the antigen involved. To date this is unresolved
is likely that changes in multiple genes, interacting both but evidence of an antigen‐specific response includes:
with each other and the environment, are required for (i) limited clonality of infiltrating T cells; (ii) persistence
disease expression. of T‐cell clones in plaques over several years; and
Little is known of the genetic epidemiology of other (iii) identical T‐cell clones in tonsils and skin of patients
forms. Guttate psoriasis, which frequently has onset in with Streptococcus‐associated psoriasis [8].
childhood, is almost invariably HLA‐C:06:02 associated A primary role for Th1‐type responses has previously
[7] and thought to be closely linked pathogenetically to been postulated in disease pathogenesis but more recent
Type I plaque psoriasis. GPP, although usually manifest- observations strongly suggest that Th17 cells are likely to
ing in middle age, has been reported in infants and in be the primary pathogenic subset. Evidence includes
childhood. Families with parental consanguinity and genetic association, with IL‐23 a key cytokine in genera-
multiple affected individuals have been observed, tion of Th17 cells, abundance of IL‐23 in psoriatic tissue
­suggesting autosomal recessive inheritance, at least in and induction of a psoriatic phenotype by IL‐23 in animal
some pedigrees. models. Other cytokines downstream of IL‐23 in Th17
pathways, including IL‐17 and IL‐22, have similar func-
tional effects [9].
Environmental risk factors
Present evidence indicates that interactions between Molecular genetics
genes and the environment are important in disease cau- Very significant progress (reviewed in [10]) has been
sation. Many environmental factors have been linked to made in understanding the genetic architecture of psoria-
psoriasis and have been implicated in, for example, initia- sis, aided in great part by technological advances made
tion of the disease process and exacerbation of pre‐exist- possible by the human genome mapping project and
ing disease. However, robust evidence validating these ­subsequent large‐scale molecular genetic projects. With
factors is often lacking. They include: respect to PV, genome‐wide association studies (GWAS)
• trauma (Koebner phenomenon) have revealed significant new knowledge. Pustular forms
• infection, especially Streptococcus have benefited from methods allowing rapid cost‐­
• drugs, especially lithium, β‐blockers, antimalarials effective sequencing (next‐generation sequencing) such
• sunlight as whole exome sequencing.
• psychogenic factors. In PV, studies clearly indicate that the primary genetic
susceptibility locus is on chromosome 6p21.3. Most
­evidence indicates that the causative gene involved is
Pathogenetic mechanisms HLA‐C:06:02 although extensive linkage disequilibrium
The cardinal features of lesional psoriatic skin are: at this locus makes it difficult to be categorical.
• epidermal hyperproliferation with loss of differentiation HLA‐C:06:02 positive individuals have a five‐fold
• dilatation and proliferation of dermal blood vessels increased risk of developing psoriasis. The heritability
• cutaneous inflammation. due to HLA‐C:06:02 is greater than for all other identified
352 Section 5  Psoriasis

Gene/
Chr Function P value OR (95% CI)
locus

5q15 ERAP1 Antigen processing 2.56 × 10–11 1.27

6p21 HLA-C Ag presentation 4.06 × 10–214 4.66

1p31 IL23R IL-23 signalling 7.13 × 10–07 1.49

5q33 IL12B IL-23 signalling 4.93 × 10–11 1.39

12q13 IL23A IL-23 signalling 2.49 × 10–07 1.49

6q21 TRAF3IP2 IL-17/NF-κB signalling 5.29 × 10–20 1.36

19p13 TYK2 IL-23/NF-κB signalling 4.04 × 10–11 1.34

2p16 REL NF-κB signalling 3.59 × 10–9 1.23

5q33 TNIP1 NF-κB signalling 3.92 × 10–05 1.27

6q23 TNFAIP3 NF-κB signalling 6.54 × 10–07 1.22


SECTION 5: PSORIASIS

14q13 NFKBIA NF-κB signalling 1.52 × 10–11 1.19


Fig. 28.1  Genetic loci with strongest association to
psoriasis vulgaris as revealed by genome‐wide association
1q21 LCE3D Skin barrier 3.32 × 10–10 1.29
studies. Colour coding groups together overlapping
functions to demonstrate that such genetic studies
1p36 IL28RA IFN signalling 6.89 × 10–8 1.22 highlight specific biological pathways that associate with
psoriasis vulgaris. Chr, chromosomal localization; Gene,
2q24 IFIH1 IFN signalling 1.06 × 10–13 1.38 top candidate gene at each locus; Function, putative
function of candidate gene; P value, statistical validation of
20q13 ZNF313 IFN signalling 1.65 × 10–06 1.20 association; OR, odds ratio or size of effect; Ag, antigen;
IFN, interferon; IL, interleukin; NF, nuclear factor.

loci combined. Approximately 50% of patients with homeostasis and altered growth pattern in genetically
severe psoriasis in UK tertiary practice are HLA‐C:06:02 predisposed individuals [9]. The initiation phase involves
positive (Jonathan Barker, personal communication). a close interplay between external factors and the
HLA‐C is involved in antigen presentation to CD8 and epidermal barrier. Triggers include physical injury (which
NK T cells, implicating them in disease pathogenesis. causes Koebner phenomenon), infections and medica-
GWAS have so far identified 46 genetic loci that associ- tions, although in most cases they are never identified.
ate with PV (Fig.  28.1), explaining about 30% of the Such triggers lead to activation of innate immunity, pos-
genetic contribution to the disease [11]. Currently the sibly through the release of the antimicrobial peptide
‘missingness’ remains unexplained but multiple genetic LL37 (cathelicidin) by keratinocytes. LL37 binds with
explanations exist. Each identified locus has a low impact pathogen‐derived DNA or self‐DNA that has been
on heritability, with odds ratios (OR) of effect size released by stressed or dying cells and forms complexes
between 1 and 2. Nevertheless they provide major that activate toll‐like receptor (TLR) 9 on plasmacytoid
insights into disease biology and give specificity to dendritic cells [12]. Imiquimod has been shown to induce
immunological models of disease pathogenesis, clearly psoriasiform skin inflammation in mouse m ­ odels, indi-
identifying disease‐specific biological pathways. This cating that TLR‐7 and TLR‐8 may also be involved in pso-
knowledge has been used to important clinical benefit, riasis pathogenesis. TLR activation promotes type I
for example by identifying new drug targets including interferon (IFN) release, which, along with tumour necro-
those targeted by the biologic therapies. sis factor (TNF)‐α, IL‐6 and IL‐1β, activates local myeloid
dendritic cells, consequently inducing T‐cell‐mediated
(adaptive) inflammation. Genetic data would suggest
Pathogenetic model of psoriasis that nuclear factor (NF)‐κB mechanisms are important in
Combining immunological and genetic knowledge allows these processes [10]. Myeloid dendritic cells migrate into
the building of a pathogenetic model of psoriasis draining lymph nodes and release cytokines including
(Fig. 28.2) in which dynamic interactions between multi- TNF‐α, IL‐23 and IL‐12 that activate allogeneic T cells.
ple cell types and numerous cytokines in response to trig- GWAS studies suggest that endoplasmic reticulum amin-
gers culminate in the disruption of skin immune opeptidase (ERAP)‐1 plays a role in antigen processing
Chapter 28  Psoriasis: Aetiology and Pathogenesis 353

activate keratinocytes, resulting in the release of cytokines


Pathogens and chemokines that continue to recruit and activate
inflammatory cells and induce altered growth in keratino-
cytes and the dermal vasculature (transforming growth
factor [TGF]‐β and vascular endothelial growth factor
Tissue
[VEGF] respectively have been implicated in these
(skin)
LCE3D
specific processes).
events
Pustular psoriasis
IL28RA
Interferon While most attention has been paid to PV, the common
signalling IFIH1 form of the disease, recent genetic studies supported by
ZNF313
functional genomic experiments have provided major
insights into pathogenetic mechanisms in pustular forms
REL of psoriasis. With respect to onset in childhood, perhaps
Innate the most important example is GPP. Indeed, in von
TNIP1
immunity: Zumbusch’s original description of the disease, one case
TNFAIP3 NF-κB/TNF
pathways IL23R
had onset in very early childhood. Two studies, one using
NFKBIA classic linkage analysis [14] and a second using next‐
IL12B generation sequencing [15], identified mutations in the
Immune IL23A gene for IL36RN, a receptor antagonist and member of

SECTION 5: PSORIASIS
signalling the IL‐1 superfamily. As a consequence, signalling
TRAF3IP2
through the IL‐36 receptor is not regulated and an autoin-
Adaptive TYK2 flammatory syndrome results. Many studies from around
immunity: the world have since confirmed these findings.
ERAP1
Ag processing
& presentation References
HLA-C
1 Mahil SK, Capon F, Barker JN. Update on psoriasis immunopatho-
Differentiation genesis and targeted immunotherapy. Semin Immunopathol 2016;
IL-17
38:11–27.
2 Barker J. Psoriasis heritability: 125 years and counting. Br J Dermatol
2014;171:3–5.
3 Elder JT, Nair RP, Guo SW et  al. The genetics of psoriasis. Arch
Dermatol 1994;130:216–24.
4 Andressen C, Henseler T. [Inheritance of psoriasis. Analysis of 2035
family histories]. Hautarzt 1982;33:214–7.
5 Henseler T, Christophers E. Psoriasis of early and late onset: charac-
terization of two types of psoriasis vulgaris. J Am Acad Dermatol
1985;13:450–6.
6 Burden AD, Javed S, Bailey M et  al. Genetics of psoriasis: paternal
inheritance and a locus on chromosome 6p. J Invest Dermatol
1998;110:958–60.
7 Mallon E, Bunce M, Savoie H et al. HLA‐C and guttate psoriasis. Br J
Fig. 28.2  Schematic representation of immunopathogenesis model of Dermatol 2000;143:1177–82.
psoriasis (see text for more detail). Tissue‐specific events lead through 8 Diluvio L, Vollmer S, Besgen P et  al. Identical TCR beta‐chain rear-
interferon signalling to activation of innate immune pathways. In turn this rangements in streptococcal angina and skin lesions of patients with
leads to activation of adaptive T‐cell pathways involving specific differen- psoriasis vulgaris. J Immunol 2006;176:7104–11.
tiation pathways. Characters in columns on left and right reference 9 Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med
candidate genes listed in Fig. 28.1 to demonstrate overlap between 2009;361:496–509.
10 Mahil SK, Capon F, Barker JN. Genetics of psoriasis. Dermatol Clin
immunological pathways and identified genetic loci. Ag, antigen; IFN,
2015;33:1–11.
interferon; IL, interleukin; NF, nuclear factor; TNF, tumour necrosis factor.
11 Capon F, Burden AD, Trembath RC, Barker JN. Psoriasis and other
complex trait dermatoses: from loci to functional pathways. J Invest
Dermatol 2012;132:915–22.
and HLA‐C in antigen presentation [13]. Once activated, 12 Lande R, Botti E, Jandus C et al. The antimicrobial peptide LL37 is a
T‐cell autoantigen in psoriasis. Nat Commun 2014;5:5621.
T cells enter the circulation and move towards inflamed 13 Genetic Analysis of Psoriasis Consortium & the Wellcome Trust Case
skin through interactions with adhesion molecules Control Consortium 2, Strange A, Capon F, Spencer CC et  al.
(including P‐selectin and E‐selectin) on the endothelial A genome‐wide association study identifies new psoriasis suscepti-
cells of blood vessels. Functional immunology and genet- bility loci and an interaction between HLA‐C and ERAP1. Nat Genet
2010;42:985–90.
ics clearly indicates that the ­primary T cell involved in 14 Marrakchi S, Guigue P, Renshaw BR et al. Interleukin‐36‐receptor
psoriasis pathogenesis is ­differentiated along the IL‐17 antagonist deficiency and generalized pustular psoriasis. N Engl J
pathway. This is supported by clinical studies showing Med 2011;365:620–8.
15 Onoufriadis A, Simpson MA, Pink AE et  al. Mutations in IL36RN/
dramatic clinical benefit from therapeutic monoclonal
IL1F5 are associated with the severe episodic inflammatory skin dis-
antibodies that inhibit IL‐23 and IL‐17. The effector mol- ease known as generalized pustular psoriasis. Am J Hum Genet
ecules secreted by T cells including IL‐17 and IL‐22 then 2011;89:432–7.
354 

CHA PTER  2 9

Psoriasis: Clinical Features and Comorbidities


Derek H. Chu1 & Kelly M. Cordoro2
1
 Stanford University School of Medicine, Palo Alto, CA, USA
2
 University of California San Francisco, San Francisco, CA, USA

Clinical features, 354 Disease comorbidities, 359

Abstract Major categories include plaque, guttate, inverse, nail, pustu-


lar and erythrodermic psoriasis. In addition to arthritis, children
with psoriasis are at increased risk for other comorbidities such as
Paediatric psoriasis is a common and often chronic inflammatory
metabolic and cardiovascular disease, psychiatric disorders, social
disorder of the skin. Clinical subtypes of psoriasis are based pri-
stigma and reduced quality of life.
marily on the site of involvement and predominant morphology.

Key points receptor antagonist (IL36RN) and caspase recruitment family


member 14 (CARD14).
SECTION 5: PSORIASIS

• A diagnosis of juvenile psoriatic arthritis can be made in the


• Plaque psoriasis is the most prevalent form of psoriasis in the
presence of arthritis and psoriasis, or arthritis coupled with a
paediatric population and commonly features facial and anogenital
family history of psoriasis in a first‐degree relative, nail pitting
involvement, annular morphology and prominent pruritus.
or onycholysis, and/or dactylitis.
• Guttate psoriasis has been reported in up to 44% of children
• Emerging data suggest that children with psoriasis are at
with psoriasis and is frequently preceded by streptococcal
increased risk for metabolic and cardiovascular comorbidities,
pharyngitis or perianal infection. A subset of patients may go on
including obesity, hyperlipidaemia, diabetes and hypertension.
to develop chronic plaque psoriasis.
Age‐related screening should be performed to facilitate early
• Psoriatic napkin dermatitis is common in children under the age
intervention.
of 2 years, and can acutely progress to widespread disease, the
• Psoriasis can have detrimental psychosocial effects on paediatric
so‐called psoriatic napkin rash with dissemination.
patients regardless of the extent of disease, leading to social
• A subset of patients with early onset and severe generalized
stigma, poor quality of life, depression and anxiety.
pustular psoriasis carry genetic mutations in interleukin 36

­Clinical features Fortunately, many children have limited disease amenable


to topical treatment, although a subset of patients present
Psoriasis is a common inflammatory disorder that affects with severe and recalcitrant psoriasis requiring photo-
the skin, nails and joints. One third of cases present therapy and/or systemic medications.
in childhood [1]. Although the classic description of pso- There are multiple recognized clinical subtypes of pso-
riasis is a well‐demarcated erythematous plaque with riasis, the classification of which encompasses both pri-
­silvery‐white scale, there can be significant age‐related mary morphology and site of involvement. Major
variations in the morphology, distribution and severity. subtypes include plaque, guttate, pustular and erythro-
Compared to adults, young children tend to have more dermic. Subtypes with special features based on site
facial and anogenital involvement. Individual lesions include inverse or flexural psoriasis, napkin (anogenital)
may be smaller and thinner and commonly feature annu- psoriasis and nail psoriasis (Box 29.1) [4].
lar morphology. Pruritus is a frequent symptom in chil-
dren [2]. Psoriasis may be triggered or exacerbated by Plaque psoriasis
trauma (Koebner phenomenon), infection, stress and Plaque psoriasis is the most common form of psoriasis in
medications [3]. Paediatric psoriasis may be mild and the paediatric population, with a reported prevalence of
localized or diffuse, severe and debilitating. In most cases, 34–84% [5,6]. Classically, patients develop well‐demarcated
psoriasis is a chronic condition that follows a relapsing erythematous papules and plaques with overlying silvery
and remitting course. The unpredictable natural history, scale (Fig.  29.1), often distributed symmetrically over the
typified by periods of exacerbation and spontaneous extensor extremities, scalp and face. The removal of scale
remission, makes management rather challenging. results in pinpoint bleeding, known as the Auspitz sign.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 29  Psoriasis: Clinical Features and Comorbidities 355

Box 29.1  Clinical variants of childhood psoriasis

Plaque

Facial
Scalp
Palmoplantar
Annular
Eczema–psoriasis overlap
Linear

Guttate Fig. 29.2  Facial psoriasis – 6‐year‐old boy with psoriasis over the eyelids
Follicular/micropapular and periorbital skin.

Inverse (intertriginous)

Napkin (diaper)/anogenital

Nail

Pustular

Erythrodermic

Source: Cordoro [4]. Reproduced with permission of Elsevier.

SECTION 5: PSORIASIS
Fig. 29.3  Scalp psoriasis – 4‐year‐old girl with plaque psoriasis involving
the postauricular and occipital scalp.

it can be difficult to differentiate facial psoriasis from


eczema, unlike eczema, lesions of psoriasis on the face
tend to be more sharply marginated, more plaque‐like,
may be annular and are less pruritic [8]. Periorbital
involvement is common (Fig. 29.2). Facial psoriasis may
be accompanied by not only severe symptoms such as
itch and discomfort, but also social stigmatization.

Fig. 29.1  Plaque psoriasis – 9‐year‐old boy with multiple discrete, sharply Scalp
marginated, erythematous papules and plaques with overlying silvery‐ The scalp is the most frequent site of involvement of
white scale.
plaque psoriasis in children (Fig. 29.3). It is the initial site
of presentation of psoriasis in about 40–60% of children
and adolescents [9]. Like facial psoriasis, scalp disease can
Plaque psoriasis can be subcategorized based on location, also occur exclusively for many years. Scalp psoriasis
such as facial, scalp and palmoplantar, as well as by may present as diffuse scaling or discrete plaques, regu-
morphology, including annular, eczema–psoriasis overlap larly involving the postauricular and occipital scalp
and linear variants. although it can occur anywhere on the scalp as well as
on,  and within, the ears. The scale may become very
Facial thick and lead to matting of the hair, known as pityriasis
The face is a common site for psoriasis in children as amiantacea [10]. When severe and prolonged, this pres-
opposed to adults [7]. Facial psoriasis may arise in isola- entation can result in focal nonscarring alopecia. The alo-
tion or as part of a more disseminated eruption. In a large pecia is generally reversible with adequate treatment [3].
case series of 1262 children with psoriasis in Sydney, Perhaps the most debilitating aspect of scalp psoriasis
Australia, 38% of patients had facial involvement [8]. is  the propensity for intense pruritus. Scalp psoriasis
About 4% of cases had isolated facial plaques. Facial may  be difficult to differentiate from, and may occur
involvement is a characteristic feature of psoriatic napkin simultaneously with, seborrhoeic and atopic dermatitis.
rash with dissemination (see Inverse psoriasis). Although Tinea capitis should be ruled out with a fungal culture.
356 Section 5  Psoriasis

Fig. 29.5  Annular psoriasis – 11‐year‐old boy with numerous annular


SECTION 5: PSORIASIS

plaques on the back.

Fig. 29.4  Plantar psoriasis – 12‐year‐old boy with severe hyperkeratotic


coalescent plaques on the plantar feet.

Palmoplantar
This is an uncommon variant of psoriasis in the paediatric
population, the prevalence of which was only 4% in a
large series of 1262 cases of childhood psoriasis [8]. The
palms and soles can either have a ‘glazed’ appearance with
erythema and fissuring, or more typical thick coalescent
plaques with silvery scale (Fig. 29.4). Both presentations
typically have a sharp demarcation at the medial and lat-
eral borders of the palms and soles.

Annular
In contrast to adults with psoriasis, children more often
present with annular and serpiginous plaques on the face,
trunk and extremities (Fig.  29.5). The annular plaques
may be studded with peripheral pustules and these chil-
dren are at risk of generalized pustular psoriasis. Annular
presentations of psoriasis may be confused clinically with Fig. 29.6  Eczematous psoriasis – 9‐year‐old boy with overlapping features
tinea corporis and nummular dermatitis. of psoriasis and eczema consisting of diffuse xerosis, follicular accentuation
and discrete scaly pink patches.

Eczema–psoriasis overlap
A small subset of patients present with overlapping fea- Linear
tures of both eczema and psoriasis. Variably referred to as This is a rare form of childhood psoriasis. Only one case
eczematous psoriasis or psoriasiform eczema, these patients was identified in a child >2 years of age in the case series
present with either an admixture of eczematous and pso- by Morris et  al [8]. Patients manifest with psoriasiform
riatic lesions, or have individual lesions with an interme- plaques following the lines of Blaschko. Histopathological
diate morphology (Fig.  29.6). These children typically evaluation may be necessary to differentiate this subtype
have an atopic background, or have a family history of from inflammatory linear verrucous epidermal naevus
atopy and psoriasis. (ILVEN).
Chapter 29  Psoriasis: Clinical Features and Comorbidities 357

Fig. 29.7  Guttate psoriasis – 12‐year‐old boy with numerous ‘drop‐like’ Fig. 29.8  Napkin (diaper) psoriasis – 19‐month‐old girl with well‐defined
scaly papules on the trunk. shiny red plaque with minimal scale involving the perineum. There is early

SECTION 5: PSORIASIS
dissemination of disease with formation of erythematous plaques on the
abdomen.

Guttate psoriasis
Guttate psoriasis is the second most common variant of
psoriasis in children and adolescents, with a reported fre- have more typical plaques elsewhere on the body. Infants
quency of 6.4–44% [5]. Patients present with an abrupt in particular will manifest with a napkin dermatitis or
onset of small <1 cm ‘drop‐like’ scaly papules primarily anogenital disease.
on the trunk, proximal extremities and face (Fig.  29.7).
Group A β‐haemolytic streptococcal pharyngitis or peria- Napkin/anogenital psoriasis
nal infection 1–2 weeks prior to onset of the eruption is Psoriatic napkin dermatitis, or ‘napkin psoriasis’, is a
frequently noted [11]. Recent data suggest that guttate common presentation in children under the age of 2
psoriasis in children may have a variable prognosis years. In fact, 45% of psoriasis patients in this age group
depending on the presence or absence of streptococcal will manifest with napkin rash [8]. In patients present-
infection as a trigger. Ko et al. found that preceding strep- ing with other variants of psoriasis, more than 20% have
tococcal pharyngitis predicted guttate morphology and a history of psoriatic napkin rash [8]. Well‐defined,
eventual resolution of the psoriasis [12], while Mercy glazed‐appearing red plaques in the napkin area, often
et al. found that preceding streptococcal pharyngitis pre- with attenuated scale or devoid of scale altogether and
dicted guttate morphology but not disease severity. In the involving the creases, characterize this subtype of pso-
latter study, initial guttate morphology in the absence of riasis (Fig. 29.8). The distribution and morphology of the
streptococcal infection predicted progression to severe eruption can help to distinguish psoriasis from other
psoriasis [13]. Therefore, although guttate psoriasis may napkin eruptions including irritant and candidal derma-
spontaneously resolve after 3–4 months, a subset of titis and acrodermatitis enteropathica. As these condi-
patients will go on to develop classic, and possibly severe, tions can overlap, the most helpful distinguishing
plaque psoriasis. A very small subset of patients with gut- feature is finding more typical psoriasis elsewhere on
tate psoriasis may present with 1–3 mm micropapular, the body. A subset of patients with napkin psoriasis
follicular‐based lesions alone or in the presence of more acutely progress from localized to widespread disease,
typical guttate morphology. Guttate psoriasis is often so‐called psoriatic napkin rash with dissemination.
confused with pityriasis rosea, a viral exanthem that fol- Reports suggest that those with psoriatic napkin rash
lows Langer’s lines on the trunk to create a ‘Christmas with dissemination are at higher risk of developing
tree’ pattern. A biopsy, if necessary, will differentiate ­classic psoriasis later in life [15].
between the two conditions.
Nail psoriasis
Inverse psoriasis Nail psoriasis has been reported to occur in up to 40% of
Inverse or flexural psoriasis, in contrast to chronic children and adolescents [13]. Nail changes may arise in
plaque psoriasis, has a distinct morphology with bright isolation or can precede, coincide with, or develop after
red or shiny erythema, frequent maceration and mini- the onset of cutaneous disease [14]. Pitting of the nails
mal scale [14]. Common sites of involvement include is  most frequently observed (Fig.  29.9). Additional
the  neck, groin and axillae. In addition, patients may features include onycholysis, oil spots, subungual
­
358 Section 5  Psoriasis

Fig. 29.11  Pustular psoriasis – 8‐year‐old boy with generalized pustular


psoriasis of von Zumbusch.
Fig. 29.9  Nail psoriasis – 11‐year‐old girl with pitting, oil spots and distal
onycholysis of the fingernails.

A proportion of patients with generalized pustular


SECTION 5: PSORIASIS

psoriasis harbour autosomal recessive mutations in the


interleukin (IL)‐36 receptor antagonist (IL36RN). This
recently described monogenic autoinflammatory syn-
drome is referred to by the acronym DITRA, (deficiency
of interleukin‐36 receptor antagonist) [17]. Aberrant
IL‐36RN structure and function leads to unregulated
secretion of inflammatory cytokines, generalized pustulo-
sis and unopposed activation of inflammatory pathways
[18]. This variant of generalized pustular psoriasis typi-
cally occurs in the absence of concomitant plaque psoria-
sis. More recently, gain‐of‐function mutations have been
discovered in caspase recruitment family member 14
Fig. 29.10  Pustular psoriasis – 14‐year‐old boy with discrete pustules at
(CARD14), a mediator of NF‐κB signalling that predis-
the periphery of bright red annular plaques on the arm. poses patients to earlier onset disease and severe gener-
alized pustular psoriasis refractory to treatment [19]. Of
note, CARD14 mutations have also been identified in
hyperkeratosis, splinter haemorrhages, onychodys- overlap presentations of psoriasis and pityriasis rubra
trophy and trachyonychia. The presence of nail disease pilaris [20].
has  been associated with psoriatic arthropathy (see Deficiency of IL‐1 receptor antagonist (DIRA) is another
Disease comorbidities). rare, autosomal recessive, neonatal onset, life‐threatening
autoinflammatory disease that may be confused with early
Pustular psoriasis onset pustular psoriasis. Patients present with dissemi-
Pustular psoriasis is rare, but does occur, in children of all nated pustules soon after birth, along with periostitis,
ages [14]. Pustular psoriasis in the paediatric age group sterile multifocal osteomyelitis and elevated inflammatory
may be localized or generalized and often follows a markers [21]. IL‐1 antagonists are life‐saving therapies for
relapsing and remitting course. Discrete and coalescent these patients.
sterile superficial pustules arise on an erythematous base
and may form annular plaques (Fig. 29.10). Another vari- Erythrodermic psoriasis
ant common in children is the development of pustules at This is an extremely rare form of psoriasis in paediatric
the periphery of pre‐existing psoriatic plaques [16]. patients and may arise de novo or in a child with pre‐
Generalized forms of pustular psoriasis, including the existing plaque psoriasis (Fig. 29.12). Psoriasis should be
von Zumbusch variant, are sometimes associated with considered in the differential diagnosis of an erythroder-
systemic symptoms such as fever, malaise, arthralgias mic infant or child. Characterized by diffuse erythema
and elevated inflammatory markers (Fig. 29.11). Localized and scaling involving >90% of the body surface area in
forms, including acrodermatitis continua of Hallopeau severe cases, erythrodermic psoriasis can be compli-
and palmoplantar pustulosis, are characterized by severe cated by temperature dysregulation, hypernatraemic
involvement of the digits and nail unit and palms and dehydration, hypoalbuminaemia, infection and cardiac
soles, respectively. failure [22].
Chapter 29  Psoriasis: Clinical Features and Comorbidities 359

c­ ommon presentation. The presence of dactylitis favours


a diagnosis of psoriasis over other arthritides. A relatively
milder form of disease commonly associated with spon-
dyloarthritis and HLA‐B27 positivity tends to affect ado-
lescent males, while a more severe and recalcitrant form
peaks at 2 years of age and is frequently characterized by
a positive anti‐nuclear antibody (ANA) and female pre-
disposition [25]. Rheumatoid factor is typically negative.
One study suggests that there is a significantly increased
risk of joint involvement in patients of all ages with eryth-
rodermic, generalized pustular, inverse and nail psoriasis
[26]. Of note, asymmetrical anterior uveitis has been
reported in up to 17% of children with psoriatic arthritis,
underscoring the importance of ophthalmic evaluation in
the setting of psoriatic arthritis [27].

Metabolic and cardiovascular
Obesity
Paediatric patients with psoriasis appear to have a higher
Fig. 29.12  Erythrodermic psoriasis – 10‐year‐old boy with erythroderma in prevalence of excess adiposity and obesity when com-
context of prior plaque psoriasis. pared with children without psoriasis [28]. In a large mul-

SECTION 5: PSORIASIS
ticentre international cross‐sectional study of 614 children
aged 5–17 years with a 6‐month or more history of psoria-
­Disease comorbidities sis, the odds ratio (OR) of obesity (body mass index [BMI]
≥95th percentile) and excess adiposity (BMI ≥85th percen-
Sufficient evidence exists to underscore the importance
tile) when compared with controls was 4.29 and 2.65,
of carefully evaluating and monitoring paediatric psori-
respectively [29]. Moreover, those with severe psoriasis,
asis patients for potential extracutaneous comorbidities.
based on physician global assessment and an affected
Psoriatic arthritis is the most widely recognized. Patients
body surface area of greater than 10%, had an overall
are also at higher risk for components of the metabolic
higher risk for obesity than those with mild psoriasis (OR
syndrome, especially obesity, as well as dyslipidaemia,
4.92 vs. 3.6). Waist circumference, a surrogate marker for
diabetes and hypertension. Of equal importance is the
central adiposity, was also greater in psoriatic patients. A
potentially detrimental effect of psoriasis on a child’s
>90th percentile waist circumference was found in 21.2%
quality of life. Children with psoriasis may suffer from
of patients with severe psoriasis compared to only 14%
poor self‐esteem, depression and anxiety. Historically,
with mild psoriasis and 9.3% of controls [29]. Some
the rate of counselling and screening for paediatric pso-
authors postulate that obesity may predispose suscepti-
riasis comorbidities has been low [23]. In order to inter-
ble individuals to the development of psoriasis, perhaps
vene early and potentially mitigate any long‐term health
related to a systemic pro‐inflammatory state mediated by
consequences, it is critical to actively screen patients
excess adipose tissue. In fact, one study demonstrated
and  identify risk factors for these potential disease
that being overweight and obese preceded psoriasis by at
comorbidities.
least 2 years in 93% of children with psoriasis [30]. It is,
however, still unclear whether weight loss can help to
Psoriatic arthritis
reduce psoriasis severity.
The exact prevalence of psoriatic arthritis in children is
not known as historically there has been a lack of uni-
versally accepted diagnostic criteria. Prevalence esti- Dyslipidaemia
mates vary widely and range from 5% to 40% [10]. In Emerging data suggest that dyslipidaemia is associated
recent years, the International League of Associations for with psoriasis not only in adults, but also in children. In a
Rheumatology (ILAR) criteria have become the diagnos- cross‐sectional study of 710 949 patients aged 2–19 years
tic standard for juvenile psoriatic arthritis. A diagnosis enrolled in Kaiser Permanente Southern California health
can be made in the presence of arthritis and psoriasis, or plan, including 1350 patients diagnosed with psoriasis,
arthritis coupled with a family history of psoriasis in a the mean total cholesterol, low‐density lipoprotein cho-
first‐degree relative, nail pitting or onycholysis, and/or lesterol, triglycerides and alanine aminotransferase were
dactylitis [24]. significantly higher in children with psoriasis compared
Cutaneous disease often precedes the onset of joint dis- to children without psoriasis after adjusting for BMI [31].
ease, although arthritis may be the initial manifestation in There is a lower high‐density lipoprotein level in paediat-
up to 15% of patients [3]. The peak onset of disease is ric patients with psoriasis compared to controls [32].
between the ages of 9 and 12 years [14]. Psoriatic arthritis Additionally, there is an association between psoriasis
is clinically heterogeneous, with asymmetrical oligoar- and higher apolipoprotein B concentrations, decreased
ticular arthritis of the hands and feet being the most large high‐density lipoprotein particles, and reduced
360 Section 5  Psoriasis

cholesterol efflux capacity [33]. These compositional and Table 29.1  Paediatric psoriasis comorbidity screening recommendations
functional abnormalities in lipoproteins reveal a tendency
towards atherogenesis even early in the course of disease. Psoriasis Screening recommendations
comorbidity

Diabetes
Arthritis Periodic history and physical examination to assess
The literature supports psoriasis as an independent risk for: (i) joint pain/swelling; (ii) joint stiffness after
factor for diabetes in adults. Further studies are needed to rest; (iii) dactylitis; and (iv) limp
determine whether this is also true in the paediatric popu- Overweight/obesitya Annual BMI starting at age 2 years
lation. In a small study of 20 children aged 9–17 years with Dyslipidaemia Universal lipid screen starting at age 9–11 and
moderate to severe psoriasis and 20 age‐ and sex‐matched 17–21 years
controls, patients with psoriasis had a statistically signifi- Additional/repeat testing if patient has other
personal or familial cardiovascular risk factors
cant higher mean fasting blood glucose of 91.1 mg/dL
Diabetes Overweight and ≥10 years old:
compared to 82.9 mg/dL [34]. However, no subject met the Fasting glucose every 2 years if patient has risk
fasting blood glucose requirement for diabetes. In a large factors for insulin resistance
cross‐sectional survey of 1.3 million patients aged 0–20 Obese and ≥10 years old:
years registered in a German health insurance organiza- Fasting glucose every 2 years
tion, of whom 33 981 carried a diagnosis of psoriasis, the Hypertension Annual blood pressure starting at age 3 years
Nonalcoholic fatty Overweight and ≥10 years old:
prevalence ratio of diabetes in those with psoriasis versus
liver disease AST/ALT every 2 years if patient has risk factors for
those without psoriasis was 2.01 [35]. As such, high‐risk insulin resistance
children should be considered for routine diabetes screen- Overweight and ≥10 years old:
ing (see Screening recommendations). AST/ALT every 2 years
SECTION 5: PSORIASIS

Mood disorders/ Annual screen for mood disorders and substance


Hypertension substance abuse abuse
Elevated blood pressure represents another component of
the metabolic syndrome, and may be associated with Source: [40–42].
a
 Overweight, BMI ≥85th and <95th percentile; obese, BMI ≥ 95th
childhood psoriasis. A prevalence ratio for hypertension
percentile.
of 1.89 in paediatric psoriasis patients compared to those ALT, alanine transaminase; AST, aspartate transaminase; BMI, body
without psoriasis has been reported [35]. Further studies mass index.
are needed to confirm whether psoriasis is an independent
risk factor for hypertension in children and adolescents.
Screening recommendations
Nonalcoholic fatty liver disease Routine monitoring of children with psoriasis and
There are multiple adult studies reporting the association screening for potential comorbidities will help identify
between psoriasis and nonalcoholic fatty liver disease problems at an early age when prevention of downstream
(NAFLD). To date, investigations in children are lacking. sequelae may be possible. The data available for comor-
Given the association of obesity and insulin resistance bidities of paediatric psoriasis suggest that age‐related
with NAFLD [36], children with these comorbidities screening as recommended by the American Academy of
should be screened for the development of liver disease. Paediatrics is sufficient, unless signs or symptoms arise
that point to the need for additional evaluation. These
Psychosocial recommendations are summarized in Table 29.1 [40–42].
Psoriasis can have profound psychosocial effects on chil-
dren and adolescents, regardless of the extent of disease. ­References
Patients commonly report a reduced quality of life as a 1 Raychaudhuri SP, Gross J. A comparative study of pediatric onset
psoriasis with adult onset psoriasis. Pediatr Dermatol 2000;17:174–8.
result of stigmatization and embarrassment, pruritus, 2 Benoit S, Hamm H. Childhood psoriasis. Clin Dermatol
sleep disturbance, interference with daily activities and 2007;25:555–62.
problems with treatment [37]. They are also at increased 3 Tollefson MM. Diagnosis and management of psoriasis in children.
Pediatr Clin North Am 2014;61:261–77.
risk for psychiatric disorders. In a retrospective study, 4 Cordoro KM. Management of childhood psoriasis. Adv Dermatol
Kimball et al. identified psoriasis as an independent risk 2008;24:125–69.
factor for psychiatric comorbidities in individuals less 5 Shah KN. Diagnosis and treatment of pediatric psoriasis: current and
than 18 years of age, and in particular found a 25–30% future. Am J Clin Dermatol 2013;14:195–213.
6 Lewkowicz D, Gottlieb AB. Pediatric psoriasis and psoriatic arthritis.
greater risk of depression and anxiety subsequent to diag- Dermatol Ther 2004;17:364–75.
nosis [38]. Patients with psoriasis were also more likely to 7 Rogers M. Childhood psoriasis. Curr Opin Pediatr 2002;14:404–9.
receive antidepressant and anxiolytic medications. A sep- 8 Morris A, Rogers M, Fischer G, Williams K. Childhood psoriasis: a
clinical review of 1262 cases. Pediatr Dermatol 2001;18:188–98.
arate retrospective cohort study by Kim et  al. revealed
9 Howard R, Tsuchiya A. Adult skin disease in the pediatric patient.
that a younger age at diagnosis of psoriasis seemed to be Dermatol Clin 1998;16:593–608.
associated with depression, social discrimination and 10 Bhutani T, Kamangar F, Cordoro KM. Management of pediatric pso-
higher lifetime Dermatology Life Quality Index (LT riasis. Pediatr Ann 2012;41:e1–7.
11 Honig PJ. Guttate psoriasis associated with perianal streptococcal dis-
DLQI) [39]. It is important to note that treatments we use ease. J Pediatr 1988;113:1037–9.
in daily clinical practice can positively impact the quality 12 Ko H‐C, Jwa S‐W, Song M et al. Clinical course of guttate psoriasis:
of life in juvenile psoriasis [37]. long‐term follow‐up study. J Dermatol 2010;37:894–9.
Chapter 29  Psoriasis: Clinical Features and Comorbidities 361

13 Mercy K, Kwasny M, Cordoro KM et al. Clinical manifestations of 28 Gutmark‐Little I, Shah KN. Obesity and the metabolic syndrome in
pediatric psoriasis: results of a multicenter study in the United pediatric psoriasis. Clin Dermatol 2015;33:305–15.
States. Pediatr Dermatol 2013;30:424–8. 29 Paller AS, Mercy K, Kwasny MJ et al. Association of pediatric pso-
14 Bronckers IMGJ, Paller AS, van Geel MJ et al. Psoriasis in children and riasis severity with excess and central adiposity: an international
adolescents: diagnosis, management and comorbidities. Paediatr cross‐sectional study. JAMA Dermatol 2013;149:166–76.
Drugs 2015;17:373–84. 30 Becker L, Tom WL, Eshagh K et al. Excess adiposity preceding pediat-
15 Neville EA, Finn OA. Psoriasiform napkin dermatitis‐a follow‐up ric psoriasis. JAMA Dermatol 2014;150:573–4.
study. Br J Dermatol 1975;92:279–85. 31 Koebnick C, Black MH, Smith N et al. The association of psoriasis and
16 Liao PB, Rubinson R, Howard R et al. Annular pustular psoriasis— elevated blood lipids in overweight and obese children. J Pediatr
most common form of pustular psoriasis in children: report of three 2011;159:577–83.
cases and review of the literature. Pediatr Dermatol 2002;19:19–25. 32 Au S, Goldminz AM, Loo DS et al. Association between pediatric psoria-
17 Hussain S, Berki DM, Choon S‐E et  al. IL36RN mutations define a sis and the metabolic syndrome. J Am Acad Dermatol 2012;66:1012–3.
severe autoinflammatory phenotype of generalized pustular psoria- 33 Tom WL, Playford MP, Admani S et al. Characterization of lipoprotein
sis. J Allergy Clin Immunol 2015;135:1067–1070.e9. composition and function in pediatric psoriasis reveals a more athero-
18 Marrakchi S, Guigue P, Renshaw BR et al. Interleukin‐36‐receptor genic profile. J Invest Dermatol 2016;136:67–73.
antagonist deficiency and generalized pustular psoriasis. N Engl J 34 Goldminz AM, Buzney CD, Kim N et al. Prevalence of the metabolic
Med 2011;365:620–8. syndrome in children with psoriatic disease. Pediatr Dermatol
19 Sugiura K. The genetic background of generalized pustular psoriasis: 2013;30:700–5.
IL36RN mutations and CARD14 gain‐of‐function variants. J 35 Augustin M, Glaeske G, Radtke MA et al. Epidemiology and comor-
Dermatol Sci 2014;74:187–92. bidity of psoriasis in children. Br J Dermatol 2010;162:633–6.
20 Inoue N, Dainichi T, Fujisawa A et al. CARD14 Glu138 mutation in 36 Nobili V, Alkhouri N, Alisi A et al. Nonalcoholic fatty liver disease: A
familial pityriasis rubra pilaris does not warrant differentiation from challenge for pediatricians. JAMA Pediatr 2015;169:170–6.
familial psoriasis. J Dermatol 2016;43:187–9. 37 Oostveen AM, de Jager ME, van de Kerkhof PC et al. The influence of
21 Aksentijevich I, Masters SL, Ferguson PJ et al. An autoinflamma- treatments in daily clinical practice on the Children’s Dermatology
tory disease with deficiency of the interleukin‐1–receptor antago- Life Quality Index in juvenile psoriasis: a longitudinal study from the
nist. N Engl J Med 2009;360:2426–37. Child‐CAPTURE patient registry. Br J Dermatol 2012;167:145–9.
22 Griffiths CE, Barker JN. Pathogenesis and clinical features of psoria- 38 Kimball AB, Wu EQ, Guérin A et al. Risks of developing psychiat-

SECTION 5: PSORIASIS
sis. Lancet 2007;370:263–71. ric disorders in pediatric patients with psoriasis. J Am Acad
23 Swary JH, Stratman EJ. Identifying performance gaps in comorbidity Dermatol 2012;67:651–7.e1–2.
and risk factor screening, prevention, and counseling behaviors of 39 Kim GE, Seidler E, Kimball AB. Effect of age at diagnosis on chronic
providers caring for children with psoriasis. Pediatr Dermatol quality of life and long‐term outcomes of individuals with psoriasis.
2015;32:813–8. Pediatr Dermatol 2015;32:656–62.
24 Stoll ML, Lio P, Sundel RP, Nigrovic PA. Comparison of Vancouver 40 Barlow SE. Expert Committee Recommendations Regarding

and International League of Associations for rheumatology classifi- the  Prevention, Assessment, and Treatment of Child and
cation criteria for juvenile psoriatic arthritis. Arthritis Rheum Adolescent Overweight and Obesity: Summary Report. Pediatrics
2008;59:51–8. 2007;120(suppl 4):S164–S192.
25 Stoll ML, Punaro M. Psoriatic juvenile idiopathic arthritis: a tale of 41 Expert Panel on Integrated Guidelines for Cardiovascular Health and
two subgroups. Curr Opin Rheumatol 2011;23:437–43. Risk Reduction in Children and Adolescents, National Heart, Lung,
26 Ferrándiz C, Pujol RM, García‐Patos V et al. Psoriasis of early and late and Blood Institute. Expert panel on integrated guidelines for cardio-
onset: a clinical and epidemiologic study from Spain. J Am Acad vascular health and risk reduction in children and adolescents: sum-
Dermatol 2002;46:867–73. mary report. Pediatrics 2011;128(suppl 5):S213–S256.
27 Southwood TR, Petty RE, Malleson PN et al. Psoriatic arthritis in chil- 42 Osier E, Wang AS, Cordoro K et al. Pediatric psoriasis comorbidity
dren. Arthritis Rheum 1989;32:1007–13. screening guidelines. JAMA Dermatol 2017;153:698–704.
362 

CHA PTER 3 0

Psoriasis: Classification, Scores and Diagnosis


Nirav Patel & Megha Tollefson
Departments of Dermatology and Pediatrics, Mayo Clinic, Rochester, MN, USA

Classification, 362 Scores, 363 Diagnosis, 363

Abstract a clinical scoring tool such as the Psoriasis Area and Severity Index
(PASI) and the Physician Global Assessment (PGA) can be used to
measure disease severity. The PGA has been validated in children
Psoriasis in the paediatric population can be classified into a vari-
and is easiest to use. Diagnosis is largely made on a clinical basis,
ety of morphologies. The plaque subtype is the most common, fol-
but skin biopsy, laboratory work and radiography may be indicat-
lowed by guttate psoriasis. Once a diagnosis has been established,
ed depending on the clinical scenario.

Key points scoring systems for paediatric psoriasis, with the PGA having
SECTION 5: PSORIASIS

validation for use in children.


• Diagnosis of psoriasis is almost always made clinically, but
• Psoriasis can be classified into a number of subtypes with plaque
histology and laboratory investigation can be helpful in
psoriasis being the most common.
confirming a diagnosis and aiding in evaluation of systemic
• Guttate and inverse psoriasis are seen more commonly in the
findings.
paediatric population compared to adults.
• The Psoriasis Area and Severity Index (PASI) and the Physician
Global Assessment (PGA) are the two most commonly used

Classification
Inverse or flexural psoriasis occurs more commonly in
Psoriasis in children may have a variety of morphologies. children than adults. Napkin psoriasis is a variant of this
As with adults, the plaque subtype is the most common and is a common presentation of psoriasis in children
[1–4]. Often the scalp and extremities are most commonly under the age of 2. A family history of psoriasis may be
involved, with finer scaling than is seen in adult plaque present in these patients [3,9–11]. This location poses
psoriasis [3]. challenges with treatment due to urine, stool and friction
Scalp psoriasis is often seen in children, and is more in the area, but often improves after potty training.
common in girls [1-3]. It can be an isolated finding or be Significant nail psoriasis can be seen in children, but
seen in conjunction with plaque psoriasis and may be is less common compared to adults [4,12], and its pres-
extremely symptomatic and challenging to treat but does ence does not correlate with psoriasis severity. Pure
not correlate with psoriasis severity [1]. nail‐only involvement is rare, and pitting is the most
Guttate psoriasis is the second most common form seen common finding followed by ridging and discoloura-
in children and is more commonly seen in the paediatric tion due to subungual hyperkeratosis. The fingernails
population as compared to adults [1–3]. Development of tend to have more involvement than toenails [4]. Nail
guttate psoriasis is often closely linked to a preceding involvement is more common in boys, suggesting koeb-
Streptococcus pyogenes infection causing pharyngitis or nerization as an aetiology, and in those with later onset
perianal dermatitis [2,5–7]; streptococcal infection may disease. An association of nail psoriasis with psoriatic
also reside in the inguinal folds. Guttate psoriasis may arthritis has not been observed in the paediatric popula-
resolve spontaneously or after clearance of infection, but tion [2,13].
some children may progress to plaque psoriasis. In those Pustular psoriasis is a rare variant of psoriasis and is
patients who progress to plaque type psoriasis after an seen even less frequently in the paediatric population
initial guttate presentation, a more severe phenotype may [1,3,4]. When it is seen in children, an annular morphol-
be seen as compared to patients who did not present with ogy is more common. Pustular psoriasis can be associated
a guttate morphology [2,3,8]. with streptococcal infection and may have a more benign

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 30  Psoriasis: Classification, Scores and Diagnosis 363

course than in adults [14–16]. The erythrodermic form of (BSA) and may also underestimate erythema in darker
psoriasis is similarly rare and presents with full‐body skin types. The PASI also does not factor in disease involv-
redness with or without the specific features of psoriasis ing the nails, acral surfaces, genitals or face [20,23,24].
[2,4]. Palmoplantar psoriasis is a form of acral psoriasis PASI scores correlate only moderately with quality of life
that is only occasionally seen in the paediatric population; (QoL) scores in children [22,23].
it is far more common in adults [3,12]. The PGA has multiple variations, but the most com-
Psoriatic arthritis, a rare comorbidity of psoriasis, par- monly used form is a five‐point scoring system assess-
ticularly in the paediatric population, is thought to have a ing plaque thickness, scale and degree of erythema.
bimodal peak of onset at age 2 and in puberty [17]. The PGA is inherently more simple and easier to use
Arthritis has not been linked to severity of skin disease compared to the PASI, and probably better suited for
[2]. Both linear and congenital psoriasis have also been tracking psoriasis outcomes outside the clinical trial
described as very rare variants [18,19]. setting. The PGA has been validated for use in chil-
dren [23,25]. Due to the lack of BSA in the score, a
patient may continue to have an unchanged score,
Scores even with improvement of BSA, if the character of the
Several scoring systems or clinical tools can be used to remaining plaques is unchanged [23]. Similar to the
measure disease burden in patients with psoriasis. The PASI, the PGA correlates only moderately with QoL
Psoriasis Area and Severity Index (PASI; Box 30.1) and the scores in children [21,22].
Physician Global Assessment (PGA) are the two best
studied and most commonly used tools [20]. The PASI is
more detailed than the PGA, is better studied and is Diagnosis

SECTION 5: PSORIASIS
the most commonly used tool in clinical trials. However, The diagnosis of psoriasis is almost always made clinically.
the PASI, although used in studies, is not validated in the Classic psoriasis in children is very similar in appearance
paediatric population [21,22]. to the adult counterpart with well‐defined, erythematous
It is therefore mainly used for older children and plaques with silvery scale. There are many variants and the
adolescents only. It is less sensitive for disease that is mild differential diagnosis of psoriasis is site and morphology
to moderate and involving minimal body surface area dependent (Table 30.1). If the correct diagnosis is in ques-
tion, a biopsy can be performed but may not always be
Box 30.1  The Psoriasis Area and Severity Index diagnostic. Classic histology demonstrates regular acan-
thosis, a diminished granular layer, parakeratosis, dilation
(a) Divide body into four areas: head, arms, trunk to groin, and legs of papillary dermal vessels, Munro microabscesses (collec-
to top of buttocks tions of neutrophils in the stratum corneum) and spongi-
(b) Generate an average score for the erythema, thickness and scale form pustules of Kogoj (neutrophilic pustules in the
for each of the four areas (0 = clear; 1–4 = increasing severity)a epidermis). If there is concern for an inciting factor or for
(c) Sum scores of erythema, thickness and scale for each area
comorbidities, laboratory testing and imaging may be per-
(d) Generate a percentage for skin covered with psoriasis for each area
and convert that to a 0–6 scale (0 = 0%; 1 = <10%; 2 = 10–<30%;
formed (Box 30.2). In patients suspected of having a strep-
3 = 30–<50%; 4 = 50–<70%; 5 = 70–<90%; 6 = 90–100%) tococcal infection, or in patients presenting with the guttate
(e) Multiply score of item (c) above times item (d) above for each form of psoriasis, investigation of antistreptolysin titre and
area and multiply that by 0.1, 0.2, 0.3, and 0.4 for head, arms, a throat, inguinal fold or perianal culture may be appropri-
trunk and legs, respectively ate [7]. Patients with generalized pustular psoriasis should
(f) Add these scores to get the PASI score be evaluated for leucocytosis with neutrophilia, renal or
hepatic impairment, disturbed electrolyte balance and low
a
 Erythema, induration and scale are measured on a 0–4 scale (none,
calcium. If psoriatic arthritis is suspected, imaging of the
slight, mild, moderate, severe).
Source: Feldman 2005 [24]. Reproduced with permission from BMJ Pub-
affected joints is appropriate as well as a set of parameters
lishing Group Ltd. to rule out other causes of joint involvement (e.g. infection,
autoimmune diseases) [26].
364 Section 5  Psoriasis

Table 30.1  Cutaneous psoriasis variants

Type Age Clinical findings Differential diagnosis

Chronic plaque Any Well‐defined erythematous plaques, Nummular eczema


silvery white scale Atopic dermatitis
Auspitz sign Allergic contact dermatitis
Extensor surfaces, trunk, periumbilical Tinea corporis
May be follicular or annular Pityriasis rubra pilaris
SECTION 5: PSORIASIS

Scalp Any Well‐defined erythematous scaly Seborrhoeic dermatitis


plaques Tinea capitis
Tinea amiantacea Atopic dermatitis
Often extends onto forehead

Guttate Children, Small, “drop‐like” <1 cm Pityriasis rosea


adolescents, erythematous scaly papules Tinea corporis
young adults Trunk, extremities, face, scalp Pityriasis rubra pilaris
Nummular eczema

(Continued)
Table 30.1  Continued

Type Age Clinical findings Differential diagnosis

Inverse Younger children Erythematous plaques with little scale, Intertrigo


possible maceration Seborrhoeic dermatitis
Flexural areas and face Erythrasma
Contact dermatitis

Napkin Ages 0–2 Well‐defined bright red plaques in Irritant contact dermatitis
napkin area Allergic contact dermatitis
Little scale, may be macerated Intertrigo

SECTION 5: PSORIASIS
Involves inguinal folds Candidal napkin dermatitis
Acrodermatitis enteropathica

Nail Any age, more Pitting, onycholysis, oil spots, Onychomycosis


common in subungual hyperkeratosis, Pityriasis rubra pilaris
older children trachyonychia Lichen planus
and adults May or may not have skin signs of
psoriasis

(Continued)
Table 30.1  Continued

Type Age Clinical findings Differential diagnosis

Pustular Older children and Superficial sterile pustules on Candida infection


adults erythematous skin Dyshidrotic eczema
May have annular configuration Staphylococcal scalded skin
May be localized to palmoplantar syndrome
areas Blistering dactylitis
Tinea infection

Erythrodermic Any Generalized erythema Minimal scale Staphylococcal scalded skin


Very rare syndrome
Pityriasis rubra pilaris
Cutaneous T‐cell lymphoma
Atopic dermatitis
SECTION 5: PSORIASIS

Congenital At birth Any of the above morphologies Atopic dermatitis


Seborrhoeic dermatitis
Bacterial infection
Candida infection
Dermatophyte infection
Langerhans cell histiocytosis
Erythema toxicum
Transient neonatal pustular
melanosis
Immunodeficiency syndromes
Ichthyosis

Source: Adapted from Tollefson 2014 [11].

Box 30.2  Psoriasis investigation

Physical parameters Radiology

• Height, weight, body mass index, blood pressure in all patients • Ultrasound and/or radiographs of affected joints in those suspected
to have psoriatic arthritis
Laboratory investigations
Histology
• Guttate – culture swab from appropriate location for group A
Streptococcus, ASL • Consider skin biopsy in cases of uncertain diagnosis or atypical presentation
• Pustular, generalized – full blood count, CRP, LFTs
ANA, anti‐nuclear antibodies; ASL, antistreptolysin; CRP, C‐reactive
• Psoriatic arthritis – ESR, CRP, RF, ANA protein; ESR, erythrocyte sedimentation rate; LFTs, liver function tests;
• Concern for metabolic syndrome – fasting blood glucose and lipid RF, rheumatoid factor.
profile, blood pressure measurement
Source: Adapted from Tollefson 2014 [11].
Chapter 30  Psoriasis: Classification, Scores and Diagnosis 367

References 15 Liao PB, Rubinson R, Howard R et  al. Annular pustular psoria-
1 Tollefson MM, Crowson CS, McEvoy MT, Maradit Kremers H. sis—most common form of pustular psoriasis in children: report
Incidence of psoriasis in children: a population‐based study. J Am of three cases and review of the literature. Pediatr Dermatol
Acad Dermatol 2010;62:979–87. 2002;19:19–25.
2 Mercy K, Kwasny M, Cordoro KM et al. Clinical manifestations of 16 Farber EM, Nall L. Childhood psoriasis. Cutis 1999;64:309–14.
pediatric psoriasis: results of a multicenter study in the United 17 Stoll ML, Zurakowski D, Nigrovic LE et al. Patients with juvenile
States. Pediatr Dermatol 2013;30:424–8. psoriatic arthritis comprise two distinct populations. Arthritis
3 Morris A, Rogers M, Fischer G, Williams K. Childhood psoriasis: a Rheum 2006;54:3564–72.
clinical review of 1262 cases. Pediatr Dermatol 2001;18:188–98. 18 Lehman JS, Rahil AK. Congenital psoriasis: case report and litera-
4 Kumar B, Jain R, Sandhu K et al. Epidemiology of childhood psoria- ture review. Pediatr Dermatol 2008;25:332–8.
sis: a study of 419 patients from northern India. Int J Dermatol 19 Seitz CS, Garbaravičienė J, Bröcker EB, Hamm H. Superimposed lin-
2004;43:654–8. ear psoriasis: differential therapeutic response of linear and nonlinear
5 Patrizi A, Costa AM, Fiorillo L, Neri I. perianal streptococcal derma- lesions. Clin Exp Dermatol 2009;34:e177–9.
titis associated with guttate psoriasis and/or balanoposthitis: a 20 Puzenat E, Bronsard V, Prey S et  al. What are the best outcome
study of five cases. Pediatr Dermatol 1994;11:168–71. measures for assessing plaque psoriasis severity? A systematic
6 Honig PJ. Guttate psoriasis associated with perianal streptococcal dis- review of the literature. J Eur Acad Dermatol Venereol 2010;24
ease. J Pediatr 1988;113:1037–9. (suppl 2):10–6.
7 Telfer NR, Chalmers RJG, Whale K, Colman G. The role of strepto- 21 Schäfer I, Hacker J, Rustenbach SJ et  al. Concordance of the
coccal infection in the initiation of guttate psoriasis. Arch Dermatol Psoriasis Area and Severity Index (PASI) and patient‐reported out-
1992;128:39–42. comes in psoriasis treatment. Eur J Dermatol 2010;20:62–7.
8 Ko HC, Jwa SW, Song M et  al. Clinical course of guttate psoriasis: 22 De Jager MEA, van de Kerkhof PCM, de Jong EMGJ, Seyger MMB.
Long‐term follow‐up study. J Dermatol 2010;37:894–9. A cross‐sectional study using the Children’s Dermatology Life
9 Beylot C, Puissant A, Bioulac P et al. Particular clinical features of Quality Index (CDLQI) in childhood psoriasis: negative effect on
psoriasis in infants and chidren. Acta Derm Venereol Suppl quality of life and moderate correlation of CDLQI with severity
(Stockh) 1979;87:95–7. scores. Br J Dermatol 2010;163:1099–101.
10 Hutton KP, Orenberg EK, Jacobs AH. Childhood psoriasis. Cutis 23 Robinson A, Kardos M, Kimball AB. Physician Global Assessment
1987;39:26–7. (PGA) and Psoriasis Area and Severity Index (PASI): why do both? A

SECTION 5: PSORIASIS
11 Tollefson MM. Diagnosis and management of psoriasis in chil- systematic analysis of randomized controlled trials of biologic agents
dren. Pediatr Clin North Am 2014;61:261–77. for moderate to severe plaque psoriasis. J Am Acad Dermatol
12 Nanda A, Kaur S, Kaur I, Kumar B. Childhood psoriasis: an epide- 2012;66:369–75.
miologic survey of 112 patients. Pediatr Dermatol 1990;7:19–21. 24 Feldman SR. Psoriasis assessment tools in clinical trials. Ann
13 Mallbris L, Larsson P, Bergqvist S et al. Psoriasis phenotype at disease Rheum Dis 2005;64(suppl 2):ii65–8.
onset: clinical characterization of 400 adult cases. J Invest Dermatol 25 Pascoe VL, Enamandram M, Corey KC et  al. Using the Physician
2005;124:499–504. Global Assessment in a clinical setting to measure and track patient
14 Cassandra M, Conte E, Cortez B. Childhood pustular psoriasis elic- outcomes. JAMA Dermatol 2015;151:375–81.
ited by the streptococcal antigen: a case report and review of the lit- 26 Mease PJ. Psoriatic arthritis: update on pathophysiology, assess-
erature. Pediatr Dermatol 2003;20:506–10. ment and management. Ann Rheum Dis 2011;70(suppl 1):i77–84.
368 

CHA PTER  3 1

Psoriasis: Management
Marieke M.B. Seyger
Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands

Introduction, 368 Systemic therapies, 370


Topical treatments, 368 Summary, 374

Abstract this age group in particular, long‐term safety of treatments is


crucial and one of the most important concerns for dermatologists
The management of paediatric psoriasis is challenging as many treating children and adolescents with psoriasis.
treatments are not approved for use in this age group, and In this chapter, the efficacy and safety of all available treatments
evidence on efficacy and safety of most treatments is limited. In for paediatric psoriasis are presented, including some practical
addition, international evidence‐based guidelines are lacking. Even advice. Based on the available evidence, a treatment algorithm
mild forms of psoriasis may exert a significant burden of morbidity, is proposed that can serve as general guidance for the treating
and therefore adequate and effective treatment is important. In physician.
SECTION 5: PSORIASIS

Key points • Narrow‐band UVB (NB‐UVB) should be used cautiously in the


paediatric population, particularly in young and fair‐skinned
children.
• Topical corticosteroids and/or vitamin D analogues or a
• Methotrexate is the conventional systemic drug of choice
compound formulation are standard therapeutic agents for
for children and adolescents with moderate to severe plaque
topical therapy.
psoriasis.
• If available, dithranol in a daycare setting should be commenced
• The biologics represent a promising therapeutic alternative to
before systemic treatments are considered.
conventional systemic treatment options.

Introduction are influenced by many factors such as age, severity,


l­ocation and type of psoriasis, previous treatments, QoL,
In almost one third of cases the onset of psoriasis is before practicality of the regimen, costs, accessibility, benefits
adulthood [1,2]. The management of paediatric psoriasis and risks of individual treatments. However, in this
is challenging, as many treatments are not approved for ­vulnerable age group, long‐term safety is one of the most
use in this age group, and evidence on efficacy and safety important concerns for all dermatologists treating
of most treatments is limited [3]. In addition, international ­children with psoriasis [8].
evidence‐based guidelines are lacking [4].
Another challenge in the treatment of paediatric psoria-
sis is adherence, which is especially a problem in adoles-
Topical treatments
cents and even more so when topical agents are used.
Therefore, education of patients and family members The majority of children with psoriasis can be managed
about the chronicity of the disease and the rationale of the with topical treatments. Corticosteroids, vitamin D ana-
treatment is important, as is taking patient preferences logues, calcineurin inhibitors and dithranol are used for
into consideration in selecting a treatment [5]. Timing of the treatment of mild to moderate paediatric psoriasis.
the first return visit may also be a practical tool to enhance Topical corticosteroids are generally considered to be first
patient adherence [6]. choice [9], although others prefer to start with vitamin D
Paediatric psoriasis has a negative impact on the qual- analogues [10]. In order to increase efficacy and decrease
ity of life (QoL), self‐esteem and psychosocial develop- potential side‐effects, it is advised to use treatments inter-
ment of those affected, and treatments can positively mittently, preferably in a combination of treatments (e.g.
influence QoL [7]. Even mild forms of psoriasis may exert topical corticosteroid 4 days per week combined with a
a significant burden of morbidity, and therefore adequate vitamin D analogue 3 days per week). As adjunctive
and effective treatment is important. Treatment choices agents, emollients can be used to reduce scaling, dryness

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 31  Psoriasis: Management 369

and itch. Coal tar preparations are one of the oldest combination with topical corticosteroids is necessary to
­treatment modalities for psoriasis, but due to lack of evi- achieve maximal effect. An often used regimen is the ‘week-
dence on effectiveness, unpleasant side‐effects (smell, end scheme’, where corticosteroids are used on weekend
staining of clothes) and concern among some physicians days, and vitamin D analogues the other days [14].
about contamination with polyaromatic compounds are A commercially available compounded formulation con-
now less and less frequently used [10]. The most common taining calcipotriol and betamethasone dipropionate has
topical treatments for paediatric psoriasis, which are been developed. Recently, four studies on the efficacy and
largely not approved for use in children and adolescents, safety of calcipotriol/betamethasone dipropionate oint-
are reviewed here. ment and gel in children and adolescents with psoriasis
were performed [15–18]. The two‐compound formulation
Pretreatment was found to be effective in all four studies, with mild
In order to permit access of active ingredients to inflamed adverse effects. In the two daily clinical practice studies
skin, intense scaling must be addressed first. Emollients with a 48‐week follow‐up, striation of the skin was reported
are used to decrease scaling and dryness and are also in up to 5% of patients. It is, however, questionable whether
used to decrease pruritus. Daily application of an these striae were steroid‐induced, as those patients were
­emollient (ointment) is advised. In addition, keratolytics adolescents (pubertal growth) and/or obese [15,16]. In both
(salicylic acid, lactic acid, urea) are helpful in removing phase II, prospective, open‐label, multicentre, single‐arm,
layers of thick scale. Salicylic acid should be avoided in 8‐week studies, no serious adverse events, including clini-
infants and children aged under 2 years because of the cally relevant changes in calcium metabolism, were
risk of percutaneous salicylism, and many restrict usage observed [17,18]. Daily application should be limited to
to children over 6 years of age [6]. 4 weeks, followed by alternation of the compound formula-

SECTION 5: PSORIASIS
tion at weekends with vitamin D analogues on weekdays.
Topical corticosteroids
Topical corticosteroids are the most commonly prescribed Calcineurin inhibitors
medication and are generally first‐line for the treatment of Tacrolimus (0.03% and 0.1% ointment) and pimecrolimus
active plaques in paediatric psoriasis [9]. However, evi- 1% cream are only licensed for use in atopic dermatitis in
dence on the efficacy and safety of topical corticosteroids children. A few studies show beneficial effects of calcineu-
in this specific patient group is limited. In a few small rin inhibitors (especially tacrolimus 0.1%) in the treat-
studies, topical corticosteroids have been shown to be effi- ment of intertriginous and facial psoriasis in paediatric
cacious, with relatively mild side‐effects [4,11]. Potential patients. Apart from pruritus, adverse effects were not
side‐effects include local telangiectasias, a­ trophy, striae of reported. Generally, it is advised to avoid combining
the skin and, if used extensively, s­ uppression of the hypo- ­calcineurin inhibitors with phototherapy or extreme sun
thalamic–pituitary–adrenal axis. Therefore corticosteroids exposure due to a possibly increased risk of ultraviolet
should be used with caution through intermittent and (UV) light‐related skin tumours [4,19].
rotational application [2,6]. High‐potency corticoster-
oids should be avoided in flexural areas, whereas ultra- Dithranol (anthralin, cignolin)
potent steroid ointment should only be used for a short Dithranol treatment for psoriasis has a long history (over
period, preferably restricted to the palms and soles. In 100 years) and is therefore one of the oldest and safest
the majority of cases, however, a modern class III topical topical antipsoriatic treatments [14,20]. Only a few stud-
steroid (e.g. mometasone) is ­ effective and sufficient ies have described the efficacy and safety of dithranol in
for  the initial treatment of most cases of paediatric the treatment of paediatric psoriasis [21–23]. In these
psoriasis. studies, dithranol was shown to be very effective with
good or excellent results achieved in most patients. In a
Vitamin D analogues and combination prospective observational study, a reduction in Psoriasis
with corticosteroids Area and Severity Index (PASI) score of 69.3% was found
Calcipotriol (calcipotriene in the USA) and calcitriol are using short‐contact dithranol cream [21]. Short‐contact
vitamin D3 analogues that are effective and reasonably dithranol cream at increasing concentrations (between
well tolerated treatment options for children with plaque‐ 0.01% and 4%) and application periods between 15 and 45
type psoriasis [4]. The most common side‐effects are minutes (until irritation develops) maintains efficacy but
localized skin irritation and pruritus, particularly when diminishes the risk of irritation and staining [2]. It
used on the face and flexural areas. However, calcitriol involves daily treatment with a median duration of
has been found to be less irritating than calcipotriol if 2 months. For adequate instructions and to avoid unnec-
used in intertriginous psoriasis [2]. To prevent the unlikely essary permanent staining of any object, it is preferable to
event of hypercalcaemia, it is important to confine the carry out some of the applications in a daycare setting.
application area to 30% of the body surface, not exceeding Visits to a daycare setting (once a week) can be combined
the maximum recommended dose of 45 g/week per m2 with telemedicine consultations (once a week) without
[12,13]. If larger areas are affected, calcipotriol must losing efficacy or safety [21]. Irritation and transient stain-
be used on alternating areas. Vitamin D analogues can be ing of the skin are the most often mentioned side‐effects.
used as monotherapy in very mild cases with thin A detailed protocol for the use of short‐contact dithranol
patches and plaques. In the majority of patients, however, cream can be downloaded at www.radboudumc.nl/
370 Section 5  Psoriasis

dithranol. As long‐term safety is important in the treatment


Box 31.1  Indications for systemic treatment in paediatric
of paediatric psoriasis, this valuable alternative topical
psoriasis
treatment should not be disregarded in the treatment
­regimen of paediatric psoriasis. If possible, it should be Paediatric ‘Rule of tens’
commenced before systemic treatments are considered [22]. • PASI ≥10
and/or
Phototherapy • BSA ≥10
Due to concerns about potential long‐term side‐effects, and/or
UV therapy in pre‐adolescents should be avoided wher- • CDLQI ≥10
ever possible. In adolescents, UVB is the preferred form of
BSA, body surface area; CDLQI, Children’s Dermatology Life Quality
phototherapy, with narrow‐band UVB (NB‐UVB, 311– Index; PASI, Psoriasis Area and Severity Index.
313 nm) being more effective than broad‐band and less
erythrogenic [24]. The combination of psoralens and UVA
(PUVA) is contraindicated for the treatment of children also gives the opportunity to start a systemic treatment in
and will therefore not be discussed in this chapter [10,20]. a child with less severe psoriasis but severely affected
As potential long‐term carcinogenic effects of NB‐UVB QoL. The CDLQI and the validated cartoon version of the
have not been assessed, it should be used cautiously in CDLQI for younger children are available in a variety of
the paediatric population, particularly in young and languages and can be downloaded from the website
in fair‐skinned children [4,10,20]. It can be considered in of  Cardiff University [http://www.cardiff.ac.uk/
adolescents who have failed topical treatment, especially dermatology/quality‐of‐life/childrens‐dermatology‐life‐
in patients who cannot receive systemic therapy for their quality‐index‐cdlqi/cdlqi‐information‐and‐instructions/).
SECTION 5: PSORIASIS

moderate to severe psoriasis [25]. See also Chapters 16 and 30.


The last few years, evidence has accumulated that NB‐
UVB demonstrates potential benefit in the treatment of Methotrexate
plaque and guttate paediatric psoriasis [25]. In several Methotrexate (MTX) has been used to treat (adult) psoriasis
studies, half of the treated patients received more than for decades and also has a long track record of safe use in the
90% improvement of their PASI scores whereas no treatment of juvenile idiopathic arthritis [30]. MTX has the
improvement was found in approximately 10% of advantage over ciclosporin, fumaric acid and retinoids of
patients. Most studies were performed in adolescents, clearing or improving psoriatic arthritis. Despite the fact that
and the majority had Fitzpatrick skin phototype III or IV MTX is probably the most widely prescribed conventional
[25,26]. Short‐term side‐effects of phototherapy include systemic drug for the treatment of paediatric psoriasis, the
erythema, xerosis, pruritus, blistering, photoactivation of evidence on its safety and efficacy in this particular patient
herpes virus and anxiety. Long‐term side‐effects include group is limited. Until recently, all ­evidence was derived
premature photoageing and carcinogenesis [25]. from retrospectively collected case series [3]. Two studies
presented prospective data on MTX use in paediatric plaque
psoriasis. A single‐group, prospective, observational daily
Systemic therapies clinical practice study showed ≥75% improvement in PASI
Systemic therapies can be divided into conventional (PASI75) in 33.3% of patients at week 24 with a maximum
­treatments and the newer ‘biologic’ treatment options. MTX dose between 0.14 and 0.63 mg/kg once weekly [31].
The best known conventional systemic treatments for More or less similar effectiveness data were found in a pro-
paediatric psoriasis are methotrexate, ciclosporin (cyclo- spective, multicentre, randomized, double‐blind trial inves-
sporine), retinoids and fumaric acid esters. To date, the tigating the safety and efficacy of MTX versus two doses of
biologics etanercept, adalimumab and ustekinumab are adalimumab [32]. In this trial PASI75 was achieved in 32.4%
approved for use in children and adolescents with psoria- of patients treated with MTX (0.1–0.4 mg/kg up to 25 mg/
sis by the European Medicines Agency (EMA). Systemic week) at week 16. MTX is usually dosed once weekly at a
therapies should only be used in paediatric patients with dose of 10–15 mg/m2 BSA [20]; less commonly, the dose is
moderate to severe psoriasis that does not respond to top- adapted according to bodyweight (0.2–0.4 mg/kg/week)
ical treatments, including dithranol, if available. The ‘rule (see Box  31.2). Approximately 3 months after therapeutic
of tens’ [27], used in adults to indicate moderate to severe control is achieved, tapering to the minimum effective dose
psoriasis, can also be applied in paediatric psoriasis as an is ­recommended to minimize possible side‐effects [19]. The
indication for the use of systemic therapy [10] (Box 31.1). most common side‐effects reported in children and
This modified ‘paediatric rule of tens’ would be PASI ≥10 ­adolescents are nausea, vomiting, transient elevation of liver
and/or body surface area (BSA) ≥10 and/or CDLQI ≥10. enzymes and fatigue. Bone marrow suppression, pulmo-
Because paediatric psoriasis can have a significant impact nary toxicity, infections and hepatotoxicity including liver
on QoL, it is advised that the CDLQI be used in daily fibrosis have only rarely been reported in children.
­clinical practice [28]. This commonly used dermatology‐ Laboratory testing on a regular basis is recommended [2].
specific QoL questionnaire is validated for children Addition of folic acid supplementation helps protect against
between 4 and 16 years. It is a simple, self‐administered side‐effects without significant alteration of ­efficacy [33,34].
10‐item questionnaire [29] which not only provides more It is still a subject of debate whether different folic acid
insight into the QoL of paediatric psoriasis patients but administration regimens (ranging from 1–5 mg daily to
Chapter 31  Psoriasis: Management 371

­ aediatric psoriasis. The majority of this evidence comes


p
Box 31.2  Recommendations for methotrexate (MTX) therapy
from three recently published retrospective studies, in
• Dosage: once weekly 10–15 mg/m2 body surface area or which ciclosporin was reported to be effective and safe
0.2–0.4 mg/kg/week [38–40]. Only one study used PASI as a severity measure
• Folic acid: 1 mg 6 days/week avoiding MTX day, or 5 mg 24 h after and reported achievement of PASI75 in 39.5% of patients
MTX at week 16, whereas 39.5% were non‐responders [39].
• Monitoring: Dosages range between 2 and 5 mg/kg/day in two sepa-
Pretreatment: complete blood count, liver enzymes, serum rate doses, with a gradual tapering after improvement.
creatinine, hepatitis A/B/C, (varicella) Rebounds or relapses after tapering of the dose are occa-
Follow‐up (week 1, 3, 5, 8, 12 and every 3 months): blood sionally seen [19].
count, liver enzymes
Commonly mentioned side‐effects are increase in
• Tapering down is advised: after clearance with steps of 2.5 mg
serum creatinine level, hypertension, hypertrichosis,
every 6–8 weeks
headache, gingival hyperplasia and gastrointestinal dis-
turbances [19,39]. Close monitoring of blood pressure and
5–10 mg once a week, 24 h after MTX administration) can renal function is required. In one retrospective study in
influence the occurrence of side‐effects. Administration of paediatric psoriasis, ciclosporin was stated to be the treat-
1 mg folic acid supplementation 6 days a week, avoiding the ment with the highest rate of severe side‐effects requiring
MTX day, seems favourable over once weekly 5 mg with termination of treatment of all systemic treatments [38].
respect to gastrointestinal side‐effects [35]. Vomiting and In particular, nephrotoxicity, immunosuppression and the
nausea can be controlled by changing the route of adminis- potential risk of developing malignancies are of some
tration (from oral to subcutaneous or intramuscular) and/or concern in children [8].

SECTION 5: PSORIASIS
by adding antiemetics [36]. In most instances, ciclosporin is considered an ideal
drug for the control of unstable disease, because of its
Retinoids rapid onset of action [19]. It seems to be especially ­effective
In a systematic evidence‐based review on systemic treat- in erythrodermic, pustular and palmoplantar psoriasis
ments in paediatric psoriasis, retinoids were found to be [38,41]. It is recommended that ciclosporin be reserved for
primarily effective in pustular and erythrodermic psoria- use in carefully selected children, and the lowest possible
sis [3]. This was recently confirmed by a retrospective dose and shortest treatment period should be used [6,42].
study in 18 children with pustular psoriasis [37]. However,
a large French retrospective study also showed good Fumaric acid esters
(clearance of >75% of lesions) or partial (clearance of Fumaric acid esters (FAEs) are used in various European
50–75%) response to acitretin in 78 children with plaque countries for the treatment of psoriasis in adults.
psoriasis [38]. Doses of acitretin ranging from 0.5 to 1 mg/ Skilarence is indicated for the treatment of moderate to
kg/day are considered appropriate, but retinoid dose severe plaque psoriasis in adults in need of systemic
should be kept as low as possible to limit side‐effects, medicinal therapy. Data on the efficacy and safety of FAEs
particularly mucocutaneous dryness. Conversely, dose
­ in children and adolescents are scarce and consist of a few
reduction may lead to inadequate efficacy [8] and hence case reports, retrospective case series and one small pro-
discontinuation of therapy. Frequently reported short‐ spective case series [3,43,44]. In a large retrospective
term side‐effects of acitretin are cheilitis, pruritus, hair German multicentre study in 127 patients, of whom 75.6%
loss and skin fragility [4]. Other reported adverse effects had plaque psoriasis, PASI was available in 59 patients at
are blepharoconjunctivitis, cataracts, myalgias and baseline and improved from 17.3 (mean) at baseline to 9.0
arthralgias. Blood monitoring is necessary to detect eleva- at 3 months and 7.7 at 6 months. In more than 80% of
tion of liver transaminases and triglycerides. Long‐term patients in this study, FAEs were the first systemic treat-
use of retinoids in higher dosages (>1 mg/kg/day) has ment used [44]. In a prospective case series of 14 patients,
been associated with hyperostosis resembling diffuse PASI improved from 10.5 (mean) at baseline to 8.6 at week
idiopathic skeletal hyperostosis, premature epiphyseal
­ 12 and 4.9 at week 24. These patients had recalcitrant
closure, and ligamentous calcification. However, ­evidence plaque psoriasis, with the majority being treated with
linking radiological skeletal abnormalities to long‐term, other systemic treatments before commencing FAEs [43].
low‐dose acitretin is conflicting, and abnormalities are In general, dosing of FAEs in the paediatric population is
likely to be rare if interval therapy is applied [8]. Retinoids according to the standardized progressive dose regimen
are known to be potent teratogens, with a delayed applied in adult patients, increasing the daily dose (30 mg)
­clearance of 3 years. It is advised to reserve retinoids for by one tablet per week. At week 4, an increase of one tab-
short‐term treatment of especially pustular or erythroder- let of 120 mg per week is started, to a maximum daily
mic psoriasis in infants and male adolescents [4]. dose of six tablets (720  mg/day), based on clinical
response and tolerability [45].
Ciclosporin Gastrointestinal complaints, flushing, lymphocytope-
Another alternative for the systemic treatment of psoria- nia, eosinophilia and increased liver enzymes were the
sis in children and adolescents is ciclosporin. In the past most common reported side‐effects of FAEs [43,45].
few years, new evidence has become available on the There is an increased concern about the possibility
efficacy and safety of ciclosporin in the treatment of
­ of FAE‐induced lymphocytopenia and manifestation of
372 Section 5  Psoriasis

­rogressive multifocal leucoencephalopathy (PML)


p treatments in paediatric psoriasis is warranted. If
which has been described in adults [46,47]. Therefore, it t­ reatment with biologics of a child with severe psoriasis
is extremely important to closely monitor laboratory is being considered, it is important to discuss the benefits
abnormalities and follow the recommendations from and risks of this treatment in a balanced way with the
the updated European S3‐guideline on FAE‐induced child and its caregivers.
leucocytopenia and/or lymphocytopenia [48].
Etanercept
Biologics Etanercept was approved by the EMA in 2009 for the
The biologics etanercept, adalimumab and ustekinumab treatment of children ≥6 years who are suffering from
are approved for use in paediatric psoriasis by the EMA severe, chronic plaque psoriasis refractory to or intolerant
(Table  31.1). In contrast, the US Food and Drug of other systemic agents or phototherapy [55]. The US
Administration (FDA) has only approved etanercept FDA approved this drug for children ≥4 years in 2016. The
and ustekinumab for use in children and adolescents. dose regimen of this subcutaneously administered drug
They represent a promising therapeutic alternative to is 0.8 mg/kg to a maximum of 50 mg once weekly or
conventional treatment options. Several randomized 0.4 mg/kg twice weekly [56].
controlled trials have been published recently, with high The majority of evidence on the efficacy and safety of
efficacy rates and reassuring short‐term safety profiles. etanercept in the treatment of paediatric psoriasis comes
In addition, biologics offer the convenience of less fre- from a phase III double‐blind randomized controlled trial
quent ­dosing and laboratory monitoring than the con- comparing etanercept 0.8 mg/kg weekly to placebo in 211
ventional systemic treatments. However, these results patients aged 4–16 years. At week 12, 57% of patients
need to be balanced against the considerable financial treated with etanercept reached PASI75, compared to 11%
SECTION 5: PSORIASIS

costs and, more importantly, the relative lack of long‐ of the placebo‐treated patients. A PASI90 response was
term safety data in this indication [3]. Paradoxical TNF‐ reached in 27% of etanercept‐treated patients, compared
inhibitor‐associated psoriasis has also been reported as a to 7% in the placebo group [56]. In a 5‐year, open‐label
complication of these therapies among both children extension study, efficacy rates were maintained in those
and adults, and may in some cases require substitution who remained in the study for up to 264 weeks [57]. The
of alternative treatment options [49,50]. most common side‐effects included infections such as
An increased risk of lymphoma and other malignan- upper respiratory tract infections, nasopharyngitis and
cies in children and adolescents with arthritis, inflamma- headache. No deaths, malignancies or opportunistic
tory bowel disease or sarcoidosis treated with TNF‐α infections were reported, and in the 5‐year follow‐up no
inhibitors is hypothesized [42,51,52]. However, a clear new safety concerns were observed [57].
causal relationship has not been established because the
contribution of underlying illnesses and concomitant use Adalimumab
of other immunosuppressant agents to the risk of malig- In the EU, subcutaneous adalimumab is indicated for the
nancies is still not fully characterized [53,54]. To date, treatment of severe chronic plaque psoriasis in children
there have not been any reports of malignancies in paedi- and adolescents from 4 years of age who have had an
atric psoriasis patients treated with biologics. In the inadequate response to or are inappropriate candidates
future, as the evidence on the long‐term safety of for topical therapy and phototherapies. In children and
­biologics in paediatric psoriasis increases, there might be adolescents, the recommended dosage is 0.8 mg/kg (up
an expanding role for the use of biologic agents in the to a maximum of 40 mg per dose) weekly for the first two
treatment of this population. Until then, more long‐term doses and every other week thereafter [58].
safety data are indispensable and a prospective, interna- Apart from a few case reports, the evidence on efficacy
tional registry on the safety and efficacy of systemic and safety of adalimumab in paediatric psoriasis has only

Table 31.1  EMA‐approved biologics in paediatric psoriasis [55,58,59]

Indication Age Dosage Interval

Etanercept Severe chronic plaque psoriasis refractory to or ≥6 years 0.8 mg/kg Once weekly
intolerant of other systemic agents or phototherapy (max 50 mg/dose)
Adalimumab Patients with severe chronic plaque psoriasis who have ≥4 years 0.8 mg/kg First two doses weekly, then every
had an inadequate response to or are inappropriate (max 40 mg/dose) other week (week 0, 1, 3, 5, etc.)
candidates for topical therapy and phototherapies
Ustekinumab Patients with moderate to severe plaque psoriasis who ≥12 years <60 kg: 0.75 mg/kg Week 0, 4, then every 12 weeks
are inadequately controlled by or intolerant to other ≥60 kg to ≤100 kg: 45 mg
systemic therapies or phototherapies >100 kg: 90 mg

Monitoring of biologics:
•  Pre‐treatment: complete blood count, liver enzymes, serum creatinine, C‐reactive protein, hepatitis A/B/C, varicella, PPD and/or IGRA, chest X‐ray, and
updated vaccinations. HIV and pregnancy test only if at risk/childbearing age.
•  Follow‐up (week 4, 12, and every 3–6 months): complete blood count, liver enzymes. PPD or IGRA annually.
IGRA, interferon‐gamma release assay; PPD, purified protein derivative.
Chapter 31  Psoriasis: Management 373

recently been described in the literature. In a randomized, 21.6 %) or PASI100 response (18.4 vs. 2.7%). The most
double‐blind, multiperiod study, children and adoles- f­ requently reported adverse events were infections; rates
cents with severe plaque psoriasis were randomly were similar for adalimumab and methotrexate (51.9% vs.
assigned 1 : 1 : 1 to receive adalimumab (0.8 mg/kg up to 54.1%, respectively) [32].
40 mg or 0.4 mg/kg up to 20 mg) at week 0, then every
other week from week 1, or methotrexate weekly (0.1– Ustekinumab
0.4 mg/kg up to 25 mg/week thereafter) for 16 weeks Ustekinumab is approved by both the EMA and the US
[32]. Overall, 114 patients aged 5–18 years were rand- FDA for the treatment of moderate to severe plaque pso-
omized (adalimumab 0.8  mg/kg, n = 38; adalimumab riasis in paediatric patients from the age of 12 years or
0.4 mg/kg, n = 39; methotrexate, n = 37). At week 16, sig- older, who are inadequately controlled by or intolerant to
nificantly more patients receiving adalimumab 0.8 mg/kg other systemic therapies or phototherapies [59]. The rec-
versus methotrexate achieved PASI75 (57.9% vs. 32.4%). ommended dose for adolescents <60 kg is 0.75 mg/kg, for
Of patients receiving adalimumab 0.4  mg/kg, 43.6% those with a weight ≥60 to ≤100 kg the dose is 45 mg, and
achieved PASI75. At 16 weeks, a higher proportion of patients >100 kg should be given 90 mg. Ustekinumab
patients in the adalimumab 0.8 mg/kg group than the should be administered at week 0 and 4, then every
methotrexate group achieved a PASI90 response (28.9 vs. 12 weeks thereafter.

TCS and vit D Consider


Facial/flexural
analogues topical calcineurin
psoriasis?
(or two-compound) inhibitors

SECTION 5: PSORIASIS
Dithranol

Narrow-band
Adolescent Yes
UVB

No Consider retinoids
Pustular or
erythrodermic
or
psoriasis ciclosporin
(rapid action required)

Methotrexate

MTX ineffective or
Fumaric acid
contraindicated

Biologicals

Etanercept Adalimumab Ustekinumab

Fig. 31.1  Treatment algorithm for the management of paediatric psoriasis. MTX, methotrexate; TCS, topical corticosteroids.
374 Section 5  Psoriasis

In a phase 3, multicentre, double‐blind, placebo‐­ conventional systemic treatment options. Because their
controlled study in adolescents (aged 12–17) with moder- dosing and indications in paediatric psoriasis differ, they
ate to severe plaque psoriasis, patients (n = 110) were empower the possibility of patient‐tailored treatment.
randomly assigned to ustekinumab standard dosing (SD;
0.75 mg/kg [≤60 kg], 45 mg [>60–≤100 kg], and 90 mg References
[>100 kg]) or half‐standard dosing (HSD; 0.375–mg/kg 1 Augustin M, Glaeske G, Radtke MA et  al. Epidemiology and
[≤60 kg], 22.5 mg [>60–≤100 kg], and 45 mg [>100 kg]) at comorbidity of psoriasis in children. Br J Dermatol 2010;162:633–6.
2 Bronckers IM, Paller AS, van Geel MJ et  al. Psoriasis in children
weeks 0 and 4 and every 12 weeks through week 40 or and adolescents: diagnosis, management and comorbidities.
placebo at weeks 0 and 4 with crossover to ustekinumab Paediatr Drugs 2015;17:373–84.
SD or HSD at week 12 [60). Significantly greater propor- 3 van Geel MJ, Mul K, de Jager ME et al. Systemic treatments in pae-
tions of patients receiving ustekinumab achieved PASI75 diatric psoriasis: a systematic evidence‐based update. J Eur Acad
Dermatol Venereol 2015;29:425–37.
(HSD, 78.4%; SD, 80.6%; placebo, 10.8%) or PASI90 (HSD, 4 de Jager ME, de Jong EM, van de Kerkhof PC, Seyger MM. Efficacy
54.1%; SD, 61.1%; placebo, 5.4%) at week 12. Through and safety of treatments for childhood psoriasis: a systematic litera-
week 40, all 110 patients received more than one injection ture review. J Am Acad Dermatol 2010;62:1013–30.
5 Tan X, Feldman SR, Chang J, Balkrishnan R. Topical drug delivery
of ustekinumab; among these, 81.8% reported an adverse systems in dermatology: a review of patient adherence issues. Expert
event through week 60. Infections and infestations was the Opin Drug Deliv 2012;9:1263–71.
most common category of adverse event, with nasophar- 6 Shah KN. Diagnosis and treatment of pediatric psoriasis: current and
yngitis (34.5%), upper respiratory tract infection (12.7%), future. Am J Clin Dermatol 2013;14:195–213.
7 Oostveen AM, de Jager ME, van de Kerkhof PC et al. The influence of
and pharyngitis (8.2%) occurring most often [60]. treatments in daily clinical practice on the Children’s Dermatology
Life Quality Index in juvenile psoriasis: a longitudinal study from the
Child‐CAPTURE patient registry. Br J Dermatol 2012;167:145–9.
Summary 8 Marqueling AL, Cordoro KM. Systemic treatments for severe pediat-
SECTION 5: PSORIASIS

ric psoriasis: a practical approach. Dermatol Clin 2013;31:267–88.


A flowchart is depicted in Fig. 31.1. This treatment algo- 9 Tollefson MM. Diagnosis and management of psoriasis in children.
rithm can serve as general guidance for the physician. Pediatr Clin North Am 2014;61:261–77.
A  quick progression to the next step and/or deviation 10 Stahle M, Atakan N, Boehncke WH et al. Juvenile psoriasis and its
clinical management: a European expert group consensus. J Dtsch
from this algorithm can be considered based on, for exam- Dermatol Ges 2010;8:812–8.
ple, the severity of psoriasis, recalcitrance of lesions, 11 Kimball AB, Gold MH, Zib B, Davis MW; Clobetasol Propionate
­contraindications, psychological burden, side‐effects or Emulsion Formulation Foam Phase III Clinical Study Group.
accessibility of treatments. Clobetasol propionate emulsion formulation foam 0.05%: review of
phase II open‐label and phase III randomized controlled trials in ster-
The initial treatment of children and adolescents with oid‐responsive dermatoses in adults and adolescents. J Am Acad
psoriasis is with topical corticosteroids and/or vitamin D Dermatol 2008;59:448–54, 454.e1.
analogues. A compound formulation containing calcipo- 12 Oranje AP, Marcoux D, Svensson A et  al. Topical calcipotriol in
childhood psoriasis. J Am Acad Dermatol 1997;36:203–8.
triol and betamethasone dipropionate can also be used 13 Bhutani T, Kamangar F, Cordoro KM. Management of pediatric pso-
once‐daily for a maximum of 4 weeks, followed by riasis. Pediatr Ann 2012;41:e1–7.
­alternation of the compound formulation at weekends 14 Sticherling M, Augustin M, Boehncke WH et al. Therapy of psoria-
and vitamin D analogues on weekdays. In very young sis in childhood and adolescence  –  a German expert consensus.
J Dtsch Dermatol Ges 2011;9:815–23.
children and in flexural areas, high‐potency corticoster- 15 Oostveen AM, de Jong EM, Donders AR et al. Treatment of paediat-
oids should be avoided. In flexural and facial psoriasis, ric scalp psoriasis with calcipotriene/betamethasone dipropionate
topical calcineurin inhibitors could be a good alternative. scalp formulation: effectiveness, safety and influence on children’s
quality of life in daily practice. J Eur Acad Dermatol Venereol
If available, application of dithranol (in a daycare set-
2015;29:1193–7.
ting) should be the next step in the treatment of paediatric 16 van Geel MJ, Mul K, Oostveen AM et  al. Calcipotriol/betametha-
psoriasis. With the emerging possibilities of telemedicine sone dipropionate ointment in mild‐to‐moderate paediatric psoria-
consultations, this treatment modality becomes achievable sis: long‐term daily clinical practice data in a prospective cohort.
Br J Dermatol 2014;171:363–9.
for larger groups of children. Due to concerns about poten- 17 Gooderham M, Debarre JM, Keddy‐Grant J et al. Safety and efficacy of
tial long‐term side‐effects, UV therapy in pre‐­adolescents calcipotriol plus betamethasone dipropionate gel in the treatment of
should be avoided wherever possible. In adolescents, NB‐ scalp psoriasis in adolescents 12‐17 years of age. Br J Dermatol
UVB is the preferred form of phototherapy. 2014;171:1470–7.
18 Eichenfield LF, Ganslandt C, Kurvits M, Schlessinger J. Safety and
Methotrexate is the conventional systemic drug of efficacy of calcipotriene plus betamethasone dipropionate topical
choice for children and adolescents with moderate to suspension in the treatment of extensive scalp psoriasis in adoles-
severe plaque psoriasis. If MTX is ineffective or contrain- cents ages 12 to 17 years. Pediatr Dermatol 2015;32:28–35.
19 Fotiadou C, Lazaridou E, Ioannides D. Management of psoriasis in
dicated, fumaric acid esters can be considered as an adolescence. Adolesc Health Med Ther 2014;5:25–34.
­alternative conventional systemic. In infants and male 20 Höger PH, Hamm H. [Psoriasis vulgaris in children and adolescents.
adolescents with pustular or erythrodermic psoriasis, Pathogenesis, clinical picture and therapy]. Hautarzt 2015;66:267–76.
short‐term treatment with retinoids can be considered. 21 Oostveen AM, Beulens CA, van de Kerkhof PC et al. The effective-
ness and safety of short‐contact dithranol therapy in paediatric pso-
Because of its rapid onset of action, ciclosporin is consid- riasis: a prospective comparison of regular day care and day care
ered an ideal drug for the control of unstable erythroder- with telemedicine. Br J Dermatol 2014;170:454–7.
mic, pustular and palmoplantar psoriasis for a short 22 de Jager ME, van de Kerkhof PC, de Jong EM, Seyger MM. Dithranol
therapy in childhood psoriasis: unjustifiably on the verge of falling
treatment period.
into oblivion. Dermatology 2010;220:329–32.
The biologics etanercept, adalimumab and usteki- 23 Zvulunov A, Anisfeld A, Metzker A. Efficacy of short‐contact therapy
numab represent a promising therapeutic alternative to with dithranol in childhood psoriasis. Int J Dermatol 1994;33:808–10.
Chapter 31  Psoriasis: Management 375

24 Green C, Ferguson J, Lakshmipathi T, Johnson BE. 311 nm UVB pho- 43 van Geel MJ, van de Kerkhof PC, Oostveen AM et al. Fumaric acid
totherapy  –  an effective treatment for psoriasis. Br J Dermatol esters in recalcitrant pediatric psoriasis: A prospective, daily clini-
1988;119:691–6. cal practice case series. J Dermatol Treat 2016;27:214–20.
25 Lara‐Corrales I, Ramnarine S, Lansang P. Treatment of childhood 44 Reich K, Hartl C, Gambichler T, Zschocke I. Retrospective data col-
psoriasis with phototherapy and photochemotherapy. Clin Med lection of psoriasis treatment with fumaric acid esters in children
Insights Pediatr 2013;7:25–33. and adolescents in Germany (KIDS FUTURE study). J Dtsch
26 Zamberk P, Velazquez D, Campos M et al. Paediatric psoriasis – narrow- Dermatol Ges 2016;14:50–8.
band UVB treatment. J Eur Acad Dermatol Venereol 2010;24:415–9. 45 Balak DM, Oostveen AM, Bousema MT et al. Effectiveness and safety
27 Finlay AY. Current severe psoriasis and the Rule of Tens. Br J Dermatol of fumaric acid esters in children with psoriasis: a retrospective
2005;152:861–7. analysis of 14 patients from The Netherlands. Br J Dermatol
28 de Jager ME, van de Kerkhof PC, de Jong EM, Seyger MM. A cross‐ 2013;168:1343–7.
sectional study using the Children’s Dermatology Life Quality Index 46 Mrowietz U, Reich K. Case reports of PML in patients treated for
(CDLQI) in childhood psoriasis: negative effect on quality of life and ­psoriasis. N Engl J Med 2013;369:1080–1.
moderate correlation of CDLQI with severity scores. Br J Dermatol 47 Nieuwkamp DJ, Murk JL, van Oosten BW et al. PML in a patient with-
2010;163:1099–101. out severe lymphocytopenia receiving dimethyl fumarate. N Engl J
29 Lewis‐Jones MS, Finlay AY. The Children’s Dermatology Life Quality Med 2015;372:1474–6.
Index (CDLQI): initial validation and practical use. Br J Dermatol 48 Nast A, Gisondi P, Ormerod AD et al. European S3‐Guidelines on
1995;132:942–9. the systemic treatment of psoriasis vulgaris – Update 2015 – Short
30 Klotsche J, Niewerth M, Haas JP et al. Long‐term safety of etanercept version  –  EDF in cooperation with EADV and IPC. J Eur Acad
and adalimumab compared to methotrexate in patients with juvenile Dermatol Venereol 2015;29:2277–94.
idiopathic arthritis (JIA). Ann Rheum Dis 2016;75:855–61. 49 Mazloom SE, Yan D, Hu JZ et al. TNF‐α inhibitor‐induced psoriasis: A
31 van Geel MJ, Oostveen AM, Hoppenreijs EP et al. Methotrexate in decade of experience at the Cleveland Clinic. J Am Acad Dermatol
pediatric plaque‐type psoriasis: Long‐term daily clinical practice 2018;pii: S0190‐9622(18)33089–5. doi:10.1016/j.jaad.2018.12.018.
results from the Child‐CAPTURE registry. J Dermatolog Treat 50 Eickstaedt JB, Killpack L, Tung J et  al. Psoriasis and psoriasiform
2015;26:406–12. eruptions in pediatric patients with inflammatory bowel disease
32 Papp K, Thaçi D, Marcoux D et  al. Efficacy and safety of adali- treated with anti‐tumor necrosis factor alpha agents. Pediatr Dermatol
mumab every other week versus methotrexate once weekly in chil- 2017;34:253–60.

SECTION 5: PSORIASIS
dren and adolescents with severe chronic plaque psoriasis: a 51 Diak P, Siegel J, La Grenade L et al. Tumor necrosis factor alpha block-
randomised ­double‐blind phase 3 trial. Lancet 2017;390:40–9. ers and malignancy in children: forty‐eight cases reported to the Food
33 van Ede AE, Laan RF, Rood MJ et al. Effect of folic or folinic acid sup- and Drug Administration. Arthritis Rheum 2010;62:2517–24.
plementation on the toxicity and efficacy of methotrexate in rheuma- 52 Gu Y, Nordstrom BL. The risk of malignancy among biologic‐naïve
toid arthritis: a forty‐eight week, multicenter, randomized, double‐blind, pediatric psoriasis patients: A retrospective cohort study in a US
placebo‐controlled study. Arthritis Rheum 2001;44:1515–24. claims database. J Am Acad Dermatol 2017;77:293–301.e1.
34 Prey S, Paul C. Effect of folic or folinic acid supplementation on 53 Luu M, Cordoro KM. The evolving role of biologics in the treatment
­methotrexate‐associated safety and efficacy in inflammatory disease: of pediatric psoriasis. Skin Therapy Lett 2013;18:1–4.
a ­systematic review. Br J Dermatol 2009;160:622–8. 54 McCroskery P, Wallace CA, Lovell DJ et al. Summary of worldwide
35 Bronckers IMGJ, Seyger MMB, West DP et al; Psoriasis Investigator pediatric malignancies reported after exposure to etanercept. Pediatr
Group (PsIG) of the Pediatric Dermatology Research Alliance and Rheumatol Online J 2010;8:18.
the  European Working Group on Pediatric Psoriasis (EWGPP). 55 Pfizer Limited. Enbrel (etanercept) Summary of Product Information.
Safety  of systemic agents for the treatment of pediatric psoriasis. Available at: https://www.ema.europa.eu/documents/product‐
JAMA Dermatol 2017;153(11):1147–57. information/enbrel‐epar‐product‐information_en.pdf (accessed 14
36 Lara‐Corrales I, Xi N, Pope E. Childhood psoriasis treatment: e­ vidence November 2018).
published over the last 5 years. Rev Recent Clin Trials 2011;6:36–43. 56 Paller AS, Siegfried EC, Langley RG et al. Etanercept treatment for
37 Popadic S, Nikolic M. Pustular psoriasis in childhood and adolescence: children and adolescents with plaque psoriasis. N Engl J Med
a 20‐year single‐center experience. Pediatr Dermatol 2014;31:575–9. 2008;358:241–51.
38 Charbit L, Mahe E, Phan A et al. Systemic treatments in childhood 57 Paller AS, Siegfried EC, Pariser DM et al. Long‐term safety and effi-
psoriasis: a French multicentre study on 154 children. Br J Dermatol cacy of etanercept in children and adolescents with plaque psoriasis.
2016;174:1118–21. J Am Acad Dermatol 2016;74:280–7 e3.
39 Di Lernia V, Stingeni L, Boccaletti V et al. Effectiveness and safety 58 AbbVie. Ltd. Humira (adalimumab) Summary of Product Information.
of cyclosporine in pediatric plaque psoriasis: a multicentric retro- Available at: https://www.ema.europa.eu/documents/product‐
spective analysis. J Dermatolog Treat 2016;27:395–8. information/humira‐epar‐product‐information_en.pdf (accessed 14
40 Bulbul Baskan E, Yazici S, Tunali S, Saricaoglu H. Clinical experience November 2018).
with systemic cyclosporine A treatment in severe childhood psoriasis. 59 Janssen‐Cilag. International NV. Stelara (ustekinumab) Summary of
J Dermatolog Treat 2016;27:328–31. Product Information. Available at: https://www.ema.europa.eu/
41 Posso‐De Los Rios CJ, Pope E, Lara‐Corrales I. A systematic review of documents/product‐information/stelara‐epar‐product‐information_
systemic medications for pustular psoriasis in pediatrics. Pediatr en.pdf (accessed 14 November 2018).
Dermatol 2014;31:430–9. 60 Landells I, Marano C, Hsu MC et  al. Ustekinumab in adolescent
42 Wright NA, Piggott CD, Eichenfield LF. The role of biologics and patients age 12 to 17 years with moderate‐to‐severe plaque psoria-
other systemic agents in the treatment of pediatric psoriasis. Semin sis: results of the randomized phase 3 CADMUS study. J Am Acad
Cutan Med Surg 2010;29:20–7. Dermatol 2015;73:594–603.
 377

C HA PTER   32

Pityriasis Rubra Pilaris


Liat Samuelov & Eli Sprecher
Department of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel

Abstract surrounding islands of normal‐appearing skin, and orange‐coloured


palmoplantar keratoderma. On histopathology, alternating paraker-
atosis and orthokeratosis in a checkerboard pattern, hypergran-
Pityriasis rubra pilaris (PRP) is a rare inflammatory papulosqua-
ulosis, broad thickening of the rete ridges, thick suprapapillary
mous disorder of unknown aetiology. PRP occurs equally in men
plates and follicular hyperkeratosis are typical features, although
and women and affects all races. Most cases are sporadic, however
repeated biopsies may be required to demonstrate them. Most cas-
in a minority of cases autosomal dominant inheritance has been
es are self‐limiting but a chronic course with frequent relapses and
described caused by mutations in CARD14. PRP has been classified
recalcitrant disease can rarely occur. Milder cases are best treated
into six types based on age of onset, clinical features and prog-
with topical treatments while systemic therapies are reserved for
nosis. Classical PRP features keratotic follicular papules, salmon‐
more severe and extensive disease.
coloured erythematous plaques covered with fine powdery scales,

Key points The classic juvenile subtype (type III) is characterized by


keratotic follicular papules coalescing into salmon‐coloured
erythematous plaques, surrounded by islands of normal‐
• Although most cases of pityriasis rubra pilaris (PRP) are sporadic,
appearing skin, and palmoplantar keratoderma. The atypical

PAPULOSQUAMOUS DISORDERS
up to 6.5% of all cases are inherited in an autosomal dominant
juvenile subtype (type V) which appears at birth or during
fashion.
infancy is characterized by hyperkeratotic follicular papules,
• Familial PRP results from gain‐of‐function mutations in CARD14,
ichthyosiform features and scleroderma‐like changes on the
encoding caspase recruitment domain family member 14, which

SECTION 6: OTHER
palms and soles. Most familial cases belong to the atypical
is a known activator of NF‐κB signalling. Although most cases of
juvenile type.
sporadic PRP do not carry germline mutations in CARD14, they
• Typical but not pathognomonic histopathological features include
do demonstrate evidence of heightened NF‐κB signalling.
alternating parakeratosis and orthokeratosis, hypergranulosis,
• Six subtypes of PRP have been recognized, three of which affect
broad thickening of the rete ridges, thick suprapapillary plates
children and adolescents.
and follicular hyperkeratosis.
• The circumscribed juvenile PRP subtype (type IV) represents
• Remission usually occurs within 3 years in the classic and
the most common variant before adulthood, and is associated
circumscribed juvenile types. In contrast, the atypical juvenile
with well‐demarcated thick erythematous scaly plaques with
type is characterized by a protracted and often lifelong course.
follicular keratotic papules mostly on the knees and elbows.

Introduction. Pityriasis rubra pilaris (PRP) is a rare History. PRP was first described by Devergie in 1856 and
chronic inflammatory papulosquamous disorder. PRP is named pityriasis pilaris [1,2]. Later on, the appearance of
subdivided into six subtypes based on clinical features, a follicular eruption in association with psoriasis pal­
age of onset and prognosis. Types I and II are typically maris, pityriasis capitis and pityriasis rubra as part of the
seen in adults and account for more than half of the cases. same disorder led to this constellation of manifestations
Types III–V are the forms most commonly seen in the being termed PRP in 1889 by Besnier [1]. The familial
paediatric population while type VI includes patients occurrence of PRP was first described by De Beurmann
infected with the human immunodeficiency virus (HIV). and Bith in 1910 [3].
PRP is characterized by the association of diffuse orange‐
coloured palmoplantar keratoderma (PPK), small erythe­ Epidemiology. PRP occurs worldwide and affects all
matous hyperkeratotic follicular papules which may ethnic groups and both sexes [4]. The prevalence may
coalesce into confluent erythematous scaly plaques vary with ethnicity. PRP is seen in approximately 1 in
(exfoliative erythroderma) surrounding islands of nor­ 5000 individuals diagnosed with skin disease in the USA
mal skin. There is great variability in presentation and and the UK [5,6], while in India a prevalence of 1 in
disease course. Treatment is challenging due to resist­ 50 000 has been reported [7]. The disease is characterized
ance to therapy and frequent relapses, although sponta­ by three peaks of onset: early childhood (0–10 years),
neous regression is common. late childhood (11–19 years) and adulthood (40–60 years)

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
378  Section 6 Other Papulosquamous Disorders

[3,6,8–16]. Although in adults males and females are mineral, both composed of calcium magnesium carbon­
equally affected, in children some studies suggest a ate. PRP has also been suggested to represent a delayed‐
higher prevalence in males [17]. Up to 6.5% of all PRP‐ type hypersensitivity reaction to microbial or chemical
affected individuals report a positive family history agents [62–65]. Environmental factors may also play a
[3,18,19], with onset at birth or within the first years role in the disease pathogenesis. Indeed, photoaggrava­
of life [18–23]. tion could be a possible eliciting factor in some cases of
PRP, particularly in type I [1,66,67], since photosensitivity
Pathogenesis. The pathogenesis of PRP remains elusive and worsening with ultraviolet (UV)A and UVB have also
and there is no consistent association between PRP and been reported [68–70].
specific diseases or laboratory findings. A number of the­ A positive family history exists in up to 6.5% of all PRP
ories have been advanced based on associations with cases (mostly type V PRP) with evidence of autosomal
laboratory abnormalities or preceding events, although dominant inheritance and partial penetrance [18–
most cases occur without any obvious foregoing event 21,23,71–73]. Autosomal recessive inheritance has been
[1,3,9,17,19,24–27]. described [22]. In addition, an occurrence of PRP in
PRP has been postulated to be associated with defective monozygotic twins has been reported [24]. Familial PRP
vitamin A metabolism given the clinical and histological has recently been shown to result from gain‐of‐function
resemblance between PRP and phrynoderma [5]. In addi­ mutations in CARD14, which encodes the caspase recruit­
tion, defective synthesis of retinol‐binding protein (RBP), ment domain family member 14, a known activator of
a specific carrier protein of vitamin A, has been proposed nuclear factor kappa B (NF‐κB) signalling [74–76].
as a biochemical marker for PRP [28,29]. However, the CARD14 is upregulated in the epidermis of familial PRP
serum level of vitamin A and RBP is often normal in PRP patients with concomitant evidence of heightened NF‐κB
patients, and treatment with high doses of vitamin A has signalling activity, increased transcription of NF‐κB target
met with variable success [30–37]. genes and aberrant immunological activation [20]. NF‐κB
A role for infections in aetiology has been proposed promotes keratinocyte viability during differentiation
PAPULOSQUAMOUS DISORDERS

based on clinical observations showing antecedent bacte­ [77,78], and mice displaying constitutive activation of
rial or viral infections mostly in juvenile PRP (e.g. upper NF‐κB develop a generalized skin disorder histologically
respiratory tract infection, diarrhoea, acute staphylococ­ featuring abnormal epidermal differentiation, T‐cell infil­
SECTION 6: OTHER

cal or streptococcal infections) [8,9,17,38–40]. This associ­ tration and the formation of microabscesses. CARD14
ation has been proposed to be due to immune system germline mutations are rarely found in sporadic PRP
dysregulation with an abnormal response to antigenic cases [79–81]. Nevertheless, NF‐κB signalling is activated
triggers [5,8]. PRP has been reported following various in the epidermis of patients with sporadic PRP, even in
viral infections and vaccinations including Epstein–Barr the absence of pathogenic CARD14 mutations [81], sug­
virus (EBV), cytomegalovirus (CMV), varicella, rubella, gesting that NF‐κB activation is key to the phenotype,
herpes simplex viruses [8,41–46] and measles–mumps– independent of the underlying cause. Mutations in
rubella (MMR), diphtheria–pertussis–tetanus (DPT) and CARD14 have also been identified in familial psoriasis
oral poliovirus vaccines [47,48]. Moreover, the association indicating that, beyond morphological (clinical and histo­
between PRP and HIV infection is well established [49– pathological) differences, psoriasis and PRP may share a
52], and follicular inflammation secondary to infection of common pathophysiology [20,24,82,83].
the hair follicle by HIV has been suggested as a triggering
event [51]. The concomitant appearance of Kawasaki dis­ Clinical features. PRP often represents a diagnostic chal­
ease and PRP has also been reported [53]. lenge and is commonly confused with other papulosqua­
Drugs have also been proposed as possible aetiological mous disorders, especially psoriasis. Diagnosis is based
agents. PRP‐like eruption, mostly simulating the classic on clinical features and histopathology.
adult type, has been reported following exposure to sev­ PRP was originally classified by Griffiths in 1980 into
eral drugs including labetalol, simvastatin, ramipril, five types based on age of onset, clinical course, morpho­
imatinib, multikinase inhibitors (ponatinib and sorafenib) logical appearance and prognosis [6] (Table 32.1). A sixth
and antiviral drugs (telaprevir and sofosbuvir) [54–60]. category, HIV‐associated PRP, was designated in 1995 by
The PRP‐like eruption appeared 1–12 weeks after the Miralles et  al. to account for the prevalence of PRP in
beginning of treatment. Imatinib is known to interfere HIV‐infected individuals and the atypical clinical find­
with regulatory T cells and the T‐cell receptor signalling ings and prognosis seen in those cases compared to the
pathway by reducing the phosphorylation of the leuco­ classic adult type [50]. Despite its limitations, including
cyte‐specific protein tyrosine kinase. Since enhanced the absence of clear correlation between clinical findings
activity of T‐suppressor cells and impaired T‐helper func­ and prognosis [40,84,85] and the existence of alternative
tion have been documented in PRP [61], imatinib phar­ systems such as the classification system proposed by
macological activity may be directly related to the onset Piamphongsant and Akaraphant [86], the Griffiths classi­
of the disease [56]. Similarly, sorafenib interferes with fication remains widely used [86,87].
RAF/MEK/ERK signalling, which may underlie altera­
tions in keratinocyte proliferation and differentiation [59]. Type I PRP: classic adult
One report described the appearance of PRP following Fifty five percent of PRP cases belong to the classic adult
exposure to dolomite, a sedimentary carbonate rock and a type I. This form of PRP carries the best prognosis, and
Chapter 32  Pityriasis Rubra Pilaris 379

Table 32.1  Classification of pityriasis rubra pilaris in adults and children according to Griffith

Type % Age of Clinical features Prognosis


onset

I Classic adult 55 Adults Follicular hyperkeratotic papules spreading caudally into red‐ 80% remission within 3 years
orange confluent plaques covered with fine powdery scale;
islands of sparing; diffuse red‐orange waxy PPK
II Atypical adult 5 Adults Ichthyosiform scale; sparse scalp hair; eczematous changes; Chronic
coarse PPK
III Classic juvenile 10 5–10 years Same as classic adult type 16% remission within 3 years;
CR may be seen in a year
but long duration possible
IV Circumscribed 25 3–10 years Well‐demarcated thick erythematous plaques with hyperkeratotic 32% remission within 3 years.
juvenile follicular papules over the knees and elbows; PPK Protracted course possible
V Atypical juvenile 5 0–4 years Hyperkeratotic follicular papules; ichthyosiform features; Persistent
scleroderma‐like changes on palms and soles
VI HIV‐associated Rare V Erythematous follicular papules; follicular plugging with the Poor
formation of spicules; associated with acne conglobata,
hidradenitis suppurativa and lichen spinulosus‐like eruption

CR, complete response; HIV, human immunodeficiency virus; PPK, palmoplantar keratoderma; V, variable.

although it is severe and disabling in some cases, more than resembling ichthyosis vulgaris especially on the legs,
80% of patients experience remission within 3 years [6,88]. sparse scalp hair, eczematous changes and coarse
Whereas in juvenile cases lesions usually start on the palmoplantar keratoderma with lamellated scale. This
­

PAPULOSQUAMOUS DISORDERS
lower half of the body, in adults PRP typically begins with type is characterized by a long clinical course of 20 years
upper body involvement (mostly scalp and face) and pro­ or more [6].
gresses gradually in a caudal direction towards the lower

SECTION 6: OTHER
parts of the body in a few weeks or months [6,88]. Primary Type III PRP: classic juvenile
lesions are small erythematous follicular keratotic pap­ Approximately 10% of PRP patients are affected by the
ules with an erythematous halo, which coalesce into con­ classical juvenile subtype, which is considered to be the
fluent erythematous to salmon‐coloured scaly plaques juvenile counterpart of PRP type I. Disease course and
surrounding islands of normal skin (‘islands of sparing’, lesion morphology and distribution are similar to type I,
previously termed ‘nappes claires’), which is a character­ but onset is typically between 5 and 10 years of age. In
istic feature [2,5]. Scales may be fine and powdery (scalp addition, febrile illnesses and trauma have been reported
and face), or coarse, especially on the lower part of the to precede the appearance of type III PRP in several
body. The rash is usually symmetrical and bilateral and cases  [3,9,38,41]. The eruption starts on the head, upper
often evolves into an exfoliative erythroderma, the fre­ trunk or neck as red‐orange macules covered with fine
quency and incidence of which vary among the different bran‐like scales (especially over the scalp and face).
types [5,17,34,86]. PPK usually appears within weeks and Within a few weeks or months, further macules appear
typically features a prominent red‐orange colour and with the formation of perifollicular erythematous papules
waxy appearance associated with painful fissures. with a central keratotic plug. Lesions coalesce and spread
Skin involvement may be associated with itching, espe­ in a cephalocaudal direction, resulting eventually in exfo­
cially in a generalized distribution, and it might be com­ liative erythroderma with up to 1 cm islands of sparing.
plicated by Kaposi varicelliform eruption [89,90]. While one series reported the development of erythro­
Oral mucosa involvement, mostly the buccal mucosa, derma in 17% of cases, this was not observed by others
gingivae and tongue, may include white plaques and [17,87]. Cephalic involvement is characteristic in children
nodules and erythematous lesions covered with white and appears in 40% of cases. It may extend towards the
streaks that may be associated with pain and irritation neck and the upper torso in a cape‐like appearance with
and resemble lichen planus [91–93]. Nail findings consist sharp borders, and large adhesive scales overlying the
of thick nail plates with a rough surface, yellow‐brown typical red‐orange erythema [87]. These features are asso­
discolouration, splinter haemorrhages, atrophy, longitu­ ciated with prominent PPK with a characteristic yellow‐
dinal ridging and subungual hyperkeratosis [6,93,94]. orange hue; when seen on the soles, this is referred to as
Ectropion has been reported in association with erythro­ ‘keratodermic sandal’. The keratoderma in children is
derma, as have other ocular complications such as periph­ characterized by oedema (often disabling) with sharply
eral ulcerative keratitis and corneal perforation [95–98]. demarcated borders and extension towards the dorsal
Reactive lymphadenopathy is not uncommon. aspect of the palms and soles. The Achilles tendon is fre­
quently involved [87]. Widespread eruptions are often
Type II PRP: atypical adult accompanied by pruritus and a burning sensation. The
Atypical adult type II PRP affects approximately 5% of Koebner phenomenon may be seen in approximately 10%
PRP patients. It is characterized by ichthyosiform scales of children with PRP [87,99].
380  Section 6 Other Papulosquamous Disorders

Nail involvement is relatively common in juvenile PRP spontaneously in 3 years while other series reported a pro­
(Fig. 32.1), appearing in 13–33% of cases. The nails may tracted course longer than 3 years [3,6,40]. Partial healing of
become thickened, yellowish and curved. In addition, the lesions may result in transformation into type IV [3],
subungual hyperkeratosis, longitudinal striae, ‘half and and mixed type III and IV cases have been reported [102]. A
half’ nails with terminal hyperaemia and splinter haem­ case of classic juvenile PRP evolving into a classic adult type
orrhages may also be seen [17,87,100]. Although hair and after a long latency period has also been described [103].
teeth are usually unaffected, mucous membrane involve­
ment may be observed with a diffuse whitish appearance Type IV PRP: circumscribed juvenile
of the buccal mucosa [101]. The circumscribed juvenile type represents the most com­
Ectropion may be evident in severe cases and was mon juvenile variant, and the second most common vari­
reported in up to 13% of juvenile PRP cases in one ant overall, accounting for approximately 25% of all cases,
series [17]. although a few paediatric PRP series have identified PRP
The eruption reaches a peak and often begins to resolve type III as the most common type in children, which prob­
spontaneously after several months to a year. Although ably reflects geographical or ethnic differences [17,62].
complete remission may be seen in a year, Griffiths Type IV affects prepubertal children and young adults. It
originally estimated that 16% of type III patients clear
­ is characterized by well‐demarcated erythematous scaly
plaques with follicular accentuation and plugging on the
knees and elbows and occasionally the anterior shins.
Transient erythematous macules and grouped follicular
papules with keratin plugs may appear on other parts of
the body (e.g. trunk and scalp) and hyperkeratosis may
be found over bony prominences (Fig. 32.2). Palmoplantar
involvement is characteristic but may be absent [104].
Dermoscopy examination of the lesions over the extensor
PAPULOSQUAMOUS DISORDERS

surfaces, which can help in differentiating the lesions


from psoriasis vulgaris, reveals multiple whitish keratotic
follicular plugs and a yellow peripheral keratotic ring,
SECTION 6: OTHER

surrounded by erythema with some linear vessels [105].


The clinical course of the disease is marked by remissions
and exacerbations even when mild and localized, and the
3‐year remission rate of 32% is substantially less than the
Fig. 32.1  Nail dystrophy in juvenile PRP. Source: Courtesy of Prof. Peter Itin. 81% seen in the classical adult type [3,6]. Gelmetti et al.

(a) (b)
Fig. 32.2  (a) PRP manifests in a young child with circumscribed plaques over the trunk, (b) featuring prominent follicular accentuation. Source: Courtesy of
Prof. Peter Itin.
Chapter 32  Pityriasis Rubra Pilaris 381

proposed classification into an acute form that resolves in enteropathy and membranous nephropathy have also
6 months, an intermediate form that disappears after 1 been reported [137,138]. Moreover, both PRP and associ­
year, and a chronic form that lasts more than a year and ated arthritis have been found to improve following
may evolve into erythroderma (types I or III) [87]. ­retinoids and tumour necrosis factor (TNF)‐α blockade
therapy [127,139], although not in every case [126].
Type V: atypical juvenile Dermatomyositis may present as a PRP‐like cutaneous
The atypical juvenile type occurs in up to 5% of PRP eruption, a variant known as Wong dermatomyositis. Skin
patients and is the rarest form of the juvenile types. It lesions with both clinical and histopathological features of
appears at birth or during infancy and manifests as hyper­ PRP may occur simultaneously with, before, or after the
keratotic follicular papules, widespread erythema and myositis [125,132,140–142]. It has been postulated that in
scales, and scleroderma‐like changes on the palms, soles cases of erector pili muscle myositis, the hair cycle is
and particularly the digits [18,23,106,107]. Most cases of altered by abnormalities of infundibular keratinization
familial PRP belong to this type, although familial type III and subsequent follicular keratotic plugging [143,144].
and type IV have been reported [88,108], in addition to a PRP is also associated with autoimmune hypothyroid­
combination of type V and other types within the same ism, and thyroid hormone replacement has been shown
family [18]. Type V usually persists throughout life. to bring about rapid and complete remission of PRP. It
has  been suggested that the occurrence of PRP in those
Type VI: HIV‐associated patients is related to the fact that thyroid hormone defi­
This subtype of PRP was added to the classification by ciency inhibits the transformation of carotene to vitamin
Miralles et al. in 1995 because HIV patients with PRP pre­ A which has been suggested to play a role in PRP patho­
sent with different clinical features and have a poorer genesis [131,133,145].
prognosis than patients with type I [50]. This type may be PRP can develop in conjunction with immunoglobulin
the presenting symptom of HIV infection although it A deficiency and common hypogammaglobulinaemia.
usually appears in patients with known HIV. It is charac­ Moreover, enhanced activity of T‐suppressor cells and

PAPULOSQUAMOUS DISORDERS
terized by erythematous follicular papules and prominent impairment of T‐helper function have been described
follicular plugging with the formation of spicules distrib­ in  PRP, suggesting an underlying immune mechanism
uted symmetrically over the extensor surfaces of the [61,146,147].

SECTION 6: OTHER
body, mainly the face and trunk [49,50]. Its association Although a coincidental finding cannot be ruled out,
with acne conglobata, hidradenitis suppurativa and lichen PRP has been identified in several patients with Down
spinulosus‐like eruption has led to the concept of HIV‐ syndrome and one of these cases was associated with
associated follicular syndrome [109]. In contrast with vitiligo [25–27].
type I, the occurrence of nail involvement and PPK is
variable. PRP type VI is notoriously resistant to treatment. Histopathology and  laboratory findings. The histo­
Complete remission may be seen with antiretroviral pathological findings in PRP are characteristic but not
­therapy although relapses are frequent [110]. specific or pathognomonic, emphasizing the need for
clinicopathological correlation. Early PRP lesions and
Associated diseases. PRP may appear in conjunction cases of erythroderma can be difficult to diagnose histo­
with other benign cutaneous disorders including eruptive logically because of subtle pathology [148]. Nevertheless,
seborrhoeic keratosis (mainly in the context of an erythro­ pathology is important for ruling out other diseases in the
dermic flare of PRP), sebaceous and glandular diseases, differential diagnosis, mainly psoriasis.
hidradenitis suppurativa and dermatitis herpetiformis The epidermis shows acanthosis with a ‘checkerboard’‐
[1,111–114]. like pattern of alternating orthokeratosis and parakera­
An association of PRP with several noncutaneous con­ tosis, focal or confluent hypergranulosis as well as thick
ditions, mostly malignancies and autoimmune diseases, suprapapillary plates with broad thickening of the rete
has been postulated. There are several reports of PRP ridges (Fig. 32.3). Dilated hair follicles with a keratotic plug
associated with various malignancies including renal cell and ‘shoulder parakeratosis’ (parakeratosis on both sides
carcinoma, hepatocellular carcinoma, squamous cell car­ of the keratin plug) are a characteristic finding [149,150]. A
cinoma, adenocarcinoma, bronchogenic carcinoma, laryn­ sparse superficial lymphohistiocytic perivascular and peri­
geal carcinoma, Merkel cell carcinoma and aggressive follicular infiltrate may be seen in the underlying papillary
cutaneous malignancies (e.g. Kaposi sarcoma, malignant dermis in conjunction with limited vascular dilation. The
melanoma, multiple basal cell carcinoma) [91,115–121]. In infiltrate does not contain neutrophils but eosinophils
some cases, PRP has been considered a paraneoplastic and plasma cells may be seen, possibly supporting the
manifestation [91,116,122,123] as resolution has coincided theory of a delayed‐type hypersensitivity reaction to
with curing the associated neoplasm. antigenic triggers [62–64]. Munro microabscesses, a dimin­
PRP has been diagnosed in association with autoim­ ished granular layer and elongated rete ridges usually seen
mune diseases including myasthenia gravis, coeliac in psoriasis are absent. Although circumscribed juvenile
sprue, lupus erythematosus, dermatomyositis, systemic PRP exhibits more psoriasiform features than classic PRP,
sclerosis, autoimmune thyroiditis and hypothyroidism, it lacks Munro microabscesses [150].
seronegative arthritis and rheumatoid arthritis [124–136]. Other less common histological findings have been
Single cases of PRP in conjunction with protein‐losing reported in PRP. Acantholysis with or without dyskeratosis
382  Section 6 Other Papulosquamous Disorders

Table 32.2  Features differentiating pityriasis rubra pilaris (PRP)


from psoriasis

Features PRP Psoriasis

Clinical
Scalp scaling Diffuse, fine, Well‐demarcated
powdery erythematous plaques with
thick, silvery, adherent scale
Keratoderma Diffuse, red‐orange, Mostly focal, well‐demarcated
waxy erythematous plaques with
thick adherent scale
Follicular Common Not common
accentuation
Islands of Common Not common
sparing
Arthritis Absent May be present
Nail changes No pitting or oil Present
Fig. 32.3  Skin biopsy showing alternating ortho‐ and parakeratosis, stains
epidermal hyperplasia and a mononuclear infiltrate (H&E, ×400).
Source: Courtesy of Prof. Peter Itin. Histopathological
Granular layer Hypergranulosis Hypogranulosis
Rete ridges Short and broad Elongated
has been reported in several case reports and series Suprapapillary Thick Thin
plates
[62–64,90,151]. Magro and Crowson demonstrated acan­
Follicular plugging Common Not common
tholysis in 25 of 32 biopsies of PRP, while a more recent Munro Very rare Common
PAPULOSQUAMOUS DISORDERS

case series found acantholysis in 22% of biopsies microabscesses


obtained from patients with type I [63,151]. In addition, Vascular dilatation Mildly increased Significantly increased
pemphigus‐like acantholysis has been reported in PRP Dermal infiltrate Lymphocytes and Mainly PMNs
SECTION 6: OTHER

[55,152]. Hashimoto and Fedoronko suggested that monocytes.


Eosinophils possible
acantholysis in PRP is induced by proteolytic enzymes
Acantholysis May be present Not common
and urea in eccrine sweat located in the keratin‐plugged
acrosyringia [153], a theory not confirmed by others Electron microscopy
[154]. A lichenoid infiltrate rather than a sparse superfi­ Tonofilaments in the granular layer Decreased Diminished
cial perivascular infiltrate and lichen nitidus‐like find­ Keratohyalin granules Normal to increased Diminished
ings have been reported in isolated cases of PRP
[63,64,153,155]. Accordingly, the presence of eosinophils, Treatment
focal acantholysis and lichenoid infiltrate should not Response to UVB Variable Good
exclude a diagnosis of PRP.
Immunoblot analysis demonstrates abnormal keratin Disease course
expression in the epidermis of PRP patients including Prognosis Mostly self‐limited Chronic with exacerbations
KRT6/KRT16 expression, abnormal KRT14 expression,
and expression of a 45 kDa acidic keratin not normally PMN, polymorphonuclear neutrophil; UVB, ultraviolet B.
expressed in epidermis, consistent with abnormal epider­
mal differentiation [23]. Moreover, the epidermis of PRP
patients demonstrates a hyperkinetic state with turnover
time between normal and that found in psoriasis longitudinal clinical observation and repeated biopsies
[32,149,156–158], and overexpression of p53 in PRP epi­ may be necessary.
dermis [159]. The main differential diagnosis in both classic and
Electron microscopy findings include a decreased localized forms of juvenile PRP is psoriasis. A clinical
number of tonofilaments and desmosomes, enlarged course with caudal progression of the lesions, islands
intercellular spaces, parakeratosis with lipid‐like vac­ of sparing and typical yellow‐orange confluent kerato­
uoles, a large number of keratinosomes, splitting of derma, the characteristic histopathological findings
the basal lamina, a reduced number of tonofilaments with lack of Munro microabscesses, poor response to
in the stratum granulosum and parakeratosis with phototherapy, recalcitrance to topical corticosteroid
irregular keratinization, vacuoles and lipoid droplets treatment and a relatively shorter disease duration
[33,149]. all  favour a diagnosis of PRP over psoriasis [5]. Key
­differences between psoriasis and PRP are given in
Differential diagnosis. Although no specific diagnostic Table 32.2.
features exist for PRP, the diagnosis can be established Additional differential diagnoses include cutaneous T‐cell
with a high degree of certainty in the presence of typical lymphoma, seborrhoeic dermatitis (especially ­during early
clinical and histopathological features. Not uncommonly, scalp involvement in PRP types I and III), hypersensitivity
Chapter 32  Pityriasis Rubra Pilaris 383

Management of juvenile pityriasis rubra pilaris

Mild disease Severe disease

Emollients
Topical treatments Topical corticosteroids
Emollients Topical treatments Vitamin D analogues
Topical corticosteroids Keratolytic agents
Vitamin D analogues Tazarotene*
Keratolytic agents Calcineurin inhibitors*
Tazarotene*
Calcineurin inhibitors*
NB-UVB
Phototherapy Re-PUVA*
UVA1*
(+/– systemic retinoids)
Bath PUVA*

Systemic retinoids - 1st line


Methotrexate - 2nd line
Other oral therapies:
• Azathioprine
Systemic therapies • Cyclosporine A
• Fumaric acid
Biologics:
• Infliximab
• Etanercept

PAPULOSQUAMOUS DISORDERS
• Adalimumab
• Ustekinumab**

Fig. 32.4  Management of juvenile pityriasis rubra pilaris. *Reported in individual cases; **first‐line in patients with activating mutations in CARD14. NB‐UVB,

SECTION 6: OTHER
narrow‐band UVB; Re‐PUVA, oral retinoids + photochemotherapy.

reactions, subacute cutaneous lupus erythematosus and


Treatment. No treatment has been reported as univer­
Wong‐type dermatomyositis. The circumscribed juvenile
sally effective in PRP. The fact that spontaneous remis­
type may be confused with pemphigus foliaceus, Grover
sions are not uncommon complicates the interpretation
disease or epidermal naevus [63,104]. Atopic dermatitis
of  uncontrolled clinical trials, while controlled trials are
and Netherton syndrome may be confused with PRP
not feasible given the rarity of the disease. In addition,
[19,23,71,160], particularly in children. Moreover, in the
given the generally favourable course, aggressive systemic
paediatric population, in whom PRP may follow an infec­
treatment is usually not warranted. The management of
tion, the differential diagnosis also includes scarlet fever,
juvenile PRP is summarized in Fig. 32.4.
staphylococcal scalded skin syndrome and Kawasaki
disease.
The circumscribed form of PRP might be confused Topical treatments
with other dermatoses associated with follicular plugging Topical treatments are the mainstay of therapy in milder
as a main feature including keratosis pilaris, follicular cases of juvenile PRP and include emollients, topical cor­
mycosis fungoides, discoid lupus erythematosus, Darier ticosteroids, vitamin D analogues, keratolytic agents (e.g.
disease, lichen planopilaris, lichen spinulosus, phryno­ formulations containing urea, salicylic acid, lactic acid or
derma and lichen scrofulosorum [3]. In particular, α‐hydroxy acid) and tazarotene alone or in combination,
the  atypical juvenile form should be differentiated with variable response.
from  autosomal recessive congenital ichthyosis (ARCI), Emollients, including equal parts of white soft paraffin
ichthyosis vulgaris and erythrokeratoderma variabilis and liquid paraffin alone or in a combination with 20%
(EKV). The presence of follicular keratotic papules with propylene glycol and 10% urea for keratoderma, are
keratin plugs and the red‐orange colour of the lesions always beneficial.
distinguish PRP from nonbullous ichthyosiform eryth­ PRP is generally not steroid‐responsive but corticoster­
roderma, and the prominent erythema favours PRP over oids can be helpful in cases of exfoliative erythroderma,
ichthyosis vulgaris. EKV can be associated with a and topical corticosteroids may lead to complete resolu­
positive family history and symmetrically‐distributed
­ tion in juvenile PRP [16,17].
­migratory well‐demarcated red‐orange plaques with fine Calcipotriol has been shown to be effective in PRP and
scale in conjunction with more persistent hyperkeratotic resulted in disappearance of the inflammatory infiltrate
plaques and PPK. on histology [161]. However, absorption of vitamin D
The differential diagnoses of erythroderma and kerato­ analogues is a limiting factor in children with PRP.
derma in the setting of PRP are described in Chapters 10 The use of keratolytics is also limited in children due to
and 127. burning, stinging and systemic absorption.
384  Section 6 Other Papulosquamous Disorders

Although reported only in individual cases, a sustained cases when single therapy has not been shown to be
remission was achieved in a child with juvenile circum­ e­ ffective, although it increases the risk of hepatotoxicity
scribed PRP with topical tazarotene [160], pimecrolimus [5,11,179]. Low‐dose MTX may be effective in treating
1% has been shown to be effective in scalp and face PRP‐induced lower eyelid cicatricial ectropion [97].
involvement in PRP [162] and topical capsaicin was effec­
tive in a case of severe pruritus resistant to topical corti­ Other oral treatments
costeroids and antihistamines [163]. Refractory adult and juvenile cases with no response to
Clinicians should be aware that a generalized form of retinoids and MTX might respond to low‐dose short‐term
PRP was described in a few cases following topical treat­ treatment with ciclosporin (cyclosporine A) [180–182],
ment with imiquimod, a toll‐like receptor agonist and and azathioprine has been used with variable clinical
immune response modifier, used for actinic keratosis and response [173,183]. Fumaric acid treatment in increasing
viral warts [164]. dosages was very effective in a child with type V PRP,
recalcitrant to various treatment modalities. The sug­
Phototherapy gested mechanism of action is related to the immune‐
Although PRP can be aggravated by UV exposure [68–70], modulating and inhibitory effects on keratinocyte
phototherapy may be an effective treatment. Successful proliferation and activation of fumaric acid [184].
treatment of PRP with narrow‐band UVB (NB‐UVB) alone
or in combination with retinoids has been reported Biologics. Recently, there has been an increasing number
[102,165,166]. Oral retinoids in combination with photo­ of reports demonstrating the effectiveness and safety of
chemotherapy (Re‐PUVA) [163] or UVA1 [167] may also biologic therapies in PRP, many of which have been tried
be an effective option, and in a case of UVB‐provoked PRP, given PRP’s similarities to psoriasis [8,24,185,186]. TNF‐α
good results were achieved with bath PUVA [168]. Some blockers were the first biologic therapies investigated in
authors reported improvement with the Goekerman regi­ PRP. Those agents are known to target the NF‐κB signal­
men [17], although this was not confirmed by others [34]. ling pathway and response to these drugs is in part
PAPULOSQUAMOUS DISORDERS

determined by polymorphisms in genes encoding vari­


Systemic therapy ous elements of the NF‐κB pathway, explaining their ben­
Systemic therapy should be considered for severe and eficial effect in cases of familial PRP with CARD14
SECTION 6: OTHER

extensive disease. Systemic retinoids seem to be the most mutation [19,187]. Infliximab has been shown to be effec­
effective therapy, while other therapies including metho­ tive mostly in adults with erythrodermic or refractory
trexate (MTX), azathioprine, ciclosporin (cyclosporine) PRP as monotherapy or in combination with adjunctive
and fumaric acid have been reported with variable systemic treatments [188–196]. However, less favourable
response [17]. Systemic corticosteroids are not effective in results were published in two cases  [197,198]. A few
PRP, and their discontinuation can result in disease flare reports of protracted type III PRP with significant
[17,169]. improvement of both skin lesions and nail changes have
been reported with infliximab [199,200]. Etanercept
Retinoids administered alone or in combination with other thera­
Isotretinoin (13‐cis‐retinoic acid) is given in a daily oral pies in adults with PRP led to favourable responses
dose of 0.75–2 mg/kg while acitretin is given in a single [139,197,201–203]. Successful treatment was reported in
oral daily dose of 0.5–1 mg/kg [17,34,170–172]. A thera­ two cases of juvenile and familial PRP [19,204]. Similar
peutic trial of retinoids is considered adequate with at promising results were achieved also with the third TNF
least 4–6 months of treatment with careful monitoring of inhibitor, adalimumab, in adults with PRP [205–211]. A
lipid profile, liver function tests and, where appropriate, 17‐year‐old boy unresponsive to etanercept was success­
bone disease. A single case of extraspinal hyperostosis has fully treated with adalimumab [212]. In addition, usteki­
been reported after 13 years of oral retinoid therapy in a numab has proved to be effective in cases of recalcitrant
child with PRP [173]. Recent reports of individual cases, PRP [72,76,213–217]. As ustekinumab is directed against
one small series in adults and one case of circumscribed the p40 subunit of both interleukin (IL)‐12 and IL‐23
juvenile PRP demonstrated favourable response to treat­ which are known to activate the NF‐κB pathway induced
ment with alitretinoin (9‐cis‐retinoic acid) [107,174–177]. by CARD14, it may be considered as the first‐line treat­
Treatment with systemic retinoids may shorten disease ment option in patients with activating mutations in
course but short‐term treatment, which is safe and well CARD14 [20,60,72].
tolerated, is required given the fact that most cases in chil­
dren are self‐limited [170,178]. A clinical response is typi­ Other treatments
cally seen in 14–16 weeks [34]. Three adult patients, one with type II PRP and two with
erythrodermic PRP not responsive to various therapies,
Methotrexate were successfully treated with extracorporeal photopho­
Oral MTX at a dose of 10–15 mg/week can be considered resis (ECP), two of them in combination with either acitre­
in refractory cases, but variable responses have been tin or ciclosporin [218,219]. One patient with recalcitrant
reported and a greater benefit has been seen in adults type II PRP was treated with intravenous immunoglo­
than in children [6,17,34,173]. The combination of sys­ bulin (IVIG) at a dosage of 2 g/kg (over 3 days) every 3
temic retinoids and MTX has been reported successful in weeks, and demonstrated a dramatic improvement [220].
Chapter 32  Pityriasis Rubra Pilaris 385

Patients with HIV‐associated PRP may benefit from 28 Bergamaschini L, Tucci A, Colombo A et  al. Effect of stanozolol in
patients with pityriasis rubra pilaris and retinol‐binding protein
­ ifferent combinations of antiretroviral treatment alone
d
­deficiency. N Engl J Med 1982;306:546–7.
or in combination with other treatments [110,221]. 29 Finzi AF, Altomare G, Bergamaschini L et al. Pityriasis rubra pilaris
Of note, several cases of PRP with an erythema ­gyratum and and retinol‐binding protein. Br J Dermatol 1981;104:253–6.
repens‐like appearance have been reported following 30 Stoll DM, King LE Jr, Chytil F. Serum levels of retinol binding
protein in patients with pityriasis rubra pilaris. Br J Dermatol
­
treatment with retinoids or MTX. Currently there is no 1983;108:375.
evidence that those patients have an increased risk of 31 van Voorst Vader PC, van Oostveen F, Houthoff HJ et  al. Pityriasis
malignancy, and malignancy evaluation should be age‐ rubra pilaris, vitamin A and retinol‐binding protein: a case study. Acta
and clinical‐based [222–226]. Derm Venereol 1984;64:430–2.
32 Griffiths WA. Vitamin A and pityriasis rubra pilaris. J Am Acad
Surgical management should be considered in cases of Dermatol 1982;7:555.
cicatricial ectropion in PRP [98]. 33 Kanerva L, Lauharanta J, Niemi KM et al. Ultrastructure of pityriasis
rubra pilaris with observations during retinoid (etretinate) treatment.
References Br J Dermatol 1983;108:653–63.
1 Griffiths WA. Pityriasis rubra pilaris ‐ an historical approach. Trans 34 Dicken CH. Treatment of classic pityriasis rubra pilaris. J Am Acad
St Johns Hosp Dermatol Soc 1975;61:58–69. Dermatol 1994;31:997–9.
2 Griffiths WA. Pityriasis rubra pilaris—an historical approach. 2. 35 Rothman S. Physiology and biochemistry of the keratinization pro­
Clinical features. Clin Exp Dermatol 1976;1:37–50. cess. Ill Med J 1955;108:10–13.
3 Sehgal VN, Srivastava G. (Juvenile) Pityriasis rubra pilaris. Int J 36 Kan Y, Sumikawa Y, Yamashita T. Successful treatment of pityriasis
Dermatol 2006;45:438–46. rubra pilaris with oral Vitamin A in oil (Chocola A) for an 18‐month‐
4 Gemmeke A, Schonlebe J, Koch A et al. Pityriasis rubra pilaris—a ret­ old child. J Dermatol 2015;42:1210–1.
rospective single center analysis over eight years. J Dtsch Dermatol 37 Koga M, Koga K, Nakayama J. A pediatric case of pityriasis rubra
Ges 2010;8:439–44. pilaris successfully treated with low‐dose vitamin a. Case Rep
5 Albert MR, Mackool BT. Pityriasis rubra pilaris. Int J Dermatol Dermatol 2012;4:170–3.
1999;38:1–11. 38 Mohrenschlager M, Abeck D. Further clinical evidence for involve­
6 Griffiths WA. Pityriasis rubra pilaris. Clin Exp Dermatol ment of bacterial superantigens in juvenile pityriasis rubra pilaris
1980;5:105–12. (PRP): report of two new cases. Pediatr Dermatol 2002;19:569.
7 Sehgal VN, Jain MK, Mathur RP. Pityriasis rubra pilaris in Indians. 39 Yamamoto T, Yokoyama A. Lymphocyte response to superantigen in
Br J Dermatol 1989;121:821–2. a patient with childhood‐onset pityriasis rubra pilaris. Int J Dermatol

PAPULOSQUAMOUS DISORDERS
8 Betlloch I, Ramon R, Silvestre JF et  al. Acute juvenile pityriasis 1999;38:639–40.
rubra  pilaris: a superantigen mediated disease? Pediatr Dermatol 40 Yang CC, Shih IH, Lin WL et  al. Juvenile pityriasis rubra pilaris:
2001;18:411–4. report of 28 cases in Taiwan. J Am Acad Dermatol 2008;59:943–8.

SECTION 6: OTHER
9 Davidson CL Jr, Winkelmann RK, Kierland RR. Pityriasis rubra pila­ 41 Ertam I, Sezgin AO, Kazandi A et al. A case of juvenile pityriasis rubra
ris. A follow‐up study of 57 patients. Arch Dermatol 1969;100:175–8. pilaris: could varicella be an aetiological agent? Clin Exp Dermatol
10 Caplan SE, Lowitt MH, Kao GF. Early presentation of pityriasis rubra 2009;34:e1012–3.
pilaris. Cutis 1997;60:291–6. 42 Betto P, Vassilopoulou A, Colombari R et al. Acute juvenile pityriasis
11 Clayton BD, Jorizzo JL, Hitchcock MG et  al. Adult pityriasis rubra rubra pilaris: a case report after mononucleosis infection. G Ital
pilaris: a 10‐year case series. J Am Acad Dermatol 1997;36:959–64. Dermatol Venereol 2008;143:271–3.
12 Darmstadt GL, Tunnessen WW Jr. Picture of the month. Juvenile pity­ 43 Funatsu S, Maejima H, Amoh Y et al. Pityriasis rubra pilaris followed
riasis rubra pilaris. Arch Pediatr Adolesc Med 1995;149:923–24. by severe herpes simplex 2 infection. Eur J Dermatol 2013;23:720–1.
13 Jacyk WK. Pityriasis rubra pilaris in black South Africans. Clin Exp 44 Kawara S, Miyake M, Oiso N et al. Pityriasis rubra pilaris with pre­
Dermatol 1999;24:160–3. ceding cytomegalovirus infection. Dermatology 2009;219:350–2.
14 Kierland RR, Kulwin MH. Pityriasis rubra pilaris; a clinical study. 45 Wang T, Liu J, Liu Y et al. Pityriasis rubra pilaris (PRP) with preceding
Arch Derm Syphilol 1950;61:925–30. Epstein‐Barr virus infection: a new type PRP with non‐HIV virus
15 Sehgal VN, Bajaj P, Jain S. Pityriasis rubra pilaris (PRP): report of four infection? Chin Med J (Engl) 2014;127:2391.
cases. J Dermatol 2000;27:174–7. 46 Larregue M, Champion R, Bressieux JM et al. [Acute pityriasis rubra
16 Selvaag E, Haedersdal M, Thomsen K. Pityriasis rubra pilaris: a retro­ pilaris in the child. Apropos of 4 cases]. Ann Dermatol Venereol
spective study of 12 patients. J Eur Acad Dermatol Venereol 1983;110:221–8.
2000;14:514–5. 47 Naciri Bennani B, Cheikh Rouhou H, Waton J et al. [Pityriasis rubra
17 Allison DS, El‐Azhary RA, Calobrisi SD et al. Pityriasis rubra pila- pilaris after vaccination]. Ann Dermatol Venereol 2011;138:753–6.
ris in children. J Am Acad Dermatol 2002;47:386–9. 48 Mohamed M, Belhadjali H, Hammedi F et al. Pityriasis rubra pilaris
18 Thomson MA, Moss C. Pityriasis rubra pilaris in a mother and two occurring after vaccination with diphtheria‐pertussis‐tetanus and
daughters. Br J Dermatol 2007;157:202–4. oral poliovirus vaccines. Indian J Dermatol Venereol Leprol
19 Vasher M, Smithberger E, Lien MH et al. Familial pityriasis rubra pila­ 2015;81:618–20.
ris: report of a family and therapeutic response to etanercept. J Drugs 49 Auffret N, Quint L, Domart P et al. Pityriasis rubra pilaris in a patient
Dermatol 2010;9:844–50. with human immunodeficiency virus infection. J Am Acad Dermatol
20 Fuchs‐Telem D, Sarig O, van Steensel MA et al. Familial pityriasis 1992;27:260–1.
rubra pilaris is caused by mutations in CARD14. Am J Hum Genet 50 Miralles ES, Nunez M, De Las Heras ME et al. Pityriasis rubra pila-
2012;91:163–70. ris and human immunodeficiency virus infection. Br J Dermatol
21 Kint A, De Bie S, Geerts ML et al. Pityriasis rubra pilaris, a familial 1995;133:990–3.
condition. Arch Belg Dermatol Syphiligr 1972;28:371–6. 51 Misery I, Faure M, Claidy A. Pityriasis rubra pilaris and human
22 Knudsen EA. Pityriasis rubra pilaris in identical twins. Br J Dermatol immunodeficiency virus infection—type 6 pityriasis rubra pilaris? Br
1958;70:27–9. J Dermatol 1996;135:1008–9.
23 Vanderhooft SL, Francis JS, Holbrook KA et  al. Familial pityriasis 52 Menni S, Brancaleone W, Grimalt R. Pityriasis rubra pilaris in a child
rubra pilaris. Arch Dermatol 1995;131:448–53. seropositive for the human immunodeficiency virus. J Am Acad
24 Klein A, Landthaler M, Karrer S. Pityriasis rubra pilaris: a review of Dermatol 1992;27:1009.
diagnosis and treatment. Am J Clin Dermatol 2010;11:157–70. 53 Pluchinotta FR, Martini G, Fortina AB et  al. Concomitant severe
25 Hazini AR, Rongioletti F, Rebora A. Pityriasis rubra pilaris and viti­ Kawasaki disease and pityriasis rubra pilaris and in a teenager: just a
ligo in Down’s syndrome. Clin Exp Dermatol 1988;13:334–5. coincidence? Clin Exp Rheumatol 2010;28:113.
26 Holden CA, Curley RK. Down’s syndrome and pityriasis rubra pila­ 54 Cheung EJ, Jedrych JJ, English JC. Sofosbuvir‐induced erythrodermic
ris. Clin Exp Dermatol 1989;14:332. pityriasis rubra pilaris‐like drug eruption. J Drugs Dermatol
27 Terasaki K, Kanekura T, Saruwatari H et al. Classical juvenile pityria­ 2015;14:1161–2.
sis rubra pilaris in a patient with Down syndrome. Clin Exp Dermatol 55 Gajinov ZT, Matic MB, Duran VD et al. Drug‐related pityriasis rubra
2004;29:49–51. pilaris with acantholysis. Vojnosanit Pregl 2013;70:871–3.
386  Section 6 Other Papulosquamous Disorders

56 Plana A, Carrascosa JM, Vilavella M et al. Pityriasis rubra pilaris‐like 85 Shahidullah H, Aldridge RD. Changing forms of juvenile pityriasis
reaction induced by imatinib. Clin Exp Dermatol 2013;38:520–2. rubra pilaris—a case report. Clin Exp Dermatol 1994;19:254–6.
57 Schmutz JL, Trechot P. [Telaprevir‐induced pityriasis rubra pilaris]. 86 Piamphongsant T, Akaraphant R. Pityriasis rubra pilaris: a new
Ann Dermatol Venereol 2013;140:414–5. ­proposed classification. Clin Exp Dermatol 1994;19:134–8.
58 Stalling SS, Vu JR, English JC. Telaprevir‐induced pityriasis 87 Gelmetti C, Schiuma AA, Cerri D et al. Pityriasis rubra pilaris in
rubra  pilaris‐like drug eruption. Arch DermatolArch Dermatol childhood: a long‐term study of 29 cases. Pediatr Dermatol
2012;148:1215–7. 1986;3:446–51.
59 Paz C, Querfeld C, Shea CR. Sorafenib‐induced eruption resembling 88 Griffiths A. Pityriasis rubra pilaris. Etiologic considerations. J Am
pityriasis rubra pilaris. J Am Acad Dermatol 2011;65:452–3. Acad Dermatol 1984;10:1086–8.
60 Bell SL, Patel AN, Leach IH et al. Consider triggers for pityriasis rubra 89 Cavalie M, Giacchero D, Cardot‐Leccia N et  al. Kaposi’s varicelli­
pilaris. Clin Exp Dermatol 2014;39:403–5. form eruption in a patient with pityriasis rubra pilaris (pityriasis
61 Shvili D, David M, Mimouni M. Childhood‐onset pityriasis rubra pila­ rubra pilaris herpeticum). J Eur Acad Dermatol Venereol
ris with immunologic abnormalities. Pediatr Dermatol 1987;4:21–3. 2013;27:1585–6.
62 Marrouche N, Kurban M, Kibbi AG et al. Pityriasis rubra pilaris: clin­ 90 Erdag G, Lockman D, Tromberg J et  al. A case of pityriasis rubra
icopathological study of 32 cases from Lebanon. Int J Dermatol pilaris with associated focal acantholytic dyskeratosis complicated
2014;53:434–9. by Kaposi’s varicelliform eruption. J Cutan Pathol 2011;38:919–22.
63 Magro CM, Crowson AN. The clinical and histomorphological fea­ 91 Kurzydlo AM, Gillespie R. Paraneoplastic pityriasis rubra pilaris in
tures of pityriasis rubra pilaris. A comparative analysis with psoriasis. association with bronchogenic carcinoma. Australas J Dermatol
J Cutan Pathol 1997;24:416–24. 2004;45:130–2.
64 Ko CJ, Milstone LM, Choi J et al. Pityriasis rubra pilaris: the clinical 92 Marshall J. Case of pityriasis rubra pilaris with lesions of buccal
context of acantholysis and other histologic features. Int J Dermatol mucosa. AMA Arch Derm Syphilol 1952;66:626–7.
2011;50:1480–5. 93 Martinez Calixto LE, Suresh L, Matsumura E et  al. Oral pityriasis
65 Iraji F, Siadat AH. Pityriasis rubra pilaris following exposure to dolo­ rubra pilaris. Oral Surg Oral Med Oral Pathol Oral Radiol Endod
mite. J Res Med Sci 2013;18:621–2. 2006; 101: 604–7.
66 Evangelou G, Murdoch SR, Palamaras I et al. Photoaggravated pity­ 94 Sonnex TS, Dawber RP, Zachary CB et al. The nails in adult type 1
riasis rubra pilaris. Photodermatol Photoimmunol Photomed pityriasis rubra pilaris. A comparison with Sezary syndrome and
2005;21:272–4. psoriasis. J Am Acad Dermatol 1986;15:956–60.
67 Iredale HE, Meggitt SJ. Photosensitive pityriasis rubra pilaris. Clin 95 Kitzmann AS, Goins KM, Syed NA et  al. Bilateral herpes simplex
Exp Dermatol 2006;31:36–8. keratitis with unilateral secondary bacterial keratitis and corneal
68 Kaskel P, Peter RU, Kerscher M. Phototesting and phototherapy in perforation in a patient with pityriasis rubra pilaris. Cornea
pityriasis rubra pilaris. Br J Dermatol 2001;144:430. 2008;27:1212–4.
PAPULOSQUAMOUS DISORDERS

69 Marguery MC, Durand‐Malgouyres C, Bayle‐Lebey P et  al. Photo­ 96 Paranjothy B, Shunmugam M, MacKenzie J et al. Peripheral ulcera­
sensitive and phototriggered pityriasis rubra pilaris. Photoder­matol tive keratitis in pityriasis rubra pilaris. Eye 2007;21:1001–2.
Photoimmunol Photomed 1994;10:42–5. 97 Durairaj VD, Horsley MB. Resolution of pityriasis rubra pilaris‐
70 Yaniv R, Barzilai A, Trau H. Pityriasis rubra pilaris exacerbated by induced cicatricial ectropion with systemic low‐dose methotrexate.
SECTION 6: OTHER

ultraviolet B phototherapy. Dermatology 1994;189:313. Am J Ophthalmol 2007;143:709–10.


71 Sehgal VN, Jain S, Kumar S et  al. Familial pityriasis rubra pilaris 98 Haridas AS, Sullivan TJ. Surgical management of cicatricial ectro­
(adult classic‐I): a report of three cases in a single family. Skinmed pion  in pityriasis rubra pilaris. Ophthalmic Plast Reconstr Surg
2002;1:161–4. 2016;32:e12–15.
72 Eytan O, Sarig O, Sprecher E et al. Clinical response to ustekinumab 99 Das JK, Gangopadhyay AK, Sengupta S. Pityriasis rubra pilaris with
in familial pityriasis rubra pilaris caused by a novel mutation in Koebner’s isomorphic phenomenon. Indian J Dermatol Venereol
CARD14. Br J Dermatol 2014;171:420–2. Leprol 2010;76:194–6.
73 Mercer JM, Pushpanthan C, Anandakrishnan C et al. Familial pity­ 100 Lambert DG, Dalac S. Nail changes in type 5 pityriasis rubra pilaris.
riasis rubra pilaris: case report and review. J Cutan Med Surg J Am Acad Dermatol 1989;21:811–2.
2013;17:226–32. 101 Baden HP, Roth SI. Oral lesions in pityriasis rubra pilaris. Oral Surg
74 Bertin J, Wang L, Guo Y et al. CARD11 and CARD14 are novel caspase Oral Med Oral Pathol 1968;25:691–4.
recruitment domain (CARD)/membrane‐associated guanylate kinase 102 Massa AF, Vasconcelos P, Soares de Almeida L et al. Pityriasis rubra
(MAGUK) family members that interact with BCL10 and activate NF‐ pilaris mixed type III/IV successfully treated with narrow band‐
kappa B. J Biol Chem 2001;276:11877–82. ultraviolet B. Indian J Dermatol Venereol Leprol 2015;81:435.
75 Scudiero I, Zotti T, Ferravante A et  al. Alternative splicing of 103 Hong JB, Chiu HC, Wang SH et al. Recurrence of classical juvenile
CARMA2/CARD14 transcripts generates protein variants with dif­ pityriasis rubra pilaris in adulthood: report of a case. Br J Dermatol
ferential effect on NF‐kappaB activation and endoplasmic reticulum 2007;157:842–4.
stress‐induced cell death. J Cell Physiol 2011;226:3121–31. 104 Caldarola G, Zampetti A, De Simone C et al. Circumscribed pityria­
76 Blonska M, Lin X. NF‐kappaB signaling pathways regulated by sis rubra pilaris type IV. Clin Exp Dermatol 2007;32:471–2.
CARMA family of scaffold proteins. Cell Res 2011;21:55–70. 105 Lopez‐Gomez A, Vera‐Casano A, Gomez‐Moyano E et  al.
77 Seitz CS, Freiberg RA, Hinata K et  al. NF‐kappaB determines Dermoscopy of circumscribed juvenile pityriasis rubra pilaris. J Am
­localization and features of cell death in epidermis. J Clin Invest Acad Dermatol 2015;72:S58–9.
2000;105:253–60. 106 Lernia VD, Ficarelli E, Zanelli M. Ineffectiveness of tumor necrosis
78 Wullaert A, Bonnet MC, Pasparakis M. NF‐kappaB in the regulation factor‐alpha blockers and ustekinumab in a case of type IV pityriasis
of epithelial homeostasis and inflammation. Cell Res 2011;21:146–58. rubra pilaris. Indian Dermatol Online J 2015;6:207–9.
79 Li Q, Jin Chung H, Ross N et al. Analysis of card14 polymorphisms in 107 Yun CH, Kim JS, Ryu HR et  al. Circumscribed juvenile pityriasis
pityriasis rubra pilaris: activation of NF‐kappaB. J Invest Dermatol rubra pilaris responsive to alitretinoin. Dermatol Ther 2016;29:81–3.
2015;135:1905–8. 108 Martin KL, Holland KE, Lyon V et al. An unusual cluster of circum­
80 Hong JB, Chen PL, Chen YT et al. Genetic analysis of CARD14 in non‐ scribed juvenile pityriasis rubra pilaris cases. Pediatr Dermatol
familial pityriasis rubra pilaris: a case series. Acta Derm Venereol 2014;31:138–45.
2014;94:587–8. 109 Resnick SD, Murrell DF, Woosley JT. Pityriasis rubra pilaris, acne
81 Eytan O, Qiaoli L, Nousbeck J et al. Increased epidermal expression conglobata, and elongated follicular spines: an HIV‐associated folli­
and absence of mutations in CARD14 in a series of patients with spo­ cular syndrome? J Am Acad Dermatol 1993;29:283.
radic pityriasis rubra pilaris. Br J Dermatol 2014;170:1196–8. 110 Gonzalez‐Lopez A, Velasco E, Pozo T et al. HIV‐associated pityriasis
82 Jordan CT, Cao L, Roberson ED et al. Rare and common variants in rubra pilaris responsive to triple antiretroviral therapy. Br J Dermatol
CARD14, encoding an epidermal regulator of NF‐kappaB, in psoria­ 1999;140:931–4.
sis. Am J Hum Genet 2012;90:796–808. 111 Cohen PR, Prystowsky JH. Pityriasis rubra pilaris: a review of diag­
83 Jordan CT, Cao L, Roberson ED et al. PSORS2 is due to mutations in nosis and treatment. J Am Acad Dermatol 1989;20:801–7.
CARD14. Am J Hum Genet 2012;90:784–95. 112 Gleeson CM, Chan I, Griffiths WA et al. Eruptive seborrhoeic kera­
84 Lim JT, Tham SN. Pityriasis rubra pilaris in Singapore. Clin Exp toses associated with erythrodermic pityriasis rubra pilaris. J Eur
Dermatol 1991;16:181–4. Acad Dermatol Venereol 2009;23:217–8.
Chapter 32  Pityriasis Rubra Pilaris 387

113 Sahin MT, Ozturkcan S, Ermertcan AT et  al. Transient eruptive 141 Canavan T, Sidorsky T, Doan LT et al. A case of Wong‐type dermato­
seborrhoeic keratoses associated with erythrodermic pityriasis
­ myositis with concomitant anti‐MDA5 features. J Am Acad Dermatol
rubra pilaris. Clin Exp Dermatol 2004;29:554–5. 2014;70:e62–4.
114 Scheinfeld N. Diseases associated with hidranitis suppurativa: part 2 142 Haro R, Revelles JM, Farina Mdel C et al. Wong’s dermatomyositis: a
of a series on hidradenitis. Dermatol Online J 2013;19:18558. new case and review of the literature. Int J Dermatol 2013;52:466–70.
115 Batchelor RJ, Yung A, Merchant W et  al. Pityriasis rubra pilaris as 143 de la Tribonniere X, Delaporte E, Alfandari S et al. Dermatomyositis
the initial presentation of renal cell carcinoma? Clin Exp Dermatol with follicular hyperkeratosis. Dermatology 1995;191:242–4.
2005;30:442–3. 144 Dupre A, Floutard M, Christol B et  al. [Dermatomyositis with
116 Kloos C, Muller UA, Hoffken K et al. [Paraneoplastic pityriasis rubra ­spinulosis (Wong type dermatomyositis). Histologic study. Attempt
pilaris in metastatic adenocarcinoma without diagnosable primary]. at pathogenic interpretation]. Ann Dermatol Syphiligr (Paris)
Dtsch Med Wochenschr 2002;127:437–40. 1976;103:141–9.
117 Sharma S, Weiss GR, Paulger B. Pityriasis rubra pilaris as an i­ nitial pres­ 145 Franzotti AM, Avelar JC, Cardoso TA et al. Pityriasis rubra pilar and
entation of hepatocellular carcinoma. Dermatology 1997;194:166–7. hypothyroidism. An Bras Dermatol 2014; 89: 497–500.
118 Tannenbaum CB, Billick RC, Srolovitz H. Multiple cutaneous 146 Castanet J, Lacour JP, Perrin C et al. Juvenile pityriasis rubra pilaris
malignancies in a patient with pityriasis rubra pilaris and focal
­ associated with hypogammaglobulinaemia and furunculosis. Br J
­acantholytic dyskeratosis. J Am Acad Dermatol 1996;35:781–2. Dermatol 1994; 131: 717–9.
119 Batinac T, Kujundzic M, Peternel S et al. Pityriasis rubra pilaris in asso­ 147 Taniguchi S, Kawahira K, Kaneto K et  al. Juvenile pityriasis rubra
ciation with laryngeal carcinoma. Clin Exp Dermatol 2009;34:e917–9. pilaris with isolated IgA deficiency. Eur J Pediatr 1997;156:896.
120 Huynh NT, Hunt MJ, Cachia AR et  al. Merkel cell carcinoma and 148 Walsh NM, Prokopetz R, Tron VA et al. Histopathology in erythro­
multiple cutaneous squamous cell carcinomas in a patient with derma: review of a series of cases by multiple observers. J Cutan
­pityriasis rubra pilaris. Australas J Dermatol 2002;43:48–51. Pathol 1994;21:419–23.
121 Sanchez‐Regana M, Lopez‐Gil F, Salleras M et  al. Pityriasis rubra 149 Braun‐Falco O, Ryckmanns F, Schmoeckel C et  al. Pityriasis rubra
pilaris as the initial manifestation of internal neoplasia. Clin Exp pilaris: a clinico‐pathological and therapeutic study with special ref­
Dermatol 1995;20:436–8. erence to histochemistry, autoradiography, and electron microscopy.
122 Remedios IM, Jensen JD, Beckum K et al. Paraneoplastic pityriasis Arch Dermatol Res 1983;275:287–95.
rubra pilaris as the presenting manifestation of metastatic squamous 150 Soeprono FF. Histologic criteria for the diagnosis of pityriasis rubra
cell carcinoma. J Drugs Dermatol 2014;13:610–2. pilaris. Am J Dermatopathol 1986;8:277–83.
123 Quenan S, Kaya G, Laffitte E. Pityriasis rubra pilaris and transi­ 151 Avitan‐Hersh E, Bergman R. The incidence of acantholysis in
tional  meningioma: a fortuitous association? Eur J Dermatol ­pityriasis rubra pilaris‐histopathological study using multiple‐step
2015;25:273–4. sections and clinicopathologic correlations. Am J Dermatopathol
124 Behr FD, Bangert JL, Hansen RC. Atypical pityriasis rubra pilaris 2015;37:755–8.

PAPULOSQUAMOUS DISORDERS
associated with arthropathy and osteoporosis: a case report with 152 Sebastian A, Koff AB, Goldberg LJ. PRP with subcorneal acantholy­
15‐year follow‐up. Pediatr Dermatol 2002;19:46–51. sis: case report and review. J Cutan Pathol 2010;37:99–101.
125 Caporali R, Cavagna L, Bellosta M et al. Inflammatory myopathy in 153 Hashimoto K, Fedoronko L. Pityriasis rubra pilaris with acantholysis

SECTION 6: OTHER
a patient with cutaneous findings of pityriasis rubra pilaris: a case of and lichenoid histology. Am J Dermatopathol 1999;21:491–3.
Wong’s dermatomyositis. Clin Rheumatol 2004;23:63–5. 154 Howe K, Foresman P, Griffin T et  al. Pityriasis rubra pilaris with
126 Conaghan PG, Sommer S, McGonagle D et  al. The relationship acantholysis. J Cutan Pathol 1996;23:270–4.
between pityriasis rubra pilaris and inflammatory arthritis: case 155 Rashidghamat E, Griffiths WA, Mellerio JE et  al. Pityriasis rubra
report and response of the arthritis to anti‐tumor necrosis factor pilaris with histologic features of lichen nitidus. J Am Acad Dermatol
immunotherapy. Arthritis Rheum 1999;42:1998–2001. 2015;73:336–7.
127 Fiallo P, Tagliapietra AG, Santoro G. Arthropathic pityriasis rubra 156 Hunter I, Skerrow D. The effect of increased tissue turnover on
pilaris. Br J Dermatol 1996;134:1154–5. the  keratinization of human epidermis. Biochem Biophys Acta
128 Gul U, Gonul M, Kilic A et  al. A case of pityriasis rubra pilaris 1981;674:155–9.
­associated with sacroileitis and autoimmune thyroiditis. J Eur Acad 157 Ralfs IG, Dawber RP, Ryan TJ et al. Pityriasis rubra pilaris: epidermal
Dermatol Venereol 2008;22:889–90. cell kinetics. Br J Dermatol 1981;104:249–52.
129 Lister RK, Perry JD, Cerio R. Pityriasis rubra pilaris and a seronega­ 158 Griffiths WA, Pieris S. Pityriasis rubra pilaris—an autoradiographic
tive polyarthritis. Br J Dermatol 1997;137:318–9. study. Br J Dermatol 1982; 107: 665–7.
130 Nakafusa J, Misago N, Narisawa Y. Pityriasis rubra pilaris in 159 Baran W, Szepietowski JC, Szybejko‐Machaj G. Enhanced expression
­association with polyarthritis. Dermatology 2002;205:298–300. of p53 protein in pityriasis rubra pilaris. Acta Derm Venereol
131 Orlandini V, Cogrel O, Doutre MS et al. Pityriasis rubra pilaris and 2006;86:276–7.
hypothyroidism. Efficacy of thyroid hormone replacement therapy 160 Karimian‐Teherani D, Parissa M, Tanew A. Response of juvenile cir­
in skin recovery. Br J Dermatol 2007;156:606–7. cumscribed pityriasis rubra pilaris to topical tazarotene treatment.
132 Polat M, Lenk N, Ustun H et al. Dermatomyositis with a pityriasis Pediatr Dermatol 2008;25:125–.
rubra pilaris‐like eruption: an uncommon cutaneous manifestation 161 Van de Kerkhof PC, Steijlen PM. Topical treatment of pityriasis rubra
in dermatomyositis. Pediatr Dermatol 2007;24:151–4. pilaris with calcipotriol. Br J Dermatol 1994;130:675–8.
133 Rasi A, Soltani‐Arabshahi R, Naraghi ZS. Circumscribed juvenile‐ 162 Gregoriou S, Argyriou G, Christofidou E et al. Treatment of pityria­
onset pityriasis rubra pilaris with hypoparathyroidism and brachyo­ sis rubra pilaris with pimecrolimus cream 1%. J Drugs Dermatol
nychia. Cutis 2006;77:218–22. 2007;6:340–2.
134 Menshikova AK, Kuklin VT, Men’shikova NE et al. [Systemic lupus 163 Neess CM, Hinrichs R, Dissemond J et  al. Treatment of pruritus
erythematosus associated with Devergie’s disease]. Vestn Dermatol by  capsaicin in a patient with pityriasis rubra pilaris receiving
Venerol 1985: 68–70. RE‐PUVA therapy. Clin Exp Dermatol 2000;25:209–11.
135 Randle HW, Winkelmann RK. Pityriasis rubra pilaris and celiac 164 Atanaskova Mesinkovska N, Dawes D, Sood A et  al. Acantholytic
sprue with malabsorption. Cutis 1980;25:626–7. pityriasis rubra pilaris associated with imiquimod 3.75% applica­
136 Agharbi FZ, Elbekkal A, Baybay H et  al. [Association of pityriasis tion. Case Rep Dermatol Med 2011;2011:412684.
rubra pilaris and myasthenia]. Pan Afr Med J 2011;10:7. 165 Kirby B, Watson R. Pityriasis rubra pilaris treated with acitretin
137 Sato T, Sakai H, Iida H et al. Protein‐losing gastroenteropathy in a and  narrow‐band ultraviolet B (Re‐TL‐01). Br J Dermatol
patient with pityriasis rubra pilaris. Digestion 2007;75:98. 2000;142:376–7.
138 Lin CY, Maurice PD, Cross NB. A case of pityriasis rubra pilaris asso­ 166 Vergilis‐Kalner IJ, Mann DJ, Wasserman J et  al. Pityriasis rubra
ciated with membranous nephropathy. Australas J Dermatol ­pilaris sensitive to narrow band‐ultraviolet B light therapy. J Drugs
2013;54:e1–3. Dermatol 2009;8:270–3.
139 Kim JH, Park MC, Kim SC. Etanercept‐induced clinical remission of 167 Herbst RA, Vogelbruch M, Ehnis A et  al. Combined ultraviolet A1
type II pityriasis rubra pilaris with rheumatoid arthritis. Acta Derm radiation and acitretin therapy as a treatment option for pityriasis
Venereol 2012;92:399–400. rubra pilaris. Br J Dermatol 2000;142:574–5.
140 Alcala R, Noguera‐Morel L, Clemente D et al. Pityriasis rubra pilaris‐ 168 Kaskel P, Grundmann‐Kollmann M, Schiller PI et al. Bath‐PUVA as a
like eruption with dermatomyositis and diffuse cutaneous systemic treatment for pityriasis rubra pilaris provoked by ultraviolet B. Br J
sclerosis overlap. J Eur Acad Dermatol Venereol 2016;30:1059–61. Dermatol 1999;140:769–70.
388  Section 6 Other Papulosquamous Disorders

169 Terheyden P, Hofmann UB, Hamm H et al. [Erythroderma after cor­ 195 Ruiz‐Genao DP, Lopez‐Estebaranz JL, Naz‐Villalba E et al. Pityriasis
ticosteroid withdrawal in papulosquamous dermatosis in children. rubra pilaris successfully treated with infliximab. Acta Derm
Diagnosis: pityriasis rubra pilaris, classic juvenile type]. Hautarzt Venereol 2007;87:552–3.
2001;52:1058–61. 196 Zirbs M, Kigitsidou E, Seifert F et al. Successful treatment with inf­
170 Borok M, Lowe NJ. Pityriasis rubra pilaris. Further observations of liximab despite positive tuberculosis ELISpot results in a patient
systemic retinoid therapy. J Am Acad Dermatol 1990;22:792–5. with pityriasis rubra pilaris taking prophylactic isoniazid. Clin Exp
171 Goldsmith LA, Weinrich AE, Shupack J. Pityriasis rubra pilaris Dermatol 2011;36:808–9.
response to 13‐cis‐retinoic acid (isotretinoin). J Am Acad Dermatol 197 Garcovich S, Di Giampetruzzi AR, Antonelli G et  al. Treatment
1982;6:710–5. of  refractory adult‐onset pityriasis rubra pilaris with TNF‐alpha
172 Gilgor RS, Chiaramonti A, Goldsmith LA et al. Evaluation of 13‐cis antagonists: a case series. J Eur Acad Dermatol Venereol
retinoic acid in lamellar ichthyosis, pityriasis rubra pilaris and 2010;24:881–4.
Darier’s disease. Cutis 1980;25:380–1, 385. 198 Lu R, George SJ, Hsu S. Pityriasis rubra pilaris: failure of combina­
173 van Dooren‐Greebe RJ, van de Kerkhof PC. Extensive extraspinal tion treatment with acitretin and infliximab. Dermatol Online J
hyperostoses after long‐term oral retinoid treatment in a patient 2006;12:18.
with pityriasis rubra pilaris. J Am Acad Dermatol 1995;32:322–5. 199 Dessinioti C, Vergou T, Moustou E et al. Long‐term infliximab treat­
174 Amann PM, Susic M, Gluder F et al. Alitretinoin (9‐cis retinoic acid) ment for refractory type III juvenile pityriasis rubra pilaris. Eur J
is effective against pityriasis rubra pilaris: a retrospective clinical Dermatol 2011;21:599–600.
study. Acta Derm Venereol 2015;95:329–31. 200 Ruzzetti M, Saraceno R, Carboni I et al. Type III juvenile pityriasis
175 Molin S, Ruzicka T. Treatment of refractory pityriasis rubra pilaris rubra pilaris: a successful treatment with infliximab. J Eur Acad
with oral alitretinoin: case report. Br J Dermatol 2010;163:221–3. Dermatol Venereol 2008;22:117–8.
176 Pampin A, Gomez‐de la Fuente E, Caro Gutierrez MD et  al. 201 Davis KF, Wu JJ, Murase JE et al. Clinical improvement of pityriasis
Successful treatment of atypical adult pityriasis rubra pilaris with rubra pilaris with combination etanercept and acitretin therapy.
oral alitretinoin. J Am Acad Dermatol 2013;69:e105–6. Arch Dermatol 2007;143:1597–9.
177 Schmitt L, Inhoff O, Dippel E. Oral alitretinoin for the treatment 202 Seckin D, Tula E, Ergun T. Successful use of etanercept in type I pity­
of  recalcitrant pityriasis rubra pilaris. Case Rep Dermatol riasis rubra pilaris. Br J Dermatol 2008;158:642–4.
2011;3:85–8. 203 Guedes R, Leite L. Therapeutic hotline. Treatment of pityriasis rubra
178 Brecher AR, Orlow SJ. Oral retinoid therapy for dermatologic condi­ pilaris with etanercept. Dermatol Ther 2011;24:285–6.
tions in children and adolescents. J Am Acad Dermatol 2003;49:171– 204 Cox V, Lesesky EB, Garcia BD et  al. Treatment of juvenile
82; quiz 183–6. pityriasis  rubra pilaris with etanercept. J Am Acad Dermatol
­
179 Searles AD, Lee AD, Feldman SR. Is concomitant use of methotrexate 2008;59:S113–4.
and oral retinoids dangerous? A review of the data regarding this 205 Bravo EA, Carrion L, Paucar SM et al. Successful treatment of pity­
PAPULOSQUAMOUS DISORDERS

combination. J Am Acad Dermatol 2011;64:791–3. riasis rubra pilaris with adalimumab ‐ case report. Dermatol Online
180 Rosenbach A, Lowe NJ. Pityriasis rubra pilaris and cyclosporine. J l 2014;20:22374.
Arch Dermatol 1993;129:1346–8. 206 Chiu HY, Tsai TF. Pityriasis rubra pilaris with polyarthritis treated
181 Wetzig T, Sticherling M. Juvenile pityriasis rubra pilaris: successful with adalimumab. J Am Acad Dermatol 2013;68:187–8.
SECTION 6: OTHER

treatment with ciclosporin. Br J Dermatol 2003;149:202–3. 207 O’Kane D, Devereux CE, Walsh MY et al. Rapid and sustained remis­
182 Usuki K, Sekiyama M, Shimada T et  al. Three cases of pityriasis sion of pityriasis rubra pilaris with adalimumab treatment. Clin Exp
rubra pilaris successfully treated with cyclosporin A. Dermatology Dermatol 2010;35:e155–6.
2000;200:324–7. 208 Schreml S, Zeller V, Babilas P et al. Pityriasis rubra pilaris success­
183 Hunter GA, Forbes IJ. Treatment of pityriasis rubra pilaris with aza­ fully treated with adalimumab. Clin Exp Dermatol 2010;35:792–3.
thioprine. Br J Dermatol 1972;87:42–5. 209 Walling HW, Swick BL. Pityriasis rubra pilaris responding rapidly to
184 Coras B, Vogt TH, Ulrich H et al. Fumaric acid esters therapy: a new adalimumab. Arch Dermatol 2009;145:99–101.
treatment modality in pityriasis rubra pilaris? Br J Dermatol 210 Wassef C, Lombardi A, Rao BK. Adalimumab for the treatment of
2005;152:388–9. pityriasis rubra pilaris: a case report. Cutis 2012;90:244–7.
185 Ivanova K, Itin P, Haeusermann P. Pityriasis rubra pilaris: treatment 211 Zhang YH, Zhou Y, Ball N et al. Type I pityriasis rubra pilaris: upreg­
with biologics  –  a new promising therapy? Dermatology ulation of tumor necrosis factor alpha and response to adalimumab
2012;224:120–5. therapy. J Cutan Med Surg 2010;14:185–8.
186 Petrof G, Almaani N, Archer CB et al. A systematic review of the lit­ 212 Kim BR, Chae JB, Park JT et al. Clinical remission of pityriasis rubra
erature on the treatment of pityriasis rubra pilaris type 1 with TNF‐ pilaris with adalimumab in an adolescent patient. J Dermatol
antagonists. J Eur Acad Dermatol Venereol 2013;27:e131–5. 2015;42:1122–3.
187 Lories RJ, Derese I, Luyten FP et  al. Activation of nuclear factor 213 Byekova Y, Sami N. Successful response of refractory type I adult‐
kappa B and mitogen activated protein kinases in psoriatic arthritis onset pityriasis rubra pilaris with ustekinumab and acitretin combi­
before and after etanercept treatment. Clin Exp Rheumatol nation therapy. J Dermatol 2015;42:830–1.
2008;26:96–102. 214 Chowdhary M, Davila U, Cohen DJ. Ustekinumab as an alternative
188 Adnot‐Desanlis L, Antonicelli F, Tabary T et al. Effectiveness of inf­ treatment option for chronic pityriasis rubra pilaris. Case Rep
liximab in pityriasis rubra pilaris is associated with pro‐inflamma­ Dermatol 2015;7:46–50.
tory cytokine inhibition. Dermatology 2013;226:41–6. 215 Di Stefani A, Galluzzo M, Talamonti M et al. Long‐term ustekinumab
189 Barth D, Harth W, Treudler R et al. Successful treatment of pityriasis treatment for refractory type I pityriasis rubra pilaris. J Dermatol
rubra pilaris (type 1) under combination of infliximab and metho­ Case Rep 2013;7:5–9.
trexate. J Dtsch Dermatol Ges 2009;7:1071–4. 216 Ruiz Villaverde R, Sanchez Cano D. Successful treatment of type 1
190 Drosou A, Kirsner RS, Welsh E et al. Use of infliximab, an anti‐tumor pityriasis rubra pilaris with ustekinumab therapy. Eur J Dermatol
necrosis alpha antibody, for inflammatory dermatoses. J Cutan Med 2010;20:630–1.
Surg 2003;7:382–6. 217 Wohlrab J, Kreft B. Treatment of pityriasis rubra pilaris with usteki­
191 Karadag AS, Kavala M, Ozlu E et al. Erythrodermic pityriasis rubra numab. Br J Dermatol 2010;163:655–6.
pilaris: Dramatic response to infliximab therapy. Indian J Dermatol 218 Haenssle HA, Bertsch HP, Emmert S et  al. Extracorporeal photo­
Venereol Leprol 2016;82:112. chemotherapy for the treatment of exanthematic pityriasis rubra
192 Liao WC, Mutasim DF. Infliximab for the treatment of adult‐onset pilaris. Clin Exp Dermatol 2004;29:244–6.
pityriasis rubra pilaris. Arch Dermatol 2005;141:423–5. 219 Hofer A, Mullegger R, Kerl H et al. Extracorporeal photochemother­
193 Manoharan S, White S, Gumparthy K. Successful treatment of type I apy for the treatment of erythrodermic pityriasis rubra pilaris. Arch
adult‐onset pityriasis rubra pilaris with infliximab. Australas J Dermatol 1999;135:475–6.
Dermatol 2006;47:124–9. 220 Kerr AC, Ferguson J. Type II adult‐onset pityriasis rubra pilaris suc­
194 Muller H, Gattringer C, Zelger B et  al. Infliximab monotherapy as cessfully treated with intravenous immunoglobulin. Br J Dermatol
first‐line treatment for adult‐onset pityriasis rubra pilaris: case 2007;156:1055–6.
report and review of the literature on biologic therapy. J Am Acad 221 Lerebours‐Nadal L, Beck‐Sague CM, Parker D et al. Severe, disfigur­
Dermatol 2008;59:S65–70. ing, pityriasis rubra pilaris in a woman in the Dominican Republic:
Chapter 32  Pityriasis Rubra Pilaris 389

histopathologic diagnosis and response to antiretroviral therapy. 224 Demonchy E, Lacour JP, Ortonne JP et al. Erythema gyratum repens,
J Int Assoc Provid AIDS Care 2016;15:11–14. not always a bad omen for patients. J Eur Acad Dermatol Venereol
222 Almaani N, Robson A, Sarkany R et  al. Erythema gyratum repens 2010;24:738–9.
associated with pityriasis rubra pilaris. Clin Exp Dermatol 225 Gebauer K, Singh G. Resolving pityriasis rubra pilaris resembling
2011;36:161–4. erythema gyratum repens. Arch Dermatol 1993;129:917–8.
223 Cheesbrough MJ, Williamson DM. Erythema gyratum repens, a 226 Marchetti MA, Greer KE. Pityriasis rubra pilaris treated with metho­
stage in the resolution of pityriasis rubra pilaris? Clin Exp Dermatol trexate resolving with an erythema gyratum repens‐like appearance.
1985;10:466–71. J Am Acad Dermatol 2013;69:e32–3.

PAPULOSQUAMOUS DISORDERS
SECTION 6: OTHER
390 

CHA PTER  3 3

Lichen Planus
Vibhu Mendiratta & Sarita Sanke
Lady Hardinge Medical College and associated hospitals, New Delhi, India

Abstract Classic, hypertrophic, annular and linear LP are the common


childhood variants. LP is multifactorial in origin and is thought
Lichen planus (LP) is a papulosquamous autoimmune T‐cell‐ to be a T‐cell‐mediated injury to the basement membrane which
mediated disease that affects skin, mucous membranes, hair results from infections, drugs and vaccinations. The lesions may
follicles and nails. It affects all age groups including children. last for up to 1–2 years, but the hypertrophic, mucosal, nail and
Childhood LP cases can be sporadic or familial. Classic LP is follicular LP variants may last significantly longer. The nail and
characterized by faintly erythematous, violaceous, flat‐topped, follicular variants can lead to the scarring processes of anonychia
polygonal papules that are characteristically pruritic. White and cicatricial alopecia, respectively. Treatment is multidimensional
crisscrossing lines called Wickham’s striae are seen on the and includes topical corticosteroids and calcineurin inhibitors,
surface. Several clinical variants are recognized according to the as well as systemic medications including nonsteroidal anti‐
configuration of the lesions, their morphological appearance inflammatory agents, systemic steroids, dapsone, methotrexate,
and the site involved. Vacuolar degeneration of the basement ciclosporin (cyclosporine), azathioprine and phototherapy. This
membrane with a band‐like lymphocytic infiltrate at the chapter outlines the clinical profile, aetiopathogenesis, course and
dermoepidermal junction constitutes a histopathological hallmark. treatment options in childhood LP.
PAPULOSQUAMOUS DISORDERS

Key points • Several morphological variants are described including annular,


linear, actinic, hypertrophic, atrophic, inverse, bullous, lichen
• Lichen planus (LP) is a papulosquamous disease characterized planus pemphigoides and lichen planopilaris.
SECTION 6: OTHER

by itchy, shiny, violaceous, flat‐topped, polygonal papules and • Diagnosis relies on clinical and histological features. Vacuolar
plaques. Lesions often koebnerize. degeneration of the basement membrane with a band‐
• LP can affect skin, mucous membrane, hair follicles and nails. like lymphocytic infiltrate at the dermoepidermal junction
• LP is a multifactorial disease resulting from an interplay of constitutes the histopathological hallmark.
genetic and environmental factors. The condition can be • Choice of treatment depends on the age of the patient and the
sporadic or familial. disease severity. Therapy is aimed at removal of the triggering
• Prevalence is about 0.5–2% with female to male ratio of factor, topical/oral corticosteroids, topical calcineurin inhibitors,
approximately 2 : 1. Childhood LP is relatively rare. dapsone, phototherapy and immunosuppressive agents.

Introduction. Lichen planus (LP) is a papulosquamous 1895 [2]. Darier is credited with ascribing these striae to
disease of unknown aetiology that affects the skin, hypergranulosis [3].
mucous membranes, hair follicles and nails. It is clinically The term ‘lichenoid’ refers to lesions that morphologi-
characterized by itchy, shiny, violaceous, flat‐topped, cally resemble LP (as grouped, shiny, flat‐topped papules)
polygonal papules and plaques. Vacuolar degeneration of and also describes a specific histopathological pattern
the basement membrane with a band‐like lymphocytic characterized by vacuolar degeneration of the basement
infiltrate at dermoepidermal junction constitutes the membrane along with a dense lymphohistiocytic infil-
­histopathological hallmark of the disease. trate at the dermoepidermal junction. In contrast to LP,
lichenoid lesions do not show the classic Wickham’s striae
History. Lichen planus is derived from the Greek word on their surface.
leichen meaning to lick and planus denoting ‘flat’‐topped
papules. The word leichen was earlier used to refer to a Aetiology and  pathogenesis. LP is a multifactorial dis-
form of symbiotic plant life found on rocks and trees. It ease resulting from an interplay of genetic and environ-
was von Hebra who first used the term ‘leichen ruber’ to mental factors (Box 33.1). Sporadic occurrence is more
refer to LP [1], but it was Erasmus Wilson who coined the common, but occurrence within a family and among
term ‘lichen planus’ in 1869. Wickham later described monozygotic twins points to a genetic linkage [4]. Familial
‘dots and streaks over the lesions’ as Wickham’s striae in LP comprises 1–2% of all childhood cases and has an early

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 33  Lichen Planus 391

The autoimmune mechanism of LP was first proposed


Box 33.1  Aetiological factors in the pathogenesis of lichen
by Black in 1977 based on the coexistence of LP with
planus
disorders such as myasthenia gravis, alopecia areata,
­
Predisposing factors
vitiligo and ulcerative colitis [12]. Current evidence
­
­supports that LP is a T‐cell‐mediated disease. Several trig-
Genetic association: HLA‐B7, HLA‐3, HLA‐5, HLA‐DR6, HLA‐DRB1 gers have been identified, including viruses, drugs, radia-
Gene polymorphisms tion and contact sensitivity to metals such as mercury
Autoimmunity amalgam, copper, gold, palladium and beryllium, that
alter antigenicity of epidermal keratinocytes and initiate
Infectious agents
T‐cell activation. Active LP shows increased expression of
Viral major histocompatibility complex class (MHC) I antigen,
Hepatitis B CD8+ T cells, activated macrophages and Langerhans
Hepatitis C cells. CD8+ T cells from lesional skin of LP have shown
Human herpesvirus‐7 ­cytotoxic activity against lesional and normal keratino-
Human papillomavirus cytes [13]. Keratinocyte apoptosis results from damage
caused by cytotoxic T cells [14].
Bacterial
Several reports describe LP following hepatitis B vac-
Staphylococcus
Streptococcus
cination, combined measles–mumps–rubella vaccine and
hepatitis C infection [15–18]. The possible association of
Vaccinations recombinant hepatitis B virus (HBV) vaccines with child-
hood LP or LP‐like eruptions has been scrutinized in only
Hepatitis B biopsy‐proven cases of LP over a period of 3 years, and
Measles–mumps–rubella five patients <16 years of age were reported in whom
Diphtheria–pertussis–tetanus
such an association could be supported by relevant

PAPULOSQUAMOUS DISORDERS
Polio
data  [19]. Cross‐reactivity between hepatitis B antigen
Influenza
and epitopes in the keratinocytes could possibly trigger
autoimmunity. Kanwar and De reported an average inter-

SECTION 6: OTHER
Environmental agents
val of 3 years between hepatitis B vaccination and LP [20].
Trauma LP has also been linked with upper respiratory tract infec-
Radiation exposure tion and viral exanthems by Nanda et al. [21]. However
other studies have failed to identify any precipitating
Drugs (see Box 33.4 for additional detail) ­factor in childhood LP [20,22,23].
Angiotensin‐converting enzyme inhibitors The association of LP with liver disease (chronic active
Anti‐inflammatory drugs hepatitis, hepatitis B and hepatitis C infection) continues to
Antibiotics be contentious [24] (Box  33.2) Autoimmune mechanisms
Antidiabetic agents can support the association of LP with chronic active
Antifungals hepatitis (CAH) [25]. There appears to be a relationship
Anticonvulsants between LP and hepatitis C virus as a Nigerian study found
Antimalarials higher prevalence rates among affected subjects as
Antimycobacterial agents compared to the control group [26]. A recently conducted
Diuretics meta‐analysis and systematic review of case‐control studies
Lipid‐lowering agents examined the prevalence of anti‐HCV antibodies in the
Metals serum of cases and controls to determine the association
Miscellaneous drugs between hepatitis C virus and oral LP and showed a
statistically significant difference in the proportion of HCV
Others
seropositivity among oral lichen planus patients, compared
Psychological stress with controls. This points towards a link between hepatitis
Graft‐versus‐host disease C virus infection and oral lichen planus [27].
Restorative dental materials
Gold, mercury Epidemiology. The prevalence of lichen planus is about
0.5–2% with the female‐to‐male ratio approximating 2 : 1
[28,29]. LP is common in adults whereas children comprise
less than 2–3% of the total cases [29]. LP is relatively uncom-
onset, a more generalized distribution, a longer relapsing mon in the western hemisphere, but is more c­ommonly
course and a greater propensity for mucosal involvement encountered in Asian countries including the Middle East
[5,6]. Several HLA types show a probable association with and India [20,22,30]. There is a higher prevalence of child-
LP. These include HLA‐3, HLA‐5 and HLAB‐7 [7–9]. An hood LP among those from the Indian subcontinent. In one
association between HLA‐DR1 and HLA‐DR‐10 was particular study utilizing UK census data, the majority of
reported among those of Arabic descent and HLA‐ children included in the census data were white, but 80.8%
DRB*0101 in those of Mexican descent [10,11]. of those with LP were from the Indian subcontinent [31].
392 Section 6  Other Papulosquamous Disorders

The natural course of disease in children and adults


Box 33.2  Various conditions associated with lichen planus
varies. In adults, lesions may persist from a few months to
even years and may be marked by acute flares or exacer-
Infections
bations. There is subsidence of activity in 65–90% of
Hepatitis B patients after 1 year [33]. One study revealed that patients
Hepatitis C with cutaneous lesions were more likely to have a shorter
Chronic active hepatitis course of disease compared to those having mucosal LP.
Epstein–Barr virus infection There was a recurrence of disease in a small percentage of
Helicobacter pylori patients, especially among those with generalized cutane-
ous involvement [34].
Autoimmune

Thymoma Clinical presentation.


Pemphigus LP consists of discrete or grouped, shiny, erythematous
Lupus erythematosus to violaceous, polygonal, flat‐topped, itchy papules with
Alopecia areata a whitish network of Wickham’s striae on the surface.
Hypothyroidism Lesions are distributed over flexor surfaces of the wrists,
Ulcerative colitis forearms, the back and the legs. This constitutes the clas-
Coeliac disease sic variant of LP which is the most common presenta-
Myasthenia gravis tion, representing about 42–76% of LP cases (Fig.  33.1)
Vitiligo
[20–23]. Approximately 6–28% of childhood LP cases
Psoriasis
demonstrate lesions following a linear pattern [20,22,25].
Autoimmune polyendocrinopathy
Extracutaneous sites such as the mucosae (oral and
Dermatitis herpetiformis
genital), scalp and nails can also be involved.
­
Morphoea
Dermoscopy reveals small pinpoint vessels associated
PAPULOSQUAMOUS DISORDERS

Others
with whitish striations (projections) with a ‘fern leaf’
aspect on an erythematous background. Papules may
Atopic dermatitis coalesce to form plaques of varied morphologies, such
SECTION 6: OTHER

Dyslipidaemia as annular, hypertrophic, linear and atrophic. Linear and


Psychological stress blaschkoid LP can also be observed in individual
Lichen sclerosus et atrophicus patients. Annular lesions result from convergence of LP
Anxiety/depression
papules which form round, ring‐shaped lesions with
central clearing with a peripheral, raised rim studded
with lichenoid papules. Genitalia and trunk are common
The mean age of onset varies from 7.1 to 8.4 years [20–22]. sites. Annular LP resembles granuloma annulare and
LP is rare in infants: the youngest reported case was a tinea corporis. Lichen planus hypertrophicus is noted as
3‐month‐old infant [32]. An equal sex distribution is noted, itchy, raised, indurated, hypertrophic plaques and
but boys have outnumbered girls in a ratio of 2 : 1 in some ­nodules over shins and ankles which heal with postin-
studies, possibly due to a reporting bias [23]. LP occurs in a flammatory hyperpigmentation [23]. Actinic LP is seen
sporadic manner; however, familial forms have been over the face and extremities [23]. Greyish‐white plaques
described in 1–2% of all childhood cases [5,20,21]. or patches with superficial atrophy characterize the

Fig. 33.1  Classic variant of lichen planus showing


Wickham’s striae.
Chapter 33  Lichen Planus 393

atrophic variant of LP. Nail and scalp LP, bullous LP and may affect both the nail bed and nail matrix. NLP may be
mucosal LP are less common in children. Oral LP is limited to one nail (12% of NLP cases) or affect all finger-
­characterized by violaceous plaques with a reticulate nails and toenails (10% of NLP cases). Isolated toenail LP
lacy pattern as well as erosions or ulcerative lesions is rare (6% of NLP cases) [36,37]. Characteristic features of
which can result in discomfort when eating spicy food. nail LP include longitudinal striations, ridging, nail thin-
ning and pterygium formation (Fig.  33.2a). Nonspecific
features such as pitting, yellow discolouration, dystrophy,
Clinical variants (Box 33.3)
onycholysis and onychomadesis can also be seen
Nail lichen planus (NLP) (Fig.  33.2b) [38]. A recent report identified two children
The nails are involved in approximately 19% of those with onychomadesis with generalized eruptive LP [39].
affected [20,35]. NLP is more common than mucocutane- Peluso and Tosti et al. identified three patterns of nail
ous LP in children [4]. Underdiagnosis as well as misdiag- LP in children: so‐called typical nail changes of LP, 20‐nail
nosis as onychomycosis is common, possibly due in part dystrophy (trachyonychia) and idiopathic atrophy of
to the reluctance to perform nail biopsies in children. Nail nails [37]. Trachyonychia is seen more commonly in
involvement may be isolated without cutaneous LP. It ­children than adults and may involve all 20 nails with a
lustreless, roughened and brittle nail plate. The idiopathic
variety of trachyonychia is more common in children and
many of these cases show spontaneous resolution.
Box 33.3  Clinical variants of lichen planus (LP) Trachyonychia is also seen in psoriasis, alopecia areata
and atopic dermatitis. The performance of nail biopsies in
According to morphology children is controversial. In cases where nails show typi-
Classic papulosquamous type
cal plate thinning, longitudinal ridging and fissuring, nail
Hypertrophic LP
biopsies can usually be avoided. When necessary, Kanwar
and De and Goettmann et al. suggest performing several

PAPULOSQUAMOUS DISORDERS
Atrophic LP
Eruptive LP 3‐mm punch biopsies during one procedure to increase
Linear and zosteriform LP the possibility of finding a typical histopathological image
Annular LP for LP [36,40]. Nakamura et al. analysed the dermoscopic

SECTION 6: OTHER
Actinic LP abnormalities observed in NLP and their diagnostic
Follicular LP relevance [41]. Nail biopsies are typically avoided in
­
Bullous LP ­trachyonychia as many childhood cases improve sponta-
LP pemphigoides neously over the years [42].
Miliary/micropapular LP Corticosteroids form the cornerstone of treatment. Oral
LP pigmentosus prednisone or prednisolone at 0.5 mg/kg or intramuscu-
lar triamcinolone acetonide at 0.5–1 mg/kg/month has
According to sites involved been used over a period of 5–7 months. Sixty five percent
Oral LP of patients respond to a 5–7 month course of systemic
Reticular ­corticosteroids [21]. Dapsone may represent an effective
Plaque steroid‐sparing alternative [43]. Local treatment is often
Erosive not particularly effective. Local treatment options include
Atrophic intralesional injections of triamcinolone acetonide or
Bullous ­topical corticosteroids. Prevost and English reported a
Genital LP case that responded well to topical therapy with clobet-
Erosive asol gel and tazarotene gel [44]. Pinter et al. described a
Annular good clinical response to topical alitretinoin in NLP [45].
Atrophic Relapses occur in 55% of cases and are usually observed
Hypertrophic in the first year after finishing treatment.
Vulvo‐vaginal‐gingival syndrome
Scalp LP Follicular lichen planus
Lichen planopilaris
Follicular lichen planus or lichen planopilaris (LPP) is
Nail LP
considered rare in children. In a report of 30 cases of LPP
Nail plate thinning
by Chieregato et  al., only two patients were under 18
Pterygium
years of age and both had isolated involvement of the ver-
Longitudinal ridging
tex scalp [46]. A retrospective chart review of LPP cases
Onycholysis
Longitudinal melanonychia
over 37 years identified only four cases of LP from 13 to 16
Nail dystrophy
years [47]. Scalp involvement can be patchy or diffuse.
Palms/soles LPP can be symptomatic or involve scalp dysaesthesia
Diffuse keratoderma and pruritus. Follicular papules with perifollicular scales
Yellow/red scaly plaques and violaceous plaques associated with loss of hair con-
Punctate keratosis stitute early signs. Loss of follicular ostia and epidermal
atrophy are seen in the later stages (Fig. 33.3). A hair pull
394 Section 6  Other Papulosquamous Disorders

(a)
PAPULOSQUAMOUS DISORDERS
SECTION 6: OTHER

Fig. 33.2  (a) Thinning of the nail plate in nail


lichen planus. (b) Nail lichen planus showing
(b) dystrophy of the nails.

psoriasis is known to be a potential cause of lichen plano-


pilaris. Paradoxical LPP was reported to arise de novo in
an 8‐year‐old girl undergoing treatment with adali-
mumab for her arthritis [49]. It has also been reported
after TNF‐α [50]. Trichoscopy reveals multiple irregular
cicatricial alopecic areas with perifollicular whitish‐grey
scaling associated with erythema, arborizing vessels,
absence of follicular openings and follicular plugging.
The impact of cosmetic disfigurement is immense, and it
can impair the quality of life in children.

Oral lichen planus


Oral LP in childhood is exceptionally rare (<2–3% of the
total) [51]. The majority of reported cases are from India
with some from the UK, Italy, Mexico, Africa, the Americas
and Kuwait. The buccal mucosa, tongue, lips, hard palate
and gingivae can be involved. Reticulated papules with a
Fig. 33.3  Lichen planopilaris of the scalp showing cicatricial alopecia and fine, white lacy pattern in the buccal mucosa are the most
hyperpigmentation. common manifestation [52] (Fig.  33.4a). Erosive oral LP
in  children is extremely rare (Fig.  33.4b). Occasionally
test is positive in the active stage. Patches of cicatricial ­ulcerations and, very rarely, white plaque forms of oral LP
alopecia may continue to progress inexorably, and can are present [53]. Patchy brown pigmentation of the oral
lead to a pseudo‐pelade of Brocq or the so‐called appear- mucosa may be the only manifestation of oral LP [54].
ance of ‘white footprints in the snow’ [48]. Etanercept and Squamous cell carcinoma is known to arise particularly
biologic anti‐tumour necrosis factor (TNF) therapy of in  erosive and atrophic variants of oral LP, hence early
Chapter 33  Lichen Planus 395

Fig. 33.4  (a) Reticulate oral lichen planus. (b)


Erosive oral lichen planus in children is extremely
rare. Here it is shown in an adult patient. (a) (b)

diagnosis is important. Periodic oral examination, good in some. Ultraviolet light‐induced alterations of self‐­
oral hygiene and avoidance of spicy foods are helpful antigens on keratinocytes may lead to recruitment of
adjuncts. Corticosteroids administered topically, intrale- cytotoxic T cells. Epidermal atrophy on histopathology is

PAPULOSQUAMOUS DISORDERS
sionally or systemically, depending on the age and condi- a distinguishing feature of ALP. The differential diagno-
tion of the patient and site and severity of the lesions, sis includes discoid lupus ­erythematosus, polymorphous
represent the mainstay of therapy [51,54]. A recent review light eruption, melasma and lichenoid drug eruption.

SECTION 6: OTHER
and meta‐analysis showed that topical tacrolimus is an Sun avoidance, topical corticosteroids or calcineurin
effective alternative to topical clobetasol and may be con- inhibitors form the cornerstone of management.
sidered as a first‐line therapy in the management of oral LP Antimalarials are typically given for multiple, recurring
[55]. There are, however, no controlled trials in children. lesions. ALP was described in six children including five
Treatment of oral LP with ciclosporin (cyclosporine), boys and one girl. The mean age at diagnosis was 11 years
­azathioprine, ­levamisole, griseofulvin, retinoids, hydroxy- of age. The disease started during the summertime in
chloroquine, dapsone and psoralen/UVA has been reported. five cases. The face and the upper limbs were the most
commonly affected sites, and the annular form was most
Ocular lichen planus common followed by the pigmented melasma‐like form.
There are reports of isolated ocular LP in children. In one Cheilitis was noted in three cases. The d
­ isease relapsed in
report, an 8‐year‐old girl with severe, filamentous dry one child after treatment interruption [58]. Ramírez et al.
eyes and persistent conjunctival hyperaemia with bilat- reported one case of ALP in a 9‐year‐old Colombian girl
eral progressive conjunctival symblepharon was who had an excellent response to hydroxychloroquine
described. Oral and topical ciclosporin combined with and photoprotection [59].
methotrexate and low‐dose oral steroids led to sustained
disease remission [56]. Hypertrophic lichen planus
The hypertrophic form of LP is relatively common in
Actinic lichen planus (ALP) children. Sharma and Maheshwari reported an inci-
Actinic LP, also known as LP subtropicus, LP tropicus dence of 26% in their series, while others reported a
and lichenoid melanodermatitis, is a photosensitive vari- lower incidence (8–10%) [21–23]. The hypertrophic vari-
ant of LP encountered commonly in young adults and ant presents as intensely pruritic, keratotic, dark brown
children from the Middle East and India. Its reported to grey‐blue, indurated plaques and nodules predomi-
incidence from India ranged from 2% to 11.5% [22,23]. nantly over the shins and the dorsal surface of the feet as
Four childhood cases have been described in Spanish is also seen with classic LP (Fig. 33.5). Lesions are often
children [57]. Lesions burn rather than itch, and are recalcitrant and tend to relapse. Moderately potent corti-
accompanied by a history of summer aggravation and costeroids under occlusion are used for treatment, but
associated photosensitivity. Three clinical variants recog- intermittent and regular patient re‐examinations are
nized are the plaque type, the annular variant and dys- needed to keep vigil for signs of epidermal atrophy.
pigmentation. The annular variant presents as brown to Intralesional steroids are an option for adolescents and
violaceous circular plaques with central hyperpigmenta- adults who may desire a faster response. Squamous cell
tion with a hypopigmented halo, typically appearing carcinoma, cutaneous horns and, recently, epidermal
over the dorsal aspects of hands, arms, neck, lips and inclusion cysts have been reported in association with
face. Melasma‐like ­pigmentation may be the only finding hypertrophic LP [60–62].
396 Section 6  Other Papulosquamous Disorders

Atrophic lichen planus Linear lichen planus


The atrophic subtype presents clinically as greyish‐white Linear LP is well recognized in children and represents
macules with superficial skin atrophy. This variant may approximately 6.9–9.2% of cases of paediatric LP [22,43].
be seen late in the course of disease when the induration Kabbash et al. described three adolescents with eruptive
subsides or secondary to the application of potent topical linear lesions of the back, abdomen and extremities dis-
corticosteroids. Morphoea closely clinically resembles tributed along the lines of Blaschko. Clinically, one of
atrophic LP. these lesions resembled an epidermal naevus in its mor-
phology and distribution [65]. Linear LP may occur as a
Acute, eruptive lichen planus Koebner phenomenon which results in linear distribution
Eruptive LP is infrequently encountered in children. A of lesions along lines of skin trauma. On the other hand,
study from India reported eruptive LP in 16.5% of patients true linear LP is often more extensive and follows lines of
[43]. A paediatric patient with eruptive LP was reported Blaschko (Fig.  33.6). The histopathology is similar to
by Pedraz et al. [63]. The onset of lesions is acute, and they ­classic LP. The lines follow a curvilinear S‐shaped pattern
may arise over a span of a few days to weeks with a wide- on the abdomen, V‐shaped lines along the posterior
spread, generalized distribution. This form of LP consists ­midline, linear patterns on the lower trunk and limbs and
of numerous, tiny, erythematous, skin‐coloured to viola- serpiginous patterns on the scalp and face. Linear LP is
ceous, pruritic papules over trunk, face, extremities, presumed to arise as a result of cutaneous mosaicism. It
palms, soles and oral mucosal surfaces. Lesions may last has been described in association with classic LP in a
for several months before healing with residual hyperpig- 4‐year‐old girl [66].
mentation. Generalized LP in a 7‐year‐old boy who had Other blaschkolinear conditions can be considered in
multiple white‐greyish papules and plaques was treated the differential diagnosis. Lichen striatus, inflammatory
with topical corticosteroid therapy resulting in healing linear verrucous epidermal naevus (ILVEN), linear psori-
within 4 weeks [64]. The differential diagnosis includes a asis and linear Darier–White disease can cause diagnostic
lichenoid drug eruption which can present very similarly. confusion due to their close morphological similarity
PAPULOSQUAMOUS DISORDERS

with linear LP.

Bullous LP and lichen planus (LP) pemphigoides


SECTION 6: OTHER

Bullous LP is a rare variant of LP and is diagnosed when


vesicular or bullous lesions develop within pre‐existing
lesions of LP. The bullae are formed secondary to
immune‐mediated damage to the basement membrane.
This leads to liquefactive degeneration of the basal layer
and results in the formation of subepidermal bullae in
longstanding LP. Both direct and indirect immunofluores-
cence are negative in bullous LP [67].
LP pemphigoides is a rare immunobullous disease. It
was described first by Kaposi in 1892 who coined the term
‘lichen ruber pemphigoides’ to describe a case of typical
LP complicated by widespread bullous eruption [68]. It is
extremely rare in children. Tense bullae develop over pre-
viously uninvolved skin, typically overlying extremities,
Fig. 33.5  Hypertrophic lichen planus. and may arise either simultaneously with or following

Fig. 33.6  Linear (zosteriform) lichen planus.


Chapter 33  Lichen Planus 397

development of classic, pruritic lesions of LP. Development folds [83]. Lesions are not itchy, but sometimes patients
of LP pemphigoides over pre‐existing scars on the limbs complain of a burning sensation. Rarely, LP pigmentosus
and buttocks was described in an 8‐year‐old Indian girl has been described in a linear or segmental distribution
[69]. Palmoplantar involvement is seen in about 50% of [84]. Dermoscopy reveals a mixed pattern: blue, grey or
cases. This form of LP may run a protracted course for 1–2 brown dots and brownish structureless areas. Treatment
years. Review of paediatric LP pemphigoides cases is not generally satisfactory and aims at identifying exter-
showed that it occurred on average around 12 years of age nal triggering agents such as dyes, chemicals, oils and
with a male‐to‐female ratio of 3 : 1. The mean time lag drugs. Tacrolimus is used to treat associated pigmenta-
between LP and LP pemphigoides was 7.9 weeks [70]. tion [85]. Oral dapsone and colchicine can be used to treat
Bullous LP will have classic LP lesions ­clinically, while his- widespread pigmentation.
topathology shows subepidermal bullae, presence of a
lichenoid infiltrate and negative immunofluorescence Inverse lichen planus
studies. In contrast, LP pemphigoides shows subepider- Inverse LP was first described as pruritic, hyperpig-
mal bullae filled with fibrin, eosinophils and neutrophils, mented, slightly scaly papules that were confined to flex-
without a lichenoid infiltrate. Direct immunofluorescence ural and intertriginous areas (axilla, popliteal fossae,
of the peribullous skin shows linear deposits of IgG and inframammary folds, inguinal folds and intergluteal
C3 along the basement membrane zone [71]. IgG autoanti- clefts). Mucosal, scalp, palmoplantar, nail or cuticular
bodies to either one or both of the 180 kDa bullous pem- abnormalities are usually absent. Given the hyperpig-
phigoid antigen (BPAg2, type XVII collagen) and the mentation seen in inverse LP, there may be overlap with
230 kDa pemphigoid antigens (BPAg1) can be demon- LP pigmentosus [86]. Erythroderma secondary to LP has
strated in these cases [72,73]. LP pemphigoides can be trig- also been reported [87].
gered by viral infections (e.g. varicella and hepatitis B
infection), ultraviolet B light exposure and henna tattoo- Lichenoid reactions
ing [74–77]. Medications such as ramipril, cinnarizine, Lichen planus–lupus erythematosus (lupus planus)

PAPULOSQUAMOUS DISORDERS
simvastatin, captopril, psoralen ultraviolet A therapy and overlap.  Some patients show hypertrophic papules and
antitubercular drugs can also cause LP pemphigoides [78]. plaques overlying acral areas which clinically resemble
The concept of ‘epitope spreading’ following an inflam- both LP and discoid lupus erythematosus (DLE).

SECTION 6: OTHER
matory process may cause tissue damage which releases However, histopathology and direct immunofluorescence
or exposes a previously ‘sequestered’ antigen, leading to a (DIF) may favour either LP or DLE. The eruption is
secondary autoimmune response against the newly chronic and can be difficult to manage [88].
released antigen [79]. Systemic agents including oral dap-
sone, oral glucocorticoids and methotrexate are used for
Lichenoid drug eruptions. A number of drugs including
the treatment of LP pemphigoides [80].
oral hypoglycaemic agents, antimalarials, methyldopa,
Bullous pemphigoid resembles LP pemphigoides;
angiotensin‐converting enzyme inhibitors, β‐blockers
however, bullous pemphigoid occurs in the elderly age
and anticonvulsants are commonly implicated (Box 33.4).
group and rarely in children, whereas patients with LP
Histopathology demonstrates parakeratosis, basal cell
pemphigoides are more typically middle‐aged and
vacuolization and eosinophils in the dermis.
­several reports have described LP pemphigoides in chil-
dren. In LP pemphigoides, the IgG and C3 are found in
the base of the bullae, as opposed to the roof in bullous Lichenoid or lichen planus‐like graft‐versus‐host
pemphigoid [79]. Reactivity to only 180 kDa antigen is ­disease. Approximately 100 days or so following haema-
seen in LP pemphigoides, whereas reactivity to both topoietic stem cell or peripheral blood stem cell trans-
180 kDa and 230 kDa antigens is observed in bullous pem- plantation, an eruption comprised of violaceous papules
phigoid. Immune electron microscopy has demonstrated and plaques  –  which clinically and histologically are
antibody deposits in the lamina lucida of lesions of indistinguishable from those of LP  –  may develop over
LP pemphigoides, as seen in bullous pemphigoid [80]. LP the dorsal surfaces of hands, feet and trunk. Areas heal
pemphigoides has been reported in association with with hyperpigmentation. Involvement of nails and
benign and malignant neoplasms such as lymphomas, mucous membrane may be observed. Bullous and follicu-
neuroblastoma, reticulosarcoma, chromophobe pituitary lar variants of graft‐versus‐host disease have also been
adenoma, craniopharyngioma and LP pemphigoides‐like described [89]. Sclerodermoid changes, when they occur,
variant of paraneoplastic pemphigus [81]. develop later.

Lichen planus pigmentosus Differential diagnosis. The various differential diagnos-


LP pigmentosus was first described by Bhutani et al. [82]. tic considerations are enumerated according to the clini-
It is encountered among various ethnic groups including cal type of LP in Box 33.5.
those of Arabic, Hispanic and Asian Indian background.
LP pigmentosus is more common in the third to fourth Diagnosis. Diagnosis of LP is based on the collective clin-
decades of life and is uncommon in children. LP pigmen- ical and ­histopathological features. Dermoscopy can aid
tosus begins as dark‐brown or slate grey macules in the in the diagnosis, especially in children and in nail and
sun‐exposed areas including the face, neck and flexural scalp LP, as it is noninvasive. Dermoscopic features of LP
398 Section 6  Other Papulosquamous Disorders

Box 33.4  Drugs implicated in lichenoid drug eruption

Anti‐inflammatory drugs Antidiabetics

Aspirin Chlorpropamide
Ibuprofen Glipizide
Indometacin Insulin
Leflunomide Tolazamide
Naproxen Tolbutamide
Sulindac
Sulfasalazine ACE inhibitors

Enalapril
Antibiotics
Captopril
Para‐aminosalicylic acid
Sulfamethoxazole Antimalarials
Tetracycline or its derivatives Chloroquine
Hydroxychloroquine
Antimycobacterial
Quinacrine
Dapsone
Ethambutol Diuretics
Isoniazid Chlorothiazide
Rifampicin Furosemide
Streptomycin Spironolactone

Antifungals
PAPULOSQUAMOUS DISORDERS

Miscellaneous
Amphotericin B
Allopurinol
Griseofulvin
Henna
SECTION 6: OTHER

Ketoconazole
Isotretinoin
Anticonvulsants Levamisole
Lithium
Carbamazepine Paraphenylene‐diamine
Oxcarbazepine Penicillamine
Phenytoin
Valproate sodium Metals

Lipid‐lowering agents Gold


Mercury
Gemfibrozil
Nickel
Pravastatin
Simvastatin

Box 33.5  Differential diagnosis of lichen planus (LP)

Classic papulosquamous LP Annular LP

Pigmented plane warts Granuloma annulare


Lichenoid graft‐versus‐host disease Pityriasis rosea
Lichen nitidus Psoriasis
Secondary syphilis Tinea corporis
Lupus erythematosus Lichen simplex chronicus
Papular acrodermatitis of childhood Discoid or nummular eczema
Pityriasis rosea
LP hypertrophicus
Eruptive LP
Prurigo nodularis
Lichen nitidus Lichenoid amyloidosis
Lichenoid drug reaction Tuberculosis verrucosa cutis
Guttate psoriasis Lichen simplex chronicus hypertrophicus
Pityriasis lichenoides chronica Lichenoid psoriasis
Nekam disease
Chapter 33  Lichen Planus 399

Box 33.5  Continued

Actinic LP Leucoplakia
Stomatitis migrans
Polymorphic light eruption
Autoimmune bullous diseases
Fixed drug eruption
Behçet disease
Discoid lupus erythematosus
Pachyonychia congenita
Dyskeratosis congenita
Linear LP
White sponge naevus
Lichen striatus Allergy to flavourings
ILVEN
Linear psoriasis Genital LP
Linear Darier disease
Lichen sclerosus et atrophicus
Vulvovaginal blistering diseases
LP pigmentosus
Vulval eczema
Macular amyloidosis Vulval intraepithelial neoplasia
Erythema dyschromicum perstans Cicatricial pemphigoid
Riehl melanosis
Addison disease Nail LP

Psoriasis
Atrophic LP
Alopecia areata
Lupus erythematosus Onychomycosis
Lichen sclerosus et atrophicus Idiopathic trachyonychia

PAPULOSQUAMOUS DISORDERS
Follicular LP Lichen planopilaris of scalp

Lichen scrofulosorum Discoid lupus erythematosus

SECTION 6: OTHER
Keratosis pilaris Pseudopalade of Brocq
Pityriasis rubra pilaris Morphoea
Lichen spinulosus Secondary syphilis

Oral LP Palmoplantar LP

Candidiasis Psoriasis
Aphthous stomatitis Eczema
Lupus erythematosus Focal palmoplantar keratoderma
Viral infections (herpes simplex, Coxsackie, HIV) Secondary syphilis

include grey‐blue dots, comedos, milium‐like cysts and dermoepidermal junction with pigmentary incontinence
vascular structures (red lines). Trichoscopy shows and abundant melanophages in the dermis. Artefactual
­perifollicular scales (black stars), diminished follicular separation of the dermis from the epidermis may create
ostia and white dots (red stars). Blue‐grey dots (yellow small clefts known as Max Joseph spaces. In bullous LP
arrows) around the follicular structures are seen as a the clefts are often large. Histology from lichen planopila-
‘­target’ pattern. Dermoscopic findings of perifollicular ris shows compact orthokeratosis, dilated follicular infun-
scale, white dots and reduced follicular ostia have been dibula filled with hyperkeratosis, slight hypergranulosis,
linked to lichen planopilaris. atrophic follicular epithelia with perifollicular fibrosis,
In atypical cases, a 4‐mm punch biopsy specimen can and new collagen formation. A dense lichenoid infiltrate
be obtained, ideally from a representative, active lesion. of lymphocytes and histiocytes at the dermoepidermal
Histopathology shows hyperkeratosis, irregular acantho- junction of the upper portion of the hair follicles, promi-
sis, wedge‐shaped hypergranulosis and sawtoothing of nent vacuolar degeneration and keratinocytic necrosis of
rete ridges (pointed rete ridges). Liquefactive degenera- the basal layer, plasma cells and eosinophils in varying
tion manifesting as vacuolization of the basal layer of the numbers in the infiltrate, and perifollicular fibrosis can be
epidermis results in loss of the usual morphology of the observed. Lichenoid drug eruption shows parakeratosis,
basal cells. This may result in loss of cohesiveness between eosinophils or neutrophils, with both a superficial and
the individual basal cells and between the epidermis and deep infiltrate. DIF studies of LP typically show fibrin at
the dermis. Civatte bodies or necrotic keratinocytes can the dermoepidermal junction and IgM (on the cytoid bod-
be seen in the lower epidermis and papillary dermis ies). Shaggy fibrin deposition at the dermoepidermal
where they appear as homogeneous, eosinophilic, PAS‐ junction is the single best indicator in the diagnosis of LP.
positive and diastase‐resistant bodies. There is presence Staining indicating IgG, IgA, C3 and fibrin can also be
of dense band‐like lymphocytic infiltrates hugging the seen. In lichen planopilaris, DIF may reveal deposition of
400 Section 6  Other Papulosquamous Disorders

Table 33.1  Treatment of lichen planus (LP)

Type of LP Topical Oral Others

Cutaneous LP Potent to mid potent steroids Systemic glucocorticoids Narrow‐band UVB


Tazarotene gel 0.05% Griseofulvin Broad‐band UVB
Hydroxychloroquine PUVA therapy
Azathioprine Intralesional triamcinolone
Mycophenolate mofetil acetonide
Acitretin
Dapsone
Thalidomide
Ciclosporin
Sulfasalazine
Levamisole
Nail LP Tazarotene gel 0.05% Oral corticosteroid mini‐pulse Intramatrix triamcinolone
Mid‐potency topical (betamethasone) acetonide
corticosteroids Acitretin
Oral LP Aloe vera gel Dapsone _
Triamcinolone acetonide in Acitretin
Orabase™
Viscous lidocaine
Diphenhydramine elixir
Ciclosporin rinse
Scalp LP Topical mid‐potency Hydroxychloroquine PUVA
corticosteroids Oral mini pulse
Methylprednisolone
PAPULOSQUAMOUS DISORDERS

Thiazolidinediones (pioglitazone)
Ciclosporin
Tetracycline or its derivatives
Acitretin
SECTION 6: OTHER

Mycophenolate mofetil

IgM and/or IgA, IgG and uncommonly C3 at the infun- modality can be considered: although short‐term use of
dibulum and isthmus level. Indirect immunofluorescence oral corticosteroids can provide more rapid initial relief,
of the lesional skin may show a specific fluorescence pat- ongoing steroid‐sparing therapy in the form of narrow‐
tern corresponding to the distribution of LP‐specific anti- band ultraviolet light phototherapy (NB‐UVB) or an oral
gen in the stratum spinosum and granulosum. It can be a systemic agent such as oral dapsone or oral acitretin or
useful adjunct for the diagnosis of LP, particularly in isotretinoin may need to be considered. Generalized LP,
atypical cases [90]. eruptive LP, LP pemphigoides, lichen planopilaris with
cicatricial alopecia and nail LP with destruction of nails
are also candidates for systemic corticosteroids.
Treatment. Identification of any possible triggering drugs or
Corticosteroids are administered in different regimens.
other exposures (such as to dyes or chemicals) is i­mportant
Some advocate a weekly pulse of betamethasone at
because their elimination can ameliorate the condition. The
0.2 mg/kg on two consecutive days of the week while
choice of treatment modality depends on the age of the
others recommend daily corticosteroid at 0.5  mg/kg
patient as well as the severity and type of LP (Table 33.1).
which is tapered over 4–6 weeks to arrest formation of
A  topical corticosteroid (preferably mild to moderately
new lesions. Sulfasalazine, ciclosporin, methotrexate and
potent), along with a second‐­generation oral antihistamine
azathioprine have also been used to control highly active
(levocetirizine/fexofenadine/­desloratadine), constitutes the
disease as steroid‐sparing agents. Metronidazole, pheny-
mainstay of therapy in children. However, topical tacroli-
toin, thalidomide and mycophenolate mofetil can be tried
mus or pimecrolimus can also be used for cutaneous and
in severe cases unresponsive to conventional agents.
oral LP. These drugs are also particularly useful for main-
tenance of remission and also for postinflammatory References
hyperpigmentation. Hypertrophic LP and nail LP can be 1 Wilson E. On leichen planus. J Cutan Med Dis Skin 1869;3:117–32.
more effectively treated with intradermal or intramatrix 2 River JK, Jackson R, Orizaga M. Who was Wickham and what are his
injections of corticosteroids for older patients who are striae? Int J Dermatol 1986;25:611–13.
3 Darier J. Precis de Dermatologie. Paris: Masson, 1909:118.
willing to tolerate injections. Oral LP may require corti- 4 Katzenelson V, Lotem M, Sandbank M. Familial lichen planus.
costeroids in adherent vehicles (such as Orabase™ or Dermatologica 1990;180:166–8.
equivalent) to provide prolonged activity overlying oral 5 Samman PD. Lichen planus: An analysis of 200 cases. Trans St Johns
Hosp Dermatol Soc 1961;46:36–8.
erosions and to help maintain oral hygiene. In cases where
6 Bermajo‐Fenoli A, Lopez‐Jornet P. Familial oral lichen planus: presen-
the percentage body area of LP involvement is >10% or tation of six families. Oral Surg Oral Med Oral Pathol Oral Radiol
where multiple site involvement is present, a systemic Endod 2006;102:e12–15.
Chapter 33  Lichen Planus 401

7 Saurat JH, Lemarchand F, Hors J et al. HLA markers and lymphocyto- 40 Kanwar AJ, De Dipankar. Lichen planus in children. Ind J Dermatol
toxins in lichen planus. Arch Dermatol 1977;113:1719–20. Venereol Leprol 2010;76:366–72.
8 Lowe NJ, Cudworth AJ, Woodrow JC. HL‐A antigens in lichen planus. 41 Nakamura R, Broce AA, Palencia DP et  al. Dermatoscopy of nail
Br J Dermatol 1976;95:169–71. lichen planus. Int J Dermatol 2013;52:684–7.
9 Powell FC, Rogers RS, Dickson ER, Moore SB. An association between 42 Tosti A, Piraccini BM, Cambiaghi S, Jorizzo M. Nail lichen planus in
HLA DR 1 and lichen planus. Br J Dermatol 1986;114:473–8. children: clinical features, response to treatment, and long‐term
10 White AG, Rostom AI. HLA antigens in Arabs with lichen planus. ­follow‐up. Arch Dermatol 2001;137:1027–32.
Clin Exp Dermatol 1994;19:236–7. 43 Pandhi D, Singal A, Bhattacharya SN. Lichen planus in childhood:
11 Luis‐ Montoya P, Yamamoto‐ Furusho JK, Vega‐Memije E et al. HLA‐ a series of patients. Pediatric Dermatol 2014;31:59–67.
DRB 1*0101 is associated with the genetic susceptibility to develop 44 Prevost NM, English JC. Palliative treatment of fingernail lichen

lichen planus in the Mexican Mestizo population. Arch Dermatol Res planus. J Drugs Dermatol 2007;6:202–4.
2007;299:405–7. 45 Pinter A, Patzold S, Kaufmann R. Lichen planus of nails – successful
12 Black MM. What is going on in lichen planus? Clin Exp Dermatol treatment with alitreinoin. J Dtsch Dermatol Ges 2011;9:1033–4.
1977;2:202–310. 46 Chieregato C, Zini A, Barba A, Magnanini M, Rosina P. Lichen plano-
13 Sugerman PB, Satterwhite K, Bigby M. Autocytotoxic T‐cell clones in pilaris: report of 30 cases and review of literature. Int J Dermatol
lichen planus. Br J Dermatol 2000;142:49–56. 2003;42:342–5.
14 Wenzel J, Scheler M, Proelss J et al. Type 1 interferon associated cyto- 47 Christensen KN, Lehman JS, Tollefson MM. Pediatric lichen plano-
toxic inflammation in lichen planus. J Cutan Pathol 2006;33:672–8. pilaris: clinico‐pathologic study of four new cases and a review of the
15 Ciaccio M, Rebora A. Lichen planus following HBV vaccination: A literature. Pediatr Dermatol 2015;32:621–7.
coincidence? Br J Dermatol 1990;122:424. 48 Nayar M, Schomberg K, Dawber RPR et  al. A clinicopathological
16 Tasanen K, Renko M, Kandelberg P et  al. Childhood lichen planus study of scarring alopecia. Br J Dermatol 1993;128:533–6.
after simultaneous measles‐mumps‐rubella and diphtheria‐tetanus‐ 49 Abbasi NR, Orlow SJ. Lichen planopilaris noted during etanercept
pertussis‐polio vaccinations. Br J Dermatol 2008;158:646–8. therapy in a child with severe psoriasis. Pediatr Dermatol 2009;26:118.
17 Zignego AL, Ferri C, Pileri SA et  al. Extrahepatic manifestations of 50 Jayasekera PS, Walsh ML, Hurrell D, Parslew RA. Case report of
hepatitis C infection: A general overview and guidelines and guide- lichen planopilaris occurring in a pediatric patient receiving a tumor
lines for a clinical approach. Dig Liver Dis 2007;39:2–17. necrosis factor α inhibitor and a review of the literature. Pediatr
18 Prabhu S, Pavithran K, Sobhanadevi G. Lichen planus and hepatitis Dermatol 2016;33:e143–6.
C virus (HCV) ‐ is there an association? A serological study of 65 cases. 51 Anuradha CH, Chandra Sekar P, Reddy GS et al. Oral mucosal lichen
Indian J Dermatol Venereol Leprol 2002;68:273–4. planus in children ‐ Report of three cases. J Orofacial Sci 2011;3:20–3.
19 Limas C, Limas CJ. Lichen planus in children: a possible complication 52 Luis‐Montoya P, Domínguez‐Soto L, Vega‐Memije E. Lichen planus in

PAPULOSQUAMOUS DISORDERS
of hepatitis B vaccines. Pediatr Dermatol 2002;19:204–9. 24 children with review of the literature. Pediatr Dermatol
20 Kanwar AJ, De D. Lichen planus in childhood: Report of 100 cases. 2005;22:295–8.
Clin Exp Dermatol 2010;35:257–62. 53 Sanjaya PR, Hallikeri K, Angadi PV. Disseminated form of childhood
21 Nanda A, Al‐Ajmi HS, Al‐Sabah H et al. Childhood lichen planus: lichen planus. Eastern J Med 2011;16:72–5.

SECTION 6: OTHER
A report of 23 cases. Pediatr Dermatol 2001;18:1–4. 54 Sugarman PB, Savage NW. Oral lichen planus: Causes, diagnosis and
22 Handa S, Sahoo B. Childhood lichen planus: A study of 87 cases. Int management. Aust Dent J 2002;47:290–7.
J Dermatol 2002;41:423–7. 55 Chamani G, Rad M, Zarei MR et al. Efficacy of tacrolimus and clobet-
23 Sharma R, Maheshwari V. Childhood lichen planus: A report of asol in the treatment of oral lichen planus: a systematic review and
50 cases. Pediatr Dermatol 1999;16:345–8. meta‐analysis. Int J Dermatol 2015;54:996–1004.
24 Carrozzo M, Pellicano R. Lichen planus and hepatitis C virus infec- 56 Igras E, Kennedy S, MacDermott EJ, Murphy CC. Isolated ocular
tion: an updated critical review. Minerva Gastroenterol Dietol lichen planus in a child. J AAPOS 2015;19:381–3.
2008;54:65–74. 57 Collgros H, Vicente A, Gonsalez MA et al. Childhood actinic lichen
25 Rebora A, Rongioletti F. Lichen planus and chronic active hepatitis. J planus: four cases report in Caucasian Spanish children and review of
Am Acad Dermatol 1984;10:840–1. the literature. J Eur Acad Dermatol Venereol 2016;30:518–22.
26 Ukonu AB, Augustine U. The prevalence of hepatitis C Virus (HCV) 58 Dammak A, Masmoudi A, Boudaya S et al. Childhood actinic lichen
among lichen planus patients and its clinical pattern at the University planus (6 cases). Arch Pediatr 2008;15:111–14.
of Abuja Teaching Hospital, Gwagwalada, Abuja, Nigeria. Glob J 59 Ramírez P, Feito M, Sendagorta E et  al. Childhood actinic lichen
Health Sci 2012;4:113–19. planus. Australas J Dermatol 2012;53:e10–3.
27 Alaizari NA, Al‐Maweri SA, Al‐Shamiri HM et al. Hepatitis C virus 60 Alvarez Alvarez C, Meijide Rico F, Rodríguez González L et  al.

infections in oral lichen planus: a systematic review and meta‐analy- Verrucous carcinoma of the penis arising from a lichen planus. A true
sis. Aust Dent J 2016;61:282–7. preneoplastic lesion? Actas Urol Esp 2006;30:90–2.
28 Laeijendecker R, Van Joost T, Tank B et  al. Oral lichen planus in 61 Sharma VK, Achar A, Ramam M, Singh MK. Multiple cutaneous
childhood. Pediatr Dermatol 2005;22:299–304. horns overlying lichen planus hypertrophicus. Br J Dermatol
29 Ozta P, Onder M, Ilter N, Ozta MO. Childhood lichen planus with nail 2001;144:424–5.
involvement: A case. Turk J Pediatr 2003;45:251–3. 62 Gayen T, Das A, Shome K, Bandyopadhyay D. Co‐localization of
30 Kanwar AJ, Belhaj MS. Lichen planus among Arabs: A study from lichen planus hypertrophicus and epidermal inclusion cyst: an inci-
Libiya. J Dermatol 1984;11:93–6. dent unreported hitherto. Indian J Dermatol 2015;60:503–5.
31 Balaubramaniam P, Ogboli M, Moss C. Lichen planus in children: 63 Pedraz J, Campos‐Muñoz L, Conde‐Taboada A et al. Familial erup-
Review of 26 cases. Clin Exp Dermatol 2008;33:457–9. tive generalized lichen planus in a pediatric patient. J Dermatol
32 Pusey WA. Lichen planus in an infant less than 6 months old. Arch 2010;37:910–12.
Dermatol Syph 1929;19:671–2. 64 Ott H, Frank J, Poblete‐Gutiérrez P. Eruptive lichen planus in a child.
33 Samman PD. A note on the natural history of lichen planus. Br
Pediatr Dermatol 2007;24:637–9.
J Dermatol 1956;68:175–81. 65 Kabbash C, Laude TA, Weinberg JM, Silverberg NB. Lichen planus in
34 Altman J, Perry HO. The variations and course of lichen planus. Arch the lines of Blaschko. Pediatr Dermatol 2002;19:541–5.
Dermatol 1961;84:179–91. 66 Adışen E, Onder M, Erdem O, Happle R. Superimposed linear lichen
35 Kumar V, Garg BR, Beruah MC, Vasireddi SS. Childhood lichen
planus in a 4‐year‐old girl. Int J Dermatol 2014;53:e541–2.
planus (LP). J D Dermatol 1993;20:175–7. 67 Joshi RK, Antukorala, Abanmi A, Awadi TA. Lichen planus pemphig-
36 Goettmann S, Zaraa I, Moulonguet I. Nail lichen planus: epidemio- oides. Is it a separate entity? Br J Dermatol 1994;130:537–8.
logical, clinical, pathological, therapeutic and prognosis study of 68 Kaposi M. Lichen ruber pemphigoides. Arch Derm Syphilol

67 cases. J Eur Acad Dermatol Venereol 2012;26:1304–9. 1892;24:343–6.
37 Peluso AM, Tosti A, Piraccini BM, Cameli N. Lichen planus limited to 69 Kumar P, Savant SS, Das A et al. lichen planus pemphigoides present-
the nails in childhood: case report and literature review. Pediatr ing preferentially over preexisting scars: a rare instance of isotopic
Dermatol 1993;10:36–9. phenomenon. Indian J Dermatol 2015;60:596–9.
38 Saman PD. The nails in lichen planus. Br J Dermatol 1961;73:288–92. 70 Cohen DM, Ben‐Amitai D, Feinmesser M, Zvulunov A. Childhood
39 Grover C, Vohra S. Onychomadesis with lichen planus: an under‐ lichen planus pemphigoides: A case report and review of the litera-
recognized manifestation. Indian J Dermatol 2015;60:420. ture. Pediatr Dermatol 2009;26:569–74.
402 Section 6  Other Papulosquamous Disorders

71 Davis AL, Bhogal BS, Whitehead P et al. Lichen planus pemphig- 80 Prost C, Tesserand F, Laroche L et al. Lichen planus pemphigoides: an
oides: Its relationship to bullous pemphigoid. Br J Dermatol immuno‐electron microscopy study. Br J Dermatol 1985;113:31–6.
1991;125:263–71. 81 Helm TN, Camisa C, Liu AY et al. Lichen planus associated with neo-
72 Sakuma‐Oyama Y, Powell AM, Albert S et al. Lichen planus pemphi- plasia: a cell‐mediated immune response to tumor antigens? J Am
goides evolving into pemphigoid nodularis. Clin Exp Dermatol Acad Dermatol 1994;30:219–24.
2003;28:613–16. 82 Bhutani LK, Bedi TR, Pandhi RK, Nayak NC. Lichen planus pigmen-
73 Hsu S, Ghohestani RF, Uitto J. Lichen planus pemphigoides with IgG tosus. Dermatologica 1974;149:43–50.
autoantibodies to the 180 kd bullous pemphigoid antigen (type XVII 83 Kanwar AJ, Dogra S, Handa S et al. A study of 124 Indian patients
collagen). J Am Acad Dermatol 2000;42:136–41. with lichen planus pigmentosus. Clin Exp Dermatol 2003;28:481–5.
74 İlknur T, Akarsu S, Uzun S et al. Heterogeneous disease: a child case 84 Vineet R, Sumit S, K GV, Nita K. Lichen planus pigmentosus in linear
of lichen planus pemphigoides triggered by varicella. J Dermatol and zosteriform pattern along the lines of Blaschko. Dermatol Online
2011;38:707–10. J 2015;21.
75 Flageul B, Hassan F, Pinquier L et al. [Lichen pemphigoid associated 85 Verma P, Pandhi D. Topical tacrolimus and oral dapsone combination
with developing hepatitis B in a child]. Ann Dermatol Venereol regimen in lichen planus pigmentosus. Skinmed 2015;13:351–4.
1999;126:604–7. 86 Lee MD, Schwartz LR. Inverse lichen planus. Skinmed 2013;11:355–6.
76 Goldscheider I, Herzinger T, Varga R et al. Childhood lichen planus 87 Gupta C, Sinha S, Sahoo B, Garg VK. Erythroderma secondary to
pemphigoides: report of two cases treated successfully with sys- lichen planus in a child. Indian J Dermatol Venereol Leprol
temic glucocorticoids and dapsone. Pediatr Dermatol 2014;31:751–3. 2016;82:89–91.
77 Rosmaninho A, Sanches M, Oliveira A et al. Lichen planus pemphig- 88 Van der Horst JC, Cirkel PKS, Nieboer C. Mixed lichen planus‐lupus
oides induced by a weight reduction drug. Cutan Ocul Toxicol erythematosus disease: a distinct entity? Clinical, histopathological
2011;30:306–8. and immunopathological studies in six patients. Clin Exp Dermatol
78 Chan LS, Vanderlugt CJ, Hashimoto T et  al. Epitope spreading:
1983;8:631–40.
Lessons from autoimmune skin diseases. J Invest Dermatol 89 Johnson ML, Farmer ER. Graft‐versus‐host reactions in dermatology.
1998;110:103–9. J Am Acad Dermatol 1998:;38:369–92.
79 Duong B, Marks S, Sami N, Theos A. Lichen planus pemphigoides in 90 Rao R, Shenoi SD. Indirect immunofluorescence to demonstrate

a 2‐year‐old girl: response to treatment with methotrexate. Am Acad lichen planus specific antigen (LPSA) in lichen planus. Indian
Dermatol 2012;67:e154–6. J Dermatol Venereol Leprol 2006;72:350–2.
PAPULOSQUAMOUS DISORDERS
SECTION 6: OTHER
 403

C HA PTER   34

Lichen Nitidus
Jasem M. Alshaiji
Dermatology Department and Pediatric Dermatology Unit, Amiri Hospital, Kuwait

Abstract been rarely reported. Most cases are isolated, but various diseases,
either inherited or acquired, have been reported in association
with LN. The pathogenesis of LN remains unclear. No laboratory
Lichen nitidus (LN) is an acquired inflammatory cutaneous disor-
abnormalities are found. The diagnosis is based largely on clinical
der affecting mostly school‐aged children and young adults. There
features, but the characteristic histological findings are very helpful
is no apparent gender or race predilection. Typically, it manifests
to confirm the diagnosis when needed. LN is usually a self‐limited
with multiple, grouped, glistening, skin‐coloured, dome‐shaped
disease, resolving within a few months to 1 year, but in some cases
or flat‐topped, pinhead‐sized papules. They are mostly seen on
the condition may be persistent, generalized or disfiguring, in which
the extremities, trunk and genitalia, but lesions can arise on any
case therapeutic modalities can be used including topical or systemic
­cutaneous surface. Usually asymptomatic, LN can occasionally be
agents and, occasionally, phototherapy with ultraviolet A or B.
mildly pruritic. It is most often localized, but generalized cases have

Key points • LN is most often localized, but can be generalized.


• Other variants include confluent, vesicular, haemorrhagic,
perforating, spinous follicular, linear and actinic.

PAPULOSQUAMOUS DISORDERS
• Lichen nitidus (LN) is a chronic benign inflammatory skin disorder.
• Mucous membranes, palms and soles and nail involvement are
• LN affects mainly school‐aged children and young adults.
rarely reported.
• The pathogenesis of LN is still unclear.
• No laboratory abnormalities have been consistently reported.
• There is no sex or race predilection.

SECTION 6: OTHER
• LN is diagnosed clinically, but can be confirmed by histology.
• LN manifests as multiple, skin‐coloured, dome‐shaped or
• It is a self‐limited disease, usually resolving within a few months
flat‐topped papules, seen predominantly on the trunk, upper
up to 1 year, although some persistent cases have been reported.
extremities and genitalia.
• LN is usually asymptomatic, but it can be pruritic.

The occurrence of LN during infancy is extremely rare but


Introduction. Lichen nitidus (LN) is a benign chronic
has been reported [3,4].
inflammatory skin disease affecting mainly school‐aged
children and young adults. There is no sex or race predi-
lection. Familial cases have been rarely reported. Its Pathogenesis. The pathogenesis of LN is still unclear.
pathogenesis is still unclear although an immunological There are reports of LN and lichen planus (LP) co‐
basis is suspected. It manifests with multiple, 1–2 mm, occurring in the same patient, which suggests the
glistening skin‐coloured or slightly pinkish or hypopig- controversial possibility that LN might represent a
­
mented dome‐shaped or flat‐topped papules limited to micropapular variant of LP with similar aetiologies [4].
the extremities, trunk and genitalia. No laboratory abnor- One study observed LN‐like lesions in nearly a third of
malities have been reported. The diagnosis is based patients with LP; however, histopathological and immu-
largely on clinical features, but can be confirmed by the nohistochemical patterns of LN are diagnostic and dis-
characteristic histological findings. LN is usually a self‐ tinctly different from those of LP. Immunohistochemical
limited disease resolving within a few months up to 1 staining of LN reveals an infiltrate of CD4+ and CD8+
year, although some cases can persist for longer. lymphocytes as well as macrophages and Langerhans
cells. Similar studies of LP more typically demonstrate a
History. Felix Pinkus first described lichen nitidus in predominance of CD4+ cells that display HECA‐452 (a
1907 [1]. skin homing receptor), supporting the notion that LN
and LP are distinct entities [4].
Epidemiology. LN affects mostly school‐aged children One aetiological theory proposes that LN is an
and young adults with a median age of 7 years in boys immunological response to an infectious agent or an
and 13 years in girls [2]. There is no sex or race predilection. idiopathic response to nonviable microbial antigens

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
404 Section 6  Other Papulosquamous Disorders

(viruses/bacteria) that may activate a cell‐mediated


response, initiate lymphocyte accumulation and form
inflammatory papules [5].

Clinical features. LN manifests clinically as multiple,


1–2 mm, discrete, glistening, skin‐coloured or slightly
pinkish or hypopigmented, dome‐shaped or flat‐topped
pinhead‐sized papules (Figs 34.1–34.3). They are usually
arranged in groups and are mainly seen on the upper
extremities, chest, abdomen and genitalia (especially the
glans and shaft of the penis, and prepuce), but lesions can
arise anywhere on the cutaneous surface. Usually asymp-
tomatic, LN can be mildly pruritic and disfiguring. It is
most often localized, but a generalized variant is rarely
seen [3,6–9]. Many associated diseases with generalized
LN have been reported (Box 34.1).
Other clinical variants of LN include confluent, vesicu- Fig. 34.3  Lichen nitidus on the extensor aspect of the lower limb with
lar, haemorrhagic [18], perforating [19,20], spinous follic- Koebner phenomenon. Source: Courtesy of Shehab Al‐Dhafiri, MD.
ular [7,21], linear/blaschkolinear [22–24] and actinic [4,5].
Actinic LN (ALN) is a unique, photodistributed sea-
sonal variant of LN seen mainly in children and young Box 34.1  Diseases associated with lichen nitidus [4,10–17]
adults with Fitzpatrick skin types IV–VI [4,25–27]. A case Crohn disease
Down syndrome
Human immunodeficiency virus infection
PAPULOSQUAMOUS DISORDERS

Juvenile chronic arthritis


Juvenile myelomonocytic leukaemia
Multiple endocrine neoplasia
SECTION 6: OTHER

Neurofibromatosis, type 1
Niemann–Pick disease
Postpartum thyroiditis
Russell–Silver syndrome

of ALN developing in a young Caucasian male with


Fitzpatrick skin type III has been described more recently
[28]. Bedi first reported this entity in 1978 as summertime
actinic lichenoid eruption (SALE) [29]. Isaacson et al. con-
sidered the lesions as part of the spectrum of actinic LP
(ALP) [30]. Kanwar and Kaur, and Hussain, proposed the
Fig. 34.1  Lichen nitidus on the forehead – actinic variant. Source: term actinic lichen nitidus [31–33].
Courtesy of Shehab Al‐Dhafiri, MD. ALN is clinically and histopathologically different from
actinic LP. There are similarities to a pinpoint papular
variant of polymorphous light eruption (PMLE), so both
PMLE and ALN can be regarded as variants of the same
entity in this author’s opinion [4].
As with LP, the Koebner phenomenon can be observed
and is a hallmark of LN [5,34].
Involvement of the mucous membranes, palms and
soles and nails is rarely reported [5,18,35–40].
When LN is present at this site, the oral cavity shows
minute grey‐white flat papules on the buccal mucosa or
tongue [40–42]. LN of the palms may differ from classic
LN in its clinical appearance: hyperkeratotic fissured and
pitted papules/plaques with central plugs or more rarely
purpuric lesions and lesions resembling pompholyx may
be observed. It tends to occur at an older age with mean
age at onset being 45 years (age range: 37–52), and to be
accompanied by plantar lesions [18,35–38]. Classic LN
Fig. 34.2  Lichen nitidus on the extensor aspect of the upper limb. Source: lesions at other sites usually accompany the palmoplantar
Courtesy of Shehab Al‐Dhafiri, MD. lesions, and nail dystrophy associated with the palmar
Chapter 34  Lichen Nitidus 405

Box 34.2  Cutaneous diseases reported in association Box 34.3  Differential diagnosis of lichen nitidus
with lichen nitidus [5,21,43,44]
Classic form [35,45–49]
Lichen planus
Erythema nodosum Keratosis pilaris
Segmental vitiligo Lichen planus
Lichen spinulosus Follicular eczema
Lichen striatus Contact dermatitis to parthenium or nickel
Psoriasis vulgaris Molluscum contagiosum
Atopic dermatitis Verruca plana
Tattooing Pityriasis rubra pilaris
Phrynoderma
Lichen simplex chronicus
Keratosis follicularis (Darier disease)
lesions has been reported [36]. Nail involvement is rarely Secondary syphilis
reported and can precede the onset of skin lesions. Unlike Id reaction
nail LP, nail LN is more common in children, and is less Lichen scrofulosorum
severe with no pterygium formation or anonychia [39]. Prurigo nodularis
The nail changes when present include thickening, lon- Psoriasis
gitudinal ridging, pitting, distal detachment, rippling, Granuloma annulare (GA)
trachyonychia and median canaliform dystrophy in the Langerhans cell histiocytosis (LCH)
absence of nail bed tumours or a history of trauma [39,40]. Papular mucinosis
Clues to the diagnosis include the presence of subtle
papules on the affected digit and swelling with Perforating lichen nitidus [19]
hyperpigmentation of the nail fold due to nail matrix Primary perforating disorders (such as Kyrle disease, reactive

PAPULOSQUAMOUS DISORDERS
involvement [39]. perforating collagenoses, elastosis perforans serpiginosa and
Rarely, LN can be associated with various other cutane- perforating folliculitis)
ous diseases as noted in Box 34.2. Secondary perforating dermatoses (such as GA and calcinosis cutis)

SECTION 6: OTHER
The simultaneous occurrence of molluscum contagio-
sum (MC) or condyloma acuminatum and LN has been Actinic lichen nitidus [26,27]
rarely reported and may be coincidental [45]. Actinic lichen planus
Actinic folliculitis
Differential diagnosis. There are many cutaneous disor- Follicular eczema
ders that can clinically mimic the classic and variant Sarcoidosis
forms of LN (Box 34.3). Mucinosis (lichen myxoedematosus)

Laboratory/pathology findings. No laboratory abnor- Palmoplantar lichen nitidus [35,36,38]


malities have been reported.
Arsenical keratosis
The diagnosis is largely based on clinical features, but Darier disease
can be confirmed by histopathological features, which are Naevoid basal cell carcinoma
quite characteristic (Figs 34.4 and 34.5). Porokeratotic eccrine ostial and dermal duct naevus
The histology demonstrates a sharply circumscribed Chronic eczema and pompholyx
inflammatory infiltrate within the dermis that generally Keratodermas
spans approximately four to five dermal papillae. The bor- Palmar lichen planus
ders are marked by extensions of epidermal rete ridges, Pitted keratolysis
giving a so‐called ‘ball‐in‐claw’ appearance. The infiltrate
is composed of lymphocytes and epithelioid histiocytes.
The overlying epidermis is thin and parakeratotic with
basal cell hydropic degeneration and cytoid bodies [50]. cells. The basal cell layer overlying the top of the dilated
The accumulation of S‐100+ CD1a+ Langerhans cells and dermal papilla is poorly refractile, possibly because of the
CD68+ macrophages/histiocytes in the papillary dermis is depletion of melanocytic cells [52]. In cases of palmoplan-
a typical feature of LN [51]. In addition to these immuno- tar LN, if typical LN lesions at other regions are not pre-
histochemical findings, factor XIIIa+ and fascin+ cells in sent, dermoscopy can be a valuable tool for confirming
the dermis have been found in cases of generalized spinous the diagnosis [53]. In plantar LN, linear scales in parallel
follicular LN with perifollicular granuloma [7]. are discontinued by oval, well‐defined depressions sur-
In vivo imaging with confocal laser scanning micros- rounded by ring‐shaped, silvery‐white scales. The long
copy (CLSM) is a useful diagnostic tool that can be uti- axes of the depressed patterns are parallel to the lines of
lized before performing skin biopsy. CLSM reveals round, scales. Fewer and finer scales on the surface of depressed
enlarged, well‐circumscribed dermal papillae that are areas are found. In palmar LN, a similar structure of
heavily laden with individual highly refractive cells, depressions and arrangement of scales is found, but the
­consistent with melanophages and possibly mononuclear depressions are flatter and have longer patterns [53].
406 Section 6  Other Papulosquamous Disorders

Treatment. Treatment of LN is often unnecessary given


its asymptomatic nature and tendency for spontaneous
improvement. However, treatment may be required for
persistent and generalized cases and for individual
patient cosmetic concerns.
For symptomatic localized cases, mid‐ to high‐
potency topical corticosteroids or topical calcineurin
inhibitors and antihistamines may be effective.
Successful treatment of penile LN in a 20‐year‐old man
has been reported with topical pimecrolimus 1% cream
applied twice daily for 2 months [54]. Ablative laser
therapy for penile LN should be avoided due to the
potential for scarring [55].
For actinic LN, additional measures including sun-
screen use and sun avoidance may help prevent
development of new lesions. Facial ALN in a 23‐year‐
old female was successfully treated with hydroxy-
chloroquine 200 mg twice daily for 4 weeks and then
a maintenance dose of 200 mg per day for a total of 6
months [27].
For generalized LN, topical corticosteroids and
­calcineurin inhibitors may be helpful [47,56]. Successful
treatment of generalized LN in an 8‐year‐old child with
pimecrolimus 1% cream twice daily for 8 weeks has been
PAPULOSQUAMOUS DISORDERS

reported [47]. Systemic agents that have been reported as


having some success include systemic retinoids (acitretin,
isotretinoin), astemizole, dinitrochlorobenzene/diphe-
SECTION 6: OTHER

Fig. 34.4  Histology of lichen nitidus (×4). Source: Courtesy of Mohammad


Al‐Enezi, MD. nylcyclopropenone immunotherapy, systemic steroids,
ciclosporin (cyclosporine), itraconazole, isoniazid and
enoxaparin [4,15,47,57]. Ultraviolet light phototherapy in
the form of narrow‐band UVB and PUVA has been
reported to have some therapeutic benefit [4,15,47,58].
While these may be reasonable considerations in some
adult patients, many of these modalities may not be
entirely suitable for use in children.

Prognosis. LN is a chronic but self‐limited disease and the


lesions usually resolve spontaneously within several
months to 1 year.

References
1 Pinkus F. Uber eine neue knochtenformige Hauteruption: Lichen
nitidus. Arch Dermatol Syph (Berlin) 1907;85:11–36.
2 Chiu HY, Chiu HC. A florid, tiny, discrete eruption on the penis.
JAMA, 2012;308:1264–5.
3 Alexander KCL, Jeffrey NG. Generalized lichen nitidus in identical
twins. Case Rep Dermatol Med 2012;2012:1–3.
4 Payette MJ, Weston G, Humphrey S et al. Lichen planus and other
lichenoid dermatoses: Kids are not just little people. Clin Dermatol
2015;33:631–43.
5 Cho EB, Kim HY, Park EJ et al. Three cases of lichen nitidus associated
with various cutaneous diseases. Ann Dermatol 2014; 26:505–9.
6 Pinto‐Almeida T, Fernandes IC, Machado S et al. Diffuse small glistening
papules in a child. J Paediatr Child Health 2015;51:461–3.
7 Summe HS, Greenlaw SM, Deng A et al. Generalized spinous follicular
lichen nitidus with perifollicular granuloma. Pediatr Dermatol 2013;
30:e20–e21.
8 Gao F, Miyamoto M, Ogata H et  al. Generalized lichen nitidus in a
Japanese girl. J Dermatol 2012:39:185–7.
9 Hernandez‐Martin A, Prada ID, Colmenero I, Torrelo A. Generalized
flesh‐colored papules in a 5‐year‐old boy. Arch Dermatol 2010; 146:1419–24.
10 Teixeira VB, Coutinho I, Cardoso JC, Tellhechea O. Generalized lichen
Fig. 34.5  Histology of lichen nitidus (×10). Source: Courtesy of nitidus in a boy with Niemann‐Pick disease type B. An Bras Dermatol
Mohammad Al‐Enezi, MD. 2013;88:977–8.
Chapter 34  Lichen Nitidus 407

11 Kanai C, Terao M, Tanemura A et  al. Generalized lichen nitidus in 35 Park SH, Kim SW, Noh TW et  al. A case of palmar lichen nitidus
Russell‐Silver syndrome. Pediatr Dermatol 2013;30:150–1. presenting as a clinical feature of pompholyx. Ann Dermatol
12 Dogan S, Memis P, Ersoy‐Evans S et  al. Generalized lichen nitidus 2010;22:235–7.
associated with neurofibromatosis type 1 and juvenile myelomono- 36 Cakmak SK, Unal E, Gonul M et al. Lichen nitidus with involvement
cytic leukemia. Int J Dermatol 2016;55:e592–e594. of the palms. Pediatr Dermatol 2013;30:e100–e101.
13 Botelho LF, Magalhaes JP, Ogawa MM et al. Generalized lichen niti- 37 Khandpur S, Kathuria SD, Gupta R et  al. Hyperkeratotic pitted

dus associated with Down’s syndrome‐case report. An Bras Dermatol plaques on the palms and soles. Indian J Dermatol Venereol Leprol
2012;87:466–8. 2010;76:52–5.
14 Wanat KA, Elenitsas R, Chachkin S et al. Extensive lichen nitidus as a 38 Podder I, Mohanty S, Chandra S, Gharami RC. Isolated palmar lichen
clue to underlying Crohn’s disease. J Am Acad Dermatol 2012; nitidus‐a diagnostic challenge: first case from Eastern India. Indian J
67:e218–e220. Dermatol 2015;60:308–9.
15 Bilgili SG, Karadag AS, Calka O et  al. A case of generalized 39 Tay EY, Ho MS, Chandran NS et al. Lichen nitidus presenting with
lichen nitidus successfully treated with narrow‐band ultraviolet nail changes‐case report and review of the literature. Pediatr
B treatment. Photodermatol Photoimmunol Photomed 2013;29: Dermatol 2015;32:386–8.
215–17. 40 Chu J, Lam J. Lichen nitidus. CMAJ 2014;186:e688.
16 Bercedo A, Cabero MJ, Garcia‐Consuegra J et al. Generalized lichen 41 Russell B. Lichen nitidus with involvement of tongue. Br J Dermatol
nitidus and juvenile chronic arthritis: an undescribed association. 1965;77:280–1.
Pediatr Dermatol 1999;16:406–7. 42 Costa de Araujo P, Lechien JR. Lichen nitidus on the dorsal tongue.
17 Shan SJ, Chen J, Zhang L et al. Generalized lichen nitidus associated Ear Nose Throat J 2016;95:143.
with postpartum thyroiditis. Eur J Dermatol 2011;21:614–16. 43 Lestringant GG, Piletta P, Feldmann R et al. Coexistence of atopic der-
18 Ikenberg K, Pflugfelder A, Metzler G, Biedermann T. Thirty‐year matitis and lichen nitidus in three patients. J Dermatol 1996;192:
history of palmar eruptions: a quiz. Palmar purpuric lichen nitidus. 171–3.
Acta Derm Venereol 2011;91:108–11. 44 Shan SJ, Xia Z, Chen J et al. Widespread lichen nitidus associated with
19 Vijaya B, Sunila, Manjunath GV. Perforating lichen nitidus. Indian J tattoo. Eur J Dermatol 2013;23:123–4.
Pathol Microbiol 2010;53:162–3. 45 Lockerman S, Braun‐Falco M. Lichen nitidus and molluscum conta-
20 Zussman J, Smart CN. Perforating lichen nitidus. Am J Dermatopathol giosum: A pure coincidence? J Dermatol 2011;38:412–13.
2015;37:406–8. 46 De D, Jindal R, Kanwar A. Contact dermatitis to parthenium simulat-
21 Park JS. Lichen nitidus and lichen spinulosus or spinous follicular ing lichen nitidus. Indian J Dermatol Venereol Leprol 2010;76:286–7.
lichen nitidus? A second case. Clin Exp Dermatol 2011;36:553–61. 47 Farshi S, Mansouri P. Letter: Generalized lichen nitidus success-
2 2 Grillo E, Perez B, Vano‐Galvan S et al. Unknown: Congenital lin- fully treated with pimecrolimus 1 percent cream. Dermatol Online

PAPULOSQUAMOUS DISORDERS
ear whitish papules in a 3‐year‐old boy. Dermatol Online J J 2011;17:11.
2013;19:9. 48 Tanyildizi T, Akarsu S, Ilknur T et al. Disseminated eruptive intersti-
23 Oiso N, Kawada A. Blaschkolinear lichen nitidus. Eur J Dermatol tial granuloma annulare mimicking lichen nitidus. Eur J Dermatol
2016;26:100–1. 2011;21:644–64–5.

SECTION 6: OTHER
24 Aravind M, Do TT, Cha HC et al. Blaschkolinear acquired inflamma- 49 Lozano Masdemont B, Gomez‐Recuero Munoz L, Villanueva Álvarez‐
tory skin eruption, or blaschkitis, with features of lichen nitidus. Santullano A et al. Langerhans cell histiocytosis mimicking lichen niti-
JAAD Case Rep 2016;2:102–4. dus with bone involvement. Australas J Dermatol 2017;58:231–3.
25 Blalock TW, Kannan S, Davis LS. Actinic lichen nitidus. Dermatol 50 Shockman S, Lountzis N. Dermatopathology Diagnosis: Lichen niti-
Reports 2010;2:e10 dus. Cutis 2013;92:288, 297–8.
26 Dar NR, Rao SU. Facial lichen nitidus actinicus. Australas J Dermatol 51 Nakamizo S, Egawa G, Miyachi Y, Kabashima K. Accumulation of
2012;53:e16–e17. S‐100+ CD1a+ Langerhans cells as a characteristic of lichen nitidus.
27 Bouras M, Benchikhi H, Ouakkadi A, Zamiati S. Facial actinic
Clin Exp Dermatol 2011;36:800–17.
lichen nitidus successfully treated with hydroxychloroquin: a case 52 Liu H, Chen S, Shi Z, Zhang F. In vivo imaging of lichen nitidus with
report. Dermatol Online J 2013;19:7. confocal laser scanning microscopy. Arch Dermatol 2011;147:142.
28 Solano‐Lopez G, Argila DD, Fraga J, Dauden E. Actinic lichen nitidus 53 Qian G, Wang H, Wu J et  al. Different dermoscopic patterns of
in a Caucasian European patient. J Eur Acad Dermatol Venereol 2014; palmoplantar and nonpalmoplantar lichen nitidus. J Am Acad
­
28:1401–6. Dermatol 2015;73:e101–3.
29 Bedi TR. Summertime actinic lichenoid eruption. Dermatologica 54 Lee WJ, Park OJ, Won CH et al. Penile lichen nitidus successfully treated
1978;157:115–25. with topical pimecrolimus 1% cream. J Dermatol 2013;40:499–500.
30 Isaacson D, Turner ML, Elgart ML. Summertime actinic lichenoid 55 Teichman JM, Sea J, Thompson IM, Elston DM. Noninfectious penile
eruption (lichen planus actinicus). J Am Acad Dermatol 1981;4:404–11. lesions. Am Fam Physician 2010;81:167–74.
31 Kanwar AJ, Kaur S. Lichen nitidus actinicus. Pediatr Dermatol
56 Park J, Kim JI, Kim DW et al. Persistent generalized lichen nitidus
1991;8:94–5. successfully treated with 0.03% tacrolimus ointment. Eur J Dermatol
32 Hussain K. Summer time actinic lichenoid eruption, a distinct
2013;23:918–19.
entity, should be termed actinic lichen nitidus. Arch Dermatol 57 Topal IO, Gokdemir G, Sahin IM. Generalized lichen nitidus:

1998;134:1302–3. Successful treatment with systemic isotretinoin. Indian J Dermatol
33 Kanwar AJ, Kaur S. Lichen nitidus actinicus. Arch Dermatol
Venereol Leprol 2013;79:554.
1999;135:714. 58 Nakamizo S, Kabashima K, Matsuyoshi N et al. Generalized lichen
34 Ghoshal L, Barua JK. White pinhead‐like eruptions on the back of a nitidus successfully treated with narrowband UVB phototherapy.
10‐year‐old. J Paediatr Child Health 2013;49:501. Eur J Dermatol 2010;20:816–17.
408 

CHA PTER  3 5

Lichen Striatus
Franck Boralevi1 & Alain Taïeb2
1
 Pediatric Dermatology Unit, Hôpital Pellegrin‐Enfants, Bordeaux, France
2
 Department of Dermatology and Paediatric Dermatology, Reference Center for Rare Skin Diseases, Hôpital Saint André, Bordeaux, France

Abstract coloured flat‐topped papules, typically with a lichenoid appearance,


arising along a Blaschko’s line. Multiple lesions may occur simultane-
Lichen striatus is a transient linear eruption that arises along ously. Hypochromic, or more rarely hyperchromic sequelae may occur.
Blaschko’s lines and lasts several months to a few years, eventu- Treatment is usually not needed. Pathological examination typically
ally healing spontaneously. Children are much more often affected shows a mixed spongiotic and lichenoid pattern, with a perivascular
than adults, and females are significantly more frequently affected and periadnexal lymphocytic infiltrate. Lichen striatus and blaschkitis
than males. The lesions consist of erythematous or sometimes skin‐­ belong to the same clinical and pathological spectrum.

Key points • Light microscopy shows a mixed spongiotic–lichenoid histological


pattern.
• Lichen striatus is a benign and self‐limited disease. Usually no
• Lichen striatus (LS) is a transient linear eruption that arises along
treatment is required.
Blaschko’s lines.
• Similarly to other linear conditions, LS may be considered a
• LS most frequently occurs in children, with a mean age of 3.5.
superimposed segmental manifestation of polygenic skin disorders.
PAPULOSQUAMOUS DISORDERS

• Usually, a single linear lesion occurs and exhibits a self‐limited


• To avoid confusion with other lichenoid conditions, the name
course of several months’ duration.
‘blaschkitis’ may also be used.
SECTION 6: OTHER

lichenoides [10], lichen striatus [11,12], lineare neurodermatitis


Introduction. Lichen striatus is a transient but prolonged
[13], ­dermatosis linearis pruriens [14] and lineären Ekzem [15].
linear ­eruption more frequently observed in children
The authors’ own review of these original sources suggests
than  in adults [1]. Usually a single linear lesion occurs
that some of the patients described may have actually had
along the developmental lines of Blaschko (Fig. 35.1) and
inflammatory linear verrucous epidermal naevus (ILVEN),
exhibits a self‐­limited course and a duration varying from
linear lichenification or linear porokeratosis [12,14].
several weeks to 2–3 years. Postinflammatory hypopig-
Later, in the 1960s, Gianotti and Frugis, from a study of
mentation or hyperpigmentation may persist even longer.
nine cases, suggested that viral diseases may trigger
Relapses rarely occur. Several conditions such as atopic
lichen striatus [16]. Jackson, in his review, emphasized the
dermatitis or vitiligo may be associated. Familial cases
distribution along the lines of Blaschko [17,18], and this
have been reported. Characteristically, light microscopy
was revisited by Grosshans [19,20], who proposed the
shows a mixed spongiotic–lichenoid histological pattern,
name blaschkitis or Blaschko dermatitis (blaschkite in
frequently with a lymphocytic infiltrate along eccrine
French) as a more precise name. More recently, in order to
ducts or glands [2]. The pathogenesis of this Blaschko’s
underline the distribution of this disorder and its relation
line ­disorder remains unclear, with suggestions that a
to a latent developmental abnormality, Taïeb et  al. sug-
postzygotic genomic event or that epigenetic mosaicism
gested the name blaschkolinear acquired inflammatory
could be involved, associated with a triggering event,
skin eruption (BLAISE) [1]. The acronym BLAISE has not
possibly a viral infection.
been widely used since the 1990s but its use could be
encouraged as it emphasizes the concept of a spectrum
History. Senear and Caro are credited with the description including LS and blaschkitis [21].
and naming of lichen striatus [3]. In their paper, published
in the middle of the twentieth century, they reviewed the Epidemiology. LS most frequently occurs in children 1–10
literature and compiled the numerous terms that have years of age, with a mean age of 3.5 in the largest series
been used to describe this disorder: éruption lichénoïde [4], (Table 35.1) [1,22–25]. Cases have been reported in infants
naevus linearis [5], trophonévrose lichénoïde en bande linéaire from the first months of life [25,26], but LS may occur at
[6], ­neurodermite zoniforme [7], linear cutaneous eruption [8], any age from infancy to adulthood in all ethnic groups
­systematisierter Lichenifikation [9], dermatosis linearis [20,26]. It is more common in children than in adults and

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 35  Lichen Striatus 409

to a common trigger [39]. Pathological findings suggest


that cytotoxic events eventually eliminate mutant cells
through a cell‐mediated immunological process involv-
ing CD8+ cytotoxic T cells. The late occurrence in many
cases suggests a breakdown of immune tolerance towards
a cell clone that was hitherto quiescent. The time course of
the disease may vary according to the duration of the
scavenging process. The abnormal immune status usually
associated with atopy might contribute to the develop-
ment of LS.
Similarly, other immune or autoimmune disorders
have already been shown to follow the lines of Blaschko
[40,41]. These are considered as superimposed segmental
manifestations of polygenic skin disorders and include
linear psoriasis, fixed drug eruption, atopic dermatitis,
lupus erythematosus, vitiligo, pemphigus vulgaris,
chronic graft‐versus‐host disease, granuloma annulare,
lichen planus, lichen nitidus and lichen planopilaris [39].
The peripheral blood hypereosinophilia described in one
case before the onset of LS might be considered an early
part of the immune response [1].
What might stimulate the sudden loss of tolerance to an
abnormal cutaneous clone in patients with this acquired
disorder? Perhaps an acute event causes the aberrant

PAPULOSQUAMOUS DISORDERS
clone to express a novel membrane antigen, as has been
hypothesized in autoimmune disease. Several precipitat-
ing factors have been identified in isolated cases, with LS

SECTION 6: OTHER
arising after skin trauma or sunburn [42,43], topical wart
therapy [25], contact dermatitis or an infectious disease
Fig. 35.1  Typical presentation of lichen striatus arising here on the upper
(flu‐like fever, tonsillitis, varicella [44]), or bacillus
limb of a young child, with an erythematous, linear and slightly papular
lesion along a line of Blaschko.
Calmette–Guérin (BCG) or yellow fever vaccination
[2,45–47]. One case arising after a bumblebee sting has
been recently reported [48].
most common in younger children (2–3 years) than in
The simultaneous occurrence of LS in pairs of (even
older ones, as reporting bias may have led to the prepon-
nonrelated) siblings and the seasonal influence men-
derance of older children in the past literature [3]. LS is
tioned in several papers provide some circumstantial evi-
significantly more common in females, with a sex ratio of
dence of an infective trigger associated with intrinsic
1 : 2.1 [3,25,27,28]. There are a few reports of familial cases,
susceptibility [29–31]. Increased quantities of IL‐1β in the
mainly arising in siblings, and more rarely concomitant
lesions [32] could support an in situ inflammasome‐
cases in a child and one of the parents [30–34]. The asso-
driven process triggered by infectious pathogens. Based
ciation with atopic dermatitis has been widely noted in
on histological findings, the primary target of the inflam-
several papers, especially among Italian or Spanish series
matory reaction in LS is the keratinocyte in the basal and
that showed signs of atopy in 48–61% of cases [24,25]. In
suprabasal layers. The occurrence along the lines of
one series, 84% of patients with LS had a positive history
Blaschko also favours the keratinocyte as the target cell
of atopic disorders [27]. A few cases of LS with psoriasis
[1]. Familial occurrence is unlikely via the somatic muta-
have been published [35], and more recently the associa-
tion mechanism because these mutations take place by
tion with vitiligo has been reported, with onset of vitiligo
chance. Epigenetic mosaicism has been suggested to con-
and LS at around the same time in a young girl, suggest-
tribute to skin diseases following the lines of Blaschko
ing a common trigger or a common background [36].
[38]. These skin diseases might be caused by visualization
of the actions of transposable elements that are partly
Pathogenesis. Lichen striatus is characterized by a mixed expressed and partly silenced at an early developmental
lichenoid and spongiotic histological pattern with a dis- stage. Epigenetic rather than genomic mechanisms fit bet-
tribution along the lines of Blaschko. Current embryologi- ter the familial observations of lichen striatus.
cal theory suggests that Blaschko’s lines correspond to the
direction of growth of clones of epidermal and dermal
cells derived from precursor cells [37,38]. The segmental Clinical features. Lichen striatus is not rare, but it may
involvement may be explained by an early postzygotic frequently go undiagnosed [1,25,27]. LS is an acquired
event in the form of loss of heterozygosity involving and not a congenital condition, even though it may appear
genes that may lead to an inappropriate immune reaction early in infancy. The lesions are distributed on the limbs
410 Section 6  Other Papulosquamous Disorders

Table 35.1  Lichen striatus clinical characteristics, including age, sex ratio, localization of the lesions, mean duration from the review of the largest LS series
PAPULOSQUAMOUS DISORDERS

n Male : female Mean age Seasonality Localization Atopy Mean Relapses


ratio (range) duration

Sittart et al., 1989 [22] 53 1 : 2.3 2.5 yr Spring and summer Limbs 92% 20% — —
SECTION 6: OTHER

(1–40 mo)
Taïeb et al., 1991 [1] 18 1 : 1 3 yr — Limbs 66% 33% 9.5 mo 5%
(0.5–14 yr) Trunk 33% (6/18) (4 wk–4 yr) (1/18)
Kennedy and Rogers, 1996 [23] 61 1 : 2 2.9 yr Spring and summer Limbs 77% — 12 mo —
(0.8–9 yr) Trunk 11% (4 mo–4 yr)
Zhang and McNutt, 2001 [28] 37 1 : 1.6 17.5 yr — Limbs 76% — — —
(1.3–49 yr)
Hofer, 2003 [29] 18 1 : 2 44 yr — Trunk 77% — 8.7 mo 28%
(adults only) Limbs 55%
Taniguchi et al., 2004 [27] 89 1 : 3 — Spring Limbs 85% 20% — —
(1 mo–14 yr) Trunk 10%
Patrizi et al., 2004 [25] 115 1 : 2.1 4.4 yr Cold seasons Limbs 62% 61% 6 mo 4.3%
(1 mo–13 yr) Trunk 25% (70/115) (5/115)
Head 15%
Paramiquel et al., 2006 [24] 24 1 : 1 3.4 yr Spring and summer Limbs 73% 48% 8.5 mo —
(1 mo–2 yr)

c35.indd 410 9/7/2019 2:34:49 AM


Chapter 35  Lichen Striatus 411

in two thirds of the cases, with a slight preponderance of


lesions on the inferior limbs (Fig.  35.2) compared to the
upper limbs (Fig. 35.1), on the trunk (Fig. 35.3) in around
20% of cases, and on the face (Fig. 35.4) in the remaining
5–10% of cases (Table 35.1) [49]. Pruritus can be moderate
but is uncommon, reported by about 10% of the patients.
Clinical ­features of atopy and minor signs of atopic der-
matitis may be associated; more rarely psoriasis or viti-
ligo is a feature (Fig.  35.5). The mean duration is 6–12
months (range 4 weeks to 10 years; median around 8
months). Relapses may occur within the first 4 years in
about 5% of cases in children and more frequently in
adults [1,25].
The most characteristic feature is the linear arrange-
ment of inflammatory lesions, ranging in extent from a Fig. 35.3  Lichen striatus on the trunk.
few centimetres to an entire limb or half of the trunk. The
lesions consist of pink, erythematous or skin‐coloured,
flat‐topped papules, with a frequent lichenoid appear-
ance (Fig. 35.6) and sometimes vesicular with eczematous
features [40]. Wickham’s striae are absent. A perforating
variant has been described [50].
The linear eruption appears in 2–3 weeks and usually
disappears in several months, leaving transient or pro-
longed depigmentation in about 50% of cases (Fig. 35.7) [1].

PAPULOSQUAMOUS DISORDERS
Hyperpigmentation has also been noted [51]. The lesions
may form in a single line or occur in grouped divided lines.
Streaks may be discontinuous. Hemicorporeal forms with

SECTION 6: OTHER
multiple involved lines occur rarely, and bilateral forms Fig. 35.4  Facial lichen striatus.
(Fig. 35.8) appear exceptionally [52]. Facial involvement is
more common than previously thought [25] but can be dif-
ficult to diagnose if a limited portion of a Blaschko’s line is
involved because of less frequent access to biopsy. In a

Fig. 35.2  Lichen striatus on the posterior aspect of the inferior limb, here
with a double erythematous line. Fig. 35.5  Association of lichen striatus and vitiligo in a young girl.
412 Section 6  Other Papulosquamous Disorders
PAPULOSQUAMOUS DISORDERS

Fig. 35.6  Lichen striatus – skin‐coloured flat‐topped papules with rarely


encountered linear vesicular lesions.
SECTION 6: OTHER

Fig. 35.8  Lichen striatus with multiple involved lines of Blaschko on the
trunk of an infant. This clinical form of LS (or blaschkitis) is characterized
by a shorter course and complete healing.

dystrophy that may be associated with longitudinal lines


or nail groove, onychorrhexis, koilonychias and distal
onycholysis. Linear papules may coexist near the nail fold.
Nail involvement may appear before typical cutaneous
lesions have been observed, or can be isolated, and can
­persist after healing of the skin lesion [53–56]. Follicular
involvement with transient focal hair loss may also exist.

Prognosis. Lichen striatus is a benign disease. Resolution


begins in a few weeks to a few months. Residual hypopig-
mentation  may constitute a cosmetic problem in darkly
pigmented skin.

Differential diagnosis. Lichen striatus, with its character-


istic linearity and history of acquired onset, is a distinc-
tive cutaneous disorder. Box 35.1 lists the major differential
diagnoses. Biopsy may be helpful to differentiate LS from
the other disorders.
The lesions of LS differ from the pruritic and usually
monomorphic keratotic papules of lichen planus, which
Fig. 35.7  Hypopigmented skin lesion as a sequela of lichen striatus. can also occur in a linear distribution [57,58]. In contrast,
pruritus is rare in LS. Lichen planus usually eventuates in
series of facial LS, it was noted that the average age and sex hyperpigmentation, and LS more frequently in hypopig-
ratio of facial LS was similar to that of LS of the limbs, with mentation [59]. Clinical variants of linear lichen planus,
a similar natural history [49]. Nail involvement (Fig. 35.9) including vesicular lichen planus and linear lichen plano-
is not uncommon and consists of linear, segmental or total pilaris, have been described [60,61].
Chapter 35  Lichen Striatus 413

Fig. 35.10  Typical linear psoriasis on the trunk and right upper limb in a
young girl, with hyperkeratotic papules surrounded by a hypochromic halo.

Linear porokeratosis is a chronic disease, and the


c­haracteristic rim of dyskeratosis around the primary
­circular or oval lesions is easily detected, especially using
dermoscopy. Linear Darier disease also has a chronic
course and typical histopathological features. Linear
lupus ­erythematosus is extremely rare and has not been
described in children.

PAPULOSQUAMOUS DISORDERS
In examining patients with the hypopigmented stage of
Fig. 35.9  Skin‐coloured linear papules on the dorsal part of the hand with LS, hypomelanosis of Ito and linear vitiligo must be
­considered [61]. However, a history of a previous inflam-

SECTION 6: OTHER
partial involvement of the right thumbnail.
matory stage will suggest the correct diagnosis. At the
onset of the eruption, cutaneous larva migrans could be
Box 35.1  Lichen striatus: differential diagnosis confused with LS.
The linear eruptions that have been described as
• Linear lichen planus
blaschkitis or Blaschko dermatitis in adults are character-
• Linear lichen nitidus
ized both clinically and histologically by eczematous
• Linear psoriasis
changes. Adult blaschkitis shows a tendency to relapse. LS
• Inflammatory linear verrucous epidermal naevus (ILVEN)
probably belongs within the spectrum of blaschkitis [62].
• Linear atopic dermatitis
• Linear porokeratosis
• Linear Darier disease Histology findings. Lichen striatus lacks specific patho-
• Linear lupus erythematosus
logical findings, but pathological examination typically
• Hypomelanosis of Ito* (hypopigmented stage)
shows a combination of spongiotic epidermal signs and
• Linear vitiligo* (hypopigmented stage)
lichenoid interface dermatitis with a perivascular and
• Cutaneous larva migrans
periadnexal lymphocytic infiltrate [66]. The largest study
* Blaschkitis is not considered as a distinct entity but as part of the LS of the pathology of LS included 41 childhood cases [67].
spectrum. The authors concluded that histological examination
could lead to a diagnosis of LS in about 50% of cases. In an
older study of 23 cases, Reed et al. considered that histo-
Linear lichen nitidus is characterized by minute pap- pathological examination was not sufficient to differenti-
ules [62]. However, a histological overlap between lichen ate LS from lichen nitidus and lichen planus [68]. More
striatus and lichen nitidus has been noted [41]. Linear precisely, the diagnosis should be based on the following
psoriasis [63] and ILVEN are characterized by larger, lin- features: (i) a focal band‐like infiltrate with (ii) variable
ear papulosquamous streaks and a psoriasiform histolog- exocytosis and (iii) a collection of histiocytes in the d
­ ermal
ical pattern not seen in lichen striatus (Fig. 35.10). papillae. Slight acanthosis and focal parakeratosis,
Linear atopic dermatitis [64] has been described in a together with satellite cell necrosis, have been described.
young girl. The patient presented with a linear arrange- Additional features of interest include perivascular lym-
ment of nummular lesions that recurred during phoid aggregates and the alignment of the infiltrate along
generalized flares of the disease. Muñoz Garza et  al.
­ hair follicles and eccrine ducts that are particularly
reported two cases of linear morphoea of the forehead ­distinctive of LS [1,2,28,66]. Occasionally, a dense deep
and nose with lesions that were initially diagnosed dermal lymphocytic infiltrate [28,69] and intraepidermal
­clinically and histologically as LS, and hypothesized that vesicles filled with Langerhans cells and CD8+ T lympho-
LS may precede or simulate linear morphoea [65]. cytes may be present. CD8+ intraepithelial lymphocytes
414 Section 6  Other Papulosquamous Disorders

are sometimes scattered but are most often located in 16 Gianotti F, Frugis F. Il lichen striatus. Aspetti clinici ed istologici di
nove casi. Giorn Ital Derm 1963;48:307–22.
clusters around necrotic keratinocytes, thus supporting
17 Jackson R. The lines of Blaschko: a review and reconsideration.
the hypothesis of a cytotoxic process directed towards Observations of the cause of certain unusual linear conditions of the
keratinocytes [1]. Most of the small lymphocytes in the skin. Br J Dermatol 1976;95:349–60.
upper dermis and epidermis are positive for CD7 [2]. 18 Blaschko A. Die Nervenverteilung in der Haut in ihrer Beziehung zu
den Erkrankungen der Haut. Beilage zu den Verhandlungen der
According to Gianotti et al. [2], the age of the lesion does Deutschen Dermatologischen Gesellschaft VII. Congress, Breslau,
not influence the ratio of nonspecific to diagnostic histo- Braumüller, Wien und Leipzig, 1901.
logical changes. In 50% of cases, nonspecific findings 19 Grosshans E. Lichen et éruptions lichénoides. Ann Dermatol
were noted. Vénéréol 1989;116:925–9.
20 Grosshans E, Marot L. Blaschkite de l’adulte. Ann Dermatol
Vénéréol 1990;117:9–15.
Treatment. Usually no treatment is required. 21 Johnson M, Walker D, Galloway W et  al. Interface dermatitis along
Careful explanation of the benign and self‐limited nature Blaschko’s lines. J Cutan Pathol 2014;41:950–4.
22 Sittart JA, Pegas JR, Sant’Ana LA, Pires MC. Lichen striatus.
of this disorder is extremely important, with mention of Epidemiologic study. Med Cutan Ibero Lat Am. 1989;17:19–21.
complete healing without skin sequelae in most cases. 23 Kennedy D, Rogers M. Lichen striatus. Pediatr Dermatol
Tacrolimus ointment has been shown to hasten the 1996;13:95–9.
­resolution of the disease and can be proposed on facial 24 Peramiquel L, Baselga E, Dalmau J et al. Lichen striatus: clinical and
epidemiological review of 23 cases. Eur J Pediatr 2006;165:267–9.
skin and on coloured skin in order to reduce the risks of 25 Patrizi A, Neri I, Fiorentini C et al. Lichen striatus: clinical and labo-
residual hypopigmentation. Topical tacrolimus has also ratory features of 115 children. Pediatr Dermatol 2004;21:197–204.
been considered for early treatment of multiple and 26 Markouch I, Clérici T, Saiag P, Mahé E. Lichen striatus avec dystrophie
unguéale chez un nourrisson. Ann Dermatol Venereol 2009;136:883–6.
spreading LS [70], but its usefulness in those with multiple 27 Taniguchi Abagge K, Parolin Marinoni L, Giraldi S et al. Lichen striatus:
areas of blaschkitis is difficult to prove because multiple description of 89 cases in children. Pediatr Dermatol 2004;21:440–3.
blaschkitis is thought to heal rapidly. Pimecrolimus has 28 Zhang Y, McNutt NS. Lichen striatus. Histological, immunohisto‐
also been proposed in cases of relapsing and itchy LS in chemical, and ultrastructural study of 37 cases. J Cutan Pathol
2001;28:65–71.
adults [71]. Because of interface changes mimicking lupus 29 Hofer T. Lichen striatus in adults or ’adult blaschkitis’?. There is no
PAPULOSQUAMOUS DISORDERS

erythematosus, Lee et  al. proposed hydroxychloroquine need for a new naming. Dermatology 2003;207(1):89–92.
in cases of facial LS, which demonstrated reasonably 30 Patrizi A, Neri I, Fiorentini C et al. Simultaneous occurrence of lichen
striatus in siblings. Pediatr Dermatol 1997;14:293–5.
good results [72]. Finally, in a recently published series
SECTION 6: OTHER

31 Smith SB III, Smith JB, Ellis LE et  al. Lichen striatus: simultaneous
of  12 patients, 308‐nm excimer laser treatment was occurrence in two nonrelated siblings. Pediatr Dermatol 1997;14:43–5.
considered effective for treating hypopigmentation after 32 Racette AJ, Adams AD, Kessler SE. Simultaneous lichen striatus in
LS, with good safety profiles [73]. siblings along the same Blaschko line. Pediatr Dermatol 2009;26:50–4.
33 Karempelis PS, Cely SJ, Davis LS. Lichen striatus in a mother and son.
Int J Dermatol 2014;53:e366.
References 34 Yaosaka M, Sawamura D, Iitoyo M et al. Lichen striatus affecting a
1 Taïeb A, El Youbi A, Grosshans E et al. Lichen striatus: a Blaschko mother and her son. J Am Acad Dermatol 2005;53:352–3.
linear acquired inflammatory skin eruption (BLAISE). J Am Acad 35 Errichetti E, Stinco G, Trevisan G, Patrone P. Lichen striatus associated
Dermatol 1991;25:637–42. with psoriasis vulgaris treated with oral acitretin. Indian Dermatol
2 Gianotti R, Restano L, Grimalt R et al. Lichen striatus – a chame- Online J 2014;5:501–3.
leon. A histopathological and immunohistological study of 41 36 Chong JH, Mermin D, Goussot JF, Taïeb A. Vitiligo co‐existing with
cases. J Cutan Pathol 1995;22:18–22. lichen striatus: more than a coincidence? J Eur Acad Dermatol
3 Senear FE, Caro MR. Lichen striatus. Arch Dermatol Syph Venereol 2017;31:e200–e202.
1941;43:116–33. 37 Happle R. Genetische Interpretation streifenförmiger Hautanoma‐
4 Du Castel. Eruption lichénoïde, suite de traumatisme sur le trajet du lien. Hautarzt 1978;29:357–63.
nerf cubital. Ann Dermatol Syph 1893;4:1188–93. 38 Happle R. Lyonisation and the lines of Blaschko. Hum Genet
5 Unna PG. Naevi lineares. In: The Histopathology of the Diseases of 1985;70:200–6.
the Skin (trans. N. Walker). Edinburgh: W.F. Clay, 1896:1146. 39 Happle R. Superimposed segmental manifestation of polygenic
6 Balzer F, Mercier R. Trophonévrose lichénoïde en bande linéaire sur le skin disorders. J Am Acad Dermatol 2007;57:690–9.
trajet du nerf petit sciatique. Ann Dermatol Syph (Paris) 1898;9:1898. 40 Bolognia JL, Orlow SJ, Glick SA. Lines of Blaschko. J Am Acad
7 Brocq L. Neurodermite zoniforme. In: La Pratique Dermatologique. Dermatol 1994;31:157–90.
Paris: Masson, 1902:154. 41 Grosshans EM. Acquired blaschkolinear dermatoses. Am J Med
8 Pinkus F. Über eine besondere Form strichförmiger Hautausschläge. Genet 1999;85:334–7.
Dermatol Ztschr 1904;11:19–27. 42 Ciconte A, Bekhor P. Lichen striatus following solarium exposure.
9 Söllner. Ein Fall von systematisierter Lichenifikation, als beitrag zur Australas J Dermatol 2007;48:99–101.
Kenntnis metamerischer Hautaffektionen. Arch Dermatol Syph 43 Gougerot E, Lortat‐Jacob E. Eczéma linéaire déclenché par une
(Berlin) 1905;73:147–62. teinture à base de paraphénylène diamine. Hypothèse de zones
10 Kirsch O. Zur Kenntnis einer Spezies strichförmiger Hauterkrankung fragiles naeviques linéaires invisibles. Bull Soc Fr Dermatol Syph
(dermatosis linearis lichenoides). Arch Dermatol Syph (Berlin) 1934;41:405–6.
1910;101:343–54. 44 Hafner C, Landthaler M, Vogt T. Lichen striatus (blaschkitis) follow-
11 Fantl G. Lichen striatus. Beitrag zur Kenntnis der strichförmigen ing varicella infection. J Eur Acad Dermatol Venereol 2006;20:1345–7.
Hauterkrankungen. Dermatol Wochnschr 1914;58:593–9. 45 Karouni M, Kurban M, Abbas O. Lichen striatus following yellow
12 Kreibich C. Lichen striatus der Bauchhaut. Arch Dermatol Syph fever vaccination in an adult woman. Clin Exp Dermatol 2017;42:823–4.
(Berlin) 1927;154:84. 46 Patri P. Lichen striatus bilaterale. Giorn Ital Derm Vener
13 Alexander A. Die Lineare Neurodermitis (Lichenoides Ekzem in Einer 1983;118:101–3.
Voigtschen Grenzlinie). In: Jadassohn J (ed.) Handbuch der Haut‐ und 47 Hwang SM, Ahn SK, Lee SH, Choi EH. Lichen striatus following
Geschechtskrankheiten, vol. 6/1. Berlin: Springer, 1927:402–3. BCG vaccination. Clin Exp Dermatol 1996;21:393–4.
14 Zoon JJ. Zwei Fälle von Dermatosis linearis pruriens. ‘Lichénification 48 Unal E, Balta I, Bozkurt O. Lichen striatus: after a bite by bumblebee.
géante’ und Naevus linearis. Acta Derm Venereol 1928–29;9:490–501. Cutan Ocul Toxicol 2015;34:171–2.
15 Leipner S. Ein Betrag zum lineären Ekzem. Dermatol Wochnschr 49 Mu EW, Abuav R, Cohen BA. Facial lichen striatus in children:
1936;102:729–31. retracing the lines of Blaschko. Pediatr Dermatol 2013;30:364–6.
Chapter 35  Lichen Striatus 415

50 Pujol RM, Tuneu A, Moreno A et al. Perforating lichen striatus. Acta 62 Prigent F, Cavelier‐Balloy B, Lemarchand‐Venencie F et  al. Lichen
Derm Venereol (Stockh) 1988;68:171–3. nitidus linéaire. Ann Dermatol Vénéréol 1989;116:814–15.
51 Hauber K, Rose C, Brocker EB et al. Lichen striatus: clinical features 63 Atherton DJ, Kahana M, Russel‐Jones R. Naevoid psoriasis. Br J
and follow‐up in 12 patients. Eur J Dermatol 2000;10:536–9. Dermatol 1989;120:837–41.
52 Gelmetti C, Cerri D, Gianotti R et al. Bilateral multiple lichen striatus. 64 Taïeb A. Linear atopic dermatitis (naevus atopicus): a pathogenetic
Pediatr Dermatol News (Bari) 1988;7:97. clue? Br J Dermatol 1994;131:134–5.
53 Baran R, Dupré A, Lauret P et  al. Le lichen striatus onychodys- 65 Muñoz Garza FZ, Manubens Mercadè E, Roè Crespo E et al. Linear
trophique. A propos des 4 cas avec revue de la littérature. Ann morphea mimicking lichen striatus in its early presentation. Pediatr
Dermatol Vénéréol 1979;106:885–90. Dermatol 2016;33:e23–6.
54 Goskowick MO, Eichenfield LF. Onychodystrophy with lichen 66 Graham JN, Hossler EW. Lichen striatus. Cutis 2016;97:86, 120–2.
­striatus. Pediatr Dermatol 1994;11:282–3. 67 Staricco RG. Lichen striatus. Arch Dermatol 1959;79:311–24.
55 Tosti A, Peluso AM, Misciali C et  al. Nail lichen striatus: clinical 68 Reed RJ, Meek T, Ichinose H. Lichen striatus: a model for
features and long‐term follow‐up of five patients. J Am Acad the  ­histologic  spectrum of lichenoid reactions. J Cutan Pathol
Dermatol 1997;36(6 Part 1):908–13. 1975;2:1–18.
56 Brys AK, Bellet JS. Persistent lesion of the thumbnail of a 3‐year‐old 69 Miquel C, Brousse N, de Prost Y et  al. Lichen striatus: évaluation
boy. Pediatr Dermatol 2016;33:95–6. des critères histologiques dans cas. Ann Pathol 2000;20:308–12.
57 Pautrier LM. Le lichen plan. In: Darier J et al. (eds) Nouvelle Pratique 70 Jo JH, Jang HS, Park HJ et  al. Early treatment of multiple and
Dermatologique, vol. 7. Paris: Masson, 1936:454–5. spreading lichen striatus with topical tacrolimus. J Am Acad
58 Mérigou D, Léaute‐Labrèze C, Louvet S et al. Lichen planus in chil- Dermatol 2007;57:904–5.
dren. Role of the campaign for hepatitis B vaccination. Ann Dermatol 71 Campanati A, Brandozzi G, Giangiacomi M et al. Lichen striatus in
Vénéréol 1998;125:399–403. adults and pimecrolimus: open, off‐label clinical study. Int J Dermatol
59 Herd RM, McLaren KM, Alridge RD. Linear lichen planus and 2008;47:732–6.
lichen striatus  –  opposite ends of a spectrum. Clin Exp Dermatol 72 Lee MW, Choi JH, Sung KJ et  al. Linear eruptions of the nose in
1993;18:335–7. ­childhood: a form of lichen striatus? Br J Dermatol 2000;142:1208–12.
60 Katz M, Weinrauch L. Differentiating vesicular linear lichen planus 73 Bae JM, Choo JY, Chang HS et  al. Effectiveness of the 308‐nm
and lichen striatus. Cutis 1987;40:151–3. excimer laser on hypopigmentation after lichen striatus:
­
61 Küster W, Kind P, Hölzle E et al. Linear lichen planopilaris of the face. A  ­retrospective study of 12 patients. J Am Acad Dermatol
J Am Acad Dermatol 1989;21:131–2. 2016;75:637–9.

PAPULOSQUAMOUS DISORDERS
SECTION 6: OTHER
416

CHA PTER 3 6

Pityriasis Rosea
Antonio A.T. Chuh1,2 & Vijay Zawar3,4
1
 Department of Family Medicine and Primary Care, The University of Hong Kong and Queen Mary Hospital, Pokfulam, Hong Kong
2
 JC School of Public Health and Primary Care, The Chinese University of Hong Kong and the Prince of Wales Hospital, Shatin, Hong Kong

3
 Skin Diseases Centre, Nashik, India

4
 Department of Dermatology, MVP’s Dr Vasantrao Pawar Medical College and Research Centre, Nashik, Maharashtra, India

Abstract Many children exhibit atypical variants of this exanthem. Vali-


dated diagnostic criteria are available and are particularly appli-
cable for children with marginal symptoms. Classification systems
Pityriasis rosea is a self‐limited exanthem likely to be related to
are available for delineating subclasses, and allow for choice of
viral infections, particularly endogenous reactivation or primary
treatment modalities as well as subgroup analyses in laboratory‐
infection of human herpesviruses 7 and 6.
based and clinical investigations.
A prodrome of coryzal symptoms is common. In around 20–
Pityriasis rosea usually does not have significant impacts on
30% of childhood cases, a herald patch is seen. The secondary
children and adolescents. Symptomatic treatments usually suffice.
generalized eruption then occurs 2–3 weeks later, with usually
The benefits of active interventions with antiviral agents are being
smaller macules with peripheral collarette scaling on the trunk
­actively investigated.
and proximal aspects of the extremities. The orientation of most
lesions follows lines of skin creases. Spontaneous remission then
occurs 2–12 weeks after the eruption of the herald patch or the
generalized eruption.
PAPULOSQUAMOUS DISORDERS

Key points • Diagnostic criteria and two versions of classifications are


SECTION 6: OTHER

available. These tools are useful clinically and in research


settings.
• Pityriasis rosea is highly likely to be caused by viral infections.
• Active interventions are unnecessary for most children with this
• Endogenous reactivations or primary infections of human
exanthem. Many clinical trials are being conducted.
herpesviruses 7 and 6 are highly to be the cause of many, but
not all, cases of this exanthem in children and adolescents.

intermediate variety between pityriasis simplex and pity-


Definition and  history. Pityriasis rosea is a subacute
riasis rubra [3]. However, he only described the macular
self‐limited eruption mainly affecting children and
variety of pityriasis rosea, not the usual annular variety,
young adults. It is characterized by a distinctive skin
which was first described by the French dermatologist
eruption with a herald patch at the onset followed by
Pierre‐Antoine‐Ernest Bazin (1807–1878) in 1862 [3].
secondary eruptions oriented along the lines of skin
Jean Baptiste Emile Vidal (1825–1893), another French
cleavage presenting as smaller, discrete, round to oval
dermatologist, described a similar condition termed
skin lesions with collarette scaling. The lesions mainly
pityriasis circiné et marginé in 1882 [3]. Some dermatolo-
affect the trunk and proximal aspects of extremities.
gists consider this clinical entity to be a special form of
Spontaneous remission usually occurs within 6–12
pityriasis rosea, with fewer and larger lesions often local-
weeks of onset of the rash. Pityriasis rosea is suspected
ized at the axillae or groins [4], while others maintained
to be related to viral infections, especially human her-
that pityriasis circinata and pityriasis circinata et margin-
pesvirus (HHV)‐7.
ata should be synonyms of pityriasis rosea [2]. Louis‐
Anne‐Jean Brocq drew attention to the ‘primitive patch’
History. The French dermatologist Camille‐Melchior in 1887 [5]. Alfred Blaschko described peripheral collar-
Gibert (1797–1866) is generally credited with the first ette scaling in 1899 [5]. Colcott Fox reported five children
accurate description of pityriasis rosea in 1860 [1], with pityriasis rosea in 1884 [2].
although Robert Willan (1757–1812) and Erasmus Wilson
(1809–1884), both British dermatologists, described sim-
ilar rashes termed erythema annulatum and lichen Epidemiology. Pityriasis rosea is very common. The inci-
annulatus serpiginosus respectively [2]. Gibert described dence ranges from 0.39 [6] to 4.8 [7] per 100 dermatology
five varieties of pityriasis: simple, rosea, rubra, versi- patients. The overall incidence upon pooling data from 18
color and nigra, and considered pityriasis rosea to be an studies [6–23] is 0.64 per 100 dermatology patients.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 36  Pityriasis Rosea 417

The occurrence of pityriasis rosea in children is similar Most of these studies were performed in adult patients.
to that in young adults [24,25]. Pityriasis rosea can occur in In a previous study on three children with pityriasis
infants [26]. The youngest reported patient was 3 months rosea, no virological evidence of HHV‐7 and ‐6 primary
old [27]. Most patients are between the age of 10 and 35 infection or endogenous reactivation was found [65],
[28]. The incidence for paediatric patients has been while a recent study found that most children with pity-
reported as 1.02 per 100 paediatric dermatology patients riasis rosea have serological findings of previous HHV‐7
(below 14 years) [11]. For adolescents and young adults (61%) and ‐6 (84%) infections. For those children who
(10–29 years), the prevalence was reported to be 0.6% [15]. have IgM against HHV‐7 (9%) and ‐6 (16%) detected,
most have viraemia of these viruses [66]. Moreover, for
Aetiology. The aetiology of pityriasis rosea is unknown. children who have viraemia [66], the virus loads are
An infectious agent is suspected owing to the apparently higher than those of adults with pityriasis rosea [48].
programmed clinical course of the disease: prodromal Autoimmunity has been suspected to play a role in the
symptoms, primary herald patch, secondary eruptions pathogenesis [67]. In adults, T‐lymphocytotoxic antibodies
followed by complete spontaneous remission. [68], anti‐nuclear antibodies [69], rheumatoid factor [23],
Particularly strong clinical evidence for a viral aetiol- anti‐Ro autoantibodies (SS‐A) [69], thyroglobulin autoanti-
ogy is that most sufferers do not have a second eruption bodies [69], thyroid peroxidase antibodies [69] and riboso-
in their lifetime, and, for the small proportion of patients mal P protein [69] have been reported to be more prevalent
who do have a relapse, virtually all do not exhibit the her- in patients. Autoimmune diseases associated with pityriasis
ald patch [29]. This is highly suggestive of immunological rosea include undifferentiated connective tissue disease,
memory after the first episode of this eruption. autoimmune thyroiditis and chronic autoimmune urticaria
Particularly strong epidemiological evidence for an [69]. The role of autoimmunity in the immunopathogenesis
infectious aetiology is the detection of significant tempo- of pityriasis rosea in children is as yet unknown.
ral [30,31] and spatial–temporal [16] clustering of patients Studies on the association of atopic dermatitis and
with pityriasis rosea. asthma with pityriasis rosea revealed controversial find-

PAPULOSQUAMOUS DISORDERS
Various viruses (cytomegalovirus [32,33], Epstein– ings [17,24,70]. The pertinence of atopy and genetic pre-
Barr virus [32,33], influenza and parainfluenza viruses disposition to atopy in this eruption is unknown.
[33,34], adenovirus [33], respiratory syncytial virus

SECTION 6: OTHER
[33], parvovirus B19 [35], picornavirus [36–39]) and Pathology. Lesional histopathological changes are nonspe-
bacteria (Legionella longbeachae, L. micdadei, L. pneu- cific for pityriasis rosea. They include focal caps of spongi-
mophila [40,41], Mycoplasma pneumoniae [33,41–43], osis, perivascular lymphocytic infiltrates, vascular dilation
Chlamydia pneumoniae [41]) have been suspected to be and patchy parakeratosis [28] (Fig. 36.1). These changes are
associated with pityriasis rosea. However, evidence for similar for the herald patch and the generalized eruption.
these microbes having a causative association with pit- Intraepidermal dyskeratotic cells with aggregations of
yriasis rosea is not definitive. tonofilaments, numerous vacuoles and intracytoplasmic
Endogenous reactivation, and, to a lesser extent, pri- desmosomes were reported to be rather specific features
mary infection with HHV‐7 and ‐6 are particularly signifi- [71]. These cytological changes are akin to those seen in
cant as the likely aetiology of pityriasis rosea. HHV‐7 viral infections such as herpes simplex and varicella zoster,
DNA was detectable by nested polymerase chain reaction and are therefore believed to substantiate the suggestion of
in the skin, plasma and peripheral blood mononuclear viruses being the culprit [72]. Lesional histopathological
cells of all patients with pityriasis rosea in two reports changes in children are similar to those in adults.
[44–46]. HHV‐like particles have been detected by elec-
tron microscopy in lesional biopsies of patients with the
eruption [47]. HHV‐7 pp85 antigen expression has been
detected in lesional biopsies of patients [48]. A significant
inverse correlation was also reported between HHV‐7
virus neutralizing antibody titres and high HHV‐7 virus
load in the plasma of patients [48]. In the absence of virus
seroconversion, which is accepted to be the gold standard
for primary herpesvirus infections, these serological find-
ings indicate endogenous reactivation of HHV‐7.
More evidence for the relevance of these viral infections
in adults with pityriasis rosea comes from the detection of
high virus loads of HHV‐7 and ‐6 in patients with persis-
tent pityriasis rosea [49], and the association of endoge-
nous reactivation of HHV‐7 and ‐6 in miscarrying women
with pityriasis rosea [50].
Other studies [51–64] have reported conflicting results on
the detection of herpesvirus‐7 and ‐6 DNA in lesional biopsies, Fig. 36.1  Lesional biopsy of a secondary lesion in pityriasis rosea, revealing
plasma and peripheral blood mononuclear cells in patients. focal spongiosis with perivascular lymphocytic infiltrates (periodic acid–Schiff
The negative results of these studies are not yet explained. stain, ×400).
418 Section 6  Other Papulosquamous Disorders

The herald patch is a primary lesion of pityriasis rosea are typically spared in children. The orientation of lesions
and presents as an erythematous large plaque with a is typically along lines of skin cleavage (Fig.  36.3) [76].
characteristic collarette scale. This is often misdiagnosed Oral and pharyngeal involvement in children (35%) is
as dermatophytic infection. more common than in adults (9%) (risk ratio: 2.19, 95%
In the peripheral blood in affected adults, elevated confidence interval [CI]: 1.30–3.69; calculated by the
erythrocyte sedimentary rate, decreased T lymphocytes authors of this chapter) [66].
and increased B lymphocytes have been reported [24]. Forty eight percent of children with pityriasis rosea have
Similar changes were noted for children with pityriasis systemic symptoms, as compared to 69% of adults (risk
rosea [65]. Autoantibodies were reported to be associated ratio: 0.70, 95% CI: 0.55–0.89) [66]. Most children do not
with pityriasis rosea in adults [68,69]. These findings were experience intense pruritus [74]. The impact of the rash on
not reported for children [65]. The roles of inflammatory the quality of life of children with pityriasis rosea is signifi-
mediators in pityriasis rosea are also being explored [73]. cantly less than for children with atopic dermatitis [75].
The total rash duration for children (average of 16 days) is
Clinical features. Pityriasis rosea in children is very simi- significantly shorter than that for adults (average of 45
lar clinically to the presentation in adults [66,74]. A herald days) [66]. Complications are rare in children and adoles-
patch is present in less than half of all sufferers. However, cents. When complications do occur, they are more likely
the time lapse between the herald patch and the second- to be related to the drug treatments rather than the disease
ary eruption (around 4 days) might be much shorter for [15]. Most children have no relapse in their lifetime.
children as compared to that in adults (around 2 weeks)
[66]. The exact incidence of the herald patch in children is Atypical presentations. Atypical features include atypical
not known. The herald patch is a solitary round to oval rash morphology (vesicular, purpuric, haemorrhagic and
lesion most commonly affecting the trunk and upper urticarial), atypical rash size (pityriasis rosea gigantea for
arms. It is annular in configuration with a raised border of exceptionally large lesions, papular pityriasis rosea for
fine, adherent scales (Fig. 36.2) [28]. exceptionally small lesions), atypical rash distribution
PAPULOSQUAMOUS DISORDERS

The secondary eruption occurs 1–3 weeks after onset of (distal aspects of extremities), atypical number of lesions,
the herald patch. Secondary lesions have a similar mor-
phology to the herald patch, although they are usually
SECTION 6: OTHER

smaller. Collarette scaling is a characteristic feature of


lesions. The scaling is circinate or oval, and fine fragments
of scales are attached only at the periphery of lesions. In
children, the centre of the lesions might also be covered
with fine scales which are lichenoid‐like with diagonal
lines [75].
In children with typical pityriasis rosea, only the trunk
and proximal aspects of the four extremities are involved.
This distribution pattern is traditionally termed a T‐shirt‐
and‐shorts or bathing suit pattern [18]. Palms and soles

Fig. 36.2  Herald plaque of a child with pityriasis rosea, which presents as Fig. 36.3  Truncal lesions of pityriasis rosea in an adolescent boy showing
an erythematous large plaque with a characteristic collarette scale. This is scaly round and oval plaques on the trunk. The herald patch is arrowed in
often misdiagnosed as dermatophytic infection. red. Orientation of the lesions is along lines of skin creases.
Chapter 36  Pityriasis Rosea 419

Box 36.1  The diagnostic criteria of pityriasis rosea [78,79]. Box 36.2  The classification of pityriasis rosea. This classification is
These diagnostic criteria are applicable to infants and children applicable to infants and children with pityriasis rosea
with pityriasis rosea
Axis 1 – Prodromal symptoms and herald patch
A patient is diagnosed as having pityriasis rosea if:
Prodromal symptoms – Present (coryza, fever, generalized muscle
• On at least one occasion or clinical encounter, he/she has all the
pain, fatigue, malaise, arthralgia, gastrointestinal symptoms),
essential clinical features and at least one of the optional
transient/barely noticeable, absent or unreliable history from patient
clinical features, and
• On all occasions or clinical encounters related to the rash, he/she Herald patch – Absent, obscure, single, multiple, gigantic, lonely
does not have any of the exclusional clinical features (herald patch as the only manifestation)

The essential clinical features are: Axis 2 – Number and distribution of lesions


• Discrete circular or oval lesions
• Scaling on most lesions, and Number – Few lesions (less than five, oligolesional), many lesions,
• Peripheral collarette scaling with central clearance on at least two suberythrodermic, erythroderma due to pityriasis rosea
lesions Distribution – Relatively unilateral, absolutely unilateral, localized,
regional, mucosal (mainly oral and genital), acral, pityriasis rosea
The optional clinical features are:
inversus (mainly involving the flexures), shoulders and hips (limb‐girdle
• Truncal and proximal limb distribution, with less than 10% of
pityriasis rosea), fewer but larger lesions localized to axillae and
lesions distal to mid‐upper‐arm and mid‐thigh
groins (pityriasis circinata et marginata of Vidal), actinic (photodis-
• Orientation of most lesions along skin cleavage lines, and
tributed), photospared
• A herald patch (not necessarily the largest) appearing at least 2
days before eruption of other lesions, from history of the patient
Axis 3 – Size, morphology and orientation of lesions
or from clinical observation
Size – Gigantic (pityriasis rosea gigantea of Darier) against miniature
The exclusional clinical features are:

PAPULOSQUAMOUS DISORDERS
Morphology – Papular, papulo‐squamous, papulo‐vesicular, vesicular,
• Multiple small vesicles at the centre of two or more lesions
lichenoid, urticarial, erythema multiforme‐like, punctate/purpuric/
• Two or more lesions on palmar or plantar skin surfaces, and
haemorrhagic, follicular
• Clinical or serological evidence of secondary syphilis

SECTION 6: OTHER
Orientation – Typical (along the skin creases), atypical (not along the
skin creases), scattered, blaschkoid (along lines of Blaschko),
segmental (along dermatomes)
atypical site of lesions (oral cavity) and atypical severity
of symptoms. Atypical rashes are fairly common in adults Axis 4 – Symptoms and clinical course
[77] but are uncommon in children [75]. Papular pityriasis
rosea is the most common atypical feature in children. Symptoms – Severely pruritic (pityriasis rosea irritata), slightly pruritic,
moderately pruritic, nonpruritic, asymptomatic
Diagnostic criteria and  classification. Validated diag- Clinical course – Too short (less than 2 weeks), too long (more than 6
nostic criteria are available for the diagnosis of children months)
with marginal clinical features [78,79] (Box 36.1), and Recurrent pityriasis rosea
adoption of these might enhance diagnosis as well as
assure high homogeneity when recruiting children for Axis 5 – Seasonal morphological variations
noninterventional studies [80]. Two modern classifi- Psoriasiform in summer/crusted/haemalis form in winter
cations [81,82] (Box 36.2) also cater for children with
pityriasis rosea.

Drug‐induced pityriasis rosea‐like rashes. Numerous


patients, adults and children alike, have been reported to stages of erythema multiforme, guttate hypomelanosis
have pityriasis rosea‐like eruption. The clinical manifesta- and pityriasis lichenoides [84].
tions of these patients usually do not fulfil the diagnostic Papular pityriasis rosea in children has to be differenti-
criteria. These rashes should not be confused with pityria- ated from viral rashes including roseola infantum and
sis rosea, typical or atypical, which fulfils the diagnostic rubella. Vesicular pityriasis rosea is uncommon in
criteria [83]. These patients should not be recruited to children. Differential diagnoses of this variant include
laboratory or clinical studies and clinical trials. The man- pompholyx, bullous erythema multiforme, chickenpox,
agement should follow that for drug eruptions. dermatitis herpetiformis, linear immunoglobulin A dis-
ease and pemphigus vulgaris [77].
Differential diagnosis. Typical pityriasis rosea in children
should be diagnosable clinically. Common differential Treatment. Pityriasis rosea in children and adolescents
diagnoses for children and adolescents include guttate is a self‐limited disease. Unlike their adult counterparts,
psoriasis, lichen planus, nummular dermatitis, parapso- this eruption usually has little to no impact on the qual-
riasis, pityriasis versicolor, tinea corporis, viral rashes and ity of life of children [74]. The aim of treatment is thus
drug eruptions [28]. Uncommon differential diagnoses mainly to relieve pruritus, not to modify the course of
are secondary syphilis for adolescents, and the early the disease.
420 Section 6  Other Papulosquamous Disorders

Oral aciclovir, in various dosages, has demonstrated 21 Ayanlowo O, Akinkugbe A, Olumide Y. The pityriasis rosea calendar:
a 7 year review of seasonal variation, age and sex distribution. Nig Q
efficacy in treating adults with pityriasis rosea [72,85–91].
J Hosp Med 2010;20:29–31.
The results of studies on the use of macrolides for adult 22 Ganguly S. A clinicoepidemiological study of pityriasis rosea in
patients vary [92–96]. In any case, macrolides are likely to South India. Skinmed 2013;11:141–6.
be of lower efficacy than aciclovir [87,89,97]. The efficacy 23 Özyürek GD, Alan S, Cenesizoğlu E. Evaluation of clinico‐
epidemiological and histopathological features of pityriasis rosea.
of ultraviolet radiation is inconclusive [98,99], and it Postepy Dermatol Alergol 2014;31:216–21.
should not be a conventional treatment modality for chil- 24 Björnberg A, Hellgren L. Pityriasis rosea. A statistical, clinical and
dren with pityriasis. laboratory investigation of 826 patients and matched healthy controls.
For children with little or no pruritus and a limited Acta Derm Venereol 1962;42(suppl 50):1–50.
25 Jawade SA, Chugh VS, Gohil SK et al. A clinico‐etiological study of
number of lesions, no treatment is generally required. For dermatoses in pediatric age group in tertiary health care center in
those with moderate pruritus, topical calamine lotion South Gujarat Region. Indian J Dermatol 2015;60:635.
and/or topical emollients might be considered. For chil- 26 Hendricks AA, Lohr JA. Pityriasis rosea in infancy. Arch Dermatol
1979;115:896–7.
dren with severe pruritus affecting their quality of life, 27 Hyatt H. Pityriasis rosea in a three month old. Arch Pediatr
oral sedating antihistamines might be given as a single 1960;77:364.
nocturnal dose. Topical corticosteroids of mild to moder- 28 Truhan AP. Pityriasis rosea. Am Fam Physician 1984;29:193–6.
ate potency might be considered for short durations for 29 Drago F, Ciccarese G, Rebora A, Parodi A. Relapsing pityriasis
rosea. Dermatology 2014;229:316–8.
children with particularly severe pruritus. 30 Chuh A, Lee A, Molinari N. Case clustering in pityriasis rosea – a
A Cochrane systematic review found that oral erythro- multi‐center epidemiologic study in primary care settings in Hong
mycin might be effective for adult patients, but further Kong. Arch Dermatol 2003;139:489–93.
31 Chuh A, Molinari N, Sciallis G et al. Temporal case clustering in pity-
clinical studies are necessary before erythromycin or
riasis rosea  –  a regression analysis on 1379 patients in Minnesota,
other macrolides can be formally recommended for adult Kuwait and Diyarbakýr, Turkey. Arch Dermatol 2005;141:767–71.
patients [100]. At the present stage of knowledge, these 32 Chuh AAT. The association of pityriasis rosea with cytomegalovirus,
treatment modalities cannot be routinely recommended Epstein–Barr virus and parvovirus B19 infections – a prospective case
control study by polymerase chain reaction and serology. Eur J
for children with pityriasis rosea. Dermatol 2003;13:25–8.
PAPULOSQUAMOUS DISORDERS

33 Bonafé JL, Icart J, Perpère M et  al. Histopathologic, ultrastructural,


References immunologic and virologic study of Gibert’s pityriasis rosea. Ann
1 Feinstein A, Kahana M. Pityriasis rosea of Gibert. J Am Acad Dermatol Dermatol Venereol 1982;109:855–61.
SECTION 6: OTHER

1987;16:1260. 34 Hudson LD, Adelman S, Lewis CW. Pityriasis rosea. Viral comple-
2 Percival GH. Pityriasis rosea. Br J Dermatol 1932;44:241–53. ment fixation studies. J Am Acad Dermatol 1981;4:544–6.
3 Klauder JV. Pityriasis rosea with particular reference to its unusual 35 Marcus‐Farber BS, Bergman R, Ben Porath E et al. Serum antibodies to
manifestations. JAMA 1924;82:178–83. parvovirus B19 in patients with pityriasis rosea. Dermatology
4 Sarkany I, Hare PJ. Pityriasis rotunda (pityriasis circinata). Br J 1997;194:371.
Dermatol 1964;76:223–7. 36 Raskin J. Possible dermatropic virus associated with pityriasis rosea.
5 Weiss L. Pityriasis rosea – an erythematous eruption of internal ori- Preliminary study. Acta Derm Venereol 1968;48;474–81.
gin. JAMA 1903;41:20–8. 37 Metz J. An electron microscopic investigation of the pityriasis rosea. J
6 De Souza Sittart JA, Tayah M, Soares Z. Incidence pityriasis rosea of Cutan Pathol 1977;4:228.
Gibert in the Dermatology Service of the Hospital do Servidor Publico 38 Neubert U, Ring J. Immunological monitoring in pityriasis rosea. 17th
in the state of Sao Paulo. Med Cutan Ibero Lat Am 1984;12:336–8. International Congress of Dermatology, Berlin, 1987:464.
7 Olumide Y. Pityriasis rosea in Lagos. Int J Dermatol 1987;26:234–6. 39 Aractingi S, Morinet F, Mokni M et  al. Absence of picornavirus
8 Vollum DI. Pityriasis rosea in the African. Trans St Johns Hosp genome in pityriasis rosea. Arch Dermatol Res 1996;289:60–1.
Dermatol Soc 1973;59:269–71. 40 Gjenero‐Margan I, Vidovic R, Drazenovic V. Pityriasis rosea Gibert:
9 Jacyk WK. Pityriasis rosea in Nigerians. Int J Dermatol detection of Legionella micdadei antibodies in patients. Eur J Epidermiol
1980;19:397–9. 1995;11:459–62.
10 Ahmed MA. Pityriasis rosea in the Sudan. Int J Dermatol 41 Chuh AAT, Chan HHL. Prospective case–control study of chlamydia,
1986;25:184–5. legionella and mycoplasma infections in patients with pityriasis
11 Giam YC. Skin diseases in children in Singapore. Ann Acad Med rosea. Eur J Dermatol 2002;12:170–3.
Singapore 1988;17:569–72. 42 Hudson LD, Adelman S, Lewis CW. Pityriasis rosea. Viral comple-
12 Harman M, Aytekin S, Akdeniz S, Inaloz HS. An epidemiological ment fixation studies. J Am Acad Dermatol 1981;4:544–6.
study of pityriasis rosea in Eastern Anatolia. Eur J Epidemiol 43 Ishibashi A, Ueda I, Fujita K. Pityriasis rosea Gibert and Mycoplasma
1998;14:495–7. pneumoniae infection. J Dermatol 1985;12:97–100.
13 Nanda A, Al‐Hasawi F, Alsaleh QA. A prospective survey of pediatric 44 Drago F, Ranieri E, Malaguti F et al. Human herpesvirus 7 in pityria-
dermatology clinic patients in Kuwait: an analysis of 10,000 cases. sis rosea. Lancet 1997;349:1367–8.
Pediatr Dermatol 1999;16:6–11. 45 Drago F, Ranieri E, Malaguti F, Battifoglio ML. Human herpesvirus 7
14 Tay YK, Goh CL. One‐year review of pityriasis rosea at the National in patients with pityriasis rosea. Electron microscopy investigations
Skin Centre, Singapore. Ann Acad Med Singapore 1999;28:829–31. and polymerase chain reaction in mononuclear cells, plasma and skin.
15 Traore A, Korsaga‐Some N, Niamba P et al. Pityriasis rosea in second- Dermatology 1997;195:374–8.
ary schools in Ouagadougou, Burkina Faso. Ann Dermatol Venereol 46 Drago F, Ranieri E, Malaguti F, Battifoglio ML. Human herpesvirus
2001;128:605–9. 7 in patients with pityriasis rosea. Electron microscopy investiga-
16 Messenger AG, Knox EG, Summerly R et al. Case clustering in pity- tions and polymerase chain reaction in mononuclear cells, plasma
riasis rosea: support for role of an infective agent. BMJ (Clin Res Ed) and skin. Dermatology 1997;195:374–8.
1982;284:371–3. 47 Drago F, Malaguti F, Ranieri E et al. Human herpes virus‐like parti-
17 Chuang TY, Ilstrup DM, Perry HO, Kurland LT. Pityriasis rosea in cles in pityriasis rosea lesions: an electron microscopy study. J
Rochester, Minnesota, 1969 to 1978. J Am Acad Dermatol Cutan Pathol 2002;29:359–61.
1982;7:80–9. 48 Broccolo F, Drago F, Careddu AM et al. Additional evidence that pity-
18 Abercrombie GF. Pityriasis rosea. Proc Roy Soc Med 1962;55:556–7. riasis is associated with reactivation of human herpesvirus‐6 and ‐7. J
19 Cheong WK, Wong KS. An epidemiological study of pityriasis rosea Invest Dermatol 2005;124:1234–40.
in Middle Road Hospital. Singapore Med J 1989;30:60–2. 49 Drago F, Broccolo F, Ciccarese G et al. Persistent pityriasis rosea: an
20 Sharma L, Srivastava K. Clinicoepidemiological study of pityriasis unusual form of pityriasis rosea with persistent active HHV‐6 and
rosea. Indian J Dermatol Venereol Leprol 2008;74:647–9. HHV‐7 infection. Dermatology 2015;230:23–6.
Chapter 36  Pityriasis Rosea 421

50 Drago F, Broccolo F, Javor S et al. Evidence of human herpesvirus‐6 75 Cavanaugh RM Jr. Pityriasis rosea in children. A review. Clin
and ‐7 reactivation in miscarrying women with pityriasis rosea. J Pediatr (Phila) 1983;22:200–3.
Am Acad Dermatol 2014;71:198–9. 76 Chuh AAT. Rash orientation in pityriasis rosea – a qualitative study.
51 Watanabe T, Sugaya M, Nakamura K, Tamaki K. Human herpesvi- Eur J Dermatol 2002;12:253–6.
rus 7 and pityriasis rosea. J Invest Dermatol 1999;113:288–9. 77 Chuh A, Zawar V, Lee A. Atypical presentations of pityriasis rosea – case
52 Watanabe T, Kawamura T, Jacob SE et al. Pityriasis rosea is associ- presentations. J Eur Acad Dermatol Venereol 2005;19:120–6.
ated with systemic active infection with both human herpesvirus‐7 78 Chuh AA. Diagnostic criteria for pityriasis rosea: a prospective
and human herpesvirus‐6. J Invest Dermatol 2002;119:793–7. case control study for assessment of validity. J Eur Acad Dermatol
53 Vág T, Sonkoly E, Kárpáti S et al. Avidity of antibodies to human Venereol 2003;17:101–3.
herpesvirus 7 suggests primary infection in young adults with pity- 79 Chuh A, Lee A, Zawar V. The diagnostic criteria of Gianotti‐Crosti
riasis rosea. J Eur Acad Dermatol Venereol 2004;18:738–40. syndrome: are they applicable to children in India? Pediatr
54 Vág T, Sonkoly E, Kemeny B et al. Studies on the novel association of Dermatol 2004;21:542–7.
human herpesvirus‐7 with skin diseases. Orv Hetil 2003;144:1623–9. 80 Chuh A, Zawar V, Sciallis GF, Lee A. The diagnostic criteria of pity-
55 Canpolat Kirac B, Adisen E, Bozdayi G et al. The role of human her- riasis rosea and Gianotti‐Crosti syndrome – a protocol to establish
pesvirus 6, human herpesvirus 7, Epstein–Barr virus and cytomegalo- diagnostic criteria of skin diseases. J R Coll Physicians Edinb
virus in the aetiology of pityriasis rosea. J Eur Acad Dermatol Venereol 2015;45:218–25.
2008;14:6. 81 Zawar V, Chuh A. Follicular pityriasis rosea. A case report and a
56 Kempf W, Adams V, Kleinhans M et al. Pityriasis rosea is not associ- new classification of clinical variants of the disease. J Dermatol
ated with human herpesvirus 7. Arch Dermatol 1999;135:1070–2. Case Rep 2012;6:36–9.
57 Yoshida M. Detection of human herpesvirus 7 in patients with pity- 82 Drago F, Ciccarese G, Rebora A et al. Pityriasis rosea: a comprehen-
riasis rosea and healthy individuals. Dermatology 1999;199:197. sive classification. Dermatology 2016;232:431–7.
58 Yasukawa M, Sada E, Machino H, Fugita S. Reactivation of human 83 Chuh AA. Nomenclature of drug‐induced pityriasis rosea‐like
herpesvirus 6 in pityriasis rosea. Br J Dermatol 1999;140:169–70. rashes. Neuropsychiatr Dis Treat 2015;11:2547–9.
59 Kosuge H, Tanaka‐Taya K, Miyoshi H et al. Epidemiological study of 84 Fox BJ, Odom RB. Papulosquamous diseases: a review. J Am Acad
human herpesvirus‐6 and human herpesvirus‐7 in pityriasis rosea. Br Dermatol 1985;12:597–624.
J Dermatol 2000;143:795–8. 85 Drago F, Vecchio F, Rebora A. Use of high‐dose acyclovir in pityriasis
60 Offidani A, Pritelli E, Simonetti O et  al. Pityriasis rosea associated rosea. J Am Acad Dermatol 2006;54:82–5.
with herpesvirus 7 DNA. (correspondence) J Eur Acad Dermatol 86 Castanedo‐Cazares JP, Lepe V, Moncada B. Antivirals for pityriasis
Venereol 2000;14:313–14. rosea. Photodermatol Photoimmunol Photomed 2004;20:110.
61 Chuh AAT, Chiu SSS, Peiris JSM. Human herpesvirus 6 and 7 DNA 87 Ehsani A, Esmaily N, Noormohammadpour P et al. The comparison

PAPULOSQUAMOUS DISORDERS
in peripheral blood leukocytes and plasma in patients with pityria- between the efficacy of high dose acyclovir and erythromycin on
sis rosea by polymerase chain reaction – a prospective case control the  period and signs of pitiriasis rosea. Indian J Dermatol 2010;
study. Acta Derm Venereol 2001;81:289–90. 55:246–8.
62 Wong WR, Tsai CY, Shih SR, Chan HL. Association of pityriasis rosea 88 Rassai S, Feily A, Sina N, Abtahian S. Low dose of acyclovir may be

SECTION 6: OTHER
with human herpesvirus‐6 and human herpesvirus‐7 in Taipei. an effective treatment against pityriasis rosea: a random investiga-
J Formos Med Assoc 2001;100:478–83. tor‐blind clinical trial on 64 patients. J Eur Acad Dermatol Venereol
63 Karabulut AA, Kocak M, Yilmaz N, Eksioglu M. Detection of human 2011;25:24–6.
herpesvirus 7 in pityriasis rosea by nested PCR. Int J Dermatol 89 Amatya A, Rajouria EA, Karn DK. Comparative study of effective-
2002;41:563–7. ness of oral acyclovir with oral erythromycin in the treatment of
64 Yildirim M, Aridogan BC, Baysal V, Inaloz HS. The role of human pityriasis rosea. Kathmandu Univ Med J (KUMJ) 2012;10:57–61.
herpes virus 6 and 7 in the pathogenesis of pityriasis rosea. Int J Clin 90 Ganguly S. A randomized, double‐blind, placebo‐controlled study of
Pract 2004;58:119–21. efficacy of oral acyclovir in the treatment of pityriasis rosea. J Clin
65 Chuh AA, Peiris JS. Lack of evidence of active human herpesvirus 7 Diagn Res 2014;8:YC01–4.
(HHV‐7) infection in three cases of pityriasis rosea in children. Pediatr 91 Das A, Sil A, Das NK et  al. Acyclovir in pityriasis rosea: An
Dermatol 2001;18:381–3. observer‐blind, randomized controlled trial of effectiveness,
66 Drago F, Ciccarese G, Broccolo F et  al. Pityriasis rosea in children: safety and tolerability. Indian Dermatol Online J 2015;6:181–4.
clinical features and laboratory investigations. Dermatology 92 Sharma PK, Yadav TP, Gautam RK et al. Erythromycin in pityriasis
2015;231:9–14. rosea: a double‐blind, placebo‐controlled clinical trial. J Am Acad
67 Burch PRJ, Rowell NR. Pityriasis rosea – an auto‐aggressive disease? Dermatol 2000;42:241–4.
Br J Dermatol 1970;82:549–60. 93 Rasi A, Tajziehchi L, Savabi‐Nasab S. Oral erythromycin is ineffec-
68 Hosokawa H, Horio S, Takiuchi Y et al. Naturally occurring T lym- tive in the treatment of pityriasis rosea. J Drugs Dermatol
phocytotoxic antibody in viral and related skin diseases. Acta Derm 2008;7:35–8.
Venereol 1984;64:275–80. 94 Amer A, Fischer H. Azithromycin does not cure pityriasis rosea.
69 Chuh AAT. A prospective case control study of autoimmune markers Pediatrics 2006;117:1702–5.
in patients with pityriasis rosea. Clin Exp Dermatol 2003;28:449–50. 95 Pandhi D, Singal A, Verma P, Sharma R. The efficacy of azithromycin
70 Chuang TY, Perry HO, Ilstrup DM, Kurland LT. Recent upper respira- in pityriasis rosea: a randomized, double‐blind, placebo‐controlled
tory tract infection and pityriasis rosea: a case–control study of 249 trial. Indian J Dermatol Venereol Leprol 2014;80:36–40.
matched pairs. Br J Dermatol 1983;108:587–91. 96 Bukhari IA. Oral erythromycin is ineffective in the treatment of pity-
71 Okamoto H, Imamura S, Aoshima T et al. Dyskeratotic degeneration riasis rosea. J Drugs Dermatol 2008;7:625.
of epidermal cells in pityriasis rosea: light and electron microscopic 97 Amatya A, Rajouria EA, Karn DK. Comparative study of effective-
studies. Br J Dermatol 1982;107:189–94. ness of oral acyclovir with oral erythromycin in the treatment of
72 Kempf W, Burg G. Pityriasis rosea – a virus‐induced skin disease? pityriasis rosea. Kathmandu Univ Med J (KUMJ) 2012;10:57–61.
An update. Arch Virol 2000;145:1509–20. 98 Arndt KA, Paul BS, Stern RS, Parrish JA. Treatment of pityriasis
73 Drago F, Ciccarese G, Broccolo F et  al. The role of cytokines, rosea with UV radiation. Arch Dermatol 1983;119:381–2.
chemokines, and growth factors in the pathogenesis of pityriasis 99 Leenutaphong V, Jiamton S. UVB phototherapy for pityriasis rosea:
rosea. Mediators Inflamm 2015;2015:438963. a bilateral comparison study. J Am Acad Dermatol 1995;33:996–9.
74 Chuh AAT. Quality of life in children with pityriasis rosea  –  a 100 Chuh AA, Dofitas BL, Comisel GG et al. Interventions for pityriasis
­prospective case control study. Pediatr Dermatol 2003;20:474–8. rosea. Cochrane Database Syst Rev 2007;2:CD005068.
423

C HA PTER   37

Pyodermas and Bacterial
Toxin‐mediated Syndromes
James R. Treat1, Christian R. Millett2, Warren R. Heymann3 & Steven M. Manders3
1
 Children’s Hospital of Philadelphia and Dermatology Section, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
2
 Forefront Dermatology, Vienna, VA, USA
3
 Division of Dermatology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Camden, NJ, USA

Pathophysiology, 423 Localized cutaneous staphylococcal and Toxin‐mediated staphylococcal and


Epidemiology, 425 streptococcal infections: pyodermas, 426 streptococcal disease, 430

Abstract recognized clinically. SA and GAS most commonly cause localized


cutaneous infections but can lead to serious systemic diseases such
as cellulitis, staphylococcal skin syndrome and necrotizing fasciitis.
Cutaneous bacterial infections in children are common but
It has become more important to consider culturing the skin to
­important to recognize as they can lead to significant sequelae.
establish bacterial sensitivities due to the increase in SA that is
Staphylococcus aureus (SA) and Group A Streptococcus (GAS) are
meticillin resistant.
the most common causes of infection. Skin infection with these
bacteria typically presents with pustules and crusting and can be

Key points • Staphylococcus aureus and group A streptococci can release


toxins that lead to widespread erythema and, in the case of toxic
shock, severe systemic sequelae.
• Superficial bacterial skin infections often present with pustules,
• Staphylococcal scalded skin syndrome is differentiated from
bullae or crusting, and swab cultures can help establish antibiotic

SECTION 7: BACTERIAL SKIN


Stevens–Johnson syndrome by the lack of mucosal involvement
sensitivities.
in staphylococcal scalded skin syndrome.

INFECTIONS
Pathophysiology
exotoxins, which are particularly potent virulence factors
Streptococci because they function as superantigens. Unlike traditional
Group A Streptococcus (GAS) is a common infectious agent antigens, superantigens have the ability to stimulate
in children, having a large number of virulence factors immune cells without undergoing antigen processing
responsible for a broad range of disease. It is a Gram‐ and presentation by antigen‐presenting cells (APCs) [3].
positive organism that is seen in chains on Gram stain. Conventional antigens are processed within the APC, and
On blood agar, GAS displays characteristic β‐haemolysis protein fragments of the antigen are then expressed on
due to the haemolysin streptolysin S. GAS has numerous the cell surface in the groove of the major histocompati-
surface and extracellular factors that confer virulence, of bility type II complex (MHCII). The antigen–MHCII com-
which the cell surface M protein is the main antigenic plex then interacts with the T cell receptor in a very
determinant. It aids in adherence but most importantly specific, antigen‐restricted fashion. The segment of T cells
enables the bacterium to evade phagocytosis. GAS classi- bearing the receptor that corresponds to the antigen is
fication is based on genotyping of the M protein. There then activated, with resultant cytokine production and
are currently approximately 180 emm sequence types and specific immune activation.
800 emm subtypes described, but new types and subtypes Whereas conventional antigens require recognition of
are still being identified [1]. all five elements of the T cell receptor (Va, Ja, Vb, Db, Jb),
Invasive GAS disease is defined by the isolation of the recognition sequence for superantigens is almost
GAS from a normally sterile body site, and includes entirely dependent on Vb only. Because only a limited
­entities such as streptococcal toxic shock syndrome and amount of Vb genes exist, a given superantigen–T cell
necrotizing fasciitis [2]. These infections depend in large interaction may lead to the activation of 5–30% of the
part on the ability of the bacteria to produce streptococcal entire T cell population, whereas conventional antigens

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
424 Section 7  Bacterial Skin Infections

activate approximately 0.01–0.1% of the body’s T cells. syndrome (SSSS) and bullous impetigo [5]. TSST‐1,
This large‐scale activation of T cells by superantigens among other toxins produced by Staph. aureus, can also
leads to massive cytokine production, especially that of cause TSS, an acute life‐threatening illness.
tumour necrosis factor (TNF)‐α, interleukin (IL)‐1 and Staph. epidermidis is a commensal organism on the skin.
IL‐6. These cytokines, especially TNF‐α and IL‐1, have Although it cannot lead to purulent infections, it is an
been shown to mediate clinical effects such as fever, ery- important cause of neonatal sepsis and central line infec-
thematous rash, emesis, hypotension, tissue injury and tions, especially in immunosuppressed hosts.
shock. Certain superantigen‐mediated illnesses appear
to induce a large‐scale depletion of particular Vb sub- Meticillin‐resistant Staphylococcus
sets; this may result from apoptosis of initially activated aureus (MRSA)
T cells [4]. In 1942, the first penicillin‐resistant Staph. aureus isolate
Host antibacterial and antitoxic immunity to GAS is an was observed, and in 1961, Staph. aureus developed meti-
important aspect in the pathogenesis of cutaneous and cillin resistance due to the acquisition of the mecA gene. It
systemic infections. Antibacterial immunity is related to has been suggested that MRSA originated in vivo through
the type‐specific M component of GAS. For example, the horizontal transfer of the mecA gene between two
infection with type 4 Streptococcus is followed by staphylococcal species. During the last 45 years, various
development of specific neutralizing antibody for M type hospital‐associated MRSA (HA‐MRSA) clones have dis-
4‐bearing organisms, but not against heterologous types. seminated worldwide. In addition, community‐associated
Antitoxic immunity follows exposure to erythrogenic tox- MRSA (CA‐MRSA) clones have become much more prev-
ins such as the streptococcal pyrogenic exotoxins (SPEs). alent [6]. Skin and soft tissue infections, especially due to
Most commonly, these toxins produce the exanthem and MRSA, have increased, leading to more hospitalizations [7].
constitutional manifestations of scarlet fever; rarely they Meticillin resistance occurs by the presence of the
may cause toxic shock syndrome (TSS). A child with anti- mecA gene, which encodes penicillin‐binding protein 2a.
bacterial immunity to a strain with a particular M type Penicillin‐binding protein is a transpeptidase that restores
will not develop clinical disease. Conversely, in the cell wall biosynthesis of MRSA in the presence of β‐lactams.
absence of M‐type specific immunity, an infected child The mecA gene is carried on the staphylococcal methicillin
will develop scarlet fever if he or she lacks immunity to resistance gene cassette, or staphylococcal cassette chro-
the specific erythrogenic toxin. Furthermore, failure to mosome mec (SCCmec). CA‐MRSA and HA‐MRSA have
produce an adequate humoral immunity to toxins can different types of cassettes and antibiotic resistance.
SECTION 7: BACTERIAL SKIN

lead to recurrent toxin‐mediated disease. CA‐MRSA typically has SCCmec type IV, and is generally
The immune response to various streptococcal exo- susceptible to most non‐β‐lactam antibiotics. In contrast,
products has been used in several diagnostic serological HA‐MRSA usually carries SCCmec types I, II or III, and is
tests, most notably the response to streptolysin O. The resistant to aminoglycosides, clindamycin and macrolides.
INFECTIONS

antistreptolysin O (ASO) titre is generally elevated in Infection with HA‐MRSA can occur in the community so
patients recovering from a recent streptococcal infection. Staph. aureus susceptibilities can no longer be reliably
This test may be useful in certain settings to confirm or predicted without culturing the bacteria.
support a diagnosis. In addition, there are two important A cloned strain of CA‐MRSA called USA300 defies this
late sequelae of GAS infections that appear 1–3 weeks generalization of antibiotic resistance. USA300 carries the
later: glomerulonephritis and rheumatic fever. The patho- genes that encode resistance to macrolides, lincosamides,
genesis remains poorly understood, and appears to be streptogranin B, tetracycline, doxycycline and mupirocin
best explained by an abnormal immune response or [8]. The USA300 strain has predominated in outbreaks
hypersensitivity to streptococcal antigens. throughout the USA, suggesting that it possesses viru-
lence or transmissibility factors that confer unusual path-
Staphylococci ogenicity. Recent sequencing of the complete genome of
Staphylococci are Gram‐positive cocci found in clusters this strain identified a mobile genetic element, termed
on Gram stain. Staphylococcus aureus is usually a tran- arginine catabolic mobile element (ACME). It has been
sient pathogenic organism on the skin, and can asymp- hypothesized that the products of this gene cluster
tomatically colonize the nasal mucosa. In certain enhance the capacity of USA300 strains to survive at low
situations, however, Staph. aureus can cause skin or pH on human skin and within phagocytic cells [9].
invasive infections via expression of a wide array of Other toxins produced by MRSA also contribute to the
virulence factors, including wall teichoic acid (TA) and pathogenesis of skin and soft tissue infections. Panton–
surface proteins. These promote adherence to damaged Valentine leukocidin (PVL) is a cytotoxin of CA‐MRSA
tissue and diminish neutrophil function and immune involved in the development of abscesses, cellulitis and
response. Staph. aureus also secretes exotoxins and furuncles. PVL lyses leucocytes by facilitating calcium
enzymes that can contribute to a variety of cutaneous channel opening and pore formation, and increases IL‐8
and systemic infections, including four haemolysins (a‐, secretion which leads to an inflammatory response and
b‐, d‐ and g‐toxins). Staphylococcal exfoliative toxins tissue necrosis. The genes that encode PVL have been
(ETs), including ETA, ETB and ETD, disrupt desmo- found in almost all CA‐MRSA isolates [8].
glein‐1, a desmosomal cadherin expressed in the upper Risk factors for acquisition of HA‐MRSA include a his-
epidermis. This results in staphylococcal scalded skin tory of hospitalization, surgery, dialysis or residence in a
Chapter 37  Pyodermas and Bacterial Toxin‐mediated Syndromes 425

long‐term care facility within 1 year before the MRSA 9 Gorwitz R. A review of community‐associated methicillin resistant
Staphylococcus aureus skin and soft tissue infections. Pediatr Infect
culture date, presence of an indwelling device at the time
Dis J 2008;27:1–7.
of culture, or a previous history of MRSA infection or 10 Stryjewski M, Chambers H. Skin and soft tissue infections caused by
­colonization. CA‐MRSA has been defined by some as an community‐acquired methicillin‐resistant Staphylococcus aureus.
MRSA infection with onset in the community in a patient Clin Infect Dis 2008;46:S368–77.
11 Patel, A, Hill E, Simpson, E, Hanafin J. Reversion of methicillin‐
lacking established HA‐MRSA risk factors. However, it is resistant Staphylococcus aureus skin infections to methicillin‐
generally not possible to determine with certainty where susceptible. AMA Dermatol 2013;149:1167–71.
MRSA was initially acquired [9]. Staph. aureus can persist 12 Fritz SA1, Hogan PG, Camins BC et al. Mupirocin and chlorhexidine
as a colonizer for years, leading to misclassification of the in Staphylococcus aureus in patients with community‐onset skin and
soft tissue infections. Antimicrob Agents Chemother 2013;57:559–68.
source. Indeed, some ‘community‐onset’ infections may 13 Milstone AM, Elward A, Song X et al. Daily chlorhexidine bathing to
in fact be caused by hospital‐acquired strains, leading to reduce bacteraemia in critically ill children: a multicentre, cluster‐
a  blurring of the distinction between CA‐MRSA and randomised, cross‐over trial. Lancet 2013;381:1099–106.
HA‐MRSA. Nevertheless, SCCmec type IV and PVL
have been useful molecular markers to identify true Epidemiology
CA‐MRSA strains.
Infections caused by CA‐MRSA predominantly affect Streptococci
children and young adults. Direct contact with infected In the past two decades, there has been a re‐emergence of
patients, colonized subjects or a contaminated environ- serious streptococcal infections. Surface proteins, host
ment is implicated in the transmission of CA‐MRSA infec- factors and toxin production have all contributed to the
tion. Crowding and sharing of personal items appear to renewed virulence of these bacteria. The re‐emergence of
be important factors. Transmission has occurred through invasive GAS infections, such as necrotizing fasciitis and
activities in which direct contact is common and, although streptococcal TSS, has been closely linked with the
originally proven in people such as football players, wres- renewed prevalence of GAS bearing M‐1 and M‐3 surface
tlers and military personnel, it is now recognized that any proteins. Host factors in the general population appear to
activity involving direct skin contact can lead to transmis- be partially responsible for the re‐emergence of strepto-
sion [10]. Nasal colonization has also been identified as a cocci as major pathogens. Very young or immunocompro-
risk factor for infection [9]. mised persons are at high risk for infection with these
Culture of purulent material establishes the diagnosis bacteria. However, a large percentage of patients who
and allows susceptibility testing to guide therapy. Patients have serious disease are otherwise healthy. This is postu-

SECTION 7: BACTERIAL SKIN


infected with MRSA can have reversion to meticillin‐ lated to be due to the absence of previous exposure to
susceptible Staph. aureus so re‐culturing should be con- these more virulent strains of bacteria, as an absence of
sidered with each infection [11]. Subsequent cultures of protective antibody appears to predispose persons to
the nares and perianal area can also determine carrier infection. Bacterial toxin production has also been shown

INFECTIONS
states. There are many regimens that have been used for to be a highly important factor in the increased frequency
decolonization but none has proven universally success- of serious streptococcal and staphylococcal disease, with
ful. A large meta‐analysis of MRSA eradication showed toxins such as streptococcal pyrogenic exotoxin‐A (SPEA)
that mupirocin applied topically to the nasal area for being linked with the increased prevalence of serious
1 week was highly successful at eradicating nasal ­carriage, invasive GAS infections in the USA [1].
but this does not necessarily indicate clearance of the Around the world, there are an estimated 1.7 million
throat and anal colonization. Combining application of new cases per year and 500 000 deaths per year from seri-
mupirocin to the nasal and anal area with chlorhexidine ous GAS disease. In addition, there are over 100 million
gluconate washes may be more effective at eradicating cases of less serious skin infections per year. Since the
MRSA [12,13]. 1980s, severe GAS diseases have been increasing in inci-
dence and severity. The incidence of invasive GAS dis-
References ease in most industrialized countries is between 2.5 and
1 Steer A, Danchin M, Carapetis J. Group A streptococcal infections in 3 per 100 000, and mortality rates vary between 10% and
children. J Pediatr Child Health 2007;43:209–13. 20%. The burden of severe GAS disease is predominantly
2 Martin J, Green M. Group A streptococcus. Semin Pediatr Infect Dis
2006;17:140–8. in developing countries and impoverished populations.
3 Burnett A, Domachowske J. Therapeutic considerations for children It is estimated that more than 660 000 cases of invasive
with invasive group A streptococcal infections: a case series report and disease resulting in more than 160 000 deaths occur glob-
review of the literature. Clin Pediatr 2007;46:550–5.
ally each year, most in developing countries. The peak
4 Manders S. Toxin‐mediated streptococcal and staphylococcal dis-
ease. J Am Acad Dermatol 1998;39:383–98. incidence of these infections occurs in infants and the
5 Iwatsuki K, Yamasaki O, Morizane S, Oono T. Staphylococcal cutane- elderly [2].
ous infections: invasion, evasion, and aggression. J Dermatol Sci
2006;42:203–14.
6 Deurenberg R, Stobberingh E. The evolution of Staphylococcus aureus. Staphylococci
Infect Genet Evol 2008;8:747–63. The incidence and prevalence of MRSA infections have
7 Lopez MA, Cruz AT, Kowalkowski MA, Raphael JL. Trends in resource also been increasing. Over the last 20 years, there have
utilization for hospitalized children with skin and soft tissue infec-
been reports from many healthcare facilities documenting
tions. Pediatrics 2013;131:e718–25.
8 Kil E, Heymann W, Weinberg J. Methicillin‐resistant Staphylococcus increasing numbers of CA‐MRSA infections treated at the
aureus: an update for the dermatologist. Cutis 2008;81:227–32, 247–52. facility, increasing proportions of all MRSA infections that
426 Section 7  Bacterial Skin Infections

are community associated, and increasing proportions of (impetigo contagiosa) is the most common form of pyo-
all community‐acquired Staph. aureus infections that are derma and is usually due to GAS, whereas bullous impe-
meticillin resistant. To date, reported CA‐MRSA infec- tigo is usually due to Staph. aureus [1]. Nonbullous impetigo
tions have disproportionately affected children, young represents a host response to the infection, whereas
adults and low socioeconomic groups [3]. Nasal coloni- staphylococcal exfoliative toxin causes bullous impetigo.
zation can be an important source of reinfection with Impetigo most often affects children 2–5 years of age,
Staph. aureus. Colonization with Proteobacteria and although it can occur in any age group. Impetigo is the
Corynebacterium spp. seems to be protective against Staph. most common bacterial skin infection among children
aureus colonization [4]. Eradication of Staph. aureus coloni- and is more common in those receiving dialysis. The
zation is challenging because the throat, perineum and diagnosis usually is made clinically and can be confirmed
nares can all be colonized. In one study the throat was the by Gram stain and culture. The infection usually heals
most common site colonized followed by perineum and without scarring and can resolve within several weeks
then nares [5]. In addition to humans, pets can be colo- even if left untreated [2].
nized and can become a source of reinfection. Dogs were Impetigo usually is transmitted through direct contact.
found to be most commonly colonized in their mouth [6]. In many cases there is antecedent cutaneous trauma, such
Between 1998 and 2004, 44.9% of non‐intensive care unit as an insect bite or minor scratch. Bacterial colonization of
Staph. aureus strains were meticillin resistant. Furthermore, the nares, axilla or perineum may serve as a reservoir for
the percentage of meticillin resistance among patients in infection. Impetigo is more common in tropical climates
intensive care units infected with Staph. aureus was 59.5% and in crowded living conditions, and can be associated
in 2003, an 11% increase in resistance from 1998 to 2002. with poor hygiene. Certain dermatoses, most notably
Meticillin resistance has also spread to the outpatient set- atopic dermatitis, are associated with higher Staph. aureus
ting, and between 1998 and 2004, 25% of Staph. aureus colonization rates, leading to frequent bacterial superin-
infections were meticillin resistant [7]. There has also been fection. Outbreaks of impetigo may also occur in the new-
an increase in resistance to topical therapies. There is born nursery setting.
geographical variability in resistance patterns but one Nonbullous impetigo begins as an erythematous mac-
paediatric study showed that Staph. aureus resistance ule or papule, which can progress to pustules and ero-
rates to mupirocin and retapamulin were 9.8% and 9.5% sions. Serous and purulent drainage forms a characteristic
respectively [8]. honey‐coloured crust. Any serum that extrudes to the sur-
face and dries can appear honey coloured. Therefore the
SECTION 7: BACTERIAL SKIN

References serous drainage from exuberant contact dermatitis such


1 Manders S. Toxin‐mediated streptococcal and staphylococcal disease. as from urushiol (poison ivy or oak) is often mistaken for
J Am Acad Dermatol 1998;39:383–98. impetigo. Individual lesions of impetigo can enlarge to 1
2 Steer A, Danchin M, Carapetis J. Group A streptococcal infections in
or 2 cm, but satellite lesions typically appear in the vicin-
INFECTIONS

children. J Pediatr Child Health 2007;43:209–13.


3 Gorwitz R. A review of community‐associated methicillin resistant ity due to spread by autoinoculation. Coalescence of these
Staphylococcus aureus skin and soft tissue infections. Pediatr Infect lesions may produce wider areas of crusted involvement.
Dis J 2008;27:1–7.
The face, especially around the mouth and nares, and the
4 Johnson RC, Ellis MW, Lanier JB et  al. Correlation between nasal
microbiome composition and remote purulent skin and soft tissue perineum are the most common sites. Recently, a strain of
infections. Infect Immun 2015;83:802–11. enterovirus (coxsackie A6) has been recognized to cause a
5 Kumar N, David MZ, Boyle‐Vavra S et al. High Staphylococcus aureus similar picture wherein widespread small vesicles with
colonization prevalence among patients with skin and soft tissue infec-
tions and controls in an urban emergency department. J Clin Microbiol
individual crusts can simulate impetigo. The eruption can
2015;53:810–15. be especially widespread when it superinfects pre‐
6 Iverson SA, Brazil AM, Ferguson JM et al. Anatomical patterns of colo- existing atopic dermatitis [3]. Although typically localized,
nization of pets with staphylococcal species in homes of people with widespread impetigo may occur, especially in the setting of
methicillin‐resistant Staphylococcus aureus (MRSA) skin or soft tissue
infection (SSTI). Vet Microbiol 2015;176:202–8. underlying atopic dermatitis or immunosuppression.
7 Kil E, Heymann W, Weinberg J. Methicillin‐resistant Staphylococcus Itching and mild discomfort may occur with impetigo, but
aureus: an update for the dermatologist. Cutis 2008;81:227–32, 247–52. systemic complaints are rare.
8 McNeil JC, Hulten KG, Kaplan SL, Mason EO. Decreased suscepti-
bilities to retapamulin, mupirocin, and chlorhexidine among
Bullous impetigo is mainly caused by toxin‐producing
Staphylococcus aureus isolates causing skin and soft tissue infections phage group II Staph. aureus. It is a localized form of SSSS,
in otherwise healthy children. Antimicrob Agents Chemother with subcorneal epidermolysis caused by one of the two
2014;58:2878–83. staphylococcal exfoliative toxins. These toxins are local-
ized to the area of infection and Staph. aureus can be cul-
tured from the blister contents (unlike the denuded skin
Localized cutaneous staphylococcal
and erosions in generalized SSSS). Superficial vesicles
and streptococcal infections:
progress to rapidly enlarging, flaccid bullae with sharp
pyodermas
margins. Ruptured bullae are replaced with yellow crust-
Impetigo ing. The peripheral peeling at the perimeter of the crust
Pyoderma refers to a localized purulent infection of the (caused by desmoglein‐1 cleavage) is very helpful in mak-
skin and soft tissues. Impetigo is a common type of super- ing the clinical diagnosis. Bullous impetigo is most fre-
ficial pyoderma that is characterized by inflammation and quently seen in infants and favours moist intertriginous
infection localized in the epidermis. Nonbullous impetigo areas such as the perineum, axillae and neck folds. It
Chapter 37  Pyodermas and Bacterial Toxin‐mediated Syndromes 427

appears to be less contagious than nonbullous impetigo, The clinical lesions of folliculitis begin as small pustules
and cases usually are sporadic. in follicular orifices, often accompanied by a halo of
Impetigo has an excellent prognosis. The infection may ­perifollicular erythema. Pustules may rupture followed
be self‐limited, but it can spread and persist on the skin, by crust formation at the follicular opening. The papulo-
remaining contagious to others. Acute poststreptococcal pustular eruption is often regionalized, and lesions are
glomerulonephritis is an uncommon but serious compli- typically located on the trunk, buttocks and extremities.
cation of nonbullous impetigo. Treatment with antibiotics Pruritus is the most common symptom of folliculitis.
does not seem to have any effect on this risk. Other rare Although prominent excoriations are usually not seen,
but potential complications include sepsis, osteomyelitis, children may excoriate each pustule leaving behind only
arthritis, endocarditis, pneumonia, cellulitis, lymphade- papules and crusts. Systemic signs and symptoms are rare.
nitis, guttate psoriasis, Henoch–Schönlein purpura, ery- The histopathology of folliculitis reveals neutrophilic
thema nodosum, TSS and SSSS [2]. pustules within the hair follicle. A perifollicular infiltrate
Treatment of impetigo is appropriate to achieve a composed of lymphocytes and histiocytes may also be
faster resolution of infection, prevent more serious com- present. Folliculitis may be acute or chronic with a ten-
plications and limit the spread of infection to others. dency for recurrence. The prognosis for superficial folli-
Patients with localized disease may be treated with topi- culitis is excellent, and the process may be self‐limited.
cal antiseptics, mupirocin, bacitracin and retapamulin. Deeper forms of folliculitis can result in dermal scarring
Oral antibiotics are recommended for more extensive and alopecia. Although uncommon, folliculitis can lead to
disease. Given the relative prevalence of staphylococcal cellulitis or lymphadenitis. Rarely, folliculitis caused by
impetigo, non‐β‐lactamase penicillins such as amoxicillin toxigenic staphylococcal strains can produce TSS.
and penicillin VK are not good choices for oral therapy In normal hosts, folliculitis may resolve spontaneously
unless the impetigo is culture proven to be exclusively without systemic treatment. Good hygiene is the best way
due to GAS. The most appropriate oral antibiotics for the to prevent this type of infection. For superficial and lim-
majority of cases of impetigo include penicillinase‐ ited folliculitis, topical antiseptics (e.g. chlorhexidine) or
resistant penicillins, cephalosporins or clindamycin. topical preparations of erythromycin or clindamycin are
Staphylococcal resistance is increasing so cultures and usually effective. Mupirocin ointment is also effective, but
sensitivities should dictate therapy, especially in immu- the rate of mupirocin resistance is increasing in proportion
nosuppressed patients or those with recalcitrant, widely to its use in mild cutaneous infections which has a nega-
disseminated disease. tive impact on its important role in elimination of nasal

SECTION 7: BACTERIAL SKIN


Ecthyma is considered to be an ulcerated form of MRSA carriage. Widespread or deep inflammatory follicu-
impetigo, and can be a consequence of failure to effec- litis usually requires oral antistaphylococcal antibiotics.
tively treat impetigo. The infection extends into the
­dermis to produce shallow ulcerations that may heal Furunculosis

INFECTIONS
with scarring. It is mainly caused by GAS. Because GAS Deep folliculitis with extensive perifollicular inflammation
can lead to small vesicles, pustules and erosions, it can and abscess formation produces a furuncle (boil). Isolated
simulate herpes simplex infections, especially in patients furuncles are common, and the term ‘furunculosis’ refers to
with atopic dermatitis. Treatment for ecthyma usually a condition with multiple, recurrent furuncles. Furuncles
requires systemic antibiotics such as β‐lactamase‐resistant are inflammatory nodules associated with a hair follicle
penicillins or cephalosporins in order to cover for the that extend into the dermis and the subcutaneous tissue.
possibility of coinfection with Staph. aureus. Systemic They usually affect moist, hairy, friction‐prone areas of
therapy is recommended because ecthyma is more the body, such as the face, axilla, neck and buttocks. These
­commonly seen in patients with underlying immuno- lesions are firm and tender, and may spontaneously
deficiency or widespread atopic dermatitis as well as drain purulent material. Fever and other constitutional
in  those living under poor hygienic conditions. GAS symptoms are rarely present. The most common causa-
ecthyma may also be an underrecognized source of tive microorganism is Staph. aureus.
rheumatic heart disease [4]. When the subcutaneous infection extends to involve
multiple furuncles, the lesions are called carbuncles. This
Folliculitis multiseptate coalescence can be painful, and constitu-
Bacterial folliculitis is a specialized form of pyoderma in tional signs and symptoms, including fever and malaise,
which the infection is anatomically confined to the hair are often present. Severe complications, such as bacterial
follicle and perifollicular structures. By far the most com- endocarditis, have been reported. If systemic involve-
mon bacterial pathogen in cases of childhood folliculitis ment of any type is suspected, evaluation should include
is  Staph. aureus. Superficial staphylococcal folliculitis is blood cultures as well as Gram stain and culture of puru-
very common and is also known as Bockhart impetigo. lent material [5].
Predisposing conditions that increase the number of skin Smaller furuncles can be treated with the application of
surface bacteria, such as occlusion, overhydration and warm, moist towels. Incision and drainage is usually
maceration, may lead to the development of folliculitis. required for larger furuncles and carbuncles. In addition,
Similar to impetigo, folliculitis is more common in tropi- the increased incidence of MRSA dictates a consideration
cal climates and in settings characterized by crowded of empirical coverage of MRSA in these infections in addi-
­living conditions and poor hygiene. tion to incision and drainage. Antibiotic choices include
428 Section 7  Bacterial Skin Infections

clindamycin, doxycycline (in children older than 9 years) report of erysipelas complicating a microcystic lymphatic
and trimethoprim–sulfamethoxazole (TMP‐SMX). Patients malformation [7].
who have recurrent furuncles may require eradication Prompt, aggressive treatment is indicated for erysipe-
of Staph. aureus from the nares, axilla and perineum with las, and parenteral antibiotics are generally preferred for
antibacterial cleansing agents such as chlorhexidine in initial management. Once stabilized, patients can be
addition to mupirocin ointment or oral clindamycin to treated with oral therapy. Penicillin remains the treatment
decrease risk for future infection. of choice for erysipelas, but numerous alternatives are
available, including the β‐lactamase‐resistant penicillins
Cellulitis and erysipelas and cephalosporins.
Cellulitis is an infection of the dermis and subcutaneous
fat that typically manifests as warm, tender, poorly Botryomycosis
demarcated areas of erythema. Occasionally, bullae and Botryomycosis is a chronic suppurative granulomatous
even necrosis may be present. Constitutional symptoms bacterial infection of the skin and subcutaneous tissue. It
may include malaise, fever and chills. In adults, cellulitis presents with clinical and histological features similar to
is often caused by Staph. aureus or Strep. pyogenes and is actinomycosis or mycetoma, although the causative
typically located on the lower extremities. In paediatric organism is usually Staph. aureus. The bacteria become
patients, cellulitis is most often caused by Staph. aureus encapsulated, which not only protects them from systemic
and frequently affects the face and neck, although other antibiotics but also prevents proliferation to a full patho-
areas can be involved. Multiorganism cellulitis with logical state. The bacterial inoculum usually remains low
anaerobes and Gram‐negative bacteria tends to occur in enough that abscesses do not develop but a chronic granu-
patients with chronic ulcers secondary to diabetes, venous lomatous reaction develops with sinuses, fistulas and
insufficiency or pressure. ulceration. Small yellow granules may be noted in the exu-
Small breaks in the skin, as can occur with minor date, and the causative bacteria are embedded within
trauma, injection drug use, body piercing or bites, serve these granules. The extremities are most commonly
as portals of entry for bacteria. Tinea pedis is a common affected, while head and neck involvement is unusual [8].
fungal infection that predisposes to bacterial cellulitis. The term ‘botryomycosis’ came from the Greek botrys,
Haematogenous spread of bacteria from other sites to the meaning bunch of grapes, because of the characteristic
skin occurs most commonly in the immunosuppressed groups of granules that resemble grapes, and mycosis,
patient. Lymphadenitis, subacute bacterial endocarditis because the origin of the disease was first thought to be a
SECTION 7: BACTERIAL SKIN

and glomerulonephritis are potential, but uncommon, fungus. However, botryomycosis is mainly caused by
complications of cellulitis. In most cases, the clinical pres- Staph. aureus, with Pseudomonas aeruginosa ranking second
entation is sufficient for an accurate diagnosis. Blood cul- in frequency. Most cases are reported in hospitalized
tures and cultures of bullae or ulcers should be obtained patients with concomitant diseases. The major associated
INFECTIONS

but may not always yield the pathogen. Imaging is not predisposing factors are skin trauma, previous surgery,
usually necessary, but can be helpful in distinguishing diabetes mellitus, liver disorders, systemic steroids, alco-
cellulitis from more severe infections such as necrotizing holism and cystic fibrosis.
fasciitis [6]. If necrotizing fasciitis is suspected, emergent Whenever botryomycosis is suspected, a Gram stain
surgical consultation is warranted. should be used to look for bacterial masses. Two cultures
Treatment in immunocompetent patients involves oral should be done: one for fungi and one for bacteria. A
antibiotic therapy targeting Staph. aureus and Strep. pyogenes. biopsy should be taken for histopathological examination
Options include cephalexin, clindamycin and dicloxacillin and to determine the shape and staining characteristics of
(for Staph. aureus). In those who are more severely ill, initial the granule, which differentiate it from actinomycosis
intravenous antibiotics are indicated, such as cefazolin, and mycetoma [9]. Histopathological findings of botryo-
clindamycin, oxacillin or vancomycin [5]. Gram‐negative mycosis include eosinophilic granules in a suppurative
bacteria and opportunistic organisms should be consid- focus. The centre of the granules is basophilic, but the
ered in immunocompromised individuals and people periphery is eosinophilic. They are usually 1–3 mm and
with diabetes. When multiorganism cellulitis is suspected, stain with periodic acid–Schiff (PAS), Gram and Giemsa
broad‐spectrum antibiotics should be instituted [6]. stains. A chronic inflammatory response composed of
Erysipelas is a distinctive superficial variant of cellulitis, neutrophils, lymphocytes, eosinophils, plasma cells,
characterized by striking erythema and a firm, well fibroblasts and histiocytes is present [8].
defined, advancing border. It is primarily caused by GAS. Treatment of botryomycosis traditionally involves a
Infants, young children and older adults are most com- combination of antibiotic and surgical therapy. Antibiotic
monly affected by erysipelas. Fever, chills, nausea and treatment should be prolonged, usually for weeks, and
vomiting are frequent, and may be associated with bac- based on the causal agent isolated. Surgical excision and
teraemia. Although most common on the lower legs, drainage of lesions can speed resolution.
­erysipelas may occur in any location. It is particularly
dangerous on the face due to the risk of septic sinus Blastomycosis‐like pyoderma
thrombosis. Recurrent erysipelas most frequently occurs Blastomycosis‐like pyoderma is a rare chronic pyoderma
in lymphoedematous extremities. Disrupted lymphatics that presents as verrucous plaques with multiple draining
put patients at higher risk of erysipelas; there is a recent sinuses, and heals with a cribriform scar [10]. Typical
Chapter 37  Pyodermas and Bacterial Toxin‐mediated Syndromes 429

vegetating skin lesions are difficult to distinguish tissue. Early surgical intervention for fasciotomy and
­clinically from blastomycosis. Staph. aureus is the most debridement, along with an aspirate for Gram stain
common cause but β‐haemolytic streptococci, Pseudomonas and  culture, is usually indicated. Antibiotic choices
spp., Proteus mirabilis and E. coli have also been reported should cover the variety of potential causative organisms,
[11]. Blastomycosis‐like pyoderma probably represents unless a definite bacterium can be isolated. The mortality
an  excessive tissue reaction in the context of altered rate for NF ranges from 35% to 40%; however, prompt
immunity. It has been reported in patients with systemic intervention lowers the rate to 12% [14].
immunosuppression due to malnutrition, alcoholism,
lymphoma, chronic myeloid leukaemia and human Blistering distal dactylitis
immunodeficiency virus (HIV) [10]. The initial lesion Blistering distal dactylitis (BDD) is an infection mani-
­usually begins at sites of trauma, and presents as large fested by acral pustules and bullae on erythematous bases
verrucous plaques with multiple pustules and elevated and, classically, as an individual bulla on the distal finger.
borders. Histology shows pseudoepitheliomatous hyper- This helps differentiate it from herpetic whitlow which is
plasia and multiple abscesses [11]. Cultures and biopsy usually a multiloculated honeycomb blister. BDD typi-
may be helpful in distinguishing blastomycosis‐like cally occurs on the proximal phalanx, palm or sole, and
pyoderma from blastomycosis. can present as multiple individual bullae. The bullae are
Treatment for blastomycosis‐like pyoderma includes typically oval and 1–3 cm in diameter and evolve into ero-
topical and systemic antibiotics, intralesional corticoster- sions when they denude. A culture of the blister fluid is
oids, surgical excision and local ablative measures such diagnostic. It most commonly occurs in infants and chil-
as  electrodesiccation and curettage or carbon dioxide dren; cases in adults have mainly been reported in immu-
laser. There are also several case reports of response to nosuppressed patients and those with diabetes.
acitretin [10]. GAS is the most frequently reported cause of BDD,
although occasional cases are linked to Staph. aureus, espe-
Necrotizing fasciitis cially if multiple bullae or erosions are present. BDD is
Necrotizing fasciitis (NF) is an aggressive soft tissue infec- associated with very low morbidity. When it is suspected,
tion involving the subcutis and fascia, with a rapid and ful- treatment is with incision, drainage and culture of bullae
minant progression. NF in adults is frequently polymicrobial, and a course of β‐lactamase‐resistant antibiotics [15].
but in children it is usually a monomicrobial infection most
commonly caused by GAS. Because of a significant mortal- Perianal streptococcal dermatitis

SECTION 7: BACTERIAL SKIN


ity rate, early diagnosis and initiation of aggressive surgical Perianal streptococcal dermatitis is an infection that pre-
and supportive therapy are critical for survival. Any suspi- dominantly affects younger children and is most fre-
cion of NF warrants immediate hospitalization, surgical quently caused by GAS. It has a male preponderance, and
consultation and critical care support [12]. the typical age group is between 6 months and 10 years.

INFECTIONS
Predisposing conditions for the acquisition of NF The signs and symptoms of PSD include rectal pain, pru-
include trauma, previous surgery, diabetes mellitus, ritus ani, painful defaecation, rectal bleeding and enco-
immunosuppression, renal failure, arteriosclerosis, odon- presis. The cutaneous findings in acute infections are
togenic infection and malignancy. In infants, the risk fac- patchy or plaque‐like, sharply demarcated, moist ery-
tors include omphalitis, mastitis, fetal scalp monitors, thema in a uniform perianal distribution extending
mammitis and necrotizing enterocolitis [13]. 2–4 cm from the anal verge. Satellite pustules may also be
Although NF usually involves an extremity, it can often observed. Perianal bacterial cultures can confirm the
involve the trunk in children. Localized painful erythema diagnosis. Staphylococcal perianal dermatitis can occur,
and oedema rapidly progress over hours to days, with but is far less common than streptococcal perianal derma-
development of cyanosis, blistering and necrosis. titis. Perianal streptococcal dermatitis is an underrecog-
Untreated, NF may progress to deep gangrene and nized cause of flares of guttate psoriasis.
sloughing of tissue. Systemic signs and symptoms include Oral therapy with penicillin V is considered first‐line
high fever, anxiety, altered mental status, tachypnoea, therapy. Alternatives include cefalexin, erythromycin
tachycardia and hypocalcaemia [14]. Vesiculation, ecchy- or  other macrolides. A duration of therapy from 14 to
mosis, crepitus, anaesthesia and necrosis are indicative of 21  days is recommended. Additional topical antiseptics
advanced disease. Severe pain out of proportion to skin (e.g. chlorhexidine) may accelerate bacterial clearance [16].
findings, rapidly spreading oedema, bullae formation,
mental status changes, marked leucocytosis or elevated Streptococcal intertrigo
creatinine kinase level all suggest NF more than cellulitis. Intertrigo is caused by friction of opposing skin surfaces
Anaesthesia of overlying skin may be present prior to the in a moist environment. Young infants are especially sus-
appearance of skin necrosis [12]. ceptible because of their abundant skinfolds. Secondary
In cases of suspected NF, an MRI scan of the affected infection with Candida albicans is common, but GAS must
extremity can help in diagnosis but should not delay sur- also be considered. A sharply demarcated, intensely ery-
gical consultation if there is high suspicion. MRI typically thematous, weeping eruption in intertriginous areas is
shows widespread soft tissue oedema and subfascial gas characteristic of both candidal and GAS intertrigo. Certain
formation. Frozen tissue sections show massive polymor- clinical features can help to differentiate these two condi-
phonuclear infiltrate in the fascia and subcutaneous tions. The appearance of satellite lesions surrounding the
430 Section 7  Bacterial Skin Infections

main eruption favours Candida infection, whereas the In all of these cases, the source location and dose of
presence of pain, bright red colour and a foul odour sug- toxin have been shown to directly influence the clinical
gests GAS intertrigo. GAS can occur in multiple skinfolds response. In addition, host factors such as local pH, glu-
including the neck, axillae, inguinal folds and popliteal cose level, oxygen level, age, and presence or absence of
fossae simultaneously. Patients with GAS intertrigo may antibodies will have a direct impact on the clinical expres-
also be irritable or have low‐grade fevers. Bacterial cul- sion of toxin‐mediated illness. Certain physical signs are
ture can establish the diagnosis. frequently present in toxin‐mediated illness. These
Simple intertrigo will respond to measures that mini- include strawberry tongue, acral erythema with desqua-
mize moisture and reduce friction, such as drying of the mation, and an erythematous eruption with frequent per-
skinfolds, barrier creams or zinc pastes. Candidal inter- ineal accentuation. These signs have been described in
trigo can be treated with topical anti‐yeast medications TSS, scarlet fever and recurrent perineal erythema, among
such as ketoconazole or nystatin. For GAS intertrigo, a others. The frequent clinical overlap between toxin‐medi-
10‐day course of penicillin successfully eliminates the ated diseases has been attributed to the significant degree
infection in most instances. Recurrences may occur and of sequence homology not only between toxins produced
require retreatment [17]. by the same bacterial strain, but also due to the similari-
ties at a molecular level between many streptococcal and
References staphylococcal toxins. Ultimately, though, the phenotypic
1 Steer A, Danchin M, Carapetis J. Group A streptococcal infections in expression of a given toxin‐mediated disease is depend-
children. J Pediatr Child Health 2007;43:209–13.
ent not only on the toxin itself, but also on host factors [1].
2 Cole C, Gazewood J. Diagnosis and treatment of impetigo. Am Fam
Physician 2007;75:859–64.
3 Mathes EF, Oza V, Frieden IJ et al. “Eczema coxsackium” and unusual Staphylococcal scalded skin syndrome
cutaneous findings in an enterovirus outbreak. Pediatrics 2013;132: SSSS is a potentially life‐threatening, toxin‐mediated
e149–57.
4 Parks T, Smeesters PR, Steer AC. Streptococcal skin infection and
manifestation of localized infection with certain strains of
rheumatic heart disease. Curr Opin Infect Dis 2012;25:145–53. staphylococci. The syndrome is characterized by bright
5 Lopez F, Lartchenko S. Skin and soft tissue infections. Infect Dis Clin red, tender skin, especially periorificial and in the neck,
North Am 2006;20:759–72. axillary and inguinal folds, that denudes with lateral
6 Kroshinsky D, Grossman M, Fox L. Approach to the patient with pre-
sumed cellulitis. Semin Cutan Med Surg 2007;26:168–78. pressure (Nikolsky sign). In more severe cases, fever,
7 Neri I, Montanari F, Baraldi C et al. Erysipelas as a superinfection of tachycardia and hypotension may be present.
an oral lymphangioma. J Pediatr 2014;165:205. SSSS is predominantly a disease of infancy and early
8 Ellerbe D, Parsons D, Cook P. Botryomycosis: improved therapy for a
SECTION 7: BACTERIAL SKIN

childhood, with most cases seen before the age of 5 years.


difficult infection. Int J Pediatr Otorhinolaryngol 1997;41:363–9.
9 Bonifaz A, Carrasco E. Botryomycosis. Int J Dermatol 1996;35:381–8. This age distribution may be due to renal immaturity
10 Nguyen R, Beardmore G. Blastomycosis‐like pyoderma: successful leading to decreased clearance of the toxin or lack of pro-
treatment with low‐dose acitretin. Australas J Dermatol 2005; tective antibodies against the toxin. Adults with SSSS
INFECTIONS

46:97–100.
11 Sawalka S, Phiske M, Jerajani H. Blastomycosis‐like pyoderma. Indian
have rarely been reported; predisposing factors in this
J Dermatol Venereol Leprol 2007;73:117–19. population include renal failure, malignancy, immuno-
12 Bingol‐Kologlu M, Yildiz R, Alper B et  al. Necrotizing fasciitis suppression, chronic alcohol abuse or HIV infection.
in  ­children: diagnostic and therapeutic aspects. J Pediatr Surg Most toxigenic strains of Staph. aureus causing SSSS
2007;42:1892–7.
13 Hsieh WS, Yang PH, Chao HC, Lai JY. Neonatal necrotizing fasciitis: belong to phage group II, types 71 and 55. SSSS results
a  report of three cases and review of the literature. Pediatrics from the effects of one of the exfoliative (also known as
1999;103:e53. epidermolytic) toxins (ETs). As noted previously, bullous
14 Manders S. Toxin‐mediated streptococcal and staphylococcal disease.
J Am Acad Dermatol 1998;39:383–98.
impetigo results when toxin is produced and exerts its
15 Scheinfeld N. Is blistering distal dactylitis a variant of bullous impe- effect locally. In generalized forms of SSSS, the toxin dif-
tigo? Clin Exp Dermatol 2007;32:314–16. fuses from an infected focus and, in the absence of specific
16 Jongen J, Eberstein A, Peleikas H et al. Perianal streptococcal dermati- antitoxin antibody, spreads haematogenously to produce
tis: an important differential diagnosis in pediatric patients. Dis Colon
Rectum 2008;51:584–7. its widespread effects. In children the infectious focus is
17 Honig P, Frieden I, Kim H, Yan A. Streptococcal intertrigo: an usually in the nasopharynx or conjunctivae; therefore, if a
underrecognized condition in children. Pediatrics 2003;112:1427–9. primary source of infection cannot be found, the nares
and rectum should be cultured to establish antibiotic sus-
ceptibilities. An appropriate antibody response to ET
Toxin‐mediated staphylococcal
appears to limit clinical disease expression to bullous
and streptococcal disease
impetigo; an inadequate humoral immune response may
Children commonly present with exanthems. Although predispose patients to development of SSSS.
viral exanthems account for the majority, staphylococcal Childhood cases of SSSS are primarily associated with
and streptococcal toxin‐mediated diseases are important solely ETA production, but the frequency of the different
disorders to identify. Early recognition and treatment of toxins in adult cases is unclear [1]. Both ETs produce blis-
these syndromes can significantly reduce the morbidity tering and denudation by disruption of the epidermal
and potential mortality. In this section, staphylococcal granular cell layer. ETs have been shown to be glutamate‐
scalded skin syndrome (SSSS), staphylococcal TSS, strep- specific serine proteases that specifically bind and cleave
tococcal TSS, streptococcal scarlet fever and recurrent desmoglein‐1. Desmogleins are cadherin‐type cell–cell
toxin‐mediated erythema will be discussed. adhesion molecules found in desmosomes, and play a
Chapter 37  Pyodermas and Bacterial Toxin‐mediated Syndromes 431

critical role in maintaining epithelial integrity. Desmogleins which occurs in a variety of clinical settings and has a ten-
are also affected in the autoimmune blistering disease dency to recur [5]. Currently, the incidence of nonmen-
pemphigus. Desmoglein‐1 is targeted by IgG autoanti- strual TSS exceeds that of menstrual TSS. Most cases of
bodies in pemphigus foliaceus, which shows superficial nonmenstrual TSS occur in the postoperative setting but
epidermal blisters with identical histological findings TSS has also been described in association with influenza,
to SSSS [2]. sinusitis, tracheitis, intravenous drug use, HIV infection,
Clinical features of SSSS include fever, irritability, skin cellulitis, burn wounds, allergic contact dermatitis, gynae-
tenderness and scarlatiniform erythema with accentua- cological infection and the postpartum period.
tion in flexural areas. Within 24–48 hours, flaccid blisters In both settings, TSS occurs when infection with toxin‐
and erosions develop. These blisters are caused by the producing strains of Staph. aureus induces a systemic
toxin and thus are sterile. The Nikolsky sign is character- inflammatory response in those lacking protective
istically present. Mucosal lesions do not occur because antibodies.
desmoglein‐3 is compensatory in the mucosa and not tar- TSST‐1 is one of the significant mediators of patho-
geted by the ETs. However, involvement of the kerati- genicity in TSS, and it is generated by strains from virtu-
nized external lip with crusting and fissuring is frequently ally all menstrual and many nonmenstrual cases of TSS.
encountered. The lack of mucosal involvement is the However, nonmenstrual cases of TSS can be due to strains
main differentiator from Stevens–Johnson syndrome of Staph. aureus that do not produce TSST‐1 but instead
(SJS). SJS is also typically caused by medicines and a produce staphylococcal enterotoxins. TSST‐1 and staphy-
rare disease in infancy. In contrast to SSSS, SJS produces lococcal enterotoxin B act as superantigens, and can lead
full‐thickness epidermal necrosis and histologically to large‐scale activation of T cells as well as massive
­demonstrates subepidermal separation, rather than the cytokine production.
intraepidermal split characteristic of SSSS. Clinically, both menstrual and nonmenstrual TSS have
Sepsis is rare in children but common in (immunocom- similar features. Fever, rash, desquamation, hypotension
promised) adults. and multiple organ involvement are the hallmarks of both
Therapy for SSSS should be directed towards eradica- variants. The eruption of TSS is defined as ‘diffuse macu-
tion of staphylococci from the focus of infection, which lar erythroderma’; however, a scarlatiniform eruption,
generally involves intravenous penicillinase‐resistant often with flexural accentuation, is frequently present.
penicillins. Usually, oral antibiotic therapy can be substi- Erythema and oedema of palms and soles, hyperaemia of
tuted within several days. In addition to antibiotics, topi- conjunctiva and mucous membranes, and strawberry

SECTION 7: BACTERIAL SKIN


cal supportive skin care and appropriate attention to fluid tongue are often noted. Desquamation of the palms and
and electrolyte management in the face of disrupted bar- soles, as seen in many bacterial toxin‐mediated disorders,
rier function will usually ensure rapid recovery. The mor- usually follows the onset of the illness by 1–2 weeks.
tality rate is low in children (3%) but exceeds 50% in Importantly, in nonmenstrual TSS caused by a postopera-

INFECTIONS
adults, especially in the setting of immunocompromise tive infection, the classic signs of localized infection such
[1]. Recent studies have documented that SSSS is more as erythema, tenderness and purulence may be absent
commonly caused by meticillin‐susceptible Staph. aureus from the site of infection, thereby making clinical diagno-
than would be predicted based on local antibiotic sensi- sis challenging. Multiple organ involvement may include
tivities. In addition, a higher percentage of clindamycin the gastrointestinal, muscular, renal, hepatic, haemato-
resistance was found. Therefore, if clindamycin is initi- logical or central nervous systems [1].
ated in order to decrease toxin production, a penicillinase‐ Intense conjunctival hyperaemia is a very frequent and
resistant penicillin or cephalosporin should be used to characteristic finding in TSS, but it also occurs in Kawasaki
provide double coverage until susceptibilities can be disease, erythema multiforme, Rocky Mountain spotted
established [3,4]. fever and adenoviral and enteroviral infections. These
It is vital to recognize the potential for epidemic SSSS in diseases, along with streptococcal toxic shock syndrome
newborn nurseries, and identification of healthcare work- (STSS) and early SSSS, constitute the differential diagno-
ers who are colonized or infected with toxigenic Staph. sis of TSS. In children, the distinction between Kawasaki
aureus is an integral part of management. Control meas- disease and TSS can be very difficult, especially early in
ures should be applied, including strict enforcement of the course of illness. The characteristic thrombocytosis
chlorhexidine handwashing, oral antibiotic therapy for and lymphadenopathy of Kawasaki disease are the most
infected workers, and mupirocin ointment for eradication helpful distinguishing features, but may not always be
of persistent nasal carriage. present.
Prompt intervention is the key to the successful man-
Staphylococcal toxic shock syndrome agement of the multiorgan involvement in TSS. Removal
Toxic shock syndrome (TSS) is defined as an acute and of infected foreign bodies, drainage of infected sites and
potentially fatal illness characterized by high fever, a dif- institution of penicillinase‐resistant antistaphylococcal
fuse erythematous rash, hypotension, involvement of antibiotics are essential to eradicate the focus of toxin‐
three or more organ systems and desquamation of the producing organisms. Massive volume replacement may
skin 1–2 weeks after onset. TSS is usually classified into be needed in the setting of severe intravascular volume
two categories: menstrual TSS, originally described in depletion. Cardiovascular support may be necessary,
association with tampon use; and nonmenstrual TSS, including both inotropic and antiarrhythmic measures.
432 Section 7  Bacterial Skin Infections

Paediatric patients may require ventilatory support for more than 50% of patients with STSS, as compared with
respiratory distress more often than adult patients. less than 15% in TSS. In addition, mortality rates are more
Metabolic acidosis, hypomagnesaemia, hypocalcaemia than five times higher in STSS.
and hypophosphataemia may accompany renal disease, Management of STSS is similar to that of TSS.
requiring aggressive monitoring and management. Supportive therapy, vasopressors and antibiotics are the
cornerstones of treatment. The increasingly reported clin-
Streptococcal toxic shock syndrome ical resistance of streptococci to penicillin, as well as the
In the late 1980s, a disease similar in appearance to TSS, difficulty in distinguishing STSS from TSS in some cases,
yet caused by invasive streptococci, was recognized. This suggests the need for adequate antimicrobial coverage for
streptococcal toxic shock syndrome (STSS) was found to both staphylococci and penicillin‐resistant streptococci.
share many clinical features with staphylococcal TSS, Consideration should be given to clindamycin, erythro-
including multiorgan involvement, toxicity, conjunctival mycin, cephalosporins or other agents as deemed appro-
hyperaemia and profound hypotension. In the majority of priate by clinical presentation and culture results [1].
cases, toxin‐producing group A streptococci have been Surgical drainage, debridement, fasciotomy or amputa-
isolated, with SPE‐A production being most closely linked tion may be necessary.
with invasive disease. However, group A streptococci
producing SPE‐B, SPE‐C, streptococcal superantigen and Streptococcal scarlet fever
mitogenic factor, as well as non‐group A streptococci, Scarlet fever is caused by pyrogenic exotoxin‐producing
have been found to be causative in individual cases of group A β‐haemolytic streptococcal infections. Epidemics
STSS as well. In a similar manner to classic TSS, the clini- of scarlet fever occurred in the first half of the twentieth
cal signs of STSS are postulated to be mediated by mas- century, but it is no longer the major public health threat
sive cytokine release as a result of toxin/superantigen that it was in the past. Morbidity, mortality and sequelae
activity. In addition, streptolysin O, produced by 100% of are much less serious today, not only because of the
streptococcal strains associated with STSS, has been dem- development of antibiotics, but also because of changes
onstrated to act synergistically with SPE‐A [1]. in the streptococci responsible for the majority of cases of
The majority of cases of STSS have occurred in young, scarlet fever. Whereas the bacteria that caused scarlet
otherwise healthy persons between 20 and 50 years of fever in the past primarily produced the more virulent
age. Most are associated with invasive streptococcal infec- SPE‐A toxin, currently SPE‐B and SPE‐C are produced by
tions with virulent (i.e. M types 1 and 3) strains causing most Strep. pyogenes organisms isolated from patients
SECTION 7: BACTERIAL SKIN

extensive soft tissue infections and leading to bacterae- with scarlet fever.
mia. (In contrast with STSS, staphylococcal TSS typically Scarlet fever remains primarily a disease of children,
is associated with occult or minor focal infections.) The with most cases occurring between the ages of 1 and 10
factors that determine whether infections with SPE‐A years. SPEs have been shown to elicit the cutaneous
INFECTIONS

lead to STSS or classic scarlet fever are uncertain. The ­manifestations of scarlet fever by enhancing delayed‐
host’s immune status for the infecting strain’s M type and type hypersensitivity to streptococcal products, thereby
toxin type is clearly important. It appears that lack of requiring previous exposure for expression of disease.
immunity to both streptococcal M type and toxin is This explains the rarity of cases of scarlet fever in infancy:
required for both scarlet fever and STSS. most infants have not had previous exposure to these
Clinically, STSS shares many features with TSS. Fever, streptococcal toxins and therefore have not generated
hypotension, myalgias, liver abnormalities, diarrhoea, antitoxin antibody. Clinical findings include the abrupt
emesis, renal dysfunction and haematological abnormali- onset of fever, sore throat, headache and chills.
ties may be present in TSS caused by either staphylococci Mucocutaneous findings include a finely papular erythe-
or streptococci. Diffuse macular erythroderma likewise matous ‘sandpaper’ rash on the trunk and extremities,
is frequently present in disease caused by both bacteria, with circumoral pallor. Pastia’s lines represent linear
and is often accompanied by mucous membrane findings, petechial streaks found especially in flexural locations
such as conjunctival injection and delayed desquamation such as the antecubital fossae, the axillae and the ingui-
of palms and soles. Nonetheless, certain important dif- nal region. Erythema and oedema of the pharyngotonsil-
ferences exist between STSS and TSS. The skin is often lar area, punctate erythematous and petechial macules
the  portal of entry in STSS, with soft tissue infections on the palate, and strawberry tongue are commonly pre-
developing in 80% of patients. The initial presentation of sent. Large, thick sheets of skin may desquamate from
STSS is often localized pain in an extremity, which rapidly the hands and feet, especially in the convalescent phase.
progresses over 48–72 hours to manifest both local and Uncommon complications include pneumonia, pericar-
systemic signs of STSS. Cutaneous signs may include ditis, meningitis, hepatitis, glomerulonephritis and rheu-
localized oedema and erythema, a bullous and haemor- matic fever. Recurrence rates of scarlet fever have been
rhagic cellulitis, necrotizing fasciitis or myositis, and gan- reported to be as high as 18%.
grene. Soft tissue involvement of this nature is distinctly Diagnosis of scarlet fever is usually apparent clinically,
uncommon in staphylococcal TSS. STSS may uncom- but can be further confirmed by supportive serologies
monly occur in the absence of cutaneous involvement; and isolation of group A streptococci from the pharynx.
in  these cases differentiation from staphylococcal TSS First‐line treatment is with penicillin; cephalosporins,
becomes more difficult. Blood cultures are positive in erythromycin and other macrolides are alternatives [1].
Chapter 37  Pyodermas and Bacterial Toxin‐mediated Syndromes 433

Recurrent toxin‐mediated (perineal) bacterial pharyngitis, isolated episodes of toxin‐mediated


erythema erythema without recurrences, and patients with episodic
Recurrent toxin‐mediated perineal erythema (RPE) is mild hypotension, fever and typical mucocutaneous find-
caused by the action of toxins produced by both staphylo- ings in the absence of full criteria for TSS have been
cocci and streptococci. Due to a homology at the molecu- described. The common feature, aside from a large degree
lar level between the toxins produced by these bacteria, of clinical overlap, has been the repeated ability to isolate
there is a substantial clinical overlap. Furthermore, a sin- toxin‐producing bacteria from normally sterile sites.
gle bacterial toxin acting as a superantigen can lead to a Given the clinical variations, it is more appropriate to
broad spectrum of clinical diseases. Testing for these tox- refer to this group of entities as ‘toxin‐mediated erythema’
ins in cases of RPE has revealed streptococcal pyrogenic although most cases are recurrent with a propensity for
exotoxins A and B as well as TSST‐1 [6]. perineal involvement [1].
The hallmark of RPE is a striking diffuse macular ery-
References
thema in the perineum occurring within 24–48 hours 1 Manders S. Toxin‐mediated streptococcal and staphylococcal disease.
after a bacterial pharyngitis. Oral mucosal changes, such J Am Acad Dermatol 1998;39:383–98.
as strawberry tongue, as well as erythema, oedema and 2 Anzai H. Production of low titers of anti‐desmoglein 1 IgG autoanti-
bodies in some patients with staphylococcal scalded skin syndrome.
convalescent desquamation of the hands and feet are
­
J Invest Dermatol 2006;126:2139–41.
­usually present as well. Systemic signs such as fever or 3 Li MY, Hua Y, Wei GH, Qiu L. Staphylococcal scalded skin syndrome
hypotension are absent; however, diarrhoea may occur. in neonates: an 8‐year retrospective study in a single institution.
Recurrences are frequent, occurring as many as 40 times. Pediatr Dermatol 2014;31:43–7.
4 Braunstein I, Wanat KA, Abuabara K et al. Antibiotic sensitivity and
Culture of the pharynx during the acute episodes reveals resistance patterns in pediatric staphylococcal scalded skin syn-
toxin‐producing Staph. aureus or Strep. pyogenes. drome. Pediatr Dermatol 2014;31:305–8.
It has become clear that RPE is part of an expanding 5 Iwatsuki K, Yamasaki O, Morizane S, Oono T. Staphylococcal cutane-
clinical spectrum of toxin‐mediated skin diseases that do ous infections: invasion, evasion, and aggression. J Dermatol Sci
2006;42:203–14.
not easily fit into previously described clinical entities. 6 Patrizi A, Raone B, Savoia F et al. Recurrent toxin‐mediated perineal
Recurrent erythroderma associated with a preceding erythema. Arch Dermatol 2008;144:239–43.

SECTION 7: BACTERIAL SKIN


INFECTIONS
434 

CHA PTER  3 8

Cutaneous Manifestations of 


Gram‐negative Infections
Saul N. Faust1, Diane Gbesemete1 & Robert S. Heyderman2
 NIHR Southampton Clinical Research Facility, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
1

 University College London, London, UK


2

Meningococcal disease, 434 Gram‐negative soft tissue Gram‐negative folliculitis, 451


Pseudomonal skin disease, 446 infections, 450

Abstract Neisseria meningitidis is a common human nasopharyngeal


commensal. In a minority of individuals it causes invasive, life‐­
threatening disease. The cutaneous features are often recognizable
Gram‐negative infections are a rare but potentially life‐threatening
at presentation and may cause significant morbidity.
cause of cutaneous lesions, often requiring urgent recognition and
Pseudomonas aeruginosa can cause both localized primary skin
treatment. This chapter discusses the pathophysiology, clinical
lesions and systemic infection with cutaneous manifestations.
features and management of the most common Gram‐negative
Pseudomonal skin infections are more commonly, but not exclu-
infections.
sively, seen in immunocompromised individuals.

Key points • Early diagnosis, early antibiotics and good supportive care are
crucial in the management of meningococcal disease.
Meningococcal disease Pseudomonal skin disease
• Neisseria meningitidis, a small encapsulated Gram‐negative • Pseudomonas aeruginosa is a Gram‐negative aerobic rod that is
SECTION 7: BACTERIAL SKIN

diplococcus, is the aetiological agent of meningococcal disease. widely distributed in the environment, particularly in aqueous
• Asymptomatic nasopharyngeal carriage is common with a peak environments.
in carriage in adolescence. • It is a common opportunistic pathogen that is responsible for
• Transmission is predominantly via respiratory droplets shed significant morbidity and mortality in immunocompromised
INFECTIONS

from asymptomatic close contacts. hosts but can also cause disease in previously fit and well
• Neisseria meningitidis causes invasive disease in a minority of individuals with a normally functioning immune system.
colonized individuals, usually soon after acquisition. • It has intrinsic resistance to many first‐line antibiotics and
• Most infections are due to serogroups A, B or C. often develops resistance to broad‐spectrum or second‐line
• Vaccines are available for serogroups A, C, W‐135, Y and, more antibiotics.
recently, serogroup B. • Skin manifestations of pseudomonal infection can be localized
• Meningococcal disease remains a major health problem worldwide primary infections or secondary manifestations of pseudomonal
and can occur as sporadic cases, outbreaks or epidemics. septicaemia.
• Although meningococcal disease can occur at any age, children • Primary, localized pseudomonal skin infection can occur in
under 5 years of age are predominantly affected with a second immunocompetent individuals who have a disruption in the
smaller peak in incidence amongst adolescents. normal skin defences against infection.
• Mortality from meningococcal disease is approximately 5–10% and • Pseudomonas septicaemia is strongly associated with
there are long‐term sequelae in up to 25% of those who survive. immunocompromise and has a very high mortality.
• The skin rash is characteristically petechial but may blanch and • Ecthyma gangrenosum is the characteristic cutaneous lesion
resemble a viral exanthem in the early stages of infection. associated with pseudomonal sepsis.

Meningococcal disease
and mortality [2,5]. Meningococcaemia is characterized
Introduction. Neisseria meningitidis is one the most by the development of a widespread purpuric rash and
­common causes of severe infection in childhood [1–3]. the progression to vascular collapse and death in up to
First described by Vieusseux [4] in 1806 following an out- 20% of cases. In his description of the rapid onset and ful-
break of epidemic meningitis in Geneva, meningococcal minant progression of meningococcal disease, Herrick [6]
disease remains an important cause of both morbidity remarked that ‘no other infection so quickly slays’. Nearly

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 435

100 years later, improved awareness, the availability of by inhibitory flora, and mucosal and systemic immunity
highly active antibiotics and advances in intensive care [22–29]. No increased risk of invasive meningococcal dis-
management have failed to alleviate our fear of meningo- ease is associated with human immunodeficiency virus
coccal infection. (HIV) infection [30,31].

Aetiology. The aetiological agent N. meningitidis is a small Epidemiology. Meningococcal disease remains a major
Gram‐negative diplococcus that is surrounded by an health problem worldwide although there is substantial
outer polysaccharide capsule, an outer membrane and an geographical variation in incidence. It is endemic in most
underlying peptidoglycan layer. The cell wall is rich in countries but occurs in small outbreaks and epidemics in
lipopolysaccharide (LPS) which, together with outer both developing and industrialized countries [3,5,14].
membrane proteins, is released as vesicles or ‘blebs’. The incidence and predominant strains seen in particu-
These blebs are thought to facilitate immune avoidance lar geographical locations change significantly over time
by binding antibodies that would otherwise bind to with cyclical epidemics occurring in some regions. Part of
whole organisms and also mediate endotoxic shock. the reason for this dynamic epidemiology is thought to be
N.  meningitidis is an obligate human commensal which antigenic variability with the organism capable of capsular
causes disease in a minority of those who carry the switching and other noncapsular antigenic changes [14].
organism [7,8]. The ‘meningitis belt’ of sub‐Saharan Africa has the
Different strains of N. meningitidis may be distinguished highest incidence with large epidemics occurring every
by their serogroups, which are based on the 13 recognized 5–10 years [32–35], affecting up to 1% of the population at
variations in capsular polysaccharide antigens (A, B, C, D, their peaks [14]. There is seasonal variation in incidence,
E, H, I, K, L, W135, X, Y, Z) [8,9]. Worldwide, most infec- rising dramatically towards the end of the dry season and
tions are due to organisms of serogroup A, B or C, falling with the onset of the rains [3,32]. Serogroup A is
although Y, W135 and X are becoming more prevalent predominant although serogroups C, Y and W135 and
[8,10–12]. N. meningitidis may be further serotyped, sero- more recently X are also seen. The incidence in North
subtyped and immunotyped on the basis of outer mem- America is currently unusually low with a predominance
brane protein and LPS expression. The bacterial genome of serogroups B and C although Y and W135 are also seen.
can be characterized using multilocus sequence typing European countries and Australasia have a predominance
(MLST) [13]. These tools are often used in conjunction of serogroup B, especially following the introduction of
when analysing the epidemic spread of disease [14]. routine vaccination against serogroup C [14]. In temper-

SECTION 7: BACTERIAL SKIN


ate climates a seasonal peak in incidence can be seen in
Meningococcal carriage the first quarter of the year [3].
Transmission of N. meningitidis is predominantly via res- Although meningococcal disease can occur at any age,
piratory droplets shed from asymptomatic close contacts. children under 5 years of age are predominantly affected,

INFECTIONS
This may result in transient acquisition, prolonged car- with the highest attack rates being seen in those less than
riage in the nasopharynx (colonization) or invasive dis- 1 year old. In many countries, a second smaller peak in
ease [8]. A recent meta‐analysis looking at nasopharyngeal incidence has also been observed among teenagers aged
carriage in European countries showed that the preva- 15–19 years [3,36].
lence of carriage increased from 4.5% in infants, with a At the beginning of the twentieth century when the
sharp rise in adolescence, peaking at 23.7% at 19 years of only treatment available was a crude horse serum, the
age before gradually decreasing again throughout adult- mortality among patients with meningococcal septicae-
hood [15]. Nasopharyngeal carriage results in the produc- mia was often over 90% [5,37]. With the introduction of
tion of antibodies [7]. sulfonamides and penicillin in the late 1930s and 1940s,
there was a dramatic reduction in this mortality in coun-
Invasion tries around the world [5]. Until the mid 1990s the mortal-
In a minority of cases N. meningitidis penetrates the ity associated with meningococcal infection remained
mucosal layer, reaches the bloodstream and causes inva- unchanged at around 10% overall, rising to between 40%
sive disease. This usually happens in the first 2 weeks fol- and 70% in those patients in severe shock [5,37]. More
lowing acquisition [8] and is rare following prolonged recently, in industrialized countries where early disease
carriage. A number of hypervirulent strains are responsi- recognition and aggressive intensive care have become
ble for the majority of invasive disease, with the presence common, the mortality even in children presenting in
of a capsule being a particularly important virulence fac- shock has decreased to between 5% and 10% [2,38]. Up to
tor [14]. Other bacterial properties that might influence a quarter of those who survive an episode of meningococ-
invasion include the presence of pili, outer membrane cal disease may have significant ongoing sequelae [39].
proteins and immunoglobulin (Ig) A1 proteases [16–19].
The most important host factor conferring protection Pathophysiology. Between 1947 and 1976, the cutaneous
from invasive disease is the presence of serum bacteri- manifestations of fulminant meningococcal septicaemia
cidal activity [20,21]. Host factors implicated in the devel- were investigated in a series of postmortem and skin
opment of invasive disease include poor socioeconomic biopsy studies [40–46]. Although the appearances
circumstances, overcrowding, secretor status, smoking, described probably represent the final common pathway
respiratory tract infection, nasopharyngeal colonization for a number of possible pathogenic mechanisms, a wide
436 Section 7  Bacterial Skin Infections

Endothelial Vasoconstriction
injury

Collagen tPA
exposure

Serotonin ADP aPC


nϕ thromboxane A2 PS TM
GAG nϕ
nϕ TAFI
loss Platelet PAI Haemostatic
release nϕ tPA
reaction, plug
aggregation
and fusion
Platelet
TF Plasmin
phospholipid
mϕ mϕ AT
TFPI
a
Xa a
TFPI Xa Fibrin
Va Xa Vllla Thrombin Vllli
a Xa X Va a
IXa Vi
X IXa AT PS
TF Xa Xa ll aPC
nϕ E
PC E aPC P
TM P C
TM C R
THROMBIN R
generation by Protein S circulates
SELECTINS amplification in a free form (the
tether monocytes and initiated by active inhibitor) or
platelets(attached to tissue factor Activation bound to C4bBP
neutrophils) to endothelium via Xa
to provide further surface Inhibition or
area for complex formation
a = activated i = inactivated
SECTION 7: BACTERIAL SKIN

Fig. 38.1  The molecular pathogenesis of disseminated intravascular coagulation of bacterial septicaemia. ADP, adenosine 5’‐diphosphate; aPC, activated
protein C; AT, antithrombin; EPCR, endothelial protein C receptor; GAG, glycosaminoglycan; PAI, plasminogen receptor inhibitor; PC, protein C; PS, protein
S; TAFI, thrombin activatable fibrinolysis inhibitor; TF, tissue factor; TFPI, tissue factor pathway inhibitor; TM, thrombomodulin; tPA, tissue plasminogen
INFECTIONS

activator.

spectrum of histological changes were observed. These A haemostatic imbalance occurs following the activa-
ranged from fibrin–platelet thrombi, endothelial swelling tion of coagulation and dysfunction or dysregulation
and necrosis, and granulocytic infiltrates to occlusion of of  the natural regulatory pathways (Fig.  38.2) [52–54].
blood vessels, extravasation of red blood cells and exten- Endothelial prostacyclin production and the expression
sive full‐thickness necrosis. In recent years, our under- of anticoagulant glycosaminoglycans may be impaired
standing of the mechanisms underlying meningococcal [59,60]. Patients with meningococcal disease have
sepsis has improved. It has become apparent that much of increased monocyte tissue factor activity [61], decreased
the disease process that follows meningococcal invasion is tissue factor pathway inhibitor [62], a deficiency of
not due to direct toxicity by the meningococcus itself but antithrombin, protein C and protein S, and significant dis-
results from the release of endotoxin into the circulation. ruption of the fibrinolytic pathway [50,57,63–68]. We have
This triggers the activation of inflammatory cells and demonstrated that disruption of the endothelial activa-
the  elaboration of a wide range of cytokines and other tion of protein C in meningococcal sepsis is caused by
mediators [47–52]. This intense inflammatory response downregulation of thrombomodulin and endothelial pro-
is  commonly associated with platelet consumption, a tein C receptor on the endothelial cell surface (Figs 38.3
coagulopathy and intravascular thrombosis, which are and 38.4) [57].
all important markers of disease progression and a poor This dysregulation of the coagulation and fibrinolysis
prognosis [53]. However, the coagulopathy and the micro- pathways occurs together with activation of inflamma-
vascular consequences of meningococcal sepsis are more tory pathways including innate, cell‐mediated and
complex than originally thought (Fig. 38.1) [54]. With the humoral immunological processes, involving the comple-
appreciation that the vessel wall is not simply an inert con- ment cascade, the kallikrein–bradykinin system, mono-
duit [55] but performs a wide range of important homeo- cytes, neutrophils and cytokines (Fig. 38.5) [52,54,69–73].
static functions, it has become recognized that dysfunction The pathways are linked at many levels [52], together
of the vascular endothelium is key to the development leading to the capillary leak, defects in vascular tone,
and progression of both the haemostatic and inflamma- hypoperfusion, myocardial dysfunction and focal throm-
tory disorders associated with severe sepsis [54,56–58]. bosis that are responsible for the tissue damage
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 437

TFPI
PAI
Plasma
AT PC, PS

Fig. 38.2  The coagulation and fibrinolytic Production Plasmin


AT function
pathways rely on the function of endothelial APC
proteins and protein complexes. Dysfunctional
regulatory mechanisms in meningococcal disease
include endothelial (protein C pathway) and
plasma (tissue factor pathway inhibitor [TFPI],
antithrombin, plasminogen receptor inhibitor tPA
PGl2 GAGs
[PAI]) factors. AT, antithrombin; CS, chondroitin Endothelial CS, HS, DS PC
sulphate; DS, dermatan sulphate; EPCR, surface TM, EPCR
endothelial protein C receptor; GAGs,
glycosaminoglycans; HS, heparan sulphate; PC,
protein C; PGI2, prostacyclin; PS, protein S; TM,
thrombomodulin; tPA, tissue plasminogen
activator.

remains uncertain. Large numbers of meningococci can


(a) be detected in the dermal vessels of purpuric skin lesions
[40,43,46,74] and may not only be directly toxic but act as
a focus for both the inflammatory and haemostatic
responses on the vessel wall [73,75–77]. Endothelial colo-
nization has recently been demonstrated in an experi-
mental model of human skin grafted onto immunodeficient
mice. This adherence was mediated by type 4 pili and
resulted in the pathological features of thrombosis, vessel

SECTION 7: BACTERIAL SKIN


occlusion, red blood cell extravasation and tissue necrosis
[78–80]. We have identified meningococci in a number of
different cellular environments expressing key virulence
factors (Fig. 38.6) [74].

INFECTIONS
While many single‐gene defects are known to affect the
immune and coagulation responses to infection [81], a
combination of genetic predisposition and bacterial
(b) (c)
virulence factors is likely to be responsible for any one
individual’s susceptibility to invasive disease. In menin-
gococcal disease, many such single‐nucleotide polymor-
phisms have now been identified. Genetic variants of
mannose‐binding lectin might account for nearly one
third of cases of invasive disease [82] and an innate anti‐
inflammatory cytokine profile contributes to the likeli-
hood of death [83]. A specific Fc‐γ receptor polymorphism
may be essential in protecting against fulminant menin-
Fig. 38.3  Skin biopsy specimen from a patient with meningococcal sepsis.
A biopsy specimen from a purpuric lesion shows areas containing
gococcal infection [84]. Although the factor V Leiden
thrombosed vessels (right‐hand arrow in (a) and arrow in (b)) and a mutation confers a specific genetic predisposition to
perivascular infiltrate (left‐hand arrow in (a)) (haematoxylin and eosin, thrombotic disease [85], it has not been shown to be asso-
×100 (a) and ×400 (b)). (c) shows inflammatory cells (arrow) around ciated with increased mortality in meningococcaemia,
unthrombosed vessels (haematoxylin and eosin, ×400). The cellular although patients with this mutation may be at higher
infiltrate consisted of both neutrophils (identified by neutrophil‐elastase risk of thrombotic complications such as amputations and
staining) and monocytes and macrophages (identified by CD68 staining).
skin grafting [86]. An increased PAI‐1 response to tumour
necrosis factor (TNF)‐α has been associated with death in
commonly observed in association with fulminant menin- meningococcal septicaemia [87], demonstrated to be due
gococcal sepsis [52]. to a polymorphism in the PAI‐1 gene [67,68].
The release of meningococcal lipopolysaccharide has
been understood for some time to be an important trigger Clinical features. Meningococcal meningitis is the most
for the inflammatory and haemostatic processes seen in common form of the disease, is indistinguishable from
fulminant disease [8,49]. However, the importance of other forms of childhood bacterial meningitis, usually
direct bacterial invasion of the skin by the organism responds rapidly to antibiotics and is associated with
438 Section 7  Bacterial Skin Infections

(a) (b)

(c) (d)
SECTION 7: BACTERIAL SKIN
INFECTIONS

(e)

Fig. 38.4  Thrombomodulin immunostaining (immunoperoxidase stain, ×200) of skin biopsy specimens from a patient with acute meningococcal sepsis
during the initial infection (a) and 3 months later (b) is shown. The same sequence is shown for a second patient (c, d). (e) Another initial specimen from
the second patient. The arrows in (a) and (e) show unthrombosed vessels with reduced thrombomodulin staining, and the arrow in (c) shows a thrombosed
vessel with reduced thrombomodulin staining. Partial recovery of thrombomodulin expression (arrows in (b) and (d)), together with some residual
inflammatory infiltrate, is seen in both patients after 3 months.

little long‐term morbidity [88]. Meningococcal septicae- 80–90% of patients [90]. The rash usually occurs within
mia with shock is more devastating, commonly present- 12–36 hours of the onset of the disease and is characteris-
ing with nonspecific symptoms of fever, vomiting, tically petechial but may blanch early in the course of the
headache, abdominal pain and muscle aches. The rash of infection. In the classic presentation of meningococcal
meningococcal septicaemia may be very subtle in the septicaemia, the petechiae are irregular, small and often
early stages (Fig. 38.7) and its recognition is imperative if have raised centres (Fig.  38.8). Lesions commonly occur
this life‐threatening condition is to be diagnosed early on the limbs and trunk but may be found on the head,
and treated effectively. palms, soles and mucous membranes. They may also
In the early stages of meningococcal infection the rash occur on areas subjected to friction or under pressure
may resemble a viral exanthem such as rubella (see by clothing.
Fig. 38.7). This erythematous maculopapular rash is non- Progression of the rash of meningococcal septicaemia
purpuric, nonpruritic and often very transient [40,89]. has been shown to relate to the severity of the coagu-
Following a diligent search of all areas of the body, the lopathy and prognosis [45,53,91–94]. As the disease
purpuric lesions of meningococcal septicaemia are seen in becomes more fulminant, lesions coalesce to form large
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 439

Sepsis usually considered contraindicated when the clinical


diagnosis is meningococcal septicaemia or is often
n delayed when a characteristic rash suggests meningococ-
io

In
at cal meningitis, as it is frequently associated with clinical

fl
l
gu

am
a deterioration and death [2,96]. Nasopharyngeal swabs

m
Co

at
may provide evidence of meningococcal infection. Biopsy

io
n
material and aspirates from the characteristic purpuric
skin lesions of meningococcal disease provide a rich
Endothelial
injury source of organisms and have been advocated by some
workers for rapid diagnosis, but this is not usually con-
Organ sidered routine owing to the advent of modern molecular
Death
failure
techniques now available [95,97–101].
An array of rapid tests is now available to detect
N. meningitidis group A, B, C, Y and W135 antigens from
Fibrinolysis blood, CSF and urine, which may establish the diagnosis
although the sensitivity is poor [102,103]. The highly sen-
sitive polymerase chain reaction (PCR) of blood (or CSF
Fig. 38.5  The inflammatory, coagulation and fibrinolytic pathways are
linked at many levels, leading to organ failure and eventually death. where available) can make the diagnosis in an additional
60% of cases in which no positive culture has been obtained
[104–107]. The sensitivity of the new WB‐Taqman PCR on
ecchymoses and become haemorrhagic and bullous (see EDTA‐treated samples of whole blood has been demon-
Fig.  38.8). These areas of extensive skin involvement strated to be 87%, increasing case confirmation compared
may  progress to gangrene of the digits or whole limbs with previous PCR techniques from 72% to 94% [107].
(Fig. 38.9). In such severe purpura fulminans, the gangre-
nous regions are gunmetal grey or blue‐black and are Management of acute meningococcal septicaemia. The
well  demarcated [41]. The arterial pulses are usually management of meningococcal disease has been reviewed
intact but the capillary perfusion is very poor. Thrombosis, in detail elsewhere [2,90] and is detailed in Fig.  38.11.
extensive skin involvement, venous congestion and tissue However, a number of important management points
oedema may lead to the development of a limb compart- will be emphasized.

SECTION 7: BACTERIAL SKIN


ment syndrome (see Fig. 38.9). Severe tissue destruction may
be associated with muscle infarction and rhabdomyolysis. Recognition and initial therapy
The development of shock associated with meningo- It is crucial that, in the absence of another diagnosis, any
coccal septicaemia may be insidious, and is manifested febrile, ill child with a petechial rash should be considered

INFECTIONS
initially by poor peripheral perfusion, confusion and an to have meningococcal septicaemia, and treatment should
increased respiratory rate. Hypotension is a late sign as be commenced without awaiting further confirmation.
children and young adults may maintain their blood Although the majority of children may not appear desper-
pressure by vasoconstriction, despite severe hypovolae- ately ill when first seen, they may deteriorate suddenly
mia [2]. It is important to recognize meningococcal shock and therefore should be observed carefully during the first
in the early phases, well before hypoxia, acidosis and 48 hours, ideally on a paediatric intensive care unit.
hypovolaemia lead to multiorgan failure (Fig. 38.10). An
algorithm for the early recognition and management of Choice of antibiotic
meningococcal infections is given in Fig. 38.11. Penicillin remains the treatment of choice for meningo-
coccal sepsis. However, since other infections may also
Differential diagnosis. Bacteraemia with Haemophilus occasionally cause shock and a petechial rash, a third‐
influenzae, Streptococcus pneumoniae, Pseudomonas spp. and generation cephalosporin should be employed until the
overwhelming viral and fungal infections may all rarely diagnosis has been confirmed. Penicillin‐resistant strains
present with shock and purpuric rash (Fig. 38.12). Other of N. meningitidis have become a problem in Spain and
congenital and immune causes of a purpura fulminans may South Africa [108–110] but fortunately only a handful of
present with the characteristic rash but these patients are insensitive strains have been detected in the UK, the rest
rarely cardiovascularly compromised (see Chapter 146). of Europe and North America [111–118].

Laboratory diagnosis. Meningococci are very sensitive to Supportive care


chilling or drying and must be inoculated and cultured Children who present in meningococcal shock need
soon after collection. Cerebrospinal fluid (CSF) examina- immediate resuscitation, restoration of circulating vol-
tion and culture are likely to provide confirmation of the ume and adequate oxygenation [2]. In those patients with
diagnosis of meningococcal meningitis in about 80–90% concomitant meningitis, cerebral oedema and raised
of cases [95]. However, blood cultures are positive in only intracranial pressure may occur. Elective ventilation, con-
one third to one half of untreated patients, even when trol of the PCO2 and improvement of cardiac output and
accompanied by a characteristic rash or positive CSF thus cerebral perfusion are important aspects of treatment
microscopy and culture [95]. Lumbar puncture is now of such patients.
440 Section 7  Bacterial Skin Infections

(a) (b)
SECTION 7: BACTERIAL SKIN
INFECTIONS

(c)
Fig. 38.6  Skin biopsy specimen from a patient with meningococcal septicaemia. (a, b) Gram‐stained sections reveal meningococci associated with a
leucocyte (a, electronically enlarged) and within blood vessels (b, electronically enlarged). Figures were processed with Adobe Photoshop and illustrator
software. Arrows indicate Gram‐stained meningococci. (c) Immunohistochemical staining of N. meningitidis in a skin biopsy sample from a patient with
meningococcal disease showing PorA‐stained bacteria in the interstitium and blood vessel. V, blood vessel. Arrows indicate immunoperoxidase‐positive
staining of meningococci (brown) with the appropriate specific mouse monoclonal antimeningococcal antibody (nuclei were counterstained in
haematoxylin).

Management of the skin lesions ­ angrene is seen in a ‘glove and stocking’ distribution.


g
Although the majority of purpuric skin lesions heal well However, there is no clinical trial supporting its use and,
following the resolution of meningococcal sepsis, in ful- when used in large doses, this agent may cause further
minant disease it is important to prevent the extensive hypotension [54,59].
skin damage, limb loss and associated multiorgan failure Faced with a devastating illness, and the prospect of
that often occur despite prompt antibiotic therapy. limb loss and death in young children, many clinicians
Although there is no evidence from randomized con- have felt justified in attempting to utilize experimental
trolled trials, fresh frozen plasma (FFP) infusions are used treatments outside controlled trials. Purified inhibitors
to correct hypofibrinogenaemia and specific clotting such as antithrombin, fibronectin, protein C or variants
factor deficiency, and to reduce the risk of haemorrhage of  α1‐antitrypsin have been advocated by a number of
[54,119]. Cryoprecipitate may be used in patients where authors [63,120–126], as have blockers of the initial phases
hypofibrinogenaemia persists despite multiple FFP of coagulation [127–129]. Others have suggested fibrino-
­infusions. In addition, prostacyclin has been used to try lytic agents [130]. Heparin has been promoted in the past
to  reverse some vasoconstriction where impending as rational therapy for disseminated intravascular
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 441

[136] and recombinant bactericidal permeability‐increasing


protein [137]. Although previous trials of steroids in sepsis
have been disappointing [138,139] and a very old study
suggested a worse prognosis in meningococcaemia [140],
more recent data have renewed interest in low‐dose steroid
therapy [141]. A phase III trial of recombinant activated
protein C was initially shown to reduce the mortality in
adult sepsis [142], although a phase III trial in childhood
sepsis did not show similar benefit [143] and the drug has
since been withdrawn from the market globally because it
failed to show any survival benefit in subsequent adult
trials [144]. On the basis of small pharmacological series
using unactivated protein C concentrate, some have advo-
cated using this form of the drug [126,145,146]. However, it
is currently not possible to predict which patients who
have been administered protein C concentrate will be able
to activate the drug [57,145]. Even when activated protein
C is detectable in the plasma, there is evidence of an acti-
vation defect in the dermal vascular endothelium and a
theoretical risk that unactivated protein C in excess may
displace activated protein C at the endothelial protein C
receptor and thus worsen dermal vessel thrombosis [57,147].
Finally, although the use of recombinant tissue plasmi-
nogen activator (tPA) to treat severe meningococcaemia
has an apparently sound theoretical basis [65,67,68,87], in
a retrospective European study of 62 children with menin-
gococcal disease treated with tPA, 8% suffered serious
intracerebral bleeding and its use cannot now be recom-
(a) mended [148]. The adverse clinical experience demon-

SECTION 7: BACTERIAL SKIN


strated in evaluating tPA and the lack of benefit of most of
the other adjunctive therapies used to treat severe sepsis
strongly reinforce the need for new or experimental
agents to be introduced only after properly controlled

INFECTIONS
randomized clinical trials.

Surgical intervention
Small vessel thrombosis, vascular insufficiency and com-
partment syndrome can lead to peripheral ischaemia and
soft tissue necrosis, predominantly in the lower limbs [149].
In the acute phase, fasciotomy may be beneficial in
selected cases to reduce peripheral ischaemia caused by
increased pressure rather than small vessel thrombosis
[149–151] (Fig. 38.13).
(b)
As discussed more fully in Chapter 146, surgical inter-
Fig. 38.7  Early stages of meningococcal septicaemia. (a) Fine maculopapu- vention to remove necrotic skin or amputate limbs or dig-
lar rash with one early purpuric lesion on the forearm of a young infant.
its is generally not indicated during the acute phase of the
(b) Fine maculopapular rash with a few early purpuric lesions on the trunk
of a young child with septic shock.
disease but should be should be carried out once the lim-
its of tissue damage have been definitively established
[149,151]. During recovery, surgery to repair damaged
coagulation (DIC) [43,131,132]. However, the agent has skin and extremities may be required in up to 72% of
failed to produce obvious benefit in a limited number of patients [152]. These procedures range from skin grafting
open clinical trials [44,133,134] and has been associated and local debridement to microvascular flaps and ampu-
with uncontrolled bleeding. tations [152–154]. Multiple grafting procedures and scar
In recent years, understanding of the pathophysiology revision may also be required [153–156] (see Fig.  38.13).
of the coagulation and inflammatory abnormalities has The use of negative pressure wound therapy has been
led to a number of large randomized controlled trials of suggested to decrease the time from initial infection to
adjunctive agents in adult and childhood sepsis and in skin grafting, as well as reducing the invasiveness and
meningococcal disease. No survival or morbidity benefit frequency of dressing changes [149].
has been demonstrated for most of these agents, including Nutritional support and the avoidance of nosocomial
antithrombin [135], antiendotoxin monoclonal antibody wound infection are essential in the management of such
442 Section 7  Bacterial Skin Infections

(a)
SECTION 7: BACTERIAL SKIN

(b) (d)
INFECTIONS

(c)

(e)
Fig. 38.8  Purpuric lesions of meningococcal sepsis. (a) Purpuric lesions on trunk and limbs. (b) Close‐up of abdominal purpuric lesions. (c) Haemorrhagic
rash with central necrosis. (d) Severe purpura fulminans. (e) Bullous formation on a large ecchymotic area.

patients [153]. A new microsurgical technique has been Outcome of skin disease
proposed to benefit outcome, but this remains to be tested A large study from The Netherlands showed the inci-
in a larger study or reported from other centres [157]. dence of long‐term skin scarring to be 48% and of ortho-
Later surgical complications may arise several years paedic sequelae 14% in children surviving meningococcal
after the original infection. Ongoing orthopaedic follow‐ sepsis. Although the severity of these sequelae varied,
up may be required to allow early recognition and thus children with more severe scarring or who had suffered
optimal management of these late sequelae such as orthopaedic sequelae were demonstrated to have had
­physeal growth plate arrest with associated limb length more severe disease [159]. A group of UK children who
discrepancy, and joint malalignment [151,158]. had suffered amputation were studied from a daily living
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 443

(a)

SECTION 7: BACTERIAL SKIN


(b)

INFECTIONS
(d)

(e)
(c)
Fig. 38.9  Extensive cutaneous involvement in meningococcal sepsis. (a) Severe digital hypoperfusion in the early stages of meningococcal septicaemia.
(b) Digital hypoperfusion that has progressed to gangrene with demarcation 2 weeks after onset of the illness. (c) Gangrene over a proximal ecchymotic
area associated with dermal vascular thrombosis. (d) A child with septicaemic shock in the first 24 hours of the illness, showing purpura fulminans,
severe bilateral lower limb ischaemia and bilateral compartment syndromes. (e) Extensive gangrene with marked venous thrombosis of superficial and
deep vessels.
444 Section 7  Bacterial Skin Infections

to an intense mononuclear infiltration of the basal layer


with overlying atrophy. Meningococcal antigen, anti-
meningococcal antibody and complement (C3) have been
demonstrated in the skin and synovium of these patients.
Greenwood et al. [164] suggested that these skin lesions
occur as a result of Arthus‐like immune complex deposi-
tion. Prolonged antibiotic therapy offers no benefit to
patients with this allergic phenomenon and the use of
systemic corticosteroids has not been evaluated in con-
trolled trials.

Chronic meningococcaemia
In addition to meningitis and fulminant septicaemia,
occasional cases of meningococcal infection may present
as chronic meningococcaemia, pneumonia, arthritis or
ophthalmitis [165,166]. More common in teenagers and
adults [165], chronic meningococcaemia has become a
rare entity since the widespread use of antibiotics.
Untreated, the disease was often self‐limiting with a small
mortality associated with supervening meningitis or
endocarditis. Underlying immunological disorders, other
chronic diseases or specific bacterial virulence factors
have not been confirmed in the pathogenesis of this syn-
drome [165,167]. However, some have suggested that
chronic infection may be caused by less virulent menin-
gococci producing a milder host immune response [168].
Fig. 38.10  Severe meningococcal sepsis associated with a capillary leak Chronic meningococcaemia is an insidious disease,
syndrome and multiorgan failure. This child had widespread purpuric classically presenting with a low‐grade fever lasting for
lesions, poor peripheral perfusion and marked tissue oedema and required at least 1 week, flitting arthralgias or, less commonly,
SECTION 7: BACTERIAL SKIN

mechanical ventilation, inotropic support and peritoneal dialysis. arthritis [165]. The rash is recurrent and may be maculo-
papular, nodular or petechial. There is little systemic tox-
icity and no significant coagulopathy, and the presence of
functions and quality of life perspective for 3–5 years meningeal signs suggests that the disease has become
INFECTIONS

following their disease. Importantly, the degree of ampu- fulminant [165,166]. Histology shows a perivascular
tation did not predict the functional outcome, and most infiltration of lymphocytes, macrophages and a few neu-
children were reported to have effective compensation trophils. Vessel wall destruction has not been reported.
strategies with a generally good quality of life [160]. Meningococci are rarely detected in the cutaneous
However, others report considerable lasting effects on lesions. Multiple blood cultures are required for the diag-
health and health‐related quality of life 2 years following nosis of chronic meningococcaemia and may not be posi-
severe meningococcal sepsis [161]. tive until between 2 and 8 weeks into the illness. Chronic
meningococcaemia responds rapidly to systemic antibi-
Allergic complications otics and therefore should always be considered in the
Approximately 5 days after the presentation of acute differential diagnosis of vasculitic disorders, particularly
meningococcal disease, between 1.7% and 4.7% of patients Henoch–Schönlein purpura, acute rheumatic fever and
may develop a vasculitic rash that may be accompanied infective endocarditis [165,166].
by arthritis or episcleritis [162,163]. Typically, the rash
occurs either as a single lesion or as crops on the trunk, Prevention
lower limbs, deltoid areas and dorsum of the hand. Often Chemoprophylaxis
associated with persistence of fever, the lesions appear At present, recommendations are that household mem-
as  darkened skin with a blistered edge and are slightly bers and hospital workers who have come into close con-
swollen, warm and tender. These lesions progress to tact with secretions and the index case (unless already
­sterile bullae containing red cells and leucocytes which treated with ceftriaxone) should receive single‐dose cip-
ulcerate but usually heal quickly. A few patients develop rofloxacin [169] chemoprophylaxis [90,170–172]. In the
tender warm nodules similar to Osler’s nodes. UK, routine prophylaxis for day nursery contacts is not
Histologically, the dermal lesions appear oedematous recommended. It is important to recognize that eradica-
and consist of dilated small blood vessels with poly- tion of the meningococcus is not always successful and
morph and mononuclear infiltrates. Thrombosis, necrosis that recolonization of the nasopharynx may occur after
and haemorrhage may be seen in association with these initial clearance. Rifampicin can be used where ciproflox-
changes and typically organisms are neither seen  nor acin is contraindicated. Chemoprophylaxis should be
­cultured. The epidermal changes range from hyperkeratosis followed by vaccination where possible.
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections
445

Fig. 38.11  Early management of meningococcal disease in children. Source: Reproduced with permission from the Meningitis Research Foundation. © Meningitis Research Foundation (www.meningitis.org).

SECTION 7: BACTERIAL SKIN


INFECTIONS
446 Section 7  Bacterial Skin Infections

However, the immunological response to these polysac-


charides does not provide complete protection for all age
groups, and does not induce immunological memory
(immunity lasts for a maximum of 3 years). Following the
outstanding successes of the H. influenzae type b vaccine
[174,175] a conjugate vaccine against the group C menin-
gococcus has been introduced successfully in the UK and
in other European countries [176–180]. Quadrivalent con-
jugate ACW135Y vaccines have been licensed for use in
the USA and Europe [181].
The development of a vaccine for group B has been
more complicated because of its poorly immunogenic
capsule and potential cross‐reactivity [12,39]. Two vac-
cines have recently been developed. 4CMenB (Bexsero,
Fig. 38.12  Differential diagnosis of meningococcal sepsis. The hand of a GSK) is a four‐component vaccine licensed for use from
child with Streptococcus pneumoniae septicaemia. Although this infancy and is part of the national immunization schedule
presentation is rare, the confluent purpuric lesions, tissue oedema and in the UK. Trumenba (Pfizer) is a protein‐based vaccine
compartment syndrome are very similar to those seen in meningococcal now licensed in the USA and Europe for use in
septicaemia.
­adolescents and adults [20].

Pseudomonal skin disease


Introduction. Pseudomonas aeruginosa is a common oppor-
tunistic pathogen that is responsible for significant mor-
bidity and mortality in immunocompromised hosts,
particularly in those with underlying neoplastic disease.
It causes particular concern as it has intrinsic resistance to
many first‐line antibiotics and often develops resistance
to broad‐spectrum or second‐line antibiotics. The progno-
SECTION 7: BACTERIAL SKIN

sis of pseudomonal sepsis in an immunocompromised


individual is particularly poor.
The reported incidence of opportunistic pseudomonal
infection is increasing. This is because of increasing levels
INFECTIONS

of immunocompromise due to more invasive and


(a) immune‐modifying treatments and procedures, with
increased survival from illnesses affecting the immune
system [182]. Localized outbreaks have been seen in hos-
pitals. Infections in healthy hosts have also increased
due to changes in lifestyle such as the use of hot tubs and
contact lenses. The increased use of broad‐spectrum anti-
biotics has also allowed for the selection of this organism
due to its high levels of intrinsic and acquired antimi-
crobial resistance. A recent survey showed a dramatic
increase in the rate of antimicrobial resistance and among
isolates of P. aeruginosa from hospitalized children in the
USA between 1999 and 2012, with multidrug resistance
(more  than three classes) increasing from 15.4% to 26%
and carbapenemase resistance increasing from 9.4% to
20% over this time [183].
(b) Although P. aeruginosa is most commonly identified as
Fig. 38.13  Surgical intervention in meningococcal sepsis. (a) Fasciotomy a potential pathogen in the immunocompromised host, it
following the development of a compartment syndrome in a child with can also cause disease in previously fit and well individu-
purpura fulminans. (b) Extensive skin necrosis following meningococcal als with a normally functioning immune system.
septicaemia requiring multiple grafting procedures. P. aeruginosa can cause infections of many different
body systems although the most common site of infection
Vaccines is the lower respiratory tract. Other sites include the
Bivalent (A and C) and tetravalent (A, C, W135, Y) bloodstream, localized skin and soft tissue, ears and eyes.
­polysaccharide vaccines have been shown to be both Skin manifestations of pseudomonal infection can be as
safe and effective in containing small outbreaks and more localized primary infections or as secondary manifesta-
widespread epidemics of meningococcal disease [173]. tions of pseudomonal septicaemia. Skin presentations
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 447

­ athogen and frequent cause of nosocomial infections. It


p
has been found as a contaminant in hospital water sys-
tems and can easily be transferred on the hands of health-
care workers [184], allowing for easy spread within the
vulnerable population of hospitals. It is an important
cause of a variety of nosocomial infections including
bloodstream infections (the third most common Gram‐
negative pathogen grown in blood cultures) [186], hospi-
tal‐acquired pneumonia, ventilator‐associated pneumonia
and surgical site infections [187]. It is a common pathogen
in burn and wound infections – the most common Gram‐
negative pathogen isolated from infected burns [188].

Pathophysiology. P. aeruginosa is an opportunistic organ-


ism that rarely penetrates the first line of host defence of
intact skin or mucosal surfaces. It therefore rarely causes
Fig. 38.14  Ecthyma gangrenosum. Source: Dr John C. Browning, Baylor disease in the immunocompetent host. Once it has been
College of Medicine and Children’s Hospital of San Antonio, Texas, USA. allowed to gain entry it has a large variety of virulence fac-
tors that allow it to adhere to host tissues, avoid the host
vary greatly, ranging from the classical and more easily innate and acquired immune defences, become invasive
recognizable lesions of ecthyma gangrenosum (Fig. 38.14) and cause disease. The interplay of these virulence factors
to more subtle or localized infections that may imitate and the host defence systems allows colonization, local
fungal or other bacterial infections, potentially leading to infection or systemic infection to result (Table 38.1).
a delay in diagnosis and treatment. Colonization of host tissues is helped by the ability to sense
a biochemically favourable environment (chemotaxis) and
Aetiology. P. aeruginosa is the main human pathogen move towards it (motility). Organisms also need to be able
of  the family Pseudomonadaceae. It is a strictly aerobic to adhere to host surfaces. The main virulence factors that
Gram‐negative rod with a polar flagellum allowing P. aeruginosa uses for these processes are pili and flagella.
motility. It is nonfastidious in terms of its nutritional and Pili and flagella are complex bacterial cell surface struc-

SECTION 7: BACTERIAL SKIN


environmental requirements for growth, can be easily tures that project away from the cell surface [188].
­cultured on routine and selective media and will tolerate A key mechanism used by P. aeruginosa is the ability to
a wide range of temperatures. It is able to survive even grow in vivo primarily in biofilm rather than as single,
in distilled water hence it is often found in water‐related

INFECTIONS
environmental reservoirs [184].
Colonies of P. aeruginosa are irregular, fluoresce under Table 38.1  Pseudomonas aeruginosa – virulence factors
Wood’s light and have a characteristic metallic blue‐green [182,184,185,188,191–193]
appearance and grape‐like odour. A variety of pigments
are produced by Pseudomonas species. Pyoverdin is a Virulence factor Location Roles
green‐yellow pigment responsible for fluorescence
which is also produced by other species of Pseudomonas. Pili Cell surface Chemotaxis
Pyocyanin is only produced by P. aeruginosa and is respon- Motility
sible for the characteristic blue‐green colour of colonies Adhesion
Biofilm production
[182,184]. Other pigments include pyorubin, pyomelanin
Flagella Cell surface Chemotaxis
and trimethylamine, the last of which is responsible for Motility
the characteristic odour [182]. Biochemically, colonies Adhesion
produce a positive oxidase test and are unable to ferment Alginate Cell surface Adhesion
carbohydrates [184]. Biofilm formation
Colonization
Carriage. P. aeruginosa is not usually a commensal of Avoidance of host immune
response
healthy individuals although it can be found in the gas-
Lipopolysaccharide Cell surface Avoidance of host immune
trointestinal microbiome of a small proportion of the pop- response
ulation, particularly in hospitalized individuals [185]. It is Systemic inflammatory response
widely distributed in the environment, particularly in Type 3 secretion Cell surface Form protein bridge for injection
aqueous environments, and in humans it frequently colo- system of toxins into host cells
nizes areas of impaired host defence (e.g. burns) or the Type 3 secretion Injected into Host cell disruption and invasion
respiratory tract of ventilated individuals or those with system toxins host cell
Secreted enzymes Extracellular Avoidance of host immune
cystic fibrosis or bronchiectasis.
and toxins response
Invasion
Epidemiology. P. aeruginosa’s ability to survive and grow Pigments Extracellular Iron scavenging
is an important factor in its role as an opportunistic
448 Section 7  Bacterial Skin Infections

free‐floating ‘planktonic’ bacteria. Bacterial biofilms are persistently moist areas of skin, allowing the survival and
associated with specific bacterial phenotype and metabo- growth of the organism. Any local disruption in the integ-
lism associated with surface aggregation and the produc- rity of the skin can allow further invasion and disease.
tion of a specific matrix. Biofilms are more tolerant to Other pathogens can also cause similar localized skin
antibiotics and can evade host immune responses. infections which can cause diagnostic uncertainty and the
Chronic infections of the skin and wounds are due to bac- risk of initial empirical treatment not including cover
teria in biofilms [189,190]. against Pseudomonas.
Intact skin and mucosal membranes are the first line of Distinguishing characteristics of pseudomonal infec-
defence against potential pathogens and are usually suf- tion include the presence of a green exudate, which fluo-
ficient to prevent infection. P. aeruginosa is not a usual resces under Wood’s light, and a characteristic fruity
commensal of dry healthy skin although it may be found grape‐like odour [182]. Localized infection usually has a
in moist areas of skin in a minority of healthy individuals. good prognosis.
Colonization of the stratum corneum is prevented by the
lack of water, continual shedding and presence of antimi- Toe web infections
crobial lipids. These local host defence strategies can be Infection of the toe webspaces can occur due to recurrent
hindered by an area of skin being exposed to a persis- recreational or occupational immersion and inadequate
tently moist or humid environment, by frequent immer- drying, prolonged occlusion with non‐breathable foot-
sion in water or by occlusion by dressings, etc., allowing a wear, high humidity or hyperhidrosis [182]. Fungal infec-
build up of sweat. The normal skin commensals have a tions are the most common cause but P. aeruginosa can
role in the prevention of colonization by potential patho- also be a cause, usually as a bacterial superinfection
gens, and any change to the normal flora such as by the where there is underlying chronic yeast infection. This
use of antiseptics or antibiotics can allow colonization by causes erythema and erosion or maceration of the local
organisms that would otherwise be present only tran- area with pustules and hyperkeratosis with local
siently, including P. aeruginosa. This often results in local- ­discomfort or pain [184].
ized superficial infection. Any disruption to the continuity
of the skin barrier can allow organisms to colonize the Green nail syndrome
exposed surface and invade deeper structures. This can Pseudomonal infection of the soft tissues around nails can
occur with dermatitis and excoriation, ulceration, trauma, cause a chronic paronychia and/or onycholysis. The
burns, or following medical or surgical procedures. The accumulation of pyocyanin results in a characteristic dis-
SECTION 7: BACTERIAL SKIN

ubiquitous nature of P. aeruginosa and its ability to grow colouration of the nailfold, known as green nail syndrome
in nutritionally poor environments means that it is a fre- or chloronychia, which can help to distinguish pseu-
quent causative pathogen in wound infections, particu- domonal infection from the commoner fungal pathogens.
larly in burns or where poorly perfused or necrotic tissue Although characteristically green, it can be blue‐grey or
INFECTIONS

is present. Colonization of burned skin requires a tiny nearly black in colour and cannot be removed by wash-
inoculum of P. aeruginosa in comparison to the inoculum ing. Any underlying abnormality of the nail or nailbed
required to colonize healthy skin [194]. such as trauma or psoriatic change can predispose to
pseudomonal infection [182,184].
Antibiotic resistance Management of these conditions involves the avoid-
P. aeruginosa has inherent resistance to multiple antibiotic ance or removal of predisposing factors where possible,
classes and has the ability to quickly gain resistance to and on occasion may require topical antibiotic treatment
others. In 2013 the Centers for Disease Control (CDC) or nail removal [182].
classified multidrug‐resistant P. aeruginosa as a serious
threat with approximately 13% of severe pseudomonal Otitis externa
healthcare‐associated infections being resistant to multi- P. aeruginosa colonizes the external auditory canal of a
ple antibiotics, including aminoglycosides, cephalospor- small minority of healthy individuals but is commonly
ins, fluoroquinolones and carbapenems [195]. Biofilm present if there is a breakdown of the host defences
phenotypes specifically increase antibiotic tolerance and with  pre‐existing inflammation, trauma (for example
make chronic pseudomonal infections hard to treat with following ear piercings) or persistent moisture. Infection
conventional antibiotics [196]. results in tenderness, pain on movement, swelling and
a  purulent discharge [182,184]. Treatment includes
Clinical features. P. aeruginosa is associated with several removal of any debris in the external canal and topical
well‐defined skin manifestations ranging from a simple antibiotics [182].
localized skin infection to cutaneous manifestation of a Necrotizing or malignant otitis externa is a potentially
life‐threatening systemic infection. life‐threatening, invasive infection of the external ear pre-
senting with severe peri‐auricular tenderness, purulent
Primary skin infections discharge and localized lymphadenopathy. Bacterial
Primary, localized pseudomonal skin infection can occur invasion into nearby blood vessels and soft tissues can
in immunocompetent individuals who have a disruption lead to localized necrosis and can progress to mastoiditis,
in the normal skin defences against infection. This can cranial nerve abnormalities, osteomyelitis and sepsis
be due to prolonged exposure to contaminated water, or [182,184]. It is classically seen in elderly patients with
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 449

underlying diabetes mellitus but has also been described Pseudomonal sepsis can cause diffuse eruptions with a
in children with underlying immunodeficiency [197]. variety of lesions including macules, papules, vesicles,
Intravenous antibiotics and surgical debridement of petechiae or ecchymoses. Subcutaneous nodules occur
affected tissues are required [182]. rarely – these are deep fluid‐filled nodules that are often ten-
der and may spontaneously rupture, releasing fluid from
Folliculitis which the organism can be cultured [182]. Skin and soft tis-
Direct contact with water contaminated with P. aeruginosa sue infections such as cellulitis, gangrenous cellulitis and
can cause a characteristic folliculitis with discrete, large necrotizing fasciitis can be primary lesions caused by infec-
papules or pustules that are erythematous and pruritic or tion with P. aeruginosa or secondary to ­bacteraemia [182].
painful [182,184]. These lesions appear 8–48 hours after
immersion [198] and are distributed in areas of skin that Ecthyma gangrenosum
have been in contact with the water surface or at specific Ecthyma gangrenosum (Fig.  38.14) is the characteristic
areas of abrasion allowing inoculation of the organism, cutaneous lesion associated with, although not pathogno-
e.g. the plantar surface of the foot following abrasion on a monic for, pseudomonal sepsis. It is seen in approximately
pool floor (hot‐foot syndrome) [184]. Hot tubs and swim- 28% of those with Pseudomonas bacteraemia [191,204]. It
ming pools are frequent sources of infection hence the initially appears as an erythematous or purpuric macule
term ‘hot‐tub folliculitis’. P. aeruginosa is a frequent con- which evolves rapidly, over 12–24 hours, forming a blue‐
taminant of such water sources and is difficult to eradi- black haemorrhagic bulla or pustule that bursts, leaving
cate because it can withstand high temperatures and an ulcer with a necrotic centre, covered by a black scab,
chlorination [182]. Folliculitis often occurs in an otherwise and with a surrounding rim of erythema [182,191,202,205].
healthy host and usually self‐resolves within 7–10 days so These lesions can be found anywhere on the skin but
treatment with antibiotics is not usually indicated are particularly associated with moist areas such as the
[182,184,198]. There may be some systemic symptoms groin, axilla and perianal regions [182]. They almost
associated with infection such as fever and malaise but always result from haematogenous spread but can occa-
systemic spread does not usually occur in healthy hosts sionally occur in the absence of bacteraemia, presumably
[182]. Changes in local host defences (e.g. trauma, under- secondary to local inoculation.
lying skin disease or chronic tetracycline use) can increase There are case reports of previously healthy children
the risk of infection. Immunocompromised hosts are at presenting with pseudomonal sepsis and ecythma gan-
risk of progression to ecthyma gangrenosum, and treat- grenosum [202,206] and with ecythma gangrenosum as

SECTION 7: BACTERIAL SKIN


ment with systemic antibiotics may be considered [184]. an isolated lesion [207], although it is normally associated
Other organisms such as Aeromonas species have rarely with significant immunodeficiency. Apparently healthy
been implicated in cases of ‘hot‐tub folliculitis’ [199,200]. children should be investigated for potential underlying
Other forms of Gram‐negative folliculitis are discussed in immunodeficiency.

INFECTIONS
the following paragraphs. The histological features of ecthyma gangrenosum
include vasculitic changes in the dermis with bacterial
Omphalitis invasion of the adventitial and medial layers but not the
Infection of the umbilical stump in the neonatal period intima or lumen, without inflammatory changes or neu-
can be caused by a number of organisms, including trophil invasion. P. aeruginosa can be cultured from the
Pseudomonas spp., and is more commonly seen in associa- lesion itself and usually also from the blood [182].
tion with poor cord care and in settings with limited Although characteristically associated with P. aerugi-
resources. It can initially present with an area of erythema nosa, ecthyma gangrenosum has been reported in associa-
and tenderness around the umbilicus and a purulent dis- tion with infection with other species of Pseudomonas
charge but may progress to pustules on the surrounding [205], other Gram‐negative bacteria such as Escherichia
skin and then necrosis of the surrounding soft tissues. The coli, Aeromonas hydrophilia, Vibrio vulnificus, Klebsiella pneu-
direct entry of bacteria into the bloodstream will result in moniae, Citrobacteri freundii, Morganella morganii and
systemic infection and sepsis. Appropriate intravenous Stenotrophomonas maltophilia [182] and occasionally Gram‐
antibiotics should be commenced at presentation and in positive bacterial, nontuberculous mycobacterial and fun-
cases of soft tissue necrosis surgical debridement will be gal infections [205].
required [182,201].
Treatment. Treatment depends on the severity of infec-
Cutaneous manifestations of systemic infection tion and antibiotic sensitivity profile of the bacteria. It is
Pseudomonas septicaemia is strongly associated with essential to culture the bacteria if at all possible in order to
immunocompromise and has a very high mortality use the correct antibiotics due to the high incidence of
[182,184]. It has also been reported to occur in apparently antibiotic resistance in Pseudomonas and other Gram‐
immunocompetent children [202] and infants [203]. negative bacteria.
Cutaneous manifestations occur in 1.3–13% of cases [182],
and a variety of manifestations occur, some of which are Burn wound infections
very typical of pseudomonal infection and some of which Partial and full thickness burns result in disruption of the
may be mistaken for other bacterial infections, potentially mechanical barrier of the skin, and depress local and sys-
delaying appropriate treatment. temic immune responses. The presence of necrotic tissue
450 Section 7  Bacterial Skin Infections

provides an ideal environment for pathogen survival and index of suspicion is essential, particularly in those with
proliferation and the impairment of blood supply results risk factors such as underlying immunocompromise, as a
in inadequate delivery of systemic immune mediators large proportion of these infections are diagnosed and
and antimicrobial drugs. These factors result in a high treated as simple cellulitis in the early stages [217].
risk of bacterial colonization and infection of burn
wounds, with a high associated mortality and morbidity. Clinical features
Burn wounds will become colonized within a few days, Early clinical features suggestive of complicated infection
initially with Gram‐positive bacteria from the patient’s include pain and tachycardia out of proportion to the ini-
own commensal flora, and later by Gram‐negative tial appearance, with raised inflammatory markers and
­organisms and fungi [208]. P. aeruginosa is one of the lead- biochemical abnormalities and other signs of systemic
ing pathogens responsible for burn wound infections, involvement such as fever and cardiovascular compro-
although other Gram‐negative organisms can also be mise. However, a lack of systemic symptoms at presenta-
responsible [208,209]. The acute management of burn tion does not exclude a necrotizing infection [211,217].
wounds should include measures to clean the wound to Local features of progressive infection include oedema
reduce the risk of secondary bacterial infection such as surrounding the involved area, subcutaneous gas, blister-
early and adequate debridement and strict infection con- ing or bullous changes, ecchymoses and then visibly
trol practices, as well as active surveillance with regular necrotic tissue. The speed of onset can be variable depend-
clinical monitoring, inspection of the wound surface, sur- ing on the organism responsible, but is often rapid [211].
face swabs or biopsy cultures. Prophylactic topical antibi-
otic dressings have been found in a recent meta‐analysis Diagnosis
to increase burn wound infections; the role of systemic The diagnosis of a necrotizing infection is based on clini-
antibiotic prophylaxis remains unclear despite many cal suspicion of severe infection, or sepsis, with or with-
local  and regional burn management clinical guidelines out the specific clinical features mentioned previously
recommending antibiotics to prevent infection [210]. and/or the presence of a risk factor such as immunocom-
Distinguishing between infection and colonization can be promise. A scoring system (the Laboratory Risk Indicator
challenging as local and systemic inflammatory reactions for Necrotizing Fasciitis or LRINEC score) has been devel-
may be due to the initial burn wound, and surface micro- oped by Wong et al [218] as an early indicator of necrotiz-
biology may be positive in either case. Rapid eschar sepa- ing fasciitis based on laboratory values. Imaging studies
ration, changes in surrounding viable tissue, progression may reveal thickening and enhancement of the affected
tissue layers, fluid collections and visible free gas [211]. A
SECTION 7: BACTERIAL SKIN

of burn depth and systemic signs of sepsis are suggestive


of infection. Adequate debridement and appropriate anti- recent study has suggested that computed tomography
biotic treatment should be commenced as soon as possi- (CT) is superior to plain radiography in the detection of
ble when infection is suspected as it can rapidly invade these changes [219].
INFECTIONS

the surrounding and deeper tissues and progress to sepsis Positive microbiology samples will help to confirm the
and multiorgan failure [208]. diagnosis and guide antimicrobial treatment but only
76.5% of wound and 35.2% of blood cultures were posi-
Gram‐negative soft tissue infections tive in a recent review [217]. A wide variety of organisms
can be implicated, with Gram‐positive cocci found in the
Necrotizing skin and soft tissue infections majority of monomicrobial infections. However, many
Necrotizing infections can involve the more superficial cases are polymicrobial and a number of Gram‐negative
skin layers, resulting in gangrenous cellulitis, or the organisms such as E. coli, Acinetobacter spp., P. aeruginosa
deeper layers resulting in necrotizing fasciitis [211]. These and Klebsiella spp. are among those that have been cul-
aggressive infections may start insidiously but then pro- tured [211,217,220].
gress rapidly resulting in necrosis and destruction of the Macroscopic features of necrosis will be seen at the time
affected layers of skin with severe systemic effects includ- of surgical debridement with tissue appearing grey and
ing multiorgan failure and high rates of mortality of up to noncontractile and being easily dissectible with minimal
40% [211–213]. The majority of cases occur in those with resistance and a lack of bleeding. Vessel thrombosis will
significant underlying immunocompromise [184,214]. be evident on histological examination [211].
Varicella infection can be a predisposing factor in children
[215], particularly in association with the use of nonsteroi- Treatment
dal anti‐inflammatory drugs [216]. The prognosis of necrotizing soft tissue infections depends
Uncomplicated skin and soft tissue infections caused upon early recognition of disease and immediate and effec-
by Gram‐positive organisms are common and frequently tive treatment. Good supportive care with adequate fluid
managed in an outpatient setting. Complicated, or resuscitation is vital. Empirical antimicrobial treatment
necrotizing skin and soft tissue infections are much rarer, should be commenced immediately with broad‐spectrum
can be caused by a wider range of organisms including antibiotics covering Gram‐positive, Gram‐negative and
many Gram‐negative organisms and require prompt and anaerobic bacteria according to local antimicrobial
aggressive medical and surgical management. The initial guidelines.
clinical presentation can be similar, with localized skin Surgical debridement of affected tissue should be car-
swelling, erythema and pain. This means that a high ried out as soon as possible, and should be complete with
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 451

a wide margin of normal healthy tissue. A delay of 6 Herrick W. Extrameningeal meningococcal infections. Arch Intern
Med 1919;23:409–18.
debridement beyond 12 hours of presentation has been
7 Yazdankhah SP, Caugant DA. Neisseria meningitidis: an overview of
shown to significantly increase the mortality rate, the the carriage state. J Med Microbiol 2004;53(Pt 9):821–32.
incidence of complications such as septic shock or acute 8 Stephens DS, Greenwood B, Brandtzaeg P. Epidemic meningitis,
kidney disease, and the total number of surgical debride- meningococcaemia, and Neisseria meningitidis. Lancet 2007;369:
2196–210.
ments required [212]. Experimental therapies such as 9 Apicella MA. Neisseria meningitidis. In: Mandell GL, Bennett JE,
intravenous immunoglobulin and hyperbaric oxygen are Dolin R (eds) Mandell, Douglas and Bennett’s Principles and Practices
yet to be proven to reduce mortality or morbidity [211]. of Infectious Diseases, 2, 7th edn. Philadelphia: Churchill Livingstone
Elsevier, 2010:2737–52.
10 Lingappa JR, Al‐Rabeah AM, Hajjeh R et  al. Serogroup W‐135
Gram‐negative folliculitis meningococcal disease during the Hajj, 2000. Emerg Infect Dis
­
2003;9:665–71.
Folliculitis is an inflammation of the hair follicles result- 11 Moura AS, Pablos‐Méndez A, Layton M, Weiss D. Epidemiology of
meningococcal disease, New York City, 1989‐2000. Emerg Infect Dis
ing in papules or pustules, most commonly caused by the 2003;9:355–61.
Gram‐positive pathogen Staphylococcus aureus [221]. 12 Sridhar S, Greenwood B, Head C et al. Global incidence of serogroup
Folliculitis caused by infection with Gram‐negative B invasive meningococcal disease: a systematic review. Lancet Infect
organisms is a recognized complication of long‐term anti- Dis 2015;15:1334–46.
13 Maiden MC, Frosch M. Molecular techniques for the investigation of
biotic treatment in acne vulgaris [222,223]. meningococcal disease epidemiology. Mol Biotechnol 2001;18:119–34.
Gram‐negative folliculitis can be classified as Type 1 or 14 Harrison LH, Trotter CL, Ramsay ME. Global epidemiology of menin-
2 [223]. Type 1 is more common and presents with small, gococcal disease. Vaccine 2009;27(suppl 2):B51–63.
15 Christensen H, May M, Bowen L et al. Meningococcal carriage by
superficial papules or pustules, usually just on the face,
age: a systematic review and meta‐analysis. Lancet Infect Dis 2010;
and extending away from the nares. Organisms such as E. 10:853–61.
coli, Klebsiella, Enterobacter, Serratia marcescens and P. aer- 16 Virji M, Alexandrescu C, Ferguson DJ et al. Variations in the expres-
uginosa are sometimes isolated. Type 2 folliculitis presents sion of pili: the effect on adherence of Neisseria meningitidis to
human epithelial and endothelial cells. Mol Microbiol 1992;6:1271–9.
with larger and deeper cystic or nodular lesions and the 17 Virji M, Makepeace K, Ferguson DJ et al. Expression of the Opc pro-
responsible organism is most commonly Proteus mirabilis. tein correlates with invasion of epithelial and endothelial cells by
This condition is thought to be a result of disruption of Neisseria meningitidis. Mol Microbiol 1992;6:2785–95.
the normal flora of the nasal mucosa and facial skin fol- 18 Lomholt H, Poulsen K, Caugant DA, Kilian M. Molecular polymor-
phism and epidemiology of Neisseria meningitidis immunoglobulin
lowing the use of long‐term systemic antibiotics such as A1 proteases. Proc Natl Acad Sci U S A 1992;89:2120–4.
tetracyclines. Sensitive Gram‐positive organisms are 19 Nassif X, Beretti JL, Lowy J et al. Roles of pilin and PilC in adhesion of

SECTION 7: BACTERIAL SKIN


replaced by more resistant and potentially pathogenic Neisseria meningitidis to human epithelial and endothelial cells. Proc
Natl Acad Sci U S A 1994;91:3769–73.
Gram‐negative organisms in the moist environment of 20 Brendish NJ, Read RC. Neisseria meningitidis serogroup B bivalent
the nasal mucosa. In individuals with increased sebum or factor H binding protein vaccine. Expert Rev Vaccines 2015;14:
sweat production, the local environment is more moist, 493–503.

INFECTIONS
allowing the survival and multiplication of these organ- 21 Frasch CE, Borrow R, Donnelly J. Bactericidal antibody is the immu-
nologic surrogate of protection against meningococcal disease.
isms with resulting inflammation [222,223]. There is some Vaccine 2009;27(suppl 2):6.
evidence that impaired host immunity has a role in sus- 22 Stuart JM, Cartwright KA, Robinson PM, Noah ND. Effect of smoking
ceptibility, with a number of changes in immune param- on meningococcal carriage. Lancet 1989;2:723–5.
23 Goldschneider I, Gotschlich EC, Artenstein MS. Human immunity to
eters seen in affected individuals [224].
the meningococcus. II. Development of natural immunity. J Exp Med
Gram‐negative folliculitis should be considered in indi- 1969;129:1327–48.
viduals in whom what is thought to be acne worsens 24 Blackwell CC, Weir DM, James VS et al. The Stonehouse study: secre-
upon commencement of particular antibiotics, or when it tor status and carriage of Neisseria species. Epidemiol Infect
1989;102:1–10.
does not improve despite 3–6 months of treatment with 25 Moore PS, Hierholzer J, DeWitt W et  al. Respiratory viruses and
systemic tetracyclines. Treatment includes the use of mycoplasma as cofactors for epidemic group A meningococcal men-
isotretinoin which decreases the production of sebum, ingitis. JAMA 1990;264:1271–5.
making the local environment less favourable for the sur- 26 Griffiss JM. Epidemic meningococcal disease: synthesis of a hypo-
thetical immunoepidemiologic model. Rev Infect Dis 1982;4:159–72.
vival of pathogens [225]. Isotretinoin can be used topi- 27 Filice GA, Hayes PS, Counts GW et al. Risk of group A meningococcal
cally or orally but should be used under expert guidance disease: bacterial interference and cross‐reactive bacteria among
due to the potential for side‐effects [226]. mucosal flora. J Clin Microbiol 1985;22:152–6.
28 Harrison LH, Armstrong CW, Jenkins SR et al. A cluster of meningo-
coccal disease on a school bus following epidemic influenza. Arch
References Intern Med 1991;151:1005–9.
1 Eykyn SJ, Gransden WR, Phillips I. The causative organisms of septi- 29 Flaegstad T, Gutteberg T, Kristiansen BE. Antibodies to meningococci
caemia and their epidemiology. J Antimicrob Chemother 1990;25(suppl in children with meningococcal disease. Scand J Infect Dis
C):41–58. 1990;22:547–51.
2 Welch SB, Nadel S. Treatment of meningococcal infection. Arch Dis 30 Brindle R, Simani P, Newnham R et al. No association between menin-
Child 2003;88:608–14. gococcal disease and human immunodeficiency virus in adults in
3 Jones D. Epidemiology of meningococcal disease in Europe and the Nairobi, Kenya. Trans R Soc Trop Med Hyg 1991;85:651.
USA. In: Cartwright K (ed.) Meningococcal Disease. Chichester: John 31 Silber E, Sonnenberg P, Ho KC et al. Meningitis in a community with
Wiley, 1995:71–114. a high prevalence of tuberculosis and HIV infection. J Neurol Sci
4 Vieusseux G. Memoire sur la maladie qui a regne a Geneve au print- 1999;162:20–6.
emps de 1805. J Med Chir Pharm 1806:163. 32 Greenwood BM. The epidemiology of acute bacterial meningitis in
5 Cartwright K. Introduction and historical aspects. In: Cartwright K tropical Africa. In: Williams JK, Burnie J (eds) Bacterial Meningitis.
(ed.) Meningococcal Disease. Chichester: John Wiley, 1995. London: Academic Press, 1987:61–91.
452 Section 7  Bacterial Skin Infections

33 Peltola H. Burden of meningitis and other severe bacterial infections 60 Heyderman RS, Klein NJ, Shennan GI, Levin M. Reduction of the
of children in Africa: implications for prevention. Clin Infect Dis ­anticoagulant activity of glycosaminoglycans on the surface of the
2001;32:64–75. vascular endothelium by endotoxin and neutrophils: evaluation by
34 Lewis R, Nathan N, Diarra L et al. Timely detection of meningococcal an amidolytic assay. Thromb Res 1992;67:677–85.
meningitis epidemics in Africa. Lancet 2001;358:287–93. 61 Osterud B, Flaegstad T. Increased tissue thromboplastin activity in
35 Taha M‐KK, Parent Du Chatelet I et al. Neisseria meningitidis sero- monocytes of patients with meningococcal infection: related to an
groups W135 and A were equally prevalent among meningitis cases unfavourable prognosis. Thromb Haemost 1983;49:5–7.
occurring at the end of the 2001 epidemics in Burkina Faso and Niger. 62 Eling M, Stephens AC, Oragui EE et al. Tissue factor pathway i­ nhibitor
J Clin Microbiol 2002;40:1083–4. (TFPI) levels in the plasma and urine of children with meningococcal
36 Harrison LH, Pass MA, Mendelsohn AB et al. Invasive meningococcal disease. Thromb Haemost 2001;85:240–4.
disease in adolescents and young adults. JAMA 2001;286:694–9. 63 Fourrier F, Lestavel P, Chopin C et al. Meningococcemia and purpura
37 Havens PL, Garland JS, Brook MM et al. Trends in mortality in chil- fulminans in adults: acute deficiencies of proteins C and S and early
dren hospitalized with meningococcal infections, 1957 to 1987. Pediatr treatment with antithrombin III concentrates. Intensive Care Med
Infect Dis J 1989;8:8–11. 1990;16:121–4.
38 Booy R, Habibi P, Nadel S et al. Reduction in case fatality rate from 64 Hesselvik JF, Malm J, Dahlbäck B, Blombäck M. Protein C, protein S
meningococcal disease associated with improved healthcare delivery. and C4b‐binding protein in severe infection and septic shock. Thromb
Arch Dis Child 2001;85:386–90. Haemost 1991;65:126–9.
39 Nadel S. Prospects for eradication of meningococcal disease. Arch Dis 65 Brandtzaeg P, Joø GB, Brusletto B, Kierulf P. Plasminogen activator
Child 2012;97:993–8. inhibitor 1 and 2, alpha‐2‐antiplasmin, plasminogen, and endotoxin
40 Hill WR, Kinney TD. The cutaneous lesions in acute meningococcae- levels in systemic meningococcal disease. Thromb Res1990;57:271–8.
mia: a clinical and pathologic study. JAMA 1947;134:513–18. 66 Powars D, Larsen R, Johnson J et al. Epidemic meningococcemia and
41 Robboy SJ, Mihm MC, Colman RW, Minna JD. The skin in dissemi- purpura fulminans with induced protein C deficiency. Clin Infect Dis
nated intravascular coagulation. Prospective analysis of thirty‐six 1993;17:254–61.
cases. Br J Dermatol 1973;88:221–9. 67 Hermans PW, Hibberd ML, Booy R et al. 4G/5G promoter polymor-
42 Cahalane SF, Waters M. Fulminant meningococcal septicaemia. A hos- phism in the plasminogen‐activator‐inhibitor‐1 gene and outcome
pital experience. Lancet 1975;2:120–1. of meningococcal disease. Meningococcal Research Group. Lancet
43 Evans RW, Glick B, Kimball F, Bobell M. Fatal intravascular consump- 1999;354:556–60.
tion coagulopathy in meningococcal sepsis. Am J Med 1969;46:910–8. 68 Westendorp RG, Hottenga JJ, Slagboom PE. Variation in plasmino-
44 McGehee WG, Rapaport SI, Hjort PF. Intravascular coagulation in ful- gen‐activator‐inhibitor‐1 gene and risk of meningococcal septic
minant meningococcemia. Ann Intern Med 1967;67:250–60. shock. Lancet 1999;354:561–3.
45 Ferguson JH, Chapman OD. Fulminating meningococcic infections 69 Kirchhofer D, Sakariassen KS, Clozel M, et al. Relationship between
and the so‐called Waterhouse‐Friderichsen syndrome. Am J Pathol tissue factor expression and deposition of fibrin, platelets, and
1948;24:763–95. ­leukocytes on cultured endothelial cells under venous blood flow
46 Sotto MN, Langer B, Hoshino‐Shimizu S, de Brito T. Pathogenesis of conditions. Blood 1993;81:2050–8.
cutaneous lesions in acute meningococcemia in humans: light, immu- 70 Zimmerman GA, Prescott SM, McIntyre TM. Endothelial cell interac-
nofluorescent, and electron microscopic studies of skin biopsy speci- tions with granulocytes: tethering and signaling molecules. Immunol
mens. J Infect Dis 1976;133:506–14. Today 1992;13:93–100.
SECTION 7: BACTERIAL SKIN

47 Waage A, Halstensen A, Espevik T. Association between tumour 71 Wachtfogel YT, Abrams W, Kucich U et  al. Fibronectin degradation
necrosis factor in serum and fatal outcome in patients with meningo- products containing the cytoadhesive tetrapeptide stimulate human
coccal disease. Lancet 1987;1:355–7. neutrophil degranulation. J Clin Invest 1988;81:1310–6.
48 Waage A, Brandtzaeg P, Halstensen A et al. The complex pattern of 72 Bone RC. Modulators of coagulation. A critical appraisal of their role
cytokines in serum from patients with meningococcal septic shock. in sepsis. Arch Intern Med 1992;152:1381–9.
INFECTIONS

Association between interleukin 6, interleukin 1, and fatal outcome. J 73 Dixon GL, Heyderman RS, Kotovicz K et  al. Endothelial adhesion
Exp Med 1989;169:333–8. molecule expression and its inhibition by recombinant bactericidal/
49 Brandtzaeg P, Kierulf P, Gaustad P et al. Plasma endotoxin as a predic- permeability‐increasing protein are influenced by the capsulation and
tor of multiple organ failure and death in systemic meningococcal lipooligosaccharide structure of Neisseria meningitidis. Infect Immun
disease. J Infect Dis 1989;159:195–204. 1999;67:5626–33.
50 Brandtzaeg P, Sandset PM, Joø GB et al. The quantitative association 74 Harrison OB, Robertson BD, Faust SN et al. Analysis of pathogen‐host
of plasma endotoxin, antithrombin, protein C, extrinsic pathway cell interactions in purpura fulminans: expression of capsule, type IV
inhibitor and fibrinopeptide A in systemic meningococcal disease. pili, and PorA by Neisseria meningitidis in vivo. Infect Immun
Thromb Res 1989;55:459–70. 2002;70:5193–201.
51 Girardin E, Grau GE, Dayer JM et al. Tumor necrosis factor and inter- 75 Klein NJ, Ison CA, Peakman M et al. The influence of capsulation and
leukin‐1 in the serum of children with severe infectious purpura. N lipooligosaccharide structure on neutrophil adhesion molecule
Engl J Med 1988;319:397–400. expression and endothelial injury by Neisseria meningitidis. J Infect
52 Pathan N, Faust SN, Levin M. Pathophysiology of meningococcal Dis1996;173:172–9.
meningitis and septicaemia. Arch Dis Child 2003;88:601–7. 76 Dunn KL, Virji M, Moxon ER. Investigations into the molecular basis
53 Gedde‐Dahl TW, Bjark P, Høiby EA et al. Severity of meningococcal of meningococcal toxicity for human endothelial and epithelial cells:
disease: assessment by factors and scores and implications for patient the synergistic effect of LPS and pili. Microb Pathog 1995;18:81–96.
management. Rev Infect Dis 1990;12:973–92. 77 Heyderman RS, Klein NJ, Daramola OA et  al. Induction of human
54 Faust SN, Heyderman RS, Levin M. Disseminated intravascular coag- endothelial tissue factor expression by Neisseria meningitidis: the
ulation and purpura fulminans secondary to infection. Baillieres Best influence of bacterial killing and adherence to the endothelium.
Pract Res Clin Haematol 2000;13:179–97. Microb Pathog 1997;22:265–74.
55 Gimbrone M. Vascular endothelium: nature’s blood container. In: 78 Join‐Lambert O, Lecuyer H, Miller F et  al. Meningococcal interac-
Gimbrone M (ed.) Endothelium in Haemostasis and Thrombosis. tion  to microvasculature triggers the tissular lesions of purpura
Edinburgh: Churchill Livingstone, 1986:1–12. ­fulminans. J Infect Dis 2013;208:1590–7.
56 Hack CE, Zeerleder S. The endothelium in sepsis: source of and a tar- 79 Lecuyer H, Borgel D, Nassif X, Coureuil M. Pathogenesis of menin-
get for inflammation. Crit Care Med 2001;29(7 Suppl):7. gococcal purpura fulminans. Pathog Dis 2017;75(3).
57 Faust SN, Levin M, Harrison OB et al. Dysfunction of endothelial pro- 80 Melican K, Michea Veloso P, Martin T et  al. Adhesion of Neisseria
tein C activation in severe meningococcal sepsis. N Engl J Med meningitidis to dermal vessels leads to local vascular damage and
2001;345:408–16. purpura in a humanized mouse model. PLoS Pathog 2013;9:e1003139.
58 Proceedings of the Third Margaux Conference on Critical Illness: The 81 Weatherall DJ, Bell JI, Clegg JB et al. Genetic factors as determinants
Endothelium ‐ an Underrecognized Organ in Critical Illness? Sedona, of infectious disease transmission in human communities. Phil Trans
Arizona, USA. November 14‐18, 2001. Crit Care Med 2002;30(5 R Soc Lond B1988;321:327 Pathogenesis of meningococcal purpura
suppl):S179–348. fulminans 48.
59 Heyderman RS, Klein NJ, Shennan GI, Levin M. Deficiency of prostacyc- 82 Hibberd ML, Sumiya M, Summerfield JA et  al. Association of
lin production in meningococcal shock. Arch Dis Child 1991;66:1296–9. ­variants of the gene for mannose‐binding lectin with susceptibility to
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 453

meningococcal disease. Meningococcal Research Group. Lancet 111 Jones DM, Sutcliffe EM. Meningococci with reduced susceptibility to
1999;353:1049 Pathogenesis of meningococcal purpura fulminans 53. penicillin. Lancet 1990;335:863–4.
83 Westendorp RG, Langermans JA, Huizinga TW et al. Genetic influ- 112 Blondeau JM, Ashton FE, Isaacson M et  al. Neisseria meningitidis
ence on cytokine production in meningococcal disease. Lancet with decreased susceptibility to penicillin in Saskatchewan, Canada.
1997;349:1912–13. J Clin Microbiol1995;33:1784–6.
84 Bredius RG, Derkx BH, Fijen CA et al. Fc gamma receptor IIa (CD32) 113 Jackson LA, Tenover FC, Baker C et al. Prevalence of Neisseria men-
polymorphism in fulminant meningococcal septic shock in children. ingitidis relatively resistant to penicillin in the United States, 1991.
J Infect Dis 1994;170:848–53. Meningococcal Disease Study Group. J Infect Dis 1994;169:438–41.
85 Bertina RM. Factor V Leiden and other coagulation factor mutations 114 Oppenheim BA. Antibiotic resistance in Neisseria meningitidis. Clin
affecting thrombotic risk. Clin Chem 1997;43:1678–83. Infect Dis 1997;24(suppl 1):101.
86 Kondaveeti S, Hibberd ML, Booy R et  al. Effect of the Factor V 115 Temime L, Boëlle PY, Courvalin P, Guillemot D. Bacterial resistance
Leiden mutation on the severity of meningococcal disease. Pediatr to penicillin G by decreased affinity of penicillin‐binding proteins: a
Infect Dis J 1999;18:893–6. mathematical model. Emerg Infect Dis 2003;9:411–17.
87 Kornelisse RF, Hazelzet JA, Savelkoul HF et  al. The relationship 116 Mastrantonio P, Stefanelli P, Fazio C et al. Serotype distribution, anti-
between plasminogen activator inhibitor‐1 and proinflammatory biotic susceptibility, and genetic relatedness of Neisseria meningitidis
and counterinflammatory mediators in children with meningococcal strains recently isolated in Italy. Clin Infect Dis 2003;36:422–8.
septic shock. J Infect Dis 1996;173:1148–56. 117 Tapsall JW, Shultz T, Limnios E et al. Surveillance of antibiotic resist-
88 Pomeroy SL, Holmes SJ, Dodge PR, Feigin RD. Seizures and other ance in invasive isolates of Neisseria meningitidis in Australia
neurologic sequelae of bacterial meningitis in children. N Engl J Med 1994‐1999. Pathology 2001;33:359–61.
1990;323:1651–7. 118 Richter SS, Gordon KA, Rhomberg PR et al. Neisseria meningitidis
89 Marzouk O, Thomson AP, Sills JA et  al. Features and outcome in with decreased susceptibility to penicillin: report from the SENTRY
meningococcal disease presenting with maculopapular rash. Arch antimicrobial surveillance program, North America, 1998‐99. Diagn
Dis Child 1991;66:485–7. Microbiol Infect Dis 2001;41:83–8.
90 Pollard AJ, Britto J, Nadel S et al. Emergency management of menin- 119 de Jonge E, Levi M, Stoutenbeek CP, van Deventer SJ. Current drug
gococcal disease. Arch Dis Child 1999;80:290–6. treatment strategies for disseminated intravascular coagulation.
91 Kingston ME, Mackey D. Skin clues in the diagnosis of life‐ Drugs 1998;55:767–77.
threatening infections. Rev Infect Dis 1986;8:1–11. 120 Balk R, Emerson T, Fourrier F et al. Therapeutic use of antithrombin
92 Toews WH, Bass JW. Skin manifestations of meningococcal infection; concentrate in sepsis. Semin Thromb Hemost. 1998;24:183–94.
an immediate indicator of prognosis. Am J Dis Child 1974;127:173–6. 121 Giudici D, Baudo F, Palareti G et  al. Antithrombin replacement in
93 Stiehm ER, Damrosch DS. Factors in the prognosis of meningococcal patients with sepsis and septic shock. Haematologica 1999;84:452–60.
infection. Review of 63 cases with emphasis on recognition and man- 122 Emerson TE. Antithrombin III replacement in animal models of
agement of the severely ill patient. J Pediatr 1966;68:457–67. acquired antithrombin III deficiency. Blood Coagul Fibrinolysis
94 Brandtzaeg P, Dahle JS, Høiby EA. The occurrence and features of 1994;5(suppl 1):S37–45.
hemorrhagic skin lesions in 115 cases of systemic meningococcal dis- 123 Taylor FB, Emerson TE, Jordan R et al. Antithrombin‐III prevents the
ease. NIPH Ann 1983;6:183. lethal effects of Escherichia coli infusion in baboons. Circ Shock
95 Cartwright KA, Jones DM. ACP Broadsheet 121: June 1989. 1988;26:227–35.
Investigation of meningococcal disease. J Clin Pathol 1989;42:634–9. 124 Rivard GE, David M, Farrell C, Schwarz HP. Treatment of purpura

SECTION 7: BACTERIAL SKIN


96 Nadel S. Lumbar puncture should not be performed in meningococ- fulminans in meningococcemia with protein C concentrate. J Pediatr
cal disease. Arch Dis Child 2001;84:375. 1995;126:646–52.
97 van Deuren M, van Dijke BJ, Koopman RJ et al. Rapid diagnosis of 125 Lundsgaard‐Hansen P, Doran JE, Rubli E et al. Purified fibronectin
acute meningococcal infections by needle aspiration or biopsy of administration to patients with severe abdominal infections. A con-
skin lesions. Br Med J (Clin Res Ed) 1993;306:1229–32. trolled clinical trial. Ann Surg 1985;202:745–59.

INFECTIONS
98 Periappuram M, Taylor MR, Keane CT. Rapid detection of meningo- 126 White B, Livingstone W, Murphy C et al. An open‐label study of the
cocci from petechiae in acute meningococcal infection. J Infect role of adjuvant hemostatic support with protein C replacement
1995;31:201–3. therapy in purpura fulminans‐associated meningococcemia. Blood
99 Netter A, Sakabuer M. The presence of meningococci in the purpuric 2000;96:3719–24.
elements of meningococcal infection. Br J Child Dis 1917(14):101–4. 127 Taylor FB, Chang A, Ruf W et al. Lethal E. coli septic shock is pre-
100 Tompkins V. The diagnostic value of smears from purpuric lesions of vented by blocking tissue factor with monoclonal antibody. Circ
the skin in meningococcal disease. JAMA 1943(123):31–2. Shock 1991;33:127–34.
101 McLean S, Caffey J. Endemic meningococcus bacteremia in early life: 128 Angelini GD, Cooper GJ, Lamarra M, Bryan AJ. Unorthodox use of
the diagnostic value of smears from purpuric lesions. Am J Dis Child aprotinin to control life‐threatening bleeding after cardiopulmonary
1931(42):1053–74. bypass. Lancet 1990;335:799–800.
102 Kaplan SL. Antigen detection in cerebrospinal fluid—pros and cons. 129 Creasey AA, Reinhart K. Tissue factor pathway inhibitor activity in
Am J Med 1983;75:109–18. severe sepsis. Crit Care Med 2001;29(7 suppl):9.
103 Cuevas LE, Hart CA, Mughogho G. Latex particle agglutination tests 130 Zenz W, Muntean W, Gallistl S et al. Recombinant tissue plasmino-
as an adjunct to the diagnosis of bacterial meningitis: a study from gen activator treatment in two infants with fulminant meningococ-
Malawi. Ann Trop Med Parasitol 1989;83:375–9. cemia. Pediatrics 1995;96:144–8.
104 Newcombe J, Cartwright K, Palmer WH, McFadden J. PCR of 131 Colman RW, Robboy SJ, Minna JD. Disseminated intravascular coag-
peripheral blood for diagnosis of meningococcal disease. J Clin ulation (DIC): an approach. Am J Med 1972;52:679–89.
Microbiol 1996;34:1637–40. 132 Corrigan JJ, Ray WL, May N. Changes in the blood coagulation sys-
105 Kotilainen P, Jalava J, Meurman O et al. Diagnosis of meningococcal tem associated with septicemia. N Engl J Med 1968;279:851–6.
meningitis by broad‐range bacterial PCR with cerebrospinal fluid. J 133 Gérard P, Moriau M, Bachy A et al. Meningococcal purpura: report of
Clin Microbiol 1998;36:2205–9. 19 patients treated with heparin. J Pediatr 1973;82:780–6.
106 Booy R, Kroll JS. Bacterial meningitis and meningococcal infection. 134 Ockelford P. Heparin 1986. Indications and effective use. Drugs
Curr Opin Pediatr 1998;10:13–8. 1986;31:81–92.
107 Hackett SJ, Carrol ED, Guiver M et al. Improved case confirmation in 135 Warren BL, Eid A, Singer P et al. Caring for the critically ill patient.
meningococcal disease with whole blood Taqman PCR. Arch Dis High‐dose antithrombin III in severe sepsis: a randomized con-
Child 2002;86:449–52. trolled trial. JAMA 2001;286:1869–78.
108 Botha P. Penicillin‐resistant Neisseria meningitidis in southern 136 Derkx B, Wittes J, McCloskey R. Randomized, placebo‐controlled trial
Africa. Lancet 1988;1:54. of HA‐1A, a human monoclonal antibody to endotoxin, in children
109 Berrón S, Vázquez JA. Increase in moderate penicillin resistance and with meningococcal septic shock. European Pediatric Meningococcal
serogroup C in meningococcal strains isolated in Spain. Is there any Septic Shock Trial Study Group. Clin Infect Dis 1999;28:770–7.
relationship? Clin Infect Dis 1994;18:161–5. 137 Levin M, Quint PA, Goldstein B et al. Recombinant bactericidal/per-
110 Angyo IA, Okpeh ES. Changing patterns of antibiotic sensitivity and meability‐increasing protein (rBPI21) as adjunctive treatment for
resistance during an outbreak of meningococcal infection in Jos, children with severe meningococcal sepsis: a randomised trial.
Nigeria. J Trop Pediatr 1998;44:263–5. rBPI21 Meningococcal Sepsis Study Group. Lancet 2000;356:961–7.
454 Section 7  Bacterial Skin Infections

138 Bone RC, Fisher CJ, Clemmer TP et al. A controlled clinical trial of 163 Edwards MS, Baker CJ. Complications and sequelae of meningococ-
high‐dose methylprednisolone in the treatment of severe sepsis and cal infections in children. J Pediatr 1981;99:540–5.
septic shock. N Engl J Med 1987;317:653–8. 164 Greenwood BM, Whittle HC, Bryceson AD. Allergic complications
139 Jafari HS, McCracken GH. Sepsis and septic shock: a review for clini- of meningococcal disease. II. Immunological investigations. Br Med
cians. Pediatr Infect Dis J 1992;11:739–48. J 1973;2:737–40.
140 Margaretten W, McAdams AJ. An appraisal of fulminant meningo- 165 Benoit FL. Chronic meningococcemia. Case report and review of the
coccemia with reference to the Shartzman phenomenon. Am J Med literature. Am J Med 1963;35:103–12.
1958;25:868–76. 166 Leibel RL, Fangman JJ, Ostrovsky MC. Chronic meningococcemia in
141 Annane D, Sébille V, Charpentier C et al. Effect of treatment with low childhood. Case report and review of the literature. Am J Dis Child
doses of hydrocortisone and fludrocortisone on mortality in patients 1974;127:94–8.
with septic shock. JAMA 2002;288:862–71. 167 Nielsen HE, Koch C, Mansa B et al. Complement and immunoglob-
142 Bernard GR, Vincent JL, Laterre PF et  al. Efficacy and safety of ulin studies in 15 cases of chronic meningococcemia: properdin
recombinant human activated protein C for severe sepsis. N Engl J deficiency and hypoimmunoglobulinemia. Scand J Infect Dis
Med 2001;344:699–709. 1990;22:31–6.
143 Nadel S, Goldstein B, Williams MD et al. Drotrecogin alfa (activated) 168 Prins JM, Lauw FN, Derkx BH et al. Endotoxin release and cytokine
in children with severe sepsis: a multicentre phase III randomised production in acute and chronic meningococcaemia. Clin Exp
controlled trial. Lancet 2007;369:836–43. Immunol 1998;114:215–9.
144 Martí‐Carvajal AJ, Solà I, Gluud C et al. Human recombinant protein 169 Visakorpi R. Ciprofloxacin in meningococcal carriers. Scand J Infect
C for severe sepsis and septic shock in adult and paediatric patients. Dis Suppl 1989;60:108–11.
Cochrane Database Syst Rev 2012;12. 170 Pollard AJ, Begg N. Meningococcal disease and healthcare workers.
145 de Kleijn ED, de Groot R, Hack CE et al. Activation of protein C fol- Br Med J (Clin Res Ed) 1999;319:1147–8.
lowing infusion of protein C concentrate in children with severe 171 Control of meningococcal disease: guidance for consultants in com-
meningococcal sepsis and purpura fulminans: a randomized, dou- municable disease control. PHLS Meningococcal Infections Working
ble‐blinded, placebo‐controlled, dose‐finding study. Crit Care Med Group and Public Health Medicine Environmental Group. Commun
2003;31:1839–47. Dis Rep CDR Rev 1995;5:95.
146 Rintala E, Kauppila M, Seppälä OP et  al. Protein C substitution in 172 Forum HPAMaH. Guidance for public health management of
sepsis‐associated purpura fulminans. Crit Care Med 2000;28:2373–8. meningococcal disease in the UK 2018 [Available from: https://
147 Bernard G, Artigas A, Dellinger P et al. Clinical expert round table www.gov.uk/government/uploads/system/uploads/attachment_
discussion (session 3) at the Margaux Conference on Critical Illness: data/file/322008/Guidance_for_management_of_meningococcal_
the role of activated protein C in severe sepsis. Crit Care Med disease_pdf.pdf.
2001;29(7 suppl):7. 173 Frasch CE. Meningococcal vaccine: past, present and future. In:
148 Zenz W, Zöhrer B, Faust SN et al. (eds). Use of tissue plasminogen Cartwright K (ed.) Meningococcal Disease. Chichester: John Wiley,
activator (t‐PA) in children with fulminant meningococcaemia. 1995: 246–83.
Royal College of Paediatrics and Child Health Annual Meetings; 174 Shinefield HR, Black S. Postlicensure surveillance for Haemophilus
2001; York, UK. London: Royal College of Paediatrics and Child influenzae type b invasive disease after use of Haemophilus influen-
Health. zae type b oligosaccharide CRM197 conjugate vaccine in a large
149 Nectoux E, Mezel A, Raux S et al. Meningococcal purpura fulminans defined United States population: a four‐year eight‐month follow‐
SECTION 7: BACTERIAL SKIN

in children: I. Initial orthopedic management. J Child Orthop up. Pediatr Infect Dis J1995;14:978–81.
2010;4:401–7. 175 Hargreaves RM, Slack MP, Howard AJ et  al. Changing patterns of
150 Davies MS, Nadel S, Habibi P et al. The orthopaedic management of invasive Haemophilus influenzae disease in England and Wales
peripheral ischaemia in meningococcal septicaemia in children. J after introduction of the Hib vaccination programme. Br Med J (Clin
Bone Joint Surg Br 2000;82:383–6. Res Ed) 1996;312:160–1.
INFECTIONS

151 Monsell F. The skeletal consequences of meningococcal septicaemia. 176 Bose A, Coen P, Tully J et al. Effectiveness of meningococcal C conju-
Arch Dis Child 2012;97:539–44. gate vaccine in teenagers in England. Lancet 2003;361:675–6.
152 Herrera R, Hobar PC, Ginsburg CM. Surgical intervention for the 177 Lakshman R, Jones I, Walker D et  al. Safety of a new conjugate
complications of meningococcal‐induced purpura fulminans. meningococcal C vaccine in infants. Arch Dis Child 2001;85:391–7.
Pediatr Infect Dis J 1994;13:734–7. 178 Salleras L, Domínguez A, Cardeñosa N. Impact of mass vaccination
153 Hudson DA, Goddard EA, Millar KN. The management of skin with polysaccharide conjugate vaccine against serogroup C menin-
infarction after meningococcal septicaemia in children. Br J Plast gococcal disease in Spain. Vaccine 2003;21:725–8.
Surg 1993;46:243–6. 179 Ramsay ME, Andrews N, Kaczmarski EB, Miller E. Efficacy of
154 Harris NJ, Gosh M. Skin and extremity loss in meningococcal septi- meningococcal serogroup C conjugate vaccine in teenagers and
caemia treated in a burn unit. Burns 1994;20:471–2. toddlers in England. Lancet 2001;357:195–6.
155 Arévalo JM, Lorente JA, Fonseca R. Surgical treatment of extensive 180 Ramsay ME, Andrews NJ, Trotter CL et  al. Herd immunity from
skin necrosis secondary to purpura fulminans in a patient with meningococcal serogroup C conjugate vaccination in England: data-
meningococcal sepsis. Burns 1998;24:272–4. base analysis. Br Med J (Clin Res Ed)2003;326:365–6.
156 Huang DB, Price M, Pokorny J et al. Reconstructive surgery in chil- 181 Pollard AJ, Perrett KP, Beverley PC. Maintaining protection
dren after meningococcal purpura fulminans. J Pediatr against invasive bacteria with protein‐polysaccharide conjugate
Surg1999;34:595–601. vaccines. Nat Rev Immunol 2009;9:213–20.
157 Boeckx WD, Nanhekhan L, Vos GD et al. Minimizing limb amputa- 182 Silvestre JF, Betlloch MI. Cutaneous manifestations due to
tions in meningococcal sepsis by early microsurgical arteriolysis. J Pseudomonas infection. Int J Dermatol 1999;38:419–31.
Pediatr Surg 2009;44:1625–30. 183 Logan LK, Gandra S, Mandal S et al. Multidrug‐ and carbapenem‐
158 Nectoux E, Mezel A, Raux S et al. Meningococcal purpura fulminans resistant Pseudomonas aeruginosa in children, United States,
in children. II: Late orthopedic sequelae management. J Child 1999‐2012. J Pediatric Infect Dis Soc 2017;6:352–9.
Orthop 2010;4:409–16. 184 Wu DC, Chan WW, Metelitsa AI et al. Pseudomonas skin infection:
159 Buysse CM, Oranje AP, Zuidema E et al. Long‐term skin scarring and clinical features, epidemiology, and management. Am J Clin
orthopaedic sequelae in survivors of meningococcal septic shock. Dermatol 2011;12:157–69.
Arch Dis Child 2009;94:381–6. 185 Goering R, Dockrell H, Zuckerman M et  al. Mims’ Medical
160 Allport T, Read L, Nadel S, Levin M. Critical illness and amputation Microbiology, 5th edn. Edinburgh: Elsevier Saunders, 2012.
in meningococcal septicemia: is life worth saving? Pediatrics 186 Diekema DJ, Pfaller MA, Jones RN et  al. Survey of bloodstream
2008;122:629–32. infections due to gram‐negative bacilli: frequency of occurrence and
161 Buysse CM, Raat H, Hazelzet JA et al. Surviving meningococcal sep- antimicrobial susceptibility of isolates collected in the United States,
tic shock: health consequences and quality of life in children and Canada, and Latin America for the SENTRY Antimicrobial
their parents up to 2 years after pediatric intensive care unit dis- Surveillance Program, 1997. Clin Infect Dis 1999;29:595–607.
charge. Crit Care Med2008;36:596–602. 187 Bucior I, Pielage JF, Engel JN. Pseudomonas aeruginosa pili and fla-
162 Whittle HC, Abdullahi MT, Fakunle FA et al. Allergic complications gella mediate distinct binding and signaling events at the apical and
of meningococcal disease. I. Clinical aspects. Br Med J 1973;2:733–7. basolateral surface of airway epithelium. PLoS Pathog 2012;8:e1002616.
Chapter 38  Cutaneous Manifestations of  Gram‐negative Infections 455

188 Korpi F, Hashemi FB, Irajian G et al. Flagellin and pilin immuni- 208 Church D, Elsayed S, Reid O et  al. Burn wound infections. Clin
zation against multi‐drug resistant Pseudomonas aeruginosa pro- Microbiol Rev 2006;19:403–34.
tects mice in the burn wound sepsis model. Immunol Lett 2016; 209 Altoparlak U, Erol S, Akcay MN et al. The time‐related changes of
176:8–17. antimicrobial resistance patterns and predominant bacterial profiles
189 Hall‐Stoodley L, Stoodley P. Evolving concepts in biofilm infections. of burn wounds and body flora of burned patients. Burns
Cell Microbiol 2009;11:1034–43. 2004;30:660–4.
190 Wilkins M, Hall‐Stoodley L, Allan RN, Faust SN. New approaches 210 Barajas‐Nava LA, López‐Alcalde J, Roqué i Figuls M et al. Antibiotic
to the treatment of biofilm‐related infections. J Infect 2014;69 prophylaxis for preventing burn wound infection. Cochrane
(suppl 1):52. Database Syst Rev 2013(6).
191 Pier GB, Ramphal R. Pseudomonas aeruginosa. In: Mandell GL, 211 Anaya DA, Dellinger EP. Necrotizing soft‐tissue infection: diagno-
Bennett JE, Dolin R (eds) Mandell, Douglas and Bennett’s Principles sis and management. Clin Infect Dis 2007;44:705–10.
and Practices of Infectious Diseases, 2, 7th edn. Philadelphia: 212 Kobayashi L, Konstantinidis A, Shackelford S et al. Necrotizing soft
Churchill Livingstone Elselvier, 2010:2835–60. tissue infections: delayed surgical treatment is associated with
192 Sampedro I, Parales RE, Krell T, Hill JE. Pseudomonas chemotaxis. increased number of surgical debridements and morbidity. J Trauma
FEMS Microbiol Rev 2015;39:17–46. 2011;71:1400–5.
193 Andonova M, Urumova V. Immune surveillance mechanisms of the 213 Sarani B, Strong M, Pascual J, Schwab CW. Necrotizing fasciitis:
skin against the stealth infection strategy of Pseudomonas aeruginosa‐ current concepts and review of the literature. J Am Coll Surg
review. Comp Immunol Microbiol Infect Dis 2013;36:433–48. 2009;208:279–88.
194 Stieritz DD, Holder IA. Experimental studies of the pathogenesis of 214 Zhao G‐JJ, Hong G‐LL, Liu J‐QQ et al. Septic shock due to community‐
infections due to Pseudomonas aeruginosa: description of a burned acquired Pseudomonas aeruginosa necrotizing fasciitis: A case report
mouse model. J Infect Dis 1975;131:688–91. and literature review. Exp Ther Med 2014;7:1545–8.
195 Antibiotic resistance threats in the United States, 2013. Centers for 215 Totapally BR. Epidemiology and outcomes of hospitalized chil-
Disease Control and Prevention, 2013. Available at: https://www. dren  with necrotizing soft tissue infections. Pediatr Infect Dis J
cdc.gov/drugresistance/pdf/ar-threats-2013-508.pdf 2017;36:641–4.
196 Høiby N, Bjarnsholt T, Givskov M et al. Antibiotic resistance of bac- 216 Mikaeloff Y, Kezouh A, Suissa S. Nonsteroidal anti‐inflammatory drug
terial biofilms. Int J Antimicrob Agents 2010;35:322–32. use and the risk of severe skin and soft tissue complications in patients
197 Rubinstein E, Ostfeld E, Ben‐Zaray S, Schiby G. Necrotizing external with varicella or zoster disease. Br J Clin Pharmacol 2008;65:203–9.
otitis. Pediatrics 1980;66:618–20. 217 Goh T, Goh LG, Ang CH, Wong CH. Early diagnosis of necrotizing
198 Chiller K, Selkin BA, Murakawa GJ. Skin microflora and bacterial fasciitis. Br J Surg 2014;101:25.
infections of the skin. J Investig Dermatol Symp Proc 2001;6:170–4. 218 Wong CH, Khin LW, Heng KS et al. The LRINEC (Laboratory Risk
199 Saegeman V, Van Meensel B. Aeromonas associated with swimming Indicator for Necrotizing Fasciitis) score: a tool for distinguishing
pool folliculitis. Pediatr Infect Dis J 2016;35:118–19. necrotizing fasciitis from other soft tissue infections. Crit Care Med
200 Julià Manresa M, Vicente Villa A, Gené Giralt A, González‐Enseñat 2004;32:1535–41.
MA. Aeromonas hydrophila folliculitis associated with an inflatable 219 Leichtle SW, Tung L, Khan M et al. The role of radiologic evaluation
swimming pool: mimicking Pseudomonas aeruginosa infection. in necrotizing soft tissue infections. J Trauma Acute Care Surg
Pediatr Dermatol 2009;26:601–3. 2016;81:921–4.
201 Stewart D, Benitz W, Newbo CO. Umbilical cord care in the newborn 220 Wong C‐HH, Chang H‐CC, Pasupathy S et al. Necrotizing fasciitis:

SECTION 7: BACTERIAL SKIN


infant. Pediatrics 2016;138(3). clinical presentation, microbiology, and determinants of mortality. J
202 Mull CC, Scarfone RJ, Conway D. Ecthyma gangrenosum as a mani- Bone Joint Surg Am 2003;85‐A:1454–60.
festation of Pseudomonas sepsis in a previously healthy child. Ann 221 Stulberg DL, Penrod MA, Blatny RA. Common bacterial skin infec-
Emerg Med 2000;36:383–7. tions. Am Fam Physician 2002;66:119–24.
203 Pandit AM, Siddaramappa B, Choudhary SV, Manjunathswamy BS. 222 Lehrhoff S, Yost J, Robinson M et al. Serratia marcescens folliculitis

INFECTIONS
Ecthyma gangrenosum in a new born child. Indian J Dermatol and concomitant acne vulgaris. Dermatol Online J 2012;18:19.
Venereol Leprol 2003;69:52–3. 223 Leyden JJ, Marples RR, Mills OH Jr, Kligman AM. Gram‐negative
204 Whitecar JP Jr, Luna M, Bodey GP. Pseudomonas bacteremia in folliculitis—a complication of antibiotic therapy in acne vulgaris.
patients with malignant diseases. Am J Med Sci 1970;60:216–23. Br J Dermatol 1973;88:533–8.
205 Vaiman M, Lazarovitch T, Heller L, Lotan G. Ecthyma gangrenosum 224 Neubert U, Jansen T, Plewig G. Bacteriologic and immunologic
and ecthyma‐like lesions: review article. Eur J Clin Microbiol Infect aspects of gram‐negative folliculitis: a study of 46 patients. Int J
Dis 2015;34:633–9. Dermatol 1999;38:270–4.
206 Cohen N, Capua T, Bilavsky E et  al. Ecthyma gangrenosum skin 225 Boni R, Nehrhoff B. Treatment of gram‐negative folliculitis in
lesions in previously healthy children. Acta Paediatr 2015;104:8. patients with acne. Am J Clin Dermatol 2003;4:273–6.
207 Ghanaiem H, Engelhard D. A healthy 2‐year‐old child with a round 226 British National Formulary for Children. London: BMJ Group and
black skin lesion. J Pediatr 2013;163:1225–50. Pharmaceutical Press and RCPCH Publications, 2010.
456 

CHA PTER  3 9

Pitted Keratolysis, Erythrasma and Erysipeloid


Zhe Xu, Yuanyuan Xiao, Ying Liu & Lin Ma
Department of Dermatology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China

Pitted keratolysis, 456 Erythrasma, 458 Erysipeloid, 460

Abstract the groin, axilla, gluteal crease or inframammary regions, and inter-
digital spaces of the feet. Erythrasma is frequently confused with
dermatophyte infections (i.e. tinea corporis), with which it may
Pitted keratolysis (PK) is a superficial bacterial infection that
occasionally coexist. However, it can be differentiated from tinea
affects the plantar stratum corneum and is caused most frequently
infection by the characteristic ‘coral‐red’ fluorescence under Wood’s
by Corynebacterium spp., Kytococcus sedentarius, Dermatophilus
lamp illumination. Erythrasma can be treated with topical antibiot-
congolensis, Actinomyces keratolytica and Streptomyces spp. PK is
ics, including erythromycin or clindamycin. Recalcitrant cases can
not limited to barefooted populations in tropical areas but has a
be treated with oral antibiotics such as tetracyclines or macrolides.
worldwide distribution. The infection is characterized by circular,
Erysipeloid is an acute skin infection caused by Erysipelothrix
shallow erosions and pits. Areas of greater pressure, friction and
rhusiopathiae, mostly subsequent to cutaneous trauma. It is
hyperhidrosis are more commonly affected. Topical antibiotics such
­characterized clinically by an erythematous oedema, with well‐
as erythromycin and clindamycin are the first‐line treatment while
defined and raised borders, usually localized to the back of one
elimination of predisposing factors such as hyperhidrosis and pro-
hand and/or fingers. Vesicular, bullous and erosive lesions may
longed contact with water is also helpful.
also be present. In addition to cutaneous infection, E. rhusiopathiae
Erythrasma is a superficial bacterial infection of the skin caused
can cause endocarditis. The diagnosis of localized erysipeloid is
by Corynebacterium minutissimum. It is characterized by asymp-
based on the patient’s history and clinical features.
tomatic, well‐demarcated, reddish brown, slightly scaly patches in

Key points
SECTION 7: BACTERIAL SKIN

• It can be differentiated from tinea infection by the characteristic


‘coral‐red’ fluorescence seen when viewed under Wood’s lamp
illumination.
Pitted keratolysis:
• Erythrasma may be treated with topical antibiotics. Oral
• Pitted keratolysis is a superficial bacterial infection that affects
antibiotic therapy is also very effective.
INFECTIONS

the plantar epidermis.


• The clinical diagnosis of pitted keratolysis can be made by the
crateriform pitting primarily affecting the pressure‐bearing Erysipeloid:
areas of the soles and the malodour. • Erysipeloid is an occupational, acute skin infection caused by
• Symptoms include hyperhidrosis, maceration and fetidness. Erysipelothrix rhusiopathiae.
• Multiple organisms have been isolated from these lesions. • It occurs primarily in individuals who handle animals and animal
• Therapy is simple and efficacious. Topical antibiotics such as products, especially fish, shellfish, meat and poultry.
erythromycin and clindamycin are safe and systemic treatment • It is characterized clinically by an erythematous oedema, with
with erythromycin has been successful. well‐defined and raised borders, usually localized to the back of
• Reducing hyperhidrosis also plays an important role in disease one hand and/or fingers.
management. • Diagnosis of localized erysipeloid is based on the patient’s
history (occupation, previous traumatic contact with infected
animals or animal products) and clinical features (typical skin
Erythrasma:
lesions, lack of severe systemic features and rapid remission
• Erythrasma is a superficial bacterial infection of the skin caused
after treatment with penicillin or cephalosporin).
by Corynebacterium minutissimum.
• It is characterized by asymptomatic, well‐demarcated, reddish
brown, slightly scaly patches in moist, occluded, intertriginous areas.

Pitted keratolysis bacterial infection of the plantar stratum corneum [1], often
diagnosed in barefooted individuals in tropical c­limates.
Brief introduction/history. Pitted keratolysis (PK), first Castellani [2] first reported this condition from Sri Lanka in
described as keratolysis plantare sulcatum (also known as 1920 and termed it ‘keratoma plantare sulcatum’. Although
keratolysis sulcata, ringed keratolysis and keratolysis plan­ initially thought to be a condition found only in tropical
tare sulcatum), is a descriptive title for a common s­ uperficial climates, it has also been described in temperate zones. The

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 39  Pitted Keratolysis, Erythrasma and Erysipeloid 457

term ‘pitted keratolysis’ was coined by Zaias et  al. [3] in seen more commonly in tropical areas. On the palms, the
1965 and is universally accepted. lesions may present as scaly collarettes instead of pits
[9,12]. In addition to bacterial infection, sweat retention
Epidemiology and  pathogenesis. PK is a condition that and water immersion are important cofactors for the
occurs worldwide and can be seen in both temperate and development of PK. Patients may experience hyperhidro­
tropical environments [4]. Prevalence rates of the disease sis, foot odour, maceration and sometimes itching or
range from 1.5% for Korean industrial workers to burning while walking, although most cases are asympto­
42.5%  for paddy field workers in South India [5,6]. PK matic. Painful lesions have been described with erythe­
is  common in physically active individuals because matous to violaceous tender plaques in ­children [9,13].
pathological microbes thrive in their warm, sweaty shoes. The exact reason for the pain in some patients is not
Hyperhidrosis of the feet brings a softening of stratum known. In one review of 53 patients with PK, hyperhidro­
corneum, thus facilitating invasion by the corynebacte­ sis was present in 51 patients (96.2%) and malodour in
rium. The aetiology of PK is attributed to a Gram‐positive 47 patients (88.7%) [14]. A mixture of thiols, sulphides and
bac­terial infection. It is characterized by superficial thio‐esters has been shown to be the cause of the unpleas­
erosions and crater‐like pits measuring 1–3 mm in diameter ant odour [14]. Yellowish‐brown discolouration of
which are located primarily along the sole. The causative involved skin has also been reported [15].
agents are  Gram‐positive bacteria, e.g. Corynebac­terium
spp., Kytococcus sedentarius, Dermatophilus congolensis and Histology and  microbiology. Histological examination of
Streptomyces spp. [7–9]. These microorganisms produce the lesion would show multiple superficial erosions
proteases that enable them to digest the keratin and ­confined to the upper layers of the stratum corneum. These
dissolve the stratum corneum, thereby producing the shallow, crater‐like defects have sharply inclined walls and
characteristic pits or craters [10]. Predisposing factors a flat base consisting of a thin layer of stratum corneum. The
include hyperhidrosis, prolonged use of occlusive foot­ pathogenic microorganisms penetrate the hydrated ­stratum
wear, increase in skin pH, humid climate, poor foot corneum. They are rarely seen with potassium hydroxide
hygiene, obesity and immunodeficiency. preparation but are more easily demonstrated on Gram‐
stained scrapings or thin tissue sections [15]. As PK is an
Clinical features. PK occurs in both sexes and in most age acquired, superficial bacterial infection of the skin, multiple
groups. The physical examination mostly reveals multi­ organisms have been isolated from these lesions, including
ple circular, discrete and coalescing superficial erosions various species of Streptomyces and Corynebacterium,

SECTION 7: BACTERIAL SKIN


on the soles and undersurface of the toes (Fig.  39.1). In Dermatophilus congolensis, Micrococcus ­sedentarius and
addition to the circular pits, some patients manifest small K.  ­sedentarius [16–19]. Specific proteases of K. sedentarius
stratum corneum defects along the lateral plantar surface. account for the degradation of corneal callus in PK [20].
The lesions primarily affect the weightbearing aspects of

INFECTIONS
the feet, such as the toes, balls of the feet and the heels. Diagnosis. The diagnosis can be made clinically [21]. PK
The skin‐coloured, punched‐out, shallow depressions is characterized by typical malodour and pits in the
measure 1–3 mm in diameter. However, involvement of hyperkeratotic areas of the soles. It is more common in
non‐weightbearing areas has been described. The areas barefooted individuals in tropical areas, or those who
involved were the instep of the foot and dorsal aspect of have to wear occlusive shoes, such as soldiers, sailors
the toes [11]. Palmar involvement rarely occurs and is and  athletes. PK is frequently accompanied by plantar

(a) (b)
Fig. 39.1  (a) Pitted keratolysis presenting as multiple crater‐like shallow plantar pits and a macerated area. (b) Dotted blue fluorescence in the sole area on
Wood’s light examination. Source: Courtesy of Dr Hongwei Wang from Huadong Hospital affiliated to Fudan University.
458 Section 7  Bacterial Skin Infections

­ yperhidrosis. The weightbearing metatarsal parts of the


h 3 Zaias N, Taplin D, Rebell G. Pitted keratolysis. Arch Dermatol
1965;92:151–4.
feet are mostly affected. Histological examination of the
4 Eetam I, Aytimur D, Yuksel SE. Isolation of Kytococcus sedentarius
lesion shows multiple superficial erosions confined to the from a case of pitted keratolysis. Ege Tip Dergisi 2005;44:117.
upper layers of the stratum corneum. The pathogenic 5 Eun HC, Park HB, Chun YH. Occupational pitted keratolysis. Contact
microorganisms can be cultured on an appropriate Dermatitis 1985;12:122.
6 Shenoi SD, Davis SV, Rao S et al. Dermatoses among paddy field work­
medium if specific identification is desired. ers–a descriptive, cross‐sectional pilot study. Indian J Dermatol
Venereol Leprol 2005;71:254–8.
Differential diagnosis. The main differential diagnoses 7 Takama H, Tamada Y, Yano K et  al. Pitted keratolysis: clinical
­manifestations in 53 cases. Br J Dermatol 1997;137:282–5.
include tinea pedis, verrucae, punctate palmoplantar ker­
8 Tamura B, Cuce LC, Souza RL et al. Plantar hyperhidrosis and pitted
atoderma type 1 and basal cell naevus syndrome [22]. A keratolysis treated with botulinum toxin injection. Dermatol Surg
potassium hydroxide stain and Wood’s light examination 2004;30:1510–14.
are valuable in differentiating tinea pedis or other 9 Bruckner DA, Colonna P, Bearson BL. Nomenclature for aerobic and
facultative bacteria. Clin Infect Dis 1999;29:713–23.
­bacterial infection, but both entities may be present simul­ 10 Fernandez‐Crehuet P, Ruiz‐Villaverde R. Pitted keratolysis: an
taneously [23]. Dermoscopy usually reveals abundant infective cause of foot odor. CMAJ 2015;187:519.
pits with well‐marked verrucae. Punctate palmoplantar 11 Takama H, Tamada Y, Yokochi K et al. Pitted keratolysis: a discussion
keratoderma type 1 (PPKP1, OMIM #148600) is a rare of two cases in non‐weight‐bearing areas. Acta Dermatol Venereol
1998;78:225–6.
form of palmoplantar keratoderma that is autosomal 12 Zaias N. Pitted and ringed keratolysis: a review and update. J Am
dominantly inherited [24]. PPKP1 is clinically character­ Acad Dermatol 1982;7:787–91.
ized by multiple punctate hyperkeratotic papules affect­ 13 Lamberg SI. Symptomatic pitted keratolysis. Arch Dermatol
1969;100:10–11.
ing the palmar and plantar skin. The lesions typically 14 Nordstrom KM, McGinley KS, Cappiello L et al. Pitted keratolysis: the
start to appear in early adolescence but sometimes role of Micrococcus sedentarius. Arch Dermatol 1987;123:1320–5.
develop later in life. Pitting associated with the basal cell 15 Sehgal VN, Ramesh V. Crateriform depression – an unusual clinical
naevus syndrome, which is an autosomal dominant syn­ expression of pitted keratolysis. Dermatologica 1983;166:209–11.
16 Gill KA, Buckels LJ. Pitted keratolysis. Arch Dermatol 1968;98:7–11.
drome that causes pitting of the palms and soles, multiple 17 Gillum RL, Qadri SM, Al‐Ahdal MN et al. Pitted keratolysis: a mani­
basal cell carcinomas, jaw cysts and skeletal abnormali­ festation of human dermatophilosis. Dermatologica 1988;177:305–8.
ties, can be distinguished by the specific clinical features 18 Woodgyer AJ, Baxter M, Rush‐Munro FM et  al. Isolation of
Dermatophilus congolensis from two New Zealand cases of pitted
seen in this syndrome [25].
keratolysis. Australas J Dermatol 1985;26:29–35.
19 Ertam I, Aytimur D, Yuksel SE. Isolation of Kytococcus sedentarius
Treatment and prevention. Therapy for PK is simple and from a case of pitted keratolysis. Ege Tip Dergisi 2005;44:117–18.
SECTION 7: BACTERIAL SKIN

efficacious. Treatment generally consists of a combination 20 Longshaw CM, Wright JD, Farrell AM et al. Kytococcus sedentarius,
the organism associated with pitted keratolysis, produces two kera­
of hygienic measures and antimicrobials. In the past, vari­ tin‐degrading enzymes. J Appl Microb 2002;93:810–16.
ous treatments (20% aluminium chloride, formalin oint­ 21 Martin AG, Kobayashi GS. Bacterial diseases with cutaneous involve­
ment, various topical antibiotics and imidazoles) were ment. In: Freedberg IM, Eisen AZ, Wolff K et al. (eds) Dermatology in
INFECTIONS

General Medicine, 5th edn. New York: McGraw Hill, 1999:2203–4.


used with some success. A variety of studies used a com­
22 Pranteda G, Carlesimo M, Pranteda G et  al. Pitted keratolysis,
bined approach in the treatment of the disease, consisting erythromycin, and hyperhidrosis. Dermatol Ther 2014;27:101–4.
of the management of provoking factors (such as hyperhi­ 23 Schlappner OL, Rosenblum GA, Rowden G. Concomitant erythrasma
drosis) and the management of the bacterial infection and dermatophytosis of the groin. Br J Dermatol 1979;100:147–51.
24 Pohler E, Mamai O, Hirst J et  al. Haploinsufficiency for AAGAB
[26]. Topical antibiotics such as sodium fusidate, clinda­ causes clinically heterozygous forms of palmoplantar keratoderma.
mycin and erythromycin are the first line of medical treat­ Nat Genet 2012;44:1272–6.
ment. Imidazole antifungal agents are also effective 25 Cohen PR. Axillary basal cell carcinoma in patients with Goltz‐Gorlin
because they have antibacterial activity. Mupirocin oint­ syndrome: report of basal cell carcinoma in both axilla of a woman
with basal cell nevus syndrome and literature review. Dermatol
ment has been found to be effective when conventional Online J 2014;17:20(8).
therapy has failed [27], but its use should be restricted to 26 Garcia GR, Nunez PY, Arrese EJ. Queratolisis punctata emergente en
elimination of meticillin‐resistant Staphylococcus aureus los Andes Cusco‐Peru. Med Cutan Iber Lat Am 2006;34:223.
27 Vazquez‐Lopez F, Perez‐Oliva N. Mupirocine ointment for sympto­
(MRSA). Systemic treatment with erythromycin is likely matic pitted keratolysis. Infection 1996;24:55.
to be effective, but it is only rarely required [28]. 28 Eetam I, Aytimur D, Yuksel SE. Isolation of Kytococcus sedentarius
Decreasing sweat production by topical aluminium from a case of pitted keratolysis. Ege Tip Dergisi 2005;44:117.
hydroxide should be considered, and even botulinum
toxin has been used in two patients with refractory PK
due to prominent hyperhidrosis. Tamura et  al. [8] per­ Erythrasma
formed subcutaneous injection of low‐dose botulinum
toxin into multiple sites in the plantar region to induce Brief introduction/history. Erythrasma is a superficial
anhidrosis; however, this therapy should only be consid­ bacterial infection of the skin caused by Corynebacterium
ered when traditional therapies have proved ineffective. minutissimum. The skin lesions show a characteristic
‘coral‐red’ fluorescence when viewed under Wood’s light.
Before 1960, the aetiological agent of erythrasma was
References presumed to be a fungal organism. Lagana [1] first iso­
1 Naik CL, Singh G. Clinicoepidemiological study of pitted keratolysis. lated a diphtheroid bacterium from an erythrasma lesion.
Indian J Dermatol 2007;52:35.
2 Castellani A. Keratoma plantare sulcatum. J Ceylon Br Med Assoc In 1961, Sarkany et al. [2] confirmed that C. minutissimum
1920;1:12–14. was the cause of erythrasma.
Chapter 39  Pitted Keratolysis, Erythrasma and Erysipeloid 459

Epidemiology and  pathogenesis. C. minutissimum, the


aetiological agent of erythrasma, is a short, Gram‐positive
rod with subterminal granules which often colonizes the
nose, pharynx, bulbar conjunctiva, ear canal and skin sur­
face. Erythrasma begins as a proliferation of C. minutissi-
mum within the stratum corneum. Organisms can be
found both extracellularly and intracellularly. Whether
the associated scaling precedes and allows the infection to
occur or is the result of heavy colonization of the stratum
corneum is still a matter of debate.
Predisposing factors include warm, humid climate,
poor hygiene, hyperhidrosis, obesity, diabetes mellitus,
advanced age and compromised host status. The infec­
tion is more common in tropical than in temperate cli­
mates. A disciform variant exists which occurs outside the
classically involved intertriginous areas. This generalized
Fig. 39.2  Erythrasma. Well‐demarcated brownish scaly plaque in bilateral
form is most commonly seen in diabetics. It can be the intracrural areas of the groin of an adolescent boy. Source: Courtesy of Dr
presentation of type 2 diabetes mellitus and can also be Hongwei Wang from Huadong Hospital affiliated to Fudan University.
secondary to tinea pedis [3].

Clinical features. Erythrasma occurs in all age groups and


in both sexes. It is much more common in adults than in
children [4] with around 15% of cases occurring in
­children between 5 and 14 years of age. The incidence is
notably higher among institutionalized children and
­children in boarding schools as well as people living a
communal lifestyle [5].
Erythrasma occurs most commonly in moist, occluded
intertriginous areas that favour the growth of C. minutis-

SECTION 7: BACTERIAL SKIN


simum. The common locations for erythrasma are the
axilla, groin, toe webs, intergluteal and crural folds and
inframammary areas. Occasionally, it occurs on mucous

INFECTIONS
membrane such as on the labia [6]. However, disciform
erythrasma, a more generalized form, occurs in noninter­
triginous areas as well as intertriginous areas and is more
commonly seen in the tropics [7]. Although most lesions Fig. 39.3  Coral fluorescence in areas of plaque on Wood’s light examina-
are asymptomatic, mild pruritus may be present. In tion. Source: Courtesy of Dr Hongwei Wang from Huadong Hospital
patients with pruritus, irritation of lesions by scratching affiliated to Fudan University.
may cause secondary changes of excoriations and
lichenification. ­ ifferentiating these entities. However, both erythrasma
d
The characteristic skin lesions are well‐demarcated, and tinea infection may be present simultaneously [10].
irregular, reddish‐brown scaly patches in the genitocru­ Tinea versicolor may resemble erythrasma but does not
ral, axillary and inframammary regions (Fig.  39.2). tend to localize to body folds. Similarly, erythrasma and
However, in the toe webs, erythrasma frequently presents tinea versicolor may coexist and become difficult to diag­
as asymptomatic, chronic maceration with fissuring or nose [4].The disciform type may be confused with lichen
scaling of interspaces or blisters [8]. Lesions of disciform sclerosis et atrophicus and plaque‐type parapsoriasis [7].
erythrasma may have a shiny, atrophic‐appearing surface It also can be in the differential diagnosis of vulvar
and may resemble lesions of parapsoriasis en plaque or mucosal rashes [6].
lichen sclerosis et atrophicus [9]. Mild, subclinical cases
are not uncommon and are diagnosed only by the typical Histology findings. The finding of bright coral‐red fluores­
fluorescence on Wood’s light examination (Fig. 39.3). cence under Wood’s light examination (the result of porphy­
rin production by the bacteria) is the best way to make the
Differential diagnosis. Erythrasma must be distinguished diagnosis. Gentle scraping of the lesions may yield scale that
from tinea cruris, intertrigo and seborrhoeic dermatitis can be stained with methylene blue or Gram stain. Coccoid
when it occurs in the genitocrural regions and from tinea and rod‐like organisms with long filaments will be seen
pedis or other bacterial infections when it occurs in the under the oil immersion lens. Corynebacterium organisms
interdigital areas. The main differentiating symptom of exhibiting coral‐red fluorescence can be cultured on tissue
erythrasma is its lack of pruritus. A potassium hydroxide culture medium, but ­culture is not routinely performed in
stain and Wood’s light examination are valuable in clinic. Skin biopsy is rarely indicated but may be p
­ erformed
460 Section 7  Bacterial Skin Infections

when the lesions are atypical in morphology and distribu­ Erysipeloid


tion, as may be seen with disciform erythrasma.
On histological examination, corynebacteria are seen in Brief introduction. Erysipeloid, named in the past as
the horny layer in small numbers as Gram‐positive rods Rosenbach disease, Baker–Rosenbach disease, pseudoery­
and filaments. Electron microscopic examination of thin‐ sipelas, erythema serpens, erythema migrans, erysipelas
sectioned skin from patients with erythrasma has also chronicum, swine erysipelas, diamond skin disease and
demonstrated ‘diphtheroids’ in the horny layer [2]. crab cellulitis [1–5], has been recognized since the late
1800s. It was initially described by Tilbury Fox, who
Treatment and prevention. Erythrasma responds quickly named the c­ ondition erythema serpens in 1873. In 1876
to treatment with a variety of topical preparations and to Robert Koch first isolated a strain of Erysipelothrix from a
topical and systemic antibiotics. The subclinical form mouse inoculated with putrefying blood and termed the
may remain asymptomatic for years or undergo periodic organism E. muriseptica. In 1884 Anton Rosenbach discov­
exacerbations. Relapses may occur even after successful ered an organism similar to Koch’s in a patient with local­
treatment. ized cutaneous lesions and named the condition
Multiple options are available for treating erythrasma, erysipeloid. In 1909 E. rhusiopathiae was isolated from
including oral and topical therapies [11]. The most effec­ cutaneous lesions of erysipeloid [6].
tive treatment is erythromycin (250 mg, four times daily
for 14 days) with cure rates (both clinical and bacte­
Epidemiology and  pathogenesis. Erysipeloid is most
riological) as high as 100% [12]. A single 1 g dose of
commonly seen in fishermen and meat handlers but can
clarithromycin has been reported to be effective as well
occur in anyone coming into contact with the appropriate
[13]. In patients with interdigital involvement or hidden
infectious agents. The incidence of this disease was about
reservoirs, some form of local therapy is recommended,
0.3% in the past [7], but only occasional sporadic cases are
such as clindamycin applied once daily during the course
reported now because of technological advances in ani­
of oral therapy and continued for 2 weeks after the phys­
mal‐handling industries. It is uncommon in the paediatric
ical clearance of these areas [12].
population.
Antibacterial soaps can be used initially, and prophy­
E. rhusiopathiae is a rod‐shaped, Gram‐positive, faculta­
lactically once the infection has cleared. Topical treat­
tive aerobic, nonmotile, nonsporulating bacterium. It is
ments with 10–20% aluminium chloride, 2% clindamycin
found worldwide in soil, contaminated water from infected
HCl solution, erythromycin, miconazole cream or
animals and food products. It can survive in soil for several
SECTION 7: BACTERIAL SKIN

Whitfield ointment have all been shown to be effective. It


weeks. In pig faeces, the survival period of this bacterium
was reported that photodynamic treatment can be used to
ranges from 1 to 5 months. E. rhusiopathiae produces neu­
treat erythrasma in adults [14].
raminidase and hyaluronidase, the latter contributing to its
INFECTIONS

capacity for dissemination within infected tissue.


References
Erysipeloid is transmitted by several animals, particularly
1 Lagana L. Contribution to the study of the pathogenic agent of swine (up to half of healthy pigs), fish, crab and shellfish.
erythrasma and its therapy (in Greek). Acta Microbiol Hellen E.  rhusiopathiae (previously named E. insidiosa) has been
1960;5:69–75. found in several dozen species of mammals and other ani­
2 Sarkany I, Taplin D, Blank H. Erythrasma—common bacterial infec­
tion of the skin. JAMA 1961;177:130–2. mals, such as sheep, rabbits, chickens, turkeys, ducks, emus,
3 Inci M, Serarslan G, Ozer B et al. The prevalence of interdigital eryth­ pigeons, cows, guinea pigs, cats, dogs [8] and kakapo [9].
rasma in southern region of Turkey. J Eur Acad Dermatol Venereol The bacterium penetrates the skin through tiny breaks.
2012;26:1372–6.
4 Karakatsanis G, Vakirlis E, Kastoridou C et al. Coexistence of pity-
The incubation period is 2–7 days. At present, strains of
riasis versicolor and erythrasma. Mycoses 2004;47:343–5. Erysipelothrix are classified as serotypes 1–26 with sero­
5 Somerville DA, Seville RH, Cunningham RC et  al. Erythrasma in a types 2, 7 and 16 being the most commonly involved in
hospital for the mentally subnormal. Br J Dermatol 1970;82:355–60. the pathogenesis of human erysipeloid [10].
6 Wilson BB, Wagenseller A, Noland MM. An atypical presentation
of erythrasma. J Am Acad Dermatol 2012;67:e217–18.
7 Gorani A, Oriani A, Falconi Klein E et  al. Erythrasmoid pityriasis
Clinical features. Three forms of human disease have
­versicolor. Mycoses 2001;44:516–17.
8 Sariguzel FM, Koc AN, Yagmur G et  al. Interdigital foot infections: been described.
Corynebacterium minutissimum and agents of superficial mycoses. The first is the localized cutaneous form (‘true’ erysip­
Braz J Microbiol 2014;45:781–4. eloid). It initially begins on the dorsal hands or fingers as
9 Tchen JA, Ramsdell WM. Disciform erythrasma. Cutis 1983;31:541–7.
10 Morales‐Trujillo ML, Arenas R, Arroyo S. Interdigital erythrasma:
an inflammatory plaque with well‐defined raised borders
clinical, epidemiologic, and microbiologic findings. Actas (Fig. 39.4). However, lesions can occur elsewhere and have
Dermosifiliogr 2008;99:469–73. been reported on the palms, forearms, arms, face and legs
11 Chodkiewicz HM, Cohen PR. Erythrasma: successful treatment after [11–13]. The colour is characteristically bright red to pur­
single‐dose clarithromycin. Int J Dermatol 2013;52:516–18.
12 Holdiness MR. Erythrasma and common bacterial skin infections. ple and lesions may assume a ‘diamond’ shape. Vesicular,
Am Fam Physician 2003;67:254. bullous and erosive lesions have also been described [14].
13 Wharton JR, Wilson PL, Kincannon JM. Erythrasma treated with sin­ The entire lesion tends to spread centrifugally. Swelling of
gle‐dose clarithromycin. Arch Dermatol 1998;134:671–2.
14 Darras‐Vercambre S, Carpentier O, Vincent P et  al. Photodynamic
the fingers is often substantial, leading to the common
action of red light for treatment of erythrasma: preliminary results. terms ‘whale finger’ and ‘seal finger’. The lesion may be
Photodermatol Photoimmunol Photomed 2006;22:153–6. slightly pruritic or painful. Fever, arthralgia, lymphadenitis
Chapter 39  Pitted Keratolysis, Erythrasma and Erysipeloid 461

(a) (b)
Fig. 39.4  (a and b) Erysipeloid characterized by marked erythematous oedema on the right hand of a woman. Source: Courtesy of Dr Hongwei Wang from
Huadong Hospital affiliated to Fudan University.

and lymphangitis may occur [15]. Erysipeloid is often self‐ Polymerase chain reaction (PCR) assays have recently
limited and resolves spontaneously. been described for the diagnosis of human erysipeloid.
The diffuse cutaneous or generalized form is much Their specificity and sensitivity seem to be high, although
rarer than the cutaneous form. It is characterized clini­ only a few cases of human erysipeloid infection have
cally by multiple lesions with central clearing of the skin. been tested to date [17].
The presence of systemic symptoms is characteristic,
including fever, lymphadenitis, arthralgia and myalgia.
Diagnosis. The diagnosis of erysipeloid is based on: (i)
This form of the disease is also self‐limited, but the ­clinical
the patient’s occupation; (ii) previous traumatic contact

SECTION 7: BACTERIAL SKIN


course is longer and recurrences more common.
with infected animals or their products; (iii) typical
The most serious manifestation of erysipeloid is the
skin lesions (erythematous oedema with well‐defined
systemic or ‘septic’ form. Cutaneous lesions may be
and raised b ­orders, or vesicular–bullous–erosive
found consisting of red papules, some of which exhibit

INFECTIONS
lesions on an erythematous base, usually localized to
necrosis and ulceration. Systemic clinical manifestations
the back of one hand and/or fingers); (iv) lack of severe
may include septic arthritis, osseous necrosis, cerebral
systemic features; (v)  slight laboratory abnormalities;
infarction or abscess, pulmonary effusion, encephalitis,
and (vi) rapid remission after treatment with penicillin
meningitis, renal failure, peritonitis and bacterial endo­
or cephalosporin.
carditis which may be acute or subacute [16]. Morbidity
and mortality (38%) are greatly increased in those who
manifest endocarditis. Differential diagnosis. The clinical differential diagnosis
includes erysipelas, cellulitis and acute irritant/allergic
Laboratory findings. Serological investigations may reveal contact dermatitis. Erysipelas is characterized clinically
leucocytosis, slightly increased serum gammaglobulins by the acute appearance of erythematous and oedema­
and an increase in inflammatory markers (erythrocyte tous lesions, usually localized on the face or legs, often
­sedimentation rate, C‐reactive protein and α‐1 acid glyco­ accompanied by pain, fever and general malaise.
protein). Anti‐E.  ­rhusiopathiae antibodies can also be Cellulitis is more common on the face and trunk with
detected by agar gel precipitation test [10]. Histopathological painful ­diffuse swelling on the skin accompanied by
findings are nonspecific. Varying degrees of spongiosis severe systemic symptoms such as high fever, chills and
and intraepidermal vesiculation have been shown in the rigors and generalized malaise. Bacterial culture with
upper and mid dermis; a polymorphous infiltrate of Gram staining can help identify the pathogen and con­
­neutrophils, lymphocytes, ­eosinophils and plasma cells is firm the clinical diagnosis. Irritant and allergic contact
seen in the dermis. Gram staining of tissue sections usually dermatitis often occur on both hands as erythematous,
does not demonstrate the organism, possibly because the vesicular and ­pruritic lesions. The clinical course is often
organism is believed to reside in the reticular dermis and chronic and recurrent.
may be missed if skin biopsy is performed superficially. In addition to these conditions, atypical cutaneous
Another possibility is that the bacteria have lost their cell leishmaniasis should be considered by clinicians in the
walls, reverting to ‘L’ forms. differential diagnosis because histopathology and culture
Isolation of E. rhusiopathiae is very often difficult. may be negative. However a smear from the lesion show­
E. ­rhusiopathiae grows slowly in media enriched with blood ing a number of amastigotes in the intracellular and extra­
and incubated in an atmosphere containing 5–10% CO2. cellular area can identify the disease [18].
462 Section 7  Bacterial Skin Infections

4 Varella TC, Nico MM. Erysipeloid. Int J Dermatol 2005;44:497–8.


Treatment and prevention. E. rhusiopathiae is sensitive to 5 Veraldi S, Girgenti V, Dassoni F et  al. Erysipeloid: a review. Clin
a number of common a­ ntibiotics, including penicillins, Exp Dermatol 2009;34:859–62.
6 Shin SJ, Gwak WG. Erysipelothrix rhusiopathiae peritonitis in
cephalosporins (cefotaxime, ceftriaxone), tetracyclines
a  patient undergoing continuous ambulatory peritoneal dialysis.
(chlortetracycline, oxytetracycline), quinolones (cipro­ J Korean Med Sci 2010;25:1234–6.
floxacin, pefloxacin), clindamycin, erythromycin, linco­ 7 Parsons DW. Treatment of the septic hand. Proc R Soc Med
mycin, imipenem and piperacillin. It is resistant to 1962;55:445–7.
8 Brooke CJ, Riley TV. Erysipelothrix rhusiopathiae: bacteriology,
vancomycin, chloramphenicol, daptomycin, gentamicin, epidemiology and clinical manifestations of an occupational
netilmicin, polymyxin B, streptomycin, teicoplanin and ­pathogen. J Med Microbiol 1999;48:789–99.
trimethoprim–sulfamethoxazole [19]. 9 Werner K, Gartrell B, Norton SA. Erysipeloid (Erysipelothrix rhusi­
Although the cutaneous forms of the disease are self‐lim­ opathiae infection) acquired from a dead kakapo. Arch Dermatol
2011;147:1456–8.
ited in many instances, all patients should receive antibiot­ 10 Molin G, Söderlind O, Ursing J et  al. Occurrence of Erysipelothrix
ics to prevent progression to systemic disease and rhusiopathiae on pork and in pig slurry, and the distribution of spe­
endocarditis. Penicillins and cephalosporins are the first cific antibodies in abattoir workers. J Appl Bacteriol 1989;67:347–52.
11 Veraldi S, Rizzitelli G, Schianchi‐Veraldi R. Occupational cutaneous
line of treatment. A 7‐day course is appropriate, and clinical infections. Clin Dermatol 1992;10:225–30.
improvement is usually observed 2–3 days after the begin­ 12 Razsi L, Sanchez MR. Progressively enlarging painful annular plaque
ning of the treatment. Incision and drainage of skin lesions on the hand. Arch Dermatol 1994;130:1314–15.
is contraindicated because surgery has been noted to pro­ 13 Hamilton JF. Recurrent erysipeloid cellulitis of the lower extremities;
with case reports. South Med J 1954;47:778–83.
long the duration of the erysipeloid lesions [20]. Direct skin 14 Connor MP, Green AD. Erysipeloid infection in a sheep farmer with
contact with pork and fish and other aquatic products coexisting orf. J Infect 1995;30:161–3.
should be avoided to prevent accidental p ­ uncture of the 15 Nelson E. 500 cases of erysipeloid. Rocky Mt Med J 1955;52:40–2.
16 Simerkoff MS, Rahal JJ Jr. Acute and subacute endocarditis due to
skin. Improved hygiene during meat ­processing and aqua­
Erysipelothrix rhusiopathiae. Am J Med Sci 1973;266:53–7.
culture is also helpful to decrease erysipeloid prevalence. 17 Brooke CJ, McLaughlin V, Mee BJ, Riley TV: An investigation of ‘cray­
fish poisoning’ in Western Australia. Med J Aust 1999;170:288.
References 18 Mnejja M, Hammami B, Chakroun A et al. Unusual form of cutaneous
1 Gilchrist TC. Erysipeloid with a record of 329 cases of which 323 were leishmaniasis: erysipeloid form. Eur Ann Otorhinolaryngol Head
caused by crab bites or lesion produced by crabs. J Cutan Dis Neck Dis 2011;128:95–7.
1904;22:507–19. 19 Boyd AS, Ritchie C, Fenton JS. Cutaneous Erysipelothrix rhusi­
2 Klauder JV, Righter LL, Harkins MJ. A distinctive and severe form of opathiae (erysipeloid) infection in an immunocompromised child.
erysipeloid among fish handlers. Arch Dermatol 1926;14:667–78. Pediatr Dermatol 2014;31:232–5.
3 Barnett JH, Estes SA, Wirman JA et al. Erysipeloid. J Am Acad Dermatol 20 Lamphier TA. Erysipeloid infection of digits. J Fla Med Assoc
SECTION 7: BACTERIAL SKIN

1983;9:116–23. 1971;58:39–41.
INFECTIONS
 463

C HA PTER   40

Lyme Borreliosis
Susan O’Connell
Formerly Lyme Borreliosis Unit, Health Protection Agency Microbiology Laboratory, Southampton University Hospitals NHS Trust, Southampton, UK

Abstract
manifestation, mainly in older people. Borreliae can disseminate
Lyme borreliosis is caused by several Borrelia burgdorferi geno- and ­affect other tissues. The most common paediatric neurologi-
species transmitted by Ixodes (hard‐bodied) ticks. Common tick cal presentations are facial and/or other cranial nerve palsies and
bite sites include skinfolds and scalp. Erythema migrans, a rash ­viral‐like meningitis. Painful radiculopathy is common in adults.
spreading from a tick attachment site, is the most common pres- Encephalomyelitis is a rare severe presentation. Lyme arthritis
entation, occurring in 80–90% of patients studied prospectively. ­affects large joints, particularly the knee, causing large effusions.
Myalgias and arthralgias can also occur. Borrelial lymphocytoma Carditis is uncommon, usually presenting with heart block. All in-
is an ­uncommon early feature, usually located on earlobe, nip- fection stages respond to antibiotics but clinical recovery can be
ple or scrotum. Acrodermatitis chronica atrophicans is a rare late incomplete if severe damage had occurred prior to treatment.

Key points • Antibody response to Borrelia burgdorferi infection takes


several weeks to develop. Antibody tests are not indicated
• Erythema migrans is the most common clinical presentation of routinely for the diagnosis of erythema migrans but should be
Lyme borreliosis. used to confirm later‐stage presentations.
• Erythema migrans rashes can vary in appearance, related to • Long‐term outcomes of patients appropriately treated in early
infecting genospecies and duration of infection. Lyme borreliosis are excellent. Oral antibiotics (usually amoxicillin
• A history of tick exposure rather than specific tick bite should or doxycycline) are recommended as first‐line agents for most
be sought, as ticks are tiny and easily overlooked, especially if clinical presentations. Recovery may be slow or incomplete in
attached in skinfolds or on the back or scalp. some patients with later‐stage disease.

SECTION 7: BACTERIAL SKIN


Definition and cause. Lyme borreliosis (Lyme disease) is which was isolated initially from Ixodes scapularis ticks

INFECTIONS
a multisystem spirochaetal infection caused by Borrelia (deer ticks) and then from skin biopsies, blood and cere-
burgdorferi sensu lato, transmitted by hard‐bodied ticks of brospinal fluid (CSF) of affected patients [2].
the Ixodes ricinus complex. Similar rashes following tick bites, termed erythema
migrans or erythema chronicum migrans, had been
History. The term Lyme arthritis was used initially in the ­recognized in European countries for many years, and
mid‐1970s, after a cluster of cases of juvenile arthritis ­clinicians had noted associations of tick bites with neuro-
occurred in the area of Old Lyme and neighbouring com- logical presentations such as facial palsy, meningitis and
munities in rural Connecticut. Two mothers became con- radiculoneuritis. Many physicians suspected an infec-
cerned about the extremely high rate of ‘juvenile tious cause, and some had used empirical penicillin
rheumatoid arthritis’ diagnosed in the locality and alerted ­treatment, with good results [3].
the local public health authorities, prompting investiga- Another skin condition, acrodermatitis chronica
tions by ­clinicians and scientists from Yale University [1]. atrophicans, had been described by German, Swedish
They noted that the arthritis had been preceded by tick and Austrian clinicians from the late nineteenth century
bites and slowly spreading erythematous skin lesions in a onwards, and was also found to be responsive to empiri-
significant ­number of patients, thus raising suspicions of cal penicillin treatment. In the early 1980s Swedish
a tick‐­transmitted infection. It also became apparent that researchers, using culture media developed by the
the ­disease could affect other organs and systems includ- American workers, established that B. burgdorferi was the
ing the nervous system and the heart. The term Lyme cause of acrodermatitis chronica atrophicans, erythema
­disease was used to denote this expanded clinical spec- migrans and a third skin manifestation also seen in
trum. The causative organism was a previously unknown Europe, borrelial lymphocytoma. The disease is now gen-
spirochaete, subsequently named Borrelia burgdorferi, erally known in Europe as Lyme borreliosis [2,3].

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
464 Section 7  Bacterial Skin Infections

continents there is considerable variation in incidence,


Epidemiology and ecological factors. Lyme borreliosis
with focal areas of hyper‐endemicity.
(LB) is the most common arthropod‐borne infectious
Hard‐bodied ticks of the Ixodes ricinus complex are the
­disease in temperate areas of the northern hemisphere
only vectors for Lyme borreliosis. In North America the
[2,4]. Its incidence is strongly dependent on ecological
infection is transmitted by the black‐legged or deer tick
and climatic factors affecting vector ticks and the small
I. scapularis (formerly called I. dammini) and I. pacificus in
mammals and birds that are reservoir hosts for B. burg-
the Pacific coastal states [2,5]. The main European vector is
dorferi, and on human behavioural factors leading to risk
I. ricinus (commonly called the deer, sheep or castor bean
of tick bite. In North America the regions of highest LB
tick) [2,10]. In Russia and the Baltic republics the range of
incidence are north‐eastern and mid‐Atlantic seaboard
I. ricinus overlaps with that of another vector, I. persulcatus
states, from Maine to Virginia, the upper mid‐west states
(the taiga tick), which is also widespread throughout tem-
of Wisconsin and Minnesota, and some coastal areas of
perate Asia to the Far East, including Japan.
northern California. About 37 000 cases were reported in
Ixodid ticks are very susceptible to desiccation and
2015, but the true incidence may be over 300 000, based
require a high relative humidity [10]. The most favourable
on data obtained from commercial laboratories and
tick habitats are forested areas and heathland where leaf
medical insurance claims  [5]. Reported incidence in
litter and undergrowth provide protection against dry-
Canada is low but increasing, with 315 cases in 2012, and
ing. These habitats also support animals and birds that
environmental studies indicate an expanding geograph-
are the natural feeding hosts for ticks [2,10]. Ticks have a
ical range [6]. In Europe the infection occurs in wood-
three‐stage lifecycle (larva, nymph and adult), usually
land and forested regions mainly between latitudes
over 2–3 years (Fig. 40.1). At each stage they take a single
62° and 42° North (southern Scandinavia to northern
blood meal lasting from 3 to 7 days. The common feeding
Mediterranean countries), with an increasing incidence
hosts for larvae and nymphs are small and medium‐sized
from west to east. There is no standardized case‐report-
mammals such as field mice, voles, squirrels and hares,
ing system for Europe, but a review of numerous data
and ground‐feeding birds including blackbirds, robins
sources suggests that there are over 200 000 European
and pheasants. These creatures are reservoir‐competent
cases annually [4,7]. Prospective studies in several high‐
hosts for B. burgdorferi. Ticks can become infected during
endemic areas showed that about 90% are likely to be
the course of a blood meal taken from a spirochaetaemic
erythema migrans [8,9]. Within endemic regions of both
SECTION 7: BACTERIAL SKIN

Larva feeds
on host 1
Larvae seek
new host
INFECTIONS

Fully fed larva


Eggs hatch drops to ground
to larva

Host 1
Eggs laid
by female Larva moults
to nymph

Fully fed female Life cycle


drops from host of the deer tick
to ground (Ixodes ricinus)

Host 3 Host 2

Female attaches Nymph attaches


and feeds on and feeds on
host 3 host 2

Nymph moults
to adult
Fig. 40.1  Lifecycle of Ixodes ricinus ticks (after Professor Jeremy Gray and Mr Bernard Kaye). The relative size of the animals approximates their significance
as hosts for the different lifecycle stages in a typical woodland habitat.
Chapter 40  Lyme Borreliosis 465

animal and in turn can transmit infection to hosts during Male : female incidence ratios seem to be about equal or
feeds at later stages in their lifecycle, thus maintaining the with a slight preponderance of cases in males in some
spirochaetal reservoir in nature [2]. Spring and early studies [2,5,8,9]. The major risk factors for infection are
­summer is the peak period for tick feeding and there can residential or recreational activities in tick habitats.
be a secondary peak during autumn. There can be a low
level of tick feeding activity on mild winter days in some
regions, resulting in occasional cases of erythema migrans Causative organisms and  pathogenetic factors. At least
occurring outside the peak periods, as demonstrated by 20 genospecies of B. burgdorferi sensu lato have been identi-
the case illustrated in Fig. 40.3. This child had sustained a fied; the majority are not pathogenic. Until recently the only
tick bite on her scalp in late December. pathogenic genospecies found in North America was
Deer are strongly associated with the presence of ticks. B.  burgdorferi sensu stricto, however another genospecies,
They are preferred feeding hosts for adult female ticks, B.  mayonii, has been identified in a few cases of LB with
who mate after feeding, drop back into the undergrowth unusually high spirochaetaemia [12]. A wider range of
and lay between 1000 and 2000 eggs before dying. Deer pathogenic genospecies is found in Europe, p ­ redominantly
are important in the maintenance and expansion of tick B. garinii and B. afzelii, but B. burgdorferi sensu stricto occurs
populations because of their feeding role at the tick’s in some areas [7]. All pathogenic strains can cause erythema
reproductive stage but they are not competent reservoir migrans, the early skin lesion. There is evidence for some
hosts for B. burgdorferi. Increased numbers and geograph- genospecies‐dependent variations associated with later
ical ranges of deer in some regions of Europe and North manifestations and for within‐­ genospecies variation in
America have been linked to significant increases in tick pathogenicity [7,13]. B. burgdorferi sensu stricto is strongly
populations and Lyme borreliosis incidence. linked with arthritic and neurological complications,
Human beings can be incidental feeding hosts for ticks. B.  ­garinii and B. bavariensis with neuroborreliosis, and
Larval ticks pose minimal risk for borrelial transmission B.  afzelii with acrodermatitis chronica atrophicans (ACA).
as they are rarely, if ever, infected transovarially. Nymphal Occasional cases of erythema migrans are caused by
ticks are most likely to transmit infection to people, as B. ­spielmanii and there have been only rare reports of disease
they are very small (about the size of a poppy seed) and linked to B. valaisiana and B. lusitaniae. Genospecies differ-
may not be noticed, even after feeding. Their major feed- ences also have implications for vaccine development.
ing period is during late spring and early summer, when Variations in the geographical distribution of European
human outdoor recreational activity is also likely to be at borrelial genospecies affect the incidence and types of

SECTION 7: BACTERIAL SKIN


its peak. The larger adult ticks are more likely to be spot- clinical presentations of later disease seen in different
ted and removed before completing their feeds (Fig. 40.2). locations. For example, ACA occurs more frequently in
Most ticks do not carry borreliae. Infected I. ricinus ticks Scandinavia and central Europe, where B. afzelii is com-
are unlikely to transmit infection in the first 18 hours of feed- monly found. The condition is infrequently seen in the

INFECTIONS
ing and I. scapularis ticks within the first 36 hours, but trans- UK, where B. afzelii is less common. The most commonly
mission risk rises steadily as feeding continues. Early identified pathogenic genospecies in the UK is B. garinii,
removal of attached ticks greatly reduces human infection and acute neuroborreliosis is the main complication seen
risk [7,11]. There is some evidence that transmission may there. A high proportion of infected ticks in the UK carry
take place at an earlier stage during I. persulcatus blood meals. B. valaisiana, which is essentially nonpathogenic. This
Most cases of Lyme borreliosis are reported in the sum- may have some bearing on the lower overall incidence of
mer months, between May and September, but some clinically significant disease there compared to other
patients with disseminated and late presentations are parts of Europe where a higher proportion of infected
diagnosed at other times of the year. In many studies ticks carry more pathogenic genospecies.
there is a bimodal age distribution, with higher incidences B. burgdorferi is an obligate parasite with limited bio-
in the 5–19 year age group and in people over age 40. synthetic ability, relying on its host for many nutrients [2].
It does not possess the virulence factors such as toxins,
lipopolysaccharides and enzymes that are associated
with many bacterial pathogens [2]. Borreliae are highly
motile and can disseminate through tissues and bind
closely to host cell surfaces, affecting their function [2,14].
Borreliae must adapt to life in very different environ-
ments: the midgut of the tick vector at ambient tempera-
ture and the mammalian or avian natural reservoir host
from about 35 ° to 39 °C. They vary in gene expression,
leading to production of different protein components.
Some help the organisms to evade the initial defences of
the host’s innate immune system, others contribute to
evasion of the subsequent acquired immune response, at
least for some time. For example, borreliae express an
outer surface protein (Osp), called OspA within the tick,
but OspC is preferentially expressed during the early
Fig. 40.2  Ixodid tick on nail of little finger. stages of mammalian infection. Expression of another
466 Section 7  Bacterial Skin Infections

protein, VlsE, which has an elaborate system for extensive


variation over time, is required to maintain persistent
infection. Synthesis of VlsE starts at about the time that
OspC production ceases and helps the organism to evade
the host’s humoral immune response, at least for some
time [2,14].
Many clinical manifestations of Lyme borreliosis occur
largely as a consequence of the human immunopatholog-
ical response to infection. Some borrelial lipoproteins can
activate a variety of cells, including macrophages, B cells,
dendritic cells and endothelial cells, triggering inflamma-
tory responses that contribute to pathogenesis [2,7,13,15].
There is also some in vitro evidence that antibodies
­produced in response to B. burgdorferi infection may also
bind to human neural and other antigens [14].
Ixodid tick bites
Ixodid ticks attach to their hosts by barbed mouth parts.
Their saliva contains anaesthetic, anti‐inflammatory and
anticoagulant substances that allow them to attach and
complete their feed without their hosts becoming aware
of their presence [16]. Borreliae can bind immunosup-
pressive tick salivary proteins to their surfaces which can
aid the establishment of infection in the skin [2,7]. The
bites are not usually significantly painful or itchy,
although some people who have had many bites over a
prolonged period, such as forestry workers, can become
sensitized to tick saliva and develop itchy skin reactions.
This can provide some protection against B. burgdorferi
infection, by drawing early attention to the tick before Fig. 40.3  Erythema migrans in a child.
SECTION 7: BACTERIAL SKIN

there is significant risk of spirochaetal transmission.


tick attachment and undisturbed blood meals which can
Clinical features. result in borrelial transmission include skinfolds (e.g.
INFECTIONS

B. burgdorferi infection can be asymptomatic or mini- groins, armpits, backs of knees, waistband area, under
mally symptomatic, as shown by prospective studies in breasts), back and scalp. Tick bites are more frequent
areas of high prevalence [8,9,17,18]. Clinically significant around the head and neck areas (particularly the scalp) of
disease has been customarily divided into three children than adults.
stages  –  early localized, early disseminated and late Most EM rashes become evident within about 5–14
Lyme ­borreliosis – but the process should be regarded as days (range 3–30 days) of a bite. They are usually round
a continuing pathological evolution in some untreated or oval but some can have a more triangular or linear
patients rather than having distinct phases or processes. appearance, presumably determined in part by lines of
Progression to later‐stage disease is not inevitable. The skin tension. A central punctum may be present at the tick
most frequently affected tissues and organs are the skin, attachment site. The leading edge can have a stronger col-
nervous system and joints. Case definitions have been our, giving the appearance of an outer ring or border, but
published, primarily for epidemiological purposes in the is not significantly raised [7,21]. They are not usually par-
USA and for clinical use in Europe [19,20]. ticularly pruritic or painful [21]. Very itchy or painful
lesions should raise suspicion of other conditions, includ-
Skin manifestations ing severe insect bite reactions or pyogenic infections.
Erythema migrans (Figs 40.3, 40.4) Most EM rashes are homogeneous in the early stages, but
Erythema migrans (EM) is the most characteristic presen- with longer duration some can develop an area of central
tation of Lyme borreliosis, occurring in about 90% of clearing, giving a so‐called ‘bull’s‐eye’ or target‐like
symptomatic infections [8,9,21,22]. It is an erythematous, appearance. Very pale lesions may be seen more easily
usually annular lesion spreading slowly from the site of a when the skin is warm, e.g. after exercise or bathing. If left
tick bite (often unnoticed) that had occurred 2 or more untreated, EM rashes eventually resolve over weeks to
days previously [7]. This lag phase occurs because spiro- months, and usually clear within a few days of commenc-
chaetal replication is relatively slow by comparison to ing appropriate treatment.
pyogenic bacteria. Localized redness appearing within a There can be variations in EM appearance, related to
few hours of a tick bite is likely to be caused by an imme- the infecting genospecies. In North American‐acquired
diate inflammatory response or hypersensitivity reaction infections, caused by B. burgdorferi sensu stricto, rashes
and usually fades within several days. Common sites for tend to evolve more quickly, are less likely to have central
Chapter 40  Lyme Borreliosis 467

Fig. 40.4  Erythema migrans in adults. Source: Courtesy of the late Dr John White.

clearing than European‐acquired infections caused by


B. afzelii, and are more likely to be accompanied by sys-
temic symptoms and signs including malaise, myalgia,
arthralgia, headache, fever and regional lymphadenopa-

SECTION 7: BACTERIAL SKIN


thy [23]. Some have central vesiculation, which can cause
difficulties in differentiation from cellulitis, arthropod
bites or herpesvirus infections [24]. Some EM lesions
caused by B. afzelii develop slowly, with little systemic

INFECTIONS
upset, and can reach large sizes with high rates of central
clearing [23]. Infections caused by B. garinii seem to be
more virulent than those caused by B. afzelii, with more
homogeneous and faster evolving rashes and a higher
likelihood of systemic symptoms and signs [25].
Multiple erythema migrans can occur, principally in
infections caused by B. burgdorferi sensu stricto. Affected
patients have multiple smaller lesions resulting from
haematogenous spread of spirochaetes from the initial Fig. 40.5  Borrelial lymphocytoma.
lesion. They usually have significant systemic symptoms
and may also have other objective extracutaneous mani- European Lyme borreliosis, mainly associated with B afzelii
festations of Lyme borreliosis, which include facial nerve infection; only rare cases have been reported in the USA.
palsy, meningitis, carditis and arthritis [26]. It  may develop several weeks to months after a tick bite
Differential diagnosis of EM includes reactions to tick and occurs more frequently in children than adults [7,20]. It
bites or other arthropod bites, urticaria, cellulitis, granuloma presents as a bluish‐red tumour‐like skin infiltrate most
annulare, ringworm, herpes simplex or zoster, fixed drug commonly on the earlobe, ear helix, nipple or scrotum
eruptions or contact dermatitis [7,23,27]. The histological [7,30–32]. If untreated it can persist for months but usually
appearance of erythema migrans is characterized by patchy resolves over some weeks following antibiotic treatment.
perivascular infiltrates, predominantly lymphocytic, with Histological examination shows dense ­lymphocytic and
plasma cells, mast cells and occasionally eosinophils, mainly histiocytic infiltrates, frequently with germinal centres [33].
affecting the superficial dermis. Immunohistochemical Borrelial lymphocytoma lesions have occasionally been
staining may demonstrate occasional spirochaetes, but misdiagnosed as B‐cell lymphomas.
silver staining is prone to artefact [28,29].
Acrodermatitis chronica atrophicans (Fig. 40.6)
Borrelial lymphocytoma (Fig. 40.5) Acrodermatitis chronica atrophicans (ACA) is an uncom-
Borrelial lymphocytoma (also termed lymphadenosis mon manifestation of longstanding infection, seen almost
benigna cutis) is an uncommon early manifestation of exclusively in European‐acquired infections. It occurs
468 Section 7  Bacterial Skin Infections

Nervous system manifestations


Neuroborreliosis is a common complication of European‐
and North American‐acquired Lyme borreliosis, mainly
presenting within a few weeks to months of infection, in
about 10% and 20% of previously untreated patients,
often with concurrent or recent EM. It can affect both
peripheral and central nervous systems. The most com-
mon manifestation in children is isolated facial nerve
palsy without clinical signs of meningitis, although CSF
examinations of some patients show mononuclear pleio-
cytosis [39,40]. It has been postulated that the higher rate
of tick bites occurring on the head and neck region of
young children causes a relatively higher incidence of facial
palsies in children than in adults [41]. Other ­neurological
Fig. 40.6  Acrodermatitis chronica atrophicans. Source: Courtesy of the presentations in children include ‘viral‐like’ meningitis
late Dr John White. with or without facial or other cranial nerve palsies, includ-
ing sixth nerve palsy [39,40]. Rare cases have been reported
mainly in older people, more frequently in women, with of children with previously untreated Lyme neuroborrelio-
only rare reports of ACA‐like lesions in children [7,22,34]. sis presenting with raised intracranial pressure and very
Almost all cases are caused by B. afzelii infection. ACA high CSF protein levels, leading to sight‐threatening
usually occurs on extensor sites of the extremities, most complications [40,42]. Other uncommon complications
­
commonly on the lower legs, causing localized livid red include meningoencephalitis and encephalopathy.
colour changes, doughy swelling and induration of the Garin–Bujadoux–Bannwarth (GBB) syndrome is the
affected skin. Untreated lesions may progress over years, most common neurological complication in adults.
with development of hyperpigmentation and atrophy, Features include facial palsy, meningitis and radiculoneu-
giving a cigarette paper‐like appearance with loss of ritis, usually presenting as a painful radiculopathy simi-
body hair, connective tissue and fatty tissue [7,35]. There lar to that of shingles or mechanical radiculopathy. It was
may be an associated peripheral neuropathy, with numb- originally described by French physicians in 1922 and a
ness, paraesthesias and allodynia [7]. If severe, this can German neurologist in 1941 [43,44]. A more indolent pres-
SECTION 7: BACTERIAL SKIN

lead to joint damage secondary to the neurological defi- entation of radiculopathy occurs in some European
cit. The differential diagnosis depends on the duration of patients, mainly in older age groups, who present with a
the ­condition and includes vascular insufficiency, acro- gradual onset of pain increasing over months after
cyanosis, livedo reticularis, lymphoedema or chilblains. ­infection acquisition. This may result from direct spiro-
INFECTIONS

The infection responds to treatment but completeness of chaetal invasion of peripheral nerves from the initial skin
clinical recovery depends on the degree of underlying inoculation site, gradually extending to the nerve roots,
tissue damage. Histological appearance depends on the in  contrast to the more rapid onset and wider effects
duration of the lesion, with earlier inflammatory lesions seen in classic GBB syndrome, resulting from haematog-
showing perivascular infiltrates of lymphocytes and enous spread [14]. Some patients may retrospectively
plasma cells, dermal oedema and telangiectasia. recall a preceding EM. In both situations radicular pain
Epidermal atrophy and loss of epidermal appendages can be very severe and patients may require opiates, but
are characteristic findings in late‐stage ACA [29,35]. analgesia requirements usually reduce very significantly
within a short time of commencing antibiotic treatment.
Other skin manifestations Painful radiculopathy is rare in children.
Several other skin conditions, including morphoea and Meningoencephalitis is an uncommon complication in
lichen sclerosis et atrophicus, have been linked to both children and adults. Encephalomyelitis is rare,
B.  ­burgdorferi infection in a few case reports and small mainly seen in older adults. It appears to affect white
­studies, predominantly from high‐endemic areas of Europe. matter disproportionately and its clinical presentation
­
Other European and North American studies have shown may be confused with multiple sclerosis or, if particularly
no link with these conditions. These mainly early reports acute, with acute disseminated encephalomyelitis.
should be interpreted cautiously, as the laboratory tests Virtually all patients have an inflammatory CSF and
used in support of some cases relied on methods such as intrathecal production of B. burgdorferi antibodies, indi-
silver staining, which is prone to artefact, or IgM antibody cating the diagnostic value of CSF examination [20,45].
tests, which are known to have a significant risk of false‐ European experts have estimated that it occurs in less
positive reactions. Positive serology, especially in areas of than one in 1000 cases of untreated infection [46].
high prevalence (where background seroprevalence in the
population can be as high as 10–20%), should not be inter- Musculoskeletal manifestations
preted as evidence for causation of atypical lesions without Myalgias and arthralgias are common features of early
additional support from direct detection of the organism. disseminated Lyme borreliosis. Frank arthritis with
More recent studies and reviews suggest that B. burgdorferi ­synovitis was the presentation that initially drew atten-
is rarely associated with these presentations [29,36–38]. tion to the infection in Connecticut. As with other later
Chapter 40  Lyme Borreliosis 469

manifestations, it has become less common in recent Recommendations for treatment of Lyme borreliosis
years, probably because high awareness of Lyme borrelio- in pregnant women are the same as those for nonpreg-
sis in endemic areas has led to earlier recognition and nant adults, with the exception that doxycycline is
treatment. ­contraindicated [7,52,53].
Arthritis is strongly associated with infection caused by
B. burgdorferi sensu stricto, but other pathogenic genospe- Diagnosis. Diagnosis of EM is primarily clinical, based on
cies also cause occasional cases in Europe [47]. Patients recognition of the characteristic skin lesion in a patient
present initially with migratory arthralgias, developing who has had recent exposure to ticks. A specific history of
later into an asymmetrical mono‐ or oligoarthritis, most tick bite is not an essential criterion, as many tick bites go
commonly affecting the knee. Other large joints are less unnoticed. No later‐stage manifestation of Lyme borrelio-
frequently affected, and small joints of the hands and feet sis is unique to the infection; supporting evidence from
are rarely involved. Synovial effusion can cause marked the laboratory should be obtained to confirm a diagnosis
joint swelling, out of proportion to the degree of pain. of disseminated or late infection [20,52].
Patients do not have symptoms suggesting acute sepsis,
and serum inflammatory markers are usually only mildly Direct detection methods
elevated [20,48,49]. Tests for serum IgG antibodies to Direct detection methods include borrelial culture and
B.  burgdorferi are almost always very strongly positive, DNA detection. Culture is not routinely available for
and laboratory examination of synovial fluid usually diagnostic purposes, as it requires complex culture media
demonstrates a polymorphonuclear infiltrate and pres- and may take 3–6 weeks. It is valuable in providing iso-
ence of borrelial DNA in patients who have not received lates of proven pathogenicity for research purposes. It has
antibiotics. Antibiotic treatment is usually effective in an overall success rate of about 70% when applied to skin
eradicating infection from the affected joint(s) but inflam- biopsies from patients with untreated EM or ACA, but
mation may not resolve completely by the end of treat- much lower rates (<10%) from CSF or blood, unless large‐
ment, taking weeks or months for complete resolution. volume (30–40 mL) blood cultures are taken. Synovial
A minority of patients have persistent arthritis even fluid culture has rarely been successful [7,54].
after re‐treatment, without objective evidence of active Nuclear amplification methods, usually polymerase
infection. This has been termed ‘antibiotic‐refractory’
­ chain reaction (PCR), for DNA detection are more valua-
arthritis and appears to have an autoimmune component. ble in providing timely results, and a variety of target
Patients with antibiotic‐refractory arthritis are more likely DNA sequences are available [2,7,55]. Positive results

SECTION 7: BACTERIAL SKIN


than those with antibiotic‐responsive arthritis to have should be confirmed by probe hybridization or sequenc-
HLA‐DR molecules that bind B. burgdorferi outer surface ing of the amplicate, also allowing genospecies identifi-
protein A163–175, and in some patients there is also an asso- cation [7]. Overall sensitivities for skin biopsies appear to
ciation with intra‐articular steroid use prior to antibiotic be about 70% for samples from patients with EM and

INFECTIONS
treatment [50]. An early longitudinal study of 21 patients over 90% from those with ACA. In patients with acute
who did not receive any antibiotics showed that they neuroborreliosis, PCR positivity on CSF samples has
had attacks of arthritis for a median of 43 months (range been reported to range from about 70% in some American
4–76 months) [48]. A later study of patients who received studies to 30% in European patients, probably reflecting
antibiotic treatment showed that those with antibiotic‐ variations in pathogenicity associated with different
responsive disease had episodes of arthritis during a ­borrelial genospecies and in duration of disease prior to
median total time period of 4 months (range 1–51 months) sampling. Synovial tissue or fluid samples from patients
and those with antibiotic‐refractory disease had episodes with untreated Lyme arthritis have DNA detection rates
for a median total period of 16 months (range 4–73 around 70–90%, and these tests can occasionally be ­useful
months), suggesting that antibiotic treatment reduces the in assessing the need for additional antibiotic treatment
duration of inflammation even in antibiotic‐refractory in patients with refractory arthritis [50]. As with any
arthritis [50]. Nonsteroidal anti‐inflammatory agents are DNA detection test, care must be taken to avoid labora-
also helpful. tory contamination, as PCR methods are extremely sensi-
tive and are vulnerable to false‐positive results from
Lyme disease in pregnancy extraneous target DNA [56].
Three cases of maternal‐to‐fetal transmission of B. burg-
dorferi were documented in early reports, with the babies Indirect diagnostic methods
stillborn or dying within the first 48 hours after delivery. Antibody tests remain the mainstay of laboratory support
The mothers had received inadequate or no treatment for for the diagnosis of Lyme borreliosis [2,7,55,57–59]. The
EM during pregnancy. Since then several large studies antibody response to B. burgdorferi infection is slow to
have shown no increased rates of adverse outcomes of develop compared to many other bacterial or viral infec-
pregnancies and no excess of congenital abnormalities in tions, reflecting the organism’s slow replication cycle, and
Lyme‐endemic areas by comparison to nonendemic areas. early treatment can abrogate an antibody response.
An extensive review of published data concluded that an Antibody positivity rates in EM range from 30% to 80%,
adverse fetal outcome due to maternal infection with depending on duration of infection prior to treatment
B. burgdorferi at any point during pregnancy in humans is and, to a certain extent, on infecting genospecies [54].
rare [51]. Infection with B. burgdorferi sensu stricto appears to
470 Section 7  Bacterial Skin Infections

­enerate a brisker antibody response than the other


g neuroborreliosis. Examination of CSF can also give valu-
­pathogenic genospecies. About 70–90% of patients with able additional supporting evidence including lympho-
early disseminated infection are seropositive at clinical cytic pleiocytosis and raised CSF protein levels [20,45,46].
presentation, and the remainder usually seroconvert
within several weeks. Patients with late‐stage Lyme bor- Diagnostic tests that are not recommended
reliosis are very rarely seronegative; seropositivity is in A number of poorly standardized and unvalidated meth-
excess of 99%. The great majority of patients with late‐ ods are marketed, mainly from commercial ‘Lyme disease
stage infections, including ACA, Lyme arthritis and late specialty’ laboratories. They include antigen detection
neuroborreliosis, are very strongly seropositive, with tests in body fluids, PCR of urine, lymphocyte transfor-
antibodies to a wide range of borrelial antigens [7,20,54,55]. mation tests (LTTs), CD57 lymphocyte measurements and
Most current antibody screening tests are based on immunoblots performed as single‐stage tests or inter-
enzyme‐linked immunosorbent assay (ELISA) methods. preted using nonstandard criteria. They have contributed
There are significant issues related to specificities of these to many misdiagnoses in North America and Europe
tests, especially older tests based on whole‐cell‐sonicate [7,55,57,61,64].
antigens, related to the complex antigenic structure of B.
burgdorferi sensu lato. Some antigens are shared with other
spirochaetes and other flagellated organisms. False‐posi- Treatment of  Lyme borreliosis. Evidence‐based guide-
tive reactions can occur in tests on sera from patients with lines and consensus documents for the treatment of Lyme
syphilis, leptospirosis and enteric bacteria, presumably borreliosis in adults and children have been published by
caused by high levels of antibodies produced to common a number of American and European specialist societies
flagellin antigens. Nonspecific reactions, especially in and expert groups [7,52,53,58,59,65–67]. These will
IgM tests, have been well documented to occur in the undergo regular review. There are only minor variations
presence of other acute infections and also with some in drug choice, dose and duration between the guidelines.
autoimmune conditions. The most commonly recommended antibiotics for oral
Newer ELISAs based on recombinant or synthetic use are doxycycline, amoxicillin and cefuroxime axetil.
­peptide antigens are easier to standardize and have better Cefuroxime axetil is generally recommended only if there
performance characteristics, but some specificity prob- are contraindications to doxycycline and amoxicillin.
lems remain. Accordingly, authorities in North America Macrolides should be used only if the other oral agents
and Europe currently recommend using a two‐stage are contraindicated because there is a significant risk of
treatment failure, especially with erythromycin, which
SECTION 7: BACTERIAL SKIN

­testing process to minimize risk of false‐positive results


[2,7,20,54,57]. Serum samples are screened in a first‐stage has been superseded by azithromycin in more recent
test, using a sensitive ELISA method. Specimens giving ­recommendations. Ceftriaxone is the preferred agent for
reactive or indeterminate results are then tested with a parenteral use, as it can be given once daily, facilitating
INFECTIONS

second method, usually immunoblot (western blot), outpatient use.


which allows an evaluation of the sample’s reactions to a Oral treatment is recommended for EM, BL, arthritis
broad range of B. burgdorferi antigens. This assessment and ACA. Most authorities recommend oral treatment
minimizes the risk of false‐positive results overall. for isolated facial palsy without other neurological mani-
Adherence to the two‐stage recommendation is impor- festations and parenteral antibiotics for other nervous
tant, as some screening ELISAs have specificities of less system presentations. However, there is increasing inter-
than 95% and the predictive value of positive results can est in the use of oral doxycycline for neuroborreliosis fol-
be very low when tests are applied in a situation where lowing a well‐conducted Norwegian study that showed
there is a low prior probability of B. burgdorferi infection, noninferiority of outcome in adult patients treated with
leading to misdiagnosis [7,20,55]. Unfortunately, this is a doxycycline compared with those who received standard
frequent occurrence. Numerous studies have shown that therapy with ceftriaxone, confirming the findings of an
misdiagnosis is a major cause of nonresponse to treat- earlier Swedish study and providing additional evidence
ment for presumed Lyme borreliosis, usually resulting to support the use of doxycycline for neuroborreliosis
from false‐positive tests or from the application of poorly [58,68–70].
specific clinical diagnostic criteria [56,60–63]. See Tables 40.1 and 40.2 for recommendations.
An important additional factor to be considered when
evaluating serological results is the high background Prognosis. Several studies have shown excellent outcomes
seroprevalence in populations living in highly endemic for patients with appropriately treated EM [71–76]. A large
areas, reflecting residual antibodies to a previous infec- prospective study performed in Connecticut showed
tion, which may have been asymptomatic, so it is impor- excellent long‐term outcomes of children who received
tant that positive antibody results are carefully assessed appropriate treatment for EM and other manifestations
for current clinical relevance [2,7,55]. [75]. European studies have shown similar results [71,76].
Antibody tests are also useful in CSF assessment. Most Most patients with acute neuroborreliosis who receive
patients with acute neuroborreliosis have detectable anti- appropriate treatment also have very good long‐term out-
bodies to B. burgdorferi in CSF at time of presentation, and comes, but full recovery can take some weeks or months,
intrathecal antibody synthesis can also be assessed. This as seen in other conditions such as viral meningitis or
is particularly useful in the evaluation of suspected late meningoencephalitis. Patients with facial palsy may have
Chapter 40  Lyme Borreliosis 471

Table 40.1  Commonly recommended antibiotics and doses for treatment of Lyme borreliosis

Agent Route Adult dose Child dose Notes

Doxycycline Oral 100  mg 12‐hourly 4 mg/kg/day (max. 100 mg/dose) Contraindicated in children <9 years
(or 200 mg daily) (12 years in UK)
Amoxicillin Oral 500 mg–1 g 50 mg/kg/day in three divided
8‐hourly doses (max. dose 500 mg)
Cefuroxime axetil Oral 500 mg 12‐hourly 30 mg/kg/day in two divided
doses (max. dose 500 mg)
Ceftriaxone Parenteral 2 g daily 50–100 mg/kg/day as single dose
Cefotaxime Parenteral 2 g 8‐hourly 150–200 mg/kg/day in three
divided doses
Benzylpenicillin Parenteral 18–24 mIU daily in 200 000–400 000 IU/kg/day in six
six divided doses divided doses (max.
18–24 mIU/day)
Azithromycin (careful follow‐up Oral 500 mg daily 5 mg/kg/day (max. 500 mg) 5‐ to 10‐day course for EM. Not
required, see text and references recommended for disseminated or late
for further details) infection. Careful follow‐up required

Table 40.2  Indications, routes of administration and duration of treatments estimated to occur in 5–10% of patients with dissemi-
for Lyme borreliosis nated infection and is less common in children than in
adults. Six randomized controlled trials have shown no
Treatment indication Route Duration evidence of persistent borrelial infection in patients
with post‐Lyme symptoms, and no sustained benefit
Erythema migrans Oral 14 days
(range 10–21)
from prolonged antibiotic treatments, which can cause
Borrelial lymphocytoma Oral 14 days significant risks [81–83].
(range 10–21)
Isolated facial nerve Oral or parenteral 14 days ‘Chronic Lyme disease’
palsy (range 14–21) This term has been used to denote a variety of patient
Early or late Parenteral (or oral – see 14 days

SECTION 7: BACTERIAL SKIN


groups, including those with post‐Lyme syndrome and
neuroborreliosis text and references) (range 10–28)
Acrodermatitis chronica Oral 21 days
patients with incomplete clinical recovery following treat-
atrophicans (range 14–28) ment of late Lyme borreliosis, as described earlier in the
Arthritis Oral 28 days chapter. The majority of patients given a diagnosis of

INFECTIONS
Arthritis (if re‐ Oral 28 days ‘chronic Lyme disease’ include those who have had
treatment indicated) B. burgdorferi infection at some time in the past but whose
current clinical presentation is due to another unrelated
See references 7,50,52,53,58,59,65–67 for more detailed information and condition, and patients who have never had B. burgdorferi
additional recommendations for complicated cases.
infection but have been misdiagnosed through the use of
poorly specific case definitions or false‐positive results,
some residual paresis, which is usually mild [77]. Some, frequently from nonstandard laboratory tests. Studies
mainly older, patients with radiculopathies can have from referral centres have shown that many patients
­persistent paraesthesias [78]. given this diagnosis fulfil criteria for conditions such
The degree of clinical recovery of patients with late‐ as  chronic fatigue syndrome or fibromyalgia, and some
stage manifestations such as ACA or late neuroborreliosis have other serious conditions for which specific treat-
depends in part on the severity of damage incurred prior ments are indicated [61–63,81].
to treatment [7,78]. Antibiotics can eradicate the infection
but full recovery may not be possible if tissue damage has Prevention. No vaccine is currently available.
been severe. However, a historical study of untreated
patients with neuroborreliosis showed good functional Education, awareness and personal protection measures
results in the majority of the study group [79]. Public education is needed to heighten awareness of ticks
and their potential for disease transmission. This is
Persistent symptoms following treated Lyme borreliosis ­particularly important with the increasing popularity of
A minority of patients have nonspecific symptoms such outdoor recreational activities such as hiking, camping
as fatigue, musculoskeletal pain or cognitive com- and mountain biking, involving urban dwellers who may
plaints persisting after receiving appropriate treatment have little prior knowledge of the potential natural haz-
of their infection. This condition has been termed ‘post‐­ ards to which they might be exposed. The main preven-
treatment Lyme syndrome (PTLS) [2,7]. It seems to be tion measures are directed at avoidance of tick bites and
similar to fatigue syndromes documented following early removal of attached ticks, as transmission of borre-
other infections and correlates with severity of disease liae from an infected tick is very unlikely to occur in the
at presentation and with delayed treatment [80,81]. It is first few hours of a tick feed [7,52,84]. Simple measures
472 Section 7  Bacterial Skin Infections

include wearing light‐coloured long‐sleeved shirts with and has a mortality rate of about 1% [89]. A TBE vaccine is
buttoned cuffs and long trousers, using DEET‐based available. Powassan virus has caused cases of menin-
insect repellents, frequent checks of exposed skin for goencephalitis in North America. Co‐infections with
attached ticks during the day and a thorough check of the Lyme borreliosis can occur with any of these infections
body, including skinfolds (armpits, groins, backs of knees, and may produce modified disease presentations, which
under breasts, waistband area, etc.), at the end of each day can cause clinical diagnostic difficulties if the possibility
in a tick‐infested area. It is important to check the head of co‐infection is not considered.
and neck area, including the scalp, of young children.
Permethrin‐impregnated clothing can be useful for peo- Southern tick‐associated rash illness (STARI)
ple who have heavy prolonged exposure to ticks, such as This condition, also known as Masters disease, is associ-
forestry workers. Permethrin kills ticks on contact, but is ated with bites of Amblyomma americanum, the Lone Star
not suitable for application directly to the skin. tick. It presents as an EM‐like rash that is smaller and
more circular and has a greater degree of central clearing
Tick removal than that of EM. Affected patients seem to be less likely to
Attached ticks should be removed using fine‐pointed have significant systemic symptoms than those who have
tweezers, grasping the mouth parts as close to the skin as Lyme borreliosis [90]. The causative agent has not been
possible and pulling gently and steadily upwards [85]. identified. The condition has been identified predomi-
A skin disinfectant can be applied after removal to reduce nantly in Missouri but the tick vector has a broad range
risk of pyogenic infection. Retained mouth parts do not through the southeastern USA and its range appears to
increase risk of borrelial transmission, but may occasion- have increased in recent years, appearing as far north as
ally cause a foreign body reaction. The site of tick attach- Maine [91].
ment should be monitored for possible development of
rash, but it is also important to recognize that infection
References
can occur following an inapparent tick bite at another site. 1 Edlow JA. Bull’s Eye: Unraveling the Medical Mystery of Lyme
Disease, 2nd edn. New Haven, CT: Yale University Press, 2004.
Chemoprophylaxis 2 Steere AC, Strle F, Wormser GP et al. Lyme borreliosis. Nat Rev Dis
Post‐tick bite antibiotic prophylaxis is not recommended Primers 2016;2:16090.
3 Stanek G, Strle F. Lyme disease  –  European perspective. Infect Dis
routinely, but may be considered in certain circumstances. Clin North Am 2008;22:327–39.
A trial performed in an American hyper‐endemic area for 4 Van der Wijngaard CC, Hofhuis A, Simoes M et al. Surveillance per-
SECTION 7: BACTERIAL SKIN

Lyme borreliosis, using single‐dose doxycycline 200 mg spective on Lyme borreliosis across the European Union and
for adults presenting within 3 days of an I. scapularis tick European Economic Area. Euro Surveill 2017;22.
5 Centers for Disease Control and Prevention. How many people get
bite, was shown to be 87% effective, but 30% of recipients Lyme disease? Available at: https://www.cdc.gov/lyme/stats/
reported significant side‐effects. It was notable that the humancases.html (accessed 9 December 2018).
INFECTIONS

overall infection rate in placebo recipients was only 3.5% 6 Ogden NH, Koffi JK, Pelcat Y, Lindsay LR. Environmental risk from
Lyme disease in central and eastern Canada: a summary of recent sur-
in this high‐risk area [86,87]. Subgroup analysis showed veillance information. Can Commun Dis Rep 2014;40:74–82.
that people who had sustained bites from nymphal ticks 7 Hofmann H, Fingerle V, Hunfeld K‐P et al. Cutaneous Lyme borre-
that had fed for several days and had become partially liosis: Guideline of the German Dermatology Society. Ger Med Sci
engorged were most at risk of infection, emphasizing the 2017;15:Doc14.
8 La maladie de Lyme: Donnees du reseau de surveillance de la maladie
value of early removal. The findings of this trial may not en Alsace. Mars 2001–Fev 2003 (2008) Institut Veille Sanitaire.
be applicable to other areas with lower borrelial carriage Available at: www.invs.sante.fr/publications/2005/lyme_alsace/
rates in ticks, nor to the European situation, as there is index.html (accessed 9 December 2018).
9 Huppertz HI, Bohme M, Standaert SM et al. Incidence of Lyme bor-
some evidence that borrelial transmission from I. ricinus
reliosis in the Wurzburg region of Germany. Eur J Clin Microbiol
ticks may occur earlier in a feed, although the risk remains Infect Dis 1999;18:697–703.
low within the first 18 hours of attachment [7]. Several 10 Medlock JM, Hansford KM, Bormane A et  al. Driving forces for
guidelines and consensus documents provide detailed change in geographical distribution of Ixodes ricinus ticks in Europe.
Parasites &Vectors 2013;6:1
discussion on the use and limitations of chemoprophy-
11 Centers for Disease Control and Prevention. Lyme Disease/
laxis [7,52,53,66]. Transmission. Available at: https://www.cdc.gov/lyme/transmission/
index.html (accessed 9 December 2018).
12 Pritt BS, Mead PS, Hoang Johnson DK et al. Identification of a novel
Associated infections. Ixodid ticks can also transmit pathogenic Borrelia species causing Lyme borreliosis with an unusu-
ally high spirochaetaemia: a descriptive study. Lancet Infect Dis
­several other infections, including babesiosis, a protozoal 2016;16:556–64.
infection parasitizing red blood cells, and anaplasmosis, a 13 Dykhuizen DE, Brisson D, Sandigursky S et al. Short report: the pro-
rickettsial infection affecting neutrophils [52]. B. miyamo- pensity of different Borrelia burgdorferi sensu stricto genotypes to
toi, a species more closely related to B. recurrentis, the cause disseminated infections in humans. Am J Trop Med Hyg
2008;78:806–10.
cause of relapsing fever, than to B. burgdorferi, has recently 14 Rupprecht TA, Koedel U, Fingerle V, Pfister HW. The pathogenesis of
been recognized as a cause of febrile illness, myalgia, Lyme neuroborreliosis  –  from infection to inflammation. Mol Med
arthralgia and, in severe cases, meningoencephalitis [88]. 2008;14:205–12.
15 Kannian P, McHugh G, Johnson BJB et  al. Antibody responses to
In some parts of Europe a tick‐transmitted flavivirus
Borrelia burgdorferi in patients with antibiotic‐refractory, antibiotic‐
causes tickborne encephalitis (TBE), which can cause responsive or non‐antibiotic‐treated Lyme arthritis. Arthritis Rheum
severe illness in some patients, particularly the elderly, 2007;56:4216–25.
Chapter 40  Lyme Borreliosis 473

16 Anderson JF, Magnarelli LA. Biology of ticks. Infect Dis Clin North 45 Halperin JJ. Nervous system Lyme disease. Infect Dis Clin N Am
Am 2008;22:195–215. 2015;29:241–53.
17 Wilhelmason P Fryland L. Sjowall J et al. A prospective study on the 46 Halperin J, Logigian E, Finkel M et  al. Practice parameters for the
incidence of Borrelia burgdorferi sensu lato infection after a tick bite diagnosis of patients with nervous system Lyme borreliosis.
in Sweden and on the Aland islands (2008‐2009). Ticks Tick Borne Dis Neurology 1996;46:619–27.
2016;7:71–9. 47 Eiffertt H, Karsten A, Thomssen R et al. Characterisation of Borrelia
18 Feder HM, Gerber MA, Cartter ML et al. Prospective assessment of burgdorferi strains in Lyme arthritis. Scand J Infect Dis 1998;30:265–8.
Lyme disease in school‐aged population in Connecticut. J Infect Dis 48 Steere AC, Schoen RT, Taylor E. The clinical evolution of Lyme arthri-
1995;171:1371–4. tis. Ann Intern Med 1987;107:725–31.
19 Centers for Disease Control and Prevention. Lyme disease/Clinical 49 Arvikar SL, Steere AC. Diagnosis and treatment of Lyme arthritis.
description. Available at: https://wwwn.cdc.gov/nndss/conditions/ Infect Dis Clin N Am 2015;29:269–80.
lyme‐disease/case‐definition/2017/ (accessed 9 December 2018). 50 Steere AC, Angelis S. Therapy for Lyme arthritis. Strategies for the
20 Stanek G, Fingerle V, Hunfeld KP et al. Lyme borreliosis: clinical case treatment of antibiotic‐refractory arthritis. Arthritis Rheum 2006;54:
definitions for diagnosis and management in Europe. Clin Microbiol 3079–86.
Infect 2011;17:69–79. 51 Elliott DJ, Eppes SC, Klein JL. Teratogen update: Lyme disease.
21 Nadelman R. Erythema migrans. Infect Dis Clin N Am 2015;29:211–39. Teratology 2001;64:276–81.
22 Glatz N, Resinger A, Semmelweis K et al. Clinical spectrum of skin 52 Wormser GP, Dattwyler DJ, Shapiro ED et al. The clinical assessment,
manifestations of Lyme borreliosis in 204 children in Austria. Acta treatment and prevention of Lyme disease, human granulocytic ana-
Derm Venereol 2015;95:564–71. plasmosis and babesiosis. Clinical Practice Guidelines by the Infectious
23 Strle F, Nadelman R, Cimperman J et al. Comparison of culture confirmed Diseases Society of America. Clin Infect Dis 2006;43:1089–134.
erythema migrans caused by Borrelia burgdorferi sensu stricto in New York 53 Societe de Pathologie Infectieuse de Langue Francaise. Lyme borreliosis:
and by Borrelia afzelii in Slovenia. Ann Intern Med 1999;130:32–6. diagnostic, therapeutic and preventive approaches. Med Mal Infect
24 Goldberg NS, Forseter G, Nadelman RB et  al. Vesicular erythema 2007;37(S3):8153–74. Available at: www.infectiologie.com/site/medias/_
migrans. Arch Dermatol 1992;128:1495–8. documents/consensus/2006-lyme-long.pdf (accessed 9 December 2018).
25 Logar M, Ruzic‐Sabljic E, Maraspin V et al. Comparison of erythema 54 Aguero‐Rosenfeld ME, Wang G, Schwarz I, Wormser GP. Diagnosis of
migrans caused by Borrelia afzelii and Borrelia garinii. Infection Lyme borreliosis. Clin Microbiol Rev 2005;18:484–509.
2004;32:15–19. 55 Dessau RB, van Dam AP, Fingerle V et  al. To test or not to test?
26 Wormser GP, McKenna D, Carlin J et al. Brief communication: haema- Laboratory support for the diagnosis of Lyme borreliosis. Clin
togenous dissemination in early Lyme disease. Ann Intern Med Microbiol Infect 2018;24:118–24.
2005;142:751–5. 56 Molloy PJ, Persing DH, Berardi VP. False‐positive results of PCR test-
27 Feder HM, Whitaker DL. Misdiagnosis of erythema migrans. Am J ing for Lyme disease. Clin Infect Dis 2001;33:412–13.
Med 1995;99:412–19. 57 Eldin C, Raffetin A, Bouiller K et  al. Review of European and
28 Weger W, Mullegger RR. Histopathology and immunohistochemistry American guidelines for the diagnosis of Lyme borreliosis. Med Mal
of dermatoborreliosis. Acta Dermatoven APA 2001;10:135–42. Infect 2019;49:121–32.
29 Müllegger RR, Glatz M. Skin manifestations of Lyme borreliosis: 58 Deutsche Gesellschaft fur Neurologie. Leitlinien: Neuroborreliose.
diagnosis and management. Am J Clin Dermatol 2008;9:355–68. Available at: https://www.dgn.org/leitlinien/3567‐ll‐030‐071‐2018‐
30 Strle F, Pieterski‐Rigler D, Stanek G et al. Solitary borrelial lymphocy- neuroborreliose (accessed 16 January 2019).

SECTION 7: BACTERIAL SKIN


toma: report of 36 cases. Infection 1992;20:201–6. 59 National Institute for Health and Clinical Excellence. NICE guideline
31 Glatz N, Resinger A, Semmelweis K et al. Clinical spectrum of skin [NG95] Lyme disease. Available at: https://www.nice.org.uk/guidance/
manifestations of Lyme borreliosis in 204 children in Austria. Acta ng95/chapter/Recommendations (accessed 16 January 2019).
Derm Venereol 2015;95:564–71. 60 Reid MC, Schoen RT, Evans J et al. The consequences of overdiagnosis
32 Arnez M, Ruzic‐Sabljic E. Borrelial lymphocytoma in children. Pediatr and overtreatment of Lyme disease: an observational study. Ann

INFECTIONS
Infect Dis J 2015;34:1319–22. Intern Med 1998;128:354–62.
33 Maraspin V, Cimperman J, Lotric‐Furlan S et al. Solitary borrelial lym- 61 Feder HM, Johnson BJB, O’Connell S et  al. A critical appraisal of
phocytoma in adult patients. Wien Klin Wochenschr 2002;114:515–23. “chronic Lyme disease”. N Engl J Med 2007;357:1422–30.
34 Picken RN, Strle F, Picken MM et al. Identification of three species of 62 Hassett AL, Radvanski DC, Buyske S et al. Psychiatric co‐morbidity
Borrelia burgdoferi sensu lato (B burgdorferi sensu stricto, B garinii and and other psychological factors in patients with “chronic Lyme dis-
B afzelii) among isolates from acrodermatitis chronica atrophicans ease”. Am J Med 2009;122:843–50.
lesions. J Invest Dermatol 1998;110:211–14. 63 Patrick DM, Miller RR, Gardy JL et  al. Lyme disease diagnosed by
35 Brehmer‐Andersson E, Hovmark A, Asbrink E. Acrodermatitis chron- alternative methods: a phenotype similar to that of chronic fatigue
ica atrophicans: histopathologic findings and clinical correlations in syndrome. Clin Infect Dis 2015;61:1084–91.
111 cases. Acta Derm Venereol 1998;78:207–13. 64 Centers for Disease Control and Prevention. Notice to readers: cau-
36 Weide B, Walz T, Garbe C. Is morphea caused by Borrelia burgdorferi? tion regarding testing for Lyme disease. MMWR 2005;54:125.
A review. Br J Dermatol 2000;142:636–44. 65 Halperin JJ, Shapiro ED, Logigian E et  al. Practice Parameter: treat-
37 Goodlad JR, Davidson MM, Gordon P et  al. Morphea and Borrelia ment of nervous system Lyme disease (an evidence‐based review).
burgdorferi: results from the Scottish Highlands in the context of the Report of the Quality Standards Subcommittee of the American
world literature. J Clin Pathol Mol Pathol 2002;55:374–8. Academy of Neurology. Neurology 2007;69:91–102.
38 Bergler‐Czop B, Lis‐Swięty A, Brzezińska‐Wcisło L. Scleroderma lin- 66 Evison J, Aebi C, Francioli P et  al. Borreliose de Lyme. Diagnose et
earis: hemiatrophia faciei progressive (Parry–Romberg syndrome) traitement de la borreliose de Lyme chez l’adulte et l’enfant: recom-
without any changes in CNS and linear scleroderma “en coup de mandations de la Societe Suisse d’Infectiologie. Rev Med Suisse
sabre” with CNS tumor. BMC Neurol 2009;9:39. 2006;2:919–40.
39 Christen HJ, Hanefeld F, Eiffert H et  al. Epidemiology and clinical 67 Mygland A, Ljostad U, Fingerle V et al. EFNS guidelines on the diag-
manifestations of Lyme borreliosis in childhood. A prospective multi- nosis and management of European Lyme neuroborreliosis. Eur
centre study with special regard to neuroborreliosis. Acta Paediatr J Neurol 2010;17:8–16.
1993;36(suppl):1–75. 68 Ljostad U, Skogvoll E, Eikelund R et al. Oral doxycycline versus intrave-
40 Sood SK. Lyme disease in children. Infect Dis Clin N Am 2015;29:281–94. nous ceftriaxone for European Lyme borreliosis: a multicentre, noninfe-
41 Eiffert H, Karsten A, Schlott T et al. Acute peripheral facial palsy in riority, double‐blind, randomised trial. Lancet Neurol 2008;7:690–5.
Lyme disease – a distal neuritis at the infection site. Neuropediatrics 69 Karlsson M, Hammers‐Berggren S, Lindquist L et al. Comparison of
2004;35:257–73. intravenous penicillin G and oral doxycycline for the treatment of
42 Rothermel H, Hodges TR, Steere AC. Optic neuropathy in children Lyme neuroborreliosis. Neurology 1994;44:1203–7.
with Lyme disease. Pediatrics 2001;108:477–81. 70 Bremell D, Dotevall L. Oral doxycycline for Lyme neuroborreliosis
43 Garin C, Bujadoux CH. Paralysie par les tiques. J Med Lyon with symptoms of encephalitis, myelitis, vasculitis or intracranial
1922;71:765–7. hypertension. Eur J Neurol 2014;21:1162–7.
44 Bannwarth A. Chronische lymphocytäre Meningitis, entzündliche 71 Lipsker D, Antoni‐Bach N, Hansmann Y, Jaulhac B. Long term prog-
Polyneuritis und “Rheumatismus”. Arch Psychiatr Nervenkr nosis of patients treated for erythema migrans in France. Br J Dermatol
1941;113:284–376. 2002;146:872–6.
474 Section 7  Bacterial Skin Infections

72 Nadelman RB, Nowakowski J, Forseter G et al. The clinical spectrum 82 Fallon BA, Keilp JG, Corbera KM et  al. A randomised placebo
of early Lyme borreliosis in patients with culture‐confirmed erythema ­controlled trial of repeated IV antibiotic therapy for Lyme encepha-
migrans. Am J Med 1996;100:502–8. lopathy. Neurology 2008;70:992–1003.
73 Cerar D, Cerar T, Ruzic‐Sabljic E et  al. Subjective symptoms after 83 Berende A, ter Hofstede HJ, Vos FJ et al. Randomised trial for longer‐
treatment of early Lyme disease. Am J Med 2010;123:79–86. term therapy for symptoms attributed to Lyme disease. N Engl J Med
74 Wormser GP, Weitzner E, McKenna D et al. Longterm assessment of 2016;374:1209–20.
health‐related quality of life in patients with culture‐confirmed early 84 Hayes EB, Piesman J. How can we prevent Lyme disease? N Engl
Lyme disease. Clin Infect Dis 2015;61:244–7. J Med 2003;348:2424–30.
75 Gerber MA, Shapiro ED, Burke GS et al. Lyme disease in children in 85 Centers for Disease Control and Prevention. Lyme disease/Tick
southeastern Connecticut. N Engl J Med 1996;335:1270–4. removal and testing. Available at: https://www.cdc.gov/lyme/
76 Thorstrand C, Belfrage E, Bennet R et al. Successful treatment of neu- removal/index.html (accessed 9 December 2018)
roborreliosis with ten day regimens. Pediatr Inf J 2002;21:1142–5. 86 Nadelman RB, Nowakowski J, Fish D et al. Prophylaxis with single
77 Skogman BH, Glimaker K, Nordman M et  al. Longterm clinical dose doxycycline for the prevention of Lyme disease after an Ixodes
outcome after Lyme neuroborreliosis in childhood. Pediatrics
­ scapularis tick bite. N Engl J Med 2001;345:79–84.
2012;130:262–9. 87 Shapiro ED. Doxycline for tick bites? Not for everyone. N Engl J Med
78 Hansen K, Lebech AM. The clinical and epidemiological profile of 2001;345:133–4.
Lyme neuroborreliosis in Denmark 1985–1990. A prospective study of 88 Krause P, Fish D, Narasimhan S, Barbour AG. Borrelia miyamotoi
187 patients with Borrelia burgdorferi‐specific intrathecal antibody pro- infection in nature and in humans. Clin Microbiol Infect 2015;
duction. Brain 1992;115:399–423. 21:631–9.
79 Krüger H, Reuss K, Pulz M et al. Meningoradiculitis and encephalo- 89 Kaiser R. Tick‐borne encephalitis. Infect Dis Clin North Am 2008;
myelitis due to Borrelia burgdorferi: a follow‐up study of 72 patients 22:561–75.
over 27 years. J Neurol 1989;236:322–8. 90 Wormser G, Masters E, Nowakowski J et al. Prospective clinical eval-
80 Hickie I, Davenport T, Wakefield D et al. Post‐infective and chronic uation of patients from Missouri and New York State with erythema
fatigue syndromes precipitated by viral and non‐viral pathogens: migrans‐like lesions. Clin Infect Dis 2005;41:958–65.
prospective cohort study. BMJ 2006;333:575. 91 Centers for Disease Control and Prevention. STARI or Lyme?
81 Marques A. Chronic Lyme disease: a review. Infect Dis Clin North Am Available at: https://www.cdc.gov/stari/disease/index.html (accessed
2008; 22:341–60. 9 December 2018).
SECTION 7: BACTERIAL SKIN
INFECTIONS
 475

C HA PTER   41

Bartonella Infections
Sonia Kamath1 & Minnelly Luu2,1
 Department of Dermatology, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
1

 Division of Pediatric Dermatology, Children’s Hospital Los Angeles, Los Angeles, CA, USA
2

Introduction, 475 Cat scratch disease, 478


Bacillary angiomatosis, 475 Bartonellosis, 480

Abstract vascular papules and nodules with lobular vascular proliferations


of plump endothelial cells on histology. Lesions generally respond
The spectrum of Bartonella infections includes cat scratch disease well to therapy with erythromycin. Lastly, bartonellosis is caused
(CSD), bacillary angiomatosis, trench fever and bartonellosis (or by B. bacilliformis and transmitted from person to person by sand-
Carrion disease). In immunocompetent individuals, B. henselae flies in certain regions of Peru, Colombia and Ecuador. Bartonel-
infection results in CSD, characterized by self‐limited regional losis is characterized by acute and chronic presentations. The acute
lymphadenopathy after a cat scratch or bite distal to the affected Oroya fever phase may lead to severe, life‐threatening anaemia
node. Histologically, CSD manifests as a granulomatous process. and requires treatment with chloramphenicol. The chronic verruga
By contrast, B. henselae or B. quintana infection in immunocom- peruana phase is associated with less morbidity and responds well
promised hosts results in bacillary angiomatosis, comprised of to rifampicin (rifampin).

Key points • Caused by B. henselae, cat scratch disease is a common self‐


limiting cause of lymphadenopathy in immunocompetent
• The majority of Bartonella infections are caused by three children and is characterized by an inoculation lesion followed
species: Bartonella henselae, Bartonella quintana and Bartonella by regional lymphadenopathy.

SECTION 7: BACTERIAL SKIN


bacilliformis. • Bartonellosis, caused by B. bacilliformis, is geographically
• Bacillary angiomatosis consists of vascular papules or nodules limited to parts of South America and consists of an acute phase
caused by B. henselae and B. quintana, occurs primarily in (Oroya fever) and eruptive phase (verruga peruana), which
immunocompromised patients, and generally responds to respond to treatment with chloramphenicol and rifampicin,

INFECTIONS
treatment with erythromycin. respectively.

Introduction B. bacilliformis, is limited geographically to certain areas of


Peru, Colombia and Ecuador [3].
The genus Bartonella includes more than 30 different ­species
of facultative intracellular Gram‐negative bacilli. A variety
of mammals, including cats, rodents and humans, serve as Bacillary angiomatosis
reservoirs for Bartonella species, and the bacteria are often
transmitted via vectors, such as sandflies, body lice, fleas History. Compared to other Bartonella infections, bacillary
and ticks. Over fifteen of these species have been described angiomatosis was described relatively recently; however,
to cause human infections; however, three species account the research on bacillary angiomatosis has led to consid-
for the majority of Bartonella‐related pathology in humans: erable advances in the understanding of Bartonella
Bartonella henselae, Bartonella quintana and Bartonella bacilli- ­infections in general. In 1983 Stoler et  al. described the
formis [1]. The clinical manifestations associated with first case of bacillary angiomatosis in a patient with
Bartonella infection depend largely on the host’s immune acquired immunodeficiency syndrome (AIDS) and sub-
status. The more benign, self‐limited cat scratch disease, cutaneous nodules. On biopsy, these nodules showed a
caused by B. henselae, occurs in immunocompetent hosts, lobular proliferation of capillary channels. Tissue cultures
while the more severe bacillary angiomatosis, caused by failed to reveal a causative organism, but Warthin–Starry
B. henselae and B. quintana, occurs more often in immuno- stains showed diffuse infiltration of small bacillary forms
compromised hosts [2]. Lastly, bartonellosis, caused by [4]. Cockerell et  al. then reported five similar cases in

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
476 Section 7  Bacterial Skin Infections

patients with human immunodeficiency virus (HIV), Both B. quintana and B. henselae are now known to cause
describing the condition as ‘epithelioid angiomatosis’ [5]. bacillary angiomatosis. B. quintana is also the aetiological
Soon after, AIDS patients with lesions histologically simi- agent of trench fever, an illness initially described in
lar to those described by Stoler et al. were found to have a troops during World War I, which recently has re‐emerged
positive reaction to antisera from the cat scratch disease in homeless inner‐city populations and has been reported
bacillus, and the term bacillary angiomatosis along with in children [25,26]. Humans are the main reservoir for
criteria for diagnosis of the condition was defined [6]. B.  quintana with human body lice (Pediculus humanus
Subsequent studies sought to better characterize the humanus) and possibly head lice (Pediculus humanus
infectious agent responsible for bacillary angiomatosis, ­capitis) as vectors [27–29].
but culture of the organism proved to be difficult. In By contrast, cats have been identified as a major reser-
1990, polymerase chain reaction (PCR) studies of infected voir for B. henselae. A case‐control study found that trau-
tissue samples revealed a previously uncharacterized matic cat exposure, such as a bite or scratch, was strongly
organism closely related to Rochalimaea quintana (the associated with bacillary angiomatosis [30,31]. Affected
cause of trench fever) [7]. Shortly after, a new species, patients were more likely to own a kitten than a cat [31].
Rochalimaea henselae, was identified as a cause of Further studies have found B. henselae bacteraemia in pet
­persistent fever with bacteraemia, peliosis hepatis and cats of affected patients and in approximately 40% of ran-
bacillary angiomatosis in patients with HIV [8–10]. domly screened cats [32]. Fleas from infected cats also
Koehler et al. then isolated both R. henselae and R. quin- carry the bacteria [32]. Still, many patients with bacillary
tana from cutaneous bacillary angiomatosis lesions and angiomatosis have no history of cat exposure, suggesting
successfully cultured the organisms on solid agar with an that the organism may be acquired from other sources,
endothelial cell monolayer [11]. such as arthropod vectors like fleas and ticks [14,27].
The original descriptions of the causative organisms of The pathogenesis of bacillary angiomatosis occurs via
bacillary angiomatosis refer to the genus Rochalimaea, multiple mechanisms, including immune evasion, bacte-
which was previously categorized in the family rial replication, erythrocyte and endothelial cell parasit-
Rickettsiaceae and order Rickettsiales. At the time, ism, and induction of angiogenesis. Bartonella may
B.  ­bacilliformis was the only Bartonella species that had successfully evade the host immune system due to ­several
been described and was categorized in the family characteristics. The B. henselae lipopolysaccharide (LPS) is
Bartonellaceae within the order Rickettsiales. A seminal subinflammatory, resulting in at least 1000‐fold less
paper by Brenner et al. reported that the four Rochalimaea potent toll‐like receptor (TLR) 4 activation compared to
SECTION 7: BACTERIAL SKIN

species were much more closely related to Bartonella than LPS from Salmonella [27]. B. quintana LPS may also p ­ ossess
previously thought. Thus, they were reclassified under antagonistic properties for TLR4, resulting in downregu-
the genus Bartonella (B. quintana, B. henselae, B. elizabethae lation of cytokines in monocytes that are normally
and B. vinsonii), and the family Bartonellaceae was produced by TLR4 [33]. Finally, B. henselae can avoid
­
INFECTIONS

removed from the Rickettsiales order altogether [12]. The lysosomal fusion and acidification after invading
­
full spectrum of Bartonella infections is now well under- endothelial cells and macrophages, thus avoiding the
stood to include bacillary angiomatosis, cat scratch ­normal endocytic pathway and conferring an intracellu-
­disease, trench fever and bartonellosis [13]. lar survival advantage [33,34].
Both B. quintana and B. henselae parasitize erythrocytes,
Epidemiology and pathogenesis. Bacillary angiomatosis causing chronic intraerythrocytic bacteraemia [27]. Within
primarily occurs in immunocompromised patients, such endothelial cells, they have been shown to induce pro‐
as those with HIV/AIDS or on immunosuppressive angiogenic mediators such as vascular endothelial growth
­therapy; however, there have been rare reports of the factor (VEGF) and interleukin (IL)‐8 [27]. The bacteria
­condition in immunocompetent patients as well [14]. In also replicate locally within the extracellular matrix and
children, bacillary angiomatosis appears to be extremely utilize BepA effector proteins to impede apoptosis of
rare. There are very few reports of HIV‐associated bacil- endothelial cells [33,35,36]. Additionally, B. quintana and
lary angiomatosis in paediatric patients; however, the B. henselae can colonize foci distant from the primary site
diagnosis should be considered in children with HIV in of infection with preference for highly vascularized tis-
the appropriate clinical setting [15–17]. There have been sues such as the liver and spleen, which has been sup-
rare reports of bacillary angiomatosis occurring in paedi- ported by an immunocompromised murine model [27,37].
atric solid organ transplant recipients and in a child The exact reasons why B. henselae and B. quintana cause
undergoing chemotherapy for leukaemia [18,19]. Few angiogenesis more often in immunocompromised hosts
reports exist of bacillary angiomatosis occurring in immu- but cat scratch disease and trench fever in immunocom-
nocompetent children [20]. Bacillary angiomatosis has petent hosts are still poorly understood.
been reported in pregnancy, and a recent case report
­provided evidence that B. henselae can be transmitted to Clinical features. Patients with bacillary angiomatosis
children in utero or during caesarean section from a may exhibit both cutaneous and extracutaneous manifes-
mother with chronic B. henselae bacteraemia [21–23]. tations of disease. Skin eruption is considered to be the
Additionally, B.  henselae was found to be viable in red most common manifestation of bacillary angiomatosis,
blood cell units after storage at 4 °C for 35 days, raising and several cutaneous morphologies have been described
the possibility of infection through blood transfusion [24]. [38]. Most commonly, lesions are described as red or
Chapter 41  Bartonella Infections 477

­ iolaceous dome‐shaped papules, which can evolve into


v response consistent with a Jarisch–Herxheimer reaction.
larger friable nodules or tumours that bleed profusely Pretreatment with an antipyretic medication may be help-
with trauma. These lesions often ulcerate with associated ful, and these symptoms are likely to improve over time
serosanguinous exudate [13]. Less commonly, patients with continued antibiotic therapy [11,18].
have deeper subcutaneous masses, which clinically appear
as large flesh‐coloured or dusky nodules that are often
Differential diagnosis. The differential diagnosis of the
tender and may appear cellulitic. These lesions may be
cutaneous lesions of bacillary angiomatosis includes
mobile or fixed to underlying structures, and involvement
infectious and non‐infectious entities. Noninfectious dis-
of the underlying bone is common [13]. Plaque‐like lesions
orders include pyogenic granuloma, cherry angioma,
and suppurative ulcers have also been reported [14].
dermatofibroma and haemangioma. Solitary lesions can
Although in general patients with cutaneous bacillary
be difficult to distinguish clinically from pyogenic granu-
angiomatosis present with multiple lesions, the majority
loma and may require biopsy for diagnosis [39]. Kaposi
of paediatric case reports of bacillary angiomatosis have
sarcoma (KS) is perhaps the most important diagnosis to
described isolated lesions, such as a crusted violaceous
differentiate from bacillary angiomatosis, as they are both
papule or nodule [15,17,19,20,39–41]. Cutaneous lesions
more common in the immunocompromised patient.
are most often located on the trunk and extremities, but
Clinically, cutaneous bacillary angiomatosis more often
there have been unusual reports of localized bacillary
presents as a bright‐red round papule, nodule or tumour.
angiomatosis in the oral cavity [42,43]. Patients with skin
Plaque‐type lesions are more rare compared to KS in
disease alone may have associated constitutional symp-
which patients often present with macules, patches and
toms such as fever, chills, malaise and anorexia [13].
plaques. On pathology, bacillary angiomatosis is more
In addition to the skin, bacillary angiomatosis may
sharply demarcated compared to KS, the vascular spaces
involve a number of internal organs, leading to associated
are more round compared to slit‐like in KS, and the
complications and a possibly fatal course. Patients with
endothelial cells in bacillary angiomatosis are plump
extracutaneous disease generally have associated consti-
compared to spindled in KS [14].
tutional symptoms and other systemic symptoms
With respect to the infectious differential diagnosis, the
depending on the organs involved. Hepatic involvement
eruptive phase of bartonellosis very closely resembles
may present as blood‐filled cystic spaces, also known as
bacillary angiomatosis clinically; however, this infection
bacillary peliosis hepatis. Splenic peliosis has also been
occurs almost only in endemic areas in South America.
observed and may require splenectomy [44]. Patients
Other infections on the differential diagnosis include

SECTION 7: BACTERIAL SKIN


usually present with gastrointestinal symptoms such as
atypical Mycobacterium infections, tuberculosis, coccidioi-
anorexia, nausea, vomiting, diarrhoea, abdominal disten-
domycosis, cryptococcosis, histoplasmosis and sporotri-
sion and weight loss. Clinical examination often reveals
chosis [39].
fever and hepatosplenomegaly [39]. Patients often have

INFECTIONS
cytopenias and elevated serum levels of liver enzymes,
including γ‐glutamyltransferase (GGT), alkaline phos- Laboratory and histology findings. Diagnosis of cutane-
phatase, and aspartate aminotransferase (AST) [14,38]. ous bacillary angiomatosis is most often based on sugges-
Peliosis hepatis or peliosis of the spleen may be compli- tive clinical features confirmed by histology. Biopsies of
cated by organ rupture with haemoperitoneum [44]. skin lesions of bacillary angiomatosis demonstrate a
Involvement of many other organs has been reported ­lobular proliferation of small blood vessels lined by large,
with bacillary angiomatosis, including the heart, lungs, plump endothelial cells that protrude into the vessel
muscles and soft tissues, gastrointestinal tract, bone lumen (Fig.  41.1) [6,18]. Granular clumps of purplish
marrow, bone and brain [39]. Lymphadenopathy may be material are present, around which there is prominent
the sole manifestation in approximately 20% of patients neutrophilic infiltrate with leucocytoclastic debris [39].
[38]. A case report by Rostad et al. described two paediatric Staining of these granular clumps with Warthin–Starry
solid organ transplant recipients who both presented reveals masses of small bacilli (Fig.  41.2) [18]. Electron
with unilateral lymph node swelling confirmed as microscopy demonstrates pleomorphic bacilli with a
bacillary angiomatosis by biopsy [18]. Laryngeal and Gram‐negative trilaminar wall [4,6]. Histological exami-
endobronchial lesions may cause obstruction and nation of bacillary peliosis hepatis demonstrates dilated
respiratory compromise [14,45]. Bony involvement may capillaries or multiple blood‐filled cystic spaces with foci
be asymptomatic or present as focal bone pain, and of necrosis and a myxoid stroma with similar granular
radiological examination often reveals osteolytic lesions clumps and inflammation admixed [14,44,47].
[46]. Some patients may develop bacteraemia without Both B. henselae and B. quintana can be isolated from
focal infection and generally present with associated blood using tubes containing ethylenediaminetetraacetic
malaise, fatigue, anorexia and weight loss [9,10]. acid (EDTA) and from cutaneous tissue by cultivation on
Once the diagnosis of bacillary angiomatosis is made, solid agar in the presence of an endothelial cell mon-
prompt treatment is required to avoid potential morbid- olayer [8,11]. Furthermore, Bartonella DNA from tissue
ity and mortality, and appropriate antibiotic therapy usu- or blood can be amplified by PCR, which has been found
ally leads to the resolution of cutaneous lesions and to be more sensitive than culture at detecting Bartonella
visceral disease [14]. Upon initiation of treatment, some [48]. Indirect fluorescence assay (IFA) and enzyme
patients may abruptly develop fever with a systemic toxic immunosorbent assay (ELISA) have also been tested for
478 Section 7  Bacterial Skin Infections

bacillary peliosis hepatis, the recommended treatment


duration is 4 months [50]. For bacillary peliosis hepatis, the
recommended treatment duration is 4 months [50]. Therapy
should be guided by clinical response and extended
beyond clinical clearance of lesions to help decrease the
likelihood of relapse. In severe or refractory cases, a combi-
nation of doxycycline and rifampicin (rifampin) can be
used. Patients should be monitored closely for relapse after
cessation of therapy, which probably requires a longer
duration of or indefinite antibiotic therapy [1].

Cat scratch disease


History. In 1889, Parinaud described a constellation of
symptoms including fever, conjunctival granulomas and
Fig. 41.1  Haematoxylin and eosin stain of a lymph node biopsy shows preauricular lymphadenopathy, known as oculoglandu-
diffuse proliferation of small blood vessels (original magnification, ×200). lar syndrome [52]. The first official report of cat scratch
Source: Reproduced from Rostad et al., 2012 [18]. Reproduced with disease came in 1950 in patients with a history of cat con-
permission of John Wiley & Sons Ltd. tact followed by regional adenitis that spontaneously
resolved [53]. Epidemiological features of the disease
­suggested an infectious aetiology; however, the infectious
agent proved to be difficult to isolate, and studies initially
implicated Afipia felis as the causative agent [54,55].
At the same time, investigators noted similarities
between the organisms causing cat scratch disease and
bacillary angiomatosis and speculated that they might be
the same. Serological evidence linked B. henselae to cat
scratch disease when sera from 36 of 41 patients (88%)
SECTION 7: BACTERIAL SKIN

with suspected cat scratch disease were found to have


high titres of antibodies to B. henselae antigens [30].
Additionally, sera with high titres of B. henselae antibod-
ies did not cross‐react with A. felis, and only 24% of
INFECTIONS

patients with cat scratch disease had detectable A. felis


antibodies, suggesting that A. felis might not be the pri-
mary causative agent [30]. Other reports confirmed the
positive serologies to B. henselae in patients with cat
Fig. 41.2  Warthin–Starry stain demonstrates aggregates of small scratch disease and found that 81% of the cats owned by
coccobacilli (original magnification, ×400). Source: Reproduced from Rostad patients with cat scratch disease had serum antibodies
et al., 2012 [18]. Reproduced with permission of John Wiley & Sons Ltd.
that reacted with B. henselae [56]. Subsequently, B. hense-
lae has been isolated from the lymph nodes of patients
serological diagnosis of B. henselae and B. quintana; with cat scratch disease [57].
­however, difficulties with serology include suboptimal
sensitivity and false‐positive tests due to cross‐reactivity Epidemiology and  pathogenesis. B. henselae is now
between Bartonella, Coxiella and Chlamydia species. widely accepted as the primary cause of cat scratch dis-
Additionally, serologies are not reliable in immunosup- ease [14]. There have also been reports of cases caused by
pressed patients [48]. As a result, PCR is preferred as a B. clarridgeiae [58,59]. Generally, cat scratch disease occurs
faster, more sensitive and more specific test [33]. in immunocompetent patients of all ages; however, 80%
are under 21 years of age [60]. It is considered the most
Treatment. The treatment of choice for bacillary angioma- common cause of chronic benign lymphadenopathy in
tosis and bacillary peliosis hepatis is erythromycin [49]. In children and adolescents. More than 90% of patients have
children, erythromycin ethylsuccinate can be used at a history of some type of contact with cats, and 60% have
40 mg/kg per day divided into four doses (maximum 2 g a preceding cat scratch [13,14]. Patients are significantly
per day). An alternative therapy depending on the age of more likely than healthy cat‐owning controls to have at
the patient is doxycycline 100 mg twice daily [50]. An least one kitten aged 12 months or younger, to have been
observational study in HIV patients found no statistical scratched by a kitten and to have at least one kitten with
difference in the cure or relapse rate in patients treated fleas [56]. There have been rare cases of cat scratch disease
with erythromycin compared to those treated with doxy- after skin injury from a foreign body, and fleas and ticks
cycline [51]. A treatment duration of at least 3 months has may also be vectors [1]. Family outbreaks have been
been recommended for bacillary angiomatosis, and for described, often in households with cats or kittens [61].
Chapter 41  Bartonella Infections 479

As discussed previously, B. henselae has been found to be manifestations aside from the primary inoculation lesion
viable in stored red blood cell units, suggesting the occur in 5% of patients and include a transient morbilli-
­possibility of transmission by blood transfusion [24]. form eruption, erythema nodosum, erythema multiforme,
Clinical manifestations of cat scratch disease may be leucocytoclastic vasculitis and thrombocytopenic pur-
due to an immunological reaction in the lymph nodes. pura [66]. Although patients with cat scratch disease are
Although PCR assays are often positive with acute not usually systemically ill, up to 14% of cases may have
disease, culture of bacteria from the lymph nodes of
­ systemic or severe disease [50]. Less common findings
patients with cat scratch disease is difficult as there are include fatigue, headache, hepatosplenomegaly, nausea,
probably few viable B. henselae bacilli present [48]. vomiting, myalgias and arthralgias [63].
Historically, the Th1 immune response has been impli- Various other organs may be involved, including heart,
cated in cat scratch disease, as evidenced by the Bartonella lung, brain, eyes, liver, spleen, kidneys, bone and thy-
skin test, which represents a classic delayed type hyper- roid. B. henselae endocarditis has been reported, particu-
sensitivity reaction [33]. Further studies have shown larly in patients with pre‐existing damage to heart valves
increased production of Th1 cytokines, including [67]. Lung involvement manifests as pleural effusion,
interferon‐γ (IFN‐γ) and IL‐12 [62]. pneumonia and, least commonly, pulmonary nodules
[68]. Neurological changes are relatively rare and present
Clinical features. The primary cutaneous lesion in cat approximately 2 weeks after the onset of cat scratch dis-
scratch disease is most often a papule, pustule or nodule, ease. Encephalopathy, the most common neurological
usually less than 1 cm in size. This lesion occurs at the site manifestation, may present as headache, seizures, coma,
of inoculation after an incubation period that ranges from transient combative behaviour, ataxia, expressive apha-
a few days to a month [13]. Over time, the lesion may sia, transient hemiplegia or hearing loss [69,70]. Other
undergo ­ulceration. Approximately half of patients pre- neurological findings may include neuroretinitis, periph-
sent with the hand or arm as the site of inoculation [13]. eral neuritis, Guillain–Barré syndrome, and chronic
Other sites include the eye, mucous membranes and scalp inflammatory demyelinating polyradiculopathy (CIDP)
[63]. The primary lesion may last for a few days to months [69,71–73]. Parinaud oculoglandular syndrome is an
and usually resolves without treatment. atypical form of cat scratch disease that presents as a
Several weeks after inoculation (range 5–50 days), ­conjunctival granuloma at the site of inoculation with
regional lymphadenopathy proximal to the inoculation preauricular lymphadenopathy [63].
site develops, which is the hallmark of the disease In the liver and spleen, necrotizing granulomas may

SECTION 7: BACTERIAL SKIN


(Fig. 41.3) [63]. In most patients, lymphadenopathy occurs develop, and microabscesses in the kidneys have been
in the head, neck or upper extremity. Although the major- reported [47,74]. Other atypical manifestations of cat
ity of patients present with regional enlarged lymph scratch disease include osteolytic lesions and septic
nodes, approximately 30% of them have involvement of arthritis [63,75]. Lastly, a single case report recently

INFECTIONS
multiple anatomical sites with generalized lymphade- described a paediatric patient who developed autoim-
nopathy in severe cases [60]. The lymphadenopathy is mune thyroiditis associated with cat scratch disease [76].
initially tender, and up to 20% of patients develop sup-
purative lymphadenopathy, which may require drainage Differential diagnosis. The differential diagnosis of cat
or removal [64,65]. It typically resolves spontaneously scratch disease includes various causes of lymphadenop-
within 2–4 months [13]. athy, ranging from infectious to malignant. Tender lym-
Associated signs and symptoms include fever and phadenopathy may be more suggestive of a different
malaise in at least 30% of patients [63]. Dermatological infectious aetiology, such as Staphylococcus aureus, group
A β‐haemolytic streptococci, anaerobes, atypical myco-
bacteria, Mycobacterium tuberculosis, Francisella tularensis
or Brucella. Fungal aetiologies such as histoplasmosis,
sporotrichosis, toxoplasmosis and nocardial infection
may also need to be considered. Viral infections, such as
cytomegalovirus, HIV and Epstein–Barr virus, usually
cause generalized lymphadenopathy, which is less com-
mon in cat scratch disease. Noninfectious diagnoses to
consider include sarcoidosis, congenital and acquired
cysts, Kawasaki disease and Kikuchi disease. Lastly,
malignancy may need to be ruled out, especially in
patients with symptoms that are slower to resolve [66].

Laboratory and histology findings. The diagnosis of cat


scratch disease is primarily clinical. Historically, the diag-
nosis could be made if three of the following four criteria
Fig. 41.3  Inoculation lesion of cat scratch disease on the right cheek and were met: history of cat exposure with presence of a
associated submandibular lymphadenopathy in an infant. Source: Courtesy scratch or primary dermal, ocular or mucous membrane
of Dr. Andrew Margileth. lesion; a positive cat scratch disease skin test; negative
480 Section 7  Bacterial Skin Infections

studies for other causes of lymphadenopathy; and a when an outbreak of severe anaemia and fever occurred in
­characteristic biopsy sample [77]. In general, the sensitivity workers constructing a railway from Lima to Oroya [81]. The
of culture and PCR is low for detecting B. henselae in patients disease subsequently became known as Oroya fever, but the
with cat scratch disease (13% and 30%), whereas serological aetiology remained unknown. Then, in 1885, a Peruvian
diagnosis reaches up to 90% sensitivity [48]. Recent studies medical student named Daniel Carrion inoculated himself
have focused on more specific methods of serological diag- with material from a verruga peruana lesion. He died after
nosis, including an IgG ELISA that uses sarcosin‐soluble developing symptoms of Oroya fever, now known as the
proteins of B. henselae which reached almost 98% specificity acute phase of bartonellosis. Carrion’s experiment suggested
[78]. Still, it is recommended that serological studies be used that both Oroya fever and verruga peruana were two phases
as an adjunct to clinical findings. of a disease caused by one p­ athogen, leading to the eponym
Histological findings in the primary cutaneous inocula- Carrion disease [82]. The infectious agent was finally discov-
tion site include necrosis in the dermis with multiple sur- ered in 1905, when Alberto Barton identified B. bacilliformis
rounding layers of palisading histiocytes and epithelioid within erythrocytes [82].
cells and a peripheral zone of lymphocytes. In lymph
nodes, the findings may be nonspecific depending on the Epidemiology and  pathogenesis. B. bacilliformis is a
stage of disease. Initially, lymphoid hyperplasia is present. small, motile, pleomorphic coccobacillus [83]. Unlike
Subsequently, stellate granulomas may form with similar B. henselae and B. quintana, B. bacilliformis is a flagellated
morphology to those in the skin. Lastly, microabscesses organism. Although the flagella may play a role in host
develop and may become confluent. Warthin–Starry stain- colonization, its importance has not yet been clearly dem-
ing demonstrates pleomorphic bacilli within areas of onstrated [27]. B. bacilliformis exhibits a preference for
necrosis in the cutaneous and lymph node lesions [13]. colonizing cooler areas of the body, such as the vascular
bed of the skin [27].
Treatment. Cat scratch disease is usually a self‐limited Humans are the main reservoir for B. bacilliformis, and
infection with an excellent prognosis. Patients usually do person‐to‐person transmission occurs via phlebotomine
not require antibiotic therapy, and symptoms resolve sandflies (Lutzomyia species) [1]. Cases of bartonellosis are
within 2–4 months [66]. For immunocompetent mildly ill geographically limited to areas in Peru, Colombia and
patients, recommended management includes ruling out Ecuador. In outbreaks of bartonellosis, the paediatric popu-
other causes and providing supportive care. Suppurative lation has been most affected with the highest mortality rate
lymph nodes can be treated with needle aspiration or compared to other age groups [80]. When the acute phase of
SECTION 7: BACTERIAL SKIN

removal if necessary. Alternatively, for children with bartonellosis occurs during pregnancy, higher mortality has
large, bulky lymphadenopathy, therapy can be consid- been reported in both mother and fetus, and vertical trans-
ered with azithromycin at 10 mg/kg on day one followed mission from mother to baby has been reported [80,84].
by 4 days at 5 mg/kg per day dosing [50]. A single pro- Additionally, bartonellosis was reported as the cause of
INFECTIONS

spective randomized double‐blind placebo‐controlled death in an aplastic anaemia patient who developed shock
trial demonstrated a more rapid reduction in lymph node and died after receiving numerous transfusions [85].
volume with the use of azithromycin compared to p ­ lacebo Compared to B. henselae and B. quintana, the intraeryth-
in the first month of treatment [79]. Treatment with rocytic phase of infection is relatively short but much
trimethoprim–sulfamethoxazole, rifampicin, ciprofloxa-
­ more severe, leading to a potentially life‐threatening drop
cin or gentamicin has been observed to be efficacious as in haematocrit. Deformation factor, or deformin, helps
well [77]; however, a recent meta‐analysis combining the B.  bacilliformis establish erythrocyte tropism by causing
results from both studies found that treatment had no pitting and invagination of the cell membranes.
­significant effect in terms of time to achieve cure or cure Additionally, B. bacilliformis expresses haemolysin and an
rate, because all patients in both arms were cured [51]. invasion‐associated gene, which help with erythrocyte
For complicated cat scratch disease, including neurore- tropism [27]. B. bacilliformis is known to cause a transient
tinitis and encephalopathy, a combination of doxycycline cellular immunosuppression due to alterations in the
and rifampicin for 4–6 weeks is recommended [1]. number and function of T cells, which predisposes to
Suspected Bartonella culture‐negative endocarditis should infections like salmonellosis, toxoplasmosis, tuberculosis,
be treated with a 2‐week course of intravenous (IV) gen- reactivated tuberculosis and pneumococcal pneumonia
tamicin and 6‐week course of IV ceftriaxone with or without [80]. The pathogenesis of the eruptive phase of bartonel-
doxycycline for 6 weeks. Documented culture‐positive losis is not well characterized, but the current under-
B. henselae endocarditis requires similar courses of doxy- standing is that verruga ­ peruana is characterized by
cycline and IV gentamicin [50]. angioblastic hyperplasia, loss of cell‐to‐cell contact, acti-
vation of Langerhans cells and positive f­actor VIII in
Bartonellosis endothelial cells [80]. Recently, a new Bartonella species,
B.  ancashensis, has been isolated from the blood of two
History. Bartonellosis was described in Peru in the pre‐ verruga peruana patients [86].
Colombian era, and the first written reports of the disease
were by Spanish conquerors in the 1500s, describing lesions Clinical features. Bartonellosis exhibits two clinical
now typical of verruga peruana [80]. All references to the dis- phases: an acute phase (Oroya fever) and an eruptive
ease mentioned only cutaneous manifestations until 1870, phase (verruga peruana). The acute phase results from
Chapter 41  Bartonella Infections 481

invasion of the bloodstream by B. bacilliformis. Clinically, Verruga peruana usually appears 2–8 weeks after the
this manifests with fever, hepatosplenomegaly, pallor, patient has recovered from Oroya fever, but many patients
jaundice, lymph node enlargement and a systolic present with no prior symptoms. The cutaneous lesions in
murmur. Laboratory evaluation demonstrates severe
­ verruga peruana display three potential morphologies
haemolytic anaemia, leucocytosis, thrombocytopenia (Figs 41.4–41.6). First, miliary lesions present as multiple,
and hepatic involvement. Almost 70% of patients with small, pruritic, erythematous papules, most commonly
acute‐phase bartonellosis experience at least one located on the lower extremities. Involvement of the mucous
­complication [82]. Complications may include menin- membranes may also occur. Second, nodular lesions are
geal signs, seizures, infections, arthralgias, hyperbiliru- larger and fewer in number. Lastly, mular lesions are
binaemia in newborns, congestive heart failure, ­typically isolated, erythematous, deep‐seated lesions that
myocarditis, pericarditis and multiorgan dysfunction may bleed easily [82]. The most common associated signs
[80]. Weight loss and severe ­malnutrition are common and symptoms in the eruptive phase include bleeding
in children, and respiratory infections have been found of the lesion, fever, malaise, arthralgias, pallor, fever and
to be the most common paediatric complication of lymphadenopathy [80].
Oroya fever [80]. The acute phase of bartonellosis can be
fatal in 10% of patients [82].
Differential diagnosis. Acute Oroya fever should be
­ istinguished from other systemic haemotropic bacterial
d
infections. The differential diagnosis for verruga peruana
includes bacillary ­ angiomatosis, Kaposi sarcoma, pyo-
genic granulomas and acquired angiomas [82].

Laboratory and histology findings. Available diagnostic


tests for bartonellosis include blood culture, IFA, soni-
cated immunoblot, PCR, ELISA, thin blood smear and
western blot. Thin blood smear is the fastest and least
expensive method; however, a recent systematic review

SECTION 7: BACTERIAL SKIN


(a)

INFECTIONS

(b)
Fig. 41.4  (a and b) Isolated facial lesions of verruga peruana in Peruvian Fig. 41.5  Multiple vascular papules of verruga peruana on the legs of a
children. Source: Courtesy of Dr. Hector Caceres‐Rios, Instituto de Salud 15‐year‐old boy from Peru. Source: Courtesy of Dr. Hector Caceres‐Rios,
del Niño, Lima, Peru. Instituto de Salud del Niño, Lima, Peru.
482 Section 7  Bacterial Skin Infections

having symptoms, suggesting that IFA may be better for


detecting past infection [87].
Histology of verruga peruana reveals capillary and
endothelial proliferation, similar to pyogenic granuloma
(Fig.  41.7) [88]. An inflammatory infiltrate with neutro-
phils, histiocytes, plasma cells, lymphocytes and mast
cells is present (Fig.  41.8). Despite use of special stains,
organisms are usually not detected on histology.
Immunohistochemical staining reveals strong positivity
for factor VIII‐related antigen [88]. Electron microscopy
demonstrates B. bacilliformis extracellularly in the stroma,
although it has been reported to be phagocytosed in neu-
trophils, histiocytes and endothelial cells [89].

Treatment and  prevention. Chloramphenicol (dose of


75 mg/kg per day divided into 4 doses for 14 days) with a
β‐lactam is the treatment of choice for Oroya fever. In
­children aged 7–12, ciprofloxacin can be used at 250 mg
Fig. 41.6  Conjunctival lesion of verruga peruana in a girl from Peru. Source: twice daily for 10 days. For verruga peruana, the pre-
Courtesy of Dr. Hector Caceres‐Rios, Instituto de Salud del Niño, Lima, Peru.
ferred treatment is rifampicin 10 mg/kg per day for
2 weeks [50]. An observational study compared treatment
with rifampicin and streptomycin and found no statisti-
cally significant difference in achievement of good
response; however, the intramuscular route of adminis-
tration of streptomycin makes its use problematic in
­children [50,51]. Blood transfusions and other supportive
measures are necessary for severely anaemic patients.
Prevention of the disease is mostly through insecticides
for control of sandflies.
SECTION 7: BACTERIAL SKIN

Acknowledgement
We would like to thank Diana B. McShane, Heidi H. Kong
and Sarah A. Myers for their contributions and acknowl-
INFECTIONS

edge use of material from their chapter in the third e­ dition


entitled ‘Bartonella Infections: Bacillary Angiomatosis, Cat
Fig. 41.7  Histopathology of verruga peruana skin lesion showing pyogenic
Scratch Disease and Bartonellosis’.
granuloma‐like appearance. Source: Courtesy of Dr. Hector Caceres‐Rios,
Instituto de Salud del Niño, Lima, Peru.

References
1 Angelakis E, Raoult D. Pathogenicity and treatment of Bartonella
infections. Int J Antimicrob Agents 2014;44:16–25.
2 Jacomo V, Kelly PJ, Raoult D. Natural history of Bartonella infections
(an exception to Koch’s postulate). Clin Diagn Lab Immunol
2002;9:8–18.
3 Maguiña C, Gotuzzo E. Bartonellosis. New and old. Infect Dis Clin
North Am 2000;14:1–22, vii.
4 Stoler MH, Bonfiglio TA, Steigbigel RT et al. An atypical subcuta-
neous infection associated with acquired immune deficiency syn-
drome. Am J Clin Pathol 1983;80:714–8.
5 Cockerell CJ, Whitlow MA, Webster GF et al. Epithelioid angiomatosis:
a distinct vascular disorder in patients with the acquired immunodefi-
ciency syndrome or AIDS‐related complex. Lancet 1987;2:654–6.
6 LeBoit PE, Berger TG, Egbert BM et al. Bacillary angiomatosis. The
histopathology and differential diagnosis of a pseudoneoplastic
infection in patients with human immunodeficiency virus disease.
Am J Surg Pathol 1989;13:909–20.
Fig. 41.8  Histopathology of verruga peruana demonstrating capillary 7 Relman DA, Loutit JS, Schmidt TM et al. The agent of bacillary angio-
matosis. An approach to the identification of uncultured pathogens.
proliferation and mixed cellular infiltrate. Source: Courtesy of Dr. Hector
N Engl J Med 1990;323:1573–80.
Caceres‐Rios, Instituto de Salud del Niño, Lima, Peru. 8 Regnery RL, Anderson BE, Clarridge JE et al. Characterization of a
novel Rochalimaea species, R. henselae sp. nov., isolated from blood
reported that the sensitivity is very low despite having of a febrile, human immunodeficiency virus‐positive patient. J Clin
Microbiol 1992;30:265–74.
high specificity [87]. IFA was found to have good sensitiv-
9 Welch DF, Pickett DA, Slater LN et al. Rochalimaea henselae sp. nov., a
ity and specificity; however, as much as 45% of the popu- cause of septicemia, bacillary angiomatosis, and parenchymal bacillary
lation in endemic areas may have antibodies without peliosis. J Clin Microbiol 1992;30:275–80.
Chapter 41  Bartonella Infections 483

10 Slater LN, Welch DF, Hensel D et al. A newly recognized fastidious 39 Spach DH. Bacillary angiomatosis. Int J Dermatol 1992;31:19–24.
gram‐negative pathogen as a cause of fever and bacteremia. N Engl 40 Smith KJ, Skelton HG, Tuur S et  al. Bacillary angiomatosis in an
J Med 1990;323:1587–93. immunocompetent child. Am J Dermatopathol 1996;18:597–600.
11 Koehler JE, Quinn FD, Berger TG et  al. Isolation of Rochalimaea 41 Paul MA, Fleischer AB, Wieselthier JS et al. Bacillary angiomatosis in
species from cutaneous and osseous lesions of bacillary angiomato- an immunocompetent child: the first reported case. Pediatr Dermatol
sis. N Engl J Med 1992;327:1625–31. 1994;11:338–41.
12 Brenner DJ, O’Connor SP, Winkler HH et  al. Proposals to unify the 42 Tucci E, Della Rocca C, Santilli F. Localized bacillary angiomatosis in
genera Bartonella and Rochalimaea, with descriptions of Bartonella the oral cavity: observations about a neoplasm with atypical behavior.
quintana comb. nov., Bartonella vinsonii comb. nov., Bartonella Description of a case and review of the literature. Minerva Stomatol
henselae comb. nov., and Bartonella elizabethae comb. nov., and to 2006;55:67–75.
remove the family Bartonellaceae from the order Rickettsiales. Int J 43 López de Blanc S, Sambuelli R, Femopase F et al. Bacillary angiomato-
Syst Bacteriol 1993;43:777–86. sis affecting the oral cavity. Report of two cases and review. J Oral
13 Nosal JM. Bacillary angiomatosis, cat‐scratch disease, and bartonel- Pathol Med 2000;29:91–6.
losis: what’s the connection? Int J Dermatol 1997;36:405–11. 44 Perkocha LA, Geaghan SM, Yen TS et  al. Clinical and pathological
14 Adal KA, Cockerell CJ, Petri WA. Cat scratch disease, bacillary features of bacillary peliosis hepatis in association with human immu-
angiomatosis, and other infections due to Rochalimaea. N Engl J nodeficiency virus infection. N Engl J Med 1990;323:1581–6.
Med 1994;330:1509–15. 45 Foltzer MA, Guiney WB, Wager GC et al. Bronchopulmonary bacil-
15 Malane MS, Laude TA, Chen CK et al. An HIV‐1‐positive child with lary angiomatosis. Chest 1993;104:973–5.
fever and a scalp nodule. Lancet 1995;346:1466. 46 Baron AL, Steinbach LS, LeBoit PE et al. Osteolytic lesions and bacil-
16 Chitsike I, Muronda C. Bacillary angiomatosis in an HIV positive lary angiomatosis in HIV infection: radiologic differentiation from
child. First case report in Zimbabwe. Cent Afr J Med 1997;43:238–9. AIDS‐related Kaposi sarcoma. Radiology 1990;177:77–81.
17 Wanat KA, Reid E, Kamiyango W et al. Tumoral bacillary angiomato- 47 Liston TE, Koehler JE. Granulomatous hepatitis and necrotizing sple-
sis in a child with human immunodeficiency virus. JAMA Dermatol nitis due to Bartonella henselae in a patient with cancer: case report
2014;150:1015–16. and review of hepatosplenic manifestations of bartonella infection.
18 Rostad CA, McElroy AK, Hilinski JA et  al. Bartonella henselae‐­ Clin Infect Dis 1996;22:951–7.
mediated disease in solid organ transplant recipients: two pediatric 48 La Scola B, Raoult D. Culture of Bartonella quintana and Bartonella
cases and a literature review. Transpl Infect Dis 2012;14:E71–81. henselae from human samples: a 5‐year experience (1993 to 1998).
19 Myers SA, Prose NS, Garcia JA et  al. Bacillary angiomatosis in a J Clin Microbiol 1999;37:1899–905.
child undergoing chemotherapy. J Pediatr 1992;121:574–8. 49 Koehler JE, Tappero JW. Bacillary angiomatosis and bacillary peliosis
20 Zarraga M, Rosen L, Herschthal D. Bacillary angiomatosis in an in patients infected with human immunodeficiency virus. Clin Infect
immunocompetent child: a case report and review of the literature. Dis 1993;17:612–24.
Am J Dermatopathol 2011;33:513–15. 50 Rolain JM, Brouqui P, Koehler JE et al. Recommendations for treat-
21 Bellissimo‐Rodrigues F, da Fonseca BA, Martinez R. Bacillary angio- ment of human infections caused by Bartonella species. Antimicrob
matosis in a pregnant woman. Int J Gynaecol Obstet 2010;111:85–6. Agents Chemother 2004;48:1921–33.
22 Riley LE, Tuomala RE. Bacillary angiomatosis in a pregnant patient with 51 Prutsky G, Domecq JP, Mori L et al. Treatment outcomes of human
acquired immunodeficiency syndrome. Obstet Gynecol 1992;79:818–19. bartonellosis: a systematic review and meta‐analysis. Int J Infect
23 Breitschwerdt EB, Maggi RG, Farmer P et al. Molecular evidence of Dis 2013;17:e811–19.

SECTION 7: BACTERIAL SKIN


perinatal transmission of Bartonella vinsonii subsp. berkhoffii and 52 Wear DJ, Malaty RH, Zimmerman LE et al. Cat scratch disease bacilli
Bartonella henselae to a child. J Clin Microbiol 2010;48:2289–93. in the conjunctiva of patients with Parinaud’s oculoglandular syn-
24 Magalhães RF, Pitassi LH, Salvadego M et al. Bartonella henselae sur- drome. Ophthalmology 1985;92:1282–7.
vives after the storage period of red blood cell units: is it transmissible 53 Debre R, Lamy M, Jammet ML et  al. Cat scratch disease. Sem Hop
by transfusion? Transfus Med 2008;18:287–91. 1950;26:1895–904.

INFECTIONS
25 Ohl ME, Spach DH. Bartonella quintana and urban trench fever. Clin 54 English CK, Wear DJ, Margileth AM et  al. Cat‐scratch disease.
Infect Dis 2000;31:131–5. Isolation and culture of the bacterial agent. JAMA 1988;259:
26 Magnolato A, Pederiva F, Spagnut G et al. Three cases of Bartonella 1347–52.
quintana infection in children. Pediatr Infect Dis J 2015;34:540–2. 55 Brenner DJ, O’Connor SP, Hollis DG et al. Molecular characterization
27 Minnick MF, Battisti JM. Pestilence, persistence and pathogenicity: and proposal of a neotype strain for Bartonella bacilliformis. J Clin
infection strategies of Bartonella. Future Microbiol 2009;4:743–58. Microbiol 1991;29:1299–302.
28 Raoult D, Roux V. The body louse as a vector of reemerging human 56 Zangwill KM, Hamilton DH, Perkins BA et al. Cat scratch disease
diseases. Clin Infect Dis 1999;29:888–911. in Connecticut. Epidemiology, risk factors, and evaluation of a new
29 Boutellis A, Veracx A, Angelakis E et al. Bartonella quintana in head diagnostic test. N Engl J Med 1993;329:8–13.
lice from Sénégal. Vector Borne Zoonotic Dis 2012;12:564–7. 57 Dolan MJ, Wong MT, Regnery RL et  al. Syndrome of Rochalimaea
30 Regnery RL, Olson JG, Perkins BA et  al. Serological response to henselae adenitis suggesting cat scratch disease. Ann Intern Med
“Rochalimaea henselae” antigen in suspected cat‐scratch disease. 1993;118:331–6.
Lancet 1992;339:1443–5. 58 Kordick DL, Hilyard EJ, Hadfield TL et al. Bartonella clarridgeiae, a
31 Tappero JW, Mohle‐Boetani J, Koehler JE et al. The epidemiology of newly recognized zoonotic pathogen causing inoculation papules,
bacillary angiomatosis and bacillary peliosis. JAMA 1993;269:770–5. fever, and lymphadenopathy (cat scratch disease). J Clin Microbiol
32 Koehler JE, Glaser CA, Tappero JW. Rochalimaea henselae infection. A 1997;35:1813–18.
new zoonosis with the domestic cat as reservoir. JAMA 1994;271:531–5. 59 Margileth AM, Baehren DF. Chest‐wall abscess due to cat‐scratch dis-
33 Mosepele M, Mazo D, Cohn J. Bartonella infection in immunocom- ease (CSD) in an adult with antibodies to Bartonella clarridgeiae: case
promised hosts: immunology of vascular infection and vasoprolif- report and review of the thoracopulmonary manifestations of CSD.
eration. Clin Dev Immunol 2012;2012:612809. Clin Infect Dis 1998;27:353–7.
34 Kyme PA, Haas A, Schaller M et  al. Unusual trafficking pattern of 60 Moriarty RA, Margileth AM. Cat scratch disease. Infect Dis Clin
Bartonella henselae‐containing vacuoles in macrophages and North Am 1987;1:575–90.
endothelial cells. Cell Microbiol 2005;7:1019–34. 61 Gonzalez BE, Correa AG, Kaplan SL. Cat‐scratch disease occurring in
35 Schmid MC, Scheidegger F, Dehio M et  al. A translocated bacterial three siblings simultaneously. Pediatr Infect Dis J 2003;22:467–8.
protein protects vascular endothelial cells from apoptosis. PLoS 62 Resto‐Ruiz S, Burgess A, Anderson BE. The role of the host immune
Pathog 2006;2:e115. response in pathogenesis of Bartonella henselae. DNA Cell Biol
36 Siamer S, Dehio C. New insights into the role of Bartonella effector 2003;22:431–40.
proteins in pathogenesis. Curr Opin Microbiol 2015;23:80–5. 63 Shinall EA. Cat‐scratch disease: a review of the literature. Pediatr
37 Chiaraviglio L, Duong S, Brown DA et al. An immunocompromised Dermatol 1990;7:11–18.
murine model of chronic Bartonella infection. Am J Pathol 64 Munson PD, Boyce TG, Salomao DR et al. Cat‐scratch disease of the
2010;176:2753–63. head and neck in a pediatric population: surgical indications and out-
38 Mohle‐Boetani JC, Koehler JE, Berger TG et al. Bacillary angiomatosis comes. Otolaryngol Head Neck Surg 2008;139:358–63.
and bacillary peliosis in patients infected with human immunodefi- 65 Ridder GJ, Boedeker CC, Technau‐Ihling K et al. Cat‐scratch disease:
ciency virus: clinical characteristics in a case‐control study. Clin Infect Otolaryngologic manifestations and management. Otolaryngol Head
Dis 1996;22:794–800. Neck Surg 2005;132:353–8.
484 Section 7  Bacterial Skin Infections

66 Margileth AM. Dermatologic manifestations and update of cat 78 Tsuruoka K, Tsuneoka H, Kawano M et al. Evaluation of IgG ELISA
scratch disease. Pediatr Dermatol 1988;5:1–9. using N‐lauroyl‐sarcosine‐soluble proteins of Bartonella henselae for
67 Fournier PE, Lelievre H, Eykyn SJ et  al. Epidemiologic and clinical highly specific serodiagnosis of cat scratch disease. Diagn Microbiol
characteristics of Bartonella quintana and Bartonella henselae endo- Infect Dis 2012;74:230–5.
carditis: a study of 48 patients. Medicine 2001;80:245–51. 79 Bass JW, Freitas BC, Freitas AD et al. Prospective randomized double
68 Bandyopadhyay A, Burrage LC, Gonzalez BE. Pulmonary nodules in blind placebo‐controlled evaluation of azithromycin for treatment
an immunocompetent child with cat scratch disease. Pediatr Infect of cat‐scratch disease. Pediatr Infect Dis J 1998;17:447–52.
Dis J 2013;32:1390–2. 80 Huarcaya E, Maguiña C, Torres R et  al. Bartonelosis (Carrion’s
69 Carithers HA, Margileth AM. Cat‐scratch disease. Acute encephalopa- Disease) in the pediatric population of Peru: an overview and
thy and other neurologic manifestations. Am J Dis Child update. Braz J Infect Dis 2004;8:331–9.
1991;145:98–101. 81 Ricketts WE. Clinical manifestations of Carrión’s disease. Arch Intern
70 Fox JW, Studley JK, Cohen DM. Recurrent expressive aphasia Med 1949;84:751–81.
as  a  presentation of cat‐scratch encephalopathy. Pediatrics 2007; 82 Maguina C, Garcia PJ, Gotuzzo E et  al. Bartonellosis (Carrión’s
119:e760–3. ­disease) in the modern era. Clin Infect Dis 2001;33:772–9.
71 Massei F, Gori L, Taddeucci G et  al. Bartonella henselae infection 83 Kosek M, Lavarello R, Gilman RH et al. Natural history of infection
associated with Guillain‐Barre syndrome. Pediatr Infect Dis J
­ with Bartonella bacilliformis in a nonendemic population. J Infect Dis
2006;25(1):90–1. 2000;182:865–72.
72 Metz CH, Buer J, Bornfeld N et al. Bilateral Bartonella henselae neuro- 84 Tuya XL, Escalante‐Kanashiro R, Tinco C et al. Possible vertical trans-
retinitis with stellate maculopathy in a 6‐year‐old boy. Infection mission of Bartonella bacilliformis in Peru. Am J Trop Med Hyg
2012;40:191–4. 2015;92:126–8.
73 Mascarelli PE, Maggi RG, Hopkins S et  al. Bartonella henselae 85 Magalhães RF, Urso Pitassi LH, Lania BG et al. Bartonellosis as cause
infection in a family experiencing neurological and neurocognitive of death after red blood cell unit transfusion. Ultrastruct Pathol
abnormalities after woodlouse hunter spider bites. Parasit Vectors 2009;33:151–4.
2013;6:98. 86 Mullins KE, Hang J, Jiang J et al. Description of Bartonella ancashensis
74 Lin CC, Chi H, Tsai JD. Renal microabscesses: a presentation of cat sp. nov., isolated from the blood of two patients with verruga peru-
scratch disease. J Pediatr 2015;166:1544.e1. ana. Int J Syst Evol Microbiol 2015;65:3339–43.
75 Mirouse G, Journe A, Casabianca L et al. Bartonella henselae osteoar- 87 Sanchez Clemente N, Ugarte‐Gil CA, Solórzano N et al. Bartonella
thritis of the upper cervical spine in a 14‐year‐old boy. Orthop bacilliformis: a systematic review of the literature to guide the
Traumatol Surg Res 2015;101:519–22. research agenda for elimination. PLoS Negl Trop Dis 2012;6:e1819.
76 Chiuri RM, Matronola MF, Di Giulio C et al. Bartonella henselae infec- 88 Arias‐Stella J, Lieberman PH, Erlandson RA. Histology, immunohis-
tion associated with autoimmune thyroiditis in a child. Horm Res tochemistry, and ultrastructure of the verruga in Carrion’s disease.
Paediatr 2013;79:185–8. Am J Surg Pathol 1986;10:595–610.
77 Margileth AM. Antibiotic therapy for cat‐scratch disease: clinical 89 Bhutto AM, Nonaka S, Hashiguchi Y et  al. Histopathological and
study of therapeutic outcome in 268 patients and a review of the electron microscopical features of skin lesions in a patient with
­
literature. Pediatr Infect Dis J 1992;11:474–8. ­bartonellosis (verruga peruana). J Dermatol 1994;21:178–84.
SECTION 7: BACTERIAL SKIN
INFECTIONS
 485

C HA PTER   42

Mycobacterial Skin Infections


G. Sethuraman1, Tanvi Dev1 & V. Ramesh2
Department of Dermatology, All India Institute of Medical Sciences, New Delhi, India
1 

Department of Dermatology, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
2 

Introduction, 485 Leprosy in children, 496 Nontuberculous mycobacterial


Cutaneous tuberculosis, 486 infections, 497

Abstract ­ isseminated lesions are quite common in children compared to


d
adults. Systemic infection is also frequent in children. Several
­unusual patterns of skin tuberculosis are recognized in the paedi-
Mycobacterial skin infections are important to recognize in the
atric population. Leprosy in children is still prevalent (>100 cases a
paediatric population as they can cause serious morbidity. They
year are reported) in some of the African countries, the Dominican
include tuberculosis, leprosy and various nontuberculous myco-
Republic, the eastern Mediterranean and the western Pacific
bacterial infections. Of these, both tuberculosis and leprosy have
­region, especially Micronesia. Borderline tuberculoid leprosy and
caused a heavy disease burden among children, especially in devel-
indeterminate leprosy are the two common forms seen in children
oping countries, owing to several factors such as malnutrition,
and the lesions are usually macular. Nontuberculous mycobacterial
poverty, poor living conditions and emerging human immunodefi-
infections are related to environmental bacteria. The incidence of
ciency virus (HIV) infection. Cutaneous tuberculosis, mostly caused
these infections is increasing, especially in tropical climates. They
by Mycobacterium tuberculosis, can have varied clinical manifesta-
cause several clinical syndromes such as cervical lymphadenopa-
tions depending on the host immune response. Children usually
thy, skin and soft tissue infections following injury and systemic
acquire the infection from infected adults. Skin tuberculosis in
diseases.
­children can be severe with a protracted course. Multifocal and

Key points

SECTION 7: BACTERIAL SKIN


unusual variants are common in children. Scrofuloderma
progressing to scrofulous gumma, a localized disseminated form
and coexistence of several forms of tuberculosis are frequent in
• Cutaneous tuberculosis and leprosy are two important
children. Drug resistance is also an emerging problem.
mycobacterial infections in children.
• An underlying systemic focus of tuberculosis is more frequent in

INFECTIONS
• Skin tuberculosis is widely prevalent in South‐East Asian
children compared to adults.
countries, especially those that are underdeveloped.
• Leprosy in children commonly presents as the macular form, and
• Scrofuloderma is the most frequent form of skin tuberculosis,
borderline tuberculoid leprosy is frequent.
followed by lupus vulgaris. The occurrence of tuberculids, in
• Nontuberculous mycobacteria cause a spectrum of clinical
particular lichen scrofulosorum, has increased in recent years.
syndromes including cervical lymphadenopathy, injury‐related
• Compared to adults, children with skin tuberculosis have severe
skin infections and soft tissue infections. Buruli ulcer occurs
disease with significant morbidity and disability. Multifocal
mostly in Africa.
involvement with dissemination is common in children. Several

Introduction M. tuberculosis and M. bovis [1]. Besides human and bovine


Mycobacteria are slender aerobic rods that stain weakly types, MTC also includes M. africanum and M. microti.
positive with Gram stain. They have a waxy cell wall Among the MTC, M. tuberculosis is responsible for
composed of mycolic acid, which makes them acid fast, most  cases of tuberculosis in humans. M. bovis can be
i.e. they retain the staining even on treatment with a mix- pathogenic for animals, especially dairy cows. Hence,
ture of acid and alcohol. This acid‐fast nature is demon- consumption of unpasteurized milk can lead to develop-
strated by the Ziehl–Neelsen stain. Broadly, the genus ment of oropharyngeal and intestinal tuberculosis.
Mycobacterium is divided into Mycobacterium tuberculosis M. africanum infection is common in tropical Africa [1,2].
complex (MTC), Mycobacterium leprae and atypical or M. leprae causes leprosy, a chronic granulomatous
nontuberculous mycobacteria (NTM) (Table  42.1). MTC ­infection of the skin and peripheral nerves [3]. NTM, on
consists of two important tubercular bacilli, namely the other hand, encompass a variety of species which are

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
486 Section 7  Bacterial Skin Infections

Table 42.1  Classification of mycobacteria Table 42.2  Classification of skin tuberculosis in children

Organism Disease Primary tuberculosis


Tuberculous chancre
Mycobacterium tuberculosis complex Miliary tuberculosis
Multibacillary
M. tuberculosis Tuberculosis Secondary tuberculosis
M. bovis Tuberculosis Scrofuloderma
Mycobacterium leprae Leprosy Tuberculous gumma
Nontuberculous mycobacteria Orificial tuberculosis
Rapid growers Lupus vulgaris Paucibacillary

M. abscessus Localized nodules, abscesses, draining sinuses, Tuberculosis verrucosa cutis


M. fortuitum injection abscess, infection of the surgical Tuberculids
M. chelonae wounds Micropapular – lichen scrofulosorum
Slow growers Papular – papulonecrotic tuberculid
M. marinum Swimming pool granuloma Nodular – nodular tuberculid (erythema
M. ulcerans Buruli ulcer induratum)
M. avium complex Cervicofacial lymphadenopathy, disseminated
infections in HIV cases
M. haemophilum Cervicofacial lymphadenopathy, skin and soft
tissue infection
M. scrofulaceum Cervicofacial lymphadenopathy
especially lichen scrofulosorum, have been emerging as
M. kansasii Skin and soft tissue infection the second most common form of skin tuberculosis in the
M. szulgai Skin and soft tissue infection recent years, especially in developing countries [8,16].

Classification. Cutaneous tuberculosis may occur as a


ubiquitous in nature. Infections with these mycobacteria result of exogenous inoculation (tuberculous chancre,
have a wide spectrum of presentation, including lymphad- tuberculosis verrucosa cutis and rarely lupus vulgaris) or
enopathy, pulmonary, skin and soft tissue infections, and endogenous spread. The latter may develop either from
even disseminated disease, especially in immunosup- contiguous (scrofuloderma, tuberculosis orificialis cutis),
pressed individuals [1]. haematogenous (acute military tuberculosis, tuberculous
In this chapter, the skin infections caused by these three gumma, lupus vulgaris) or lymphatic spread. Based on
SECTION 7: BACTERIAL SKIN

major groups of mycobacteria will be discussed. the bacterial load it can be divided into multibacillary or
paucibacillary forms. The simple working classification of
Cutaneous tuberculosis paediatric skin tuberculosis is true tuberculosis and tuber-
culids. True tuberculosis is further subdivided into pri-
INFECTIONS

Cutaneous tuberculosis is an uncommon form of extrapul- mary and secondary forms depending on previous
monary tuberculosis. It is most commonly caused by sensitization to M. tuberculosis [7,17] (Table 42.2).
M. tuberculosis and rarely by M. bovis [4]. The clinical spec-
trum of paediatric skin tuberculosis is diverse. Most cases Lupus vulgaris
of skin tuberculosis in children have been reported from Lupus vulgaris (LV) is the most common form of skin
developing countries, in particular the South Asian tuberculosis [15]. However, in many paediatric skin
Association for Regional Co‐operation (SAARC) nations tuberculosis series it is the second most common type
and African countries. Skin tuberculosis in children is reported [7–10,12,15,16]. LV occurring as a result of
quite severe with significant morbidity and disability. ­systemic haematogenous spread of M. tuberculosis affects
the head and neck region. In tropical countries like India,
Epidemiology. Cutaneous tuberculosis accounts for about LV can also develop due to exogenous inoculation on the
0.5% of the dermatoses in Europe [4], 1.3% in Ethiopia [5], gluteal region, knees, feet and hands [7,17].
2% in Morocco [6], 0.1–0.5% in India [7–10] and 0.066– LV classically presents as slowly progressive, irregular
0.04% in Hong Kong [11]. In Ethiopia, paediatric skin and infiltrative plaques with characteristic central
tuberculosis accounted for 24% of all cutaneous tubercu- atrophic scarring and advancing margins (Figs 42.1–42.3).
losis cases [5]. In Morocco, 60% of skin tuberculosis cases The erythematous elevated edge of the lesion may show
were under the age of 30 years [6]. In Pakistan, 45% of several soft, translucent, erythematous papules and/or
cutaneous tuberculosis patients were children <10 years coalescent plaques, which on diascopy reveal yellow‐
of age, and another 37% were aged between 10 and brown apple‐jelly nodules. These nodules represent
20  years [12]. In Japan, cutaneous tuberculosis was microscopic granulomas. This particular subtype, also
observed in all age groups before 1980. Interestingly, no known as plane LV, predominantly affects the head and
case has been reported in the age group 0–19 years neck [15].
between 1981 and 2002 [13]. In India, skin tuberculosis in LV occurring on the extremities and buttocks due to the
children accounts for 18.7–54% of all cases of skin tuber- exogenous route may show longstanding larger lesions
culosis [8,14–16]. with excessive hyperkeratosis. The epidermal hyperpro-
The two most common forms of paediatric skin tuber- liferation in keratotic or psoriasiform LV is possibly due
culosis are scrofuloderma and lupus vulgaris. Tuberculids, to immunological events influenced by the presence of
Chapter 42  Mycobacterial Skin Infections 487

Fig. 42.1  Lupus vulgaris: large erythematous and infiltrated plaques with Fig. 42.2  Lupus vulgaris: large erythematous and hyperkeratotic plaque
keloidal scarring. involving bilateral gluteal region causing anal stricture.

SECTION 7: BACTERIAL SKIN


INFECTIONS
(a) (b)
Fig. 42.3  (a and b) Lupus vulgaris: multifocal involvement involving bilateral gluteal region and left knee.

concomitant chronic pyogenic infections in hot and Genital involvement is usually associated with scarring,
humid tropical climates [15]. mutilation and ulceration [15].
The natural course of LV is characterized by its chronic Several unusual variants of LV have been described.
indolent behaviour. The lesions of LV can progress to The authors have described symmetrical LV involving
become very large, sometimes affecting the entire ana- both the knees, possibly due to exogenous inoculation of
tomical region. During its course, chronic secondary mycobacteria, and also symmetrical LV developing in the
infections are common, resulting in crusting and ulcera- vicinity of a scrofuloderma sinus due to autoinoculation
tion. Such lesions may heal with hypertrophic, keloidal or tuberculosis [18,19]. Other variants include sporotrichoid,
atrophic scarring (Fig. 42.2). Sometimes, LV may result in kissing LV involving the gluteal cleft, and multifocal
irreversible contractures, such as deformities of nasal or lesions [7,17,20]. Rarely LV can also develop following
ear cartilage, flexion contractures of extremities, etc. bacillus Calmette–Guérin (BCG) vaccination [21].
Rarely, squamous cell carcinoma may develop in long- Histopathology of LV usually reveals psoriasiform epi-
standing LV [8,15,16]. dermal hyperplasia and a diffuse upper dermal granu-
Involvement of the genitalia is uncommon. Both vulval lomatous reaction composed predominantly of epithelioid
and scrotal involvement has been described in children. cells. Acid‐fast bacilli (AFB) are usually not detected [7,17].
488 Section 7  Bacterial Skin Infections

Scrofuloderma with extensive swellings, ulcers and sinus tracts forming


Scrofuloderma (SFD) is the most common pattern of skin finger‐like extensions leading to a condition known as
tuberculosis in children. It is due to the contiguous spread scrofulous gumma, which is mostly reported from India
of tuberculous infection from an underlying focus. Lymph (Fig. 42.8) [15].
nodes are the most frequently associated focus followed The authors have described symmetrical SFD involving
by bone and joints, testes and lacrimal glands. Among the bilateral ankles with underlying tuberculosis of the bones
lymph nodes, cervical and inguinal groups are often [19]. Disseminated SFD affecting the left leg and foot,
involved. Other lymph nodes such as supraclavicular, right hand and inguinal region has been reported in a
axillary, occipital, parasternal, submandibular and malnourished child who also had widespread tubercu-
epitrochlear groups may also be affected [7,17]. lous infection of several joints, lungs and cold abscess
SFD begins as a subcutaneous cold abscess (Fig. 42.4) [22]. SFD occurring in a segmental pattern is uncommon
which, over a period of time, ruptures to form an ulcer or (Fig.  42.9). Tuberculous cold abscess or SFD developing
sinus tracts (Fig. 42.5). These ulcers are typically shallow in association with other forms of skin tuberculosis such
with bluish margins and undermined edges (Fig.  42.6). as LV (Fig. 42.10), tuberculosis verrucosa cutis and lichen
There may be one or several ulcers in the affected area scrofulosorum is also recognized [7,17].
(Fig. 42.7). These ulcers and sinus tracts may heal sponta- An underlying systemic focus, including lymph nodes,
neously and leave behind the characteristic puckered lungs, intestine and bones, has been reported in about
scars that are the hallmark of healed SFD. In some cases 46% of cases of paediatric skin tuberculosis [8,23].
several adjoining groups of lymph nodes are involved A skin biopsy should be taken from the edge of the
ulcer or the sinus tract. It shows mixed cell granulomas
composed of epithelioid cells admixed with acute
inflammatory cells. Necrosis is often identified, which
may show AFB on Ziehl– Neelsen (ZN) staining [17].
SECTION 7: BACTERIAL SKIN
INFECTIONS

Fig. 42.4  Scrofuloderma: tuberculous cold abscess on the anterior neck.

Fig. 42.6  Scrofuloderma: a large shallow ulcer with bluish margin on the


clavicular area.

Fig. 42.5  Scrofuloderma: tuberculous cold abscess with multiple sinuses Fig. 42.7  Scrofuloderma: multiple ulcers overlying a tuberculous
on the inguinal region. osteomyelitic focus on the wrist.
Chapter 42  Mycobacterial Skin Infections 489

(b)

(a)

SECTION 7: BACTERIAL SKIN


INFECTIONS
(c) (d)
Figs 42.8  (a–d) Scrofulous gumma. Tuberculous cold abscesses involving inguinal, cervical and axillary groups of lymph nodes with extensive swellings,
ulcers and sinus tracts forming finger‐like extensions.

Tuberculosis verrucosa cutis also occur in children. It is often difficult to distinguish it


Tuberculosis verrucosa cutis, otherwise known as warty from the hypertrophic variant of LV. However, there is
tuberculosis, accounts for 12–15% of paediatric skin tuber- much more verrucosity, whereas there is much less mutila-
culosis. It occurs as a consequence of exogenous inocula- tion than in LV [7,15,17].
tion of M. tuberculosis in previously sensitized individuals.
Clinically, it is characterized by the presence of warty or Tuberculous gumma
verrucous plaques on the acral extremities (Fig.  42.11). Tuberculous gumma, also known as tuberculous meta-
Rarely, the scrotum and gluteal region can also be affected. static abscess, results from haematogenous dissemination
The surface may show fissuring that may extrude pus, and of the organism from a primary focus during periods of
there is often perilesional erythema. Lesions can be unilat- lowered resistance. It is often seen in malnourished and
eral or bilateral. A preceding history of trauma is often immunocompromised children. It is considered as a severe
present. Tuberculosis verrucosa cutis is mostly solitary, variant of SFD as the gummatous lesions resemble SFD
however bilateral, multifocal or segmental patterns can clinically, histologically and bacteriologically. Typically
490 Section 7  Bacterial Skin Infections

Orificial tuberculosis
Orificial tuberculosis occurs as a result of autoinoculation
of M. tuberculosis in sputum or stool onto the skin. It is
often seen in immunocompromised children. Clinically it
presents as painful and refractory necrotic ulcers in the
perioral and perianal regions. Underlying pulmonary
and intestinal tuberculosis is usually associated [7,17].

Sporotrichoid cutaneous tuberculosis


Sporotrichoid tuberculosis is a rare form of skin tuberculo-
sis that has been reported in 3% of all cases with cutaneous
tuberculosis. It mimics the subcutaneous linear lymphang-
itic form of sporotrichosis and sporotrichoid NTM.
Fig. 42.9  Scrofuloderma in a segmental pattern over the right thoracic Sporotrichoid tuberculosis occurs mainly by lymphatic
chest wall (T4). spread, which would explain the linear arrangement of
the lesions in the same limb. In all the published reports,
the sporotrichoid lesions have been described mostly in
children and at times in young adults. The efficient lym-
phatic drainage in children and their physical activity pre-
disposing them to trauma may be responsible for this rare
lymphangitic form of skin tuberculosis [20,24,25].
Sporotrichoid tuberculosis is called inverse sporotrichoid
when the proximal half of the lower limb is affected due
to retrograde lymphatic spread from the inguinal lymph
nodes [20].

Tuberculids
Tuberculids are considered to be delayed hypersensitivity
reactions to M. tuberculosis in individuals with strong
SECTION 7: BACTERIAL SKIN

immunity. Children with tuberculids often have an under-


lying focus of tuberculosis. The key diagnostic criteria
include the following: (i) absence of the organisms in the
lesional smear or culture; (ii) strongly positive Mantoux
INFECTIONS

Fig. 42.10  Unilateral tuberculosis showing cold abscess involving the groin reaction; (iii) presence of concomitant or past tuberculo-
and multifocal lupus vulgaris on the sole. sis; and (iv) clinical resolution with antitubercular treat-
ment. Although M. tuberculosis is not identified in either
smear or culture, polymerase chain reaction (PCR) tech-
niques have demonstrated mycobacterial DNA in lesional
biopsies of tuberculids [7,17].
Tuberculids can be broadly divided into true and facul-
tative tuberculids. Lichen scrofulosorum and papulone-
crotic tuberculids are true tuberculids, in which M.
tuberculosis significantly contributes to the disease patho-
genesis. Facultative tuberculids (erythema induratum
and erythema nodosum) have diverse aetiological factors
and M. tuberculosis is one of them [7,17].

Lichen scrofulosorum
Lichen scrofulosorum (LS) is characterized by lichenoid
eruption of minute papules in children and adolescents
with tuberculosis. Hebra initially described it in 1868.
Subsequently, many investigators have reported several
Fig. 42.11  Tuberculosis verrucosa cutis on the sole showing verrucous cases of LS across the world. Most LS cases are due to
plaques. pulmonary or extrapulmonary infection with M. tubercu-
losis [26,27]. It has also been reported after BCG vaccina-
tion. Rarely atypical mycobacteria such as M. szulgai and
tuberculous gumma is characterized by multiple, non- M. avium‐intracellulare complex can also cause LS [28].
tender, subcutaneous nodules that may break down to LS was a rare tuberculid, but according to reports from
form deep punched‐out ulcers and sinuses (Figs  42.12 developing countries, the prevalence of LS is increasing.
and 42.13) [7,15,17]. In two different studies from India, LS was the second
Chapter 42  Mycobacterial Skin Infections 491

SECTION 7: BACTERIAL SKIN


INFECTIONS
Fig. 42.12  (a and b) Tuberculous gumma showing multiple punched‐out ulcers with undermined edges and bluish margins.

most common pattern of paediatric skin tuberculosis, have also been described, and may be a marker of
seen in 23.5% and 33% respectively [8,16]. ­ isseminated systemic tuberculosis (Fig. 42.16) [32].
d
Clinically, LS is characterized by an asymptomatic In a large prospective study of 39 cases of LS, 82% were
eruption of lichenoid or skin‐coloured follicular and patients younger than 15 years. Twenty two (56.4%) were
­perifollicular papules, ranging from 0.5 to 3 mm in diam- boys while 17 (43.6%) were girls. The trunk was the com-
eter (Figs  42.14 and 42.15). They are mostly seen over monest site affected. An underlying tuberculous focus
the abdomen, chest, back and proximal extremities. was observed in 28 (72%) cases. Of these, 13 (33%) had
Occasionally other body sites such as genitalia, palms and lymph node tuberculosis (submandibular and cervical),
soles can also be affected. The lesions may last several 11 (28%) had pulmonary involvement, three had tubercu-
months and heal spontaneously without any scarring. loma of the brain, and one had spinal tuberculosis along
Several unusual variants such as micropustular, nodulo‐ with paravertebral cold abscess. Six patients (15%) also
ulcerative, and annular discoid plaques have been had associated cutaneous tuberculosis. All the patients
reported [26,27,29–31]. Widespread psoriasiform plaques had a strongly positive Mantoux reaction [27].
492 Section 7  Bacterial Skin Infections

Fig. 42.13  Tuberculous gumma with ocular involvement.

Fig. 42.15  Lichen scrofulosorum showing large lichenoid scaly plaques.


SECTION 7: BACTERIAL SKIN
INFECTIONS

(a)

Fig. 42.14  Lichen scrofulosorum showing lichenoid follicular papules on


the trunk.

Histologically, LS reveals noncaseating, epithelioid cell


granulomas in the upper dermis and also around the der-
mal appendages.

Papulonecrotic tuberculids
Papulonecrotic tuberculid (PNT) is a rare entity of tuber- (b)
culids. Most cases of PNT have been reported from South Fig. 42.16  (a and b) Lichen scrofulosorum showing psoriasiform plaques
Africa. In the largest series of 91 cases of PNT seen over a with pustules.
Chapter 42  Mycobacterial Skin Infections 493

(a) (b)
Fig. 42.17  (a and b) Papulonecrotic tuberculid showing papulonecrotic ulcers and varioliform scars over trunk and extremities.

vulva, perineum and trunk [35]. PNT occurring in associa-


tion with disseminated LV has also been described [36].
The vast majority of children with PNT have associated
pulmonary tuberculosis or phlyctenular conjunctivitis.

SECTION 7: BACTERIAL SKIN


Histologically, early lesions show leucocytoclastic vascu-
litis with a wedge‐shaped dermoepidermal infarct. Older
lesions may show noncaseating ill‐formed granulomas
along with lymphohistiocytic vasculitis. Immunologically,

INFECTIONS
PNT is considered as an Arthus reaction followed by a
delayed‐type hypersensitivity reaction to the intravascular
release of M. tuberculosis. During the process of immuno-
logical reactions, bacillary emboli are opsonized with anti-
bodies and complement. The subsequent complement
activation and polymorphonuclear leucocyte activation
results in the release of proteolytic enzymes leading to vas-
cular necrosis. The consequent mononuclear cell response
Fig. 42.18  Papulonecrotic tuberculid involving the glans penis with a destroys the bacilli. Sometimes the tubercle bacilli may sur-
healed sinus and a punched‐out scar. vive, leading to the development of LV [33].

period of 17 years, 26% of the cases were younger than 10 Erythema induratum of Bazin
years of age, and 21% were between 11 and 20 years [33]. Erythema induratum of Bazin (EIB) is rare in children.
However, Jordaan et  al. were the first group from the The pathogenesis appears to be similar to that of PNT, but
same region to describe in detail the clinicopathological EIB affects larger vessels. Clinically, it presents on the calf
findings of eight children with PNT [34]. as deep‐seated reddish blue nodules that break down to
PNT affects children and young adults. Typically the form an ulcer that heals with atrophic scarring [37].
lesions are distributed symmetrically on the acral extremi- Simultaneous occurrence of PNT and EIB has been
ties, ears and extensor aspect of the joints. Clinically, the described in a child [38]. Histologically, EIB shows a lobu-
lesions are papulonecrotic and/or crusted ulcers, measur- lar panniculitis with vasculitis. In about 50% of the cases
ing 1–5 mm, which may heal with atrophic or varioliform mycobacterial DNA can be isolated by PCR [37].
scars over several weeks (Fig. 42.17a and b). Continuous Nodular tuberculid is a variant in which there is
crops of new lesions may develop over a period of several involvement of deep dermis and superficial fat, in con-
months or years [34]. Involvement of genitalia is not trast to the deeper involvement in EIB. The inflamed ves-
uncommon with deep punched‐out ulcers healing with sels in nodular tuberculid are present in the superficial fat
disfigurement (Fig. 42.18). Ramdial et al. described PNT in at the dermoepidermal junction, whereas in EIB they are
a child who had adult-pattern involvement of the face, situated at a deeper level [37].
494 Section 7  Bacterial Skin Infections

Erythema nodosum than 2 years, diagnosis of tuberculous infection should


Erythema nodosum is a nonspecific entity characterized therefore not rely on the result of IGRA alone [40].
by painful subcutaneous nodules on the shins that last
for  about 2 weeks and heal with hyperpigmentation. Histopathology
Histology shows septal panniculitis without vasculitis. The characteristic histological features of cutaneous
Several aetiological factors are associated with erythema tuberculosis include the presence of tuberculoid granulo-
nodosum [7,17] (see Chapter 102). mas consisting of epithelioid cells, giant cells and lym-
phocytes. The overall clinicopathological concordance is
BCG and cutaneous tuberculosis noted in 64–85% of the cases. In general, the presence of
The BCG vaccine is a live attenuated vaccine derived from diffuse dermal epithelioid cell granulomas along with
M. bovis. It is given soon after birth and is recommended in epidermal hyperplasia suggests either LV or tuberculosis
all tuberculosis endemic countries excluding the USA and verrucosa cutis. These are paucibacillary forms in which
parts of Europe [21]. Usually it is safe to administer BCG; AFB are often not seen. In SFD and gummatous tubercu-
however local reactions are known to occur. Rarely there losis, there would be dermal mixed cell granulomas com-
may be persistent ulceration, infection, abscess and keloid posed of epithelioid cells and histiocytes admixed with
formation. A few cases may develop maculopapular rash neutrophils and eosinophils. Necrosis is often seen and
and erythema nodosum. Paradoxically, a few children AFB can be detected [7,17].
may develop LV, SFD and tuberculids [7,17].
Studies have shown that the BCG vaccine prevents Culture
approximately 50% of pulmonary tuberculosis and Culture positivity varies from 5% to 55% [17]. Vashisht
50–80% of severe and disseminated tuberculosis, includ- et al. identified the organism in 36.8% and 13.6% of biop-
ing meningeal involvement. Studies on skin tuberculosis sies in SFD and LV respectively [16]. With the use of mul-
have shown no difference in the clinical spectrum or out- tiple media and Kirchner’s liquid medium for storing the
come of skin tuberculosis in BCG‐vaccinated and unvac- tissue biopsy, the yield of organisms is better. In a com-
cinated children [7]. parative study of the radiometric BACTEC™ 460 TB cul-
ture system and Lowenstein–Jensen (LJ) medium for the
isolation of M. tuberculosis in 35 patients with cutaneous
Diagnosis. Diagnosis is usually based on the classical tuberculosis, 26 isolates of the organism were identified.
clinical morphology and supported by laboratory investi- Of these, four (11.4%) grew on LJ medium, 17 (48.5) on the
gations [39]. Demonstration of M. tuberculosis in the tissue
SECTION 7: BACTERIAL SKIN

BACTEC™ system, and five on both the media. It was


smear or biopsy is the gold standard in the diagnosis of found that the sensitivity of BACTEC™ system was sig-
skin tuberculosis. In general, all children with skin tuber- nificantly better than that of LJ medium (62.8% vs. 25.7%,
culosis should be investigated for a tuberculous focus in P <0.002). The combined sensitivity for isolation on both
INFECTIONS

the lungs, lymph nodes, bone and joints, nervous system the media was 74.3% [41].
and other relevant organs. Fine needle aspiration cytology (FNAC) is a simple and
rapid diagnostic procedure. The aspirated air‐dried smear
Screening tests can be stained with Giemsa and ZN stain. The cytomor-
The tuberculosis skin test (TST, formerly the Mantoux test) phological features in LV include cohesive epithelioid cell
is a good screening test to detect the presence or absence of granulomas along with chronic inflammatory cells, while
tuberculous infection. The test consists of an intradermal in SFD there will be predominant caseous necrosis with or
injection of 0.1 mL (5 TU) of purified protein derivative on without granulomas along with acute inflammatory cells
the volar aspect of the forearm, and reading is performed [42]. With FNAC, the identification of AFB is easier, espe-
after 48 hours. An induration of 10 mm or more is sugges- cially in SFD and lymph node tuberculosis. In an Ethiopian
tive of infection or disease. Strongly positive reactions or study of 143 cases (including children) with SFD, AFB
bullous reactions are seen in tuberculids. The test may be were identified in all the cases by ZN staining [5]. In
negative in disseminated military tuberculosis or coexist- another study of 19 cases of SFD, AFB were identified in
ent human immunodeficiency virus (HIV) infection. 79% of cases in the aspirate compared to only 16% in the
False‐positive results are observed in BCG‐vaccinated biopsy specimens [42]. Hence, FNAC can be used as an
children. Interferon‐γ release assays (IGRA) are based on alternative diagnostic test in cutaneous tuberculosis, par-
the release of interferon by blood monocytes and lympho- ticularly SFD associated with lymph node tuberculosis.
cytes upon stimulation by highly specific antigens of
M. tuberculosis. They are equally sensitive but more specific PCR
than TST and can thus avoid false‐positive results due to PCR amplification may be a useful tool for the detection
BCG vaccination or exposure to NTM. Combination of of mycobacterial DNA and is particularly helpful to dif-
TST and IGRA may increase the detection rate of children ferentiate tuberculosis from other granulomatous condi-
at high risk of progression from tuberculous infection to tions such as sarcoidosis, fungal granulomas and NTM.
tuberculous disease [40]. In children >5 years IGRA are However, there are conflicting reports of its usefulness in
applicable in all situations where TST is indicated. the diagnosis of skin tuberculosis. Several of the common
However, the sensitivity of IGRA in younger children is forms of skin tuberculosis have been confirmed by
currently not sufficiently established. In children younger PCR by various investigators. IS6110 based conventional
Chapter 42  Mycobacterial Skin Infections 495

Table 42.3  Antitubercular therapy (ATT)

Drug Dosage/kg/day Side‐effects Monitoring

First‐line drugs
Isoniazid (H) 10 (7–15) mg/kg/day, Hepatotoxicity Liver function test
max. 300 mg/day Peripheral neuropathy
Ataxia
Acne
Rifampicin (R) 15 (10–20) mg/kg/day, Hepatotoxicity Liver function test
max. 600 mg/day Orange‐coloured body fluids
Pruritus
Drug interactions (P450)
Pyrazinamide (Z) 35 (30–40) mg/kg/day, Hyperuricaemia Renal function test
max. 2000 mg Hepatotoxicity
Arthralgia
Rash, photosensitivity, pruritus
Ethambutol (E) 20 (15–25) mg/kg/day, Retrobulbar neuritis Fundus examination (contraindicated
max. 2000 mg Rash in younger children)
Second‐line drugs
Amikacin 15 mg/kg /day Ototoxicity Renal and auditory function test
single dose IM or IV Nephrotoxicity
Kanamycin 15/kg/day IM
one or two divided doses
Streptomycin 20–40 mg/kg/day IM
Max. 1000 mg/day
Ciprofloxacin 15–30 mg/kg/day PO in two GI (nausea, vomiting, abdominal CNS evaluation and
doses discomfort, diarrhoea) electrocardiographic monitoring,
Levofloxacin 10 mg/kg/day CNS (headache, dizziness, rarely blood sugar levels
Ofloxacin 10 mg/kg/day in two doses hallucinations, delirium and seizures)
Gatifloxacin 10 mg/kg/day single oral dose Skin rash/ photosensitivity
Prolongation of QT interval
Drugs for nontuberculous mycobacteria

SECTION 7: BACTERIAL SKIN


Clarithromycin 15 mg/kg/day q. 12 hourly oral GI intolerance No evaluation required
Cotrimoxazole 5–8 mg/kg of TMP and Haematological side‐effects, GI intolerance, Haemogram, liver and renal
(trimethoprim [TMP]– 25–50 mg/kg of SMZ per hypersensitivity reaction, vasculitis, renal functional tests
sulfamethoxazole [SMZ]) day q. 12 hourly oral damage, CNS disturbance rarely

INFECTIONS
Doxycycline 5 mg/kg/day q. 12 hourly oral GI intolerance, photosensitivity, hepatic Liver and renal function tests
(not in children <8 years) and renal toxicity, discolouration of teeth
Max. 200 mg/day Cutaneous pigmentation with minocycline
Minocycline 4 mg/kg/day q. 12 hourly (not
in children <8 years)
Max. 200 mg/day

CNS, central nervous system; GI, gastrointestinal; IM, intramuscular; IV, intravenous; PO, per os (oral).

PCT/nested PCR has been found to be useful in the diagnosis treatment with ATT [7,17]. Dosing schedule, side‐effects
of skin tuberculosis and superior to 16S rRNA gene based and monitoring evaluation are given in Table 42.3.
PCR [43,44]. However, others have not found PCR to be a
useful complement to the clinical and histological diagno- Drug resistance
sis [45]. Hence, the sensitivity and specificity of PCR need Multidrug resistance (MDR) is an emerging problem in
better validation in order to recommend it for diagnosing children with skin tuberculosis. Multidrug‐resistant
cutaneous tuberculosis in the routine clinical setting. strains are isolates that are resistant to rifampicin and
INH on drug sensitivity testing, with or without resist-
Treatment. The standard antitubercular therapy (ATT) ance to other drugs. MDR should be suspected in children
for skin tuberculosis consists of an initial 2 months of with poor or no response to first‐line ATT with clinical
intensive‐phase regimen with four drugs (ethambutol [E], deterioration and where other causes of treatment failure
isoniazid [INH, H], rifampicin [R] and pyrazinamide [Z]), are not proven. The predisposing factors include high dis-
followed by 4 months of maintenance with two drugs ease burden, endemicity of tuberculosis and emerging
(HR) (2 EHRZ/4HR). Pyridoxine (10–25 mg/day) should HIV co‐infection.
also be given in order to prevent INH‐related neuropathy. MDR has been reported in all common forms of skin
Children with underlying bony or other organ tuberculo- tuberculosis, namely LV, SFD and tuberculosis verrucosa
sis or associated HIV infection need longer periods of cutis [46–48]. Aggarwal et  al., in their study of drug
496 Section 7  Bacterial Skin Infections

s­ usceptibility testing conducted in Chennai, South India, Table 42.4  The proportion of childhood leprosy cases reported in different
showed resistance to one or more ATT for 12 (46.2%) of tropical countries
the 26 mycobacterial isolates. Of the 15 isolates from LV,
six resistant isolates were identified, and of the nine iso- Region Period of Age group Proportion
reporting (years) of childhood
lates from SFD, five resistant isolates including four MDR
leprosy (%)
were identified [41].
Culture and drug susceptibility testing should be done India
in all suspected cases of MDR cutaneous tuberculosis and • South India [51,52] 1990–1995 <14 33–34
these patients should be treated with second‐line ATT for • Delhi [53] 1992–2003 <15 7.71
a period of 18 months (Table 42.3). • Delhi [54] 2000–2009 <14 9.6
• Chandigarh [55] 2001–2011 <18 4.8
Brazil (Aracaju) [56] 2001–2012 <15 8.3
Leprosy in children Colombia [57] 1994–2000 <15 7
Southern Ethiopia [50] 1999–2011 <15 7.4
Leprosy is a chronic infectious disease caused by M. lep-
rae, which is a Gram‐positive obligate intracellular organ-
ism that has an affinity for Schwann cells and cells of the
reticuloendothelial system, where it occurs in clumps or reported in different tropical countries are summarized
globi. Leprosy affects mainly the skin and peripheral in Table 42.4 [50–57].
nerves, leading to physical disabilities. Clinically it mani- A history of familial contact has been reported in 0.66–
fests itself along a spectrum with two poles, namely 47% of cases [58]. This may be a concern as it implies con-
tuberculoid and lepromatous, and borderline forms in tinuing transmission of the disease.
between [2,3].
Clinical spectrum of leprosy in children. In India, hypo-
pigmented macules are the most commonly observed
Epidemiology. Leprosy has been a major public health skin lesions. Plaques are also seen, but nodular lesions are
problem but with implementation of multidrug therapy uncommon. All five types defined under the Ridley–
in 1985, the prevalence of leprosy reduced drastically by Jopling classification, namely TT, BT, BB, BL and LL, can
as much as 45%. With strong and effective control meas- occur in children (Box 42.1 and Figs 42.19–42.25). However
ures by the World Health Organization (WHO) and the most common form of leprosy reported in various
SECTION 7: BACTERIAL SKIN

national‐level programmes worldwide, the goal of elim- studies from India is BT leprosy [51–55,58,59]. In a series
ination of leprosy (defined as a reduction of leprosy of 59 cases, BT leprosy was seen in 67.8%, followed by LL
prevalence to <1 case per 10  000 population) was in 12% and BL in 10%. Twenty three cases (39%) had a
achieved at global level by the year 2000. Most countries single skin lesion, 2–5 lesions were seen in 12 (20%), and
INFECTIONS

also reached the goal of elimination at national level by >5 lesions or diffuse infiltration in 22 (37%) of the cases.
2005 [49]. Thickened nerves were observed in 48 (81.4%), of whom
The global leprosy prevalence at the end of the first 44% had single nerve and 56% had more than one nerve
quarter of 2014 was 0.32 per 10 000 population, with the trunk involvement. Disability was noted in 24 (40%) chil-
maximum prevalence of 0.62 per 10 000 population in the dren [55]. In another series of 138 cases from South India,
South East Asia Region (SEAR). The new case detection BT leprosy was seen in 66%, followed by indeterminate
rate (during 2013) globally was 3.81 per 100 000 popula- leprosy (IL) in 19.4% and TT in 10% [59]. In Brazil, pauci-
tion. SEAR has the highest number of new cases and bacillary leprosy (182, 68.4%) is more common than multi-
accounts for 72% of the global leprosy burden. Fourteen bacillary leprosy (84, 31.6%). Tuberculoid (95, 35.7%) and
main endemic countries, which include six each from IL (92, 34.6%) forms are the two most commonly reported.
SEAR and Africa and one each from America and the A total of 24 children had deformities [56]. In southern
western Pacific region, reported >1000 new cases in 2013. Ethiopia, multibacillary leprosy was the most common
India reported the highest number of new leprosy cases type, seen in 95% of the children and 84% of adolescents.
(126 913) followed by Brazil (31  044) and Indonesia Six (27.3%) children and 18 (28.6%) adolescents had
(16 856). India alone accounts for 58.85% of the global deformities [50].
­leprosy burden [49]. Overall clinicopathological concordance is seen in 76%
The percentages of children among new leprosy cases of the cases. Slit skin smear positivity ranges from 5% to
in countries reporting ≥100 cases in the different WHO 25%. Lepra reactions have been reported in 1–30% (type 1
regions are as follows: African region  –  Comoros 29%, reactions, 1.2–28%; type 2 reactions, 0.2–5.8%). However,
Niger 0.9%; America  –  Dominican Republic 9.4%, in some studies reactions are not reported. Relapse can
Argentina and Mexico 0.6%; eastern Mediterranean occur in 1–7% of the cases [58,60].
region  –  Yemen 12.3%, Sudan 2.1%; SEAR  –  Indonesia
11.9%, Nepal 4.1%; western Pacific region  –  Micronesia Diagnosis. Diagnosis is based on the classical clini-
39.5%, China 1.5% [49]. cal  morphology supported by slit skin smear and
Overall, several epidemiological studies indicate histopathology. Serological tests (anti‐phenolic gly-
­
that  childhood leprosy accounts for about 5–10% of the colipid I, PGL‐I) are often used to screen for subclinical
cases [50]. The proportions of childhood leprosy cases infections [61].
Chapter 42  Mycobacterial Skin Infections 497

Box 42.1  Spectrum of leprosy

Tuberculoid leprosy (TT) • As the disease progresses, the macules may show infiltration or
papulonodules, especially on the face and ears.
• TT affects skin and peripheral nerves. However TT seldom leads to
• Peripheral nerve involvement is asymmetrical and nerve tenderness
peripheral nerve damage or disability.
and damage is less pronounced than in BT.
• Lesions are dry, anaesthetic and hypopigmented with sharply defined
• Cases of BL that might have downgraded from BT may show features
infiltrated margins and loss of appendages. Erythema may be seen in
of nerve damage or concurrent lesions of BT.
fairer skin types.
• Lesions are usually single or few.
Lepromatous leprosy (LL)

Borderline tuberculoid leprosy (BT) • Numerous small symmetrically distributed normoaesthetic and shiny
macules become more and more infiltrated with disease progression,
• The lesions resemble TT but the marginal definition is less
leading to a waxy appearance.
­pronounced, possibly with pseudopodia or satellite lesions. The
• Sites of predilection for infiltration include the face, particularly
lesions may also show a palpable feeder nerve.
forehead, zygoma, chin and earlobes, and also the cooler dorsal areas
• Number of lesions may be up to 10 or 20.
of the body.
• Hypopigmentation, dryness and anaesthesia tend to be less pronounced.
• Infiltration may lead to loss of skin creases and extreme cases may
• In contrast to TT, nerve damage is severe and much more widespread
result in ‘leonine’ facies.
as reaction is more frequent.
• Immune zones or the warmer areas of the body such as axillae, groin,
• Untreated BT leprosy may continue with episodes of reaction or
midline of back, perineum and scalp are preferentially spared.
deformities, which might eventually heal or may progress towards the
• Symmetrical enlargement of the peripheral nerves with glove and
LL spectrum.
stocking sensory impairment occurs in later stages of disease
progression.
Mid‐borderline leprosy (BB)
• Signs of nerve damage such as wasting of muscles and/or deformities
• This is the unstable part of the spectrum and usually manifests as are an indicator of downgraded spectrum.
reactional states, either upgrading or downgrading. • Systemic involvement can be seen as there is a high bacterial load and
• Has dimorphous features with ‘punched‐out’ inner and sloping outer bacillaemia.
margins (inverted saucer appearance). Sometimes present as
‘geographical’ lesions. Indeterminate leprosy (IL)

SECTION 7: BACTERIAL SKIN


• Lesions tend to occur in symmetry.
• Presents as one or few slightly hypopigmented ill‐defined macules on
• Nerve damage is variable.
the face, buttocks or extensor aspect of extremities.
• In most cases IL lesions heal spontaneously without any sequelae.
Borderline lepromatous leprosy (BL)
• A few cases may progress either to tuberculoid or lepromatous poles.

INFECTIONS
• Earlier lesions are multiple, ill‐defined shiny macules, which are
distinct and tend to show symmetry. The intervening skin is clinically
as well as bacteriologically normal.

Treatment. The WHO recommendations for multidrug


Nontuberculous mycobacterial
therapy in children (10–14 years) are given in Table 42.5. infections
The appropriate dose for children under 10 years of age NTM are a group of AFB which encompass all mycobac-
is as follows: rifampicin, 10 mg/kg monthly; clofazimine, terial species other than M. tuberculosis complex and M.
1 mg/kg daily and 6 mg/kg monthly; dapsone, 2 mg/kg leprae. Unlike the other two groups of mycobacteria,
daily. which are pathogenic to humans, the NTM are mostly
found in soil and water. Most of these organisms are
BCG vaccination and leprosy. The role of the BCG vaccine in nonpathogenic; however some can cause infections in
­
leprosy was first described by Fernandez in 1939 who humans. Based on their cultural characteristics, they are
observed lepromin conversion among lepromin‐negative broadly divided into rapid growers and slow growers
healthy children following BCG administration. Subse- (Table 42.1). The major clinical syndromes associated with
quently several studies have been published with varying NTM infections include: lymphadenitis, pulmonary dis-
ranges of protective efficacy (20–90%). In a meta‐analysis on ease, disseminated disease and skin and soft tissue infec-
the protective aspect of BCG vaccine in the prevention of tions. Of the various skin infections, fish tank or swimming
leprosy, the author has reported that 13 (54.2%) studies dem- pool granuloma caused by M. marinum and Buruli ulcer
onstrated >50% efficacy/effectiveness and five (20.9%) stud- caused by M. ulcerans are the two species‐specific NTM
ies showed >75% protection against leprosy. Since familial disorders [1,2,63,64].
contacts play an important role in transmission in children,
a simple and inexpensive measure like BCG vaccination Epidemiology. The incidence of NTM infections has
may partly help to halt the transmission of the disease [62]. increased in the last two decades owing to increasing
498 Section 7  Bacterial Skin Infections

Fig. 42.19  Borderline tuberculoid leprosy on the face. Source: Courtesy of


Neena Khanna.
Fig. 42.21  Borderline tuberculoid leprosy in reaction on the face, showing
annular erythematous and oedematous plaque with scaling. Source:
Courtesy of Saurabh Singh.
SECTION 7: BACTERIAL SKIN
INFECTIONS

Fig. 42.20  Borderline tuberculoid leprosy with reaction. Source: Courtesy


of Neena Khanna.

nonsurgical cosmetic and implant procedures, wide-


spread use of immunosuppressive therapy and HIV
infection. Most of the published data relating to NTM are
available for adult populations in the developed coun-
tries [1]. Data for the epidemiology of NTM in children
are scarce. The overall incidence of NTM infections in
children varies between 0.3 and 1.9 per 100 000 [65]. In
Fig. 42.22  Borderline tuberculoid leprosy in downgrading reaction
developed countries, epidemiological studies have showing erythematous and oedematous plaques along with multiple
focused mainly on cervicofacial lymphadenitis, which is mid‐borderline (BB) lesions showing inverted saucer appearance on the
the most common clinical syndrome associated with extremities.
Chapter 42  Mycobacterial Skin Infections 499

Fig. 42.23  Mid‐borderline (BB) leprosy showing multiple annular plaques


with inverted saucer appearance on the buttocks. Source: Courtesy of Fig. 42.25  Indeterminate leprosy showing a hypopigmented macule with
Saurabh Singh. ill‐defined margin on the forehead. Source: Courtesy of Saurabh Singh.

SECTION 7: BACTERIAL SKIN


INFECTIONS
(a) (b)
Fig. 42.24  (a and b) Borderline lepromatous leprosy in reaction showing multiple erythematous, oedematous and infiltrated plaques over face and ear.
Source: Courtesy of Saurabh Singh.

NTM in children [1]. Blyth et  al., in their Australian of the cases had lymphadenopathy, mostly in the cervical
study of 102 children with NTM infection, reported region (87%). Of the 27 children who had a positive cul-
lymph node disease to be the most frequent (66.6% of the ture result, M. avium was isolated in 67% [66]. In Sweden,
cases), followed by skin and soft tissue infection and pul- the annual incidence of NTM infection varied from a min-
monary disease in 13.7% each. M. avium intracellulare is imum of 0.06 to a maximum of 5.7 per 100 000 children
the most frequently isolated species (48.5%) among the under 5 years. During the study period from 1969 to 1990,
NTM infections. Rapid growers such as M. abscessus, M. 390 culture‐confirmed cases were studied. The most fre-
chelonae, M. fortuitum and M. peregrinum were responsible quent disease was lymph node and soft tissue infection,
for 66.6% of skin and soft tissue infection, 50% of pulmo- and M. avium intracellulare was the aetiological agent in
nary disease and all bacteraemias [65]. In another pro- 317 (81.28%) cases. In most of the cases the cervicofacial
spective study of 61 children from the Netherlands, 92% group of lymph nodes was involved. Granulomatous
500 Section 7  Bacterial Skin Infections

Table 42.5  World Health Organization multidrug therapy for leprosy in children aged 10–14 years

Rifampicina Dapsoneb Clofaziminec Duration

Paucibacillary leprosy 450 mg monthly 50 mg once daily – 6 months


Multibacillary leprosy 450 mg monthly 50 mg once daily 150 mg monthly + 50 mg alternate days 12 months

For younger children the dose is calculated as follows:


a
 10–20 mg/kg once a month;
b
 1–2 mg/kg/day;
c
 1 mg/kg/day and 4–6 mg/kg once a month.

skin infection due to M. marinum was seen in four cases


(1.02%) and another child had M. avium intracellulare
infection [67].

Clinical spectrum of NTM infections


Cervicofacial lymphadenopathy
Cervicofacial lymphadenopathy is the most frequent
manifestation of NTM infection in immunocompetent
children. The portal of entry of infection is the orophar-
ynx via contaminated water or soil. Typically it affects
children below 5 years of age with unilateral lymphade-
nopathy. The commonly affected lymph nodes are the
Fig. 42.26  Buruli ulcer: a large deep ulcer with undermined edges
jugulodigastric, parotid, preauricular, submandibular involving the lower part of the shin along with oedematous infiltration of
and posterior triangle group. Rarely, the inguinal and the ankle region. Source: Courtesy of Bessie D.
axillary group of nodes is affected. The lymphadenopa-
thy may resolve spontaneously or progress through
SECTION 7: BACTERIAL SKIN

the  following stages in about 50% of the cases: stage 1, Buruli ulcer
painless firm lymphadenopathy; stage 2, fluctuant Buruli ulcer is one of the most devastating infections
­lymphadenopathy due to liquefaction; stage 3, violaceous caused by M. ulcerans and is the third most common
discolouration and thinning of the overlying skin; stage 4, mycobacterial infection worldwide after tuberculosis and
INFECTIONS

rupture of the swelling leading to fistula or sinus tract leprosy. It is mostly seen in the swampy rural areas of
formation [1,66,67]. western and central Africa [69]. It is estimated that more
Most of the cases are due to M. avium intracellulare and than 7000 people develop Buruli ulcer annually with the
some cases are caused by M. haemophilum, especially in highest incidence in Benin, West Africa [70]. Overall, cases
older children. In the latter, multiple groups of lymph have been reported from at least 32 different countries in
nodes are affected [1]. M. scrofulaceum infection accounts Africa, Australia, South‐East Asia, China, the western
for 60% of cases of cervical lymphadenopathy due to Pacific and Central and South America [63]. Risk factors
NTM in India [68]. for Buruli ulcer include living in proximity to slow‐flowing
In immunocompromised cases, lymphadenopathy water bodies, poor wound care and not wearing protective
is  often accompanied by disseminated mycobacterial clothing [63]. The organisms may be directly transmitted
disease [1]. from the environment to the wounded skin or indirect
Whenever NTM lymphadenitis is suspected, complete transmission by an insect vector [1].
diagnostic surgical excision is recommended, which is In Africa, most cases (75%) occur in children 15 years
also curative at the same time. The sensitivity of bacterial or younger, and the lower extremities are often affected
culture is suboptimal (41–80%) while PCR in the lymph [69]. Buruli ulcer begins as preulcerative nodules, plaques
node sample has a sensitivity of up to 72%. For M. haemo- or oedematous infiltrations which later develop into
philum, the media needs to be supplemented with iron deep necrotic ulcers with undermined edges (Fig. 42.26).
sources [1]. Typically the ulcers are painless. The lesions may spread
to an entire limb and the adjoining bones become devital-
Skin and soft tissue infections ized and necrotic with the development of sequestra and
The various NTM that cause localized infections of the osteomyelitis. Local or metastatic osteomyelitis has been
skin and soft tissue include M. fortuitum, M. abscessus, reported in 10% of the cases [69–73]. In a recent large
M. chelonae, M. ulcerans and M. marinum. Of these, Buruli series of 1227 laboratory‐confirmed cases of Buruli ulcer,
ulcers caused by M. ulcerans and fish tank or swimming 57% of patients were younger than 15 years of age.
pool granuloma due to M. marinum are the two species‐ Ninety six percent had one localization and 36% had
specific disorders. The other rapid growers produce lesions of >15 cm in diameter. The clinical spectrum
several nonspecific clinical morphologies [64]. included: ulcers in 805 (66%); plaques in 668 (54%);
Chapter 42  Mycobacterial Skin Infections 501

oedema in 307 (25%); nodules in 42 (3%); and osteomyeli- duration of therapy varies from 6 months to 12 months,
tis in 82 (7%). Permanent residual sequelae were noted in depending on the anatomical sites of involvement,
229 of 1043 (22%) cases who had a follow‐up record; severity and potential toxicity [63].
there was a significant association with multiple lesions
and osteomyelitis [69]. References
Diagnosis is usually made by PCR, culture and histopa- 1 López‐Varela E, García‐Basteiro AL, Santiago B et al. Non‐tuberculous
thology [74]. In a study of 180 cases in Australia, PCR was mycobacteria in children: muddying the waters of tuberculosis
diagnosis. Lancet 2015;3:244–56.
positive in 99% and 95% in lesional swab and biopsy, 2 Jarzembowski JA, Young MB. Nontuberculous mycobacterial infec-
respectively, and the culture yield was low (19% and 47% tions. Arch Pathol Lab Med 2008;132:1333–41.
respectively) [63]. The standard WHO‐recommended 3 Britton WJ, Lockwood DNJ. Leprosy. Lancet 2004;363:1209–19.
4 Fariña MC, Gegundez MI, Piqué E et  al. Cutaneous tuberculosis: a
treatment consists of streptomycin 15 mg/kg per day
clinical, histopathologic, and bacteriologic study. J Am Acad Dermatol
intramuscularly and oral rifampicin 10 mg/kg per day 1995;33:433–40.
for  a period of 8 weeks. It should be supported with 5 Terranova M, Padovese V, Fornari U, Morrone A. Clinical and epide-
complementary treatment that includes wound care,
­ miological study of cutaneous tuberculosis in Northern Ethiopia.
Dermatology 2008;217:89–93.
­surgical intervention, general care of the patient, etc. [69]. 6 Zouhair K, Akhdari N, Nejjam F et  al. Cutaneous tuberculosis in
The detailed recommendation is available in the WHO Morocco. Int J Infect Dis 2007;11:209–12.
treatment guidelines for Buruli ulcer (http://apps.who. 7 Sethuraman G, Ramesh V. Cutaneous tuberculosis in children.
int/iris/bitstream/handle/10665/77771/9789241503402_ Pediatr Dermatol 2013;30:7–16.
8 Pandhi D, Reddy BS, Chowdhary S et  al. Cutaneous tuberculosis
eng.pdf;jsessionid=977D9167EDBCA3FFF20154DF9E9E8 in Indian children: the importance of screening for involvement of
510?sequence=1). internal organs. J Eur Acad Dermatol Venereol 2004;18:546–51.
9 Kumar B, Muralidhar S. Cutaneous tuberculosis: a twenty‐year
­prospective study. Int J Tuberc Lung Dis 1999;3:494–500.
Mycobacterium marinum infection 10 Kumar B, Rai R, Kaur I et  al. Childhood cutaneous tuberculosis:
Swimming pool granuloma or fish tank granuloma due to a  study over 25 years from northern India. Int J Dermatol
M. marinum infection is very rare in children. The infec- 2001;40:26–32.
tion is acquired through abraded skin while handling 11 Chong LY, Lo KK. Cutaneous tuberculosis in Hong Kong: a 10‐year
retrospective study. Int J Dermatol 1995;34:26–9.
aquaria with tropical fish. Clinically it presents as a pain- 12 Bhutto AM, Solangi A, Khaskhely NM et al. Clinical and epidemio-
less papule or nodule at the site of infection. These lesions logical observations of cutaneous tuberculosis in Larkana, Pakistan.
may progress to form ulcers or abscesses. Sporotrichoid Int J Dermatol 2002;41:159–65.
13 Hamada M, Urabe K, Moroi Y, et  al. Epidemiology of cutaneous
lesions can be seen in 20% of the cases. Disseminated tuberculosis in Japan: a retrospective study from 1906 to 2002. Int J

SECTION 7: BACTERIAL SKIN


lesions can develop in immunocompromised cases. Dermatol 2004;43:727–31.
Diagnosis is usually made by PCR and histology [75]. M. 14 Umapathy KC, Begum R, Ravichandran G et  al. Comprehensive
marinum is usually susceptible to antimicrobials such as ­findings on clinical, bacteriological, histopathological and therapeutic
aspects of cutaneous tuberculosis. Trop Med Int Health 2006;11:1521–8.
rifampicin, doxycycline, minocycline, fluoroquinolones,

INFECTIONS
15 Ramesh V, Misra RS, Beena KR et al. A study of cutaneous tubercu-
clarithromycin and co‐trimoxazole (Table 42.3). losis in children. Pediatr Dermatol 1999;16:264–9.
16 Vashisht P, Sahoo B, Khurana N et  al. Cutaneous tuberculosis in
Other skin infections due to NTM children and adolescents: a clinicohistological study. J Eur Acad
Dermatol Venereol 2007;21:40–7.
Infection with the rapidly growing mycobacteria, M. for- 17 Sethuraman G, Ramesh V, Ramam M et  al. Skin tuberculosis in
tuitum, M. abscessus and M. chelonae, can present as local- ­children: learning from India. Dermatol Clin 2008;26:285–94.
ized abscess or draining sinuses at the site of puncture 18 Sethuraman G, Thappa DM, Kurvilla S. Symmetrical reinfection
lupus vulgaris. Pediatr Dermatol 1999;16:488–90.
wounds or open traumatic injuries or fractures. They can 19 Sethuraman G, Kaur J, Nag HL et al. Symmetrical scrofuloderma with
also cause nosocomial infections which include infec- tuberculosis verrucosa cutis. Clin Exp Dermatol 2006;31:475–7.
tions of intravenous or peritoneal catheters, post‐injection 20 Ramesh V. Sporotrichoid tuberculosis. Clin Exp Dermatol 2007;
abscesses, infection after liposuction or any other surgical 32:680–2.
21 Samuel A, Browning J, Campbell J et  al. Bacillus Calmette‐Guérin
procedures. Sometimes they can present as chronic gran- vaccine‐induced lupus vulgaris in a child adopted from China.
­
ulomatous infection of the bone, joints, bursae and ten- Pediatr Dermatol 2007;24:E44–46.
don sheaths, after direct inoculation of the organisms 22 Singal A, Pandhi D, Agrawal SK. Multifocal scrofuloderma with
­disseminated tuberculosis in a severely malnourished child. Pediatr
through accidental injury or puncture wounds [65,66].
Dermatol 2005;22:440–3.
Diagnosis is made by biopsy, culture and PCR. There 23 Kaur S, Thami GP, Kanwar AJ et al. Scrofuloderma, with multiple organ
are no standard treatment guidelines available for pae- involvement in a 5‐year‐old child. Pediatr Dermatol 2001;18:328–31.
diatric infections as systematically collected data on 24 Pau WS, Al Saffar H, Weinstein M et al. Sporotrichoid‐like tuberculosis.
Pediatr Infect Dis J 2009;28:1135–6.
treatment outcomes and also randomized trials are lack- 25 Remenyik E, Nagy B, Kiss M et  al. Sporotrichoid cutaneous
ing in children. Even in adults the data are scarce. The Mycobacterium tuberculosis infection in a child. Acta Derm Venereol
treatment efficacy and outcomes in adults have been 2005;85:375–6.
reported through a few case reports, case series and pro- 26 Beena KR, Ramesh V, Mukherjee A. Lichen scrofulosorum – a series of
eight cases. Dermatology 2000;201:272–4.
spective nonrandomized trials [76–79]. Based on these 27 Singhal A, Bhattacharya SN. Lichen scrofulosorum: a prospective
reports, childhood infections can be treated with a com- study of 39 patients. Int J Dermatol 2005;44:489–93.
bination of multiple chemotherapeutic agents including 28 Komatsu H, Terunuma A, Tabata N et  al. Mycobacterium avium
­infection of the skin associated with lichen scrofulosorum: report of
a macrolide base. Clarithromycin, rifampicin, sulfona- three cases. Br J Dermatol 1999;141:554–7.
mides, clofazimine, fluoroquinolones and minocycline 29 Dandale A, Gupta N, Dhurat R et al. Unusual presentation of lichen
have been tried with variable success (Table  42.3). The scrofulosorum. Indian J Dermatol Venereol Leprol 2013;79:436–8.
502 Section 7  Bacterial Skin Infections

30 Singhal P, Patel PH, Marfatia YS. Lichen scrofulosorum: A diagnosis and clinical characteristics of disease over eleven years from a tertiary
overlooked. Indian Dermatol Online J 2012;3:190–2. care hospital in North India. Lepr Rev 2014;85:296–310.
31 Gandhi V, Vij A, Bhattacharya SN. Lichen scrofulosorum on the 56 Santos VS, Santos LC, Lôbo LV et al. Leprosy and disability in chil-
­genitalia—an unusual presentation. Indian J Dermatol 2007;46:548–9. dren younger than 15 years in an endemic area of northeast Brazil.
32 Kumar U, Sethuraman G, Verma P et al. Psoriasiform type of lichen Pediatr Infect Dis J 2015;34:e44–7.
scrofulosorum: clue to disseminated tuberculosis. Pediatr Dermatol 57 Romero‐Montoya IM, Beltrán‐Alzate JC, Ortiz‐Marín DC et  al.
2011;28:532–4. Leprosy in Colombian children and adolescents. Pediatr Infect Dis J
33 Morrison JG, Fourie ED. The papulonecrotic tuberculide: from 2014;33:321–2.
Arthrus reaction to lupus vulgaris. Br J Dermatol 1974;91:263–70. 58 Palit A, Inamadar AC. Childhood leprosy in India over the past two
34 Jordaan HF, Schneider JW, Schaaf HS et al. Papulonecrotic tuber- decades. Lepr Rev 2014;85:93–9.
culid in children: a report of eight patients. Am J Dermatopathol 59 Sasidharanpillai S, Binitha MP, Riyaz N et  al. Childhood leprosy: a
1996;18:172–85. retrospective descriptive study from Government Medical College,
35 Ramdial PK, Mosam A, Mallett R et al. Papulonecrotic tuberculid in a Kozhikode, Kerala, India. Lepr Rev 2014;85:100–10.
2‐ year‐old girl: with emphasis on extent of disease and presence of 60 Palit A, Inamadar AC, Desai SS et al. Childhood leprosy in the post‐
leucocytoclastic vasculitis. Pediatr Dermatol 1998;15:450–5. elimination phase: data from a tertiary health care hospital in the
36 Senol M, Ozcan A, Aydin A et al. Disseminated lupus vulgaris and pap- Karnataka state of south India. Lepr Rev 2014;85:85–92.
ulonecrotic tuberculid: case report. Pediatr Dermatol 2000;17:133–5. 61 Barreto JG, Guimarães Lde S, Frade MA et al. High rates of undiag-
37 Jordan HF, Schneider JW, Abdulla EA. Nodular tuberculid: a report nosed leprosy and subclinical infection amongst school children in
of four patients. Pediatr Dermatol 2000;17:183–8. the Amazon Region. Mem Inst Oswaldo Cruz 2012;107:60–7.
38 Kim GW, Park HJ, Kim HS et al. Simultaneous occurrence of papulo- 62 Zodpey SP. Protective efficacy of bacillus Calmette Guérin (BCG) vac-
necrotic tuberculid and erythema induratum in a patient with pulmo- cine in the prevention of leprosy: a meta analysis. Indian J Dermatol
nary tuberculosis. Pediatr Dermatol 2013;30:256–9. Venereol Leprol 2007;73:86–93.
39 Ramesh V, Misra RS, Jain RK. Secondary tuberculosis of the skin. 63 Atkins BL, Gottlieb T. Skin and soft tissue infections caused by
Clinical features and problems in laboratory diagnosis. Int J Dermatol ­nontuberculous mycobacteria. Curr Opin Infect Dis 2014;27:137–45.
1987;26:578–81. 64 Bartralot R, García‐Patos V, Sitjas D et  al. Clinical patterns of
40 Starke JR. Interferon gamma release assay for diagnosis of tubercu- ­cutaneous nontuberculous mycobacterial infections. Br J Dermatol
losis infection and disease in children. Pediatrics 2014;134:e1763–73. 2005;152:727–34.
41 Aggarwal P, Singal A, Bhattacharya SN et al. Comparison of the radi- 65 Blyth CC, Best EJ, Jones CA et  al. Nontuberculous mycobacterial
ometric BACTEC 460 TB culture system and Löwenstein‐Jensen infection in children: a prospective national study. Pediatr Infect Dis J
medium for the isolation of mycobacteria in cutaneous tuberculosis 2009;28:801–5.
and their drug susceptibility pattern. Int J Dermatol 2008;47:681–7. 66 Haverkamp MH, Arend SM, Lindeboom JA et  al. Nontuberculous
42 Kathuria P, Agarwal K, Koranne RV. The role of fine‐needle aspiration mycobacterial infection in children: a 2‐year prospective surveillance
cytology and Ziehl Neelsen staining in the diagnosis of cutaneous study in the Netherlands. Clin Infect Dis 2004;39:450–6.
tuberculosis. Diagn Cytopathol 2006;34:826–9. 67 Romanus V, Hallander HO, Wåhlén P et al. Atypical mycobacteria in
43 Ogusku MM, Sadahiro A, Hirata MH et al. PCR in the diagnosis of extrapulmonary disease among children. Incidence in Sweden from
cutaneous tuberculosis. Braz J Microbiol 2003;34:165–70. 1969 to 1990, related to changing BCG‐vaccination coverage. Tuber
44 Obieta MP, Obieta MY, Monje VD. Polymerase chain reaction‐based Lung Dis 1995;76:300–10.
SECTION 7: BACTERIAL SKIN

detection of Mycobacterium tuberculosis DNA in formalin‐fixed, par- 68 Jindal N1, Devi B, Aggarwal A. Mycobacterial cervical lymphadenitis
affin‐embedded skin tissue specimens from Filipino patients. Int J in childhood. Indian J Med Sci 2003;57:12–15.
Dermatol 2010;49:470–2. 69 Vincent QB, Ardant MF, Adeye A et al. Clinical epidemiology of labo-
45 Tan SH, Tan BH, Goh CL et al. Detection of Mycobacterium tubercu- ratory‐confirmed Buruli ulcer in Benin: a cohort study. Lancet Glob
losis DNA using polymerase chain reaction in cutaneous tuberculosis Health 2014;2:e422–30.
INFECTIONS

and tuberculids. Int J Dermatol 1999;38:122–7. 70 Walsh DS, Portaels F, Meyers WM. Buruli ulcer (Mycobacterium
46 Ramesh V, Muralidhar S, Kumar J et  al. Isolation of drug resistant ulcerans infection). Trans R Soc Trop Med Hyg 2008;102:969–78.
tubercle bacilli in cutaneous tuberculosis. Pediatr Dermatol 71 Marion E, Obvala D, Babonneau J et  al. Buruli ulcer disease in
2001;18:393–5. Republic of the Congo. Emerg Infect Dis 2014;20:1070–2.
47 Ramesh V, Sen MK, Nair D et  al. Cutaneous tuberculosis caused 72 Bessis D, Kempf M, Marsollier L. Mycobacterium ulcerans disease
by  multidrug resistant tubercle bacilli: report of three cases. Int J (Buruli ulcer) in Mali: A new potential African endemic country.
Dermatol 2011;50:300–3. Acta Derm Venereol 2015;95:489–90.
48 Ramesh V, Sen MK, Sethuraman G et al. Cutaneous tuberculosis due 73 Boyd SC, Athan E, Friedman ND et al. Epidemiology, clinical features
to multidrug‐resistant tubercle bacilli and difficulties in clinical diag- and diagnosis of Mycobacterium ulcerans in an Australian popula-
nosis. Indian J Dermatol Venereol Leprol 2015;81:380–4. tion. Med J Aust 2012;196:341–4.
49 World Health Organization. Global Leprosy update, 2013; reducing 74 Varma‐Basil M, Garima K, Pathak R et al. Development of a novel PCR
disease burden. Weekly epidemiological record 2014;89:389–400. restriction analysis of the hsp65 gene as a rapid method to screen for
Available from: http://www.who.int/wer (accessed 11 December the Mycobacterium tuberculosis complex and nontuberculous myco-
2018). bacteria in high‐burden countries. J Clin Microbiol 2013;51:1165–70.
50 Ramos JM, Reyes F, Lemma D et al. The burden of leprosy in children 75 Doedens RA, van der Sar AM, Bitter W et  al. Transmission of
and adolescents in rural southern Ethiopia. Paediatr Int Child Health Mycobacterium marinum from fish to a very young child. Pediatr
2014;34:24–8. Infect Dis J 2008;27:81–3.
51 Shetty VP, Ghate SD, Wakade AV et  al. Clinical, bacteriological, 76 Choi WS, Kim MJ, Park DW et al. Clarithromycin and amikacin vs.
and histopathological characteristics of newly detected children with clarithromycin and moxifloxacin for the treatment of post‐acupunc-
leprosy: A population based study in a defined rural and urban area ture cutaneous infections due to Mycobacterium abscessus: a pro-
of Maharashtra, Western India. Indian J Dermatol Venereol Leprol spective observational study. Clin Microbiol Infect 2011;17:1084–90.
2013;79:512–17. 77 Parize P, Hamelin A, Veziris N et al. Induction therapy with line-
52 Selvasekar A, Geetha J, Nisha K et  al. Childhood leprosy in an zolid/clarithromycin combination for Mycobacterium chelonae
endemic area. Lepr Rev 1999;70:21–7. skin infections in immunocompromised hosts. J Eur Acad Dermatol
53 Mahajan S, Sardana K, Bhushan P et al. A study of leprosy in children, Venereol 2016;30:101–5.
from a tertiary pediatric hospital in India. Lepr Rev 2006;77:160–2. 78 Huang YC, Liu MF, Shen GH et al. Clinical outcome of Mycobacterium
54 Singhal A, Sonthalia S, Pandhi D. Childhood leprosy in a tertiary‐care abscessus infection and antimicrobial susceptibility testing. J Microbiol
hospital in Delhi, India: A reappraisal in the post‐elimination era. Immunol Infect 2010;43:401–6.
Lepr Rev 2011;82:259–69. 79 Sethi S, Arora S, Gupta V, Kumar S. Cutaneous Mycobacterium fortui-
55 Dogra S, Narang T, Khullar G et al. Childhood leprosy through the tum infection: successfully treated with amikacin and ofloxacin com-
post‐leprosy‐elimination era: a retrospective analysis of epidemiological bination. Indian J Dermatol 2014;59:383–4.
503

C HA PTER   43

Rickettsial Disease
Arun C. Inamadar & Aparna Palit
Department of Dermatology, Venereology & Leprosy, Sri B.M. Patil Medical College, Hospital & Research Centre, BLDE University, Vijayapur, Karnataka, India

Abstract tiorgan involvement contributes to the morbidity and mortality.


The specific diagnostic test is detection of rising antibody titre in
paired sera by immunofluorescence assay or enzyme‐linked immu­
Rickettsial organisms have a worldwide distribution. They are the
nofluorescence assay (ELISA). The drug of choice is doxycycline,
causative agents for typhus fever and rickettsial spotted fever. The
irrespective of patient age. Rickettsial diseases are difficult to
­
organisms have a complicated lifecycle in lower vertebrates and
­diagnose during the first week because of the lack of specific clini­
blood‐sucking arthropods. Human beings are infected accidentally
cal features and absence of specific diagnostic tests. As the disease
and human‐to‐human transmission does not occur. These organ­
is most responsive to antibiotics during this period, a high index
isms are ‘vasculotropic’, causing selective damage to endothe­
of suspicion and empirical therapy with doxycycline is essential to
lial cells, termed ‘rickettsial vasculitis’. The classic presentation is
prevent mortality.
with high fever, headache, myalgia and petechial skin rash. Mul­

• Sensitive and specific diagnostic tests are not available in the


Key points early phase of illness.
• The few ancillary laboratory findings that may diagnose
• Rickettsial infections are zoonotic illnesses with a worldwide rickettsial illness during the early phase (<7 days) are: normal or
distribution. low total leucocyte count with band cells in peripheral blood
• The prototype illnesses are Rocky Mountain spotted fever smear, thrombocytopenia, hyponatraemia, hypoalbuminaemia
(R. rickettsii) and Mediterranean spotted fever (R. conorii) in the and raised hepatic enzymes.
‘spotted fever group’, and epidemic typhus (R. prowazeki) and • A high index of suspicion is required to diagnose rickettsial fever
endemic typhus (R. typhi) in the ‘typhus group’. at presentation, even in endemic areas.
• The organisms target vascular endothelial cells causing inflam­ • Doxycycline is the treatment of choice in rickettsial illnesses,

SECTION 7: BACTERIAL SKIN


mation, apoptosis and capillary leakage, collectively known as even in children.
‘rickettsial vasculitis’. • In case of doubt, empirical therapy with doxycycline can be
• Rickettsial infections present with nonspecific signs and started at presentation in all suspected cases of rickettsial fevers.
symptoms such as fever, headache, myalgia and petechial • In uncomplicated cases the patient should become afebrile

INFECTIONS
skin rash. within 48 hours of administration of doxycycline. If this is not
• Multisystem involvement with pneumonia, meningoencephalitis, the case, the physician should reconsider the initial diagnosis.
acute renal failure and ‘disseminated intravascular coagulation’‐ • Personal protection and environmental treatment are the
like features is often present. important preventive measures.

determined by the then ongoing epidemic of louse‐borne


Introduction. Rickettsial organisms are heterogenous,
typhus fever [1].
causing febrile illnesses with variable clinical features
The upsurge of rickettsial diseases in recent years is a
ranging from mild to life‐threatening. Epidemic typhus
threat to human health and a problem for the medical
caused by louse‐borne Rickettsia prowazeki is of great
fraternity. Diagnosis of these infections is challenging
historical importance. The first epidemic of typhus fever
because of lack of ‘tell‐tale’ clinical features. Moreover,
was recorded during the fifth century, in Athens, Greece
there is no laboratory test to confirm the disease during
[1]. Napoleon’s victory march to Russia was curtailed by
the first week of illness. Although low‐cost specific therapy
the then ongoing typhus epidemic in the country which
is available, rickettsial illnesses continue to remain a
affected his troops [1]. It became a scourge during the
menace to health in many endemic areas.
First World War when >30 million people were infected
and nearly 3 million lost their lives [1–3]. It was aptly
remarked by Vladimir Lenin (1919) during the Russian Epidemiology. Rickettsial illnesses occur worldwide and
Revolution that ‘Either socialism will defeat the louse or there are endemic foci of infection in which sporadic cases
the louse will defeat socialism’ [1]. Many historians and occasional outbreaks occur. In suitable environmen­
believe that the destiny of twentieth‐century warfare was tal conditions there is re‐emergence of the infection with a

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
504 Section 7  Bacterial Skin Infections

resultant epidemic. Business or recreational travel to causative agent for scrub typhus [5,7]. There has been
various places may transport the disease to new geo­ documented emergence of new species such as R. parkeri
graphical areas. Decade‐wise frequency estimation of (2002) and R. slovaca in the spotted fever group.
Rocky Mountain spotted fever by passive surveillance Various species of Rickettsia, the vectors, hosts and
demonstrates a gradual increase in annual incidence rate reservoirs, the diseases caused by them and their geo­
(1.7 cases per million person‐years in 2000 to 14.3 cases graphical distribution, are presented in Table 43.1. These
per million person‐years in 2012) [4]. organisms are probably able to adapt to new ecosystems,
completing their lifecycle through new arthropod hosts
Agent and maintaining their virulence [8]. R. prowazekii has the
The genus Rickettsia in the family Rickettsiaceae includes ability to persist in the human reticuloendothelial system
a special group of microorganisms with properties tran­ during convalescence as a subclinical infection and may
sitional between bacteria and viruses. These are aerobic, manifest later as recrudescent typhus (Brill–Zinsser
strictly obligate, intracellular organisms with coccobacil­ disease) [9].
lary morphology [5]. They have variable Gram staining
properties but are mostly Gram negative and take on a Host
purple colour on Giemsa staining [5]. Rickettsia are una­ Human rickettsioses are considered to be zoonoses. The
ble to grow in cell‐free media and have fastidious growth natural hosts of Rickettsia are lower mammals, some
requirements. Laboratory culture of these organisms is vertebrates and arthropods. Blood‐sucking ectoparasites
undertaken in the yolk sac of developing chick embryos, such as ticks, fleas, mites and lice constitute the arthropod
and in continuous cell lines such as mouse fibroblast, hosts as well as vectors [5]. They acquire the infection
HEp‐2, HeLa, Detroit 6, etc. [5]. Isolation from patient following a blood meal from an infected/reservoir verte­
samples is better performed in animal models like guinea brate host. The ectoparasites become infective within
pigs and mice [5]. 1  week (when the organism has multiplied in their
There are more than 20 validated species; in a current alimentary canal) and remain so until they succumb to
classification system, these have been categorized into the infection [5]. Wild rats and household mice are the
four groups [6]. Orientia is another genus in the same commonest mammal hosts [5]. Human beings are not
family, and the organism Orientia tsutsugamushi is the actually involved in the natural lifecycle of rickettsial

Table 43.1  Various rickettsial species, related vectors, hosts and reservoirs, diseases caused by them and geographical distribution [3,7]
SECTION 7: BACTERIAL SKIN

Species Disease Vector Host/vertebrate Distribution


reservoir
INFECTIONS

A.  Ancestral group (tick‐borne): R. bellii, R. canadensis


B.  Typhus group
R. prowazekii Epidemic typhus Human body louse; rarely, Human, flying Central, north and south America,
Recrudescent typhus (Brill–Zinsser head louse squirrels central Africa, Asia
disease)
R. typhi Murine typhus (endemic typhus) Rat flea, (Xenopsylla cheopis) Rat Worldwide tropical and subtropical
possibly some ticks regions
C.  Spotted fever group
R. rickettsii Rocky Mountain spotted fever Tick (Dermacentor variabilis) Dogs, rabbits, birds North, central and south America
R. conorii Mediterranean spotted fever Tick Rodents, dogs Southern Europe, South and West
(boutonneuse fever), Indian Asia, India, Africa
tick typhus
R. japonica Japanese spotted fever Tick ?Rodents Japan
R. slovaca Tick‐borne lymphadenopathy Tick Lagomorphs, rodents South and East Europe, Asia
(TIBOLA), Dermacentor‐borne
necrosis, erythema and
lymphadenopathy (DEBONEL)
Other organisms in the spotted fever group (tick‐borne): R. aeschlimannii, R. africae, R. heilongjiangensis, R. helvetica, R. honei, R. massiliae, R.
montanensis, R. parkeri, R. peacockii, R. rhipicephali, R. sibirica
D. Transitional group
R. akari Rickettsial pox Gamasid mite Domestic mouse, wild Soviet Union, South Africa, Korea,
rodents Turkey, Balkan countries, North
and South America
R. felis Cat‐flea rickettsiosis Cat flea Domestic cat, rodents, Europe, North and South America,
opossum Asia
R. australis Queensland tick typhus Tick Bush rodents Tasmania, Australia
Other organisms
Orientia Scrub typhus (chigger‐borne Trombiculid mites (transmitted Rodents, birds Asia Pacific region from maritime
tsutsugamushi typhus) by mite larvae/chiggers) Russia and China to Indonesia and
North Australia to Afghanistan
Chapter 43  Rickettsial Disease 505

organisms. Human disease is accidental, occurring due to epidemic typhus are mediated by human‐to‐human
an arthropod bite [3,5]. transfer of the ectoparasites [5].
There is a close inter‐relationship between the vector,
vertebrate hosts, human activities and the transmission of Pathogenesis. In mammals, rickettsial organisms have
disease. Travel and outdoor activities such as hunting, a tropism for the vascular endothelium of small and
fishing, camping, trekking and dog‐walking increase the medium‐sized blood vessels [3]. R. rickettsii rarely may
chances of acquiring infection [7]. Epidemic typhus is infect the smooth muscle cells, where they multiply in
prevalent in particular populations such as nomads and in the cytoplasm [12]. At the site of the first host–pathogen
refugee colonies, army barracks, etc., where risk of body interaction, i.e. in eschar, mononuclear phagocytes are
louse infestation is increased [7]. The rat flea (Xenopsylla the first group of defence cells to become infected [3].
cheopis), the vector for endemic typhus, remains in an A  series of events takes place thereafter, resulting in
infective stage for a long time and bites human beings clinical manifestations.
only when the natural hosts are not available [10].
Adhesion and entry into host cells
Environment Rickettsial organisms possess genetically encoded surface
Rickettsial organisms are present ubiquitously irrespec­ cell antigens (Sca) that encode autotransporter‐like pro­
tive of geographical variations. The vector for scrub teins and are involved in host cell adhesion and/or inva­
typhus, the trombiculid mite, resides in scrub jungle and sion [3]. Well‐identified antigens include Sca0 (OmpA),
tall‐grass areas [5,7]. These mites reside in a well‐defined Sca1, Sca2 and Sca5 (OmpB). Spotted fever group (SFG)
area of soil with a favourable microecosystem, designated organisms possess two well‐characterized Sca, OmpA
as ‘mite islands’ [5]. Humans contract infection through and OmpB; typhus fever group (TG) organisms possess
chigger‐bites when accidentally entering these areas. only OmpB [3]. These Scas play varied roles in the organ­
This is of importance for the movement of soldiers during ism–host interaction which may differ species‐wise. In
warfare [5,7]. R. conorii, Sca1 acts as an adhesin without playing a role
Disease is common in spring and summer as ticks and in invasion and Sca2 mediates interaction with the target
fleas remain most active during these seasons [7]. April host cell membrane [3,9]. In R. parkeri, Sca2 acts as a
to September are the common months for the occurrence ‘formin‐like mediator of actin‐based motility’ and inter­
of Rocky Mountain spotted fever (>90% cases) [9,10]. acts with host proteins to promote survival of the organ­
Endemic typhus occurs throughout the year in the USA ism [3,9]. Sca4 (all rickettsial species) co‐localizes with

SECTION 7: BACTERIAL SKIN


with a peak during April to June [9]. Having domestic host cell vinculin at the site of adhesion and binds to and
cats/dogs and forest around the residence confers a higher activates vinculin through two vinculin‐binding sites [3].
risk of infection [9]. Rickettsial pox, the reservoir host for
which is domestic mice, prevails in urban areas [5,7,9]. Role of host cells

INFECTIONS
Multiple interactions between host cell receptors and
Transmission outer membrane ligands of Rickettsia take place for inter­
The arthropod vectors acquire the infection by ingesting a nalization [3]. Host cell nuclear DNA‐dependent protein
blood meal from an infected small mammal host. In kinase Ku70 (also located in plasma membrane and cyto­
arthropod hosts rickettsiae can maintain their lifecycle plasm) acts as the receptor for OmpB and helps in entry of
through trans‐ovarial and trans‐stadial transmission [5,7]. R. conorii [3]. In rickettsia‐infected hosts, at the site of entry,
The larvae of trombiculid mites (chiggers) harbour the ubiquitination of Ku70 occurs by recruitment of ubiquitin
organisms rather than adults and infect humans by ligase c‐cbl. Integrins (α2β1) on the host cell surface act as
repeated asymptomatic biting. receptors for OmpA [3]. Actin polymerization in host cells
Tick‐borne diseases are transmitted to a human host is another mechanism for rickettsial internalization. There
through tick saliva. Flea and lice defaecate while feeding is evidence that endocytosis, mediated by clathrin and
on hosts (flea‐feeding reflex) [11]. Entry of organisms claveolin‐2, may play a part in this process [3].
through human skin is facilitated by rubbing of infective The rickettsial membranolytic proteins haemolysin C
faecal pellets onto microtraumatized areas of skin and and phospholipase D cause rupture of phagosomal mem­
mucosa while scratching [11]. Rare modes of transmission branes and thus entry to host cytosol [3,9]. A phospholi­
in typhus fevers are inhalation of dried faeces of louse or pase A2‐like activity may also facilitate entry of R. typhi
ticks (aerosols) or conjunctival inoculation [5,7]. Food that to host cells [3,13]. In vivo, these organisms preferentially
has recently been contaminated with rat urine may be a invade the endothelial cells of small and medium‐sized
source of infection [5,7]. Very rare modes of transmission blood vessels, causing vascular damage [3].
are blood transfusion and organ transplantation [8].
In addition to tick bites, Rocky Mountain spotted Intracellular movement and cell‐to‐cell dissemination
fever can be transmitted by deeper contact via a crushed This process varies for different species. Spotted fever
or incompletely removed tick [10]. Human‐to‐human group organisms develop a ‘polar actin tail’ which helps
transmission does not occur. Body lice are temperature their intracellular movements and intercellular passage,
sensitive. They leave the body of a febrile human host in thus promoting dissemination, a key factor in pathogenesis
search of a normothermic one. In communities or geo­ [3,9]. R. typhi develop a shorter polar actin tail, resulting
graphical areas with high lice infestation, outbreaks of in limited, erratic motility. RickA, a Wiskott–Aldrich
506 Section 7  Bacterial Skin Infections

syndrome protein (WASP), regulates actin‐based motility in a blood smear. Procoagulant properties of the activated
via activation of actin nucleation and Arp2/3 complex endothelial cells result in platelet adhesion, leucocyte
recruitment [3,9]. R. prowazekii undergoes intracellular chemotaxis and consumption of coagulation factors
replication by binary fission and the infected cells bulge resulting in a clinical picture similar to disseminated
with loaded organisms resulting in necrotic cell lysis [3]. intravascular coagulation [13]. Fig. 43.1 illustrates various
Intracellular Rickettsia secrete various membrane‐associated steps in the pathogenesis of rickettsial illnesses.
transporter systems and effector proteins to modulate
host cell functions. Clinical features. Symptomatology‐wise, rickettsial
diseases are categorized into SFG and TG. The main ill­
Host immune response nesses in SFG are Rocky Mountain spotted fever (RMSF),
The selective vasculotropism of rickettsial organisms Mediterranean spotted fever (MSF) and rickettsial pox. In
predisposes mammal hosts to events such as inflamma­ TG, epidemic and endemic typhus are the main illnesses
tion of the vessel wall, loss of vascular integrity and of clinical importance. Recrudescent typhus (Brill–Zinsser
increased vascular permeability. This conglomerate effect disease) is an illness of epidemiological importance in this
of rickettsial infection is known as rickettsial vasculitis group. Scrub typhus is a closely related disorder caused
[3,9]. Simultaneously, the host immune system mounts an by genus Orientia.
anti‐inflammatory response to curb the damage caused Elicitation of a detailed history about outdoor activi­
by rickettsial vasculitis. ties, travel to endemic areas in the recent past, pets in the
In response to rickettsial infection endothelial cells family, tick/mite exposure within 15 days of illness and
undergo activation from a relatively quiescent state to other affected family members is of great importance [12].
an ‘activated phenotype’ with pro‐inflammatory and The presence of any of these helps to make an early tenta­
procoagulant properties (normally constituting an anti‐ tive diagnosis, but absence does not exclude it. Mite and
inflammatory and anticoagulant barrier) [9]. In vitro tick bites are typically painless, hence the site (eschar)
studies demonstrate an array of local changes: altered may remain obscure. Bites by nymphal stages of ticks
surface adhesiveness of platelets, increased expression of result in multiple, small erythematous papules simulating
tissue factors such as interleukin (IL)‐1α, intercellular and mosquito bite rather than an eschar [12]. The clinician
vascular cell adhesion molecules, E‐selectin, increased must search for an eschar or attached tick or mite after
synthesis of plasminogen activator inhibitor‐1, and adequate stripping [13]. Clothing should also be checked
release of von Willebrand factor from Weibel–Palade bod­ for ticks or body lice [13].
SECTION 7: BACTERIAL SKIN

ies of endothelial cells [9]. Infection with R. prowazekii The incubation period of all rickettsial diseases ranges
induces secretion of prostaglandins E2 and I2 [9]. Vascular between 5 and 15 days after a bite by the vector [6]. Any
endothelial insults eventually lead to expression of genes age group may be affected. High‐grade fever is the usual
dependent on the nuclear factor (NF)‐κB family of tran­
INFECTIONS

presenting feature in all forms of rickettsiosis and some


scription factors. This results in activation of NF‐κB and variants present with skin rash. Multisystem involvement
there is upregulation of NF‐κB binding sites in promoter and a fatal course may ensue if the disease is not treated
regions of the organisms. NF‐κB has antiapoptotic activity; at an early stage.
this maintains mitochondrial integrity of host cells and
prevents infection‐induced apoptosis of endothelial cells Cutaneous features
by preventing the activation of caspase‐8 and ‐9 [9]. There High fever and chills are the presenting features in spot­
is secretion of interferons (IFN), IFN‐α and ‐β, which have ted fevers. In some spotted fevers, e.g. African tick bite
shown inhibitory action on the growth of R. prowazekii fever or MSF, one or more eschar(s) may be demonstrable
in in vitro studies [9]. The killing of intracellular organ­ [14]. A faint erythematous skin rash evolves 3–5 days
isms in endothelial cells, macrophages and hepatocytes after the onset of fever. Rose‐red, discrete, blanchable
depends upon the concentration of IFN‐γ, IL‐1β, RANTES macules start at the wrists and ankles and spread to proxi­
and tumour necrosis factor (TNF)‐α [9]. mal extremities, trunk and face (centripetal spread)
Rickettsia‐infected vascular endothelium produces (Fig. 43.2a and b). Palms and soles are classically involved
reactive oxygen species (ROS), superoxide radical (O2−) [10,12] (Fig.  43.3a and b). In dark‐skinned patients, the
and hydrogen peroxide (H2O2), which put the host cells rash may be difficult to appreciate initially. In RMSF the
under oxidative stress [9]. There is widespread dilatation rash may sometimes be evanescent or localized, or totally
of intracellular membranes, mostly of rough endoplasmic absent (20%) [12,13,15,16]. The rash gradually turns pete­
reticulum, loss of osmoregulatory control and cell lysis chial (Fig. 43.4) or may become maculopapular. Confluent
occurring by 5–6 days of infection [9]. There is increased purpuric lesions may be seen as the disease advances and
vascular permeability due to PGE2 and PGI2 resulting indicate severe illness [12]. Ecchymotic and gangrenous
in the inflammatory oedema found during rickettsial areas simulating purpura fulminans may appear, espe­
infections [9]. cially in body areas with end‐vasculature: tip of nose,
Widespread vascular injury and capillary leak caused helix of ear, scrotum and digits [13] (Fig.  43.5). Rarely,
by rickettsial organisms affects nearly all organ systems, digital gangrene simulating ‘peripheral symmetrical
lungs, kidney, liver, spleen, meninges, brain and testes, gangrene’ may occur [13] (Fig. 43.6a and b). Periorbital
resulting in complications. There are limited organisms in oedema (Fig.  43.7) and swelling of hands and feet
the peripheral circulation and these are not demonstrable (Fig. 43.8) usually accompany the skin lesions [10].
Chapter 43  Rickettsial Disease 507

R1= SFG organism


R2= TG organism
R1 R2
= OmpA

= OmpB

R1 =SFG organism with


polar actin tail

INTEGRIN =Group of TG
Ku70 organisms multiplied
Ku70 R1 by binary fission
R1
Internalization R2
Effector proteins to modulate
R1,R2 host
host cell function
cellfunction Replication
by
endocytosis Spread of TG
Cell organisms
R1 Activated endothelial cell with bulging
Spread of SFG procoagulant & proinflammatory & lysis
R1 property
organism
cell
R1 R1 Maintains mitochondrial integrity
NF-κB
Prevents activation of caspase
upregulation pathway
Cell
R1
HOST IMMUNE
SYSTEM RESPONSE
IL-1α, IFN-γ, Prevents
Adhesion molecules IL-1β, infection-induced
Reactive E-selectin RANTES, apoptosis
oxygen Plasminogen activator inhibitor-1 TNF-α
v-WF
Prostaglandins E2 & l2
Killing of
intracellular
organism
CELL LYSIS Inflammation of
endothelial cells

SECTION 7: BACTERIAL SKIN


RICKETTSIAL EFFECT ON HOST CELLS

INFECTIONS
Fig. 43.1  Various steps in the pathogenesis of rickettsial infection.

(a) (b)
Fig. 43.2  (a and b) Initial faint erythematous rash of rickettsial spotted fever.
(a)

Fig. 43.5  Ecchymotic spots on helix of the ear.


SECTION 7: BACTERIAL SKIN
INFECTIONS

(a)

(b)
Fig. 43.3  (a and b) Rash on palms and soles.

(b)
Fig. 43.4  Petechial skin rash. Source: Image courtesy of Dr Atul Kulkarni, Fig. 43.6  (a) Gangrene of fingertips and ecchymotic areas on palm. (b)
Paediatrician. Ecchymotic areas on sole with peripheral symmetrical gangrene of feet.
Chapter 43  Rickettsial Disease 509

Fig. 43.9  Maculopapular rash on the trunk in a case of typhus fever.

Fig. 43.7  Periorbital swelling and faint skin rash on face. Eschars are painless ulcers with central, dark, adherent
crusts with surrounding erythema or scaling and regional
lymphadenopathy. Usually eschar is solitary but multiple
lesions may be present [10].

Systemic features
There is abrupt‐onset high fever, occasionally with morn­
ing remission [13]. Shaking chills, headache, myalgia,
nonproductive cough, dyspnoea, nausea and vomiting,

SECTION 7: BACTERIAL SKIN


calf pain and tenderness may accompany the fever at
presentation. Headache is typically frontal [13]. It is a
consistent feature and may be severe. Myalgia commonly
involves the muscles of the lumbar area, thigh and calf,

INFECTIONS
resulting in lower‐back and lower‐limb pain [13]. Rare
and unusual complications of MSF fever are haemophago­
cytic lymphohistiocytosis and acute pancreatitis [18,19].
In scrub typhus, headache and conjunctival injection
are the common initial presentation. Hepatosplenomegaly
and generalized lymphadenopathy are seen in most of
Fig. 43.8  Ecchymotic skin lesions on sole with swelling of the foot. the patients [13]. Multiorgan involvement may manifest
by the end of 1 week. Pneumonia is the commonest
Patients with typhus fever also present with a febrile systemic manifestation. Others include hepatitis, renal
illness. In both epidemic and endemic typhus, the skin failure, myocarditis and meningoencephalitis. Severe
rash is morphologically similar but milder in the latter. In neurological manifestations in this illness (photophobia,
contrast to SFG, the eruption starts on the trunk and blurring of vision, drowsiness, disorientation, delirium and
thereafter spreads in a centrifugal distribution (Fig. 43.9). stupor) are the origin of the name ‘typhus’ (cloudiness)
Face, palms and soles are spared in this disease [10]. [13]. Septic shock or acute respiratory distress syndrome
Rickettsial pox is characterized by a varioliform skin (ARDS) may complicate the disease course [10].
eruption. The fever is preceded by eschar formation at Immediate and long‐term residual complications (beyond
the site of mite adherence. A generalized papulovesicular 1 year) in patients suffering from rickettsial illnesses are
eruption sparing the palms and soles is characteristic of presented in Table 43.2 [12,13,20].
this disease [5,10]. The lesions heal with black crusting
and resolve without scarring. This is the mildest of the Childhood disease
rickettsial illnesses. Children may be affected by any type of rickettsial illness.
The incubation period of scrub typhus is 1–3 weeks An earlier notion that RMSF is more common in children
[10]. These patients lack a generalized skin rash. However, was not substantiated in later studies [12]. However, in a
an eschar (tache noir) at the site of a bite by chiggers recent report on an explosive outbreak of RMSF in tribal
may be noticed at presentation and is the pointer to lands of Arizona, nearly 50% of the cases were ≤10 years of
the  diagnosis [10,17]. Common body sites to search for age and this was attributed to ‘dog handling’ by children,
eschars are neck, axillae, chest, abdomen and groin. tick‐infested dogs being the reservoir of infection in that
510 Section 7  Bacterial Skin Infections

Table 43.2  Long‐term complications of the main rickettsial diseases [12,13,20]

Disease Early complications Late complications

Rocky Mountain • Interstitial pneumonia • Partial paraplegia


spotted fever (RMSF) • Pulmonary oedema • Bell’s palsy
• Acute respiratory distress syndrome • Digital/limb gangrene requiring amputation
• Abdominal pain • Deafness and blindness
• Meningoencephalitis • Loss of bladder and bowel control
• Disseminated intravascular coagulopathy‐like • Movement disorders
symptoms and peripheral gangrene • Difficulty in speech
• Hepatic failure These have been reported in patients with RMSF who had prolonged hospital
• Myocarditis stay and some of these complications developed beyond 1 year of illness
Mediterranean spotted • Haemophagocytic lymphohistiocytosis
fever (MSF) • Acute pancreatitis
Scrub typhus • Constipation
• Meningoencephalitis
• Photophobia and blurring of vision
• Acute renal failure

area [21]. The overall case fatality rate of RMSF in children at any time during the course of the illness. The
aged 5–9 years has been found to be high in the USA [22]. abdominal pain may be severe enough for the clinician
Clinical features of rickettsial infections in children to consider causes of acute abdomen, most likely acute
similar to those in adults. There are few case series appendicitis [12].
reporting rickettsial fever in paediatric patients [23–25].
The youngest patient among the reported cases was 16 Diagnosis. Diagnosis of rickettsial diseases during the
months old. Continuous, high‐grade fever extending initial few days is easily missed because of nonspecific
beyond a week and skin rash strikingly involving palms clinical features simulating viral exanthema. Even in
and soles are the consistent features in this age group. endemic areas, approximately 60–75% patients with
Onset of rash may be earlier in children as compared RMSF are diagnosed as having some other illness at first
SECTION 7: BACTERIAL SKIN

to  adults [12]. Headache is uncommon in younger visit to a doctor [12]. A history of tick bite may be avail­
­children but, if present, is often not manageable with able in less than half of the cases [13]. As the bite by an
analgesics [13]. Calf pain and tenderness are common in ectoparasite is usually painless, the sufferer may not be
children [13]. Puffiness of the face, periorbital oedema aware of an eschar. Absence of fever or rash in a patient at
INFECTIONS

and oedematous extremities are common features; gen­ presentation should not deter the clinician from making a
eralized oedema with ascites and hydrothorax may be diagnosis of rickettsial fever, as the patient may be in the
present. Various systemic features reported in paediat­ evolving phase of the disease; moreover, fever and even
ric patients are conjunctival injection (Fig. 43.10), altered rash may be absent in these patients [15,21]. A compli­
sensorium, seizures, hepatosplenomegaly, abdominal cated case of rickettsial fever may present with shock‐
pain, arthralgia and pneumonia, which may manifest related hypothermia [21].
‘Case definitions’ for diagnosis of rickettsial diseases
formulated by the Indian Council of Medical Research
(ICMR) in 2015 are listed in Box 43.1 [17].

Box 43.1  Case definitions for diagnosis of rickettsial diseases


formulated by the Indian Council of Medical Research (ICMR) in 2015
[17]

• Suspect/clinical case: Acute‐onset fever of unknown origin for a duration


of ≥5 days, ± the presence of eschar. In the presence of eschar, scrub
typhus can be suspected even if the fever duration is <5 days. Skin rash,
headache, myalgia and other systemic features may be associated.
• Probable case: A suspect/clinical case with positive Weil–Felix test
(≥1:80 titre for all three antigens) and positive enzyme‐linked
immunofluorescence assay (ELISA) for IgM antibody (>0.5 optical
density).
• Confirmed case: Detection of rickettsial DNA from sample of eschar
or polymerase chain reaction (PCR)of whole blood; or demonstration
of rising antibody titre in acute and convalescent sera by indirect
immunofluorescence assay/indirect immunoperoxidase assay.
Fig. 43.10  Conjunctival injection in an adolescent with rickettsial fever.
Chapter 43  Rickettsial Disease 511

Laboratory tests high concentration of agglutinins is also demonstrable in


Leucocytosis, thrombocytopenia and elevated serum nonrickettsial infections [17]. However, good correlation
transaminase levels are detected during the early phase of between a positive Weil–Felix test and raised IgM antibody
the disease. In RMSF, the total leucocyte count may be by IFA has been seen [13]. It is a useful option for making
normal or borderline low with immature band cells in the probable diagnoses of rickettsial diseases (spotted fever or
peripheral blood smear [12,13]. Thrombocytopenia is pre­ typhus) in resource‐poor countries [17]. The test becomes
sent in 60% patients [13]. The erythrocyte sedimentation reactive after 5–7 days of onset of fever. A 1 : 80 titre is indic­
rate is usually high. ative of possible rickettsial infection; however, this should
Hyponatraemia and hypoalbuminaemia resulting from be standardized in each endemic region [17]. Paired sera
capillary leak syndrome may be features in the acute can be tested for a fourfold rise in titre, or a single high titre
stage [12,13]. Hepatic transaminases are often raised. In (>1 : 320) can be considered diagnostic [13].
the presence of respiratory symptoms, a chest radiograph
should be obtained and may show a bilateral interstitial Skin biopsy
infiltrate or lobar consolidation. In the presence of menin­ Histopathology. A skin biopsy should be obtained from
gitis, cerebrospinal fluid analysis shows polymorphonu­ the purpuric skin rash. In patients with RMSF a haema­
clear or lymphocytic pleocytosis (<100 cells/μL) [12]. toxylin and eosin‐stained specimen shows a dense mono­
nuclear cell infiltrate around the dermal vasculature [27].
Immunofluorescence assay (IFA).  This is considered a
‘gold‐standard’ test for the diagnosis of rickettsial infec­ Immunohistochemical staining. This is performed on skin/
tion [12,17]. The sensitivity of the test depends on the involved organ biopsy specimens. Immunohistochemical
duration of disease; usually it becomes positive beyond staining from skin biopsy specimens in patients with
7–10 days. Sensitivity of IFA assay rises to 94–100% RMSF has 70% sensitivity and 100% specificity. The presence
beyond 14 days of illness, higher than any other sero­ of rickettsial antigen in skin may be focal, limiting consist­
logical test for rickettsial infection [12]. It is ideal to test ent detection.
the paired sera (acute and convalescent) at an interval of Rathi et al. have developed the Rathi, Goodman, Aghai
2–3 weeks, to demonstrate a greater than fourfold rise in (RGA) scoring system, which utilizes clinical, laboratory
titre of IgM and IgG. Availability of this test is limited to and epidemiological parameters to diagnose SFG
certain centres and it is not useful during the first week of illnesses [26]. The RGA scoring system has a total of
­
illness. In RMSF, the IgM antibody wanes by 3–4 months 35 points. It has been reported to have a sensitivity of 96.1%

SECTION 7: BACTERIAL SKIN


and IgG by 7–8 months [12]. In endemic areas the value of and specificity of 98.8% for diagnosing spotted fever at
detection of IgM and IgG antibodies against rickettsial score 14 [28].
organisms may not be very specific; anti‐R. conorii IgM
has been found to cross‐react with bacterial antigens such Differential diagnosis. The commonest differential

INFECTIONS
as Legionella spp. and Coxiella burnetii, giving rise to false diagnoses of rickettsial fever in children include various
positivity [26]. classical viral exanthems of childhood (measles, rubella,
erythema infectiosum, roseola infantum) and drug rash.
Demonstration of antibodies by ELISA.  ELISA is used The chronological order in which the fever and skin rash
more frequently in the diagnosis of rickettsial infections. appear, progress and subside in these illnesses is usually
IgM capture assay of serum confirms recent infection but distinctive. Constitutional symptoms are milder and the
the significant baseline titre needs to be determined in risk of systemic involvement is usually low. Moreover,
each endemic region [12]. the character of the rash is mostly maculopapular rather
than purpuric in these conditions. Other viruses causing
Indirect immunoperoxidase assay.  This is considered transient exanthematous illness in children are coxsacki­
equivalent to IFA assay and is another gold‐standard test evirus, echovirus and Epstein–Barr virus. Various differ­
in the diagnosis of rickettsial fever. However, its availability ential diagnoses of rickettsial illnesses and the points of
is limited and it is mostly used for research purposes [17]. differentiation are presented in Table 43.3.

PCR. Detection of rickettsial antigen by PCR can be used Risk factors and prognosis. Rickettsioses are usually benign
for early and rapid diagnosis. Blood is not a very suitable infectious illnesses but 10% of patients may develop severe
sample, and a skin or organ biopsy specimen should be complications [23]. The clinical severity depends upon
utilized for this purpose. However, this facility is again the virulence of the organism as well as host factors
available only in specific laboratories. Treatment with such as older age, black ethnicity, male gender, chronic
doxycycline decreases the sensitivity of PCR [12]. alcoholism and presence of co‐morbid illnesses [12,29].
National surveillance data (1999–2007) from the USA
Weil–Felix test.  This is a heterophile antibody test. The regarding case fatality rate in RMSF have revealed that
basis of this test is sharing of antigens between Rickettsia native Americans had more fatal outcome of the disease
and Proteus species (P. vulgaris: OX19, OX2; P. mirabilis: than other races [22,30]. Tick‐borne illnesses are more com­
OXK) and demonstration of agglutinins to these organisms mon in males, probably because of higher occupational and
in a ‘suspect case’ of rickettsial fever. This test lacks the recreational exposures in tick‐infested areas [4,29]. Children
sensitivity and specificity of a good diagnostic test as a are also at higher risk when playing outdoors.
512 Section 7  Bacterial Skin Infections

Table 43.3  Differential diagnoses of rickettsial fevers in children [10,13]

Disease Clinical features/laboratory tests Clinical features/laboratory tests not in favour of rickettsial illness
similar to rickettsial illness

Infections
Scarlet fever Fever, skin rash, vomiting, Rash characteristics: generalized, fine, punctate and erythematous; described as
lymphadenopathy, often ‘sunburn with goose pimples’; transverse streaks at skinfolds, ‘Pastia’s lines’; subsides
myocarditis and meningitis with desquamation; flushing of face with perioral pallor; white strawberry tongue
Staphylococcal toxic Fever, skin rash, vomiting, myalgia, Initially, faint skin rash present on first day, with diffuse erythema, disappears by 3 days.
shock syndrome marked oedema of hands and Further maculopapular and purpuric rash appears in second week. Characteristic
feet, thrombocytopenia desquamation of palms and soles by 10–21 days. Mucosal and conjunctival erythema
Dengue fever High fever with maculopapular skin Rash characteristics – ‘islands of normal skin in sea of red’; joint pain; haemorrhagic
rash, myalgia, thrombocytopenia symptoms and shock
Meningococcaemia Febrile child with petechial skin rash Rapid progression of illness
Meningitis CSF analysis: very low glucose level, neutrophilic pleocytosis; Gram stain: Gram‐negative
diplococci; blood culture: growth of N. meningitidis
Secondary syphilis Lesions on palms and soles Afebrile patient
Lesions on palms and soles are either hyperpigmented macules or hyperkeratotic, scaly
papules
Positive serological tests for syphilis
Colorado tick fever High biphasic fever, severe Absence of skin rash
neurological involvement,
leucopenia, thrombocytopenia
Enteric fever High fever and rose spots Rash appearing in second week of fever
Noninfectious illness
Kawasaki disease High fever, maculopapular rash, Cheilitis, nonexudative conjunctivitis, strawberry tongue, periungual and perineal
lymphadenopathy desquamation
Erythema multiforme Fever, involvement of palms and soles Target lesions
Lesions on palms and soles are pruritic and tender
Stevens–Johnson Fever, involvement of palms and Temporal correlation with drug intake
syndrome soles Typical/atypical target lesions
Lip and mucosal involvement
SECTION 7: BACTERIAL SKIN

Drug‐induced Fever, maculopapular rash Temporal correlation with drug intake


maculopapular rash Rash is devoid of haemorrhagic component
Henoch–Schönlein Purpuric skin lesions starting at ankles Palpable purpura, usually involving lower limbs; arthralgia/arthritis
purpura Fever may be present, renal Immunofluorescence of skin biopsy specimen demonstrates IgA deposit at dermal vessel
INFECTIONS

involvement, acute abdomen walls


Histopathology of skin lesions:
inflammation around dermal vessels
Thrombotic Petechial skin lesion,
thrombocytopenic thrombocytopenia
purpura

Epidemic typhus is considered one of the most severe organism and host risk factors in certain populations such
infectious diseases of human beings, the mortality rate as alcoholism and patients with concomitant illnesses
ranging from 10% to 60% [9]. Mortality directly correlates such as acute renal failure and hyperbilirubinaemia [32].
with patient age, being lower in young adults (10%) and Scrub typhus usually runs a self‐limiting course [29].
higher in patients >50 years (60–70%) [29]. Complications However, the risk of mortality ranges from 1% to 35%,
are rare in endemic typhus, which has a mortality rate depending upon the virulence of the organism, adverse
of 1% [29]. host factors and time of initiation of therapy [29].
RMSF is the most severe SFG in developed countries
and untreated cases may have 20–30% mortality by the Treatment. Approximately 50% of patients with rickettsial
second week [9]. Delay in diagnosis and initiation of infections with major systemic symptoms require hospitali­
treatment with the appropriate antibiotic is associated zation and a multidisciplinary approach.
with 4% mortality [31]. Glucose‐6‐phosphate dehydroge­ Currently, doxycycline is the treatment of choice in
nase (G6PD) enzyme deficiency is directly proportional patients of any age suffering from rickettsial fever. It is
to the severity of RMSF and fatality ensues within 5 days a rickettsiostatic drug [12]. Initially, there was reluc­
of the onset of fever [29]. tance among paediatricians to use this drug empirically
In general, MSF is considered to be a milder disease in in children suffering from rickettsioses [24]. However, in
humans that runs a benign course. However, at present children a single 5–7 day exposure to doxycycline (as in
R. conorii has a wide geographical distribution and it the treatment of rickettsial infections) is not associated
appears that the clinical course is increasing in severity. with discolouration of teeth [33]. The American Academy
This may be related to changes in the virulence of the of Pediatrics Committee for Infectious Diseases has
Chapter 43  Rickettsial Disease 513

recommended doxycycline as a life‐saving drug in the Protective clothing. Long clothing with socks and closed
treatment of ‘suspect’ or ‘confirmed’ rickettsial infection shoes prevents tick/mite adherence. Light‐coloured
in children [34]. It is also recommended in pregnant clothing is preferable in order to see a crawling ectoparasite.
women in whom there is high suspicion of rickettsial
disease [12]. It must be started early as antibiotic therapy Tick removal.  Periodic inspection of the entire body for
is most effective during the initial phase of the disease hidden, attached ticks should be performed daily in endemic
when the organism is multiplying. In a ‘suspect’ case, areas. Frequent checking and removal is beneficial as the tick
doxycycline should be started empirically [21]. If there may remain quiescent for several hours (from 2 to 20 hours)
are complicating features, doxycycline should be started before injecting the organism into a human body. An
prior to referral to a tertiary centre [17]. These measures attached tick must be removed with mouth parts intact by
prevent disease‐related morbidity and death [17]. Consi­ gentle traction using fine forceps. If the mouth parts break
dering all these factors, early administration of doxycycline off in the skin they should be removed. Use of the fingers for
is a judicious step towards the goal of patient survival. this purpose and crushing of the tick is not recommended to
Oral doxycycline at 4.5 mg/kg bodyweight in two prevent undue contamination and risk of conjunctival inoc­
divided doses is administered in children <45 kg [17]. ulation. The bite wound should be disinfected.
In an uncomplicated case without organ dysfunction, the
child becomes afebrile within 24–48 hours of starting the Use of insect repellents. N,N‐diethyl‐m‐toluamide (DEET)
drug and this may be taken as indirect evidence of rickett­ can be used on exposed skin and clothing. For children,
sial infection [13]. Failure to respond in this way may a 20–30% concentration is recommended by the American
prompt the clinician to reconsider the diagnosis [12]. Academy of Pediatrics [36]. Both DEET and benzyl ben­
Children >45 kg should be given the adult dosage of zoate have miticidal action [37]. Malathion is effective
100 mg orally twice daily. The optimum duration of oral against the rat flea [37]. A spray‐on preparation of perme­
treatment for outpatient/inpatient cases has not been thrin can be applied evenly to outer clothing and dried
established. Two recommendations for uncomplicated before wearing to kill ectoparasites.
cases are until 3 days after remission of fever and a
minimum total period of 5–7 days [12]. Cases with com­ Protection of pets
plications may require longer duration of treatment. Pet dogs must be treated with acaricides at periodic inter­
Intravenous doxycycline may be administered in hospi­ vals. Their bodies should also be checked for the presence
talized children with critical illness. The calculated dose of ticks at periodic intervals to minimize the risk of human

SECTION 7: BACTERIAL SKIN


is added to 100 mL of normal saline and slowly infused exposure.
over a period of 30 minutes. This is followed by oral
dosage for a total duration of 7–15 days. Depletion of natural hosts
Other drugs that can be used are fluoroquinolones, Vegetation around human habitats must be cleared peri­

INFECTIONS
chloramphenicol and azithromycin [17,23]. Fluoro­ odically to reduce the load of ectoparasites in the environ­
quinolone has been used at a dosage of 20–30 mg/kg ment [10]. Rodenticides and mouse traps should be
per day [23]. The dose of chloramphenicol is 50–100 mg/kg regularly used on farms and in households [10].
per day (maximum 3 g/day) administered 6 hourly. Use
of chloramphenicol is nowadays limited because of its Role of prophylactic antibiotic
life‐threatening adverse effects, enhancing the risk of In patients who have had an accidental tick bite but who
mortality in these patients. The dosage of azithromycin have no symptoms of illness, prophylactic antibiotics are
in children is 10 mg/kg bodyweight as a single dose for not indicated [12]. This is because, in an endemic area,
5 days [17]. This is the drug of choice in the presence of only 1–3% of the vector ticks are infected and <1% have
doxycycline resistance [35]. confirmed disease [12,38]. Hence, the chance of acquiring
Sulfonamides should not be administered in ‘suspect’ infection merely by a tick bite is low. Moreover, it has
cases of rickettsial fever as these may directly enhance the been shown that prophylactic antibiotics may delay the
growth of the organism. This, in combination with delay onset of symptoms of RMSF in a patient exposed to infec­
in institution of specific therapy, increases morbidity and tion, but do not prevent it [12,39].
mortality [13,17].
Role of vaccine
Prevention
Rickettsial vaccines are not yet available.
Personal protection
When visiting areas endemic for rickettsial diseases or
Notification of cases
tick‐infested geographical areas, adoption of personal
A physician diagnosing and treating a case of rickettsial
protective measures is of immense importance.
illness should notify it to the local health authorities. This
is especially relevant in naive areas for rickettsial infec­
Avoidance of tick habitats. Walking on cleared trails and
tions to prevent an outbreak of the disease.
avoiding brushing against uncut grass and weeds is
advised, especially during peak seasons for tick activity.
Children may be prevented from playing in backyards Future directions. Some inherent properties of rickettsial
with vegetation – clear ground is preferred. organisms make them suitable for illegitimate use as
514 Section 7  Bacterial Skin Infections

bioterrorism weapons [2,3]. Some nations have field‐ 16 Kirkland KB, Wilkinson WE, Sexton DJ. Therapeutic delay and
mortality in cases of Rocky Mountain spotted fever. Clin Infect Dis
tested weaponized rickettsial organisms [2]. The compli­
1995;20:1118–21.
cated arthropod‐dependent lifecycle of the organisms is 17 Rahi M, Gupte MD, Bhargava A et al. DHR‐ICMR Guidelines for
an obstacle to such attempts [2]. However, the ease of diagnosis and management of rickettsial diseases in India. Ind J
spread of diseases like epidemic typhus and spotted Med Res 2015;141:417–22.
18 Cascio A, Giordano S, Dones P et al. Haemophagocytic syndrome and
fevers, the associated lethality and the potential to destroy Rickettsial diseases. J Med Microbiol 2011;60:537–42.
humankind en masse should put endemic countries on 19 Rombola F. Mediterranean spotted fever presenting as an acute
constant alert for such eventualities. pancreatitis. Acta Gastroenterol Belg 2011;74:91–2.
Vaccines against rickettsial organisms are not currently 20 Nilsson K, Wallménius K, Hartwig S et al. Bell’s palsy and sudden
deafness associated with Rickettsia spp. Infection in Sweden. A retro­
available and remain an area to be explored. The possibil­ spective and prospective serological survey including PCR findings.
ity of antibiotic‐resistant rickettsial strains developing Eur J Neurol 2014;21:206–14.
should be checked periodically. Current advances in the 21 Traeger MS, Regan JJ, Humpherys D et al. Rocky Mountain spotted
fever characterization and comparison to similar illnesses in a highly
understanding of the pathogenesis of rickettsial disease endemic area—Arizona, 2002‐2011. Clin Infect Dis 2015;60:1650–8.
have paved the way for development of novel therapies 22 Dahlgren FS, Holman RC, Paddock CD et al. Fatal Rocky Mountain
[3,9]. This knowledge can be exploited to develop patho­ spotted fever in the United States, 1999‐2007. Am J Trop Med Hyg
genesis‐based supplemental therapy to reduce the lethality 2012;86:713–19.
23 Murali N, Pillai S, Cherian T et  al. Rickettsial infections in south
associated with the illness [9]. The reactive oxygen radicals India  –  How to spot the spotted fever? Indian Pediatr 2001;
generated in the vascular endothelium can be combated 38:1393–6.
by use of antioxidants to relieve oxidative stress in host 24 Vitaliti G, Falsaperla R, Lubrano R et al. Incidence of Mediterranean
spotted fever in Sicilian children: a clinical‐epidemiological obser­
cells. Prevention of apoptosis of vascular endothelium by
vational retrospective study from 1987‐2010. Int J Infect Dis 2015;
interrupting the activation of the caspase pathway may 31:35–40.
be another intervention [9]. 25 Purvis JJ, Edwards MS. Doxycycline use for rickettsial disease in
pediatric patients. Pediatr Infect Dis J 2000;19:871–4.
26 McQuiston JH, Wiedeman C, Singleton J et al. Inadequacy of IgM
­References antibody tests for diagnosis of Rocky Mountain spotted fever. Am J
1 Holmes F. Typhus on the Eastern Front. Available at: http://www. Trop Med Hyg 2014;91:767–70.
kumc.edu/wwi/index‐of‐essays /medicine/typhus‐on‐the‐east­ 27 Sexton DJ. Rocky Mountain spotted fever and other Rickettsial
ern‐front.html (accessed 13 December 2018). infections. In: Irvine AD, Hoeger PH, Yan AC (eds) Harper’s
2 Azad AF. Pathogenic Rickettsiae as bioterrorism agents. Clin Infect Textbook of Pediatric Dermatology, 3rd edn. Oxford: Wiley‐Blackwell,
Dis 2007;45(suppl 1):S52–5. 2011:61.1–10.
3 Sahni SK, Narra HP, Sahni A, Walker DH. Recent molecular insights 28 Rathi NB, Rathi AN, Goodman MH, Aghai ZH. Rickettsial diseases
SECTION 7: BACTERIAL SKIN

in to rickettsial pathogenesis and immunity. Future Microbiol 2013; in central India: proposed clinical scoring system for early detection
8:1265–88. of spotted fever. Indian Pediatr 2011;48:867–70.
4 Drexler NA, Dahlgren FS, Heitman KN et al. National surveillance of 29 Rathore MH, Steele RW. Rickettsial infection. Available at: http://
spotted fever group rickettsioses in the United States, 2008‐2012. Am reference.medscape.com/article/968385‐overview (accessed 13
J Trop Med Hyg 2016;94:52–60. December 2018).
INFECTIONS

5 Kapil A. Rickettsiaceae. In: Kapil A (ed.) Ananthanarayan & Paniker’s 30 Folkema AM, Holman RC, McQuiston JH, Cheek JE. Trends in clinical
Textbook of Microbiology, 9th edn. Hyderabad: University Press diagnosis of Rocky Mountain spotted fever among American Indians,
(India), 2014:405–14. 2001‐2008. Am J Trop Med Hyg 2012;86:152–8.
6 Gillespie JJ, Beier MS, Rahman MS et al. Plasmids and rickettsial evo­ 31 Walker DH, Dumler JS. Emerging and reemerging rickettsial diseases.
lution: insight from Rickettsia felis. PLoS One 2007;2:e266. N Engl J Med 1994;331:1651–2.
7 McQuiston J. Rickettsial (spotted & typhus fevers) & related infec­ 32 Sousa R, França A, Dória Nòbrega S et al. Host‐ and microbe‐related
tions (anaplasmosis & ehrlichiosis). In: CDC Health Information risk factors for and pathophysiology of fatal Rickettsia conorii infection
for International Travel 2014: The Yellow Book. New York: Oxford in Portuguese patients. J Infect Dis 2008;198:576–85.
University Press. 33 Lochary ME, Lockhart PB, Williams WT Jr. Doxycycline and staining
8 Dumler JS, Walker DH. Rocky Mountain spotted fever–changing
of permanent teeth. Pediatr Infec Dis J 1998;17:429‐‐31.
ecology and persisting virulence. N Engl J Med 2005;353:551–3. 34 American Academy of Pediatrics. Rocky Mountain spotted fever.
9 Sahni SK, Rydkina E. Host‐cell interactions with pathogenic
In: Pickering LK, Baker CJ, Overturf GD, Prober CG (eds) 2003 Red
Rickettsia species. Future Microbiol 2009;4:323–9. Book: report of the Committee on Infectious Diseases, 26th edn. Elk
10 Luoto L. Rickettsial diseases of animals transmissible to children. Grove Village, IL: American Academy of Pediatrics, 2003:532‐‐4.
Pediatrics 1958;22:384–7. 35 Kim D‐M, Yu KD, Lee JH et al. Controlled trial of a 5‐day course of
11 Azad AF, Beard CB. Rickettsial pathogens and their arthropod vectors. telithromycin versus doxycycline for treatment of mild to moderate
Emerg Infect Dis 1998;4:179–86. scrub typhus. Antimicrob Agents Chemother 2007;51:2011–15.
12 Chapman AS, Bakken JS, Folk SM et  al.; Tickborne Rickettsial 36 American Academy of Pediatrics, International travel. In: Pickering
Diseases Working Group. Diagnosis and management of tickborne LK, Baker CJ, Overturf GD, Prober CG (eds) 2003 Red Book: Report of
rickettsial diseases: Rocky Mountain spotted fever, Ehrlichioses the Committee on Infectious Diseases, 26th edn. Elk Grove Village, IL:
and Anaplasmosis—United States: a practical guide for physicians American Academy of Pediatrics, 2003:93–8.
and other health‐care and public health professionals. MMWR 37 Park K. Epidemiology of communicable diseases. In: Park K (ed.)
Recomm Rep 2006;55;1–27. Park’s Textbook of Preventive and Social Medicine, 23rd edn.
13 Rathi N, Rathi A. Rickettsial infections: Indian perspective. Indian Jabalpur, India: M/s Banarasidas Bhanot Publishers, 2015:143–59.
Pediatr 2010;47:157–64. 38 Niebylski ML, Peacock MG, Schwan TG. Lethal effect of Rickettsia
14 Rahman MS, Gillespie JJ, Kaur S et al. Rickettsia typhi possesses phos­ rickettsii on its tick vector (Dermacentor andersoni). Appl Environ
pholipase A2 enzymes that are involved in infection of host cells. PLoS Microbiol 1999;65:773–8.
Pathog 2013;9:e1003399. 39 Kenyon RH, Williams RG, Oster CN, Pedersen CE Jr. Prophylactic
15 Sexton DJ, Corey GR. Rocky Mountain “spotless” and “almost spotless” treatment of Rocky Mountain spotted fever. J Clin Microbiol
fever: a wolf in sheep’s clothing. Clin Infect Dis 1992;15:439–48. 1978;8:102–4.
515

C HA PTER   44

Endemic Treponematoses: Yaws, Pinta


and Endemic Syphilis
Herman Jan H. Engelkens
Department of Dermatology and Venereology, Ikazia Hospital, Rotterdam, The Netherlands

Abstract Emergency Fund (UNICEF) and World Health Organization (WHO)


supported mass treatment campaigns in the 1950s and 1960s,
The treponemal infections that occur in humans are venereal the incidence of the endemic treponematoses has been greatly
syphilis and the endemic treponematoses (yaws, endemic syphilis reduced. However, these diseases have not been eradicated. Latent
and pinta). The endemic treponematoses, all of which are chronic cases are still prevalent and millions of people continue to be at
relapsing tropical diseases, have similar natural histories. Young risk of acquiring the endemic treponematoses from individuals
children are at the highest risk of acquiring these nonvenereal with relapsing disease.
treponematoses. Nearly all of the new cases are found in children In 2012, WHO launched a new initiative for the eradication of yaws
less than 15 years of age. As a result of United Nations Children’s by 2020.

Key points • Children younger than 15 years are predominantly affected.


• Late‐stage sequelae may lead to severe handicap.
• There is a new 2012 WHO initiative for eradication of yaws.
• The endemic treponematoses are yaws, pinta and endemic
syphilis.

Endemic syphilis (bejel, firjal or loath) is assumed to

SECTION 7: BACTERIAL SKIN


Definition and aetiology. The treponemal infections that
have originated before 8000 bce, probably as a result of
occur in humans are venereal syphilis and the endemic
a mutation of the (ancestor) treponeme, or as an adapta­
treponematoses (yaws, pinta and endemic syphilis). The
tion to new epidemiological factors [11–14]. The sexually
causative organisms of the endemic treponematoses

INFECTIONS
transmitted form of treponematosis, venereal syphilis,
are listed in Table 44.1. These organisms belong to the
emerged in a later millennium, at a time when bodily
order Spirochaetales, family Treponemataceae and genus
contact among adults was largely restricted to sexual
­
Treponema. In the case of T. carateum, propagation in an
­contact [11,14].
animal model has not yet been achieved; this is consid­
According to Hackett [11], four different treponemal
ered to be a separate species [1,2]. At present the causa­
diseases exist, but the different treponemes share a
tive agents of the different treponematoses cannot be
common biological ancestor, probably an animal path­
distinguished serologically or morphologically [1–5].
ogen. However, it has also been suggested that the four
The endemic treponematoses, all of which are chronic
clinical entities that are recognized today are, in fact, a
relapsing tropical diseases, have similar natural histo­
single disease caused by T. pallidum (unitary theory).
ries [6–10]. Young children are at the highest risk of
Changing environmental conditions are held responsi­
acquiring these nonvenereal treponematoses. Nearly all
ble for the different epidemiological patterns of the
of the new cases are found in children less than 15 years
treponematoses [13].
of age.
In 1905, Castellani [15] discovered the presence of spi­
rochaetes in ulcers from patients with parangi (yaws) in
History. It has been postulated that treponematoses orig­ Ceylon. The microorganisms resembled those observed by
inated some 22 000 years ago on the Eurasian–African Schaudinn and Hoffmann, who described the causative
landmass [11,12]. Pinta (carate, mal del pinto, azul) is agent of venereal syphilis earlier that year. As the cutane­
believed to be the first treponematosis occurring in ous lesions in yaws frequently resemble raspberries, the
humans. Yaws (framboesia tropica, pian, parangi, paru, disease was called ‘framboesia tropica’ and skin lesions
buba) probably arose around 10 000 bce on the Afro‐Asian were called ‘frambesiomas’. The causative agent of pinta
landmass from a mutant form of pinta. This mutated was first believed to be a fungus. In 1938, a spirochaete,
organism was more adapted to tropical circumstances. T. carateum, was recognized as the causative organism.

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
516 Section 7  Bacterial Skin Infections

Table 44.1  The causative agents of the endemic treponematoses


Aetiology. Direct personal skin–skin contact is the major
Yaws Treponema pallidum ssp. pertenue (T. pertenue) route of transmission of yaws. Breaks in the skin provide
Pinta Treponema carateum (T. carateum) a site of entry for the treponemes. Transmission is facili­
Endemic syphilis Treponema pallidum ssp. endemicum (T. endemicum) tated under overcrowded conditions, where open skin
lesions are common and in areas where no protective
clothing is worn. Infectivity is highest in humid tropical
climates. The mode of transmission of pinta is not well
The World Health Organization (WHO) and the United defined. Transmission most probably occurs through
Nations Children’s Emergency Fund (UNICEF) supported direct skin or mucous membrane contact. The disease is
mass treatment campaigns against the endemic trepone­ transmitted among children [7]. Endemic syphilis is most
matoses. These were executed with great success in the probably transmitted directly or indirectly by skin–skin
1950s and 1960s [6–10]. As a result of penicillin treatment or mouth–mouth contacts with infectious lesions [8].
during these mass campaigns, the incidence of the Theoretically, indirect transmission of treponemal disease
endemic treponematoses has been greatly reduced [16– by fomites and insects is probable, but no direct evidence
20]. However, these diseases have not been eradicated. for this mode exists [7,8].
Latent cases are still highly prevalent and millions of peo­ Serological evidence indicates that there is a reservoir
ple continue to be at risk of acquiring the endemic for yaws in West African baboons (Papio cynocephalus)
treponematoses from individuals with relapsing disease [44,45]. However, the significance of this reservoir for
[16–20]. In recent years, an increasing incidence of infec­ humans is not known. As a rule, sexual transmission does
tion is being reported from endemic regions [6–10,19,20]. not have a pathogenetic role in endemic treponematoses.
In 2012, WHO launched a new initiative, the Morges
strategy, for the eradication of yaws by 2020 [21].
Pathology
Yaws
Distribution The histopathological appearance of yaws is varied. In
Yaws early yaws, papillomatous epidermal hyperplasia is
Yaws is prevalent in rural areas of warm tropical regions the main feature. In addition, there is focal spongiosis
with high humidity and heavy rainfall. The main reser­ and neutrophils migrating into the epidermis may
voirs of infection are located throughout Africa [6,19–23]. give rise to intraepidermal microabscesses. There is a
SECTION 7: BACTERIAL SKIN

An alarming resurgence of yaws has been observed in the dense ­dermal perivascular infiltrate [46,47]. In contrast
tropical countries of West and Central Africa. In Ghana, with venereal syphilis, blood vessels are usually only
Benin, Togo and the Central African Republic, the preva­ mildly affected, and little proliferation of endothelial
lence of yaws resembles that seen in the precampaign era cells is found [47]. Epidermal changes, similar to those
INFECTIONS

[24]. An increase in seroreactivity has also been reported seen in condylomata lata, can occur. Hyperkeratotic
in Nigeria, Ivory Coast and Mali. In areas of South‐East lesions ­ f requently show nonspecific characteristics
Asia and the Pacific Islands (e.g. Indonesia, Timor‐Leste, (acanthosis, hyperkeratosis and parakeratosis) and
Papua New Guinea, Solomon Islands and Vanuatu), foci only a mild infiltrate [48,49]. With silver impregnation
of infection persist. Only a few countries in the Americas or immunofluorescent staining techniques (Figs  44.1
continue to report sporadic cases. In several endemic and 44.2), treponemes can be demonstrated [48,50,51].
areas, the current extent of yaws is not fully known, and Characteristic for yaws is the epidermotropic character
there is presumably considerable underreporting in many of T. pertenue [48–51].
affected areas [25–29]. In 1995, when WHO last issued
estimates, there were at least 2.5 million cases of endemic
treponematoses, mostly yaws [26].

Pinta
Pinta is still prevalent in remote rural regions in tropical
Central and South America [8,30]. Unfortunately, precise
figures on the prevalence of this disease are difficult to
obtain [6,21,31–33]. Serological surveys in these areas are
essential for estimating the current magnitude of this
treponemal reservoir.

Endemic syphilis
Endemic syphilis still exists, especially among isolated
closed communities living under primitive, crowded and
unhygienic conditions [6,19,34–40]. Infectivity is particu­
larly present in the arid climates of the eastern hemisphere,
among nomads and seminomads in Saudi Arabia and in Fig. 44.1  Numerous treponemes (Treponema pertenue) visualized by the
Sahel countries in Africa [34–43]. Steiner silver staining method.
Chapter 44  Endemic Treponematoses: Yaws, Pinta and Endemic Syphilis 517

Fig. 44.2  Treponema pertenue in the epidermis (immunofluorescent


staining).

Pinta
The histopathological changes in pinta are largely similar
to those in yaws [49,50,52], except that ulcer formation
does not occur [49]. In the early lesion, only mild acan­
thosis is present, with migration of some lymphoid cells
into the epidermis. Basal cells show loss of melanin and
liquefaction degeneration. In early‐stage lesions, melano­
phages may be seen in the upper dermis [50]. A moderate Fig. 44.3  Early (primary) yaws: infectious skin lesions on the leg.
dermal inflammatory infiltrate consists mainly of plasma
cells and lymphocytes. Histiocytes and neutrophils may initial skin lesions, but are greater in both number and
be seen. Occasionally, slight swelling of endothelial cells size. In the tertiary stage, deformities can occur. An

SECTION 7: BACTERIAL SKIN


is present [49,50]. In the late stage, irregular acanthosis or overlap between the stages is often noted. A more prac­
epidermal atrophy can occur. Pigmentary changes vary tical system of classification defines an early stage with
with the colour of skin lesions. A lymphocytic infiltrate contagious skin lesions, and a late stage, in which the skin
and numerous melanophages may be present in the lesions are less contagious [44]. Early yaws includes the

INFECTIONS
dermis. Treponemes can sometimes be visualized in the primary and secondary stage; late yaws is equivalent to
epidermis by means of silver staining methods or immu­ the tertiary stage.
nofluorescent techniques. Dyschromic late skin lesions Early stage skin lesions, often pruritic, appear after
may contain treponemes and show an inflammatory an  incubation period of 9–90 days, with an average of
infiltrate, but achromic late skin lesions lack both of these 3  weeks [6]. Classically, the initial presentation of yaws
features [50]. consists of one or more nontender papules which later
become crusted and ulcerated (Fig.  44.3). The lower
Endemic syphilis
extremities are a site of predilection. The early papules
The histopathological picture closely resembles that of
can evolve into ulcerated and papillomatous lesions,
venereal syphilis. In the early stage, the dermal infiltrate
containing numerous treponemes.
is primarily perivascular, and plasma cells and lympho­
Disseminated lesions may occur together with or after
cytes predominate. Slight vascular changes may be
the disappearance of primary lesions. These are some­
present. In early endemic syphilis, granulomas consisting
times preceded or accompanied by malaise, fever and
of epithelioid cells and multinuclear giant cells are seen
generalized lymphadenopathy. These early (secondary)
[50]. Silver or immunofluorescent staining methods may
stage skin lesions often resemble the initial lesions
assist in visualization of the treponemes in the early
(Fig. 44.4). Macules, papules and nodules can occur. Palms
stage. In the late stage, treponemes are rarely seen.
and soles often show hyperkeratosis (crab yaws). In the
early stage, bone and joint manifestations, particularly
Clinical features osteitis and periostitis, may already be noted [53]. Lesions
Yaws in the early stage of yaws usually disappear spontane­
Historically, several systems have been used to classify ously, sometimes leaving slight pigmentary changes.
the clinical stages of yaws [6]. The most frequently used However, severe secondary infection can be life‐threatening
system defines four stages (primary, secondary, tertiary and scarring is common. In the warm and rainy seasons,
and latent), equivalent to those seen in venereal syphilis. cutaneous lesions are more florid and abundant than
In the primary stage, skin lesions develop at the site of during drier periods of the year. During the dry season,
infection. Secondary stage skin lesions, a result of the patients may present with atypical macular cutaneous
widespread dissemination of treponemes, resemble the lesions.
518 Section 7  Bacterial Skin Infections

Fig. 44.5  Sabre tibia. This irreversible condition is caused by chronic untreated


periostitis. Source: Perine et al. 1984 [6]. Reproduced with permission of WHO.

Fig. 44.4  Multiple ulcerocrustopapillomatous skin lesions in early


(secondary) yaws. Dark‐field examination revealed the presence of many
SECTION 7: BACTERIAL SKIN

treponemes.

After the early skin manifestations have subsided, a


INFECTIONS

latent period of variable duration follows. The presence


of latent infection can be detected by the presence of a
positive serological test. This period may be interrupted
by one or more relapses of skin lesions. In the majority of
patients latency is lifelong. However, an estimated 10%
of patients develop a destructive late stage after 5–10
years or longer. Irreversible lesions of bone, cartilage,
soft tissue and the skin then develop. Manifestations
of late yaws include gangosa (destructive ulcerative
rhinopharyngitis), gummata, juxta‐articular nodes, con­
tractures and sabre tibiae (Figs 44.5 and 44.6). Gondou,
the rarely occurring exostoses of nasal and adjacent
bones, can also develop early in the course of the dis­
ease. Hyperkeratotic lesions may be observed in both
late and early yaws [6].
It has been generally assumed that no neurological or
cardiovascular involvement occurs in yaws. However, a
review in 1986 suggested that cardiovascular and neuro­
logical disease, similar to that in syphilis, may be seen in
patients with yaws [22,54,55]. In one study, ocular and
neurological abnormalities were described in patients
with late yaws [55]. Cerebrospinal fluid abnormalities,
optic atrophy, abnormal pupils and perivascular sheath­
ing were noted. As confusion between yaws and other
treponematoses is possible, and as there is no serological Fig. 44.6  Gummatous osteoperiostitis. Monodactylitis is characteristic of
test to differentiate between these diseases, additional late yaws bone lesions. Deformity of the fingers may result. Source: Perine
research is required [54]. et al. 1984 [6]. Reproduced with permission of WHO.
Chapter 44  Endemic Treponematoses: Yaws, Pinta and Endemic Syphilis 519

Pinta most common manifestations. Condylomata lata, compa­


The early stage of pinta includes the development of rable to those in yaws and sexually transmitted syphilis,
initial lesions and more generalized secondary lesions; occur frequently. In the early stage, a painful osteoperios­
the late stage includes a tertiary and late latent phase. titis, similar to yaws, may be observed. After early lesions
There may be considerable overlap between stages. After have subsided, the patient enters latency. In this stage,
an incubation period of several weeks to months, the ini­ treponemal infection can only be detected by positive
tial lesion, usually a papule or an erythematosquamous serological tests. In some patients, the late stage may
plaque, develops on exposed skin. This primary lesion develop at a young age [36,42]. In this stage, involvement
may become pigmented, hyperkeratotic and scaly, and of skin, bones and cartilage may lead to severe destruc­
may be accompanied by local lymphadenopathy. Primary tion, especially of the nose and palate. The larynx may
lesions of pinta disappear spontaneously. be solely affected [43]. In patients with nasopharyngeal
After several months or even years, more extensive and involvement, gummata may progress to destructive
generally smaller lesions may appear, which are similar in chronic ulceration, similar to rhinopharyngitis mutilans.
morphology to the initial lesions. These so‐called ‘pintids’ Neurological and cardiological involvement is extremely
may persist for years or resolve and then recur. Striking rare or perhaps nonexistent [42,43,56,57].
changes in skin pigmentation may result [6].
In late (tertiary) pinta, disfiguring pigmentary changes, Differential diagnosis. The diagnosis of endemic trepone­
achromia, skin atrophy and hyperkeratoses are common. matosis must be based on clinical, geographical, epidemi­
The pigmentary alteration may vary from lesion to lesion, ological and laboratory findings. At this point in time, it is
resulting in a sometimes lifelong mottling of the skin not possible to distinguish between the causative agents of
(Fig. 44.7). Skin lesions may turn red, white, bluish, violet the different treponemal diseases on serological, morpho­
or brown. Severe mutilation does not occur; it is assumed logical or biochemical grounds. Positive serological tests
that the skin is the only organ affected by this chronic in all but the very early stage, the presence of treponemes
treponemal disease [6]. Pinta is considered the most in dark‐field examination of exudates of cutaneous lesions
benign of the endemic treponematoses, with a good and the histopathological features confirm the diagnosis.
prognosis. No cardiological or neurological symptoms Diagnosis can be difficult in geographical regions where
have been described [6,7,55]. There is no known congeni­ venereal syphilis and/or endemic treponematoses are
tal form [7]. simultaneously prevalent, particularly for patients who
are in latency and the late stage (Table 44.2). Several other

SECTION 7: BACTERIAL SKIN


Endemic syphilis diseases in the tropics can resemble yaws, pinta and
Endemic syphilis may be divided into an early and a late endemic syphilis (Tables 44.3–44.5).
stage. The early stage includes primary and secondary
symptoms, and the late stage includes tertiary and late Laboratory tests. For the endemic treponematoses, the

INFECTIONS
latent disease. In contrast with the other treponematoses, same treponemal and nontreponemal tests designed
and perhaps because of the small inoculum, primary for the serodiagnosis of venereal syphilis are used. The
lesions may go unrecognized. In the primary phase, the treponemal tests are the T. pallidum haemagglutination
oropharyngeal mucosa is frequently involved. Frequently, assay (TPHA), the fluorescent treponemal antibody‐
endemic syphilis first manifests in the secondary stage absorbed test (FTA‐abs) and most enzyme‐linked immu­
and resembles secondary venereal syphilis. Patches on nosorbent assay (ELISA) tests. The nontreponemal tests
the mucous membranes, angular stomatitis, nonpruritic are the rapid plasma reagin (RPR) test and the Venereal
skin eruptions and generalized lymphadenopathy are the Disease Research Laboratory (VDRL) test. In remote regions,
only the nontreponemal tests are practical. In reference

Table 44.2  Some characteristics of treponematoses

Venereal Yaws Endemic Pinta


syphilis syphilis

Source of Sexual School, School, Social


infection partner house house
Age Adults 2–15 years 2–15 years 10–30 years
Type of climate All Humid/warm Dry/warm Dry/warm
Early genital Often Rare Rare Very rare
lesions
Neurological Sometimes No? No? ?
complications
Cardiovascular Sometimes No? No? ?
complications
Congenital Occasional No? No? ?
disease
Fig. 44.7  Pigmentary changes in pinta.
520 Section 7  Bacterial Skin Infections

Table 44.3  Differential diagnosis of yaws laboratories, the treponemal tests can be used to confirm
reactive nontreponemal tests. In the field, the most prac­
Skin lesions Venereal syphilis tical means for collecting and transporting blood for
Endemic syphilis serological tests is the filter paper method [58]. The
Pinta temporal relation between infection and reactivity, and
Leprosy
the persistence of positive serologies after treatment, are
Impetigo
Ecthyma the same in the endemic treponematoses and sexually
Tungiasis transmitted syphilis.
Tropical ulcers Recent progress in DNA technology, using Escherichia
Chromomycosis coli‐derived recombinant T. pallidum proteins, has led to
Cutaneous leishmaniasis the development of tests that can be used for sexually
Sarcoidosis, psoriasis
transmitted syphilis as well as for the endemic trepone­
Vitamin deficiencies
Scabies
matoses [59–61]. However, shared antigens give rise to
Viral infections such as mollusca contagiosa cross‐reactive antibodies common to all treponemal
or plantar warts diseases, precluding a differentiation of these diseases
Bone lesions Venereal syphilis on the basis of serological tests.
Endemic syphilis Demonstration of treponemes by dark‐field examination
Tuberculosis of exudates from early stage skin lesions can assist in diag­
Bacterial osteomyelitis
nosing treponemal disease, but there are no differentiating
Sickle cell anaemia
Rhinopharyngeal lesions Espundia (mucocutaneous leishmaniasis)
morphological criteria among the treponematoses.
Rhinosporidiosis
Rhinoscleroma Treatment. The drug of choice is long‐acting penicillin.
Tuberculosis Treatment consists of 600 000 U benzathine penicillin for
Leprosy all patients and contacts (for children under 10 years) and
South American blastomycosis
1 200 000 U (for older individuals) by single intramuscular
injection. No other drug is effective in a single dose.
Family members, contacts of patients and patients with
Table 44.4  Differential diagnosis of the skin lesions of pinta
latent infection should receive the same dose as those
with active disease [62]. For patients who are allergic to
SECTION 7: BACTERIAL SKIN

Venereal syphilis, yaws and endemic syphilis penicillin, alternative therapies are tetracycline and eryth­
Skin disorders with changes in pigmentation (vitiligo, pityriasis versicolor, romycin [63]. The newer tetracycline and erythromycin
pityriasis alba, erythema dyschromicum perstans, Riehl melanosis, analogues have not yet been tested on a large scale for the
chloasma, discoid lupus erythematosus) treatment of endemic treponematoses.
INFECTIONS

Eczema, psoriasis, tinea corporis, chronic pellagra, leprosy Change in titre of quantitative nontreponemal tests is
the major parameter used to measure response to therapy.
Treponemal tests may remain positive for life despite
Table 44.5  Differential diagnosis of endemic syphilis treatment. Strains of T. pallidum that are resistant to peni­
cillin have not yet been reported [63,64].
Skin lesions Venereal syphilis Endemic treponematoses are still prevalent in various
Yaws and pinta regions of the world, especially among people living in
Psoriasis unhygienic circumstances in inaccessible regions with
Eczema inadequate medical facilities [8,19,65]. Public health
Pityriasis rosea
concern about the endemic treponematoses diminished
Lichen planus
Leprosy, mycoses
after the mass campaigns, and a resurgence of endemic
Herpes simplex treponematoses has been documented [19,65,66]. Millions
Condylomata acuminata of people are again at risk of contracting these infections.
Perlèche and aphthae Owing to the ever‐increasing frequency of worldwide
Many diseases presenting with a generalized rash travel and migration, nonvenereal treponematoses can
Late stage Malignancies (carcinoma, mycosis fungoides, easily be transported to nonendemic areas, confronting
leukaemias, Bowen disease)
the local healthcare provider with a diagnostic dilemma.
Lupus vulgaris
Toxicodermas
A positive treponemal test result in a person originating
Bromoderma and iododerma from an endemic region must arouse the suspicion of one
Infiltrated types of rosacea and lupus of the non‐sexually‐transmitted treponemal infections [67].
Erythematosus, facial granuloma In the modern era, yaws and endemic syphilis may pre­
Nasopharyngeal lesions Yaws sent in a milder ‘attenuated’ form or an atypical form
Venereal syphilis [38,68–73] with less florid skin lesions. In areas with a low
Leishmaniasis
prevalence, solitary, small, flat, dry and hidden lesions of
Histoplasmosis
Rhinoscleroma short duration are seen. It has been argued that endemic
Rhinosporidiosis treponematoses demonstrate extremely variable disease
severity, depending on the frequency of infection in a
Chapter 44  Endemic Treponematoses: Yaws, Pinta and Endemic Syphilis 521

given community [68]. This phenomenon may be caused Early recognition and treatment of patients with endemic
by the widespread use of antibiotics, the improvement of treponematoses can prevent patients from developing
social conditions, a mutation or an adaptation of the later stage manifestations. Without institution of therapy,
causative organism or altered immune responses of the late‐stage sequelae may lead to severe handicap.
host. A modified form of pinta has not yet been reported.
The rarity or absence of congenital infection and of neu­ References
rological and cardiovascular involvement in the endemic 1 Smibert RM. Genus III Treponema Schaudinn 1905, 1728. In: Krieg NR,
Holt JG (eds) Bergey’s Manual of Systematic Bacteriology. Baltimore,
treponematoses (as opposed to venereal syphilis) is MD: Springer, 1984:50–2.
intriguing [55,56]. One theory suggests that the long time‐ 2 Fohn MJ, Wignall FS, Baker‐Zander SA et al. Specificity of antibodies
span between initial infection and pregnancy prevents from patients with pinta for antigens of Treponema pallidum subspecies
pallidum. J Infect Dis 1988;157:32–7.
the development of congenital infection in the offspring 3 Turner TB, Hollander DH. Biology of the Treponematoses. WHO
of an infected woman. However, other sources suggest Monograph Series 35. Geneva: World Health Organization, 1957.
that congenital infection does occur. As positive treponemal 4 Ovcinnikov NM, Delektorskij VV. Treponema pertenue under the elec­
tests in the newborn may be the result of the transplacental tron microscope. Br J Vener Dis 1970;46:349–79.
5 Hovind‐Hougen K, Birch‐Andersen A, Jensen HJS. Ultrastructure of
passage of immunoglobulin G (IgG), and as a serological cells of Treponema pertenue obtained from experimentally infected
test to differentiate yaws from venereal syphilis is not hamsters. Acta Path Microbiol Scand Sect B 1976;84:101–8.
available, further study of this question is needed. 6 Perine PL, Hopkins DR, Niemel PLA et  al. Handbook of Endemic
Treponematoses: Yaws, Endemic Syphilis and Pinta. Geneva: World
The worldwide acquired immune deficiency syndrome
Health Organization, 1984.
(AIDS) epidemic has led to new problems in the diagnosis 7 Gonçalves AP, Basset A, Maleville J. Tropical treponematoses. In:
and treatment of venereal syphilis in human immunode­ Canizares O, Harman RRM (eds) Clinical Tropical Dermatology, 2nd
ficiency virus (HIV)‐infected patients. To date, no reports edn. Oxford: Blackwell Scientific Publications, 1992:129–50.
8 Luger AFH. Endemic treponematoses. In: Parish LC, Gschnait F (eds)
of simultaneous HIV infection and endemic treponematosis Sexually Transmitted Diseases. A Guide for Clinicians. Berlin: Springer‐
have been published. Endemic treponematoses occur Verlag, 1989:32–58.
‘where the highways end’, regions where HIV infection 9 Engelkens HJH, Judanarso J, Oranje AP et al. Endemic treponematoses.
has not yet been introduced [74–76]. I. Yaws. Int J Dermatol 1991;30:77–83.
10 Engelkens HJH, Niemel PLA, van der Sluis JJ et al. Endemic trepone­
It has not been possible to cultivate T. pertenue in vitro matoses. II. Pinta and endemic syphilis. Int J Dermatol 1991;30:231–8.
for sustained periods. In experimental studies, hamsters 11 Hackett CJ. On the origin of the human treponematoses. Bull WHO
and rabbits are most frequently used for propagation of 1963;29:7–41.
12 Rothschild BM. On the antiquity of treponemal infection. Med
the treponemes [77]. Turner and Hollander [3] described Hypotheses 1989;28:181–4.

SECTION 7: BACTERIAL SKIN


a slightly different type of reaction to T. pertenue in 13 Hudson EH. Treponematosis in perspective. Bull WHO 1965;
hamsters, compared with T. pallidum. Previous genetic 32:735–48.
evidence indicated that T. pallidum and T. pertenue are 14 Hollander DH. Treponematosis from pinta to venereal syphilis revis­
ited: hypothesis for temperature determination of disease patterns.
indistinguishable [78]. However, Noordhoek showed that

INFECTIONS
Sex Transm Dis 1981;8:34–7.
T. pertenue and T. pallidum differ in at least one nucleotide 15 Castellani A. On the presence of spirochaetes in two cases of ulcerated
in a homologous antigen [79]. It is hoped that further parangi (yaws). Br Med J 1905;ii:1280.
16 Anonymous. Programme for the Control of the Endemic Trepone­
research will lead to the development of specific tests
matoses, VDT/EXBUD/87.1. Geneva: World Health Organization,
to  differentiate the treponematoses morphologically August 1987.
or  serologically. Reliable serological screening in field 17 Willcox RR. Evolutionary cycle of the treponematoses. Br J Vener Dis
­circumstances is of utmost importance. 1960;36:78–91.
18 Editorial. Endemic treponematoses in the 1980s. Lancet 1983;ii:551–2.
During relapses, individuals with latent treponema­ 19 Mitja O, Marks M, Konan DJP et al. Global epidemiology of yaws:
tosis will periodically present with infectious lesions. a systematic review. Lancet Glob Health 2015;3:e324–31.
All contacts exposed are at risk of contracting the disease. 20 Luger A, Meheus A. Drastische Ausbreitung endemischer Trepone­
Early detection and treatment campaigns are crucial. matosen. [Alarming spread of endemic treponematoses.]. Z Hautkr
1988;63:463–4.
Continuing surveillance by seroepidemiological evaluation 21 WHO. Eradication of yaws – the Morges strategy. Wkly Epidemiol
is also urgently needed. As gathered from past experi­ Rec 2012;87:189–200.
ence, treatment campaigns, continuing health education 22 Koff AB, Rosen T. Nonvenereal treponematoses: yaws, endemic syphilis
and pinta. J Am Acad Dermatol 1993;29:519–35.
and improvement of social and medical conditions can all 23 Sehgal VN, Jain S, Bhattacharya SN et al. Yaws control/eradication.
contribute to halting the spread of the treponematoses Int J Dermatol 1994;33:16–20.
and, it is hoped, will lead to their eradication [80–86]. 24 Widy‐Wirski R. Surveillance and control of resurgent yaws in the
The WHO launched a new initiative in 2012 (the Morges African region. Rev Infect Dis 1985;7(suppl 2):227–32.
25 Catterall RD, Hume JC. Summary and recommendations. Rev Infect
strategy) for the eradication of yaws by 2020, after the Dis 1985;7(suppl 2):343–50.
publication of a randomized controlled trial showing that 26 Maurice J. WHO plans new yaws eradication campaign. Lancet
a single dose of azithromycin was as effective as intra­ 2012;379:1377–8.
27 Zahra A. Yaws in Southeast Asia: an overview. Rev Infect Dis
muscular benzathine benzylpenicillin in a study of mass
1985;7(suppl 2):245–50.
treatment of yaws in Papua New Guinea by Mitja et al. 28 Sanyakorn CK. Situation of control of yaws in the Southeast Asia
[21,87,88]. region: past, present, and future. Southeast Asian J Trop Med Public
Health 1986;17(suppl):3–7.
29 Sosroamidjojo S, Rai K, Suesen N. Yaws in Indonesia. Southeast Asian
Prognosis. After penicillin treatment, early infectious J Trop Med Public Health 1986;17(suppl):19–34.
lesions of yaws and endemic syphilis usually heal within 30 St John RK. Yaws in the Americas. Rev Infect Dis 1985;7(suppl 2):
2 weeks. The skin lesions of early pinta heal more slowly. 266–72.
522 Section 7  Bacterial Skin Infections

31 Hopkins DR, Flórez D. Pinta, yaws, and venereal syphilis in Colombia. 61 Hardy PH. Prospects for improved laboratory diagnoses of trepone­
Int J Epidemiol 1977;6:349–55. mal infections and species differentiation. Rev Infect Dis 1985;7(suppl 2):
32 Falabella R. Nonvenereal treponematoses: yaws, endemic syphilis 300–4.
and pinta. J Am Acad Dermatol 1994;31:1075. [letter] 62 Ijsselmuiden OE. Development and evaluation of modern enzyme
33 Castro LGM. Nonvenereal treponematoses: yaws, endemic syphilis immunoassays for comprehensive syphilis serology. Thesis. Rotterdam,
and pinta. J Am Acad Dermatol 1994;31:1075–6. [letter] The Netherlands, 1989.
34 Fuchs J, Milbradt R, Pecher SA. Tertiary pinta: case reports and over­ 63 Report of a WHO Scientific Group. Treponemal Infections. WHO Tech
view. Cutis 1993;51:425–30. Rep Series, 1982:674.
35 Touré IM. Endemic treponematoses in Togo and other West African 64 Brown ST. Therapy for nonvenereal treponematoses: review of the
states. Rev Infect Dis 1985;7(suppl 2):242–4. efficacy of penicillin and consideration of alternatives. Rev Infect Dis
36 Pace JL. Treponematoses in Arabia. Saudi Med J 1983;4:211–20. 1985;7(suppl 2):318–26.
37 Basset A, Maleville J. Les tréponématoses endémiques (pian et béjel): 65 Stapleton JT, Stamm LV, Bassford PJ Jr. Potential for development of
des maladies utiles à connaître. Méd Afr Noire 1984;31:533–6. antibiotic resistance in pathogenic treponemes. Rev Infect Dis
38 Pace JL, Csonka GW. Endemic non‐venereal syphilis (bejel) in Saudi 1985;7(suppl 2):314–17.
Arabia. Br J Vener Dis 1984;60:293–7. 66 WHO. Programme for the Control of the Endemic Treponematoses,
39 Csonka G, Pace J. Endemic nonvenereal treponematosis (bejel) in VDT/EXBUD/87.1. Geneva: World Health Organization, August
Saudi Arabia. Rev Infect Dis 1985;7(suppl 2):260–5. 1987.
40 Gazin P, Meynard D. Enquête clinique et sérologique sur le béjel au 67 Engelkens HJH, Oranje AP, Stolz E. Early yaws, imported in The
nord du Burkina Faso. Bull Soc Pathol Exot 1988;81:827–31. Netherlands. Genitourin Med 1989;65:316–18.
41 Autier P, Delcambe JF, Sangaré D et  al. Etudes sérologiques et 68 Vorst FA. Attenuating endemic treponematosis. Thesis. Amsterdam,
cliniques sur la tréponématose endémique en République du Mali. The Netherlands, 1974.
Ann Soc Belge Méd Trop 1989;69:319–30. 69 Vorst FA. Clinical diagnosis and changing manifestations of trepone­
42 Abbas SMA, Satwekar SR. Positive treponematosis in Saudi antena­ mal infections. Rev Infect Dis 1985;7(suppl 2):327–31.
tals and their perinatal outcome over a 7‐year period. Saudi Med J 70 Niemel PLA, Brunings EA, Menke HE. Attenuated yaws in Surinam.
1989;10:301–4. Br J Vener Dis 1979;55:99–101.
43 Basset A, Maleville J, Basset M. Aspects de la syphilis endémique des 71 Niemel PLA, Sadal S, van der Sluis JJ. Yaws in Suriname. Rev Infect
Touareg du Niger. Bull Soc Pathol Exot 1969;62:80–92. Dis 1985;7(suppl 2):273–5.
44 Pace JL, Csonka GW. Late endemic syphilis: case report of bejel with 72 Fegan D, Glennon M, Macbride‐Stewart G et al. Yaws in the Solomon
gummatous laryngitis. Genitourin Med 1988:64:202–4. Islands. J Trop Med Hyg 1990;93:52–7.
45 Hackett CJ. The transmission of yaws in nature. J Trop Med Hyg 73 Csonka GW. Clinical aspects of bejel. Br J Vener Dis 1953;29:95–103.
1957;60:159–68. 74 Engelkens HJH, van der Sluis JJ, Stolz E. Syphilis in the AIDS era. Int
46 Fribourg‐Blanc A, Niel G, Mollaret HH. Confirmation sérologique et J Dermatol 1991;30:254–6.
microscopique de la tréponémose du cynocéphale de Guinée. Bull Soc 75 Engelkens HJH, Stolz E. Framboesia tropica en andere endemische
Pathol Exot 1966;59:54–9. treponematosen. In: Faber WR, Naafs B (eds) Import Dermatologie.
47 Engelkens HJH, Vuzevski VD, Judanarso J et al. Early yaws: a light Nieuwegein: Glaxo, 1991:107–12.
microscopic study. Genitourin Med 1990;66:264–6. 76 Engelkens HJH, Niemel PLA, Stolz E. De endemische treponema­
48 Engelkens HJH, ten Kate FJW, Vuzevski VD et al. Primary and second­ tosen in de jaren negentig. Memisa Med 1993;59:31–7.
SECTION 7: BACTERIAL SKIN

ary syphilis: a histopathological study. Int J STD AIDS 1991;2:280–4. 77 Schell RF, Le Frock JL, Babu JP et al. Use of CB hamsters in the study
49 Williams HU. Pathology of yaws especially the relation of yaws to of Treponema pertenue. Br J Vener Dis 1979;55:316–19.
syphilis. Arch Pathol 1935;20:596–630. 78 Miao RM, Fieldsteel AH. Genetic relationship between Treponema
50 Hasselmann CM. Comparative studies on the histopathology of pallidum and Treponema pertenue, two noncultivable human patho­
syphilis, yaws, and pinta. Br J Vener Dis 1957;33:5–12. gens. J Bacteriol 1980;141:427–9.
INFECTIONS

51 Lever WF, Schaumburg‐Lever G. Treponemal diseases. In: Lever WF, 79 Noordhoek GT. Syphilis and yaws: a molecular study to detect and
Schaumburg‐Lever G (eds) Histopathology of the Skin, 7th edn. differentiate pathogenic treponemes. Thesis. Utrecht, The Netherlands,
Philadelphia: J.B. Lippincott, 1992:352–9. 1991.
52 Engelkens HJH, ten Kate FJW, Judanarso J et  al. The localisation of 80 Antal GM, Causse G. The control of endemic treponematoses. Rev
treponemes and characterisation of the inflammatory infiltrate in skin Infect Dis 1985;7(suppl 2):220–6.
biopsies from patients with primary or secondary syphilis, or early 81 Meheus A. Integration of yaws control and primary health care. Rev
infectious yaws. Genitourin Med 1993;69:327. Infect Dis 1985;7(suppl 2):284–8.
53 Pecher SA, Azevedo EB. Aspectos histopatológicos da pinta terciária. 82 Hopkins DR. Control of yaws and other endemic treponematoses:
Med Cutan Ibero Lat Am 1987;15:239–42. implementation of vertical and/or integrated programs. Rev Infect
54 Engelkens HJH, Ginai AZ, Judanarso J et al. Case report 724. Skeletal Dis 1985;7(suppl 2):338–42.
Radiol 1992;21:194–7. 83 Capuano C, Ozaki M. Yaws in the Western Pacific Region. A review of
55 Román GC, Román LN. Occurrence of congenital, cardiovascular, vis­ the literature. J Trop Med 2011;2011:642832.
ceral, neurologic, and neuro‐ophthalmologic complications in late 84 Anselmi M, Araujo E, Narváez A et  al. Yaws in Ecuador: impact of
yaws: a theme for future research. Rev Infect Dis 1986;8:760–70. control measures on the disease in the Province of Esmeraldas.
56 Lawton Smith J, David NJ, Indgin S et  al. Neuro‐ophthalmological Genitourin Med 1995;71:343–6.
study of late yaws and pinta. II. The Caracas project. Br J Vener Dis 85 Meheus A, Antal GM. The endemic treponematoses: not yet eradi­
1971;47:226–51. cated. Rapp Trimest Statist Sanit Mond 1992;45:228–37.
57 Basset A. Pian et béjel ou syphilis endémique. Tempo Méd Afr 86 Farnsworth N, Rosen T. Endemic treponematosis: review and update.
1982;32:13–20. Clin Dermatol 2006;24:181–190.
58 Tabbara KF, Al Kaff AS, Fadel T. Ocular manifestations of endemic 87 Mitja O, Hays R, Rinaldi AC et al. New treatment schemes for yaws:
syphilis (bejel). Ophthalmology 1989;96:1087–91. the path toward eradication. Clin Infect Dis 2012;55:406–12.
59 Perine PL, Nelson JW, Lewis JO et al. New technologies for use in the 88 Mitja O, Hays R, Ipai A et  al. Single‐dose azithromycin versus
surveillance and control of yaws. Rev Infect Dis 1985;7(suppl 2):295–9. benzathine benzylpenicillin for treatment of yaws in children in
60 Stamm LV, Bassford PJ Jr. Cellular and extracellular protein antigens Papua New Guinea: an open‐label, non‐inferiority, randomised
of Treponema pallidum synthesized during in vitro incubation of freshly trial. Lancet 2012;379:342–7.
extracted organisms. Infect Immunol 1985;47:799–807.
523

C HA PTER   45

Tropical Ulcer
Vibhu Mendiratta & Soumya Agarwal
Department of Dermatology, Lady Hardinge Medical College and associated hospitals, New Delhi, India

Abstract vesicle/pustule at the site of trauma most occur on the lower leg)
followed by ulceration with perilesional oedema, purulent discharge
and pain. Untreated, the ulcer shows necrotic slough and invasion
Tropical ulcer (TU) is a synergistic bacterial infection caused by
of deeper subcutaneous tissues, fascia and bone. Histopathology is
Bacillus fusiformis and Treponema spp. A high prevalence of TU
necessary to exclude malignancy in the chronic stage. Complications
is recorded in Africa, South‐East Asia and South America. Current
include septicaemia, osteomyelitis, contractures and squamous cell
trends show a declining prevalence owing to better awareness of
carcinoma development. Treatment includes local cleaning with
environmental and personal hygiene, health education and avail­
soap and water/hydrogen peroxide, nonadherent antiseptic
ability of effective antimicrobial treatment. Aetiological factors
dressings, penicillin/macrolide antibiotics, metronidazole and, if
include minor, superficial trauma to skin, exposure to muddy
necessary, skin grafting.
water, poor hygiene and malnutrition. Clinically TU begins as a

Key points • Diagnosis is mainly based on clinical grounds. It can be


confirmed by dark‐field examination and culture of the causative
• Tropical ulcer (TU) is a painful, acute infective ulcer caused by organisms. Histopathology is required in chronic cases to rule
synergistic bacterial infection of Fusobacterium, spirochaetes out malignancy.
and/or anaerobic bacteria. • TU should be differentiated from other infective (Buruli ulcer,
• The incidence of TU is currently declining due to improved yaws, leishmaniasis, leprosy, deep fungal infections) and
socioeconomic conditions and better hygiene practices. The noninfective (traumatic, sickle cell anaemia, atherosclerosis,
prevalence is high in certain areas of Africa and South‐East Asia. vasculitis, drug‐induced) causes of leg ulceration.
• Predisposing factors include minor, superficial trauma to skin, • TU can be complicated by secondary bacterial infection, osteo­

SECTION 7: BACTERIAL SKIN


exposure to muddy water, poor hygiene and malnutrition. myelitis, malignant transformation, deformity and lymphoedema
• The disease has no sex predilection in childhood, but males due to scarring.
are more commonly affected in the adult age group. The most • The mainstay of treatment is systemic antibiotics including
common age group involved is 5–20 years. penicillin, streptomycin and metronidazole. It should be

INFECTIONS
• The most common site affected is the lower legs. TU starts as a supplemented with repeated antiseptic dressings, adequate
tiny vesicle/pustule that breaks down rapidly to form a tender, rest, improved nutrition and local cleaning with soap, water
round–oval, foul‐smelling ulcer that bleeds easily on touch. and/or hydrogen peroxide.

in the Transvaal in southern Africa. This author demon-


Introduction
strated the presence of Vincent’s bacilli alone or in asso-
Definition
ciation with Spirillum in the pus from ulcers affecting
Tropical skin ulcer (TU), also known as Naga sore, ulcus
mineworkers and hypothesized that some local infection
tropicum, Aden ulcer, Jungle rot, Malabar ulcer, and tropi-
of a minor wound was responsible for the ulcers [1].
cal phagedena, is a painful and disabling condition of the
Similar mixed isolates comprising fusiform bacilli,
lower leg and foot affecting primarily adolescents and
­spirochaetes and staphylococci were obtained from TU
young adults. TU is defined as a painful, acute inflamma-
in a series of 38 patients in India [2].
tory, rapidly spreading ulceration of skin and subcutaneous
TU has been referred to by different names in the past
tissue affecting the lower extremities which is accompanied
such as ‘Naga sore’ by Fox [3], ulcus tropicum by Panja [4]
by necrosis, sloughing oedema and foul‐smelling discharge.
and tropical phagedenic ulcer by MacAdam [5]. Both
The ulcer is characterized by an undermined edge and
Burnie and MacAdam suspected that infection had a
considerably raised margins [1–5].
causative role and both demonstrated that it was possible
to produce a TU by inoculating pus from affected indi-
History viduals into healthy volunteers [6]. TU was considered
Vinson was the first to mention tropical ulcers when epidemiologically important during World Wars I and II
describing them in 1857 as occurring in slaves aboard when returning European troops got infected, and reporting
a  ship. Later, features of tropical ulcers that affected of these cases by treating doctors diverted medical
mineworkers were studied by Spencer Lister in an epidemic attention to the subject [7]. Early reports from the 1930s to

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
524 Section 7  Bacterial Skin Infections

1950s are on record from South Africa and India that including many aerobes such as Staphylococcus aureus, can
described TU as acute, painful, inflammatory ulceration also be found. The reservoir of F. ulcerans is not fully
over the lower legs that was associated with great known, but it has been isolated from mud and stagnant
morbidity [4]. Earlier reports may lack evidence as to the water in endemic areas [13].
design but TU was first recognized as a distinct clinico- Panja [4] claimed that the identification of fusiform
morphological entity in tropical areas. The aetiology is bacilli in granulation tissue confirms that they cause
multifactorial, and still not known completely. Historical ulceration in TU. In a later study, Panja experimentally
suggestions throughout the literature support the belief produced TU in 11 volunteers after injecting pure cultures
that TU occurs only in people who are undernourished of fusiform bacilli obtained from ulcer swabs, further
and live in poor conditions [8,9]. In 1936, Clements supporting his theory that infection was truly responsible
described them as ‘one of the diagnostic garbage heaps for ulceration [4]. Results of the four‐country survey
of tropical medicine’ and argued that this was ‘a definite performed in the 1980s also indicated that fusobacteria
disease with its own signs and histopathology’ [8]. In were actively involved in the aetiology of tropical ulcera-
1963, Loewenthal defined a tropical phagedenic (meaning tion [18]. Further laboratory experiments found that these
‘to eat’) ulcer as ‘an acute, specific, localised necrosis of organisms produced acidic butyrate as a metabolite and
skin and subcutaneous tissues endemic in, but not con- were cytotoxic to cells in vitro, which might contribute
fined to, the tropical regions’. After the acute stage a to ‘rapid tissue destruction and ulcer formation’ [19].
chronic, nonspecific ulcer may persist [9]. Later, Tumwine Electron microscopy of the epidermis and dermis of
et al. gave a more detailed clinical definition of TU [10]. TU biopsies revealed fusobacteria deep in the dermis,
supporting Panja’s findings [4,20]. Gross destruction of
Epidemiology. TU is worldwide in distribution and collagen was observed, particularly near clusters of bacte-
cases are found in almost all tropical and subtropical ria. Oedema and huge numbers of polymorphonuclear
areas. A higher prevalence is noted in Nigeria, Uganda, leucocytes supported the suspicion that necrosis was
Democratic Republic of Congo, West Africa, the Bantu of caused by toxins released from fusobacterium [20].
southern Africa, Sri Lanka, Indonesia, Java, Papua New Factors like poor hygiene, exposure to muddy water
Guinea, the Pacific Islands, Madagascar and South and and malnutrition seem to predispose to the bacterial
Central America.The disease occurs mostly in sporadic infection and also facilitate spread of the ulcer. A wet trop-
and endemic forms, although outbreaks of almost ical climate is conducive to the easy spread of infection. In
epidemic proportions have been noted from Assam,
­ the presence of a susceptible host who sustains superficial
SECTION 7: BACTERIAL SKIN

Cochinchina, Malaya, certain parts of Africa, the Solomon skin trauma through a scratch/insect bite or abrasions
Islands and Melanesia [11]. India was among the initial from thorns/tall grass, the bacteria are inoculated into
regions to report TU; however, there has been a steady the skin leading to acute, painful ulcerations of the skin
decline in numbers over the last decade. (Box  45.1). TU is encountered more commonly in rural
INFECTIONS

All age groups from the age of 5 to 70 years can have areas where improperly clad farmers go to the fields and
TU; however, the majority of cases of TU are between bear a high occupational risk of sustaining minor skin
5 and 20 years. The disease shows no sex predilection in injuries. Contamination of minor abrasions sustained dur-
childhood, but occurs more commonly in men in adults ing farming by muddy water, dirt, or local applications of
in most countries. A review of 230 cases of TU from a cow dung, tobacco or many other locally available indig-
hospital in Haiti between 1963 and 1973 found ulcers enous products, promotes secondary infection by other
affecting both men and women (males more than females) bacteria, which produces chronic inflammation and
with a median age of 21 years, median duration of 2 years resultant lack of healing of the ulcers.
and healing time of 2 days to 18 years [12]. Robinson et al.
described 170 patents with 241 ulcers affecting the lower Clinical features. The clinical presentation of TU is similar
legs in predominantly male children aged between 5 and in all geographical locations. TU begins as a tiny vesicle
14 years. On the other hand, women were more com- that turns into a pustule over a couple of days. The vesiculo-
monly affected in Papua New Guinea. In the majority, the pustule ruptures to form a round to oval ulcer usually
ulcers were of less than 6 months’ duration [13]. Other located below the knee on the anterolateral side of the leg
retrospective data from Cook Island and from Zimbabwe or the ankle.The rapid breakdown of the initial preulcerative
recorded the predominance of ulcers in 10‐ to 19‐year‐old
males [10,14]. An epidemiological survey from Ethiopia
confirmed the age and sex distribution of TU in 6‐ to 15‐
Box 45.1  Tropical ulcer: predisposing factors
year‐old males [15]. A declining trend has been noted in
the prevalence of TU, but certain geographical areas still Hot and humid climate
continue to register significant numbers of cases. Trauma
Lack of footwear
Pathogenesis. TU is a synergistic bacterial infection that Malnutrition
Deficiency of micronutrients (e.g. zinc)
occurs secondary to invasion of skin by at least two
Overcrowding
organisms, one of which is a Fusobacterium spp., usually Poor socioeconomic conditions
F. ulcerans, while others include spirochaetes or some Poor hygiene
anaerobic bacteria [16,17]. In chronic TU, different bacteria,
Chapter 45  Tropical Ulcer 525

papule to form a TU constitutes one of the characteristic equinovarus. The tibia and fibula may become thin and
features of TU [13]. atrophic. Lymphoedema can result from scarring.
The ulcer bleeds easily on touch. It has circular, oedem-
atous margins that are raised above the surrounding skin.
Diagnosis. The diagnosis of TU is based on clinical his-
The ulcer base is granulomatous and shows a foul‐smelling
tory and examination. The presence of a round to oval,
grey slough (Fig. 45.1). Edges become rolling, along with
painful ulcer over the lower third of the leg discharging
evident perilesional pigmentation in the chronic stage.
pus and blood and covered with greyish slough should
There is usually no regional lymphadenitis.
arouse clinical suspicion of TU. Bacillus fusiformis and
The majority of TU remain confined to the skin only.
Borrelia vincentii have been found in more than 30% of TU.
Some may rapidly invade deeper tissues such as subcuta-
B. fusiformis is a cigar‐shaped Gram‐negative organism
neous tissue, fascia, tendons, muscles and bones in a span
that grows on an anaerobic medium containing serum
of a few weeks to months if neglected. Untreated ulcers
agar or serum broth. B. vincentii is a delicate organism,
can assume a large size of up to 15 cm. The duration of the
5–10 µm in length, with three to eight irregular spirals.
ulcer may range from 6 months to 15 years.
It is motile, Gram‐negative and stains with silver impreg-
nation. It is best demonstrated by dark–field examination
Complications (Box 45.2)
and is cultured on anaerobic medium consisting of serum
Superimposed secondary bacterial infections, septicae-
agar or broth. Many other Gram‐negative and ‐positive
mia and gangrene are acute complications. Malignant
bacteria can also be isolated in the chronic stage of a TU.
degeneration is rare before 20 years of ulcer duration, but
Histopathological examination of the ulcer shows a
may occur in about 2–9% of chronic TU. Squamous cell
chronic inflammatory infiltrate with necrosis and fibrosis.
carcinoma with lymph node spread is known to arise in
Features of vasculitis are absent. Pseudoepitheliomatous
the ulcer and has been reported. It may also develop at
hyperplasia is prominent in the late stage and simulates
the site of a healed ulcer scar. Healing of the ulcer may be
squamous cell carcinoma. Imaging (radiography/ ultra-
followed by flexion deformity of the knee and talipes
sonography) of the limb will reveal soft tissue thickening
and a periosteal reaction that later produces a thickened
sclerotic layer known as ivory osteoma.

Differential diagnosis. TU mimics a host of other tropical

SECTION 7: BACTERIAL SKIN


infections that cause leg ulcers. Buruli ulcer caused by
Mycobacterium ulcerans is common in children but they are
in good health and there is no pain. Yaws and cutaneous
leishmaniasis should also be differentiated.

INFECTIONS
Castellani’s ‘tropicaloid’ ulcer is caused by Corynebacterium
spp. and closely mimics TU.
Veldt sores or Barcoo rot and leg ulcers in sickle cell
anaemia over the inner side of the leg near the ankle also
should be differentiated from TU (Box 45.3).

Treatment. There is a dearth of literature on the treatment


of TU. General measures such as resting the affected
part, elevation of the limb and institution of adequate
diet are some important steps. Simple protective meas-
ures such as wearing clothing that fully covers the body
and boots to cover the feet are helpful in prevention.
Minor abrasions/cuts sustained during farming or acci-
dentally must be cleansed with clean water (which should
be preferably boiled for 15 minutes as for drinking) and
Fig. 45.1  A large chronic tropical ulcer with oedematous margins, serous
followed by the application of gentian violet. This will
discharge and unhealthy base over the lateral malleolus. help get rid of the infection. It is imperative also to treat
any other underlying chronic disease.
The patient with a TU should preferably be admitted
Box 45.2  Tropical ulcer: complications to hospital. While the results of the antibiotic sensitivity
are awaited, penicillin and streptomycin 1 g daily should
Severe debilitating pain in the acute phase be given for about 7–10 days [21]. Long‐chain penicillins
Scarring and contractures resulting in crippling deformities can be given once/twice a week to minimize injections.
Lower limb amputation
Alternatively, an oral tetracycline or erythromycin in
Squamous cell carcinoma at ulcer site
Hepatitis infection
divided doses can be used in children (after development
Septicaemia if untreated of the permanent dentition in the case of tetracyclines) or
in patients who cannot be admitted. Metronidazole also
526 Section 7  Bacterial Skin Infections

All patients are seen on at least four occasions and


Box 45.3  Tropical ulcer: differential diagnosis
hospitalized patients require a minimum of 15 days’ ther-
Acute leg ulcers
apy compared to 10 days if lesions are grafted early. Skin
grafting produces excellent responses but the drop‐out
Traumatic ulcer rate is high. There is a strong need for a uniform approach
Impetigo to the management of ulcerated skin lesions along with
Ecthyma ready access to grafting facilities to reduce the require-
Cellulitis ment for long‐term dressings and the risk of chronic
Drug‐induced (hydroxycarbamide, ergotamine, iododerma) ulcer
ulceration.
Radiation‐induced ulcer
Prevention of minor abrasions seems to be vital in
preventing TU and the wearing of long‐legged trousers
Chronic leg ulcers
is recommended. If a minor trauma occurs, first aid
Buruli ulcer including thorough cleansing of the lesion with soap
Veldt sore and water should be performed followed by an occlusive
Cutaneous leishmaniasis dressing.
Tuberculosis
Leprosy
References
Pyoderma gangrenosum
1 Lister FS. Aetiology of tropical ulcer. Transvaal Med J 1911;7:25–6.
Deep fungal infections 2 Rao RV, Kini MG, Subrahmanyan KS. Tropical ulcers in Madras city.
Yaws Ind Med Gaz 1949;84:88–92.
Vasculitis 3 Fox ECR. Naga sore. Ind J Med Res 1920;8:694–8.
4 Panja G. Aetiology and treatment of ulcus tropicum. Ind J Med Res
Squamous cell carcinoma
1945;33:11–16.
Sickle cell anaemia 5 MacAdam I. Tropical phagedenic ulcer in Uganda. J Roy Coll Surg
Venous/arterial ulcer Edinb 1966;11:196–205.
6 Burnie RM. Observations on tropical ulcer. West Afr Med J
1931;4:77–86.
7 Smyth EA. The treatment of tropical ulcer. J Royal Army Medical
has similar efficacy and can be used as a single therapeutic Corps 1946;87:141–2.
agent in a dose of 250 mg every 8 hours for 10 days. TU 8 Clements FW. Tropical ulcer with special reference to its aetiology.
Med J Australia 1936;2:615–44.
responds quickly with relief of pain and appearance of 9 Loewenthal LJ. Tropical phagedenic ulcer: a review. Int Rev Trop
SECTION 7: BACTERIAL SKIN

healthy granulation tissue [22]. Combination therapy Med 1963;2:267–91.


with metronidazole and amoxicillin–clavulanic acid can 10 Tumwine JK, Dungare PS, Tswana SA, Maoneke WR. Tropical
ulcers in a remote area in Zimbabwe. Cent Afr J Med 1989;
also be used [23]. 35:413–16.
Various other treatment modalities have been reported 11 Roy DN. Tropical ulcer. Brit Encyl Med 1949;12:320.
INFECTIONS

to be successful for TU: 12 Ariyan S, Krizek TJ. Tropical ulcer. Plast Reconstr Surg 1975;55:
• Antibiotics: in the early stages, penicillin or metronidazole 324–9.
13 Robinson DC, Adriaans B, Hay RJ et al. The clinical and epidemio-
is used in combination with topical antibiotics or antisep- logical features of tropical ulcer (tropical phagadenic ulcer). Int J
tics such as framycetin sulfate, neomycin, polymyxin B, Dermatol 1988;27:49–53.
bacitracin, nitrofurazone and povidone–iodine. 14 Kubersi T, Koteka G. An outbreak of tropical ulcer in the Cook Islands.
Am J Trop Med Hyg 1980;29:291–7.
• Improved nutrition and vitamins.
15 Bulto T, Muskel FH, Fisseha G. Skin lesions in resettled and indige-
• Nonadherent dressings. nous populations in Gambela with special emphasis on the etiology
• Large infected ulcers may require debridement under of tropical ulcer. Ethiopian Med J 1993;35:75–82.
anaesthesia. 16 Adriaans B, Hay RJ, Drasar B et al. The infectious aetiology of tropi-
cal ulcer–a study of the role of anaerobic bacteria. Br J Dermatol
• Skin grafting may be helpful in advanced cases to 1987;118:31–7.
ensure the lesion does not progress to the chronic stage. 17 Citron DM. Update on the taxonomy and clinical aspects of the genus
• In extreme cases, amputation is necessary. fusobacterium. Clin Infect Dis 2002;35(suppl 1):S22–7.
In the late stages of the ulcer, after infection has been ade- 18 Adriaans B, Drasar BS. The isolation of fusobacteria from tropical
ulcers. Epidemiol Infect 1987;99:361–72.
quately treated, pinch grafting of the skin is a good alternative 19 Adriaans B, Garelick H. Cytotoxicity of Fusobacterium ulcerans.
to secondary intention healing. The procedure is simple and J Med Microbiol 1989;29:177–80.
can be easily carried out by health workers in rural areas [24]. 20 Adriaans B, Hay R, Lucas S et  al. Light and electron microscopic
features of tropical ulcer. J Clin Pathol 1987;40:1231–4.
In specialized centres, wide excision of the infected 21 Ngu VA. Tropical ulcer. BMJ 1967;1:283–5.
bone and tendon can be carried out in chronic cases with 22 Yesudian P, Thambiah AS. Metronidazole in the treatment of tropical
associated underlying osteomyelitis. A split‐skin graft or phagedenic ulcers. Int J Dermatol 1979;18:755–7.
a full‐thickness pedicle graft is used to provide cover for 23 Aribi M, Breuillard F, Poirriez J. Guess what! Eur J Dermatol 1999;
9:321–2.
the resulting defect [25]. 24 Morris GE, Hay RJ, Srivastava A et al. The diagnosis and management
Watkinson et  al. [26] conducted a double‐blind trial of tropical ulcer in East Sepik province of Papua New Guinea. J Trop
on the use of oral zinc supplements as an adjunctive Med Hyg 1989;92:215–20.
25 Goodacre TEE. Plastic surgery in a rural African Hospital: spectrum
treatment for TU and concluded that, although zinc sup-
and implications. Ann R Coll Surg Engl 1986;68:42–44.
plements significantly elevated plasma concentrations of 26 Watkinson M, Aggett PJ, Cole TJ. Zinc and acute tropical ulcers in
zinc, they had no effect on ulcer healing. Gambian children and adolescents. Am J Clin Nutr 1985;41:43–51.
527

C HA PTER   46

Superficial Fungal Infections


Peter Mayser1 & Yvonne Gräser2
 Clinic of Dermatology, Allergology and Venereology, Justus Liebig University (UKGM), Giessen, Germany
1

 The National Reference Laboratory for Dermatophytes, Universitätsmedizin – Charité, Institute of Microbiology and Hygiene, Berlin, Germany
2

Introduction, 527 Candidosis (candidiasis), 545 Less common superficial fungal


Dermatophytoses, 527 Malassezia‐associated diseases, 550 infections, 556

Abstract carriers. Onychomycosis, although uncommon in children, increases


with age. Risk factors are sporting activities and infected parents.
Superficial mycoses are among the most common skin infections. Recalcitrant candidosis may be indicative of immune deficiency. Among
Molecular methods can accelerate diagnosis. In childhood, tinea children in the tropics, pityriasis versicolor is not an uncommon disease.
capitis, especially caused by anthropophilic dermatophytes, is of Furthermore Malassezia yeasts seem to be associated mainly with the
rising importance. Oral therapy is essential. Index cases due to head, neck and face types of atopic eczema/dermatitis syndrome, cor-
T. tonsurans, T. violaceum and M. audouinii warrant screening of all responding to the numerous sebaceous glands found in this region,
family members and close contacts and treatment of asymptomatic but it remains to be clarified whether this is also applicable to children.

Key points • Tinea capitis, especially when caused by anthropophilic


dermatophytes, is of rising importance in childhood. For
eradication of the organism, resulting in both clinical and
• Superficial mycotic infections are among the most common skin
mycological cure as quickly and safely as possible and reduction
infections in childhood.
of transmission to others, oral therapy is essential.
• Molecular methods can accelerate diagnosis.

Introduction skin and keratinized tissues such as hair and nails. The
severity of the infection depends on the dermatophyte
Superficial fungal infections are common in both temperate species involved and the immunological response of the

SECTION 8: FUNGAL SKIN


and tropical climates and although the majority occur in host. The clinical appearance of the lesions varies accord­
all age groups, some are seen only rarely in children. For ing to the site of infection and the causative organism.
these superficial infections, fungal invasion is restricted

INFECTIONS
to the skin, hair, nails and mucous membranes. In the
skin, these fungi are found only in the stratum corneum History. The first recognition that a fungus could be
and, very exceptionally, in the epidermis. Some of these the cause of an infection was in 1837, when Remak [1]
fungal infections show characteristic and diagnostic clini­ observed the presence of filaments and spores in material
cal features but, in others, laboratory procedures are from favus, a chronic infection of the scalp. In 1839,
required to differentiate them from other dermatoses. The Schoenlein [2] described the filaments as being those of
principal infections and their causative agents are listed moulds, and when the fungus was isolated by Remak [3]
in Table 46.1. The taxonomy of dermatophytes has been in 1845, he classified it in the genus Achorion and named it
renewed recently. The taxonomic names applied in this A. schoenleinii (now Trichophyton schoenleinii). In 1841,
chapter are based on current species conceptualizations. Gruby [4] also published on the fungal nature of favus; he
Table 46.2 shows the old and new species concepts in this recognized the different types of hair invasion in cases of
group of fungi. tinea capitis, giving the name Microsporum audouinii to
the fungus causing ectothrix‐type infection. In 1910,
Sabouraud [5] published his classic book Les Teignes,
Dermatophytoses representing the accumulation of his work on taxonomy,
morphology, diagnosis and treatment of dermatophy­
Definition. Dermatophytoses are infections caused by a toses. The dermatophytes were classified into four
specialized group of fungi, the dermatophytes, which are genera  –  Achorion, Epidermophyton, Microsporum and
able to invade and colonize the stratum corneum of the Trichophyton – on the basis of the clinical manifestations of

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
528 Section 8  Fungal Skin Infections

Table 46.1  Principal superficial infections and their causative agents A further development in taxonomy was the discovery of
the sexual states of some dermatophytes [7], which placed
Infection Agent the dermatophytes in their correct systematic position.
They are classified as Euascomycetes, order Onygenales
Dermatophytosis Microsporum spp.
and family Arthrodermataceae. Recently, molecular
Trichophyton spp.
Epidermophyton spp.
methods have been introduced to re‐evaluate the taxonomy
Nannizzia spp. of these three genera [8,9] and according to the new poly­
Pityriasis versicolor Malassezia spp. phasic species concept in dermatophytes, the number of
Tinea nigra Hortaea werneckii recognized species is now about 40. Not all cause human
White piedra Trichosporon spp. infections, as most of them have been isolated only from
Black piedra Piedraia hortae soil or other keratin‐containing substrates. The applica­
Neoscytalidium infections Neoscytalidium dimidiatum
tion of molecular methods in the last 20 years has revolu­
Neoscytalidium hyalinum
tionized fungal taxonomy, and phylogeny is nowadays
the mainstay for systematics. Extensive application of
Table 46.2  New and old species concept within the dermatophytes several gene markers has shown that the main topology
of the Arthrodermataceae seems to be stable, but does not
New species Synonymized taxa entirely correspond with morphology [8,9]. Therefore in
concept 2017 de Hoog et al. [9] confined Trichophyton, Microsporum
and Epidermophyton to mainly anthropophilic and zoo­
Mammal‐associated dermatophytes philic species located in derived clusters while the highly
Trichophyton T. mentagrophytes var. granulosum, A. benhamiae diverse geophilic species were located in the middle and
benhamiae
the bottom of the tree and were reclassified in Nannizzia,
T. bullosum Identical
T. concentricum Identical
Lophophyton and Arthroderma.
T. equinum All varieties of T. equinum A notable event in the study of dermatophytes was the
T. erinacei T. mentagrophytes var. erinacei report by Gentles [10] in 1958 of the successful treatment
T. eriotrephon Identical of experimental dermatophytoses in guinea pigs by the
T. interdigitale T. mentagrophytes var. interdigitale, goetzii, oral administration of griseofulvin. Subsequent clinical
nodulare, T. krajdenii trials proved the efficacy of griseofulvin in human derma­
T. mentagrophytes T. mentagrophytes var. mentagrophytes,
tophyte infections, and it remained the only systemic
granulosum, T. verrucosum var. autotrophicum,
A. vanbreuseghemii
therapy available for many years.
T. quinckeanum T. mentagrophytes var. quinckeanum, T. langeronii,
T. sarkisovii Aetiology, epidemiology and  pathogenesis. The most
T. rubrum T. fischeri, T. kanei, T. raubitschekii frequent dermatophyte species reported from human
T. schoenleinii Identical infections comprise three each in the genera Microsporum
T. simii A. simii
SECTION 8: FUNGAL SKIN

and Nannizzia, 17 in the genus Trichophyton and one


T. soudanense T. gourvilii, T. megninii
T. tonsurans All varieties of T. tonsurans
Epidermophyton species. Dermatophytes are not part of
T. verrucosum Remaining varieties of T. verrucosum the normal skin flora but have had a long‐lasting associa­
INFECTIONS

T. violaceum All varieties of T. violaceum, T. yaoundei tion with the human host. They can be divided into three
Epidermophyton Identical groups depending on their natural habitat.
floccosum • anthropophilic species, which affect only humans
Microsporum M. langeronii, M. rivalieri • zoophilic species, whose normal hosts are animals but
audouinii
which also infect humans
M. canis M. distortum, M. equinum, A. otae
M. ferrugineum Identical
• geophilic species, which are found in soil and are only occa­
sionally pathogenic for humans and animals (Box 46.1).
Main geophilic dermatophytes
Nannizzia gypsea M. appendiculatum, M. gypseum, A. gypseum
N. incurvata M. gypseum, M. incurvatum, A. incurvatum
N. fulva K. longifusus, M. boullardii, M. ripariae, M. fulvum, Box 46.1  Ecology of dermatophytes
A. fulvum
Anthropophilic Zoophilic Geophilic
E. floccosum M. canis N. fulva
M. audouinii N. gypsea
the diseases they caused as well as the morphological
M. ferrugineum T. equinum N. incurvata
characteristics of the fungi. In subsequent years, many T. concentricum T. erinacei
new species and genera were described, separated only T. interdigitale
on the grounds of small differences in fungal morphology T. rubrum T. mentagrophytes
or by the type of lesions they produced. T. soudanense T. benhamiae
By 1934, over 300 species in 40 genera had been pro­ T. schoenleinii T. simii
T. tonsurans T. verrucosum
posed. However, in 1934, Emmons [6] found that, on the
T. violaceum T. quinckeanum
basis of spore morphology, the dermatophytes could T. eriotrephon
be divided naturally into three genera, Microsporum, T. bullosum
Trichophyton and Epidermophyton, comprising 19 species.
Chapter 46  Superficial Fungal Infections 529

i­solated from African immigrants in the UK and other


Epidemiology. Changes in the prevalence of infections
parts of Europe while the indigenous population rarely
caused by different species are important because infec­
becomes infected by these two organisms. Whether racial
tions caused by these species vary in virulence and in
differences play a role in the epidemiology of geographi­
response to therapy. The two most widely distributed
cally restricted species is not understood. T. schoenleinii
anthropophilic species are Trichophyton rubrum and
was previously considered as a cosmopolitan anthropo­
Trichophyton interdigitale. Of these, T. rubrum, since its
philic species and was widespread in Europe but is now
introduction from the Far East to Europe and other coun­
mainly isolated from parts of Africa and the Near East.
tries after World War II, has become the most common
Before the 1960s, M. audouinii was the dominant cause
cause of skin and nail infections in most countries. In con­
(over 50%) of fluorescent scalp infections in Europe, the
trast, Trichophyton concentricum, the cause of tinea imbri­
USA and parts of Africa. Subsequently, the number of
cata, is endemic only in the South Pacific, the south‐eastern
M. audouinii infections markedly decreased in both Europe
periphery of Asia and parts of Latin America and Mexico,
and America. In a survey covering the years 1979–1981
and cases of this infection are rarely seen elsewhere
in the USA [12], it accounted for only 0.3% of the total
(Tables 46.3 and 46.4).
isolations of dermatophytes. It has been suggested that
The aetiology of tinea capitis worldwide is known
this decline reflects the success of griseofulvin in treating
to  change periodically. Microsporum ferrugineum is an
infections caused by this fungus. In parts of Africa,
example of an anthropophilic agent of tinea capitis
however, M. audouinii remains endemic and skin infec­
whose ­distribution changed. In Japan, where prior to the
tions, as well as scalp infections, are common.
1970s M. ferrugineum predominated, the zoophilic species
Trichophyton violaceum and Trichophyton tonsurans,
Microsporum canis is now isolated more frequently. M. fer-
although recognized as cosmopolitan, are far more com­
rugineum remains a common cause of infection in Korea,
mon in some parts of the world than others. T. violaceum
parts of China and north‐west Africa. Children of Korean
has always been the dominant cause of scalp infection in
immigrants to eastern Europe after World War II who had
the Indian subcontinent. It is also prevalent in North and
tinea capitis were found to be affected by M. ferrugineum,
East Africa, where 10–30% of the child population is
but this species appears not to have spread to the indig­
infected [13], and due to immigration is found with
enous population [11]. Similarly, the anthropophilic
increasing incidence in Europe [14].
­species Trichophyton soudanense was originally considered
Over the last 40 years, T. tonsurans has become the
geographically restricted to West Africa, and is now
most common causative agent of tinea capitis in the
USA, particularly in African‐American children living in
Table 46.3  Cosmopolitan dermatophytes urban areas. One report from Chicago [15] found that this
species accounted for 96% of all cases seen from 1976 to
Fungus Site of infection 1980. This was confirmed in a large survey from 1999 to
2002 by Foster et al. who found that T. tonsurans achieves
Anthropophilic E. floccosum Foot, groin, nail

SECTION 8: FUNGAL SKIN


near exclusionary proportions as the causal agent of tinea
T. interdigitale Foot, groin, nail
capitis in the USA [16].
T. rubrum Foot, nail, groin, body
T. tonsurans Scalp, body T. tonsurans is endemic in Mexico, Puerto Rico and

INFECTIONS
Zoophilic M. canis Scalp, body other Latin American countries, and it has been suggested
T. benhamiae Scalp, body that it was spread to the USA by immigrants from such
T. mentagrophytes Scalp, body areas. The fungus is also now endemic among the aboriginal
T. verrucosum Scalp, beard, body populations of parts of Australia [17]. The aetiology of
Geophilic N. fulva Scalp, body
tinea capitis in Europe has shown significant changes
N. gypsea Scalp, body
N. incurvata Scalp, body
during the last 30 years. In the 1980s, the dominant patho­
gens were zoophilic, with M. canis predominant. In the
early 1990s, a rise in the number of anthropophilic derma­
tophytes causing scalp ringworm was noticed in some
Table 46.4  Geographically limited species urban areas of north‐western Europe, particularly those
with large communities of Caribbean or African ancestry.
Fungus Site of Distribution
This trend has continued, and a survey performed under
infection
the auspices of the European Confederation of Medical
M. audouinii Scalp Africa
Mycology [18] confirmed a dramatic change from pre­
M. ferrugineum Scalp Far East, Africa dominantly zoophilic to anthropophilic infections in a
T. concentricum Body Western Pacific, South‐East Asia, South number of major European cities. The children involved
America are predominantly of African ancestry, but they are not
T. erinacei Body Europe, New Zealand, Asia, Africa recent immigrants; in fact the vast majority were born in
T. quinckeanum Body Asia, Africa, Europe Europe. Several anthropophilic species are involved.
T. soudanense Scalp Africa, Asia
In the UK and in Amsterdam, T. tonsurans is dominant.
T. schoenleinii Scalp Asia, Africa
T. violaceum Scalp, body, Africa, Asia In a 25‐year survey in Bristol, UK, the contributions of
nail T. tonsurans and T. violaceum to total dermatophyte isola­
tions increased tenfold, and in 2003 both accounted for
530 Section 8  Fungal Skin Infections

Table 46.5  Animal hosts of zoophilic dermatophytes scalp infections. Some of the asymptomatic carriers may
later develop clinical infections. Parents of children with
Fungus Major animal host(s) tinea capitis are also likely to be asymptomatic carriers of
the infecting fungus [23,24]. In the study by White et al.
M. canis Cats, dogs, horses
[25] of 209 household contacts of patients with tinea capi­
T. equinum Horses
T. erinacei Hedgehogs
tis caused by T. tonsurans, 44.5% had silent fungal carriage
T. mentagrophytes Rodent, cats, sheep on the scalp. Therefore all household contacts of patients
T. quinckeanum Camels, rodent with tinea capitis should be offered screening to eradicate
T. benhamiae Guinea pigs, rabbits a potential reservoir of infection.
T. simii Monkeys Constant exposure to possible infection may also influ­
T. verrucosum Cattle ence the incidence of disease. In closed communities, it
T. bullosum Donkeys
has been shown that the number of cases of tinea pedis
increases with the time of exposure to possible sources of
infection. For example, in one study carried out in a resi­
86% of all dermatophytes isolated from tinea capitis. In dential school, it was found that the incidence of toe space
contrast, M. canis prevalence decreased by 90% [19]. infections increased with the number of years the boys
In  Paris and its environs, however, T. soudanense and had been in the school [26]. New boys on entering the
M. audouinii predominate [18]; the incidence of the latter school had a 5% prevalence of infection; after 1 year, this
also increased in Switzerland [20]. had risen to 19% and after 2 years to 36%, whereas 54% of
Human infections caused by zoophilic species are the boys were infected if they had been at the school for
restricted to areas where the host animal is present. 4 years or more.
M.  canis, commonly associated with cats and dogs, has
always been a worldwide cause of human infection. In Pathogenesis. Dermatophytes penetrate keratinized tis­
many parts of Europe, and even in rural areas of countries sues by a combined enzymatic and physical attack on
that have seen such a dramatic rise in anthropophilic host tissues. They have been shown to produce enzymes,
infections in the major cities, this organism remains the such as keratinases, and in vitro invading dermatophyte
most frequent cause of tinea capitis. The countries report­ hyphae have been shown to be flattened, branched fronds
ing the highest incidence of M. canis infections are mainly adapted to force themselves between keratinocytes [27].
in the Mediterranean but also bordering countries such as Infection is transmitted by arthroconidia, spores formed
Austria, Hungary, Germany and Poland [21]. Other zoo­ by segmentation of fungal filaments (hyphae) that adhere
philic species with a wide distribution include Trichophyton to the skin, germinate and invade the stratum corneum.
benhamiae (guinea pigs, rabbits and other rodents) [22] For this infection to occur, some slight trauma or abrasion
and T. mentagrophytes (with a large range of animal hosts is necessary. Other factors that may play a role in the
inclusive of cats), T. verrucosum (mainly cattle) and T. equi- establishment of infection are skin surface factors such as
num (horses). pH and carbon dioxide tension; a rise in the latter has
SECTION 8: FUNGAL SKIN

By contrast, T. quinckeanum seems to occupy the same been shown to promote growth of fungi. Once established
distribution area as T. schoenleinii does [8]. Thus this spe­ on glabrous skin, the hyphae grow centrifugally to form
INFECTIONS

cies is very rare in Europe and other parts of the world. ‘ringworm‐like’ lesions. Different disease patterns are
Similarly, Trichophyton simii, found on monkeys in India, determined by the site of infection and the causative fun­
produces human infection only in these countries gus. The pathogenesis of experimentally induced and
(Table 46.5). naturally acquired scalp infections has been studied by
Kligman [28]. After initial infection of the scalp skin, the
Transmission and  source of  infection. Transmission of hyphae grow down into the space between the hair shaft
infection is usually indirect, occurring through infected and follicle wall. The hair is then invaded at about mid‐
skin scales and hairs shed from infected human or animal follicle level. The intrapilary hyphae grow down towards
hosts in which the infective spores (arthroconidia) may the hair bulb but stop at the upper limit of the keratoge­
remain viable for many months. Scalp infections may also nous zone, known as Adamson’s fringe. Hyphae within
be acquired through the use of contaminated combs and the hair are carried upwards by the growth of the hair, but
hairbrushes. Clothing, towels and bed linen are all possi­ the newly keratinized shaft is invaded by the fungus as
ble sources of infection. It is not difficult to isolate fungi soon as it is formed, at a rate of about 0.3 mm daily. When
from the floors of showers, changing rooms and dormito­ the invading fungus is one that produces an ectothrix
ries. It has been shown that in schools with outbreaks of infection (e.g. M. audouinii), the intrapilary hyphae
tinea capitis, many children in the same classroom as an emerge through the hair surface and fragment into masses
infected child carry the causative fungus on their scalps. of spores. The infected hair grows upwards and usually
This scalp carriage is believed to represent contamination breaks off a few millimetres above the scalp surface. In an
by spores shed from the infected child. It is usually tran­ endothrix infection (e.g. T. tonsurans), the intrapilary
sient, as once clinical cases are removed from the school, hyphae break up to form spores, which become rounded
the carrier rate decreases and then disappears. Never­ and fill the cortex of the hair. The invaded hairs break off
theless, these findings illustrate that there is widespread close to the scalp surface because of the greater degree of
dissemination of fungal spores from children who have shaft damage (‘black dot ringworm’). Favus is the name
Chapter 46  Superficial Fungal Infections 531

given to a chronic type of endothrix invasion of the hair within the epidermis can be observed [34]. Certain unsat­
caused by T. schoenleinii in which the hyphae grow irregu­ urated fatty acids present in sebum are inhibitory to the
larly down the hair shaft and do not fragment into masses growth of some dermatophytes in vitro. Their presence in
of spores. The shaft is not weakened and long lengths of postpubertal sebaceous secretions is thought to be an
infected hair are carried beyond the follicle by the growth important factor in preventing tinea capitis in adults.
of the hair. Degeneration of the hyphae leads to the for­ Tinea capitis in children may clear spontaneously at
mation of elongated air spaces in the hair. puberty, a time when greater quantities of sebum are
Infection of skin, hair and nails can persist only if the formed. Cell‐mediated immunity is thought to play an
fungus can grow into newly formed keratin as quickly as important role in defence against dermatophyte infection
old keratin is shed. Fungus‐specific factors may include [35]. Zoophilic and geophilic dermatophytes induce a
adaptation to the host, release of enzymes, production of delayed‐type hypersensitivity cell‐mediated response,
toxins and/or release of immune modulating agents. In which usually results in recovery and subsequent protec­
general, the better the fungus is adapted to its host, the tion against re‐infection. Clinically this might be repre­
milder the inflammatory response tends to be. Since sented by the characteristic healing from the centre of a
dermatophytes are almost exclusively localized in kerati­ classic ringworm lesion. The response is characterized by
nized tissues, this may require particular metabolic elevated levels of the key cytokines IL‐12 and interferon
activities. The spectrum of acidic proteases secreted by (IFN)‐γ, which trigger T‐helper 1 (Th1) cells for the activa­
dermatophytes is similar to that of Aspergillus, but differs tion of macrophages as effector cells. In contrast,
by multiple endoprotease members of the S8 (subtilisins), anthropophilic species (e.g. T. rubrum, T. interdigitale and
M35 (deuterolysin) and M36 (fungalysins) families [29,30]. T. tonsurans) tend to be associated with less inflammatory
Surprisingly, most of the proteases secreted in vitro dur­ but more chronic and persistent infections. These infections
ing keratin digestion in protein media were not detected are correlated with poor specific delayed‐type hypersen­
in vivo during the establishment of an infection. sitivity, elevated specific IgE and IgG4, and IgE‐mediated
Transcriptome analysis from guinea pigs infected by the immediate hypersensitivity (IH), with the production of
dermatophyte T. benhamiae and proteomic analyses of Th2 cytokines by mononuclear leucocytes [27,36].
proteins extracted from infected nail beds of patients with Infections by dermatophytes have been usually associ­
onychomycosis by T. rubrum revealed that Sub1, Sub2, ated with extrinsic conditions such as immunosuppres­
Sub7 and especially Sub6, not detected in vitro, were the sion, diabetes mellitus, advanced age, environmental
major proteases during infection [31,32]. Additional factors (e.g. humidity, occlusive footwear), contaminated
secreted proteases, including the closely related Sub7 and objects and surfaces as well as direct transmission by
the dipeptidyl peptidase DppV, were found. These results infected individuals or animals. However, it is well
from in vivo experiments are of particular interest since known that people are not equally susceptible to infec­
Sub6 and DppV in T. rubrum were previously identified tion, even when they have the same risk factors. There is
and described as the major dermatophyte allergens, Tri r2 some evidence for a familial or genetic predisposition to

SECTION 8: FUNGAL SKIN


and Tri r4, respectively [33].Tri r2 induces delayed‐type dermatophytosis [37]. The observations include special
hypersensitivity (DTH) reactions, which are assumed to forms of the disease in small isolated communities (e.g.
be associated with highly inflammatory lesions in derma­ tinea imbricata in Polynesians) and a high incidence of

INFECTIONS
tophytosis. In addition to a protective role of endogenous tinea in certain families, suggesting genetic susceptibility.
microbiota, host factors that play a role in the inflamma­ Specific defects in molecules that are important in innate
tion process include the site of entry, nonspecific defence and adaptive immunity have been shown for the Dectin‐1
mechanisms and both the adaptive and innate immune receptors, the caspase recruitment domain‐containing
response [27]. In more detail these factors comprise an protein 9, the transcription factor signal transducer and
increase in cell proliferation, the production of noncellular activator of transcription, the major histocompatibility
antimicrobial agents derived from epidermal keratino­ complex class II genes and the DEFB4 (defensin beta 4
cytes, complement‐related mechanisms and a specific gene) [37].
immune response involving the activation of immune‐
competent cells and a subsequent increase in Langerhans Pathology. Most dermatophyte infections are confined to
cells, T‐cell‐mediated delayed immune reaction and the the stratum corneum, nails and hair. In general, three dif­
production of antibodies. Innate immunity in superficial ferent changes in the stratum corneum can be associated
dermatophytosis implies the action of keratinocytes, with dermatophyte infections: the presence of neutro­
neutrophilic granulocytes and macrophages. Keratino­ phils, compact orthokeratosis and the presence of the
cytes may induce an acute and early response to infection, ‘sandwich sign’, which refers to the presence of hyphae
releasing a variable spectrum of cytokines upon initial ‘sandwiched in’ between an upper but normal bas­
interaction with a dermatophytic agent. These include ketweave stratum corneum and a lower layer of recently
interleukin (IL)‐8, a potent chemoattractant for neutrophils produced stratum corneum which is abnormal in being
which can kill dermatophytes, and the pro‐inflammatory compact orthokeratotic and parakeratotic in type [38].
tumour necrosis factor (TNF)‐α. Following an inflammatory In noninflammatory scaling lesions, hyperkeratosis
reaction, damage can be observed within the epidermal with little dermal infiltrate is present. In more inflamma­
barrier, epidermal proliferation is enhanced, and enhanced tory lesions, in the early phase, there is a dense neutrophil
expression of keratinocyte‐derived antimicrobial peptides accumulation in the stratum corneum and around the
532 Section 8  Fungal Skin Infections

hair follicles. When there is hair follicle invasion, fungal shows a characteristic type of endothrix invasion in which
fragments are surrounded by phagocytes or giant cells in hyphae and air spaces are found in the hair but very few
the dermis. spores are present. Another feature of scalp infections that
helps to distinguish the invading fungi is the bright‐green
Clinical features. The clinical features of dermatophyte fluorescence of infected hairs seen under an ultraviolet
infection depend on the body site infected, the causative (UV) lamp with maximal emission at 365 nm (Wood’s lamp)
organism and the immune status of the host. The degree of when Microsporum species are responsible. However, nega­
inflammation of the epidermis results from an immunologi­ tive fluorescence does not exclude Microsporum infection.
cally mediated reaction to the diffusion of fungal antigens Favus‐infected hairs fluoresce a dull green, but hairs invaded
from the stratum corneum. The amount of scaling is the result by other Trichophyton species show no fluorescence.
of increased epidermal replacement following inflammation.
Dermatophyte infections are commonly described Small‐spored ectothrix infections
using the Latin word ‘tinea’ followed by the Latin name When the causative fungi are anthropophilic (e.g.
of the site; for example, scalp ringworm is ‘tinea capitis’. M.  audouinii, M. ferrugineum), slowly extending circular
patches with partial hair loss occur in the scalp, with only
Tinea capitis mild erythema and scaling (Fig. 46.1). More inflammatory
Scalp ringworm is primarily a disease of childhood. The lesions with a greater degree of scaling occur when zoo­
incidence among adults is low, but adult cases of T. ton- philic or geophilic species are responsible (e.g. M. canis,
surans infection may be seen, for example in adults with N. gypsea) (Fig. 46.2).
acquired immune deficiency syndrome (AIDS). Although
the clinical appearance is in part dependent on the fungus Large‐spored ectothrix infections
responsible for the infection, there is always hair loss, with These infections are produced by the zoophilic species
varying degrees of scaling and erythema. Inflammation is T. mentagrophytes, T. benhamiae, T. quinckeanum and
generally more intense when zoophilic fungi are responsi­ T. verrucosum and are usually more inflammatory than
ble. However, an inflammatory lesion that becomes swol­ those caused by M. canis. Solitary lesions are typical
len and pustular, referred to as a kerion, may be produced with swollen and pustular areas (kerion) containing loose
by both zoophilic and anthropophilic fungi. With the hairs (Fig. 46.3).
exception of the two species Epidermophyton floccosum and
T. concentricum, all dermatophytes are able to invade hair, Endothrix infections
although T. rubrum is rarely reported as a cause of tinea Circular patches of alopecia with mild scaling and ery­
capitis in countries with a temperate climate. Scalp infec­ thema are characteristic. The infected hairs break off at
tions may be classified by the way in which dermato­ the surface of the scalp and in dark‐haired subjects give
phytes form spores after invading the hair shaft. a ‘black dot’ appearance (Fig.  46.4). In some patients,
In ectothrix infections, the spores are seen predomi­ scaling becomes widespread with a generalized erythema
SECTION 8: FUNGAL SKIN

nantly outside the hair shaft and may be either small resembling seborrhoeic dermatitis and leading to progres­
(2–3 µm) and arranged in masses, or larger (3–5 or 5–10 µm) sive hair loss.
and usually occurring in straight chains on the surface of Sometimes a kerion develops followed by scarring and
INFECTIONS

the hair. Small‐spored ectothrix invasion is produced by permanent alopecia; this has been seen most frequently
Microsporum species and the larger ectothrix by T. menta- when T. violaceum was the invading organism but also
grophytes, T. benhamiae and T. verrucosum respectively. In occurs in cases of T. tonsurans infection (Fig. 46.5).
endothrix infections, the spores (4–8 µm) are found within
the hair shaft, and this type of infection is produced by
Trichophyton spp., e.g. T. tonsurans (Table 46.6). Favus, a
chronic form of tinea capitis caused by T. schoenleinii,

Table 46.6  Types of hair invasion

Species Spore arrangement Size (µm)

Microsporum M. audouinii Ectothrixa


Nannizzia M. canis Ectothrixa
N. gypsea Ectothrixa 2–3
M. ferrugineum
Trichophyton T. quinckeanum Ectothrix 3–5
T. verrucosum Ectothrix 5–10
T. soudanense Endothrix
T. tonsurans Endothrix 4–8
T. violaceum Endothrix
T. schoenleinii Endothrixa

 Infected hairs fluoresce under Wood’s lamp.


a
Fig. 46.1  Tinea capitis caused by Microsporum audouinii.
Chapter 46  Superficial Fungal Infections 533

Fig. 46.2  Tinea capitis caused by M. canis and fluorescence under Wood’s lamp.

SECTION 8: FUNGAL SKIN


INFECTIONS
Fig. 46.3  Tinea capitis caused by Trichophyton mentagrophytes. Fig. 46.5  Tinea capitis with kerion caused by T. tonsurans.

Favus, a chronic scalp disease mainly caused by


T.  schoenleinii, is characterized by crusted, cup‐shaped
lesions (known as scutula) consisting of hyphae and
spores, which develop around follicular openings
(Fig.  46.6). Infections frequently produce considerable
scarring and alopecia and, unlike other forms of tinea
capitis, may persist into adult life. Patients are frequently
seen in families, with different generations being infected.
Tinea capitis is generally more common in boys than
girls, possibly because shorter hair allows easier access
for infecting fungal spores. However, in a survey of
fungal infections in the USA, more girls (59%) than
boys (41%) were found to have tinea capitis caused by
T. tonsurans [15].
The prevalence of a particular dermatophyte as the
Fig. 46.4  Tinea capitis caused by T. violaceum. cause of tinea capitis varies from country to country
534 Section 8  Fungal Skin Infections

Fig. 46.7  Tinea facei caused by T. mentagrophytes.


Fig. 46.6  Favus of the scalp.

Box 46.2  Geographical distribution of principal dermatophytes causing tinea capitis

Europe North and South America Australia Africa Middle East Asia
M. canis T. tonsurans M. canis T. violaceum M. canis T. violaceum
T. tonsurans M. canis T. tonsurans T. soudanense T. schoenleinii M. ferrugineum
T. tonsurans
M. audouinii

(Box  46.2). It should be noted, however, that children


whose parents and grandparents were immigrants from
other countries tend to be infected with the causative
fungi from their country of origin, irrespective of how
long they have been resident in their adopted country.

Tinea faciei
SECTION 8: FUNGAL SKIN

The face is an unusual site for a fungal infection except


when representing spread from another site, particularly
INFECTIONS

the scalp. Lesions are similar to those found on other


parts of the body (Fig. 46.7). They have been reported in
newborns and infants, and may be caused by anthropo­
philic or zoophilic species. In the USA, it has been reported
that the face was the most common site for skin lesions of
the body caused by T. tonsurans [15].

Tinea corporis
This term is used for infections of the trunk and limbs,
which are characterized by circular lesions arising by
centrifugal spread of the fungus from the initial site
of  infection. The degree of inflammation depends on
whether the fungus is zoophilic or anthropophilic. A zoo­
philic species such as M. canis produces erythematous Fig. 46.8  Tinea corporis caused by M. canis.
scaly circinate lesions, often with a raised border and
pustules; hairs in the involved area may become infected the infant’s family or infected pets are usually the source
(Fig.  46.8). In contrast, fungi of human origin such as of infection.
T. rubrum or T. tonsurans cause scaly and not very erythe­
matous lesions, although there is usually a clearly defined Tinea incognito
margin. Children of all ages can be infected, and rare Tinea incognito describes an infection in which the usual
cases have been recorded in the newborn. Many different clinical signs of tinea have been altered by the application
fungi have been implicated, including M. canis [39], of potent topical corticosteroids. The active margin of the
T.  mentagrophytes [40] and T. tonsurans [15]. Members of lesion may be lost, pruritus may be decreased or absent,
Chapter 46  Superficial Fungal Infections 535

Fig. 46.9  Tinea imbricata.

scaling may be absent, and the rash typically becomes


widespread. Pustules and papules are prominent [41].
Fig. 46.10  Tinea pedis caused by T. interdigitale.
Tinea imbricata
This is a type of chronic tinea corporis caused by T. con-
centricum. The fungus, and the disease it produces, has a
very restricted geographical distribution, being found
only in the western Pacific area (e.g. Papua New Guinea),
South‐East Asia and Amazonia. The infection is often
acquired during infancy and occurs in all ages. It is most
frequently observed on the trunk and limbs. The lesions
are very characteristic, consisting of extensive and persis­
tent rings of concentric scales (Fig. 46.9).

Tinea cruris
This infection is rare before puberty but is frequently seen
in adolescent males. The clinical features are erythema­

SECTION 8: FUNGAL SKIN


tous and scaly lesions extending symmetrically from the
groin down the inner surface of the thighs with a margin­
ated edge sometimes containing pustules; lesions are

INFECTIONS
often very itchy. The infection may spread to the perianal
area and buttocks. The causative fungus is T. rubrum, T.
interdigitale or E. floccosum. The last is almost always the
cause of outbreaks in groups of sportspeople or in people
living in close communities (e.g. boarding schools) and
Fig. 46.11  Tinea pedis in an infant caused by T. rubrum.
using shared bathing facilities. Under these circum­
stances, it is not difficult to isolate the causative fungus
from towels, bedding or clothing. clinical features depend on the fungus responsible. When
Annular rashes in the napkin area may also be caused T. interdigitale is the causative organism, small clear vesi­
by dermatophytes and have to be distinguished from cles may develop which eventually rupture, dry and
other forms of napkin dermatitis [42]. form an irregular scaly edge. Fine, dry scaling on the
soles is typical of T. rubrum infections. These tend to
Tinea pedis (athlete’s foot) become chronic and widespread, affecting the heels,
This is the most common form of tinea in temperate cli­ sides and dorsa of the feet.
mates. The causative fungi are all anthropophilic, and T. The incidence of tinea pedis in children before the age
rubrum is now the most common species isolated in the of puberty is low, although there are reports of cases in
majority of countries, having in the past three decades children less than 4 years old [43] (Fig. 46.11). Surveys of
supplanted T. interdigitale and E. floccosum. schoolchildren in the UK have recorded incidences rang­
The infection usually starts in the toe spaces, often ing from 2.2% in 7‐ to 10‐year‐old boys to 6.6% in 11‐ to
between the fourth and fifth toe, and is characterized by 14‐year‐old boys [44]. In Denmark, 15‐year‐old children
peeling, maceration and fissures (Fig. 46.10). In the acute showed an incidence of tinea pedis of 3.7% [45]. A 20‐year
phase, the lesions are usually itchy. Spread of the infection survey of tinea pedis in children between 3 and 13 years
may occur to the toes and soles of the feet, where the of age in Italy found only 80 cases compared with over
536 Section 8  Fungal Skin Infections

2500 cases in adults studied concurrently, children there­ established, the nail plate becomes friable and thickened.
fore representing only 3.1% of cases [46]. A study in France In developed countries, T. rubrum is the most common
looking at dermatophytosis in children over a 5‐year cause; in areas where T. tonsurans, T. violaceum or T. sch-
period recorded only two cases of tinea pedis in infants oenleinii is endemic [54], these fungi may produce similar
less than 2 years old but 19 cases among children aged infections.
between 2 and 10 years [47]. In Korea, a study over a
3‐year period examined 102 children with foot dermatitis Superficial white onychomycosis. This is an uncommon
and identified 21 cases of tinea pedis, based on positive type of nail damage produced by a fungus invading the
direct microscopy [48]. Seven of the children were less superficial surface of the nail plate, with minimal penetra­
than 4 years old and eight were between the ages of 5 and tion of the nail. White patches that are easily removed by
9 years. The reason for the different incidences in children scraping are produced in the nail. This type of infection
and adults is not known. Risk factors such as frequent use is mainly seen on toenails and is usually caused by
of swimming baths are the same in both age groups. T. interdigitale.
Parents of children with tinea pedis are frequently found
to have chronic dermatophyte infections. As in adults, Proximal subungual onychomycosis.  Fungal invasion
tinea pedis occurs more frequently in males than in females. occurs from the skin to the proximal nail bed and white
The clinical features in children are similar to those found spots occur under the nail; these may extend distally to
in adults, although vesicular lesions are perhaps more involve all layers of the nail (Fig. 46.12). It is more often
common and bullous lesions can also occur [49]. seen with yeast infection and rarely caused by a dermato­
phyte, but it has been particularly associated with patients
Tinea manuum with AIDS [55].
This is again rare in children. Lesions usually follow a
primary foot infection and T. rubrum is the usual cause. Endonyx onychomycosis. This is seen with infection due
Diffuse hyperkeratosis and peeling of the palms and to dermatophytes that cause endothrix scalp infections,
fingers are the most common features, often affecting notably T. soudanense. The nail plate is scarred with pits
only one hand, for reasons that are not understood. and lamellar splits. Invasion occurs from the top surface
Spread may occur to the dorsum of the hand when lesions but the fungus penetrates deeply into the nail plate [56,57].
have a more marked edge.
Total dystrophic onychomycosis. The entire nail plate is
Tinea unguium destroyed, leaving a thickened and abnormal nail bed.
Onychomycosis is uncommon in children, the incidence This may occur as a consequence of any of the three types
increasing with age. In a British study, only one case of of onychomycosis. Although the majority of toe‐ and fin­
tinea unguium was found in a survey of 494 schoolchil­ gernail infections in children are caused by anthropophilic
dren aged 5–10 years, an overall prevalence of 0.2% [50]. fungi (Fig.  46.13), occasionally zoophilic species such as
SECTION 8: FUNGAL SKIN

In the USA, a 3‐year survey of children under 12 years M. canis [58] and T. equinum [59] are responsible.
revealed 26 cases of onychomycosis [51]. Six children also
had tinea pedis. In France, a survey reported four cases of Mykids
INFECTIONS

onychomycosis in children up to 2 years old and a further An ‘id’ reaction is a secondary immunological reaction to
41 cases in children between 2 and 10 years [47]. Ninety circulating antibodies or activated T lymphocytes that are
nine cases of onychomycosis in children and adolescents directed against microbial antigens [60]. Superficial
were diagnosed in North Poland, representing 19.8% fungal infections are the most common cause of so‐called
of  all mycologically confirmed superficial mycoses (500
cases). Fingernail onychomycosis was recognized pre­
dominantly in children under 3 years of age (52 cases)
while 47 patients were affected by toenail onychomycosis,
mostly due to T. rubrum. The incidence increased steadily
with age [52].
As distinct from adults, most cases of onychomycosis in
children show no sign of infection in the adjacent skin sites.
However, the parents of children with tinea unguium are
frequently found to be infected and therefore are the most
likely source of infection [52,53]. Children with infection of
a fingernail by an anthropophilic organism causing tinea
capitis should be examined for scalp infection.
There are five types of nail involvement.

Distal and lateral subungual onychomycosis. This is the


most common form of fungal nail dystrophy. Invasion
starts at the distal end and side of the nail and then Fig. 46.12  Proximal subungual onychomycosis caused by T. mentagrophytes,
spreads proximally up the nail. As the infection becomes assisted by the habit of thumb sucking.
Chapter 46  Superficial Fungal Infections 537

1 Diagnosis based on clinical impression only is often


misleading. Clinically indistinguishable lesions may be
produced by organisms that are not always responsive
to the same form of therapy. For example, T. rubrum and
Neoscytalidium dimidiatum may cause clinically identical
infections of the skin and nails but the former is sensi­
tive to griseofulvin, whereas the latter is resistant.
2 The administration of oral antimycotics without identi­
fication of the aetiological agent is questionable.
3 Therapy options for Trichophyton spp. and Microsporum
spp., as well as for anthropophilic and zoophilic
species, may be different.
4 Identification of a zoophilic dermatophyte species
allows conclusions on the animal source of infection,
Fig. 46.13  Tinea unguium caused by T. rubrum.
which also has to be treated.
5 Early start of causative therapy.
6 For epidemiological reasons.
‘mykids’. Two major types of dermatophytids, a mykid
caused by a dermatophyte, have been described: (i) Collection of specimens
generalized lichenoid eruptions of grouped or scattered Skin lesions are best collected using a blunt scalpel.
follicular papules on the chest, trunk and back, mainly in Infected hairs (minimum of 10) may be removed with
children during the course of tinea capitis (‘lichen tricho­ flat‐ended forceps and infected hair stumps by scraping
phyticus’); and (ii) dyshidrotic and vesicular eczema on with a scalpel. Nails (3 mg or 20 small clippings) are often
the hands (palms and/or fingers) associated with tinea thickened and nail clippers are necessary to cut through
pedis (mainly caused by T. interdigitale). Other forms the whole thickness; subungual debris may also be
include nodular erythema/erythema nodosum, diffuse removed with a scalpel. The scalp may be sampled using
erythema and pustule formation. A set of diagnostic criteria either a shampoo brush or a toothbrush [63] when scaling
were defined to identify a dermatophytid reaction: is the predominant sign (mainly in cases of infection with
1 Primarily there is a proven focus of dermatophytic anthropophilic species).
infection elsewhere.
2 Fungal forms are not present in the site of the mykid. Microscopy
From this site, culture assays remain sterile and direct Samples are placed in a drop of 20–30% potassium or
mycological examinations as well as histopathological sodium hydroxide (KOH/NaOH) or tetra‐ethylammoni­
preparations are negative. umhydroxide (TEAH) on a microscope slide. Softening of
3 The dermatophytid symptoms spontaneously resolve

SECTION 8: FUNGAL SKIN


the tissue can be hastened by incubation in a wet chamber
after an acute course, provided that the primary focus for 20–30 min (1–2 h for nails) at room temperature. The
of fungal infection is eliminated. incorporation of dimethyl sulphoxide (DMSO) in KOH
4 The patients are sensitized to dermatophyte antigens

INFECTIONS
(DMSO 40 mL, distilled water 60 mL, KOH 30 g) may per­
and show a positive skin test response to an extract pre­ mit more rapid examination without heating. Unstained
pared from cultures of fungi of the genus Trichophyton preparations are generally satisfactory for the demonstra­
(‘Trichophytin’). tion of fungi in keratin, but chlorazole black E may be
A dermatophytid may worsen after effective, mainly sys­ added to DMSO to aid in the differentiation of hyphae
temic antimycotic therapy has been initiated and should from common artefacts such as cotton fibres, elastic fibres
not be confused with a drug reaction [60]. Topical (or or ‘mosaic fungus’. The last is an artefact caused by depo­
occasionally, if very severe, oral) corticosteroids may pro­ sition of cholesterol and other material around the periphery
vide symptomatic relief. of the epidermal cells. Calcofluor white (10–40 mg
Blankophor®, 95 mL NaOH 0.5 M, 5 mL DMSO), a stain
Diagnosis. The diagnostic procedures for superficial that fluoresces green on excitation with UV light, can also
fungal infection are changing. At present the microscopic be added to an equal volume of KOH. The stain is selec­
observation of fungal elements in specimens of infected tive for fungi and is helpful for the detection of scanty
skin, hair or nail followed by species identification using fungal elements in clinical material. The preparation must
culture is still widely used. However, molecular methods be examined with a fluorescence microscope.
to diagnose the causative agent play an increasing Infected skin scrapings show branching hyphae, often
role. Polymerase chain reaction (PCR) techniques and segmenting into chains of arthroconidia (Fig.  46.14).
matrix‐assisted laser desorption/ionization time‐of‐flight If  hairs are infected, the size and arrangement of the
(MALDI‐TOF) analysis [61,62] are able to detect dermato­ spores will contribute to the identification of the derma­
phyte and nondermatophyte species directly from clinical tophyte species involved (Figs 46.15–46.18) (see Table 46.6)
specimens or from a grown culture, respectively [61,62]. but an accurate species identification is not possible.
It is important to establish the correct diagnosis for the It should be noted that microscopy of skin, hair and nails
following reasons: is more often positive than culture.
538 Section 8  Fungal Skin Infections

Fig. 46.17  Endothrix invasion of hair by T. tonsurans (30% KOH).


Fig. 46.14  Hyphae and arthroconidia of dermatophyte in skin (30% KOH).

Fig. 46.18  Favus hair showing hyphae and air spaces infected by


T. schoenleinii (30% KOH).
SECTION 8: FUNGAL SKIN

(peptone 1%, dextrose 2–4%, agar 1.5%, pH 5.6). It is


recommended that the clinical material is inoculated on
INFECTIONS

Fig. 46.15  Small‐spored ectothrix invasion of hair by M. canis (30% KOH). two agar plates, one without (for moulds) and one with
gentamicin or chloramphenicol (0.005%) which is added
to reduce bacterial contamination and the medium is
made selective for dermatophytes by adding cyclohex­
imide (0.05%). Dehydrated media are commercially
available that include these antibiotics either incorpo­
rated in the agar powder (Mycobiotic Agar, Difco/Gibco;
Mycosel™, BBL™) or in separate vials (Dermasel™,
Unipath). Dermatophyte test medium (DTM) or so‐called
Taplin agar are not real alternatives because the alkalini­
zation of the media (red colour) is also induced by geo­
philic dermatophytes (e.g. A. terrestre) or moulds.
The macro‐ and micromorphology of the dermatophyte
colony is used for species identification, inclusive of the
texture of the colony, the surface colour and production of
pigments seen on the reverse side of the Petri dish. The
Fig. 46.16  Large‐spored ectothrix invasion of hair by T. verrucosum (30% KOH). three genera of dermatophytes are characterized by the
type of multicellular spores (macroconidia) produced on
culture (Fig. 46.19):
Culture 1 Microsporum and Nannizzia genus: fusiform
Petri dishes are satisfactory for the culture of dermato­ macroconidia;
phytes but quite time consuming (2–6 weeks). Sabouraud 2 Trichophyton genus: cylindrical macroconidia;
dextrose agar (SDA) is the medium most commonly used 3 Epidermophyton genus: pyriform macroconidia.
Chapter 46  Superficial Fungal Infections 539

(a) (b)

(c)

SECTION 8: FUNGAL SKIN


Fig. 46.19  (a) Microsporum, (b) Trichophyton and (c) Epidermophyton macroconidia.

The shape and arrangement of unicellular spores

INFECTIONS
(microconidia) and the presence of other structures such other members of the T. rubrum complex) are not reliably
as chlamydoconidia, spirals and so on are other features differentiated by any molecular target, inclusive of mark­
that aid in the identification of a dermatophyte [64]. ers with high discriminatory power, such as microsatellites
[65]. These factors should be kept in mind by anyone per­
Molecular diagnostics forming dermatophyte identification or reading the results.
A review on the new species concept of dermatophytes by In the past, an increased number of PCR strategies were
de Hoog et al. is available [9]. The taxon names applied in published and manufactured kits were developed which
this chapter are based on the new species concept. Table 46.2 can be applied in routine diagnostics to detect dermato­
shows the old and new species concepts in this group of phytes directly from clinical specimens. In general, the
fungi. It is recommended to strictly rely on the new con­ selection of the right molecular technique depends on its
cept, at least when molecular methods for identification are analytical sensitivity and specificity, corresponding inter­
used. For instance, routine laboratories that entirely rely on nal controls (positive, negative, extraction and amplifica­
conventional diagnostics are unable to further differentiate tion controls) and last but not least on effective DNA
between species of the T. mentagrophytes complex, i.e. T. extraction methods [66]. At the moment, commercially
mentagrophytes, T. benhamiae, T. interdigitale, T. erinacei and available kits for the detection of T. rubrum, T. interdigitale,
T. quinckeanum. Furthermore, it has been shown that E. floccosum (the main agents of onychomycosis and tinea
misidentifications are possible when using phenotypic fea­ pedis), M. canis, M. audouinii and N. gypsea at species level
tures. Given that the former T.  mentagrophytes complex are useful. Other pathogens of dermatophytoses are identi­
consists of species that are distinguished at many molecu­ fied at genus level only (e.g. Trichophyton spp.). In addition,
lar targets, it would be very confusing to use the name a detection reaction that identifies the dermatophytes as a
T. mentagrophytes for all isolates. The question would arise: group (Arthrodermataceae) should be included for the
which T. mentagrophytes is meant? In turn, species that have identification of new species and of geophilic dermato­
been synonymized (e.g. T. raubitschekii, T. megninii and phytes that are nonpathogenic [67].
540 Section 8  Fungal Skin Infections

Comparision of conventional and molecular Tinea cruris


diagnostic methods Tinea cruris should be differentiated from flexural pso­
Many studies have compared the sensitivity and speci­ riasis, seborrhoeic dermatitis, erythrasma and Candida
ficity of microscopy, culture and molecular methods intertrigo.
(conventional and real‐time PCR). The results uniformly
reveal that even at species level molecular methods are up Tinea pedis
to 21% more sensitive than direct microscopy and culture Other causes of interdigital scaling are Candida infection,
[68,69]. erythrasma and Gram‐negative and staphylococcal
A further advantage of molecular methods is the very infections.
short time period needed to diagnose the pathogen and
to start the appropriate therapy (24–48 hours compared Tinea manuum
to 2–6 weeks when using culture). The results of any Tinea manuum should be distinguished from eczema and
mycological detection system (culture and molecular) psoriasis.
need to be discussed against the background of clinical
plausibility. Saprophytic fungi without any clinical rele­ Tinea unguium
vance may be detected in culture if the medium is suit­ Psoriasis, eczema and Candida infection of the nails should
able. Using molecular methods, the detection of such be considered in the differential diagnosis of tinea
microorganisms will succeed only when specific prim­ unguium. Features of psoriasis of the nail are involvement
ers/probes have been developed. Thus rarely occurring of the same nails of the opposite hand or foot, pitting of the
dermatophyte species as well as yeast and moulds which nails and onycholysis; psoriasis is often present elsewhere.
are potential pathogens in about 5% of onychomycosis
cases may be identified using appropriate methods, e.g. Treatment. Although certain forms of dermatophytosis
the microarray technique. are self‐limiting, most types require treatment. Topical
In summary, PCR strategies supplement conventional therapy is used for localized lesions not involving hair or
methods at present but will be able to substitute for nails. Oral therapy is essential for chronic and widespread
microscopy and culture in the near future. infections and for those involving the scalp or nail.
The main antifungal drugs used in the treatment of der­
Further methods for the differentiation matophyte infections include the following.
of dermatophytes
The identification of dermatophyte species grown in Griseofulvin
culture is possible using MALDI‐TOF analysis [61,62]. This is an antifungal antibiotic derived from a number of
Identification of dermatophytes directly from clinical Penicillium species. It is fungistatic and affects nuclear
specimens is not possible. A prerequisite for identification division by binding to microtubular proteins. It also acts
using this method is that MALDI‐TOF platforms have as an inhibitor of nucleic acid synthesis. It is selectively
SECTION 8: FUNGAL SKIN

been validated by a sufficient number of strains of all rele­ concentrated in the stratum corneum, where it inhibits
vant dermatophyte species. The differentiation of phylo­ fungal growth sufficiently to prevent invasion of newly
genetically closely related species is, however, difficult, formed keratin. Griseofulvin was the first oral drug avail­
INFECTIONS

e.g. T. tonsurans/T. equinum, T. mentagrophytes/T. interdig- able for the treatment of dermatophytosis. Its activity is
itale or M. canis/M. ferrugineum. restricted to the dermatophytes and its clinical use is
limited to infections caused by these fungi. Given the
superior activity obtained with newer agents, griseoful­
Differential diagnosis
vin is rarely used now except for some scalp infections.
Tinea capitis
Scalp scaling without hair loss occurs in a number of con­ Azoles
ditions. Seborrhoeic dermatitis, psoriasis or pityriasis These are synthetic drugs that act by inhibition of ergos­
amiantacea may appear similar to tinea capitis. In pityria­ terol biosynthesis by fungal cytochrome P450 enzymes.
sis amiantacea, large numbers of thick scales are present Ergosterol is an essential constituent of the fungal cell
around hairs, which do not break. In alopecia areata, membrane. The imidazoles comprise a large number of
where there are patches of hair loss, there is usually no compounds primarily for topical use such as clotrima­
scaling and a number of broken tapering hairs (‘exclama­ zole, miconazole, econazole, bifonazole, isoconazole,
tion mark’ hairs) are present. Dermoscopy may be helpful oxiconazole, sulconazole, terconazole, tioconazole, fenti­
in differentiating tinea capitis (‘comma hairs’) from alope­ conazole and ketoconazole. They have a broad spectrum
cia areata (exclamation mark hairs) [70]. of antifungal activity including dermatophytes and yeasts
such as Candida and Malassezia species. Some also show
Tinea corporis activity against Gram‐positive bacteria.
Tinea corporis may be mistaken for other annular lesions The triazoles itraconazole and fluconazole are used
such as eczema, annular erythemas, psoriasis and granu­ orally for the treatment of superficial fungal infections,
loma annulare. Dermatophyte infections usually have a having a broad spectrum of activity similar to the imida­
distinct edge where scaling is more obvious, and the zoles. In many countries they are not licensed at present
lesions itch. for use in children.
Chapter 46  Superficial Fungal Infections 541

Allylamines dosages of griseofulvin (>18 mg/kg daily). When broken


These synthetic compounds block the formation of ergos­ down by species, the mean efficacy rates for Trichophyton
terol in the cell membrane by inhibition of squalene epox­ spp. and Microsporum spp. were 67.6%  ± 
9% and
idase. They have a broad spectrum of activity in vitro. 88.1% ± 5% respectively [72]. The drug should be taken
Naftifine shows antifungal activity against both dermato­ after meals. An oral suspension is available in some
phytes and yeasts and is used topically in the treatment of countries.
superficial fungal infections. It also has anti‐inflammatory Adverse reactions that have been reported with gri­
activity. Terbinafine is available for topical and oral use. seofulvin include drowsiness, headache and nausea;
Although topically effective against both yeasts and photosensitivity and urticarial reactions are also occa­
­dermatophytes, terbinafine, when given orally, is effective sionally observed. The use of selenium sulfide shampoo,
only against dermatophytes. Against these fungi, it shows 2.5% twice weekly, has been shown to reduce the frequency
primary fungicidal activity. Oral terbinafine is licensed of positive cultures from scalps when used with griseo­
for use in children only in some countries. It is available fulvin [73]. Ketoconazole shampoo may also be helpful
as a cream, a solution, a 250 mg tablet, and in the form of in conjunction with griseofulvin.
packaged granules. A 125 mg tablet is now available in The use of the allylamine terbinafine and the triazoles
some countries for treating children. itraconazole and fluconazole in the treatment of tinea
capitis has been reviewed by Gupta et  al. [74], who
Morpholines summarized many of the open study and trial data.
These compounds strongly inhibit ergosterol biosynthesis These drugs generally appear to have equal efficacy
at two different enzymatic stages specific for fungal cells with griseofulvin, and are effective with shorter courses
(the reductase and isomerase stage). Amorolfine, a phenyl of therapy.
propyl morpholine derivative, has in vitro activity against Few significant adverse effects have been reported. As
a variety of fungi including dermatophytes and yeasts; it active levels of drug remain in the keratinized tissues for
has fungicidal activity against dermatophytes. It is inac­ weeks after therapy has ceased, pulse and intermittent
tive when given systemically and is thus only available therapy have been reported to be successful with itracon­
for topical use. azole and fluconazole.
Terbinafine has been shown to be effective in the treat­
Miscellaneous ment of tinea capitis caused by Trichophyton spp. The fol­
Other miscellaneous compounds available for topical treat­ lowing dosages are commonly recommended:
ment include tolnaftate, ciclopiroxolamine, haloprogin • children weighing <20 kg, 62.5 mg
and derivatives of undecylenic acid. • children weighing 20–40 kg, 125 mg
• children weighing >40 kg, 250 mg.
Treatment of specific conditions With T. tonsurans, doses are generally given once daily
Tinea capitis. The British Association of Dermatologists’ for 4 weeks. Some reports suggest that a 2‐week course

SECTION 8: FUNGAL SKIN


guidelines for the management of tinea capitis were may be effective. The experience with terbinafine in
published in 2014 [71]. They also include a patient infor­ infants and neonates is been limited [75,76]. Terbinafine
mation leaflet (available on the British Association of is well tolerated in children between 2 and 17 years of

INFECTIONS
Dermatologists’ [BAD] website, http://www.bad.org.uk). age, with few adverse events. Gastrointestinal discom­
Oral therapy is essential to achieve the aims of treatment: fort and skin rashes have been reported. There is some
1 Eradication of the organism, resulting in both a clinical evidence that terbinafine is less effective in infections
and mycological cure as quickly and safely as possible; caused by Microsporum spp. and that these may need
2 Alleviation of symptoms; longer courses of therapy and higher doses [77–81]. In a
3 Prevention of scarring; and comparative study of a new paediatric formulation of
4 Reduction of transmission to others. terbinafine hydrochloride oral granules with griseofulvin
Adjuvant therapies such as antifungal shampoos or oral suspension, clinical cure was higher for terbinafine
topical antimycotics should be routinely combined. At than for griseofulvin, but the difference was not found to
present, griseofulvin is the only antifungal licensed for be statistically significant. Subgroup analyses revealed
oral use in tinea capitis in most parts of the world. that terbinafine was significantly better than griseofulvin
However cumulative evidence now demonstrates that among patients with T. tonsurans and vice versa with
newer antifungal agents have higher response rates and M. canis [82].
are safe and more cost‐effective. Itraconazole is also not yet licensed for oral use in tinea
Griseofulvin (10–15 mg/kg daily) should be given for capitis in many countries but has been used quite exten­
at least 6 weeks. In some cases, particularly with T. ton- sively in open studies and trials. It is available in capsule
surans infections, longer courses and sometimes higher form or as a suspension. It should not be given with a
doses (20 mg/kg daily) of griseofulvin therapy may be meal and can cause diarrhoea because of the presence of
needed [24]. M. canis infections may also need longer cyclodextrin. For continuous therapy, itraconazole is gen­
courses [71]. In a meta‐analysis of clinical trials of griseo­ erally given as 3–5 mg/kg per day for 4–6 weeks. In the
fulvin use in tinea capitis, the overall mean effective cure majority of the studies, the efficacy was high. In one study,
at 4–6 weeks post treatment was 73.4% ± 7%. Higher effi­ 50 children from six countries with tinea capitis caused by
cacy rates appeared to be reported with the use of higher either M. canis or T. tonsurans were included. The majority
542 Section 8  Fungal Skin Infections

of cases were treated with 100 mg of itraconazole daily for for 2 weeks and the patients were assessed at 4 weeks
approximately 5 weeks [83]. In total, 38 patients (76%) and, if clinically indicated, a further 1 week of treatment
were considered healed at the end of the study. Another was given. Terbinafine was given as follows: >40 kg,
study [84] compared 100  mg daily itraconazole and 250 mg per day; 20–40 kg, 125 mg per day; and <20 kg,
500 mg daily griseofulvin in the treatment of tinea capitis. 62.5 mg per day. The itraconazole dose was 5 mg/kg per
There were 17 patients in each group and the majority of day and fluconazole 6 mg/kg per day. At 12 weeks’ follow‐
infections were caused by M. canis. Treatment was given up, effective treatment was seen in 92% of the griseofulvin
for 6 weeks, and at follow‐up 8 weeks after the end of ther­ group, 94% of the terbinafine group, 86% of the itracona­
apy, 80% of patients in each group were cured. However, zole group and 84% of the fluconazole group. Overall,
another study in which 25 patients with T. tonsurans infec­ 6 weeks of griseofulvin therapy appears similar in efficacy
tion completed a course of 4 weeks of 100 mg itraconazole to 2–3 weeks’ therapy with the newer antifungals.
daily together with selenium sulfide shampoo resulted in Additional measures include the following [25,71]:
only 10 cures (40%) [85]. In one study, 163 children with 1 Children receiving appropriate therapy should be
tinea capitis caused by M. canis and previously unsuc­ allowed to attend school or nursery.
cessful therapy with terbinafine were treated with 5 mg/ 2 Index cases due to anthropophilic species, especially
kg per day given in a continuous regimen either as a cap­ T.  tonsurans, T. violaceum and M. audouinii, warrant
sule or an oral suspension. In all children, there was both screening of all family members and close contacts and
clinical and mycological cure after a mean treatment treatment for positive cases.
period of 39 ± 12 days [86]. In infants, itraconazole proved 3 In asymptomatic carriers (no clinical infection, culture
to be a safe and effective treatment option for M. canis‐ or PCR positive) with a high spore load, systemic treat­
induced tinea capitis [87]. ment is generally justified. If the spore load is low,
Pulse therapy has been reported to be successful. In a carriage may be eradicated with topical treatment
small study involving 10 children [88], the subjects took alone, but close follow‐up is needed, with repeated
5 mg/kg itraconazole daily for 1 week followed by 2 weeks pathogen detection, to ensure that treatment has been
off between consecutive pulses. One, two or three pulses effective. In an outbreak, additional infection control
resulted in clinical cure in one, six and three patients measures include antifungal shampoo use, exclusion of
respectively. A subsequent larger study produced a myco­ identified cases for a short period, removal of shared
logical and clinical cure in 30 of 37 patients (81%) [89]. items, and enhanced decontamination of fomites [94].
Few data are available for the use of fluconazole in See Table 46.7 and Box 46.3 for therapeutic recommenda­
tinea capitis. In one study, 48 patients with Trichophyton tions for tinea capitis.
tinea capitis were treated with 6 mg/kg daily for 2 weeks,
followed by 2 weeks without treatment. At 4 weeks Tinea corporis.  Unless there is widespread infection,
after the start of therapy, they were examined and those lesions usually respond well to a topical imidazole, such
with persistent clinical signs were given a further week’s as clotrimazole, or to ciclopiroxolamine, amorolfine or
SECTION 8: FUNGAL SKIN

treatment. Follow‐up at 12 weeks resulted in clinical terbinafine. These compounds are available as creams
and  mycological cure in 37 of 42 patients (88%) [90]. A and lotions and most will be effective after once or twice
subsequent larger study, including patients with both daily application for 2–4 weeks. Terbinafine cream is
INFECTIONS

Trichophyton and Microsporum infections, comprised 61 effective after 7 days of treatment applied once daily [95].
children, dosed at 8 mg/kg once weekly. At follow‐up,
complete clinical and mycological cure was seen in Tinea cruris.  Topical therapy is the same as for tinea
60 patients; of these, 47 were treated for 8 weeks, 10 for corporis.
12 weeks and three for 16 weeks. In total, three patients
suffered a mild, reversible gastrointestinal complaint and Tinea pedis. Tinea pedis will usually respond to topical
one patient showed an elevated liver function test, but treatment with one of the compounds mentioned under
this was asymptomatic and reversible [91]. In another Tinea corporis. The antifungal should be applied twice
study, fluconazole at two different doses (6 mg/kg per daily to the interdigital areas for at least 2 weeks; longer
day for 3 weeks followed by 3 weeks of placebo, flucona­ treatment is sometimes required. However, a compara­
zole 6 mg/kg per day for 6 weeks) was compared with tive study of 1 week of terbinafine 1% cream versus
standard‐dose griseofulvin (11 mg/kg per day microsize 4  weeks of clotrimazole 1% cream found the former
formulation) in tinea capitis caused mainly by T. tonsurans
(86%, 11% M. canis). At the end of treatment, both regi­
mens produced low mycological and clinical cure rates. Table 46.7  Recommendations for the oral therapy of tinea capitis
Mycological cure rates for fluconazole at 3 weeks and according to [71]: choice of drug according to organism isolated

6 weeks were 44.5% and 49.6% respectively. For griseoful­


Trichophyton tonsurans Terbinafine
vin the mycological cure rate was 52.2% [92].
Trichopython violaceum, soudanense Terbinafine
A study compared griseofulvin with terbinafine, itra­ Microsporum canis Griseofulvin or itraconazole
conazole and fluconazole in the treatment of Trichophyton Microsporum audouinii Griseofulvin or itraconazole
tinea capitis [93]. In total, 200 patients were randomized
to the four groups. Griseofulvin therapy was 20 mg/kg Source: Fuller et al. 2014 [71]. Reproduced with permission of John Wiley
per day for 6 weeks; the remaining antifungals were given & Sons.
Chapter 46  Superficial Fungal Infections 543

therapy in one infant and 19 children aged 8 years and


Box 46.3  Recommendations for the oral therapy of tinea capitis
older resulted in a complete cure rate of 80.0% (n = 20).
according to [71]: dosage of oral antifungals. (NB Terbinafine,
itraconazole and fluconazole are not licensed for oral use
The efficacy and safety profiles of terbinafine, itracon­
in childhood in many countries)
azole, griseofulvin and fluconazole in children were
similar to those previously reported for adults. When
Griseofulvin oral therapy is contraindicated, application of an anti­
fungal nail lacquer could be considered. Ciclopirox nail
>50 kg 1 g/day (single or divided dose) for 6–8 weeks
lacquer applied once daily for up to 6 months produces
<50 kg 15–20 mg/kg per day (single or divided dose) for 6–8 weeks
varying mycological cure rates from about 30% up to
Doses up to 25 mg/kg per day may be required in some cases 85.6% [100].
Alternatively, amorolfine 5% nail lacquer applied once
Terbinafine or twice weekly for up to 6 months produces cure in
approximately 40–55% of patients with mild onychomy­
<20 kg 62.5 mg/day for 2–4 weeks
cosis without nail matrix involvement. Studies in adults
20–40 kg 125 mg/day for 2–4 weeks
>40 kg 250 mg/day for 2–4 weeks
suggest that even better responses can be obtained by
combining amorolfine with either terbinafine or itracona­
zole [101].
Itraconazole

50–100 mg/day for 4 weeks, or 5 mg/kg per day for 2–4 weeks


References
1 Seeliger HPR. The discovery of Achorion schoenleinii. Mykosen
• Onychomycosis: application of an antifungal nail lacquer should be
1985;28:161–82.
considered, especially when oral therapy is contraindicated. 2 Schoenlein JL. Zur Pathologie der Impetigines. Arch Anat Phys Wiss
• Tinea corporis: topical therapy is preferred (imidazoles, such as Medical 1839:82.
clotrimazole; ciclopiroxolamine, amorolfine or terbinafine) unless 3 Remak R. Diagnostische und Pathogenetische Unterschungen in der
there is widespread infection. Klinik des Geh. Raths Dr Schoenlein auf dessen Veranlassung an‐ges­
tellt und mit Benutzung anderweitiger Beobachtungen veröffent‐
licht. Berlin: A. Hirschwald, 1845:242.
Source: Adapted from Fuller et al. 2014 [71].
4 Gruby D. Sur les mycodermes que constituent la teigne faveuse. CR
Acad Sci (Paris) 1841;13:309–12.
5 Sabouraud R. Les Teignes. Paris: Masson, 1910.
a­ ntifungal to be more effective [96]. Further studies have 6 Emmons CW. Dermatophytes: natural groupings based on the form
of spores and accessory organs. Arch Dermatol Syph 1934;30:
shown that even a single dose of terbinafine 1% film‐ 337–62.
forming formulation is as effective as the 1‐week course 7 Stockdale PM, MacKenzie DWR, Austwick PKC. Arthroderma simii
of terbinafine 1% cream [97]. sp. nov.: the perfect state of Trichophyton simii (pinoy) comb. nov.
Sabouraudia 1965;4:112–23.
If a mixed fungal and bacterial infection is suspected,
8 Gräser Y, Scott J, Summerbell R. The new species concept in dermato­
imidazoles such as clotrimazole, miconazole or econa­ phytes – a polyphasic approach. Mycopathologia 2008;166:239–56.
zole, which possess some antibacterial activity, are

SECTION 8: FUNGAL SKIN


9 de Hoog GS, Dukik K, Monod M et al. Towards a novel multilocus
preferable. If tinea pedis is of the dry scaly type, 4 weeks phylogenetic taxonomy for the dermatophytes. Mycopathologia
2017;182:5–31.
of topical treatment would be the minimum time to pro­ 10 Gentles JC. Experimental ringworm in guinea pigs: oral treatment
duce remission and the relapse rate is often high. Oral

INFECTIONS
with griseofulvin. Nature (Lond) 1958;182:476–7.
griseofulvin is the only licensed alternative for children
11 Prochacki H. Mycological flora isolated from people in Poland.
in many countries, but both oral itraconazole and terbin­ Mycopathol Mycol Appl 1970;40:65–72.
12 Sinski JT, Flouras K. A survey of dermatophytes isolated from human
afine have been shown to be effective in this type of patients in the United States 1979–1981 with chronological listings of
infection [98]. Oral terbinafine granules have recently worldwide incidence of five dermatophytes often isolated in the
been approved for use in children 4 years of age or older United States. Mycopathologia 1984;85:97–120.
13 Verhagen AR. Distribution of dermatophytes causing tinea capitis in
for the treatment of tinea capitis at the following doses: Africa. Trop Geogr Med 1974;26:101–20.
<25 kg, 125 mg once daily; 25–35 kg, 187.5 mg once daily; 14 Nenoff P, Krüger C, Ginter‐Hanselmayer G, Tietz HJ. Mycology ‐ an
>35 kg, 250 mg once daily. update. Part 1: Dermatomycoses: causative agents, epidemiology and
pathogenesis. J Dtsch Dermatol Ges 2014;12:188–209.
15 Bronson DM, Desai DR, Barskey S et  al. An epidemic of infection
Tinea unguium. This rare condition in children responds with  Trichophyton tonsurans revealed in a 20‐year survey of fungal
well to oral terbinafine even though there have been few infections in Chicago. J Am Acad Dermatol 1983;8:322–30.
clinical trials and in many countries the drug is not 16 Foster KW, Ghannoum MA, Elewski BE. Epidemiologic surveillance
of cutaneous fungal infection in the United States from 1999 to 2002.
licensed for use in children. Similarly, studies of itracona­
J Am Acad Dermatol 2004;50:748–52.
zole and fluconazole for the treatment of nail disease in 17 Rogers M, Muir D, Pritchard R. Increasing importance of Trichophyton
children are limited. Although there is faster nail growth tonsurans in childhood tinea in New South Wales. Australas J
in children and the success rates with oral therapy should Dermatol 1993;34:5–8.
18 Hay RJ, Robles W, Midgley G et  al., on behalf of the European
be higher, responses are sometimes disappointing. The Confederation of Medical Mycology Working Party on Tinea Capitis.
use of the newer antifungals in the treatment of onycho­ Tinea capitis in Europe: new perspective on an old problem. J Eur
mycosis in children was reviewed by Gupta et  al. [99]. Acad Dermatol Venereol 2001;15:229–33.
Therapy with systemic antifungals alone in children aged 19 Borman AM, Campbell CK, Fraser M et al. Analysis of the dermato­
phyte species isolated in the British Isles between 1980 and 2005 and
1–17 years resulted in a complete cure rate of 70.8% review of worldwide dermatophyte trends over the last three dec­
(n = 151), whereas combined systemic and topical antifungal ades. Med Mycol 2007;45:131–41.
544 Section 8  Fungal Skin Infections

20 Kieliger S, Glatz M, Cozzio A, Bosshard PP.Tinea capitis and tinea 50 Philpot CM, Shuttleworth D. Dermatophyte onychomycosis in chil­
faciei in the Zurich area – an 8‐year survey of trends in the epide­ dren. Clin Exp Dermatol 1989;14:203–5.
miology and treatment patterns. J Eur Acad Dermatol Venereol. 51 Chang P, Logemann H. Onychomycosis in children. Int J Dermatol
2015;29:1524–9. 1994;33:550–1.
21 Ginter‐Hanselmayer G, Weger W, Ilkit M et al. Epidemiology of tinea 52 Lange M, Roszkiewicz J, Szczerkowska‐Dobosz A et al. Onychomycosis
capitis in Europe: current state and changing patterns. Mycoses is no longer a rare finding in children. Mycoses 2006;49:55–9.
2007;50(suppl 2):6–13. 53 Bonifaz A, Saul A, Mena C et  al. Dermatophyte onychomycosis in
22 Nenoff P, Uhrlaß S, Krüger C et  al. Trichophyton species of children under 2 years of age: experience of 16 cases. J Eur Acad
Arthroderma benhamiae  –  a new infectious agent in dermatology. Dermatol Venereol 2007;21:115–17.
J Dtsch Dermatol Ges 2014;12:571–81. 54 Kalter DC, Hay RJ. Onychomycosis due to Trichophyton soudanense.
23 Babel DE, Baughman SA. Evaluation of adult carrier state in juvenile Clin Exp Dermatol 1988;13:221–7.
tinea capitis caused by Trichophyton tonsurans. J Am Acad Dermatol 55 Dompmartin D, Dompmartin A, Deluol AM et  al. Onychomycosis
1989;21:1209–12. and AIDS. Int J Dermatol 1990;29:337–9.
24 Elewski BE. Tinea capitis: a current perspective. J Am Acad Dermatol 56 Tosti A, Baran R, Piraccini BM et al. ‘Endonyx’ onychomycosis: a new
2000;42:1–20. modality of nail invasion by dermatophytes. Acta Dermatol Venereol
25 White JM, Higgins EM, Fuller LC. Screening for asymptomatic car- 1999;79:52–3.
riage of Trichophyton tonsurans in household contacts of patients 57 Baran R, Hay RJ, Tosti A et al. A new classification of onychomycosis.
with tinea capitis: results of 209 patients from South London. J Eur Br J Dermatol 1998;139:567–71.
Acad Dermatol Venereol 2007;21:1061–4. 58 Al‐Fouzan AS, Nanda A. Dermatophytosis of children in Kuwait.
26 Munro‐Ashman D, Clayton YM. Tinea pedis in adolescence. Proc Roy Pediatr Dermatol 1992;9:27–30.
Soc Med 1962;55:551–4. 59 Nicholls DSH, Midgley G. Onychomycosis caused by Trichophyton
27 Hube B, Hay R, Brasch J et  al. Dermatomycoses and inflammation: equinum. Clin Exp Dermatol 1989;14:464–5.
The adaptive balance between growth, damage, and survival. J Mycol 60 Ilkit M, Durdu M, Karakaş M. Cutaneous id reactions: a comprehen­
Med 2015;25:e44–58. sive review of clinical manifestations, epidemiology, etiology, and
28 Kligman AM. The pathogenesis of tinea capitis due to Microsporum management. Crit Rev Microbiol 2012;38:191–212.
audouinii and M. canis. J Invest Dermatol 1952;18:231–46. 61 Nenoff P, Erhard M, Simon JC et  al. MALDI‐TOF mass spectrome­
29 Burmester A, Shelest E, Glöckner G et al. Comparative and functional try – a rapid method for the identification of dermatophyte species.
genomics provide insights into the pathogenicity of dermatophytic Med Mycol 2013;51:17–24.
fungi. Genome Biol 2011;12:R7. 62 Gräser Y. Species identification of dermatophytes by MALDI‐TOF
30 Sriranganadane D, Waridel P, Salamin K et al. Identification of novel MS. Curr Fungal Infect Rep 2014;8:193–7.
secreted proteases during extracellular proteolysis by dermatophytes 63 Nasir S, Ralph N, O’Neill C et al. Trends in tinea capitis in an Irish
at acidic pH. Proteomics 2011;11:4422–33. pediatric population and a comparison of scalp brushings versus
31 Staib P, Zaugg C, Mignon B et al. Differential gene expression in the scalp scrapings as methods of investigation. Pediatr Dermatol
pathogenic dermatophyte Arthroderma benhamiae in vitro versus infec­ 2014;31:622–3.
tion. Microbiology 2010;156:884–95. 64 Kane J, Summerbell R, Sigler L et  al. Laboratory Handbook of
32 Méhul B, Gu Z, Jomard A et al. Sub6 (Tri r 2), an onychomycosis Dermatophytes. Belmont, CA: Star Publications, 1997.
marker revealed by proteomics analysis of Trichophyton rubrum 65 Gräser Y, Fröhlich J, Presber W, de Hoog GS. Microsatellite markers
secreted proteins in patient nail samples. J Invest Dermatol 2016; reveal geographic population differentiation in Trichophyton rubrum.
136:331–3. J Med Microbiol 2007;56:1058–65.
33 Woodfolk JA Allergy and dermatophytes. Clin Microbiol Rev 66 Gräser Y, Czaika V, Ohst T. Diagnostische PCR für Dermatophyten – ein
2005;18:30–43. Überblick. J Dtsch Dermatol Ges 2012;10:721–6.
34 Jensen JM, Pfeiffer S, Akaki T et al. Barrier function, epidermal dif­ 67 Jensen RH, Arendrup MC. Molecular diagnosis of dermatophyte
ferentiation, and human beta‐defensin 2 expression in tinea corporis. infections. Curr Opin Infect Dis 2012;25:126–34.
J Invest Dermatol 2007;127:1720–7. 68 Ohst T, Kupsch C, Graser Y. Detection of common dermatophytes
SECTION 8: FUNGAL SKIN

35 Almeida SR. Immunology of dermatophytosis. Mycopathologia in clinical specimens using a simple quantitative real‐time TaqMan
2008;166:277–83. polymerase chain reaction assay. Br J Dermatol 2016;174:602–9.
36 Vermout S, Tabart J, Baldo A et al. Pathogenesis of dermatophytosis. 69 Paugam A, L’ollivier C, Viguié C et al. Comparison of real‐time PCR
Mycopathologia 2008;166:267–75. with conventional methods to detect dermatophytes in samples
INFECTIONS

37 García‐Romero MT, Arenas R. New insights into genes, immunity, from patients with suspected dermatophytosis. J Microbiol Methods
and the occurrence of dermatophytosis. J Invest Dermatol 2015;135: 2013;95:218–22.
655–7. 70 Slowinska M, Rudnicka L, Schwartz RA et al. Comma hairs: a derma­
38 Ackerman AB. Histologic Diagnosis of Inflammatory Skin Disease. toscopic marker for tinea capitis: a rapid diagnostic method. J Am
Baltimore: Williams and Wilkins, 1997. Acad Dermatol 2008;59:S77–9.
39 Capesius‐Dupin C, Benaily N, Hennequin C et al. Dermatomycoses 71 Fuller LC, Barton RC, Mohd Mustapa MF et al. British Association
en pediatrie. J Mycol Med 1995;5(suppl 1):40–5. of Dermatologists’ guidelines for the management of tinea capitis
40 Monteiro CM, Ferreira JA, Goncalves HMG et al. Dermatophytosis 2014. Br J Dermatol 2014;171:454–63.
in a newborn infant: report of a case. Med Cut Ibero‐Lat Am 72 Gupta AK, Cooper EA, Bowen JE. Meta‐analysis: griseofulvin efficacy
1991;19:164–6. in the treatment of tinea capitis. J Drugs Dermatol 2008;7:369–72.
41 Ive FA, Marks R. Tinea incognito. BMJ 1968;3:149–50. 73 Allen HB, Honig PJ, Leyden JJ et  al. Selenium sulphide: adjunctive
42 Parry EL, Fosher WS, Marks JG. Diaper dermatophytosis. Am J Dis therapy for tinea capitis. Pediatrics 1982;69:81–3.
Child 1982;136:273–4. 74 Gupta AK, Hofstader SLR, Adam P et al. Tinea capitis: an overview
43 Kearse HL, Miller OF. Tinea pedis in prepubertal children: does it with emphasis on management. Pediatr Dermatol 1999;16:171–89.
occur? J Am Acad Dermatol 1988;19:619–22. 75 Kobayashi M, Yamamote O, Asahi M. Kerion celsi in an infant treated
44 English MP, Gibson MD. Studies in the epidemiology of tinea pedis. 1. with oral terbinafine. J Dermatol 2001;28:108–9.
Tinea pedis in school children. BMJ 1959;1:1442–6. 76 Ravenscroft J, Goodfield MJ, Evans EGV. Trichophyton tonsurans tinea
45 Svejgaard E, Albrectsen B, Baastrup N. The occurrence of tinea of the capitis and tinea corporis: treatment and follow‐up of four affected
feet in 15‐year‐old school children. Mykosen 1983;26:450–4. family members. Pediatr Dermatol 2000;17:407–9.
46 Terragni L, Buzzetti I, Lasagni A et al. Tinea pedis in children. Mycoses 77 Dragos V, Lunder M. Lack of efficacy of 6‐weeks treatment with oral
1991;34:273–6. terbinafine for tinea capitis due to Microsporum canis. Pediatr Dermatol
47 Reichert‐Penetrat S, Contet‐Andonneau N, Barbaud A et  al. 1997;14:46–8.
Epidemiology of dermatophytoses in children living in northeast 78 Hamm H, Schwinn A, Brautignam M et al. Short duration treatment
France: a 5‐year study. Pediatr Dermatol 2002;19:103–5. with terbinafine for tinea capitis caused by Trichophyton or Microsporum
48 Jang KA, Chi DH, Choi JH et al. Tinea pedis in Korean children. Int J species. Br J Dermatol 1999;140:480–2.
Dermatol 2000;39:25–7. 79 Koumantaki E, Kakourou T, Rallis E et al. Doubled dose of oral terbi­
49 Maroon MS, Miller OF. Trichophyton rubrum bullous tinea pedis in a nafine is required for Microsporum canis tinea capitis. Pediatr Dermatol
child. Arch Dermatol 1989;125:1716. 2001;18:339–42.
Chapter 46  Superficial Fungal Infections 545

80 Devliotou‐Panagiotidou D, Koussidou‐Eremondi TH. Efficacy and


tolerability of 8 weeks’ treatment with terbinafine in children with
Candidosis (candidiasis)
tinea capitis caused by Microsporum canis: a comparison of three Superficial candidosis is an infection of the mucous mem­
doses. J Eur Acad Dermatol Venereol 2004;18:155–9.
81 Lipozencic J, Skerlev M, Orofino‐Costa R et al. A randomized, dou­
branes, skin or nails caused by Candida species. These
ble‐blind, parallel‐group, duration finding study of oral terbinafine Candida species are normal commensals in the mouth and
and open, high‐dose griseofulvin in children with tinea capitis due gastrointestinal tract but are less frequently present on the
to Microsporum species. Br J Dermatol 2001;146:816–23. skin. The disease is worldwide in distribution. Infections
82 Elewski BE, Cáceres HW, DeLeon L et al. Terbinafine hydrochloride
oral granules versus oral griseofulvin suspension in children with may be acute or chronic with a broad clinical spectrum.
tinea capitis: results of two randomized, investigator‐blinded, mul­
ticenter, international, controlled trials. J Am Acad Dermatol History. Hippocrates, in the fourth century bce, first
2008;59:41–54.
83 Legendre R, Esola‐Macre J. Itraconazole in the treatment of tinea
described oral aphthae (thrush) in debilitated patients.
capitis. J Am Acad Dermatol 1990;23:559–60. Rosen von Rosenstein, in 1771, and Underwood, in 1784,
84 Lopez‐Gomez S, del Palacio A, van Cutsem J et al. Itraconazole ver­ in textbooks of paediatrics, recognized thrush as a condi­
sus griseofulvin in the treatment of tinea capitis: a double‐blind ran­ tion of the newborn and infants. In 1835, Veron [1] postu­
domized study in children. Int J Dermatol 1994;33:743–7.
85 Abdel‐Rahman SM, Powell DA, Nahata MC. Efficacy of itraconazole lated that newborns acquired the disease during passage
in children with Trichophyton tonsurans tinea capitis. J Am Acad through the vagina. Langenbeck was the first, in 1839, to
Dermatol 1998;38:443–6. discover the organism when he observed a fungus in
86 Ginter‐Hanselmayer G, Smolle J, Gupta A. Itraconazole in the treat­
ment of tinea capitis caused by Microsporum canis: experience in a
scrapings from a case of oral thrush in a patient with
large cohort. Pediatr Dermatol 2004;21:499–502. typhus, although he mistook the yeast as being the cause
87 Binder B, Richtig E, Weger W et  al. Tinea capitis in early infancy of typhus [2]. However, a few years later, Bennett (1844)
treated with itraconazole: a pilot study. J Eur Acad Dermatol [3] conclusively demonstrated the fungal aetiology of
Venereol 2009;23:1161–3.
88 Gupta AK, Alexis ME, Raboobee N et al. Itraconazole pulse therapy thrush. Zopf (1890) [4] named the fungus Monilia albicans,
is effective in the treatment of tinea capitis: an open multicentre from which moniliasis, the early name for candidosis
study. Br J Dermatol 1997;137:251–4. (candidiasis), originated. However, in 1923, Berkhout [5]
89 Gupta AK, Hofstader SL, Summerbell RC et  al. Treatment of tinea
showed that the Monilia species isolated from rotting fruit
capitis with itraconazole capsule pulse therapy. J Am Acad Dermatol
1998;39:216–19. or leaves differed both morphologically and physiologi­
90 Gupta AK, Adam P, Hofstader SL et al. Intermittent short duration cally from those associated with thrush and established
therapy with fluconazole is effective for tinea capitis. Br J Dermatol the genus Candida, which was accepted as a nomen conserva-
1999;141:304–6.
91 Gupta AK, Dlova N, Taborda P et  al. Once weekly fluconazole is
tum by the Eighth Botanical Congress in 1954.
effective in the treatment of tinea capitis: a prospective, multicentre
study. Br J Dermatol 2000;142:965–8. Aetiology and pathogenesis. Candida albicans is the major
92 Foster KW, Friedlander SF, Panzer H et al. A randomized controlled
aetiological agent of most clinical forms of candidosis. It
trial assessing the efficacy of fluconazole in the treatment of pediat­
ric tinea capitis. J Am Acad Dermatol 2005;53:798–809. is rarely isolated from natural sources such as soil, plants
93 Gupta AK, Adam P, Dlova N et al. Therapeutic options for the treat­ or water. Other Candida species implicated in human dis­
ment of tinea capitis caused by Trichophyton species: griseofulvin ease include mainly C. tropicalis, C. parapsilosis, C. glabrata,

SECTION 8: FUNGAL SKIN


versus the new oral antifungal agents, terbinafine, itraconazole and
fluconazole. Pediatr Dermatol 2001;18:433–8.
C. guilliermondii, C. krusei, C. lusitaniae, C. stellatoidea and
94 Gray RM, Champagne C, Waghorn D et  al. Management of a C. dubliniensis. Apart from C. glabrata, which, like C. albicans,
Trichophyton tonsurans outbreak in a day‐care center. Pediatr is a commensal yeast, and C. dubliniensis, which is associ­

INFECTIONS
Dermatol 2015;32:91–6. ated predominantly with AIDS patients, the other species
95 Evans EGV, James IGV, Joshipura RC. One week treatment of tinea
corporis and tinea cruris with terbinafine (Lamisil) 1% cream: a pla­ are often isolated from natural sources. However, fre­
cebo controlled study. J Dermatol Treat 1992;3:181–4. quent isolation of C. dubliniensis in caries‐active children
96 Korting HC, Kiencke P, Nelles S, Rychlik R. Comparable efficacy and was reported recently [6]. Candida spp. may also be iso­
safety of various topical formulations of terbinafine in tinea pedis
irrespective of the treatment regimen: results of a meta‐analysis. Am
lated from the hospital environment such as floors, wash­
J Clin Dermatol 2007;8:357–64. basins, bedding and other surfaces in wards [7]. However,
97 Ortonne JP, Korting HC, Viguié‐Vallanet C et  al. Efficacy and Candida cells show poor survival on dry surfaces and
safety of a new single‐dose terbinafine 1% formulation in patients transmission of infection by dry fomites is therefore
with tinea pedis (athlete’s foot): a randomized, double‐blind,
placebo‐controlled study. J Eur Acad Dermatol Venereol 2006; unlikely [8]. The success of C. albicans, the most prevalent
20:1307–13. and best studied Candida spp., as both commensal and
98 Hay RJ, McGregor JM, Ryatt KS et al. A comparison of 2 weeks terbi­ human pathogen depends on its genetic, biochemical and
nafine 250 mg/day with 4 weeks itraconazole 100 mg/day in plantar
morphological flexibility, which facilitates adaptation to a
type tinea pedis. Br J Dermatol 1995;132:604–8.
99 Gupta AK, Paquet M. Systemic antifungals to treat onycho- wide range of host niches. In addition, formation of bio­
mycosis in children: a systematic review. Pediatr Dermatol 2013;30: films provides additional protection from adverse envi­
294–302. ronmental conditions. Furthermore, in many host niches
100 Gupta AK, Fleckman P, Baran R. Ciclopirox nail lacquer topical solu­
tion 8% in the treatment of toenail onychomycosis. J Am Acad
Candida cells coexist with members of the human microbi­
Dermatol 2000;43(4 suppl):S70–80. ome. The resulting fungal–bacterial interactions have a
101 Baran R, Sigurgeirsson B, Berker D et  al. A multicentre, rand­ major influence on the success of C. albicans as commensal
omized, controlled study of the efficacy, safety and cost‐effective­ and also influence disease development and outcome [9].
ness of a combination therapie with amorolfine nail lacquer and
oral terbinafine compared with oral terbinafine alone for the treat­ As the most important source of Candida spp. in human
ment of onychomycosis with matrix involvement. Br J Dermatol disease is endogenous, many studies have been carried
2007;157:149–57. out on the yeast flora of the mouth, rectum, vagina and
546 Section 8  Fungal Skin Infections

skin of healthy subjects and of patients. The reported There have been many reports in the literature about
prevalence of Candida spp. from these sites depends the pathogenicity of the hyphal stage as opposed to the
largely on the sampling methods used. Imprint cultures yeast phase of C. albicans. The hyphal phase was consid­
or mouthwash samples are found to give more positive ered to be the pathogenic or parasitic phase and the yeast
findings of oral carriage than swabs. Odds [7] has calcu­ phase the saprophytic form. It is now known that the
lated from published data that the carriage rate of C. albi- transition from the oval, budding yeasts to parallel‐sided
cans in the mouths of healthy subjects ranges from 2% to hyphae is associated with adhesion to and penetration of
41%, whereas in hospitalized patients the range is from host cells and the temperature, pH and other environ­
6% to 70%, as determined by mouth swabs. Oral Candida mental factors. The ability to invade tissues and to induce
carriage also varies according to age. The mean carriage an inflammatory response is a characteristic of the species
rate of yeasts was calculated to be 17.3% in neonates, and not of a particular growth form. On body surfaces,
46.3% in infants aged 1 week to 18 months and 15.1% in the normal growth form of the organism is a yeast. Once
children over 18 months. Studies in neonatal intensive colonization and invasion of tissues have occurred, the
care units evaluating preterm infants found gastrointesti­ proportion of hyphal elements increases with the age of
nal colonization rates of 23% in 2157 infants [10] and the lesion. In superficial cutaneous candidosis, the yeasts
79.6% in 54 neonates [11]. The mean carriage rate in adults and hyphae are restricted to the stratum corneum. The
was found to be 25.1% [7]. main changes in the early stages of Candida invasion are
In the vagina, the carriage rate of C. albicans in normal the infiltration of the epithelium by neutrophils with
females is rarely found to be more than 20%. The vaginal some hyper‐ and parakeratosis. There is upper dermal
carriage of yeasts is more prevalent in pregnant than in infiltration of lymphocytes and plasma cells.
nonpregnant women, with the highest prevalence being
in the third trimester of pregnancy. The influence of oral Clinical features
contraceptives depends on the oestrogen dosage [7]. Oral candidosis
The change from a commensal status to a pathogenic This condition can be divided into a number of distinct
one is associated with a number of underlying host condi­ clinical forms [14]: (i) acute pseudomembranous candi­
tions. Factors predisposing to Candida infections include dosis; (ii) acute erythematous atrophic candidosis; (iii)
the following: chronic erythematous candidosis; and (iv) chronic hyper­
• age plastic candidosis (Candida leucoplakia).
• pregnancy
• local occlusion and maceration Acute pseudomembranous candidosis (thrush).  This
• antibiotic therapy presents with white plaques on the buccal mucosa and
• endocrine disease lateral borders of the tongue. These gradually coalesce
• immune defects and become confluent (Fig. 46.20). Lesions may spread to
• immunosuppressive therapy the throat, leading to serious dysphagia.
SECTION 8: FUNGAL SKIN

• iron and zinc deficiency. This type of oral candidosis is common in infants with
Newborn infants, especially premature infants, are very an incidence of between 4% and 8%. It is also the form
susceptible to Candida infection, probably reflecting their seen in patients who are immunocompromised, such as
INFECTIONS

immature mucosal immunity [12]. The occlusive effects


of  napkins are a cause of candidosis in babies [13], and
thumb sucking may lead to maceration of the nailfolds
and subsequent infection by Candida.
Disturbance of the normal bacterial flora by the admin­
istration of antibiotics facilitates the growth of Candida
spp. Endocrine disorders such as diabetes mellitus,
Cushing syndrome, hypoparathyroidism, hypothyroid­
ism and autoimmune polyendocrinopathy syndromes
(such as APECED) favour the development of candidosis.
Immune defects, particularly in T‐lymphocyte function,
predispose to superficial forms of candidosis, and chronic
mucocutaneous candidosis is often associated with
defects in cell‐mediated immunity [12]. In patients with
neutropenia, the development of Candida infections
reflects the inability to eliminate yeasts that have already
penetrated the skin or mucous membranes.

Pathology Successful colonization and infection by Candida


begins with the adherence of commensal organisms to
mucosal cells or keratinocytes. Adherence must take place
before the yeasts can germinate to produce germ tubes and
hyphal elements, which can penetrate host tissues. Fig. 46.20  Oral candidosis with lesions extending on the tongue surface.
Chapter 46  Superficial Fungal Infections 547

those with AIDS. The cumulative prevalence of oral


Candida infection was found to be 72% of 99 children with
perinatally acquired human immunodeficiency virus
(HIV) infection examined longitudinally for oral lesions
[15]. HIV‐positive children show the same range of
Candida spp. reported for adults who are HIV positive,
and C. dubliniensis has been isolated in several studies
[16,17]. Resistance to azole antifungals has also been dem­
onstrated in clinical C. albicans isolates from HIV‐infected
children [18,19].

Acute erythematous atrophic candidosis.  This entity is


characterized by small or large erythematous and atrophic
areas. Any part of the oral mucosa may be affected,
although the most common site is the tongue. This is a rare
condition and is usually associated with broad‐spectrum
antibiotic treatment [20] but may also develop in HIV‐
Fig. 46.21  Napkin candidosis.
positive patients.

Chronic erythematous candidosis. This is the most com­ discoloured brown or green and is often ridged. The nail
mon form of oral candidosis, usually associated with oral becomes friable and detached from the nail bed.
prostheses such as those used in orthodontics. Presenting Paronychial infection can occur in infants with a long
signs are persistent erythema and oedema of the portion of history of finger sucking, leading to a breakdown of the
the palate that is in contact with dentures. It is frequently cuticle and invasion of Candida. The fingers become red
accompanied by angular cheilitis, but this condition can with proximal and lateral nailfold swelling [25]. Candida
develop in association with any form of oral candidosis. onychomycosis has also been reported in a 5‐day‐old
neonate [26], and also at 5 weeks in a preterm infant
Chronic hyperplastic candidosis (Candida leucoplakia). with suspected C. albicans sepsis [27]. In both cases, the
This presents with lesions ranging from small translucent mothers had vaginal candidosis before delivery.
white areas to large dense opaque plaques, which cannot
be easily detached from the affected areas of the mouth or Congenital cutaneous candidosis
tongue. This condition is seen mainly in adults and is This is a rare condition in newborn infants whose mother
associated with smoking. However, hyperplastic nodules, had an intrauterine Candida infection prior to delivery.
particularly on the tongue, are often found in children The disease is characterized by multiple papules on an

SECTION 8: FUNGAL SKIN


with chronic mucocutaneous candidosis (CMC) (see erythematous base. The lesions occur on the face, neck,
Chronic mucocutaneous candidosis). trunk and limbs. This form of candidosis is believed to
occur after yeasts ascend from the vagina and penetrate

INFECTIONS
Napkin (diaper, nappy) dermatitis the amniotic membranes [28]. Odds [7] found that in 40%
Napkin dermatitis leads to approximately 20% of all of 54 cases, the pregnancy was associated with a foreign
childhood dermatology visits. Common causes include body in the uterus, either a contraceptive device or cervi­
allergic contact dermatitis, irritant contact dermatitis, cal suture. Inflammation of the fetal membranes may be
infection and psoriasis [21]. Rashes due to Candida on the present at birth with focal yellow lesions containing
buttock and in the perianal area of babies, associated with yeasts on the umbilical cord and placenta [29]. Full‐term
wearing napkins, present with erythema, scaling and infants with congenital cutaneous candidosis generally
characteristic satellite pustules (Fig. 46.21). exhibit a benign course and respond well to topical therapy;
The relationship between the presence of cutaneous some may even clear spontaneously. However, in prema­
candidosis in infants and the faecal carriage of Candida ture and low‐birthweight infants this is a serious infection,
has been frequently recorded [13,21,22]. The precise role frequently complicated by systemic involvement [30].
of Candida in napkin rash is still unclear. Candida as a pri­ Inborn errors of caspase recruitment domain‐containing
mary invader of an irritant dermatitis has been favoured protein 9 (CARD9) immunity are predisposing factors for
by some investigators [23], whereas others have found invasive candidosis [12,31].
that C. albicans was a common secondary invader of many
types of napkin dermatitis [24]. (See also Chapter 20.) Chronic mucocutaneous candidosis
Chronic mucocutaneous candidiasis (CMC) is character­
Paronychia and onychia ized by recurrent or persistent infections of the skin, nails,
These are acute or chronic infections of the nailfolds and oral and genital mucosae with Candida spp., mainly
nails caused by Candida spp. The nailfold becomes swol­ C. albicans. In recent years, inborn errors of human IL‐17
len, erythematous and painful with a discharge of pus. immunity have been demonstrated to underlie primary
Nail plate invasion may follow, infection beginning in immunodeficiencies with CMC. Various defects in recep­
the proximal portion of the nail. The nail plate becomes tors responsible for sensing of C. albicans or downstream
548 Section 8  Fungal Skin Infections

signalling to IL‐17 may lead to susceptibility to Candida Other infections.  Other chronic skin infections such as
infection. While CMC is common in patients with pro­ warts and dermatophytosis can occur in patients with
found T‐cell immunodeficiencies, CMC can also form CMC [33].
part of other immunodeficiencies in syndromic CMC, or
occur as a separate entity [12,31,32]. Prognosis. Prognosis of candidosis depends largely on
the type and severity of the predisposing factors or asso­
Oral lesions.  These are usually of the pseudomembra­ ciated disease. Oral and congenital cutaneous candidosis
nous type but may develop into chronic hyperplastic in the newborn may clear uneventfully but other forms,
lesions that present with white, adherent plaques affect­ for example CMC, are difficult to treat and will not clear
ing any mucosal surface, including the lateral borders of spontaneously.
the tongue. Angular cheilitis may be present. Oral lesions
occur in more than 90% of all CMC patients [7]. Diagnosis. In all cases of suspected candidosis, micro­
scopic and culture confirmation is necessary. Smears from
Skin lesions.  These progress from erythema to hyper­ mucosal surfaces may be examined as unstained wet
keratosis and crusted plaques (candida granuloma) preparations in saline or KOH. Heat‐fixed preparations
(Fig. 46.22). Granulomatous lesions predominate on the may be stained by Gram (Fig. 46.24) or by periodic acid–
scalp and face but can also occur on other parts of the Schiff (PAS) stain. Skin and nail specimens should be
body. placed in 30% KOH under a coverslip. Softening of the
tissue can be hastened by gentle heating. Specimens
Nails.  Fingernails become affected but rarely toenails should be examined for the presence of round or oval
(Fig. 46.23). Nail involvement is characterized by severe budding yeast cells of 2–4 µm in size. Long hyphae with
dystrophic changes with thickening, distortion and frag­ yeasts at points of constriction are also present (Fig. 46.25).
mentation of the nail plate. Material should be inoculated onto SDA. Chloram­
phenicol may be incorporated to reduce bacterial contam­
ination, but cycloheximide should be omitted as many
yeast species are sensitive to this antibiotic. Incubation is
at 37 °C for 2–5 days and plates are discarded as negative
SECTION 8: FUNGAL SKIN
INFECTIONS

Fig. 46.24  Smear showing hyphae and yeasts (Gram stain).


Fig. 46.22  Candida granuloma.

Fig. 46.23  Chronic mucocutaneous candidosis affecting a thumb nail. Fig. 46.25  Hyphae and yeasts in skin (30% KOH).
Chapter 46  Superficial Fungal Infections 549

if no growth appears after 7 days. Colonies should be


Box 46.4  General recommendations for the treatment of candidosis
examined by microscopy for typical budding yeast cells.
Selective and chromogenic media for the isolation of • Topical therapy (polyenes = nystatin, amphotericin B; azoles;
yeasts and their identification are now available com­ amorolfine) in a suitable formulation is usually sufficient.
mercially. They include CandiSelect™ 4 (Bio‐Rad, USA), • Griseofulvin is not effective.
an isolation medium on which C. albicans appear pink to • Oral candidosis: nystatin or amphotericin B oral solution.
purple and other yeasts turquoise, and BBL CHROMagar™ • Cutaneous candidosis: above‐mentioned antifungals in a cream or
Candida (Becton Dickinson, Europe; Mast Diagnostics, paste formulation.
UK), an isolation medium on which colonies of C. albicans • Chronic mucocutaneous candidosis: oral azole antifungals
according to sensitivity testing.
appear green, C. tropicalis blue, C. krusei pink and others
white to pink.
Tests for the identification of Candida spp. involve the
investigation of isolates for their ability to ferment sugars
and also to assimilate various sources of carbohydrates Oral candidosis
and nitrates. Commercial identification kits are available Oral candidosis in infants generally responds quickly to
and include API™ 20C Aux and ID 32C (both BioMerieux, topical treatment. Nystatin oral suspension (100 000 U/mL)
USA) and AuxaColor 2™ (Bio‐Rad, France). or amphotericin oral suspension (100 mg/mL) should
be  given at 4–6 hourly intervals. In older children, the
Molecular diagnostics dose of amphotericin and nystatin oral suspension can
For the identification of Candida spp. directly from clinical be increased to 1 mL at 6 hourly intervals for 2–3 weeks.
specimens, the same methods are used as for dermato­ Nystatin and amphotericin B lozenges can also be used.
phyte fungi (real‐time or conventional PCR in combina­ These compounds have a bitter taste and miconazole oral
tion with sequencing or hybridization to species‐specific gel is sometimes better tolerated. In children under 2
probes). In addition, fluorescence in situ hybridization years, 2.5 mL at 12 hourly intervals should be given, chil­
and nucleic acid sequence‐based amplification of dren between 2 and 6 years of age should have 5 mL at 12
extracted yeast cell RNA combined with species‐specific hourly intervals and those over 6 years 5 mL at 6 hourly
probes have been applied to detect Candida yeast cells intervals. The gel should be retained in the mouth for as
mainly from blood or blood cultures (for review see [34]). long as possible and treatment should be continued for at
As target genes for the differentiation of Candida yeasts, least 48 hours after all symptoms have disappeared.
the 26S and the ITS 1/2 region of the rDNA have been Oral candidosis occurring in immunosuppressed chil­
established. Even phylogenetically closely related spe­ dren may require systemic therapy, such as itraconazole
cies, e.g. C. dubliniensis and C. albicans, are delineated by solution or fluconazole or the newer azoles posaconazole
these targets. Commercially available tests are designed and voriconazole, and prophylaxis should be introduced,
to identify Candida spp. from systemic infections (mainly for example in children with acute lymphocytic leukae­
blood).

SECTION 8: FUNGAL SKIN


mia [35].

Differential diagnosis. Oral candidosis should be Cutaneous candidosis


­distinguished from lichen planus, leucoplakia, herpes

INFECTIONS
Most infections respond to topical nystatin, azoles or
stomatitis, aphthous stomatitis and erythema multiforme. amorolfine. Napkin dermatitis associated with Candida
Napkin dermatitis caused by C. albicans may resemble other infection may be treated with low‐potency corticosteroid
forms of dermatitis and psoriasis [21]. Staphylococcal preparations (1% hydrocortisone cream) in addition to a
and Gram‐negative nailfold infections are similar to topical antifungal [36]. Congenital cutaneous candidosis
Candida paronychial infections, and Candida nail infec­ responds well to topical antifungals such as nystatin or an
tions should be differentiated from dermatophytosis, azole except in infants who are premature or low birth­
psoriasis and eczema. weight who typically require systemic therapy.
Congenital cutaneous candidosis may resemble neona­
tal bacterial infections (staphylococcal infections), tran­ Paronychia and onychia
sient neonatal pustulosis and congenital syphilis. Topical application of an azole cream or solution to the
nailfold may be helpful; control of the predisposing fac­
Treatment. The majority of the antifungal drugs tor, such as finger sucking, may lead to a spontaneous
described for dermatophytosis are also effective for the cure [37]. Nail infections occurring in infants have also
treatment of most superficial forms of candidosis. been reported as recovering without therapy.
However, in addition, the polyenes, complex macrolide
substances derived from Streptomyces spp. that act by Chronic mucocutaneous candidosis
selectively binding to the fungal cell membrane, are Oral and cutaneous lesions will respond to short courses
useful topically for oral and vaginal candidosis. The of antifungal therapy, but longer courses are needed to
main polyene drugs are amphotericin B, nystatin and clear nail infections. Improvement is often transient and
natamycin. Resistance to these compounds is rare and recurrence is common. Therapy with itraconazole and
toxicity, a problem with parenteral use, is not seen with fluconazole may be helpful. Especially in systemic disease,
local application. See Box 46.4. the newly developed azoles (voriconazole, posaconazole)
550 Section 8  Fungal Skin Infections

or the echinocandins (caspofungin, anidulafungin, 28 Chapel TA, Gagliardi C, Nichols W. Congenital cutaneous candidosis.
J Am Acad Dermatol 1982;6:926–8.
micafungin) offer some advantages as they have a broader
29 Whyte RK, de Hussain Z, Sa D. Antenatal infections with Candida spe­
spectrum and a higher sensitivity against otherwise cies. Arch Dis Child 1982;57:528–35.
resistant pathogens [35]. 30 Darmstadt GL, Dinulos JG, Miller Z. Congenital cutaneous candidia­
sis: clinical presentation, pathogenesis, and management guidelines.
Pediatrics 2000;105:438–44.
References 31 Lanternier F, Cypowyj S, Picard C et al. Primary immunodeficiencies
1 Veron S. Memoire sur le muguet. Arch Gen Med 1835;8:466. underlying fungal infections. Curr Opin Pediatr 2013;25:736–47.
2 Langenbeck B. Auffindung von Pilzen aus der Schleimhaut der 32 Sparber F, LeibundGut‐Landmann S. Interleukin 17‐mediated host
Speiseröhre einer Typhus‐Leiche. Neue Not Geb Natur‐U Helik defense against Candida albicans. Pathogens 2015;4:606–19.
(Froriep) 1839;12:145–7. 33 Shama SK, Kirkpatrick CH. Dermatophytoses in patients with chronic
3 Bennett JH. On the parasitic vegetable structures found growing in mucocutaneous candidiasis. J Am Acad Dermatol 1980;2:285–94.
living animals. Trans Roy Soc Edin 1844;15:277–94. 34 Trtkova J, Raclavsky V. Molecular‐genetic approaches to identifica­
4 Zopf W. Die Pilze in morphologischer, physiologischer, biologischer tion and typing of pathogenic Candida yeasts. Biomed Pap Med Fac
und systematischer Richtung. Breslau: Trewendt, 1890. Univ Palacky Olomouc Czech Repub 2006;150:51–61.
5 Berkhout CM. De Schimmelgeschlachten Monilia, Oidium oospora 35 Koehler P, Cornely OA. Contemporary strategies in the prevention
en Torula. Dissertation. University of Utrecht, 1923. and management of fungal infections. Infect Dis Clin North Am
6 Al‐Ahmad A, Auschill TM, Dakhel R et  al. Prevalence of Candida 2016;30:265–75.
albicans and Candida dubliniensis in caries‐free and caries‐active 36 Cohen‐Wolkowiez M, Moran C, Benjamin DK Jr, Smith PB.
children in relation to the oral microbiota – a clinical study. Clin Oral Pediatric antifungal agents. Curr Opin Infect Dis 2009;22:553–8.
Investig 2016;20:1963–71. 37 Ameen M, Lear JT, Madan V et  al. British Association of
7 Odds FC. Candida and Candidosis, 2nd edn. London: Baillière Dermatologists’ guidelines for the management of onychomycosis
Tindall, 1988. 2014. Br J Dermatol 2014;171:937–58.
8 Kashbur IM, Ayliffe GAJ, George RH. The survival of Candida albicans
in moist and dry environments. J Hosp Infect 1980;1:349–51.
9 Polke M, Hube B, Jacobsen ID. Candida survival strategies. Adv Malassezia‐associated diseases
Appl Microbiol 2015;91:139–235.
10 Saiman L, Ludington E, Dawson JD et al. The National Epidemiology The genus Malassezia currently comprises 17 species of
of Mycoses Study Group. Risk factors for Candida species colonisa­
tion of neonatal intensive care unit patients. Pediatr Infect Dis J
lipophilic yeasts (M. pachydermatis, M. furfur, M. yama-
2001;20:1119–24. toensis, M. sympodialis, M. caprae, M. dermatis, M. slooffiae,
11 Gagneur A, Sizun J, Vernotte E et al. Low rate of Candida parapsilosis‐ M. japonica, M. nana, M. equina, M. obtusa, M. restricta,
related colonisation and infection in hospitalised pre‐term infants: a M.  globosa, M. cuniculi, M. brasiliensis, M. psittaci, M.
one‐year prospective study. J Hosp Infect 2001;48:193–7.
12 Netea MG, Joosten LA, van der Meer JW et  al. Immune defence arunalokei). These species belong to the resident flora of
against Candida fungal infections. Nat Rev Immunol 2015;15:630–42. human and animal skin; they are also associated with
13 Dixon PN, Warin RP, English MP. Alimentary Candida albicans and common skin diseases, but their pathogenesis is still not
napkin rashes. Br J Dermatol 1972;86:458–62.
fully elucidated (for review see [1,2]).
14 Lehner T. Classification and clinico‐pathological features of Candida
infections in the mouth. In: Winner H, Hurley R (eds) Symposium on
Candida Infections. Edinburgh: Livingstone, 1966:119–37.
15 Katz MH, Mastrucci MT, Leggott PJ et al. Prognostic significance of Pityriasis versicolor
oral lesions in children with perinatally acquired human immunode­
SECTION 8: FUNGAL SKIN

ficiency virus infections. Am J Dis Child 1993;147:45–8. Definition. Pityriasis versicolor is a chronic, mild and
16 Jabra‐Rizk MA, Falkler WA, Enwonwu CO. Prevalence of yeast
among children in Nigeria and the United States. Oral Microbiol
usually asymptomatic infection of the stratum corneum.
It is produced by the proliferation of lipid‐dependent
INFECTIONS

Immunol 2001;16:383–5.
17 Brown DM, Jabra‐Rizk MA, Falkler WA et al. Identification of Candida yeasts of the genus Malassezia, which are part of the nor­
dubliniensis in a study of HIV‐seropositive pediatric dental patients. mal flora of human skin. The infection is characterized by
Pediatr Dent 2000;22:234–8.
18 Muller FM, Weig M, Peter J et al. Azole cross‐resistance to ketocona­ the development of erythematous scaly macules in
zole, fluconazole, itraconazole and voriconazole in clinical Candida sebum‐rich areas of the skin, especially on the trunk, arms
albicans isolates from HIV‐infected children with oropharyngeal and neck. Occasionally, the face and groin are involved.
candidosis. J Antimicrob Chemother 2000;46:338–40.
The lesions may be followed by long‐lasting depigmenta­
19 Pelletier R, Peter J, Gonzalez C et  al. Emergence of resistance of
Candida albicans to clotrimazole in human immunodeficiency virus‐ tion known as pityriasis versicolor alba (PVa). The disease
infected children: in vitro and clinical correlations. J Clin Microbiol is distributed worldwide but is more prevalent in tropical
2000;38:1563–8. areas, where extensive lesions are common. It can occur at
20 Lehner T, Ward RG. Iatrogenic oral candidosis. Br J Dermatol
1970;83:161–6.
any age but is most commonly seen in young adults and
21 Klunk C, Domingues E, Wiss K. An update on diaper dermatitis. Clin is comparatively rare in children.
Dermatol 2014;32:477–87. The term ‘tinea versicolor’ is inaccurate and should not
22 Rebora A, Leyden JJ. Napkin (diaper) dermatitis and gastrointestinal be used as the causative agent is not a dermatophyte.
carriage of Candida albicans. Br J Dermatol 1981;105:551–5.
23 Leyden JJ, Kligman AM. The role of microorganisms in diaper derma­
titis. Arch Dermatol 1978;114:56–9. History. The disease was first recorded by Eichstedt [3] in
24 Dixon PN, Warin RP, English MP. The role of Candida albicans infection 1846 and also described by Sluyter [4] in 1847. Both
in napkin rashes. BMJ 1969;2:23–7.
25 Raimer SS, Petrusick TW. Superficial fungal infections in children.
authors called the infection ‘pityriasis versicolor’ but did
Dermatol Clin 1984;2:57–65. not propose a name for the causative organism. In 1853,
26 Plantin P, Jouan N, Calligaris C et al. Onychomycosis in a newborn Robin [5] named the fungus Microsporum furfur and called
due to Candida albicans with neonatal infection. Ann Dermatol the disease ‘tinea versicolor’ because he thought the fun­
Vénéréol 1992;119:213–15.
27 Abraham Z, Sujov P, Blazer S et al. Candida onychomycosis in a pre­ gus resembled the dermatophyte Microsporum audouinii.
term infant. Mykosen 1986;29:357–9. Malassez [6], in 1874, described yeast‐like cells from scalp
Chapter 46  Superficial Fungal Infections 551

lesions and in dandruff and called them ‘spores’. In Reports on the distribution of the newly recognized
1889, Baillon [7] created a new genus, Malassezia, for species suggest that M. globosa and M. restricta [21–23] are
these organisms. the most common species on normal adult skin. However,
The fastidious nutritional requirements of these lipid‐ these studies are flawed because the majority relied on
dependent yeasts made their isolation difficult, and some swabbing for sampling, and this method is unlikely to
of the early reports of their culture may be suspect. yield accurate quantitative data. Sites examined and
Castellani and Chalmers [8] cultured a yeast from human media used varied as well. This may also explain the
skin in 1913, which they named Pityrosporum ovale, fol­ different results obtained concerning the colonization of
lowing the lead of Sabouraud [9] who, in 1904, assigned children by Malassezia yeasts. Studies based on molecular
the yeasts seen in dandruff and sebaceous dermatitis to a techniques showed that M. restricta overwhelmingly pre­
new genus, Pityrosporum, as P. malassezii. In the same year, dominated at ages over 16–18 years in males and 23–29
Kraus [10], using a medium supplemented with lanolin, years in females. M. globosa and M. restricta together
also reported in vitro isolation of the yeasts, although accounted for more than 70% of Malassezia spp. regardless
the  lipophilic nature of Malassezia was first described of gender [24].
by Benham [11] only in 1939. The isolation of P. ovale by The incidence of Malassezia on clinically normal
Panja using Petroff’s egg medium is also widely regarded skin  from the back of newborn infants and those aged
as genuine [12]. In 1951, Gordon [13] isolated a round, 6 months, 1 year, 5 years, 10 years and 15 years was inves­
double‐contoured, lipophilic yeast‐like fungus and called tigated by Faergemann and Fredriksson [25]. They were
it Pityrosporum orbiculare, suggesting that it might be unable to demonstrate M. furfur in children less than
the causative fungus of pityriasis versicolor. Subsequent 1 year of age. In contrast, several later studies suggest that
studies by Keddie and co‐workers [14,15] demonstrated neonatal and infant carriage of Malassezia yeasts is not
both morphological and antigenic relationships between unusual. In a report from Thailand, Malassezia yeasts were
P. orbiculare cultured from infected scales and the M. furfur isolated from the normal skin of 47% of 150 1‐ to 5‐day‐
cells found in skin scales. old infants [26], and 37% of infants hospitalized in inten­
For a number of years there was controversy between sive care units in the USA were found to harbour Malassezia
those who believed that P. ovale and P. orbiculare repre­ on the skin [27]. A study of 245 neonates and 42 infants in
sented two distinct species and those who believed that a neonatal and medical unit demonstrated that 41 out of
the oval and round cells were aspects of a single species. the 42 infants were colonized with Malassezia yeasts on
This was apparently resolved in 1984 when Yarrow and admission. None of the neonates was colonized immedi­
Ahearn [16] placed both of these lipophilic organisms into ately after birth, but 78 (31.8%) became colonized during
a single species. As the generic name Malassezia given the study. In both infants and neonates, the ear was the
by Baillon had priority over Pityrosporum given by most common site of colonization [28].
Sabouraud, the organism was named Malassezia furfur, With respect to older children, a survey on the scalp
and this applied to both yeast and hyphal phases of carriage of Malassezia species in healthy schoolchildren

SECTION 8: FUNGAL SKIN


growth. Subsequently, Simmons and Gueho described a aged between 7 and 17 years showed that prepubertal
second lipid‐dependent species which was a normal resi­ children yielded yeasts less often than older or sexually
dent on human skin, Malassezia sympodialis [17]. The mature children [29]. This study of nearly 600 children

INFECTIONS
situation was further complicated when a detailed study also found that Caucasians harboured yeasts more often
using morphological, physiological and molecular tech­ than black children. In a study that examined skin from
niques showed that the yeasts classified as M. furfur actu­ the forehead of healthy children, 119 out of 138 children
ally comprised a number of different species and a further (87%) gave positive cultures. Children producing the
four species  –  M. globosa, M. slooffiae, M. restricta and highest number of colonies were those aged between 2
M. obtusa  –  were added to the genus [18]. By means of and 23 months and those over 9 years old [30]. In the
conventional and molecular methods, today 17 species of study of children comparing 5‐, 10‐ and 15‐year‐olds by
lipophilic yeasts (M. pachydermatis, M. furfur, M. yama- Faergemann and Fredrikson [25], the highest prevalence
toensis, M. sympodialis, M. caprae, M. dermatis, M. slooffiae, (93%) was in 15‐year‐old children.
M. japonica, M. nana, M. equina, M. obtusa, M. restricta, M. A number of factors influencing the appearance of pity­
globosa, M. cuniculi, M. brasiliensis, M. psittaci, M. arunalokei) riasis versicolor have been described [31,32]. It is well
are distinguished (for review, see [1,2]). known that the incidence of the disease is higher in warm
and humid climates, and sweating may be one factor
Aetiology and pathogenesis. The recognition of new spe­ associated with increased growth of Malassezia, leading to
cies of Malassezia makes many of the preceding studies of signs and symptoms of infection. Systemic corticosteroid
the genus difficult or even impossible to interpret, as the therapy is related to an increased incidence of pityriasis
yeast strains used have not been maintained in culture and versicolor [19]. It is also associated with Cushing syn­
cannot be related to the new species. The yeasts are part of drome [33] and immunosuppression associated with
the normal flora of the skin. Early studies found that 97% of transplantation but not with AIDS. To some extent,
clinically normal people may carry the yeast on the scalp genetic factors may play a role, as the disease occurs more
and 92% on the trunk [19,20]. The oval form was more frequently among first‐degree family members [34].
common on the scalp (‘M. ovale’), whereas the spherical However, the pathogenesis of pityriasis versicolor is not
forms were more frequent on the trunk (‘M. orbiculare’). clearly understood. Being most frequently isolated from
552 Section 8  Fungal Skin Infections

the lesions, M. globosa is discussed as the causative agent hyperkeratosis may be present but there are few or no
of pityriasis versicolor but the clinical phenomena, espe­ signs of inflammation (‘seemingly normal skin’).
cially fluorescence and change in skin colour, are not
explained. M. furfur has been shown to produce a great Clinical features. Pityriasis versicolor classically presents
number of indole pigments and fluorochromes when as asymptomatic scaly hypo‐ or hyperpigmented macular
grown with tryptophan (Trp) as sole nitrogen source, lesions which, in adults, mainly occur on the upper trunk
which may explain several clinical features of pityriasis and arms. The macules frequently coalesce, producing
versicolor [2]. In the plant pathogen Ustilago maydis, a patches of varying shapes and sizes. The colour of the
fungus phylogenetically closely related to Malassezia spp., lesions varies: in pale‐skinned individuals, the lesions
it was recently shown that the biosynthetic pathway lead­ appear fawn or brown‐coloured or occasionally red
ing to pigment production from Trp was based on the (Fig. 46.26), whereas after sun tanning they appear paler
activity of a single enzyme, Tam 1 [35]. It is a Trp ami­ or hypopigmented in comparison with the surrounding
notransferase that converts Trp to indolepyruvate (IP). tanned skin. In dark‐skinned subjects, the rash is usually
Furthermore, it was found that the indole pigments can hypopigmented (Fig. 46.27) but may be hyperpigmented.
develop spontaneously from IP and Trp without the These pigmentary changes are thought to be due to the
action of any additional enzymes. Thus, synthesis of a inhibition of melanin formation by dicarboxylic acids,
multitude of pigments produced from Trp is catalysed by such as azelaic acid, produced by the enzyme activity of
a single biosynthetic step, the activity of Tam 1. This sug­ the yeasts. However, given the characteristic occurrence
gests that utilization of spontaneously generated meta­ of pityriasis versicolor only under special conditions such
bolic by‐products might constitute an important step in as high air humidity and excessive sweating and the fact
the pathophysiology of pityriasis versicolor. This was that Malassezia yeasts belong to the normal cutaneous
underlined by a rapid impressive effect of the topically flora, the regularly produced dicarboxylic acids cannot
applied transaminase inhibitor cycloserin on the clinical serve as the sole explanation why pityriasis versicolor
course of pityriasis versicolor [36]. (alba) does not occur whenever the organism is found on
Although M. globosa is nowadays considered the main the skin [45,46].
pathogen in pityriasis versicolor, it is not able to produce Other sites of infection include the face, neck, abdo­
pigment in vitro. Hypothetically, M. globosa might have men and thighs. More extensive lesions occur in tropical
the genetic potency to produce pigment in vivo, but the
prerequisites for induction in vitro remain to be deter­
mined. Investigations based on the genome of M. globosa
might help clarify this discrepancy [37]. The comparative
rarity of pityriasis versicolor in children under 10 years of
age might suggest that physiological changes in skin
lipids during puberty could enhance fungal pathogenic­
SECTION 8: FUNGAL SKIN

ity [19]. Pityriasis versicolor is most commonly seen in


young adults with an almost equal sex distribution.
Although much less common, it does occur in children
INFECTIONS

[38–40]. Of 305 children examined in Poland, 33 (10.8%)


were found to have pityriasis versicolor. Their ages
ranged from under 1 year to 10 years old, with the highest
number affected being between the ages of 8 and 10 years;
two patients were under 1 year of age [38]. A report
from  Tunisia [33] analysed retrospectively the results of
Fig. 46.26  Pityriasis versicolor. Hyperpigmented lesions.
a  5‐year study. Of 1379 cases of pityriasis versicolor,
164 (11.8%) were in children aged 5 months to 14 years. In
children, the face is often involved, in contrast to the rar­
ity with which facial lesions occur in adults [40–42]. In a
series of 57 cases of pityriasis versicolor in children,
Terragni et al. [40] found that the face was the only site of
involvement in 18 patients (31.6%). Similarly, in the
Tunisian study, of the 164 children diagnosed, 78 (47.5%)
had facial lesions. Among children in the tropics, pityria­
sis versicolor is not an uncommon disease. There is a sud­
den resurgence of cases in the hot monsoons and even
infants are not spared [43]. The infection has also been
recorded in neonates [44].

Pathology. The yeasts and hyphae are restricted to the


outermost layers of the stratum corneum, where hyphae
invade both between and inside corneocytes. Mild Fig. 46.27  Pityriasis versicolor. Hypopigmented lesions.
Chapter 46  Superficial Fungal Infections 553

climates, covering many sites on the body surface. erythrasma (generally restricted to intertriginous areas)
Occasionally slight irritation may be noted by patients, and pinta also need to be considered.
but medical attention is generally sought for cosmetic rea­
sons. Examination of lesions under a Wood’s light reveals Molecular diagnostics
a golden‐yellow fluorescence in the majority of cases and The 17 species described currently within the genus
may reveal the presence of lesions not obviously visible to Malassezia are differentiated by sequence analysis of the
the naked eye. This fluorescence might be explained by D1/D2 domain of the 26S and the ITS 1/2 region of the
the Trp‐derived compound pityrialactone [2]. rDNA. Several conventional and real‐time ‘in‐house’ PCRs
have been developed targeting these genes to identify
Diagnosis. Pityriasis versicolor is easily diagnosed by Malassezia spp., even quantitatively [1,2]. For routine
microscopic examination of skin scales mounted in 30% diagnostics, however, the delineation of the animal‐
KOH. Both yeasts and hyphae fluoresce very strongly associated species M. pachydermatis (source of infection)
with the calcofluor technique (‘spaghetti and meatballs’; from the remaining species is necessary.
Fig. 46.28). It is the hyphae that are diagnostic of pityriasis
versicolor. In the majority of instances, the yeasts associ­ Treatment. Treatment is usually successful with either
ated with the hyphae are round, budding on a narrow topical or systemic antimycotic therapy within 2 weeks or
base, with morphology typical of M. globosa, and this less, but widespread disease and frequent relapses may
yeast is widely accepted as the main species associated be of some concern [49]. For example, the condition can
with pityriasis versicolor [21–23]. However, occasionally, be treated effectively with 2.5% selenium sulfide suspen­
and more frequently in the tropics, oval yeasts are seen sion as a shampoo applied to the skin, left on overnight
with the hyphae, suggesting that more than one species of and washed off the next morning. One or two applica­
Malassezia may be associated with the disease [47]. Culture tions are usually sufficient. It also responds to most
is not helpful as the organisms are part of the normal skin topical imidazoles (such as clotrimazole, miconazole,
flora. If required, Malassezia yeasts can be grown on a econazole) for 1–2 weeks as well as ketoconazole sham­
medium enriched with lipids, such as Dixon’s agar or poo (two or three applications).
Leeming’s medium [1,2]. Incubation should be at 32–35 °C It should be noted that, as Malassezia yeasts are part of
for 2 weeks. Hypopigmented lesions may be confused the normal flora of the skin, relapses occur very fre­
with vitiligo but there is no scaling in vitiligo and the quently. Any skin discolouration may take several months
lesions are white and usually symmetrical. In contrast to to revert to normal, and patients should be reassured that
vitiligo, there is no sunburn after exposure to UV light, this is normal and that further treatment is not required.
which might be explained by the action of the UV filter
pityriacitrin [48]. Other Malassezia (Pityrosporum) infections
Although the trunk may be affected, other sites Malassezia (Pityrosporum) folliculitis
involved include wrists, ankles, knees and hands. Other This condition is seen primarily in teenagers or young

SECTION 8: FUNGAL SKIN


pigmentary disorders, such as chloasma, in which hyper­ adult males. Lesions consist of papules or pustules, often
pigmented macules without scaling affect the face but not widely scattered on the shoulders and back [50,51]
the trunk or limbs, should also be excluded. Pityriasis (Fig. 46.29). Numerous, often round, budding Malassezia

INFECTIONS
rosea, like pityriasis versicolor, has a predilection for the yeasts are found in affected follicles, surrounded by
trunk but is more acute and rapidly spreading after the inflammatory cells, suggesting that M. globosa may again
appearance of a herald patch. Seborrhoeic dermatitis, be of significance in many instances [21]. Some workers
have suggested that follicular occlusion may be the primary
event in the development of folliculitis and that over­
growth of Malassezia is a secondary occurrence [52]. It is

Fig. 46.28  Thick‐walled yeasts and short angular hyphae in skin


(‘spaghetti and meatballs’; calcofluor preparation). Fig. 46.29  Pityrosporum folliculitis.
554 Section 8  Fungal Skin Infections

not associated with a single Malassezia species [53]. It has homologous human self‐antigens [66]. The human
to be distinguished from acne as it does not respond to proteins are capable of inducing positive skinprick and
the same treatment. Oral ketoconazole or ketoconazole atopy patch tests in patients sensitized to the Malassezia
shampoo is the most effective treatment. proteins, indicating a role of IgE‐mediated autoreactivity
in the pathogenesis of AD in a subset of patients. A com­
Malassezia (Pityrosporum) yeasts in seborrhoeic parative analysis of Malassezia flora between adults and
dermatitis children with AD showed that M. restricta was the pre­
In adults, Malassezia yeasts are found in large numbers dominant species in the children with AD, while both
in the scales of seborrhoeic dermatitis and dandruff. M. restricta and M. globosa predominated in the adults.
Seborrhoeic dermatitis is characterized by yellowish‐red The adults showed increased sensitization in terms of
lesions in seborrhoeic areas covered with greasy scales anti‐Malassezia‐specific IgE responses in the sera to
and associated with mild itching. The relationship both M. globosa and M. restricta in comparison with the
between infantile seborrhoeic dermatitis and these yeasts children [67]. In the study by Lange et al. sensitization of
is less well established. The finding of oval yeasts in infan­ 141 children with AD against a mixture of three Malassezia.
tile seborrhoeic dermatitis was first reported by Devred spp. (M. sympodialis, M. globosa and M. restricta; Pharmacia
et al. [54] in 1985. Ruiz‐Maldonado et al. [55] found P. ovale Immunocap m227) showed no significant correlation to
in 73% of infants with seborrhoeic dermatitis and in 53% age, duration or onset of the disease, but a higher risk for
of healthy infants, whereas Broberg and Faergemann [56] sensitization was found with increasing total IgE levels
cultured P. ovale significantly more often from children with [68]. Malassezia yeasts seem to be associated mainly with
seborrhoeic dermatitis (90%) than healthy children (20%). the head, neck and face type of atopic eczema/dermatitis
Since the recognition of the new species of Malassezia, a syndrome (AEDS) (head and neck dermatitis), corre­
study in Japan compared 52 patients with infantile sebor­ sponding to the numerous sebaceous glands found in
rhoeic dermatitis and 47 healthy 1‐month‐old infants. this  region. This is supported by the demonstration of
Direct counts of yeasts in skin samples from the cheek Malassezia‐specific IgE antibodies, skinprick testing and
found significantly more yeasts in the seborrhoeic derma­ atopy patch test, but in particular by the results of clinical
titis group. Cultures from the other cheek also yielded studies in which antimycotics were applied topically or
M. furfur and M. globosa at significantly higher rates than systemically for treatment of head and neck dermatitis
the seborrhoeic dermatitis group [57]. However, although [65]. It remains to be clarified whether these results are
it appears that these Malassezia yeasts are found in the also applicable to children with AD.
skin of infantile seborrhoeic dermatitis sufferers more fre­
quently than in healthy children, the importance of these Malassezia fungaemia and invasive infection
yeasts as pathogens is still unclear. Treatment of infected Malassezia spp. are rare causes of invasive infections in crit­
infants with 2% ketoconazole cream has given good ically low‐birthweight ill infants and in immunocompro­
response rates [55,58] but, as this condition has a good mised children and adults [69,70]. The clinical spectrum
prognosis without treatment, placebo studies are neces­ ranges from asymptomatic infection to life‐threatening
SECTION 8: FUNGAL SKIN

sary before the role of Malassezia in infantile seborrhoeic sepsis and disseminated disease, with intravascular cath­
dermatitis can be established with certainty. eters and administration of lipid‐supplemented parenteral
INFECTIONS

nutrition acting as the main risk factors. The overwhelm­


Neonatal cephalic pustulosis ing majority of invasive infections reported in the literature
Malassezia yeasts have been linked to a nonfollicular pus­ have been associated with M. furfur and M. pachydermatis.
tulosis of the face in neonates, first reported by Aractingi Most patients had a serious underlying disease affecting
et al. [59]. Several subsequent reports have supported the overlapping components of host responses, had indwell­
view that this condition is directly related to the presence ing vascular access, and had received parenteral nutrition
of these yeasts [60–62], and some have reported a clinical containing lipid emulsions.
response to antifungal treatment. However, only a pro­ While invasive infections may occur sporadically, in the
portion of pustule smears in these reports revealed the last decade nosocomial outbreaks of neonatal M. furfur
presence of Malassezia yeasts, and positive cultures also and M. pachydermatis infections have been widely
did not show complete correlation between pustules and reported. As shown by molecular typing methods, infants
isolation of yeasts [63,64]. Considering the now convinc­ become colonized by skin contact with parents or health­
ing evidence that the normal skin of neonates may be rap­ care workers, who may further transmit the organism
idly colonized by Malassezia yeasts in the first weeks of from an infected or colonized infant to others via their
life, more evidence is required to confirm the significance hands [71,72]. Malassezia spp. can be detected in blood
of these yeasts in this condition. and other specimens by direct microscopic examination,
by culture and by molecular methods. Detection is
Atopic dermatitis complicated by the organism’s lipid‐dependent nature as
Malassezia yeasts have been shown to act as allergens in most routinely used media do not support its growth [69].
atopic dermatitis (AD), and 13 allergens have been cloned, Use of lipid‐supplemented media may be warranted in
characterized and produced as recombinant proteins certain specimens, especially if cultures appear sterile
from Malassezia species [65]. Some of these recombinant on routine media and yeasts have been observed on
allergens show a high degree of sequence identity to microscopy.
Chapter 46  Superficial Fungal Infections 555

There is general consensus that the management of 30 Bergbrant IM, Broberg A. Pityrosporum ovale. Culture from the forehead
of healthy children. Acta Dermatol Venereol (Stockh) 1994;74:260–1.
Malassezia‐related fungaemia and sepsis requires the
31 Crespo‐Erchiga V, Florencio VD. Malassezia yeasts and pityriasis ver­
prompt removal of any indwelling catheter in addition sicolor. Curr Opin Infect Dis 2006;19:139–47.
to intravenous antifungal therapy and, less clearly, the 32 Gupta AK, Batra R, Bluhm R et al. Pityriasis versicolor. Dermatol Clin
temporary discontinuation of parenteral nutritional lipid 2003;21:413–42.
33 Canizares O, Shatin H, Kellert AJ. Cushing’s syndrome and dermato­
solutions. mycosis. Arch Dermatol 1959;80:701–12.
34 He SM, Du WD, Yang S et al. The genetic epidemiology of tinea versi­
References color in China. Mycoses 2008;51:55–62.
1 Theelen B, Cafarchia C, Gaitanis G et al. Malassezia ecology, patho- 35 Zuther K, Mayser P, Hettwer U et al. The tryptophan aminotrans-
physiology, and treatment. Med Mycol 2018;56(suppl 1):S10–25. ferase Tam1 catalyses the single biosynthetic step for tryptophan‐
2 Boekhout T, Guého E, Mayser P, Velegraki A. Malassezia and the Skin. dependent pigment synthesis in Ustilago maydis. Mol Microbiol
New York: Springer, 2009. 2008;68:152–72.
3 Eichstedt CF. Pilzbildung in der Pityriasis versicolor. Froriep’s Neue 36 Mayser P, Rieche I. Rapid reversal of hyperpigmentation in pityriasis
Notizen aus dem Gebiete der Natur‐ und Heilkunde 1846;853:270–1. versicolor upon short term topical cycloserine application. Mycoses
4 Sluyter T.De vegetabilibus organismi animalis parasitis ac de novo 2009;52:541–3.
epiphyto in pityriasi versicolore obvio. Inaugural address. Berlin: G. 37 Xu J, Saunders CW, Hu P et  al. Dandruff‐associated Malassezia
Schade, 1847. genomes reveal convergent and divergent virulence traits shared
5 Robin C. Histoire Naturelle des Vegetaux Parasites. Paris: Baillière, with plant and human fungal pathogens. Proc Natl Acad Sci U S A
1853. 2007;104:18730–5.
6 Malassez L. Note sur le champignon de la pelade. Arch Physiol 38 Michalowski R, Rodziewicz H. Pityriasis versicolor in children. Br J
1874;11:203–12. Dermatol 1963;75:397–400.
7 Baillon EH. Traite de Botanique Medicale Cryptogamique. Paris: 39 Miskeen AK, Kelkor SS, Shroff HJ. Pityriasis versicolor in children.
Octave Doin Editeur, 1889:234–5. Indian J Dermatol Venereol Leprol 1984;50:144–6.
8 Castellani A, Chalmers AJ. Manual of Tropical Medicine, 2nd edn. 40 Terragni L, Lasagni A, Oriani A et al. Pityriasis versicolor in the pae­
London: Baillière Tindall and Cox, 1913. diatric age. Pediatr Dermatol 1991;8:9–12.
9 Sabouraud R. Maladies des cuir chevelu. 11 Les maladies desquama­ 41 Bouassida S, Boudaya S, Ghorbel R et al. Pityriasis versicolor in chil­
tives. Paris: Masson, 1904. dren: a retrospective study of 164 cases. Ann Dermatol Venereol
10 Kraus A. Uber das Wesen des sogenannten Unnaschen Flaschenbazillus. 1998;125:581–4.
Arch Dermatol Syphil 1913;106:723–6. 42 Pontasch MJ, Kyanko ME, Brobell RT. Tinea versicolor of the face in
11 Benham RW. The cultural characteristics of Pityrosporum ovale: a lipo­ black children in a temperate region. Cutis 1989;43:81–4.
philic fungus. J Invest Dermatol 1939;2:187–203. 43 Jena DK, Sengupta S, Dwari BC, Ram MK. Pityriasis versicolor in the
12 Panja G. The Malassezia of the skin, their cultivation morphology and pediatric age group. Indian J Dermatol Venereol Leprol 2005;71:259–61.
species [trans.]. 7th Cong Far East Assoc Trop Med 1927;2:442–56. 44 Wyre HW, Johnston WT. Neonatal pityriasis versicolor. Arch Dermatol
13 Gordon M. Lipophilic yeast‐like organisms associated with tinea 1981;117:752–3.
versicolor. J Invest Dermatol 1951;17:267–72. 45 Thoma W, Krämer HJ, Mayser P. Pityriasis versicolor alba. J Eur Acad
14 Keddie LM, Shadomy S. Etiological significance of Pityrosporum Dermatol Venereol 2005;19:147–52.
orbiculare in tinea versicolor. Sabouraudia 1963;3:21–5. 46 Midgley G. The lipophilic yeasts: state of the art and prospects. Med
15 Sternberg TH, Keddie FM. Immunofluorescence studies in tinea Mycol 2000;38(suppl 1):9–16.
versicolor. Arch Dermatol 1961;84:999–1003. 47 Krisanty RI, Bramono K, Made Wisnu I. Identification of Malassezia
16 Yarrow D, Ahearn DG. Genus 7. Malassezia Baillon. In: Kreger‐van Rij species from pityriasis versicolor in Indonesia and its relationship
NJW (ed.) The Yeasts. A Taxonomic Study, 3rd edn. Amsterdam: with clinical characteristics. Mycoses 2009;52:257–62.
Elsevier Science, 1984:882–5. 48 De Almeida Jr H, Mayser P. Absence of sunburn in lesions of pityria­

SECTION 8: FUNGAL SKIN


17 Simmons RB, Gueho E. A new species of Malassezia. Mycol Res sis versicolor alba. Mycoses 2006;49:516.
1990;94:1146–9. 49 Hu SW, Bigby M Pityriasis versicolor: a systematic review of inter-
18 Gueho E, Midgley G, Guillot J. The genus Malassezia with description ventions. Arch Dermatol 2010;146:1132–40.
50 Back O, Faergemann J, Hornqvist R. Pityrosporum folliculitis: a com­

INFECTIONS
of four new species. Antonie Leeuwenhoek 1996;69:337–55.
19 Roberts SOB. Pityriasis: a clinical and mycological investigation. Br J mon disease of the young and middle aged. J Am Acad Dermatol
Dermatol 1969;81:315–26. 1985;12:56–61.
20 Roberts SOB. Pityrosporum orbiculare incidence and distribution on 51 Jacinto‐Jamora S, Tamesis J, Katigbak ML. Pityrosporum folliculitis in
clinically normal skin. Br J Dermatol 1969;81:264–9. the Philippines: diagnosis, prevalence and management. J Am Acad
21 Crespo‐Erchiga V, Ojeda A, Vera A et al. Malassezia globosa as the caus­ Dermatol 1991;24:693–6.
ative agent of pityriasis versicolor. Br J Dermatol 2000;143:799–803. 52 Hill MK, Goodfield MJD, Rodgers FG et  al. Skin surface electron
22 Gupta AK, Kohli Y, Summerbell RC et  al. Quantitative culture of microscopy in Pityrosporum folliculitis. Arch Dermatol 1990;126:181–4.
Malassezia species from different body sites of individuals with or 53 Akaza N, Akamatsu H, Sasaki Y, Malassezia folliculitis is caused by
without dermatoses. Med Mycol 2001;39:243–51. cutaneous resident Malassezia species. Med Mycol 2008;23:1–7.
23 Nakabayashi A, Sei Y, Guillot J. Identification of Malassezia species 54 Devred D, Agache P, Barale T et  al. Pityrosporum et dermite sebor­
isolated from patients with seborrheic dermatitis, atopic dermatitis, rheique du nourrisson. Ann Dermatol Vénéréol 1985;112:211.
pityriasis versicolor and normal subjects. Med Mycol 2000;38:337–41. 55 Ruiz‐Maldonado R, Lopez‐Matinez R, Perez Chavarria EL et  al.
24 Wu G, Zhao H, Li C et  al. Genus‐wide comparative genomics of Pityrosporum ovale in infantile seborrhoeic dermatitis. Pediatr
Malassezia delineates its phylogeny, physiology, and niche adapta- Dermatol 1989;6:16–20.
tion on human skin. PLoS Genet 2015;11:e1005614. 56 Broberg A, Faergemann J. Infantile seborrhoeic dermatitis and
25 Faergemann J, Fredriksson T. Age incidence of Pityrosporum orbiculare Pityrosporum ovale. Br J Dermatol 1989;120:359–62.
on human skin. Acta Dermatol Venereol (Stockh) 1980;60:531–3. 57 Nakabayashi A, Sei Y. Relationship between Malassezia yeast and
26 Thianprasit M, Thagerngpol K. Pityriasis versicolor. In: Gatti F, de infantile seborrhoeic dermatitis. Nippon Ishinkin Gakkai Zasshi
Vroey C (eds) Human Mycoses in Tropical Countries. Bologna: 2001;42:218–20.
Quaderni di Cooperazione Sanitaria, 1991:63–9. 58 Taïeb A, Legrain V, Palmier C et al. Topical ketoconazole for infantile
27 Powell DA, Hayes J, Durrell DE et al. Malassezia furfur skin colo­ seborrhoeic dermatitis. Dermatologica 1990;181:26–32.
nization of infants hospitalized in intensive care units. J Pediatr 59 Aractingi S, Cadranel S, Reygagne P et al. Pustulose neonatale induite
1987;111:217–20. par Malassezia furfur. Ann Dermatol Venereol 1991;118:856–8.
28 Ashbee HR, Leck AK, Puntis JWL et al. Skin colonisation by Malassezia 60 Rapelanoro R, Mortureux P, Couprie B et al. Neonatal Malassezia fur-
in neonates and infants. Infect Cont Hosp Epidemiol 2002;23:212–16. fur pustulosis. Arch Dermatol 1996;132:190–3.
29 Noble WC, Midgley G. Scalp carriage of Pityrosporum species: the 61 Niamba P, Weill FX, Sarlangue J et al. Is common neonatal cephalic
effect of physiological maturity, sex and race. Sabouraudia 1978; pustulosis (neonatal acne) triggered by Malassezia sympodialis? Arch
16:229–32. Dermatol 1998;134:995–8.
556 Section 8  Fungal Skin Infections

62 Bernier V, Weill FX, Hirigoyen V et al. Skin colonisation by Malassezia


species in neonates. A prospective study and relationship with neona­
tal cephalic pustulosis. Arch Dermatol 2002;138:215–18.
63 Bergman JN, Eichenfield LF. Neonatal acne and cephalic pustulosis. Is
Malassezia the whole story? Arch Dermatol 2002;138:255–7.
64 Ayhan M, Sancak B, Karaduman A et al. Colonization of neonate skin
by Malassezia species: relationship with neonatal cephalic pustulosis.
J Am Acad Dermatol 2007;57:1012–18.
65 Darabi K, Hostetler SG, Bechtel MA, Zirwas M.The role of Malassezia
in atopic dermatitis affecting the head and neck of adults. J Am Acad
Dermatol 2009;60:125–36.
66 Schmid‐Grendelmeier P, Scheynius A, Crameri R. The role of sensiti­
zation to Malassezia sympodialis in atopic eczema. Chem Immunol
Allergy 2006;91:98–109.
67 Takahata Y, Sugita T, Kato H et al. Cutaneous Malassezia flora in atopic
dermatitis differs between adults and children. Br J Dermatol
2007;157:1178–82.
68 Lange L, Alter N, Keller T et al. Sensitization to Malassezia in infants
and children with atopic dermatitis: prevalence and clinical character­ Fig. 46.30  White piedra hair showing nodules containing arthroconidia
istics. Allergy 2008;63:486–7. (30% KOH).
69 Marcon MJ, Powell DA. Human infections due to Malassezia spp.
Clin Microbiol Rev 1992;2:101–19.
70 Tragiannidis A, Bisping G, Koehler G, Groll AH. Minireview: nodules of white piedra are seen to be composed of
Malassezia infections in immunocompromised patients. Mycoses hyphae fragmented into rectangular or round arthrospores
2010;53:187–95.
and budding yeasts (Fig.  46.30). Infected hairs can be
71 Chang HJ, Miller H, Watkins N et al. An epidemic of Malassezia pachy-
dermatis in an intensive care nursery associated with colonization of cultured on SDA with chloramphenicol but without
health care workers’ pet dogs. N Engl J Med 1998;338:706–11. cycloheximide; yellowish or cream colonies are produced
72 Chryssanthou E, Broberger U, Petrini B. Malassezia pachydermatis fun­ within 2–3 days.
gaemia in a neonatal intensive care unit. Acta Paediatr 2001;90:323–7.

Black piedra
Less common superficial fungal This is caused by Piedraia hortae, a filamentous fungus.
infections The disease occurs in humid, wet tropical parts of the
world and is found on primates as well as humans. The
Piedra
natural habitat of this fungus is not known. Black piedra
This is an infection limited to hair shafts and is character­
is most commonly seen in young adults [5].
ized by nodular lesions composed of fungal elements.
There are two varieties: white and black piedra.
Clinical features. Black piedra is usually found only on
scalp hair. Infection is characterized by asymptomatic
White piedra
fusiform black nodules firmly attached to the hair shaft;
Until recently, the yeast‐like fungus Trichosporon beigelii
SECTION 8: FUNGAL SKIN

the cortex of the hair shaft is not penetrated. The nodules


was thought to be the causative organism. This organism
are variable in size, often visible to the naked eye, and are
is found in soil, lake water and on plants, and is occa­
rough and granular to the touch.
sionally seen as part of the normal flora of the human
INFECTIONS

skin and mouth. However, genetic analysis has shown


Diagnosis. This is readily confirmed by microscopy in
that this taxon actually represents a mixture of six distinct
30% KOH. Infected hairs show firmly adherent, black and
species [1,2]. Those associated with infection of genital or
hard, gritty nodules composed of regularly arranged,
scalp hairs are Tr. mucoides, Tr. inkin, Tr. asahii and Tr. ovoides.
thick‐walled cells and hyphae held together by a cement‐
The disease occurs in both temperate and tropical
like substance (Fig.  46.31). Dark‐brown to black velvety
regions. White piedra can affect males and females of all
colonies are produced on culture. The nodules could be
age groups, but infection of the genital hairs is thought to
confused with trichonodosis or trichorrhexis nodosa.
be more common in young men [3]. However, a survey
from equatorial Africa found that, of 449 females exam­
ined, 18% of inguinal hairs were infected by white piedra; Treatment. For both types of piedra, cutting or shaving
in the youngest age group (15–20 years), white piedra off all affected hairs is the simplest form of treatment.
was seen in 32 of 221 patients (14.5%) [4]. Infection may recur and the topical application of an imi­
dazole cream is recommended. One case of black piedra
Clinical features. The presence of irregular, soft, white successfully treated with oral terbinafine has been
nodules along the hair shafts is characteristic of white pie­ reported [6].
dra. Scalp hairs may be involved as well as beard and
axillary hairs but infection is mainly seen in genital hairs. Tinea nigra
The surrounding skin is unaffected. This is an infection of the palmar or plantar skin, charac­
terized by brown to black macules with well‐defined bor­
Differential diagnosis. The nodules produced on pubic ders. It is mainly seen in the tropics but can occur in
hair shafts may be confused with pediculosis or tricho­ Europe and the USA. It is more prevalent under the age of
bacteriosis axillaris. When examined microscopically, the 20 years. Females are three times more likely to be affected
Chapter 46  Superficial Fungal Infections 557

(a) (b)
Fig. 46.31  (a) Black piedra hairs with dark nodules. (b) Hair in 30% KOH showing nodule consisting of regularly arranged, thick‐walled cells and hyphae.

SECTION 8: FUNGAL SKIN


INFECTIONS
Fig. 46.32  Tinea nigra. Fig. 46.33  Infected skin scales showing brown, septate, irregularly
branched hyphae (30% KOH).

than males, the reason for this being unknown; one case
has been reported in a 20‐month‐old girl [7]. Predisposing yellowish or light‐brown hyphae, which are septate and
factors associated with the condition are hyperhidrosis branched, in 30% KOH preparations (Fig. 46.33). Colonies
and presence in coastal areas or hypersaline environ­ of Hortaea werneckii grow on a standard medium within
ments, where the causative agent may be picked up from 5–8 days. They are initially black with a creamy appear­
the natural habitat [8]. ance and later become filamentous.
The disease is caused by the halophilic, melanized,
yeast‐like fungus Hortaea werneckii (Horta) Nishimura Treatment. This consists of daily application of an imi­
and Miyaji, which is found in the soil and on decompos­ dazole for 2–3 weeks to avoid recurrence.
ing plant material.
Neoscytalidium infections
Neoscytalidium dimidiatum, a plant pathogen found in
Clinical features. Tinea nigra usually presents as an tropical and subtropical parts of the world, and N. hyali-
asymptomatic dark patch of skin on the palm of one hand num, a colourless form of the same fungus with lower
(Fig. 46.32) or on the sole. The lesion is solitary, with an virulence [9,10], which has only been isolated from
irregular, sharply defined margin. There is no erythema humans, cause infections of the hand and foot indis­
and only rarely is fine scaling present. tinguishable from the dry‐type infections caused by
T. rubrum; nail infection also occurs.
Differential diagnosis. The main differential diagnosis is In endemic areas, surveys have shown that N. dimidia-
an acral melanoma or naevus. Diagnosis of tinea nigra is tum is a not uncommon cause of foot infections in adults,
confirmed by the presence of hyaline, light‐coloured but only a few cases have been recorded in children.
558 Section 8  Fungal Skin Infections

Fig. 46.35  Neoscytalidium hyphae in skin (30% KOH, phase contrast).

is also fast growing with fairly high aerial mycelium,


which is white or buff cream in colour. Chains of hyaline
arthroconidia are observed on microscopy. Cultures
must be set up on SDA without cycloheximide, to which
these fungi are sensitive [16]. Infections must be distin­
guished from dermatophyte lesions as they are unre­
sponsive to the majority of antifungal drugs.
Fig. 46.34  Neoscytalidium infection of the palm.

Treatment. These nondermatophyte fungi do not respond


In Nigeria, of 64 cement workers with culture‐positive tinea to griseofulvin. Although they exhibit in vitro sensitivity
pedis, N. dimidiatum was the causative organism in 23% of to miconazole, clotrimazole, voriconazole and ampho­
cases and N. hyalinum in 8%. Family members of workers tericin B, there is insufficient evidence at present to
were also examined, and N. dimidiatum was isolated from know whether or not these compounds will prove useful
toe web lesions in two children whose fathers had this clinically [17]. These infections are extremely refractory to
infection [11]. A case of N. dimidiatum infection in a child has treatment and no consistently effective therapy can be
also been reported from a survey of children attending a recommended.
paediatric dermatology clinic in Sri Lanka [12]. Outside
SECTION 8: FUNGAL SKIN

endemic areas, the majority of cases recorded have been in Acknowledgements


immigrants. However, a few cases have been reported in The authors acknowledge use of material from the
patients who have not visited endemic areas [13,14]. excellent chapter in the 2nd edition entitled ‘Superficial
INFECTIONS

fungal infections’ written by Yvonne M. Clayton and


Clinical features. The infection presents with scaling of Mary K. Moore.
the soles and palms (Fig. 46.34) and cracking between the
toes. Nail dystrophy is common and, in the fingernails, References
onycholysis without significant thickening is present; in 1 Gueho E, Improvisi L, De Hoog GS et al. Trichosporon on humans: a
practical account. Mycoses 1994;37:3–10.
addition, nailfold swelling may occur [15]. N. dimidiatum 2 Chagas‐Neto TC, Chaves GM, Colombo AL. Update on the genus
and N. hyalinum produce similar clinical features. Lesions Trichosporon. Mycopathologia 2008;166:121–32.
may be asymptomatic. 3 Kalter DC, Tschen JA, Cernock PL et al. Genital white piedra: epidemi­
ology, microbiology and therapy. J Am Acad Dermatol 1986;14:982–93.
4 Therizol‐Ferly M, Kombila M, Gomez de Diaz M et al. White piedra
Diagnosis by morphology. Microscopic examination of and Trichosporon species in equatorial Africa. I. History and clinical
infected material in 30% KOH shows hyphae resembling aspects: an analysis of 449 superficial inguinal specimens. Mycoses
those of a dermatophyte but with more variation in 1994;37:249–53.
5 Coimbra CEA, Santos RV. Black piedra among the Zoro indians from
width along their length and a double‐contoured appear­ Amazonia (Brazil). Mycopathologia 1989;107:57–60.
ance due to retraction of cytoplasm from the hyphae 6 Gip L. Terbinafine for black piedra. Lancet 1993;341:1164.
cell wall (Fig.  46.35). Cultures of N. dimidiatum are of 7 Palmer SR, Bass JW, Mandojana R et  al. Tinea nigra palmaris and
two forms. They may be fast growing, producing a high, plantaris: a black fungus producing black spots on the palms and
soles. Pediatr Infect Dis 1989;8:49–50.
­cotton‐like aerial mycelium after 3–4 days at 26 °C, ini­ 8 Bonifaz A, Badali H, de Hoog GS et  al. Tinea nigra by Hortaea wer-
tially white but darkening to grey or black as the colony neckii, a report of 22 cases from Mexico. Stud Mycol 2008;61:77–82.
ages. Alternatively, the colonies may be slower growing 9 Machouart M, Menir P, Helenon R et al. Scytalidium and scytalidiosis:
what’s new in 2012? J Mycol Med 2013;23:40–6.
and velvety, without the development of aerial myce­
10 Ruíz‐Cendoya M, Madrid H, Pastor FJ et al. Development of murine
lium. Both are easily identified by the chains of brown models of disseminated infection by Neoscytalidium dimidiatum.
one‐ or two‐celled arthroconidia they produce. N. hyalinum Med Mycol 2010;48:681–6.
Chapter 46  Superficial Fungal Infections 559

11 Oyeka CA, Gugnani HC. Skin infections due to Hendersonula toru- 15 Hay RJ, Moore MK. Clinical features of superficial fungal infections
loidea, Scytalidium hyalinum, Fusarium solani and dermatophytes in caused by Hendersonula toruloidea and Scytalidium hyalinum. Br J
cement factory workers. J Mycol Med 1992;2:197–201. Dermatol 1984;110:677–83.
12 Perera J, Perera C. Fungal skin infections in a paediatric dermatology 16 Midgley G, Moore MK, Cook JC et al. Mycology of nail disorders.
clinic. Ceyl Med J 1993;38:75–7. J Am Acad Dermatol 1994;31:S68–74.
13 Elewski BE, Greer DL. Hendersonula toruloidea and Scytalidium hyali- 17 James JE, Santhanam J, Lee MC et al. In vitro antifungal susceptibility
num. Arch Dermatol 1991;172:1041–4. of Neoscytalidium dimidiatum clinical isolates from Malaysia.
14 Frankel DH, Rippon JW. Hendersonula toruloidea infection in man. Mycopathologia 2017;182:305–13.
Mycopathologia 1989;105:175–86.

SECTION 8: FUNGAL SKIN


INFECTIONS
560 

CHA PTER  4 7

Deep Fungal Infections


María Teresa García‐Romero
Department of Dermatology, National Institute for Pediatrics, Mexico City, Mexico

Introduction, 560 Systemic mycoses, 564


Subcutaneous mycoses, 561 Opportunistic mycoses, 569

Abstract infections. The diagnosis of deep fungal infections should be made


promptly in order to decrease morbidity and mortality. As globali-
Deep fungal infections are uncommon in children and are divided zation occurs and fungal infections, previously restricted to certain
into subcutaneous and systemic mycoses. Subcutaneous mycoses locations, are found outside those areas, and acquired immune
mainly occur in tropical climates and the causal fungi are usually deficiencies have become more common due to infections or ther-
acquired from the environment. Systemic mycoses are divided into apies related to oncological and autoimmune diseases, all physi-
those that occur in specific endemic areas and those that primar- cians should be acquainted with the presentation of deep fungal
ily occur in immunocompromised patients, called opportunistic infections in children.

Key points which occur mainly in Latin America; their treatment is


completely different.
• Sporotrichosis is a subcutaneous mycosis that occurs mainly • Opportunistic infections caused by Mucorales are increasing
in tropical climates and in children presents frequently as due to more severe immunosuppression associated with
one verrucous plaque on the face or acrally as nodules with chemotherapy regimens in children, and should be differentiated
lymphatic spread. from aspergillosis because their treatment is based on
• Mycetomas can be caused by fungi (eumycetomas), more intravenous amphotericin B and surgical debridement instead of
common in Africa and Asia, or actinomycetes (actinomycetomas) voriconazole.

Introduction from where the fungus can disseminate by haematoge­


SECTION 8: FUNGAL SKIN

Fungal infections are common in humans, and they nous and, eventually, lymphatic involvement, and reach
include the frequent superficial infections as well as the the skin and other organs. Opportunistic mycoses are
INFECTIONS

rarer, more severe and even potentially fatal deep and seen in all climates, as their main determinant is an under­
systemic infections [1,2]. Deep fungal infections are lying defect in the immune response that predisposes to
uncommon and are divided into subcutaneous and sys­ ­infection. Their causal agents are considered to have low
temic mycoses. pathogenicity, but can produce disseminated and fatal
The subcutaneous mycoses mainly occur in tropical infections if not treated early [1,3]. The causal fungi exist
­climates and are usually confined to the skin and subcuta­ as saprophytes in the environment in the form of free
neous tissue but may extend deeper, even to bone. The spores and filaments and can be inoculated through
causal fungi are usually from sources in the environment ­occupational exposure or during leisure activities, which
such as soil or plants, and they rarely culminate in sys­ makes human‐to‐human transmission exceptional. The
temic infection. severity of the infection is determined by the extent of the
Systemic mycoses are generally divided into those that exposure to the organism and by the immune status of
occur in specific endemic areas (endemic or respiratory the patient [3,4].
mycoses) that can infect anyone irrespective of their Globalization has made international travel much
immune status, and those that only occur in the immuno­ easier and more accessible, and so fungal infections
­
compromised patient, called opportunistic infections restricted to certain geographical regions can be found in
[1–3]. The endemic or respiratory mycoses are caused by travellers returning from those areas [2].
the inoculation of fungal particles, usually through the This chapter will cover the spectrum of deep fungal
respiratory tract but occasionally by direct inoculation, infections as well as two nonfungal diseases that were

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 47  Deep Fungal Infections 561

formerly considered mycoses and share clinical similari­ form is fixed cutaneous (30% of cases) [7] which presents
ties with them: nocardiosis and actinomycosis. as a plaque, nodular, verrucous or ulcerated lesion [13,14].
Other less frequent presentations include dacryocystitis
[15], earlobe infiltration and deformity [16], nasal lesions
Subcutaneous mycoses [17], palpebral lesions [18] and osteolytic lesions [19].
Sporotrichosis The differential diagnoses include leishmaniasis, cuta­
neous tuberculosis and other atypical mycobacteriosis,
Introduction. Sporotrichosis is a subacute or chronic cutaneous nocardiosis, tularaemia, chromoblastomycosis
infection caused by the dimorphic fungus Sporothrix and mycetoma.
schenckii complex, found in vegetation and organic matter
in temperate climates. It is the most common subcutane­ Laboratory and histological findings. Direct examination
ous mycosis in Latin America. Most cases involve skin in this condition is not useful because fungal structures
and subcutaneous tissues, but disseminated sporotricho­ are not usually observed with routine tests, but periodic
sis can occur in immunosuppressed patients. acid–Schiff (PAS) stain or fluorescent techniques can be
performed. The definitive diagnosis is made by fungal
Epidemiology and pathogenesis. The disease is common culture in Sabouraud dextrose agar (SDA). In cultures,
in children and young adults [5], particularly in some colonies are creamy white initially and then become
geographical locations such as Peru where 60% of cases brown‐black and membranous with radiating growth; the
occur in patients younger than 14 years old, with an inci­ characteristic microscopic features are sympodial conid­
dence of 1 per 1000 [6]. An equal sex distribution occurs. iae resembling daisy flowers. Histopathological findings
Many outbreaks have been described and the fungus has include hyperplastic epidermis and suppurative granulo­
been isolated in and transmitted by fauna, especially mas including cigar‐shaped yeasts and asteroid bodies,
rodents and cats [5–11]. which are fungal elements, surrounded by eosinophilic
Traditionally sporotrichosis was considered an occupa­ material resulting from antigen–antibody reaction
tional disease, particularly of males, with the highest ­proteins [5,13]. Other techniques have been developed in
­incidence in gardeners, florists, labourers and farmers association with outbreaks, such as enzyme‐linked immu­
who acquire the fungus through penetration of the skin; nosorbent assay (ELISA), with excellent accuracy in
however, in a high percentage of patients there is no
­ ­paediatric patients [20].
­history of trauma [7].
Treatment and prevention. Therapeutic options include
Clinical features and  differential diagnosis. There are local hyperthermia, saturated solution of potassium
four clinical forms: lymphocutaneous, fixed cutaneous, iodide (SSKI), azoles (ketoconazole, itraconazole and flu­
multifocal or disseminated, and extracutaneous (affecting conazole) and other antifungals such as terbinafine and
lungs and joints) which is rare and usually acquired by amphotericin B (for systemic disease). SSKI is the first‐

SECTION 8: FUNGAL SKIN


inhalation of the fungus. Overall, the most common form line treatment in many developing countries because of
is lymphocutaneous (70–75%) [12], characterized clini­ its effectiveness and low cost; the dose for children is
cally by an indurated papular lesion that develops about 1.5–3 g/day administered continuously until the lesions

INFECTIONS
1–4 weeks after inoculation. It subsequently turns into a are cleared and then continued for 2–4 weeks more [5,21–
nodule with or without ulceration, and further nodules 23]. Itraconazole is the azole of choice: the paediatric dose
appear following a lymphatic spreading pattern that may is 5 mg/kg per day for 3–6 months [13]. Terbinafine has
ulcerate or suppurate (Fig.  47.1). Regional lymph nodes also been shown to be effective. Other therapeutic alter­
are involved and may ulcerate. The most commonly natives include co‐trimoxazole and griseofulvin. The cure
involved areas are the upper and lower extremities and rate of sporotrichosis is high [5,7,14].
the face. In paediatric patients facial lesions are frequent
(in up to 90% of cases) [5] and the most common clinical Chromoblastomycosis

Introduction. Chromoblastomycosis is an important


s­ ubcutaneous mycosis that occurs worldwide, although it
is more common in the tropics. It is not fatal, but tends to
be very chronic and difficult to treat with potential seri­
ous complications such as lymphatic damage, scarring
and neoplastic transformation. It is caused by traumatic
implantation into the skin of dematiaceous fungi of the
genera Fonsecaea, Cladophialophora and Phialophora that are
present in soil, decaying vegetation and wood.

Epidemiology and pathogenesis. Chromoblastomycosis is


caused by fungi of the genera Fonsecaea, Cladophialophora and
Phialophora such as Fonsecaea pedrosoi, F. compacta, F. nubica,
Fig. 47.1  Lymphatic spread of sporotrichosis in a child. F. monophora, Phialophora verrucosa, Cladophialophora carrionii
562 Section 8  Fungal Skin Infections

and Exophiala jeanselmei – all saprophytes in soil and plants. examination with 10% potassium hydroxide of scrapings
It is distributed worldwide, especially in tropical and sub­ taken from the lesion, the pathognomonic sclerotic or
tropical regions of Latin America, the Caribbean, Africa and ­fumagoid cells (Medlar bodies, copper pennies) can be
Asia. F. pedrosoi is the most common agent found in tropical found, which are small, oval, dark, thick‐walled cells
forests with high rainfall such as the Amazon and elsewhere with septation or binary fission. Cultures can be grown
in Latin America. C. carrionii is the most important agent in in SDA and the microscopic characteristics depend on
dry and desert regions, such as Australia and South Africa. the species; however, since these are slow‐growing fungi,
The disease occurs frequently in young men, especially in the culture can be inconclusive. Polymerase chain reac­
rural areas. Some cases and series of affected children have tion (PCR) assays have been developed for the identifica­
been described (86% are males), with a mean age of 11 years tion of Fonsecaea species and C. carrionii. Histopathological
[24]. It is a disease with a prolonged evolution time (10 years findings are marked ­epitheliomatous hyperplasia, gran­
or more) and therefore many adult patients could have ulomatous infiltrates with multinucleate giant cells and
acquired it during childhood. More than half of the children the presence of sclerotic bodies intermixed with the infil­
reported with chromoblastomycosis had a close relative trate [14,24–27].
affected, which suggests a genetic susceptibility to develop
the disease [24]. In paediatric cases the most frequent causa­ Treatment and prevention. Treatment is difficult as the
tive agent has been found to be C. carrionii [24,25]. disease has variable cure rates and is recalcitrant,
although paediatric cases have higher cure rates (86%)
Clinical features and  differential diagnosis. Lesions of [24]. Management depends on the aetiological agent,
chromoblastomycosis start as erythematous, verrucous size and extent of lesions, and consists of long courses
papules and plaques that can progress and eventually, (6 months) of antifungal drugs combined with physical
after many years, ulcerate or become cauliflower‐like; treatments such as thermotherapy, surgery, electrodessi­
necrotic plaques have been described in infancy. The most cation or ­liquid nitrogen [26,27]. Itraconazole and terbi­
frequent location in children is the upper limbs (whereas nafine ­followed by surgery have shown good efficacy
in adults it is the lower limbs) [24], followed by the back for localized lesions [26]; other options are oral 5‐fluoro­
and lower limbs [24]. As the lesion increases in size or cytosine, 5‐fluorouracil (5FU) [24], SSKI, intralesional
heals, the central area becomes scar‐like and atrophic and amphotericin B, thiabendazole [14], vitamin D [26] and
can be severely disfiguring [25–28] (Fig. 47.2) Skin lesions ajoene in some areas of South America [24,26]. Local
may be mildly pruritic or asymptomatic unless complica­ heat therapy using pocket warmers for 6 months has
tions develop, including ulceration, secondary infection also been found to be effective, because temperatures
and lymphoedema. Rarely, chronic lesions have been over 40–42 °C kill the causative fungi [26]. The response
complicated by malignant transformation into squamous also depends on the fungal species causing the ­condition:
cell carcinoma. Chromoblastomycosis stays confined to C. carrionii and P. verrucosa respond better to a­ ntifungals
the skin and rarely invades underlying muscle or bone, than F. pedrosoi [25,26].
SECTION 8: FUNGAL SKIN

except in the context of immunosuppression [27].


The differential diagnoses are tuberculosis verrucosa Mycetoma
cutis, sporotrichosis, psoriasis and squamous cell carci­ Mycetomas are unique chronic granulomatous subcuta­
INFECTIONS

noma, which can develop in chronic ulcerated lesions [26]. neous infections caused by actinomycetes (actinomyceto­
mas) and by fungi (eumycetomas). Differentiating
Laboratory and  histological findings. Microbiological between these two categories is essential for treatment
confirmation of the diagnosis is key to differentiate and prognosis. They affect the skin but can also compro­
­chromoblastomycosis from other conditions. On direct mise fascia, tendons, muscles and bones, giving rise to
significant morbidity and disability. Mycetoma is
amongst the most common of the subcutaneous mycoses
with a worldwide distribution, predominantly in tropical
and subtropical countries (the so‐called mycetoma belt)
where this poses a significant burden on global health
[29,30]. Actinomycetomas are more common in Mexico,
Central and South America, and eumycetomas in Africa
and India [31,32].

Eumycetoma

Introduction.
Eumycetoma is, along with sporotrichosis, the most com­
mon subcutaneous mycosis worldwide. It is not frequent
in children, and there is a possible role for hormones in its
development that could explain this, as could the fact that
Fig. 47.2  Verrucous scar‐like plaque typical of chromoblastomycosis. it is a very slowly progressing condition that might go
Source: Courtesy of Prof. Roberto Arenas MD, Mexico City. unnoticed until adulthood [31].
Chapter 47  Deep Fungal Infections 563

the new posaconazole and voriconazole are effective, but


Epidemiology and  pathogenesis. At least 30 species of
should be given for 12–24 months [14], a costly option in
fungi are associated with eumycetomas, and they are
low‐resource settings. Terbinafine for 24–48 weeks has
classified according to the type of grains found in
­
been effective in some reports [31,36]. The combination of
­secretions: black or white‐grey grains. More than 90% of
itraconazole and terbinafine for 6–12 weeks before surgi­
eumycetomas worldwide are caused by Madurella myce-
cal removal and continued until remission of the disease
tomatis, M. grisea, Pseudoallescheria boydii and Leptosphaeria
is very effective and easier for compliance [31,32,34].
senegalensis. The distribution depends on the geographical
Given the high rates of drug resistance, routine drug
location: Acremonium spp. and M. grisea are common in
­susceptibility testing should be used when available [32].
South America, and M. mycetomatis has an extensive distri­
bution in India and Africa. Soil is the natural reservoir of
Actinomycetoma
most agents and the disease begins with the traumatic
introduction of the fungus through the skin. The evolution
Introduction. Actinomycetoma is a chronic subcutane­
of the disease is very slow and most patients seek medical
ous infection caused by aerobic branching actinomycetes.
help after many years [31]. There is a male predominance,
It is endemic in tropical climates with the highest inci­
and the affected patients are usually young adults,
dence occurring in Asia, Africa, Mexico, Central and
although in some regions of the world such as Sudan, a
South America.
significant number of cases in children are described
[31,33] with an incidence between 3% and 4.5% [34]. In the
Epidemiology and  pathogenesis. Worldwide, 60% of
largest series reported, most children were male (73.5%),
mycetomas are caused by actinomycetes, but in Mexico
between 10 and 14 years of age, and students (79.6%). The
and Central and South America 90% of mycetomas are
average duration of the disease was 2 years [34].
actinomycetomas. The most frequent causative agents are
Nocardia brasiliensis, N. asteroides, Actinomadura madurae,
Clinical features and differential diagnosis. Mycetomas
A. pelletieri and Streptomyces somaliensis [39].
present as tumour‐like lesions, gradually developing
Actinomycetomas are uncommon in children (4.5–9.8%
­nodules, abscesses and sinus tracts that drain a mucopu­
of all cases) [40,41] and only a few cases and series are
rulent discharge in which fungal colonies in the form of
reported in the literature. The role of hormones in disease
grains are found. The grains can be observed with the
susceptibility may explain the fewer cases of mycetoma in
naked eye in some cases or microscopically, and their
children. The median age in a paediatric series was
morphology and colour contribute to their identification
11.2 years, the mean duration of disease was 1 year, and
[31]. The most frequent location is the foot, followed by
there was male predominance (80%) and a frequent
the hands, legs and back [34]; mycetomas in the oral
­history of farm work [40].
mucosa, face and scalp have been reported [33,35–37].
Mycetomas can progress to osteomyelitis and require
Clinical features and  differential diagnosis. Actinomy­
amputation in severe cases [38].

SECTION 8: FUNGAL SKIN


cetomas are similar to eumycetomas but the clinical
The differential diagnosis includes foreign body granu­
picture is more inflammatory. There is induration and
­
lomas, sporotrichosis, chromomycosis, leishmaniasis or
increased volume, causing deformity of the affected area
cutaneous tuberculosis. In children the diagnosis of soft

INFECTIONS
with the presence of nodules, scar tissue, abscesses, fistulae
tissue neoplasia should be ruled out [33].
and purulent exudate that contains the granules (Fig. 47.3).
The most commonly affected sites in all ages are the legs,
Laboratory and histological findings. Diagnosis is con­ followed by the back and chest [40]. In children there is
firmed by direct microscopy, histopathology and cul­ often a milder presentation, even in longstanding cases,
ture. Direct microscopic observation of the grains can
contribute to identification of the causative fungus.
Culture of the secretion and grains in SDA will yield the
causative agent’s colony and PCR can also be used for
identification if culture fails. Histologically, pseudoepi­
theliomatous hyperplasia with abundant granulation
tissue and granulomatous inflammation are found, with
grains in the centre of the abscesses. Disease extent can
be assessed by radiology, ultrasonography or even
­helical computed tomography (CT) [14,31,32].

Treatment and  prevention. Eumycetomas usually


respond less well to drug treatment and therefore
­combined medical and surgical treatment is the standard
therapy, achieving better cure rates in shorter periods.
Drug treatment is given before surgery to decrease lesion
size and continued afterwards to ensure complete remis­ Fig. 47.3  Actinomycetoma in a child. Note the marked inflammation and
sion [32]. Azoles such as ketoconazole, itraconazole and volume increase with tumefaction and fistulization.
564 Section 8  Fungal Skin Infections

with one or two draining sinuses, little or no swelling and cutaneous infection occurs after traumatic or accidental
seldom bone involvement (minimycetoma) [40,42,43]. inoculation, and disseminated disease is seen in the
The differential diagnosis includes eumycetoma, spor­ immunocompromised [14].
otrichosis, tuberculosis, osteomyelitis, botryomycosis,
phaeohyphomycosis and neoplasias of the bone and soft Epidemiology and  pathogenesis. Coccidioidomycosis
tissues [39]. occurs predominantly in the western hemisphere, mainly
in the southern area of the USA and northern states of
Laboratory and histological findings. Direct microscopic Mexico. It is also endemic in some regions of Central and
examination with 10% KOH or saline reveals the size and South America such as Guatemala, Venezuela, Colombia
morphology of the grains, which help to identify the and Argentina.
causative agent. Isolation of the actinomycete is achieved The causative agent is Coccidioides, either immitis or
by culture of the discharge using Sabouraud or blood posadasii, a dimorphic fungus characterized by a sapro­
agar. Colonies grow slowly and their characteristics vary phytic or infecting phase in the form of arthrospores and
depending on the agent. On histology, suppurative a parasitic phase with spherules containing endospores in
­granulomatous inflammation is found and within this their interior. It is found in the soil and vegetation of semi‐
infiltrate the grains of the specific agent are present arid areas and introduction to the body occurs through
(Fig. 47.4) [4]. the respiratory tract, although most infected individuals
Imaging studies are useful in delineating the extent of are asymptomatic [45,46]. In endemic areas, both genders
the lesions in bone and neighbouring tissues [39]. and all ages are equally affected [47] but two peaks have
been described: in children under 5, and in adults over
Treatment and  prevention. Treatment should be only 45 years of age [48]. There is an ethnic predisposition to
medical, as surgical interventions may disseminate the severe infection, with higher rates of dissemination in the
condition. There are several regimens available: the most Philippines and in African Americans (10–175 times
effective includes co‐trimoxazole alone [39] or in combi­ higher), although the reason is not completely under­
nation with other antibiotics such as dapsone alone or stood [49]. In large case series of coccidioidomycosis in
combined with rifampicin for 6 months to 2 years [42], children, 60% were male with a median age at diagnosis
depending on the clinical response [40]. Other regimens of 5 years, ranging from 6 months to 17 years of age
include amoxicillin/clavulanate [40] or the combination [50,51]. There has been an increase in incidence in the last
of meropenem or imipenem with or without amikacin for decade with rates as high as 15 per 100 000 [52], as well as
3 weeks and repeated after a 6‐month interval, which has an increase in hospitalization rates due to the disease [53],
shown very good results [32,44]. with health‐related costs becoming a public health chal­
lenge [48,49,54–56]. Also, with intensive treatments for
patients with haematological malignancies becoming
Systemic mycoses
more common, disseminated coccidioidomycosis is being
Coccidioidomycosis
SECTION 8: FUNGAL SKIN

seen frequently in these patients who live in endemic


areas, warranting special attention to diagnose, effec­
Introduction. Coccidioidomycosis, also known as San tively treat and appropriately adjust chemotherapy plans
INFECTIONS

Joaquin Valley fever, is an endemic mycosis caused by to minimize immunosuppression [57].


two distinct species, Coccidioides immitis and C. posadasii,
both saprophytes of soil found in desert areas. The most Clinical features and differential diagnosis. In children,
frequent presentation is pulmonary; however, primary the most common clinical presentation is pneumonitis
and mediastinitis [50,51]. Infected individuals are asymp­
tomatic in 60–75% of cases, the remaining presenting a
nonspecific upper respiratory infection 1–3 weeks after
inoculation [45]. Only 1–5% develop a chronic pulmonary
infection and/or disseminated infection of other organs,
especially  –  but not exclusively  –  in the context of an
altered immune system [46]. Symptomatic patients pre­
sent with fever, chills, asthenia, anorexia, productive
cough and occasionally chest pain, all of which disappear
in about 8 weeks. In almost 5% of cases, a chronic infec­
tion develops with pulmonary infiltrates, pleural dis­
charge, cavernous lesions and coccidioidomas (fungus
balls). When the disease disseminates it affects lymph
nodes, skin, bones, joints, heart, liver, spleen, kidneys, the
central nervous system and occasionally the retina [49,58].
Symptoms vary depending on the affected site. Cutaneous
findings include abscesses, fistulae, verrucous lesions,
Fig. 47.4  Histology of an actinomycetoma showing a grain characteristic ulcers, and keloid scars [14]. Central nervous system
of Nocardia brasiliensis surrounded by a mixed inflammatory infiltrate. involvement causes headache, confusion, hydrocephaly
Chapter 47  Deep Fungal Infections 565

and diverse neurological syndromes [14]. Musculoskeletal immunosuppression, should receive amphotericin B
infection can occur in 10–50% of children with extrapul­ [69] or posaconazole, which has shown efficacy in
monary infection [59], manifesting as painful osseous patients with chronic refractory meningitis [70]. The
masses usually without preceding pulmonary infection, combination of voriconazole and caspofungin salvage
the most common location being the foot and the knee therapy for refractory paediatric coccidioidomycosis
[45,46,60]. There are reports of intraocular infection simu­ has shown excellent results [71]. For musculoskeletal
lating neoplasms [61,62], and abdominal and pelvic and sometimes thoracic disease, treatment should be a
masses simulating appendicitis and tumours [63]. combination of medical and surgical approaches
The differential diagnosis includes lung cancer, viral [46,59,72]. The duration of therapy varies on the site
infections, mycoplasma, tuberculosis, parasitic abscesses and severity of disease. Duration of 3–6 months is
[64], North American blastomycosis, histoplasmosis, asper­ ­preferred for primary pulmonary disease, with follow‐up
gillosis, sporotrichosis and cryptococcosis. Musculoskeletal for at least 1 year after diagnosis. In contrast, chronic
infection can be confused with osteomyelitis and sarcomas, pulmonary disease warrants lifelong therapy to pre­
and intra‐abdominal infection can be confused with serve lung function and avoid damage [49].
tumours, abscesses and appendicitis. Development of a vaccine against Coccidioides spp. is
under way, with cost‐effectiveness analysis showing a
Laboratory and  histological findings. The diagnosis is substantial public health benefit [53].
confirmed by the identification of the parasitic or sapro­
phytic form of the fungus. The saprophytic form are 10‐ to Histoplasmosis
100‐µm sized spherules contained by a refractile double
wall and can be seen by direct KOH examination or by Introduction. Histoplasmosis (also known as American
PAS stain of exudate, pus or cerebrospinal fluid. Special histoplasmosis, Darling disease or Ohio Valley fever) is a
stains such as PAS, Gridley and Gomori facilitate the common infection produced by the dimorphic fungus
identification of the fungus, especially when the spher­ Histoplasma capsulatum var. capsulatum. It is endemic to
ules are scarce. Culture of any specimen such as sputum certain zones and affects primarily the lungs, but skin
or tissue requires Sabouraud medium; it should be manifestations can occur as the result of haematogenous
­processed in a level III security bell because accidental spread or direct inoculation. Another type of histoplas­
infection may occur in laboratory personnel. C. immitis mosis in humans is caused by Histoplasma capsulatum var.
grows in 7–14 days as a white, downy colony but it can duboisii, also called African histoplasmosis because it is
present variations in texture and colour [14,49]. endemic to that continent. In this chapter we will refer
Histological findings include granulomas with a poly­ only to American histoplasmosis.
morph nuclear infiltrate, plasma cells, macrophages,
giant cells, areas of necrosis and the presence of spherules Epidemiology and  pathogenesis. Histoplasmosis is the
with a double refractive outer wall that vary from 10 to most common endemic mycosis causing human infection

SECTION 8: FUNGAL SKIN


100 µm in diameter [65]. [73]. Histoplasma capsulatum is a dimorphic saprophytic
Antigens such as coccidioidin (obtained from the myce­ soil fungus with a worldwide distribution. The endemic
lial phase) and spherulin (from the spherular phase) can areas are the Mississippi and Ohio river valleys of the

INFECTIONS
be used to evaluate previous exposure to the disease and USA, the rural regions of tropical Central and South
response to treatment. Coccidioidin is more widely America, and some areas in the eastern USA, southern
­available and commonly used. Serological tests can detect Europe, Africa and South‐East Asia [73–77]. In the USA
antibodies in the first phase of the infection; the most population histoplasmin skin testing is positive in 80%
­reliable is the immunodiffusion test which can be used [14]. The spores are found in soil contaminated with
both in serum and cerebrospinal fluid. Complement‐­ chicken, pigeon or bat feathers or droppings (guano).
fixation serological tests determine the level of IgG Activities associated with this infection include: mining;
­antibodies against the antigens of C. immitis [14,49]. landscaping; cleaning debris from attics, bridges or barns;
Imaging studies such as chest radiograph, CT scans tearing down structures; and hobbies such as speleology
and magnetic resonance imaging (MRI) help visualize the and caving [73,75,76,78].
extent of the fungal infection. Histoplasmosis affects all races and ages, with infants
and children commonly having the acute pulmonary or
Treatment and  prevention. For pulmonary disease, disseminated form [77–79]. Histoplasmosis is usually
a­ ntifungal therapy is frequently not required, but pro­ acquired by inhalation, but there have been rare cases of
longed courses of antifungals are generally needed in transmission from fomites, direct inoculation, solid organ
­disseminated disease. The first line of therapy is with transplant, sexual contact and wood‐burning [77,80,81].
­triazole antifungals: either fluconazole or itraconazole H. capsulatum is an intracellular pathogen with the abil­
has been effective for various forms of coccidioidomy­ ity to remain viable in tissues for years in a latent state;
cosis, including meningitis [66]. Clinical evidence sug­ thus, if cell‐mediated immunity wanes, the organism can
gests that ­ itraconazole is equivalent or superior to reactivate and cause disease [73,81]. In patients with
fluconazole in treating osteoarticular infections in rates human immunodeficiency virus (HIV)‐acquired immune
of cure and  recurrence [67,68]. Patients with dissemi­ deficiency syndrome (AIDS) the frequency of infection is
nated or refractory disease, especially in the context of higher than in the immunocompetent host, with a greater
566 Section 8  Fungal Skin Infections

risk for life‐threatening, disseminated infections, and


histoplasmosis is considered a marker of the disease
­
[79,82]. In paediatric patients who received solid organ or
haematopoietic cell transplants, the 12‐month cumulative
incidence rate for histoplasmosis is 0.102%, and the risk of
infection continues for years after transplant [83].
Children with primary or drug‐induced immunodefi­
ciency are also at a higher risk of acquiring severe forms
of the disease [84,85].

Clinical features and differential diagnosis. Histoplasmosis


is a systemic infection that can compromise multiple tis­
sues; however, in the majority of cases the lungs and reticu­
loendothelial system are affected. Illness may occur within
a few weeks following heavy exposure, or months to years
Fig. 47.5  Disseminated histoplasmosis in an immunocompromised teenager.
later following low‐inoculum exposure or even due to
reactivation of old infection during immunosuppression.
Up to 95% of affected individuals have asymptomatic dis­ characterized by a chancre‐like lesion and regional lym­
ease that is discovered by radiography of the lungs (hilar phadenopathy that resolves in several months with no
calcification) or positive skin tests to histoplasmin but 5% treatment. Widespread primary cutaneous disease has
can develop systemic illness, particularly immunocompro­ been described but is exceptional.
mised patients [75,85]. Histoplasmosis produces four major The differential diagnosis of acute pulmonary histo­
groups of clinical manifestations: (i) acute pulmonary plasmosis is acute pulmonary blastomycosis and atypical
form; (ii) chronic pulmonary form; (iii) disseminated forms; community‐acquired pneumonias. The differential
and (iv) traumatic primary cutaneous form. The most com­ ­diagnosis of chronic pulmonary histoplasmosis includes
mon forms of histoplasmosis in children are severe acute tuberculosis, nontuberculous mycobacterial infections,
pulmonary histoplasmosis, mediastinal adenitis and other endemic fungal infections and sarcoidosis.
­progressive disseminated histoplasmosis [77,78].
Acute pulmonary histoplasmosis usually follows a Laboratory and histological findings. The fungus can be
heavy inoculum exposure. It is the most common initial isolated from a variety of samples such as saliva, cerebro­
form and manifests as a flu‐like febrile disorder, with spinal fluid, blood, bone marrow, pus and biopsies. In
cough, malaise, chest pain, low‐grade fever, upper res­ direct examination of these smears stained with Wright or
piratory symptoms and pulmonary infiltration lasting for Giemsa stains, intracellular yeasts of 3–4 µm in size can be
several weeks or months. In approximately 5% of patients seen. Culture of blood, sputum or skin material in
it is accompanied by rheumatological and/or dermato­ Sabouraud or blood agar grows smooth cotton‐like white
SECTION 8: FUNGAL SKIN

logical manifestations, such as erythema nodosum and or brownish colonies in up to 4–6 weeks. Microscopic
erythema multiforme, and arthritis and arthralgias. examination of the colonies is necessary for positive iden­
INFECTIONS

Chronic pulmonary histoplasmosis resembles tubercu­ tification and shows abundant tuberculate macroconidia
losis clinically and radiographically and can also present and microconidia [14]. PCR on paraffin‐embedded tissue
with erythema nodosum and erythema multiforme. also allows the identification of the fungus.
Disseminated histoplasmosis is rare and affects mostly Biopsy of skin stained with Gomori’s methenamine sil­
young children and older individuals, as well as immu­ ver or PAS stains demonstrates a granulomatous reaction
nocompromised hosts. It has become more frequent with with epithelioid and giant cells, macrophages and histio­
the AIDS epidemic. Fulminant disease is seen during cytes, with or without caseous necrosis. Small (1–3 µm),
childhood, especially in the first year of life. This form of round budding encapsulated yeasts are found within
histoplasmosis presents with high fever, anaemia, hepat­ macrophages and grouped in extracellular masses [65].
osplenomegaly, lymphadenomegaly, diarrhoea and Serological tests (complement fixation and immunodif­
weight loss. Mucosal and cutaneous lesions are due to fusion) play an important role in the diagnosis of acute
haematogenous spread of the fungus and can be papules, pulmonary, chronic cavitary pulmonary and chronic
nodules, molluscoid lesions, indurated granulomatous or progressive disseminated histoplasmosis. However, in
­
vegetating plaques; they frequently become ulcerated immunosuppressed patients, serology is rarely useful.
and are usually multiple (Fig. 47.5). Both assays have a sensitivity of about 80%. A false‐­
Other less common forms of infection in children are negative test is seen during the first month following
cerebral [86], meningeal [87], granulomatous pleuritis acute infection, and a false‐positive test may occur in
[88] and endocarditis [89]. patients with other fungal infections and granulomatous
Primary cutaneous histoplasmosis is extremely rare processes. The histoplasmin skin test demonstrates the
and seen especially in laboratory workers. It can also be immune response of the host to the fungus, but is of little
the result of accidental inoculation of the fungus into value in endemic areas [77,90].
the skin of a patient with immunodeficiency. In the Measuring the cell‐wall polysaccharide antigen of
immunocompetent host, this form of histoplasmosis is H.  capsulatum is a sensitive diagnostic tool for patients
Chapter 47  Deep Fungal Infections 567

who have disseminated infection, and urine is preferred infected by group 2 present with isolated pulmonary
for testing because of enhanced sensitivity. Antigen involvement and constitutional symptoms, with similar
­detection is also helpful in monitoring efficacy of therapy findings replicated in a paediatric cohort [95,96].
and detecting relapses [73,90]. Examination of faecal Blastomycosis affects mainly adult men in rural areas,
mucus to detect yeast cells of the fungus has shown to be and there have been outbreaks associated with dogs,
quick and cost‐effective for diagnosing disseminated which can also be infected by the fungus. Certain ethnici­
­disease in children [91]. ties seem to have a greater incidence of the disease, such
as African Americans, Canadian aboriginals and Asian
Treatment and  prevention. Mild to moderate acute Hmong [93,94,97]. Patients of all ages are susceptible but
­ ulmonary histoplasmosis in children does not usually
p children are rarely infected by this agent: only 3–11% of
warrant treatment, but itraconazole for 6–12 weeks is reported cases occur in patients under 20 years of age
­recommended for patients who continue having symp­ [93,95], and the youngest reported surviving patient was
toms for 1 month after the infection. For moderate or 4 months of age [98].
severe pulmonary infection amphotericin B deoxycholate Rare human‐to‐human transmission has been observed
1 mg/kg daily is well tolerated, or itraconazole 5–10 mg/kg in female sexual partners of men with blastomycosis
daily in two divided doses. For progressive disseminated ­disseminated to the prostate gland, in post mortem trans­
infection treatment with amphotericin B deoxycholate mission at autopsy and perinatal infection of newborns
1 mg/kg daily for 4–6 weeks is recommended, or the [98]. In contrast to other fungal infections usually seen in
same dose for 2–4 weeks followed by itraconazole immunocompromised patients, B. dermatitidis is a true
5–10 mg/kg daily in two divided doses to complete pathogen that often infects immunocompetent individu­
3 months of therapy. Longer therapy may be needed for als [65,95].
patients with severe disease, immunosuppression or pri­
mary immunodeficiency syndromes. Lifelong suppres­ Clinical features and differential diagnosis. The clinical
sive therapy with itraconazole 5 mg/kg daily may be spectrum of blastomycosis ranges from asymptomatic
required in immunosuppressed patients and in those subclinical infection to disseminated or even fulminant
who relapse despite adequate treatment [74]. Voriconazole disease.
and posaconazole are considered second‐line agents Following an incubation period of 2–6 weeks, pulmonary
because there are no treatment trials as yet using these blastomycosis develops, although asymptomatic infection
agents [92]. occurs in at least 50% of cases. Acute pulmonary disease
Most cases of histoplasmosis resolve spontaneously and mimics community‐acquired bacterial pneumonia with
have a good prognosis, with minimal pulmonary calcifica­ uni‐ or bilobal infiltrates, and typically resolves spontane­
tion, but disseminated histoplasmosis carries a serious ously. Chronic pulmonary blastomycosis is an indolent pro­
prognosis with a mortality rate as high as 95%, especially cess that tends to progress insidiously with fever, weight
in young infants or immunocompromised hosts. loss and night sweats and cavitary or mass‐like lesions on

SECTION 8: FUNGAL SKIN


radiographs. It is indistinguishable from tuberculosis, other
Blastomycosis fungal infections and cancer. Only a few patients experience
disseminated disease, where the skin and subcutaneous

INFECTIONS
Introduction. Blastomycosis, also known as Gilchrist tissue are the most common sites of extrapulmonary
­
­ isease or North American blastomycosis, is a chronic
d involvement, ­followed by bones and joints, the genitouri­
granulomatous disease of the lungs, skin and subcutane­ nary tract and the central nervous system [93].
ous tissue produced by the dimorphic fungus Blastomyces Cutaneous lesions occur in 60% of patients with
dermatitidis. extrapulmonary blastomycosis and may be single or mul­
tiple and affect any part of the body [99,100]. They tend to
Epidemiology and  pathogenesis. Blastomycosis is seen be nodules or plaques that enlarge and ulcerate or become
most frequently in the central eastern and mid‐western verrucous or crusted, sometimes developing abscesses
USA and Canadian provinces, and occasionally in Central and fistulizing through the overlying skin. Erythema
and South America and isolated regions of Asia and nodosum may be associated with pulmonary blastomy­
Africa. Within endemic areas the annual incidence can be cosis [93].
up to 101.3 per 100 000 [93,94]. The fungus grows as a Primary cutaneous blastomycosis (also known as inoc­
nonpathogenic mould in soil and decaying wood and its ulation blastomycosis) is a rare infection resulting either
conidia convert to pathogenic yeasts in the tissues. from laboratory accidents or animal bites. After inocula­
Occupational and recreational activities that involve tion, an erythematous, indurated area with a chancre
­disruption of soil or wood, frequently along streams or associated with painful lymphadenopathy appears in
rivers, can result in exposure to the conidia. These are 1–2 weeks, but spontaneously heals [93].
inhaled and phagocytosed by macrophages and neutro­ In children, the disease tends to present as acute pneu­
phils in the lungs, but some can become yeasts evading monia, but extrapulmonary dissemination is common
the immune response, become pathogenic and dissemi­ (38%) with skeletal involvement as well as cutaneous and
nate causing systemic infection [93,95]. Two species of central nervous system manifestations being frequent,
Blastomyces have been identified: group 1 (B. gilchristii), associated with higher morbidity and mortality rates
and group 2 (B. dermatitidis). The majority of patients [93,95,101–103].
568 Section 8  Fungal Skin Infections

The diagnosis of blastomycosis is difficult and ­frequently It is also known as South American blastomycosis or Lutz–
delayed, which highlights the importance of the dermatol­ Splendore–de Almeida disease.
ogist recognizing blastomycotic skin lesions, which, along
with pneumonia unresponsive to treatment, are highly Epidemiology and  pathogenesis. Paracoccidioidomycosis
suggestive of the disease in endemic areas [95,97,101]. is endemic in Mexico and South America, with most cases
The differential diagnosis of cutaneous blastomycosis occurring in Brazil (where the reported incidence is 1–10
is wide and includes scrofuloderma, lupus vulgaris, squa­ cases per 100 000 per year, making the disease an impor­
mous cell carcinoma, keratoacanthoma, tertiary syphilis, tant public health concern), Colombia and Venezuela.
leprosy, bacterial pyoderma, pyoderma gangrenosum, Even though both sexes are equally exposed to the fun­
leishmaniasis and other fungal infections. gus, paracoccidioidomycosis is more common in males in
a 15 : 1 ratio [106,107]. Approximately 10 million people
Laboratory and histological findings. Direct examination are infected, 2% of whom will develop the disease. It
with potassium hydroxide of the purulent discharge from occurs more frequently in adults, although in a large
a lesion or sputum often demonstrates the characteristic series up to 18.5% of patients were children, and 9.7%
thick double‐walled, round yeast with wide‐based bud­ were younger than 4 years [106,108].
ding. Fungal cultures require a 1‐ to 4‐week incubation Transmission is through inhalation of the fungal spores,
period for growth of a slow‐growing brown, wrinkled but the skin and oropharyngeal mucous membranes can
yeast colony at body temperature (37 °C) and a mycelial be inoculated after trauma with contaminated materials.
form at room temperature (25 °C), producing a white, There is no evidence of human‐to‐human transmission. The
fluffy colony on SDA. incubation period is variable, from weeks to years. There
Histopathological examination of tissue specimens is a correlation of T‐cell depression with the severity of
with use of Gomori’s methenamine silver or PAS stain paracoccidioidomycosis, and hormonal, genetic and
demonstrates a suppurative noncaseating granulomatous nutritional factors all play a role in the development of
reaction with the distinctive yeasts inside macrophages: the infection and clinical disease [108,109].
round to oval, 8–15 µm in diameter, with a thick refractile
wall located intra‐ and extracellularly [65]. An enzyme Clinical features and differential diagnosis. Two clinical
immunoassay (EIA) for the detection of a polysaccharide forms of paracoccidioidomycosis are currently recog­
cell‐wall antigen in urine, serum, bronchoalveolar lavage nized: the acute juvenile form, and the chronic adult form.
and cerebrospinal fluid has improved rapid diagnosis of The acute juvenile form occurs in 25% of cases and
blastomycosis, with high sensitivity but low specificity affects mainly children and adolescents [106]. It is charac­
because of cross‐reactivity. Serological testing has poor terized by disseminated infection that progresses rapidly
sensitivity and specificity, but an EIA using the BAD‐1 with reticuloendothelial system involvement and a Th2
antigen of the fungus is promising. Real‐time PCR assays immunological response. Skin lesions are infrequent, and
for rapid diagnosis are being used with very high sensi­ the only mucocutaneous manifestations are gingival
SECTION 8: FUNGAL SKIN

tivity and specificity [93,104]. oedema with recession and tooth loss, or infiltrated viola­
ceous plaques reminiscent of sarcoidosis [110]. Mortality
Treatment and  prevention. Current guidelines empha­ is as high as 5–11% [14,108,111,112].
INFECTIONS

size that all patients with blastomycosis should receive Most adult patients affected by paracoccidioidomyco­
antifungal treatment because of the high likelihood of sis have the chronic form, characterized by a Th1 response,
progression or recurrence of the infection. Itraconazole is which is the result of primary pulmonary infection and
first‐line therapy for mild to moderate disease for a can be indolent and self‐limiting (75%) or chronic and
­minimum of 6–12 months. Other options are fluconazole, progressive. It initially presents with mucosal nodules,
voriconazole or posaconazole, which have been used suc­ vegetations or ulcers (called mulberry‐like stomatitis),
cessfully in a few reports. For moderately severe to severe which can progress to tissue destruction and ulceration
blastomycosis, treatment should be with amphotericin B and respiratory obstruction [108,113].
until improvement is noted in 1–2 weeks, followed by The main differential diagnoses of the acute juvenile
itraconazole or voriconazole for 12 months. For acute res­ disease are lymphoproliferative disorders or sarcoidosis
piratory distress syndrome (ARDS), methylprednisolone [110,111]; for the classic chronic adult form with oral
can be added or extracorporeal membrane oxygenation lesions, squamous cell carcinoma, leishmaniasis, tubercu­
provided. Central nervous system disease and infection losis, syphilis, sarcoidosis and Wegener granulomatosis
in immunosuppressed patients should be treated as may be considered.
severe blastomycosis [105]. Overall, mortality is 4–6%,
but if ARDS occurs it can be as high as 89% [93]. Laboratory and  histological findings. Diagnosis is
mainly clinical. Mycological direct examination shows
Paracoccidioidomycosis (South American yeast‐like cells with a helm‐like arrangement (Fig. 47.6).
or Brazilian blastomycosis) Histology shows pseudoepitheliomatous hyperplasia
with intraepithelial microabscesses and a granulomatous
Introduction. Paracoccidioidomycosis is the most frequent reaction. Spores of P. brasiliensis are round, 6–20 µm in
systemic mycosis in Latin America and is caused by the ther­ diameter, and develop multiple buds in their periphery
mally dimorphic fungus Paracoccidioides brasiliensis complex. that give the characteristic ‘pilot’s wheel’ configuration.
Chapter 47  Deep Fungal Infections 569

Epidemiology and  pathogenesis. All species of Candida


are distributed widely in nature, but C. albicans, the most
frequent one, is an endosaprophyte of mammal and bird
gastrointestinal tract, C. tropicalis is found in oropharynx
and C. glabrata in vagina. They become pathogenic when
cellular immunity fails and/or when the normal flora is
altered. The incidence of invasive candidiasis has steadily
risen because of increasing use of multiple antibiotics and
invasive procedures such as parenteral nutrition and cen­
tral venous catheters. Any source of immune compromise
(e.g. primary immunodeficiency, haematological malig­
nancy, solid organ or stem cell transplantation, HIV‐AIDS
or medication) is associated with disseminated infection by
this yeast [119]. Incidence rates have decreased signifi­
Fig. 47.6  Direct examination of exudate showing spherical spores with cantly in the general population following strict protocols
multiple buds in a pilot’s wheel configuration characteristic of P. brasiliensis. for central line insertion, but in the paediatric population
Source: Courtesy of Prof. Alejandro Bonifaz, Mexico City. >1 year of age they have increased from 2.0 to 2.4 per
100 000 person‐years in a period of 5 years in the USA [120].
Worldwide there is a shift from C. albicans towards non‐
They are found frequently in multinucleated giant cells, albicans species such as C. parapsilosis and C. tropicalis, with
but also inside the microabscesses [65,108]. an associated increase in mortality and antifungal resist­
P. brasiliensis grows very slowly in special media with ance. In large studies, the median age for disseminated
antibiotics. Serological examination by immunodiffu­ candidiasis in children was 3.5 years and the most common
sion or complement fixation and skin intradermal tests underlying diagnoses were oncological, gastrointestinal
with paracoccidioidin are used mainly for epidemio­ and cardiac diseases. The organs most commonly affected
logical studies or to monitor treatment [114]. The were lung, liver, kidney, brain, spleen, eye and heart [121].
­technique of double immunodiffusion, because of its Independent risk factors that have been identified are per­
sensitivity of over 80% and specificity of over 90%, has sistently positive blood cultures for Candida spp. with a
been considered the main tool for the diagnosis of para­ central venous catheter in place, immunosuppression and
coccidioidomycosis. A latex immunoassay for the gp43 a stay in the intensive care unit) [122].
antigen of the f­ ungus has shown high diagnostic accu­
racy, low cost and reduced assay time [115]. Molecular
Clinical features and  differential diagnosis. In patients
identification of P.  brasiliensis has been successfully
with altered cell‐mediated immunity, systemic candidae­
achieved by immunohistochemistry and PCR amplifi­
mia is the most frequent presentation and has a high mor­

SECTION 8: FUNGAL SKIN


cation of ribosomal DNA.
tality if not treated promptly. The second in frequency is
chronic disseminated candidiasis (7%), followed by
Treatment and prevention. The first line of treatment is mucocutaneous candidiasis (particularly in patients with

INFECTIONS
itraconazole, followed by fluconazole and, less preferred, HIV). Patients with systemic or disseminated candidiasis
ketoconazole, until clinical remission with 50% of the present with persistent unexplained fever, various degrees
dose continued for 1 year thereafter. Sulphonamide deriv­ of multiorgan dysfunction that can progress to severe
atives can also be used, such as co‐trimoxazole every ­sepsis or shock, and skin lesions [123]. The skin lesions are
12 hours until clinical remission and 50% of the dose for commonly erythematous papules, pustules or nodules that
1 year thereafter. Voriconazole has been trialed compared often coalesce; crusty and erosive lesions can also be found
to itraconazole, with good results. Amphotericin B is the (Fig.  47.7) [121,124,125]. Patients with chronic dissemi­
drug of choice for severe or ­ recalcitrant cases nated candidiasis present with persistent fever unrespon­
[14,107,108,116]. Efforts are under way to develop a thera­ sive to broad‐spectrum antibiotics, abdominal distention
peutic vaccine using peptides, purified antigens, attenu­ and hepatosplenomegaly. The diagnosis is usually delayed
ated yeast cells and/or monoclonal antibodies [117]. as imaging studies are not always helpful [126].
Other clinical presentations include meningitis, endo­
carditis, endophthalmitis, ecthyma gangrenosum‐like
Opportunistic mycoses lesions and renal candidiasis in neonates [124,127,128].
Candidiasis The differential diagnosis should be made with bacte­
rial infections and opportunistic fungal infections such as
Introduction. Candida spp. is the most common pathogen aspergillosis, fusariosis or cryptococcosis.
causing disseminated infection in immunocompromised
children, especially those with primary immunodeficien­ Laboratory and histological findings. Direct examination
cies that involve CARD9 deficiency [118]. The most with KOH or iodide of any exudate or secretion will show
­common isolated species is C. albicans, but resistant strains abundant yeasts of 2–4 µm in diameter and pseudohy­
of C. kruzei and C. glabrata are increasing in frequency. phae or real hyphae. Culture on Sabouraud medium at
570 Section 8  Fungal Skin Infections

Cryptococcosis

Introduction.
Cryptococcosis (also known as torulosis, European blasto­
mycosis or Busse–Buschke disease) is an invasive fungal
infection caused by an encapsulated yeast, Cryptococcus neo-
formans. The organism produces infection following inhala­
tion, disseminates haematogenously, and manifests with a
great variety of signs, both cutaneous and extracutaneous.
Cryptococcus neoformans var. neoformans is associated
with immune cellular depression and has become an
increasingly prevalent pathogen among immunocompro­
mised patients. Cryptococcus neoformans var. gattii infects
immunocompetent individuals more frequently than
Fig. 47.7  Chronic disseminated candidiasis in a malnourished child. immunosuppressed hosts [132]. Mortality is high, espe­
cially when diagnosis is delayed [133,134]. Several forms of
non‐neoformans cryptococci (C. laurentii, C. albidus) have
rarely been associated with human infection [135,136].
room temperature should be done quickly, and smooth
shiny white creamy colonies that eventually become
­rugged or membranous will grow. On microscopic exami­ Epidemiology and pathogenesis. Cryptococcus neoformans
nation of the colonies, spherical or ovoid budding yeast is an encapsulated round or oval yeast, 3–10 µm in diame­
with mycelia will be found (only C. glabrata does not pro­ ter. Although more than 30 species are included in the
duce hyphae). Biochemical testing methods such as genus Cryptococcus, the pathogenic yeasts involved in
CHROMagar™ Candida are based on specific enzymes of cryptococcosis are C. neoformans var. neoformans, C. neofor-
the various species and allow the identification of the mans var. gattii and C. neoformans var. grubii. The environ­
most common species as well as other yeasts such as mental reservoir for C. neoformans is primarily pigeon
Rhodotorula, Cryptococcus, Trichosporum and Geotrichum. droppings. C. gattii is found predominantly in eucalyptus
On histopathology findings include pseudoepitheli­ and other trees such as firs and oaks. Because of its
omatous hyperplasia, granulomatous abscesses and ­environmental reservoir, infection by C. ­neoformans occurs
blastospores that are best seen with PAS, Gomori–Grocott worldwide, whereas the distribution of C. gattii and
or methenamine silver stain. C. ­grubii is more limited to tropical and subtropical regions.
There are multiple available diagnostic molecular Cryptococcosis is uncommon in infancy, with a calculated
­techniques such as specific DNA probes, electrophoresis, incidence of 1 in 100 000 in the general paediatric popula­
enzyme restriction analysis, PCR and in situ hybridiza­ tion and 47 in 100 000 in HIV‐infected children, which is
SECTION 8: FUNGAL SKIN

tion. PCR is the most sensitive analysis available and is much lower than in adults. A bimodal peak in incidence
used widely for the diagnosis of disseminated or systemic has been described, affecting children <1 year of age and
candidiasis [4]. the 5–10 years age group. Paediatric patients with HIV‐
INFECTIONS

AIDS and cryptococcosis are usually severely immuno­


compromised with low CD4 counts [137]. Cryptococcal
Treatment and  prevention. Amphotericin B remains
infection in immunocompetent children is much less
the first choice in critically ill children, at 0.6–1 mg/kg
common, even though a high proportion of children
­
per day intravenously [123]. Echinocandins such as
are  seropositive to the fungal proteins by the age of 2
caspofungin or micafungin are also used with good
[132,134,138–141]. Neonatal cryptococcosis due to vertical
results; paediatric doses are 25–50 mg/m 2 daily and
or peripartum transmission has also been reported,
3–10 mg/kg daily, respectively. After initial intensive
although it is very rare [142].
treatment, fluconazole at 6–12 mg/kg daily for several
weeks should follow [4,127]. Treatment should be
­initiated promptly after blood cultures are taken and Clinical features and differential diagnosis. The portal of
susceptibility patterns identified. Voriconazole has entry is primarily through inhalation of the fungus from
been used as salvage treatment of invasive candidiasis the environment, although direct inoculation into tissue
that was resistant to other azoles with a response rate following trauma occasionally occurs. Once the fungus
of 56% [129]. There are reports of adjunctive use of enters the human body, it can produce either latent infec­
interferon‐γ immunotherapy in cases of primary tion or acute disease. In many circumstances, the yeast
immunodeficiencies [130]. Catheters and intravascu­ can remain dormant for years in hilar lymph nodes or
lar devices should be cultured and replaced [124]. pulmonary foci in an asymptomatic individual, and then
Even with appropriate treatment, mortality remains grow and disseminate outside these sites when the local
high at 40–50%. cellular immunity is suppressed.
Vaccine development is under way, using live attenu­ Disseminated cryptococcosis with cutaneous or
ated strains, proteins, cell‐wall extracts, and combination mucocutaneous manifestations occurs in 10–15% of cases
of polysaccharides with proteins [131]. following primary pulmonary disease, especially in
Chapter 47  Deep Fungal Infections 571

immunocompromised patients. It is one of the most


Treatment and prevention. Untreated cryptococcosis is
common opportunistic infections in individuals with
­
fatal in 90% of patients. Treatment of C. neoformans and
HIV‐AIDS. Other conditions that favour the spread of
C. gattii is determined by the host’s immune status and
cryptococcosis are acute leukaemia, solid organ trans­
organ involvement and includes the use of antifungal
plantation, diabetes mellitus, connective tissue diseases,
agents, CSF drainage and occasionally surgical resec­
chronic pulmonary diseases and treatment with corticos­
tion [134].
teroids or immunosuppressive monoclonal antibodies
Fluconazole, itraconazole and amphotericin B in com­
[143–146].
bination with flucytosine (5FC) are effective therapies for
Cryptococcosis has a predilection for the central
cryptococcosis [150]. Treatment must be started early in
nervous system (CNS), producing meningitis, menin­
­
order to improve the prognosis. Another option is to
goencephalitis and abscesses of the brain. The CNS and
­combine amphotericin B and 5FC for 2 weeks and then
respiratory tract are the organs most commonly involved
continue treating with fluconazole for a minimum of
­
in C. neoformans and C. gattii infections. Other affected
10  weeks [4]. Isavuconazole has been used with good
organs include prostate, eye, bone, urinary tract, biliary
results [151,152].
tract and blood [147].
Cryptococcal infection cannot be eradicated in patients
Cutaneous cryptococcosis is the third most common
with HIV‐AIDS unless the immune status improves with
clinical presentation and can be either a primary cutane­
antiretroviral therapy. Fluconazole prophylaxis has been
ous infection from direct inoculation, or a secondary
shown to be effective for preventing cryptococcosis in
lesion as part of disseminated disease. Primary crypto­
AIDS patients who have low CD4 counts [<100 cells/μL],
coccosis manifests as a nodule or abscess after trauma
and it is also effective for preventing relapses of crypto­
[148] and secondary cryptococcosis presents mainly on
coccosis [139].
the face and scalp, with papules mimicking acne or mol­
Vaccines and/or immune therapies to induce protection
luscum contagiosum. On rare occasions, cryptococcosis
and improve the host’s immune response to C. neoformans
presents as deep abscesses or vegetating ulcerations,
are under development [139].
nodules, vesicles, ­pustules, sinuses, cellulitis and even
panniculitis [134,149].
Differential diagnosis includes molluscum contagio­ Zygomycosis
sum and acne, pyoderma gangrenosum, and other oppor­
tunistic infections, mainly systemic candidosis. Introduction. Zygomycosis infection is associated with
fungi of the Zygomycetes class and the order Mucorales
Laboratory and  histology findings. Direct microscopic (the term mucormycosis should be discouraged because
examination for the presence of encapsulated yeasts using Mucor spp. are rarely the cause of zygomycosis). Most of
an India ink preparation is a rapid and inexpensive diag­ the species involved in these infections are saprophytes
nostic test. This simple technique is 80% sensitive in that live in the soil or in plants or animals and acciden­
tally infect the human body under special conditions

SECTION 8: FUNGAL SKIN


AIDS‐related meningitis and 50% in immunocompetent
hosts [132,139]. (opportunistic infections). Zygomycosis or phycomycosis
Culture in SDA grows smooth mucoid white colonies also includes another type of mycosis, the entomophtho­
ramycoses caused by members of Entomophthoraceae

INFECTIONS
developing in 24–48 hours.
On histopathology, a dermal and hypodermal inflam­ [4]. These are rare and mostly endemic to tropical regions,
matory granulomatous infiltrate is found, with rounded not opportunistic infections; thus they will not be covered
PAS‐positive budding yeast cells with capsules, 2–20 µm in this chapter.
in size, found within clear mucoid spaces [65]. Stains such
as methenamine silver, Alcian blue or mucicarmine help Epidemiology and  pathogenesis. Opportunistic infec­
visualize the yeast cells [4]. tions due to the Mucorales are caused by species of
Cryptococcus polysaccharide capsular antigen can be Mucor, Rhizopus, Absidia, Rhizomucor, Saksenaea, Mortierella,
detected in most fluids and tissues of the body by a latex Cunninghamella and Syncephalastrum. These fungi are
agglutination test. The sensitivity of this test is reported to ubiquitous in nature and can be found in vegetation,
be 93–100% and it has a specificity of 93–98%. A latex bread and soil. Classically, they infect patients with dia­
agglutination test in CSF or serum is rapid, specific, non­ betic acidosis [153], extensive burns and/or malnutrition,
invasive and virtually diagnostic of meningoencephalitic but in the past decades they have become an increasing
or disseminated cryptococcosis if high titres are found. cause of infection in patients with neutropenia due to
Latent asymptomatic individuals can be discovered ­haematological malignancies who are treated prophylac­
through positive cryptococcal skin testing for delayed tically with voriconazole against aspergillosis [154]. They
hypersensitivity to Cryptococcus or through a serological are also more common in patients with corticosteroid or
test for the fungus. Serum cryptococcal antibodies are not immunosuppressive therapy, organ transplantation and
helpful in diagnosing and deciding treatment for crypto­ HIV‐AIDS [155], and even in immunocompetent patients
coccosis, because of the poor sensitivity and specificity of [156–158]. They affect both genders and all ages equally
this method. Its positivity can appear later during the [159]. Along with aspergillosis, they are the second or
treatment, or it can be negative in patients with disease third most frequent fungal infection after candidiasis in
confined to the skin [134,139]. immunosuppressed patients [156,160]. The most common
572 Section 8  Fungal Skin Infections

route of infection is through inhalation of spores but, in inflammation, necrosis, vascular invasion and different
diabetic acidosis and burns, infection can develop under degrees of thrombosis, and collections of broad, ribbon‐
occlusive bandages or after trauma; and there have been like, nonseptated hyphae branching at 90° angles
outbreaks in hospitals and large institutions [161,162]. (Fig. 47.9). Tissue should be sent for direct examination
The most frequently observed organisms are Rhizopus and fungal culture in order to identify the specific
arrhizus, R. oryzae and R. microsporus, Absidia corymbifera, fungus causing the infection and its susceptibility
­
A. ramosa and Mucor pusillus [159,163,164]. ­patterns. Culture of infected tissue in Sabouraud’s agar
grows colonies in 1–5 days. The diagnosis by culture
Clinical features and differential diagnosis. Opportunistic alone is difficult because of the widespread nature of
zygomycosis caused by Mucorales is a serious, fulminant these agents as contaminants [172].
infection responsible for rapid death in susceptible hosts.
It can affect many different organs: rhinocerebral, pulmo­ Treatment and prevention. Treatment should be started
nary, gastrointestinal, cutaneous or disseminated. The promptly. Ideally it should combine surgical debridement
rhinocerebral form is the most common; infection starts at and antifungal treatment with intravenous amphotericin
the nasal mucosa and then disseminates to the orbit and B or posaconazole [153,159]. Isavuconazole has been tried
brain. Necrotic ulcers of the palate are an important sign, and has comparable results with amphotericin B [173].
as is facial cellulitis. The fungi have a predilection to The echinocandins and other triazoles such as ravucona­
invade blood vessels, leading to thrombosis, haemor­ zole and voriconazole are considered inactive against
rhage, necrosis and infarction [159,163,165]. Mucorales [174]; however, there are reports of successful
The primary cutaneous form usually follows inocula­ combined treatment with caspofungin and posaconazole
tion or contact with contaminated material after trauma, or amphotericin B, suggesting that they have additional
natural disasters or accidents. It is a rapidly progressive or synergistic antimucoral effects [175]. Relapses can
infection characterized by tissue destruction and blood occur after successful treatment [176]. It is necessary to
vessel invasion. Skin lesions are usually tender erythema­ simultaneously correct the predisposing abnormalities
tous or purpuric nodules or patches that rapidly progress and monitor patients closely.
to necrosis and invade peripheral tissues (Fig. 47.8) [166]. Zygomycosis caused by Mucorales is a serious, life‐
Haematogenous dissemination is not rare, and the threatening disorder that leads to death in most
mortality rate, although lower than in other forms of
­ ­disseminated cases. Early diagnosis is the cornerstone of
mucormycosis, is high even after treatment has been successful treatment, increasing survival from approxi­
started (up to 32%) [155,156,167]. mately 40–80% [177].
Rare presentations such as acute bilateral renal artery
thrombosis in a healthy young adult [168], a pulsatile Aspergillosis
nodule [169], renal mass [170] and mediastinal mass have
been described [171]. Introduction. Aspergillus spp. are ubiquitous fungi that
SECTION 8: FUNGAL SKIN

The differential diagnosis is with pyoderma gangreno­ very frequently cause opportunistic infections in
sum, septicaemia, bacterial cellulitis, necrotizing fasciitis immunocompromised patients, especially those with
and meningitis. leukaemias and lymphomas. It is the second most
INFECTIONS

­frequent opportunistic fungal infection, after candidia­


Laboratory and  histological findings. The diagnosis is sis. In immunocompetent people, Aspergillus spp. can
made histologically from a skin or tissue biopsy. induce allergic responses such as sinusitis and asthma
The  ­
typical findings are suppurative granulomatous [178,179].

Fig. 47.9  Skin biopsy stained with Gomori’s methenamine silver showing


Fig. 47.8  Purpuric plaque that rapidly progressed into a necrotic ulcer in a broad, ribbon‐like, nonseptated hyphae branched at 90° angles typical of
child with acute leukaemia and zygomycosis. Mucorales fungi.
Chapter 47  Deep Fungal Infections 573

Epidemiology and  pathogenesis. Members of the Laboratory and  histological findings. Clinical suspicion
Aspergillus genus can be found in fomites, the atmosphere is crucial, but the definitive diagnosis is histological and
and organic materials, with a worldwide distribution. mycological through culture of tissue. On histological
Aspergillus fumigatus is the most common species isolated examination of a biopsy stained with Gomori’s methena­
from cultures (50–60%), followed by A. flavus, A. niger and mine silver, septate hyphae that branch at acute angles are
A. terreus (10–15% each); other rare Aspergillus spp. repre­ found within a suppurative granulomatous infiltrate and
sent less than 2% of isolates. Inhalation is the most com­ the deep arterial vessels of the skin; however, sensitivity is
mon route of entry of Aspergillus spores. Primary cutaneous not high. Immunoperoxidase staining using the monoclo­
aspergillosis is rare, following either direct physical nal antibody EB‐A1 on formalin‐fixed, paraffin‐embedded
­inoculation or occlusive dressing at the site of indwelling tissue permits detection of Aspergillus spp. even when
catheters. More often the skin is involved due to haema­ cultures remain negative. Aspergillus grows rapidly in
­
togenous dissemination from a pulmonary focus. ­cultures in SDA, but this is not conclusive because of fre­
Aspergillosis has emerged as an important morbid­ quent contamination with this ubiquitous agent. Thus,
ity‐ and mortality‐causing agent in immunocompro­ diagnosis must rely on noninvasive and indirect methods
mised children with deficiencies of phagocytic cells, such as fungal antigen detection and amplification of fun­
especially those receiving myelosuppressive antineo­ gal nucleic acid sequences. PCR assays have a sensitivity
plastic chemotherapy for acute leukaemias, recipients and specificity of 84%, but these increase when at least two
of haematopoietic stem cell or solid organ transplants, positive specimens are used [194]. Aspergillus antigen
or patients with primary immunodeficiencies such as detection was a synonym for the galactomannan ELISA
chronic granulomatous disease [178–184]. Large studies until the modern 1,3 β‐glucan test was developed.
report that up to 75% of paediatric invasive aspergillo­ Circulating galactomannan antigens may be present in the
sis cases occur in this setting, increasing mortality rates blood in detectable levels very early in the course of inva­
from 1% to 21% [184]. Secondary, acquired or induced sive aspergillosis in immunosuppressed patients and can
deficiencies of T‐cell‐dependent defence mechanisms be detected by enzyme immunoassay in serum [184].
and comorbidities such as cytomegalovirus disease are Specific DNA can be detected by PCR assay
also involved but appear to be less important [184]. [178,179,182,189,195]. There are conflicting results con­
Patients with cystic fibrosis have increased rates of cerning sensitivity and specificity of all these assays in
allergic bronchopulmonary aspergillosis and Aspergillus children because data are limited, but the galactomannan
bronchitis [185]. assay has acceptable sensitivity and specificity [181,184].
Other studies include pulmonary CT scan, which in
Clinical features and  differential diagnosis. Invasive children should be used selectively, and bronchoalveolar
aspergillosis principally involves the respiratory tract. lavage.
Facial swelling, epistaxis, proptosis, cranial nerve abnor­

SECTION 8: FUNGAL SKIN


malities or bone erosion are signs of fungal sinusitis. Treatment and prevention. Mortality is high if aspergillosis
Fever, cough and dyspnoea are frequent nonspecific find­ is not treated promptly, so treatment should be started
ings of pulmonary aspergillosis. Poorly controlled based on risk profile and available diagnostic testing. The

INFECTIONS
Aspergillus infection may lead to extension to local struc­ gold standard of systemic antifungal treatment is voricona­
tures and haematogenous dissemination that can involve zole 8–14 mg/kg per day divided in two doses as the
any organ. ­primary therapy for invasive aspergillosis [189] or ampho­
Cutaneous aspergillosis can be primary, secondary tericin B 1–1.5 mg/kg per day for at least 6–12 weeks to
through haematogenous spread from a distant focus (up avoid recurrence [182]. For children under 2 years of age,
to 11% of these patients have skin manifestations) or only liposomal amphotericin B is approved at 3 mg/kg per
affected by spread from contiguous tissues (e.g. nasal day. Choices for second‐line treatment in patients older
­septum abscesses). Lesions usually arise around catheters, than 2 years include amphotericin B lipid complex, ampho­
oxygen saturation monitors, intravenous lines or sites that tericin B colloidal dispersion and caspofungin [184].
were covered with tape or dressings. Clinically, lesions are Combination therapy with two antifungal agents such as
erythematous plaques or nodules that can develop into voriconazole and liposomal amphotericin B has shown
necrotic eschars or purpuric bullae. Disseminated second­ good results with higher survival rates [196]. However, the
ary aspergillosis of the skin is similar to meningococcae­ use of voriconazole significantly increases the risk of cuta­
mia, with scattered purpuric nodules and papules over neous squamous cell carcinoma, and the risk increases as
the entire surface of the body [121,186–189]. the duration of the treatment increases, so special attention
Some less frequent clinical presentations of aspergillo­ should be given to these patients and they should be coun­
sis are laryngeal [190], orbital [180], bronchial following selled appropriately [197]. Prophylactic treatment with
foreign body aspiration [191], osteomyelitis [192] and posaconazole or itraconazole significantly improves sur­
otomycosis [193]. vival and reduces the risk of invasive aspergillosis in
Aspergillosis must be differentiated from other dis­ patients with leukaemia and post stem cell transplantation
eases causing nodules and ulceration, such as bacterial [182,184,195]. Early debridement of localized aspergillosis,
septicaemia, systemic candidosis, nocardiosis, fusariosis, such as sinusitis, cutaneous disease and osteomyelitis,
zygomycosis and vasculitis. should be performed for a better prognosis. Invasive
574 Section 8  Fungal Skin Infections

a­ spergillosis caused by A. terreus has a worse prognosis


Epidemiology and  pathogenesis. Actinomycosis is a
than non‐terreus species, regardless of treatment used [198].
ubiquitous disease. Its incidence has decreased due to
better oral hygiene. It affects both genders with a slight
Nocardiosis predominance in females, and all ages equally.
Predisposing factors include dental procedures, dental
Introduction. Nocardiosis is a disease caused by aerobic cavities, intrauterine devices and bad dental hygiene. The
actinomycetes, particularly Nocardia asteroides, N. brasilien- most frequently implicated agent is Actinomyces israelii
sis and N. otitidis‐caviarum. Primary cutaneous nocardiosis (80%); less frequent are A. naeslundii, A. viscosus, A. odon-
is rare in both children and adults, and is usually associ­ tolyticus, Arachnia propinica and Bifidobacterium eriksonii
ated with an underlying immunological condition, [4,14,201].
although it can present in healthy patients [199].
Clinical features and  differential diagnosis. The infec­
Epidemiology and pathogenesis. Nocardiosis is a cosmo­ tion may involve both bone and soft tissues. There are
politan disease, affecting mainly adult males, and is ­different types according to the location: cervicofacial,
acquired by inhalation or through cutaneous inoculation. pulmonary, abdominal and pelvic.
There is usually a history of trauma, and the interval Cervicofacial actinomycosis is the most frequent. It is
between this and the appearance of the lesion is very vari­ rare in childhood but several cases have been reported.
able. Nocardiosis affects healthy individuals and those who Trauma or dental disease can usually be implicated. In
are immunodeficient due to any cause. The most common children tooth eruption may provide a portal of entry and
species isolated are N. asteroides and N. brasiliensis [4,199,200]. infection spreads slowly to contiguous tissue. It is charac­
terized by an increase in volume, tissue deformity and
Clinical features and  differential diagnosis. Clinical sinus tract formation with purulent discharge that con­
manifestations are primarily pulmonary as acute or tains parasitic forms called ‘sulphur granules’ (Fig. 47.10).
chronic pneumonia in 70%. Dissemination to the CNS The pulmonary form manifests as fever, cough and
occurs in one quarter of cases and causes brain abscesses expectoration. The abdominal and pelvic forms are com­
or meningeal symptoms. Skin lesions are nodules or monly mistaken for appendicitis, and in chronic cases the
plaques with abscesses and fistulae. Primary cutaneous skin is involved with abscesses and purulent draining
nocardiosis is exceptional and occurs after trauma. It is sinuses [4,14,201].
characterized by pustules or nodules that spread with a The differential diagnosis should be made with tuber­
lymphangitic (sporotrichoid) pattern away from the inoc­ culosis, mycetoma, lymphomas and odontogenic sinus
ulation site. They are very tender and inflammatory, and tracts.
the patients are frequently febrile [4,199,200].
Laboratory and  histological findings. The diagnosis
Laboratory and  histological findings. The diagnosis is can be established through a direct examination of the
SECTION 8: FUNGAL SKIN

made by direct examination of the discharge from lesions exudate and visualization of the sulphur granules,
or sputum with Gram and Ziehl–Neelsen stains, where which are multilobulated masses of yellow‐white
partially acid‐fast filamentous branching structures will micromycelia. A biopsy should be stained with Gram,
INFECTIONS

be found. Culture of material obtained from the lesions in PAS or Gomori–Grocott as in all chronic, purulent and
SDA yields characteristic white cerebriform colonies. granulomatous processes. The granules can be distin­
Histologically, there is pseudoepitheliomatous hyperpla­ guished from Nocardia spp. with Ziehl–Neelsen stain
sia and suppurative granulomatous inflammation. With because Actinomyces granules are not acid fast. Culture
Gram and Ziehl–Neelsen stains the filaments of Nocardia confirms the species but must be done in an anaerobic
spp. are demonstrated [4,199,200]. medium [4,14].
The differential diagnosis includes pulmonary tuberculo­
sis, blastomycosis, coccidioidomycosis, and histoplasmosis;
cutaneous lesions can be confused with actinomycosis,
mycetoma and botryomycosis.

Treatment and prevention. The treatment of choice is co‐


trimoxazole in doses of 10 mg/kg per day for 3 months.
Other antibiotics employed are minocycline, amikacin,
cephalexin, erythromycin and imipenem [4,199,200].

Actinomycosis

Introduction. Actinomycosis is a chronic, suppurative


endogenous infection caused by filamentous anaerobic
actinomycetes that reside in the oral mucosa, dental cavi­ Fig. 47.10  Increased volume and tumefaction of the right mandibular
ties, tonsil crypts and vagina. area, characteristic of cervicofacial actinomycosis.
Chapter 47  Deep Fungal Infections 575

27 Ameen M. Chromoblastomycosis: clinical presentation and manage­


Treatment and  prevention. The treatment of choice for ment. Clin Exp Dermatol 2009;34:849–54.
actinomycosis is 4–6 weeks of penicillin, clindamycin 28 Hofmann H, Choi SM, Wilsmann‐Theis D et al. Invasive chromoblas­
tomycosis and sinusitis due to Phialophora verrucosa in a child from
being a second choice, for several weeks to avoid recur­
northern Africa. Mycoses 2005;48:456–61.
rence. Other options are co‐trimoxazole, minocycline, 29 van de Sande WW. Global burden of human mycetoma: a systematic
amoxicillin‐clavulanate or cephalosporins [4,14,201]. review and meta‐analysis. PLoS Negl Trop Dis 2013;7:e2550.
30 Fahal A, Mahgoub el S, El Hassan AM et  al. Head and neck myce­
toma: the Mycetoma Research Centre experience. PLoS Negl Trop Dis
References 2015;9:e0003587.
1 Hay RJ. Fungal infections. Clin Dermatol 2006;24:201–12. 31 Estrada R, Chavez‐Lopez G, Estrada‐Chavez G et al. Eumycetoma.
2 Lupi O, Tyring SK, McGinnis MR. Tropical dermatology: fungal Clin Dermatol 2012;30:389–96.
tropical diseases. J Am Acad Dermatol 2005;53:931–51. 32 Ameen M, Arenas R. Developments in the management of myceto­
3 Arenas R, Moreno‐Coutino G, Welsh O. Classification of subcuta­ mas. Clin Exp Dermatol 2009;34:1–7.
neous and systemic mycoses. Clin Dermatol 2012;30:369–71. 33 Hussain Mirza S, Hussain Gardezi A, Khan Y, Wiqar MA.
4 Arenas R. Illustrated Medical Micology. Mexico: McGraw‐Hill, Subcutaneous facial mycosis in a child due to Madurella mycetoma­
2008:401 pp. tis. J Pak Med Assoc 2007;57[9]:466–8.
5 Vasquez‐del‐Mercado E, Arenas R, Padilla‐Desgarenes C. 34 Fahal AH, Sabaa AH. Mycetoma in children in Sudan. Trans R Soc
Sporotrichosis. Clin Dermatol 2012;30:437–43. Trop Med Hyg 2010;104:117–21.
6 Pappas PG, Téllez I, Deep AE et al. Sporotrichosis in Peru: Description 35 Frey D, Lewis MB. Mycetoma of the scalp in an aboriginal child.
of an area of hyperendemicity. Clin Infect Dis 2000;30:65–70. Australas J Dermatol 1976;17:7–8.
7 da Rosa AC, Scroferneker ML, Vettorato R et al. Epidemiology of 36 Chiapello LS, Dib MD, Nuncira CT et al. Mycetoma of the scalp due
sporotrichosis: a study of 304 cases in Brasil. J Am Acad Dermatol to Microsporum canis: hystopathologic, mycologic, and immunoge­
2005;52:451–9. netic features in a 6‐year‐old girl. Diagn Microbiol Infect Dis
8 Dooley DP, Bostic PS, Beckius ML. Spook house sporotrichosis. A 2011;70:145–9.
point‐source outbreak of sporotrichosis associated with hay bale props 37 Joshi A, Acharya S, Anehosur VS et al. Oral eumycetoma of infancy: a rare
in a Halloween haunted‐house. Arch Intern Med 1997;157:1885–7. presentation and a brief review. J Craniomaxillofac Surg 2014;42:35–40.
9 Barros MB, Costa DL, Schubach TM et  al. Endemic of zoonotic 38 El‐Sobky TA, Haleem JF, Samir S. Eumycetoma osteomyelitis of the
sporotrichosis: profile of cases in children. Pediatr Infect Dis J calcaneus in a child: a radiologic‐pathologic correlation following
2008;27:246–50. total calcanectomy. Case Rep Pathol 2015;2015:129020.
10 Bastos de Lima Barros M, de Oliverira Schubach A, Gutierrez 39 Welsh O, Vera‐Cabrera L, Welsh E, Salinas MC. Actinomycetoma
Galhardo MC, et  al. Sporotrichosis with widespread cutaneous and advances in its treatment. Clin Dermatol 2012;30:372–81.
lesions: report of 24 cases related to transmission by domestic cats in 40 Bonifaz A, Ibarra G, Saul A et al. Mycetoma in children: experience
Rio de Janeiro, Brazil. Int J Dermatol 2003;42:677–81. with 15 cases. Pediatr Infect Dis J 2007;26:50–2.
11 Silva MB, Costa MM, Torres CC et  al. [Urban sporotrichosis: a
41 Munoz F, García M, Mayorga J, Barba J. Micetomas. Estudio epide­
neglected epidemic in Rio de Janeiro, Brazil]. Cad Saude Publica miologico de 13 anos en el occidente de Mexico [1981‐1993]. Rev Mex
2012;28:1867–80. Dermatol 1999;39:13–17.
12 Mata‐Essayag S, Delgado A, Colella MT et al. Epidemiology of sporo­ 42 García‐Vargas A, Navarro C, Mayorga JA et al. Minimicetoma en un
trichosis in Venezuela. Int J Dermatol 2013;52:974–80. pre‐escolar mexicano. Reporte de un caso y revisión de la literatura.
13 Ramos‐e‐Silva M, Vasconcelos C, Carneiro S, Cestari T. Dermatol Pediatr Lat 2004;2:54–8.
Sporotrichosis. Clin Dermatol 2007;25:181–7. 43 Perez‐Elizondo AD, del Pino‐Rojas GT, Bonifaz A. Make your diagno­
14 Arenas R, Estrada R. Tropical Dermatology. Georgetown, TX: sis. Rev Argent Dermatol 2011;92.
Landes Bioscience, 2001. 44 Ameen M, Arenas R, Vasquez del Mercado E et al. Efficacy of imipe­
15 Freitas DF, Lima IA, Curi CL et al. Acute dacryocystitis: another clini­ nem therapy for Nocardia actinomycetomas refractory to sulfona­

SECTION 8: FUNGAL SKIN


cal manifestation of sporotrichosis. Mem Inst Oswaldo Cruz mide. J Am Acad Dermatol 2010;62:239–46.
2014;109:262–4. 45 Ho AK, Shrader MW, Falk MN, Segal LS. Diagnosis and initial man­
16 Lee H, Kim DY, Lee KH et  al. Deformity of the earlobe caused by agement of musculoskeletal coccidioidomycosis in children. J Pediatr
fixed cutaneous sporotrichosis in a pediatric patient. Int J Dermatol Orthoped 2014;34:571–7.

INFECTIONS
2015;54:e187–9. 46 Waterman BR, Waterman SM, McCoy AC, Cameron CD. Coccidioidal
17 Zhang Y, Pyla V. Nasal sporotrichosis in children. Int J Dermatol osteomyelitis of the patella. Orthopedics 2010;33.
2014;53:e342–3. 47 Canteros CE, Toranzo A, Ibarra‐Camou B et al. [Coccidioidomycosis
18 Zhang Y, Pyla V. Palpebral sporotrichosis. Int J Dermatol 2014;53:e356–7. in Argentina, 1892‐2009]. Rev Argent Microbiol 2010;42:261–8.
19 Ribeiro BN, Ribeiro RN, Penna CR, Frota AC. Bone involvement by 48 Baptista Rosas RC, Riquelme M. [The epidemiology of coccidioido­
Sporothrix schenckii in an immunocompetent child. Pediatr Radiol mycosis in Mexico]. Rev Iberoam Micol 2007;24:100–5.
2015;45:1427–30. 49 Twarog M, Thompson GR. Coccidioidomycosis: Recent updates.
20 Bernardes‐Engemann AR, de Lima Barros M, Zeitune T et  al. Semin Respir Crit Care Med 2015;36:746–55.
Validation of a serodiagnostic test for sporotrichosis: a follow‐up 50 McCarty JM, Demetral LC, Dabrowski L et al. Pediatric coccidioido­
study of patients related to the Rio de Janeiro zoonotic outbreak. Med mycosis in central California: a retrospective case series. Clin Infect
Mycol 2015;53:28–33. Dis 2013;56:1579–85.
21 Yamada K, Zaitz C, Framil VM, Muramatu LH. Cutaneous sporotri­ 51 Cid Chavez DM, Ruiz Pedraza MD, Sanchez Sanchez LM et  al.
chosis treatment with potassium iodide: a 24 year experience in Sao [Clinical and immunological characteristics in pediatric patients
Paulo State, Brazil. Rev Inst Med Trop Sao Paulo 2011;53:89–93. with coccidioidomycosis in northeastern Mexico]. Gac Med Mex
22 Macedo PM, Lopes‐Bezerra LM, Bernardes‐Engemann AR, Orofino‐ 2013;149:541–7.
Costa R. New posology of potassium iodide for the treatment of cuta­ 52 Fisher BT, Chiller TM, Prasad PA et al. Hospitalizations for coccidioi­
neous sporotrichosis: study of efficacy and safety in 102 patients. J Eur domycosis at forty‐one children’s hospitals in the United States.
Acad Dermatol Venereol 2015;29:719–24. Pediatr Infect Dis J 2010;29:243–7.
23 Ramirez Soto MC. Effects of potassium iodide in concentrations of 53 Chu JH, Feudtner C, Heydon K et  al. Hospitalizations for endemic
TSH, tT3 and tT4 in serum of subjects with sporotrichosis. Mycoses mycoses: a population‐based national study. Clin Infect Dis
2014;57:460–5. 2006;42:822–5.
24 Perez‐Blanco M, Hernandez Valles R, Garcia‐Humbria L, Yegres F. 54. Barnato AE, Sanders GD, Owens DK. Cost‐effectiveness of a potential
Chromoblastomycosis in children and adolescents in the endemic vaccine for Coccidioides immitis. Emerg Infect Dis 2001;7:797–806.
area of the Falcon State, Venezuela. Med Mycol 2006;44:467–71. 55 Sondermeyer G, Lee L, Gilliss D et al. Coccidioidomycosis‐associated
25 Pradeepkumar NS, Joseph NM. Chromoblastomycosis caused by hospitalizations, California, USA, 2000‐2011. Emerg Infect Dis
Cladophialophora carrionii in a child from India. J Infect Dev Ctries 2013;19:1590–7.
2011;5:556–60. 56 Hector RF, Rutherford GW, Tsang CA et al. The public health impact
26 Torres‐Guerrero E, Isa‐Isa R, Isa M, Arenas R. Chromoblastomycosis. of coccidioidomycosis in Arizona and California. Int J Environ Res
Clin Dermatol 2012;30:403–8. Public Health 2011;8:1150–73.
576 Section 8  Fungal Skin Infections

57 Bravo R, Pelayo‐Katsanis LO, Shehab ZM, Katsanis E. Diagnostic and 88 Zahn M, Hesson M, Morton R, Wheat LJ. Granulomatous pleuritis
treatment challenges for the pediatric haematologist oncologist in caused by histoplasmosis in a healthy child. Pediatr Pulmonol
endemic areas for coccidioidomycosis. J Pediatr Hematol Oncol 2011;46:729–31.
2012;34:389–94. 89 Riddell Jt, Kauffman CA, Smith JA et  al. Histoplasma capsulatum
58 La Via WV, Koulouri S, Ross LA et al. Right atrial mass in a child with endocarditis: multicenter case series with review of current diagnos­
disseminated coccidioidomycosis. Pediatr Infect Dis J 2005;24:470–2. tic techniques and treatment. Medicine 2014;93:186–93.
59 Sheppard JE, Switlick DN. Coccidioides immitis osteomyelitis of the 90 Kauffman CA. Histoplasmosis: a clinical and laboratory update.
radius presenting as Ewing’s sarcoma. Orthopedics 2008;31:607. Clin Microbiol Rev 2007;20:115–32.
60 Torres‐Najera M, de la Garza‐Galvan S, Cerda‐Flores RM et  al. 91 Vega W, Almeida R, Mino G et al. A quick and cost‐effective method
[Osteoarticular coccidioidomicosis. Clinical and pathological study of for diagnosing disseminated histoplasmosis in children. Diagn
36 Mexican patients]. Rev Invest Clin 2006;58:211–16. Microbiol Infect Dis 2007;57:405–8.
61 Char DH, Crawford JB, Bertolucci G, Cole T. Intraocular coccidioido­ 92 Dhawan J, Verma P, Sharma A et al. Disseminated cutaneous histo­
mycosis simulating a neoplasm. Br J Ophthalmol 2012;96:218–19. plasmosis in an immunocompetent child, relapsed with itracona­
62 Stanga SD, Dajud MV. Visual changes in a 4‐year‐old. Clin Pediatr zole, successfully treated with voriconazole. Pediatr Dermatol
2008;47:959–61. 2010;27:549–51.
63 Smith G, Hoover S, Sobonya R, Klotz SA. Abdominal and pelvic 93 Castillo CG, Kauffman CA, Micelli MH. Blastomycosis. Infect Dis
­coccidioidomycosis. Am J Med Sci 2011;341:308–11. Clin North Am 2016;30:247–64.
64 El Dib NA, Eldessouky NM, El Sherbini SA et al. Disseminated coccidioi­ 94 Roy M, Benedict K, Deak E et  al. A large community outbreak of
domycosis in a 5‐year‐old Sudanese boy. J Trop Pediatr 2014;60:260–3. blastomycosis in Wisconsin with geographic and ethnic clustering.
65 Fernandez‐Flores A, Saeb‐Lima M, Arenas R. Morphological find­ Clin Infect Dis 2013;57:655–62.
ings  of deep cutaneous fungal infections. Am J Dermatopathol 95 Frost HM, Anderson J, Ivacic L, Meece J. Blastomycosis in chil­
2014;36:531–56. dren: an analysis of clinical, epidemiologic, and genetic features.
66 Saitoh A, Homans J, Kovacs A. Fluconazole treatment of coccidioidal J Pediatric Infect Dis Soc 2017;6:49–56.
meningitis in children: two case reports and a review of the literature. 96 Meece JK, Anderson JL, Gruszka S et al. Variation in clinical pheno­
Pediatr Infect Dis J 2000;19:1204–8. type of human infection among genetic groups of Blastomyces
67 Homans JD, Spencer L. Itraconazole treatment of nonmeningeal ­dermatitidis. J Infect Dis 2013;207:814–22.
­coccidioidomycosis in children: two case reports and review of the 97 Dworkin MS, Duckro AN, Proia L et al. The epidemiology of blasto­
literature. Pediatr Infect Dis J 2010;29:65–7. mycosis in Illinois and factors associated with death. Clin Infect Dis
68 Galgiani JN, Catanzaro A, Cloud GA et  al. Comparison of oral 2005;41:e107–11.
­fluconazole and itraconazole for progressive, nonmeningeal coccidi­ 98 Pelly L, Al Juaid A, Fanella S. Severe blastomycosis in infants.
oidomycosis. A randomized, double‐blind trial. Ann Intern Med Pediatr Infect Dis J 2014;33:1189–91.
2000;133:676–86. 99 Abdalla M, Sax PE, Mostaghimi A et al. Clinical problem‐solving. On
69 Yorgin PD, Rewari M, al‐Uzri AY et al. Coccidioidomycosis in adoles­ the nose. N Engl J Med 2015;373:955–61.
cents with lupus nephritis. Pediatr Nephrol 2001;16:77–81. 100 Taub AL, Nelsen DD, Nasser R, Stratman EJ. Perianal North
70 Schein R, Homans J, Larsen RA, Neely M. Posaconazole for chronic American blastomycosis. Cutis 2015;96:E23–6.
refractory coccidioidal meningitis. Clin Infect Dis 2011;53:1252–4. 101 Brick KE, Drolet BA, Lyon VB, Galbraith SS. Cutaneous and dis­
71 Levy ER, McCarty JM, Shane AL, Weintrub PS. Treatment of pediatric seminated blastomycosis: a pediatric case series. Pediatr Dermatol
refractory coccidioidomycosis with combination voriconazole and 2013;30:23–8.
caspofungin: a retrospective case series. Clin Infect Dis 2013;56:1573–8. 102 Johnson RR, Vora SS. Blastomycotic multifocal arthritis and osteo­
72 Connelly MB, Zerella JT. Surgical management of coccidioidomycosis myelitis in the urban setting. J Rheumatol 2013;40:1627–9.
in children. J Pediatr Surg 2000;35:1633–4. 103 Head AJ, Myers LK, Thompson JD et al. Disseminated blastomycosis
73 Kauffman CA. Histoplasmosis. Clin Chest Med 2009;30:217–25. presenting as oligoarticular septic arthritis in a 12‐year‐old girl.
74 Wheat LJ, Freifeld AG, Kleiman MB et al. Clinical practice guide­ Arthritis Rheum 2005;53:138–41.
lines for the management of patients with histoplasmosis: 2007 104 Mongkolrattanothai K, Peev M, Wheat LJ, Marcinak J. Urine antigen
SECTION 8: FUNGAL SKIN

update by the Infectious Diseases Society of America. Clin Infect detection of blastomycosis in pediatric patients. Pediatr Infect Dis J
Dis 2007;45:807–25. 2006;25:1076–8.
75 Wheat LJ, Kauffman CA. Histoplasmosis. Infect Dis Clin North Am 105 Chapman SW, Dismukes WE, Proia LA et al. Clinical practice guidelines
2003;17:1–19, vii. for the management of blastomycosis: 2008 update by the Infectious
INFECTIONS

76 Kauffman CA. Endemic mycoses: blastomycosis, histoplasmosis, and Diseases Society of America. Clin Infect Dis 2008;46:1801–12.
sporotrichosis. Infect Dis Clin North Am 2006;20:645–62, vii. 106 Bellissimo‐Rodrigues F, Machado AA, Martinez R. Paracoccidioido­
77 Adderson EE. Histoplasmosis. Pediatr Infect Dis J 2006;25:73–4. mycosis epidemiological features of a 1,000‐cases series from a
78 Butler JC, Heller R, Wright PF. Histoplasmosis during childhood. hyperendemic area on the southeast of Brazil. Am J Trop Med Hyg
South Med J 1994;87:476–80. 2011;85:546–50.
79 Arango M, Castaneda E, Agudelo CI et al. Histoplasmosis: results of 107 Batista da Matos W, Campelo dos Santos GM, Boucas da Silva VE
the Colombian national survey, 1992‐2008. Biomedica 2011;31:344–56. et al. Paracoccidioidomycosis in the State of Maranhão, Brazil: geo­
80 Haselow DT, Safi H, Holcomb D et al. Histoplasmosis associated with graphical and clinical aspects. Rev Soc Bras Med Trop 2012;45:385–9.
a bamboo bonfire‐‐Arkansas, October 2011. MMWR 2014;63:165–8. 108 Ramos‐E‐Silva M, Saraiva Ldo E. Paracoccidioidomycosis.
81 Ashbee HR, Evans EG, Viviani MA et al. Histoplasmosis in Europe: Dermatol Clin 2008;26:257–69, vii.
report on an epidemiological survey from the European Confederation 109 Bellissimo‐Rodrigues F, Bollela VR, Da Fonseca BA, Martinez R.
of Medical Mycology Working Group. Med Mycol 2008;46:57–65. Endemic paracoccidioidomycosis: relationship between clinical presen­
82 Mata‐Essayag S, Colella MT, Rosello A et al. Histoplasmosis: a study tation and patients’ demographic features. Med Mycol 2013;51:313–18.
of 158 cases in Venezuela, 2000‐2005. Medicine 2008;87:193–202. 110 Nascimento CR, Delanina WF, Soares CT. Paracoccidioidomycosis:
83 Kauffman CA, Freifeld AG, Andes DR et  al. Endemic fungal infec­ sarcoid‐like form in childhood. An Bras Dermatol 2012;87:486–7.
tions in solid organ and haematopoietic cell transplant recipients 111 Taicz M, Rosanova MT, Bes D et  al. [Paracoccidioidomycosis in
enrolled in the Transplant‐Associated Infection Surveillance Network pediatric patients: a description of 4 cases]. Rev Iberoam Micol
[TRANSNET]. Transpl Infect Dis 2014;16:213–24. 2014;31:141–4.
84 Yilmaz GG, Yilmaz E, Coskun M et al. Cutaneous histoplasmosis in a 112 da Costa Carneiro R, Grassi de Miranda B, Neto CC et al. Juvenile
child with hyper‐IgM. Pediatr Dermatol 1995;12:235–8. paracoccidioidomycosis in urban area: report of two cases. Bras J
85 Hunstad DA, French AR. Histoplasmosis in a child with JRS on low‐ Infect Dis 2010;14:77–80.
dose methotrexate. Rheumatology (Oxford) 2007;46:177–8. 113 Sposto MR, Scully C, de Almeida OP et al. Oral paracoccidioidomy­
86 Nino‐Serna L, Restrepo‐Gouzy A, Garces‐Samudio C, Trujillo‐ cosis. A study of 36 South American patients. Oral Surg Oral Med
Honeysberg M. [Cerebral histoplasmosis in immunocompetent chil­ Oral Pathol 1993;75]:461–5.
dren]. Rev Neurol 2013;56:444–6. 114 Magalhaes EM, Ribeiro Cde F, Damaso CS et al. Prevalence of para­
87 Schuster JE, Wushensky CA, Di Pentima MC. Chronic primary central coccidioidomycosis infection by intradermal reaction in rural areas
nervous system histoplasmosis in a healthy child with intermittent in Alfenas, Minas Gerais, Brazil. Rev Inst Med Trop Sao Paulo
neurological manifestations. Pediatr Infect Dis J 2013;32:794–6. 2014;56:281–5.
Chapter 47  Deep Fungal Infections 577

115 Dos Santos PO, Rodrigues AM, Fernandes GF et al. Immunodiagnosis 144 Chopra S, Capoor MR, Mallik R et al. Pulmonary Cryptococcosis in HIV‐
of paracoccidioidomycosis due to Paracoccidioides brasiliensis sero‐negative patients: case series from India. Mycoses 2015;58:288–93.
using a latex test: detection of specific antibody anti‐gp43 and 145 Martinez‐Longoria CA, Rubio‐Perez NE, Rios‐Solis JE, Garcia‐
­specific antigen gp43. PLoS Negl Trop Dis 2015;9:e0003516. Rodriguez F. Severe meningoencephalitis co‐infection due to
116 Shikanai‐Yasuda MA. Paracoccidioidomycosis treatment. Rev Inst Cryptococcus neoformans and Mycobacterium tuberculosis in a
Med Trop Sao Paulo 2015;57(suppl 19):31–7. child with systemic lupus erythematosus. Int J Infect Dis
117 Taborda CP, Uran ME, Nosanchuk JD, Travassos LR. 2015;33:106–8.
Paracoccidioidomycosis: challenges in the development of a vaccine 146 Cavalcante EG, Montoni JD, Oliveira GT et  al. Fatal cryptococcal
against an endemic mycosis in the Americas. Rev Inst Med Trop Sao meningitis in a juvenile lupus erythematosus patient. Rev Bras
Paulo 2015;57(suppl 19):21–4. Reumatol 2014;54:155–8.
118 Pilmis B, Puel A, Lortholary O, Lanternier F. New clinical pheno­ 147 Zhang C, Du L, Cai W et al. Isolated hepatobiliary cryptococcosis man­
types of fungal infections in special hosts. Clin Microbiol Infect ifesting as obstructive jaundice in an immunocompetent child: case
2016;22:681–7. report and review of the literature. Eur J Pediatr 2014;173:1569–72.
119 Munoz P, Aguado JM. [Invasive yeast diseases in solid organ trans­ 148 Lenz D, Held J, Goerke S et al. Primary cutaneous cryptococcosis in
plant recipients]. Rev Iberoam Micol 2016;33:152–9. an eight‐year‐old immunocompetent child: how to treat? Klin
120 Cleveland AA, Harrison LG, Farley MM et al. Declining incidence Padiatr 2015;227:41–4.
of Candidemia and the shifting epidemiology of Candida resist­ 149 Kothiwala SK, Prajapat M, Kuldeep CM, Jindal A. Cryptococcal pan­
ance in two US Metropolitan Areas, 2008‐2013: Results from niculitis in a renal transplant recipient: case report and review of lit­
Population‐Based Surveillance. PLoS One 2015;10:e0120452. erature. J Dermatol Case Rep 2015;9:76–80.
121 Marcoux D, Jafarian F, Joncas V et al. Deep cutaneous fungal infec­ 150 Coelho C, Casadevall A. Cryptococcal therapies and drug targets:
tions in immunocompromised children. J Am Acad Dermatol 2009; the old, the new and the promising. Cell Microbiol 2016;18:792–9.
61:857–64. 151 Thompson GR 3rd, Rendon A, Dos Santos RR et al. Isavuconazole
122 Zaoutis TE, Greves HM, Lautenbach E et al. Risk factors for dissemi­ treatment of cryptococcosis and dimorphic mycoses. Clin Infect Dis
nated candidiasis in children with candidemia. Pediatr Infect Dis J 2016;63:356–62.
2004;23:635–41. 152 Segal BH, Herbrecht R, Stevens DA et al. Defining responses to therapy
123 Singhi S, Deep A. Invasive candidiasis in pediatric intensive care and study outcomes in clinical trials of invasive fungal diseases:
units. Indian J Pediatr 2009;76:1033–44. Mycoses Study Group and European Organization for Research and
124 Bruce Diemond J, Lopez C, Huerta Romano F, Montiel Castillo C. Treatment of Cancer consensus criteria. Clin Infect Dis 2008;47:674–83.
Fungal [Candida] infections in the immunocompromised pediat­ 153 Dayal D, Jain P, Kumar R et  al. Clinical spectrum and outcome of
ric patient. Drugs Today 2008;44(suppl 4):45–51. invasive filamentous fungal infections in children with Type 1 diabe­
125 Paramythiotou E, Frantzeskaki F, Flevari A et  al. Invasive fungal tes: North Indian experience. Clin Pediatr Endocrinol 2015;24:51–7.
infections in the ICU: how to approach, how to treat. Molecules 154 García‐Romero MT, García‐Méndez J, Arenas R et al. Zygomycosis
2014;19:1085–119. in two hematologic cases. Case Rep Infect Dis 2011;2011:181782.
126 Donker AE, Mavinkurve‐Groothuis AM, van Die LE et al. Favorable 155 Cantatore‐Francis JL, Shin HT, Heilman E, Glick SA. Primary cuta­
outcome of chronic disseminated candidiasis in four pediatric patients neous zygomycosis in two immunocompromised children. Pediatr
with haematological malignancies. Med Mycol 2012;50:315–19. Dermatol 2007;24:6.
127 Ben Ameur S, Hentati Y, Ben Dhaoui M et al. [Neonatal renal can­ 156 Kennedy KJ, Daveson K, Slavin MA et  al. Mucormycosis in
didiasis: a case report]. Arch Pediatr 2014;21:287–90. Australia: contemporary epidemiology and outcomes. Clin
128 Beasley K, Panach K, Dominguez AR. Disseminated Candida tropi­ Microbiol Infect 2016;22:775–81.
calis presenting with ecthyma‐gangrenosum‐like lesions. Dermatol 157 Angali RK, Jeshtadi A, Namala VA, Gannepalli A. Fatal rhino‐orbito‐
Online J 2016;22. cerebral mucormycosis in a healthy individual. J Oral Maxillofac
129 Ostrosky‐Zeichner L, Oude Lashof AM, Kullberg BJ, Rex JH. Pathol 2014;18:460–3.
Voriconazole salvage treatment of invasive candidiasis. Eur J Clin 158 Pozo Laderas JC, Pontes Moreno A, Pozo Salido C et al. [Disseminated
Microbiol Infect Dis 2003;22:651–5. mucormycosis in immunocompetent patients: A disease that also

SECTION 8: FUNGAL SKIN


130 Buddingh EP, Leentjens J, van der Lugt J et  al. Interferon‐gamma exists]. Rev Iberoam Micol 2015;32:63–70.
immunotherapy in a patient with refractory disseminated candidia­ 159 Bonifaz A, Tirado‐Sanchez A, Calderon L et al. Mucormycosis in
sis. Pediatr Infect Dis J 2015;34:1391–4. children: a study of 22 cases in a Mexican hospital. Mycoses
131 Wang XJ, Sui X, Yan L et  al. Vaccines in the treatment of invasive 2014;57(suppl 3):79–84.

INFECTIONS
candidiasis. Virulence 2015;6:309–15. 160 Park JS, Cho SH, Youn SK et al. Epidemiological characterization of
132 Moretti ML, Resende MR, Lazera MS et al. [Guidelines in crypto­ opportunistic mycoses between the years 2006 and 2010 in Korea.
coccosis—2008]. Rev Soc Bras Med Trop 2008;41:524–44. J Microbiol Biotechnol 2016;26:145–50.
133 Aye C, Henderson A, Yu H, Norton R. Cryptococcosis ‐ The Impact 161 Cheng VC, Chen JH, Wong SC et al. Hospital outbreak of pulmonary
of Delay to Diagnosis. Clin Microbiol Infect 2016;22:632–5. and cutaneous zygomycosis due to contaminated linen items from
134 Maziarz EK, Perfect JR. Cryptococcosis. Infect Dis Clin North Am substandard laundry. Clin Infect Dis 2016;62:714–21.
2016;30:179–206. 162 Davoudi S, Graviss LS, Kontoyiannis DP. Healthcare‐associated out­
135 Cheng MF, Chiou CC, Liu YC et al. Cryptococcus laurentii fungemia breaks due to Mucorales and other uncommon fungi. Eur J Clin
in a premature neonate. J Clin Microbiol 2001;39:1608–11. Invest 2015;45:767–73.
136 Molina‐Leyva A, Ruiz‐Carrascosa JC, Leyva‐Garcia A, Husein‐ 163 Dehority W, Willert J, Pong A. Zygomycetes infections in pediatric
Elahmed H. Cutaneous Cryptococcus laurentii infection in an haematology oncology patients: a case series and review of the lit­
immunocompetent child. Int J Infect Dis 2013;17:e1232–3. erature. J Pediatr Hematol Oncol 2009;31:911–19.
137 Meiring ST, Quan VC, Cohen C et  al. A comparison of cases of 164 Langford S, Trubiano JA, Saxon S et  al. Mucormycete infection or
paediatric‐onset and adult‐onset cryptococcosis detected through colonisation: experience of an Australian tertiary referral centre.
population‐based surveillance, 2005‐2007. AIDS 2012;26:2307–14. Mycoses 2016;59:291–5.
138 Goldman DL, Khine H, Abadi J et al. Serologic evidence for Cryptococcus 165 Kursun E, Turunc T, Demiroglu YZ et al. Evaluation of 28 cases of
neoformans infection in early childhood. Pediatr 2001;107:E66. mucormycosis. Mycoses 2015;58:82–7.
139 Chayakulkeeree M, Perfect JR. Cryptococcosis. Infect Dis Clin North 166 Al‐Zaydani IA, Al‐Hakami AM, Joseph MR et al. Aggressive cutane­
Am 2006;20:507–44, v–vi. ous zygomycosis caused by Apophysomyces variabilis in an immu­
140 Correa Mdo P, Oliveira EC, Duarte RR et al. [Cryptococcosis in children nocompetent child. Med Mycol Case Rep 2015;10:11–13.
in the State of Para, Brazil]. Rev Soc Bras Med Trop 1999;32:505–8. 167 Lelievre L, Garcia‐Hermoso D, Abdoul H et al. Posttraumatic mucor­
141 Warnatz K. Review: Cryptococcosis in HIV‐negative immunodefi­ mycosis: a nationwide study in France and review of the literature.
ciency. Clin Adv Hematol Oncol 2008;6:448–52. Medicine 2014;93:395–404.
142 Soni ZJ, Reubenson G, De Maayer T et al. Neonatal cryptococcosis: 168 Annigeri RA, Parameswaran A. Zygomycosis presenting as acute
beware of false‐positive results. J Pediatric Infect Dis Soc bilateral renal artery thrombosis in a healthy young male. J Assoc
2012;1:250–3. Physicians India 2015;63:77–9.
143 Henao‐Martinez AF, Beckham JD. Cryptococcosis in solid organ 169 Porto DA, Kohen LL. Pulsatile nodule on the ventral wrist. J Am
transplant recipients. Curr Opin Infect Dis 2015;28:300–7. Acad Dermatol 2015;73:e129–30.
578 Section 8  Fungal Skin Infections

170 Dhaliwal HS, Singh A, Sinha SK et al. Diagnosed only if considered: 187 Naeem F, Rubnitz JE, Hakim H. Isolated nasal septum necrosis
isolated renal mucormycosis. Lancet 2015;385:2322. caused by Aspergillus flavus in an immunocompromised child.
171 Khatami A, Outhred AC, Britton PN et  al. Mediastinal mass in a Pediatr Infect Dis J 2011;30:627–9.
healthy adolescent at The Children’s Hospital at Westmead, 188 Idris N, Lim LH. Nasal eschar: a warning sign of potentially fatal
Australia. Thorax 2015;70:194–7. invasive fungal sinusitis in immunocompromised children. J Pediatr
172 Lass‐Florl C. Zygomycosis: conventional laboratory diagnosis. Clin Hematol Oncol 2012;34:e134–6.
Microbiol Infect 2009;15(suppl 5):60–5. 189 Cadena J, Thompson GR 3rd, Patterson TF. Invasive aspergillosis:
173 Marty FM, Ostrosky‐Zeichner L, Cornely OA et  al. Isavuconazole current strategies for diagnosis and management. Infect Dis Clin
treatment for mucormycosis: a single‐arm open‐label trial and case‐ North Am 2016;30:125–42.
control analysis. Lancet Infect Dis 2016;16:828–37. 190 Saha A, Saha K, Chatterjee U. Primary aspergillosis of vocal cord:
174 Skiada A, Pagano L, Groll A et al. Zygomycosis in Europe: analysis Long‐term inhalational steroid use can be the miscreant. Biomed J
of 230 cases accrued by the registry of the European Confederation 2015;38:550–3.
of Medical Mycology [ECMM] Working Group on Zygomycosis 191 Mitchell CA, Kreiger P, Goff C, Shah UK. Pediatric foreign body aspira­
between 2005 and 2007. Clin Microbiol Infect 2011;17:1859–67. tion: A nidus for Aspergillus colonization. Int J Pediatr Otorhinolaryngol
175 Sheybani F, Naderi HR, Sarvghad M et al. How should we manage a 2015;79:938–40.
patient with invasive mucormycosis who develops life‐threatening 192 Gamaletsou MN, Rammaert B, Bueno MA et al. Aspergillus osteo­
reaction to amphotericin B? Report of two cases and literature myelitis: epidemiology, clinical manifestations, management, and
review. Med Mycol Case Rep 2015;8:29–31. outcome. J Infect 2014;68:478–93.
176 De Leonardis F, Perillo T, Giudice G et al. Recurrent rhino‐ocular‐cer­ 193 Ho HC, Hsiao SH, Lee CY, Tsai CC. Treatment of refractory
ebral mucormycosis in a leukemic child: a case report and review of Aspergillus otomycosis with voriconazole: case series and review.
pediatric literature. Pediatr Rep 2015;7:5938. J Laryngol Otol 2014;128:547–51.
177 Walsh TJ, Skiada A, Cornely OA et al. Development of new strategies 194 Arvanitis M, Ziakas PD, Zacharioudakis IM et al. PCR in diagnosis
for early diagnosis of mucormycosis from bench to bedside. Mycoses of invasive aspergillosis: a meta‐analysis of diagnostic performance.
2014;57(suppl 3):2–7. J Clin Microbiol 2014;52:3731–42.
178 Erjavec Z, Kluin‐Nelemans H, Verweij PE. Trends in invasive fungal 195 Del Bono V, Mikulska M, Viscoli C. Invasive aspergillosis: diagnosis,
infections, with emphasis on invasive aspergillosis. Clin Microbiol prophylaxis and treatment. Curr Opin Hematol 2008;15:586–93.
Infect 2009;15:625–33. 196 Domenech C, Leick‐Courtois C, Bienvenu AL et al. Improvement
179 Segal BH. Aspergillosis. N Engl J Med 2009;360:1870–84. in the outcome of invasive aspergillosis in a pediatric hematol­
180 Aggarwal E, Mulay K, Menon V et al. Isolated orbital aspergillosis in ogy  department: a 10‐year review. J Pediatr Hematol Oncol
immunocompetent patients: a multicenter study. Am J Ophthalmol 2015;37:560–5.
2016;165:125–32. 197 Mansh M, Binstock M, Williams K et al. Voriconazole exposure and
181 Frange P, Bougnoux ME, Lanternier F et al. An update on pediatric risk of cutaneous squamous cell carcinoma, Aspergillus coloniza­
invasive aspergillosis. Med Mal Infect 2015;45:189–98. tion, invasive aspergillosis and death in lung transplant recipients.
182 Maschmeyer G, Haas A, Cornely OA. Invasive aspergillosis: epi­ Am J Transplant 2016;16:262–70.
demiology, diagnosis and management in immunocompromised 198 Hachem R, Gomes MZ, El Helou G et  al. Invasive aspergillosis
patients. Drugs 2007;67:1567–601. caused by Aspergillus terreus: an emerging opportunistic infection
183 Lanternier F, Cypowyj S, Picard C et al. Primary immunodeficiencies with poor outcome independent of azole therapy. J Antimicrob
underlying fungal infections. Curr Opin Pediatr 2013;25:736–47. Chemother 2014;69:3148–55.
184 Tragiannidis A, Roilides E, Walsh TJ, Groll AH. Invasive aspergil­ 199 Stefano PC, Noriega AL, Kobrin AL et  al. Primary cutaneous
losis in children with acquired immunodeficiencies. Clin Infect nocardiosis in immunocompetent children. Eur J Dermatol
Dis 2012;54:258–67. 2006;16:406–8.
185 Armstead J, Morris J, Denning DW. Multi‐country estimate of different 200 Vasarinsh P. Primary cutaneous nocardiosis. Arch Dermatol
manifestations of aspergillosis in cystic fibrosis. PloS One 2014;9:e98502. 1968;98:489–93.
186 Torrelo A, Hernandez‐Martin A, Scaglione C et al. Aspergilosis cutánea 201 Badgett JT, Adams G. Mandibular actinomycosis treated with oral
SECTION 8: FUNGAL SKIN

primaria en un niño con leucemia. Act Dermosifiliogr 2007;98:276–8. clindamycin. Pediatr Infect Dis J 1987;6:221–2.
INFECTIONS
 579

C HA PTER   48

Molluscum Contagiosum
Joachim J. Bugert1, Ali Alikhan2 & Tor Shwayder3
1
 Institut für Mikrobiologie der Bundeswehr, München, Germany
2
 University of Cincinnati, Department of Dermatology, Cincinnati, OH, USA
3
 Department of Dermatology, Henry Ford Hospital, Detroit, MI, USA

Abstract and 77% in different geographical locations and subpopulations,


s­ uggesting many infections are either subclinical or not reported.
MC is normally self‐limiting with few lesions disappearing sponta-
Molluscum contagiosum (MC) is the most common poxvirus infec-
neously without need for treatment. Extensive cases are seen in
tion circulating in humans. Prevalence/incidence studies indicate
immunosuppressed individuals and those with atopic dermatitis.
the infection is frequently seen in general practice with a case
MC can persist for years, and treatment should be considered in
incidence rate of 1295/100 000 person‐years in the UK. Mollus-
those cases.
cum contagiosum virus (MCV) seroprevalences vary between 6%

Key points • MC lesions are, once detected, cleared by an overwhelming


immune response.
• MCV enters basal keratinocytes and causes hyperproliferation
• Molluscum contagiosum (MC) is a self‐limiting poxviral infection
of epidermal cells.
caused by molluscum contagiosum virus (MCV).
• MCV is secreted through the umbilicated lesion centre onto the
• MCV induces an antibody response which does not prevent
skin surface and then transmitted by smear infection.
re‐infection.
• Diagnosis can be made by clinical inspection, extrusion of papule
• MC seroprevalence is between 6% and 77% depending on
contents and histopathology.
enzyme‐linked immunosorbent assay (ELISA) format, and
• Destructive treatment methods including laser/cryotherapy,
population studied.
topical cytotoxins, and curettage are not adequately evaluated
• MC lesions persist for up to 3 years because they are temporarily
to be included in the 2013 Cochrane report.
shielded from immune effectors through sequestration and
• Imiquimod is ineffective.
production of immune evasive molecules, including inhibitors of
signalling pathways and apoptosis.

detected [3,4]. A Turkish study detected MCV‐1 in 100%


Aetiology. Molluscum contagiosum (MC) is a benign
(61/61) of cases examined [5]. In Japan, MCV‐1 and ‐3
self‐limiting infection of the skin caused by a member
are most commonly found, whereas MCV‐2 and ‐4
of the poxvirus family, molluscum contagiosum virus
occur in only 2% of cases [6]. MCV‐2 occurs primarily
(MCV). MCV‐1/80–U60315 is the type member of the
in sexually transmitted MC [7]. There may also be vari-
genus Molluscipoxvirus and has a linear double‐

SECTION 9: VIRAL SKIN


ation in prevalence of the subtypes in those infected with
stranded DNA genome of approximately 190 000 base
the human immunodeficiency virus (HIV) compared
pairs [1]. The infection appears to be specific to the
with immunocompetent individuals [8].

INFECTIONS
human host, and MCV cannot be grown in tissue
culture or embryonated chicken eggs. Unlike human
papillomaviruses or herpesviruses, it does not inte- Epidemiology. MC is a common infection, occurring
grate into the host genome or develop a latent state. worldwide and demonstrating variation in geographical
Restriction endonuclease analysis of the genomic distribution of viral subtypes [3,4,6]. The exact prevalence
DNA from various sources has revealed four closely is not known, but an increase in sexually transmitted MC
related viral subtypes, MCV‐1–4. There is no appar- was reported in both the UK and USA between 1960 and
ent relationship between viral subtype, morphology 1980 [9]. Incidence has been previously reported at
or anatomical distribution [2,3]. However, there between 2% and 10% [10,11], and the incidence rate in
appears to be marked geographical variation in the dis- those 14 years or younger in a large British study was
tribution of subtypes. MCV‐1 and its variants account 1265/100 000 person‐years [12]. Of note, in the USA, visits
for 98% of cases in the USA, while MCV‐1 and ‐2 pre- to dermatologists comprised the majority (71%) of healthcare
dominate in the UK, with MCV‐3 only rarely being utilization units for MC [13].

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
580 Section 9  Viral Skin Infections and Opportunistic Infections

Local outbreaks of MC occur and have been reported


in children attending swimming pools [14,15] and on
one occasion among 34 children from a kibbutz [16].
High incidence usually reflects increased exposure to
the virus. Sexual transmission from skin to skin surface
suggests that close contact is required to facilitate
spread of infection. MC rarely occurs before the age of
1 year, perhaps because of less direct contact with other
children [14], but this might also be explained by mater-
nally transmitted immunity to the infant or a prolonged
incubation period. Nonetheless, congenital transmis-
sion has been reported, presenting as lesions on the
extremities and scalp [7].
Age (young school‐aged children), living in close prox-
imity, skin‐to‐skin contact, sharing of fomites, residence
in tropical climates and swimming pool use are associ- Fig. 48.1  Seventeen‐year‐old white male with a subtle immune defect
with widespread molluscum and flat warts.
ated with higher rates of MC, while the child’s sex, sea-
sonality and hygiene do not show an association [10].
Viral transmission via fomites inside the pool (e.g. kick- Concomitant atopic dermatitis must not be confused
boards) or outside the pool (e.g. bath towels), or both, with new‐onset dermatitis that may occur around the
may explain the general correlation between MC and mollusca [28]. It is unclear whether application of topical
swimming pool exposure [10]. corticosteroids encourages the infection to flourish [29],
A prospective community cohort study by Olsen and or reduces the numbers of lesions [27]. Inflammation of
co‐workers in 2015 described MC as one of the 50 most MC lesions may indicate immune responses leading to
prevalent diseases worldwide with very little epidemio- viral clearance.
logical information available. The authors found a rate of Other immunosuppressants such as tacrolimus [30,31],
MC consultations in primary care for children aged 0–14 pimecrolimus [32] and methotrexate [33] can also facili-
years of 9.5 per 1000. In one in ten children with MC, the tate spread of an infection. Extensive and persistent MC
condition is likely to have a substantial effect on their also occurs in the immunosuppressed, including organ
quality of life [17]. allograft recipients and patients infected with HIV [34–36].
In a 2014 systematic literature review by the same In the latter group, facial involvement is common
authors, most epidemiological data on MC were found to (Fig. 48.1) [37]. Of children with acquired immune deficiency
be of poor quality. However, the picture emerging is that syndrome (AIDS) in a Romanian orphanage, 74% had MC
children aged between 0 and 14 years have an incidence of the head or face [38]. These observations support the
rate of 12–14 episodes per 1000 per year, with a point importance of cell‐mediated immunity in eliminating or
prevalence in children aged 0–16 years of between 5.1% controlling spread of the infection.
and 11.5% as per meta‐analysis. Children aged 1–4 years
exhibit the highest incidence. Swimming and eczema are Pathology. The virus enters the basal keratinocytes,
associated with the presence of MC, but the causal rela- where it causes an increase in cell turnover that extends to
tionships are unclear [18]. the suprabasal region. Higher in the stratum spinosum,
SECTION 9: VIRAL SKIN

Recent enzyme‐linked immunosorbent assay (ELISA)‐ the mitotic rate decreases as viral DNA synthesis increases.
based studies show that seroprevalences in general popu- Proliferating cells within the follicular epithelium in par-
INFECTIONS

lations vary from 6% in Japan [19] to 14.8% in Germany ticular form lobulated epithelial masses compressing the
[20], while seroprevalences up to 77% are observed in dermal papillae.
immunosuppressed individuals [21,22]. However, the basal layer is not disrupted. Cytoplasmic
aggregations of viral material appear as large hyaline
Relation to disease state bodies (molluscum bodies), and these eventually destroy
Clinical impressions suggest that MC occurs commonly the cells, particularly at the centre of each lobule. In a fully
in those with atopic dermatitis [23,24]. In large epidemio- developed lesion, a cavity develops in which there are
logical studies, up to 24.2% of MC patients were diag- large numbers of molluscum bodies (Fig. 48.2).
nosed with concomitant atopic dermatitis [25,26]. Little inflammatory infiltrate is seen in the adjacent der-
MCV seroconversion was more common in dermato- mis, although in chronic MC a granulomatous infiltrate
logical and autoimmune disorders than in immunocom- may occur, possibly secondary to the discharge of papule
promised patients or patients with multiple sclerosis [20]. contents into the upper dermis [39]. Scanning electron
A retrospective medical chart review of 696 patients microscopy revealed a number of different gross mor-
(mean age, 5.5 years) with MC found that 259 patients phologies of molluscum but all had a dense distribution
(37.2%) had a history of atopic dermatitis. Patients with of small protrusions with similar ultrastructure [40].
atopic dermatitis had higher numbers of MC lesions and In a synopsis of observations available so far, the MCV
an increased likelihood of molluscum dermatitis (50.6% lifecycle (Fig. 48.3) starts with attachment of virus particles
vs. 31.8% [27]). to keratinocytes, probably followed by macropinocytosis
Chapter 48  Molluscum Contagiosum 581

structural gene products and assembly are extremely


efficient in vivo and sequestered in the ‘viral colony sac’, a
cellular membrane compartment in the cytoplasm of
infected cells observed on electron microscopy [48].
Cellular organelles are pushed to the side and eventually
disappear altogether in the spinous layer of infected
­epidermis. The final step of viral egress is a destructive
process characterized by disintegrating cells, with virus
particles, membranous vesicles and other cellular debris
forming a characteristic plug‐like debrisome in the umbil-
icated centre of MC lesions (Fig.  48.2). This plug can be
extruded by pressure, leading to release of infectious
virus and autoinoculation as well as transmission through
smear infection. This process is similar to holocrine secre-
tion from sebaceous glands and possibly associated with
hair follicles, leading to the hypothesis that the initially
(a) infected cells are follicular stem cells [21]. Reattachment
and reinfection of MCV in neighbouring skin areas may
then occur, producing further MC lesions.
Immune response
Good correlation was found between the presence of
antibodies to MCV and clinical infection [49,50]. More
recently, ELISAs were found to be specific and sensitive in
the detection of antibodies to MCV [20,51]. However, as
with viral warts, it seems that depressed cell‐mediated
immunity (Th1), secondary to anti‐inflammatory viru-
lence factors produced by the MCV, rather than humoral
immunity (Th2) is of greater importance in disease
control [45,52].
MCV may evade detection and destruction by encod-
ing proteins suppressing the immune system, including
chemokine inhibitors (mc148 [53]), IFN‐β inhibitors
(mc160 [54]), FLICE/apoptosis inhibitors (mc066/mc159
(b)
[55,56]), as well as homologues of CD150/SLAM, which
Fig. 48.2  (a and b) Haematoxylin and eosin stained section of a fully is involved in signalling pathways essential to viral
developed molluscum lesion. Proliferating cells within the follicular
immunity (mc002, mc161 and mc162 [57]). The pathologi-
epithelium form lobulated epithelial masses. Cytoplasmic aggregations of
viral material appear as large hyaline bodies (molluscum bodies).
cal appearance of the papular stage of resolving MC
shows a dense mononuclear cell infiltrate hugging the
epidermis [58], suggesting that a cell‐mediated immune

SECTION 9: VIRAL SKIN


[41]. Release of the virus core leads to transcription of mechanism is involved in this process. There is evidence
early mRNA which has been observed in vitro [42]. Early for humoral immunity directed against MCV [19,20]

INFECTIONS
gene products are partially secreted, suppress innate through the ubiquitin‐proteasome system, followed by
immune responses and may have systemic effects (mc159 apoptosis [59] and complete clearance as the consequence
and 160, NF‐κB, interferon [IFN] [1]). The infection is up of an overwhelming immune response [43].
to this point undetected by the immune system and the
lesion can develop to the immunologically undisturbed Clinical features. An incubation period for MCV of
state [43]. However, leakage of virus components through 2 weeks to 6 months has been reported [60].
the basal membrane or limited viraemia [44] at any stage This is also reflected in the seropositivity rate in a
would explain polymerase chain reaction (PCR) detection cohort of 289 German adults and children aged 0–40
in serum as well as serological responses (mc084 [20]) and years; seropositivity was low in children below the age of
further control by cellular immune effectors and clear- 1 (4.5%), peaked in children aged 2–10 years (25%) and
ance [43]. If cellular immune control is missing, extensive fell again in older populations (11–40 years: 12.5% [20]).
MC is the consequence [45]. Uncoating and DNA replica- The rate of general practitioner consultations in chil-
tion are the next steps in cells now dividing due to viral dren aged 1–4 years and 5–9 years was found to be 13.1
interference with the cell cycle (mc007 [46]). This step is and 13.0 (males) and 13.0 and 13.9 (females) per 1000
not well understood and does not happen in any in vitro respectively, as compared to 9.5 per 1000 in children aged
cell culture system tested so far. A cellular factor that is 0–14 years [25].
only present/induced in infected epidermis may be In a prospective community cohort study of 306 chil-
required [47]. Viral DNA replication, translation of late dren with MC [17] the mean time to resolution was
582 Section 9  Viral Skin Infections and Opportunistic Infections

Egress – destructive

‘Debrisome - plug’

Infection - attachment

‘Viral colony sac’

Epidermal differentiation
Macropinocytosis/core release

Late gene products

Early gene products Uncoating/DNA replication/mitosis?

Spinous cell
N
Basal cell

Basal membrane

Secreted viral factors - antigen/leak?

Virion

Viral genome

Viral core protein/lateral bodies

Early gene product

Late gene product

Viral colony sac

Intra/extracellular vesicles

Fig. 48.3  Schematic diagram of the lifecycle of molluscum contagiosum virus in human epidermis. N, nucleus.

c­ entre (Fig.  48.4). Lesions vary in size from 1 to 10 mm


although occasionally giant lesions are seen (primarily in
the setting of HIV infection). Growth occurs over several
weeks and eventually spontaneous remission will ensue.
SECTION 9: VIRAL SKIN

Spontaneous remission is heralded by inflammation,


suppuration and crusting (Fig.  48.5) which destroys the
lesion, leaving a small atrophic scar. Unlike varicella, the
INFECTIONS

molluscum pox scar is transitory. Most cases of MC


resolve within 6–9 months, but they can occasionally per-
sist for several years. However, studies on children in Fiji
show that no individual lesion persists for more than
2 months. In rare cases, primarily in the setting of immu-
nodeficiency, widely disseminated and grossly disfiguring
MC can occur in children (see Fig. 48.1) [61].
The lesions are often distributed on sites of friction
from clothing (e.g. around the neck, axillae or trunk); in
Fig. 48.4  Classic molluscum on the upper back of a 6‐year‐old white male. one large epidemiological study, the trunk was the most
commonly reported location for lesions, and half of the
patients presented with lesions in more than one anatomical
13.3 months. Eighty (30%) of 269 cases had not resolved region (Fig. 48.6) [26]. In tropical climates, involvement of
by 18 months, 36 (13%) had not resolved by 24 months, the limbs is more common. MC does occur in the anogeni-
and transmission to other children in the household was tal region too, but this does not necessarily signify sexual
recorded in 102 (41%) of 250 cases. spread in children (Fig. 48.7). More unusual sites include
The morphology of an individual lesion is a dome‐shaped the scalp and face, although the latter distribution is com-
papule, flesh‐coloured or pearly, with an umbilicated mon in those infected with HIV [28]. On the soles and
Chapter 48  Molluscum Contagiosum 583

Fig. 48.5  Inflammation of molluscum heralding resolution in a 5‐year‐old Fig. 48.7  Molluscum and human papillomavirus in a 20‐month‐old white
white female. female with eczematous reaction.

Fig. 48.8  KOH of a molluscum lesion. Low power. Note enlarged


keratinocytes in pseudo‐saccules leading to central pore.

MCV infection. Herpes simplex virus (other than the

SECTION 9: VIRAL SKIN


vesicular form), human papillomavirus, benign naevi,
fibrous papules, adnexal tumours, milia, juvenile xan-

INFECTIONS
thogranuloma, pyogenic granuloma, bacillary angioma-
Fig. 48.6  Three‐year‐old white male with multiple mollusca on his chest.
tosis, histoplasmosis, sebaceous naevi, comedones,
furuncles, syringomas, Penicillium marneffei infection,
mucosal surfaces, the appearance is atypical [62–64]. basal cell carcinoma, keratoacanthoma and varicella zos-
Around 10% of patients develop eczema around the ter virus should also remain in the differential [61,69].
lesions, but this disappears as the infection resolves [65]. If diagnosis is not straightforward, other techniques
Ocular manifestations of MC include dermal lid lesions, may be of assistance. The easiest is a 10% KOH smear;
conjunctival lesions, follicular conjunctivitis, intraocular simple light microscopy will demonstrate a papule with
lesions and chronic conjunctivitis or keratoconjunctivitis pseudo‐saccules gathered symmetrically around the cen-
[66,67]. If left untreated, vascular infiltration and scarring tral pore. These pseudo‐saccules have ever‐increasing
of the peripheral cornea may occur [68]. keratinocyte cell size as one looks from the base to the
centre of the specimen (Fig.  48.8). In addition to KOH
Diagnosis. This is generally straightforward, but large smear, biopsy, real‐time PCR of swab samples (with
solitary lesions in adults may occasionally be mistaken pyrosequencing to differentiate between MCV‐1 and
for basal cell carcinoma. Expression of a white cheesy MCV‐2) [70], dermoscopy [71] and reflectance confocal
substance by lateral pressure with forceps on an individ- microscopy (RCM) [72] may also help to diagnose MC
ual lesion is characteristic. In the immunosuppressed [69]. Dermoscopy reveals a polylobular, white‐yellow,
patient, cutaneous cryptococcal infection may mimic amorphous structure in the centre surrounded by a crown
584 Section 9  Viral Skin Infections and Opportunistic Infections

of vessels [73]. RCM demonstrates a round, well‐circum- to each lesion with the blunt, wooden end of an applicator
scribed lesion with central round cystic areas teeming stick and rinsed off after 2–6 hours, or earlier in the case of
with bright refractile material [72]. ­discomfort. Another study of 54 patients demonstrated a
96% improvement rate and a parental satisfaction rate of
Treatment. In minor infections, it is reasonable to await 78% [83]. Several applications may be required to success-
spontaneous resolution, as this is least distressing to a fully treat lesions. In our experience (TS), two to six visits
child. There is a risk, however, of widespread cutaneous usually cure even the most widespread cases, and cantha-
dissemination, discomfort, bacterial superinfection, acute ridin has been used safely on the face and anogenital
inflammatory and chronic granulomatous reactions and region without adverse events. Careful application of the
scar formation [28]. Spontaneously resolving lesions that cantharidin with a wooden applicator stick should be fol-
become secondarily infected may require topical antisep- lowed by immediate drying with a fan. Adjacent skin
tic application. Furthermore, left untreated, lesions can should be held apart until completely dry. If the skin is
persist for several months to several years. thin or in an intertriginous area, less contact time
Active treatments for MC include destructive methods, is needed (2 hours). The eyelids can be treated if the child
occlusion, and cytotoxic, immune‐modulating and antivi- is calm and can hold still with their eyes closed until the
ral preparations. Unlike varicella scars, molluscum scars cantharidin is completely dried. Avoid the area immediately
are usually few and resolve completely. Molluscum scar adjacent to the eye margins. Avoid at all costs dripping
keloids have not been noted in the literature. cantharidin into the eye itself. Baby shampoo is recom-
mended to wash the area afterwards to avoid the eye‐
Destructive methods stinging sensation of regular soaps.
Cryotherapy and curettage. Cryotherapy is a very effective This preparation is not available in some countries,
therapy, inducing an ice ball on the lesion itself but not on including the UK, and may have some adverse events,
the surrounding skin [60,74,75]. Application of EMLA including exaggerated blistering, pain, pruritus, tempo-
cream (a eutectic mixture of prilocaine and lidocaine) rary hypopigmentation and secondary infection [83].
1  hour before the procedure may facilitate treatment,
which should not, however, be inflicted on an unwilling Keratolytics.  Keratolytics, which disintegrate des-
recipient! The use of phenol is not recommended because mosomes and hemi‐desmosomes, are also an effective
of scarring and potential toxicity/carcinogenicity. treatment for MC. However, one large study examining a
Curettage of single or a few lesions can be carried out, combination cream of salicylic acid 16.7% and lactic acid
once again using EMLA cream for local anaesthesia [76,77]. 16.7% found it to be quite irritating, resulting in only a
In order to avoid the risk of methaemoglobinaemia, the 32.1% patient and parent satisfaction rate even with the
recommended upper quantities of EMLA cream per appli- 100% cure rate by two visits [28]. Adapalene 0.1% gel [84]
cation need to be taken into consideration (see Chapter 173). and salicylic acid 12% gel [85] may also be effective treat-
In one large study of four treatments, curettage was ment options. Again, caution is necessary in order to
found to be the most effective, associated with the fewest avoid systemic toxic effects of transcutaneously absorbed
adverse effects, and rated highest for patient satisfaction salicylic acid (see Chapter 5).
among patients and parents [19]. In this study curettage
was done under general anaesthesia. Curettage had an Electrosurgery. Electrosurgery has been suggested using
80% cure rate after one visit and 96% after two visits. Of a disposable 23‐gauge or thicker bore hypodermic needle
note, in this same study, after two visits, topical canthari-
SECTION 9: VIRAL SKIN

as the effective probe [86]. Pain and possible scarring


din had an 80% cure rate and topical keratolytics had a must be considered when using this modality.
100% cure rate. These treatments did not require anaes-
INFECTIONS

thesia. Another curettage study found a high risk of treat- Topical potassium hydroxide. KOH aqueous solution in
ment failure at weeks 4 and 8; risk factors for treatment concentrations of 5–20% to all lesions is another treatment
failure were the number of lesions at day 0, the number of option for MC as it effectively dissolves keratin [87].
involved anatomical sites, and concomitant atopic derma- Applied once daily at bedtime, this solution achieved
titis [78]. Furthermore, topical anaesthesia (e.g. EMLA) complete clearance after a mean period of 17 days in 27
and occasionally systemic sedation may be required to Indian children [87]. Ten percent KOH also demonstrated
perform the procedure. efficacy, with lesions in 70% of children completely clear-
Recently, a modified curettage technique has been ing in a double‐blind, placebo‐controlled study [88].
described, involving first opening the epithelial surface of
the lesion with a 30‐gauge needle followed by gently Laser treatment.  Flashlamp‐pumped pulsed‐dye laser
scraping away the viral core with a fox curette [79]. This therapy is a treatment for MC [89–91], although rand-
may be less traumatic and forceful compared with tradi- omized controlled trials are lacking. In most cases, only
tional curettage. one or two treatments are needed for complete lesion
resolution. Furthermore, the face, trunk and extremities
Cantharidin.  Cantharidin, a vesicant produced by can all be treated with this modality, and side‐effects are
Coleoptera, has been widely used for MC in the USA [80–82]. minimal (e.g. local pain, itching, transient hypo‐ or hyper-
In a series of 300 children [80] it was shown to be pigmentation). EMLA cream under occlusion prior to
extremely effective and well tolerated, applied carefully treatment is advisable in younger children.
Chapter 48  Molluscum Contagiosum 585

Duct tape. Typically used for common warts, duct tape occlusion Other antiretroviral agents, such as zidovudine,
is also effective in treating MC [92]. It is nondestructive, lamivudine and nevirapine, have successfully cleared
painless, affordable and convenient. Complete treatment MC in those with comorbid AIDS [105]. There is no mech-
may take several weeks to months. anistic explanation for this effect.

Topical cytotoxic therapy.  Topical therapy with podo- Other treatments


phyllotoxin has been found to give good results, a 0.5% These include 10% solution (v/v) of essential oil of
cream resulting in a 92% rate of resolution [93]. The epi- Australian lemon myrtle (Backhousia citriodora) [106],
dermal irritation that results is thought to disrupt the pro- 0.0001–0.1% topical diphencyprone solution [107], and
tein–lipid membrane around the molluscum, thereby photodynamic therapy with 20% 5‐aminolaevulinic
facilitating an immune response. Systemic toxicity after acid [108].
transcutaneous resorption has been reported [94]. The The 2006 Cochrane review on MC treatment, updated
substance is not licensed for use in children. in 2010 [109] stated that many treatments have been
used for MC but a clear evidence base supporting them
is lacking. Eleven studies with a total number of 495
Immune‐modulating treatments participants were evaluated, of which nine described
Immune modulators. Imiquimod, a member of the imi- the effects of topical treatments and one each described
dazoquinoline family, stimulates the release of proinflam- systemic and homeopathic interventions, respectively.
matory cytokines such as IFN‐β, IFN‐α, tumour necrosis No treatment was convincingly effective, while many
factor (TNF)‐α, interleukin (IL)‐1, IL‐3, IL‐6, IL‐8 and common treatments for MC, such as physical destruc-
IL‐10 [28] by Th1 lymphocytes and is a therapeutic ago- tion, did not fulfil the criteria to be included in the
nist of toll‐like receptor (TLR)7 [95]. Its use in molluscum Cochrane review.
infection is not recommended. In a letter to Lancet
Infectious Diseases in 2014, Kenneth Katz pointed out that
imiquimod had been tested in two large but unpublished Conclusion. MC is a common, worldwide poxvirus infec-
randomized trials (IMQ14194 and 1495‐ [2006]), and tion that usually undergoes spontaneous resolution. As a
found to be ineffective in 702 individuals 2–12 years of general rule, reports concerning MC treatment cures,
age, of whom 470 had received imiquimod 5% cream [96]. sometimes in the medical literature and almost always in
Independently, Yaldo and co‐workers reported in 2015 the lay press and on the internet, are usually without pla-
that MC lesions show no detectable expression of TLR 7 and cebo control. Multiple small controlled studies have been
8, and TLR7 agonists may therefore be ineffective [97]. conducted to support various treatments; however, the
only treatment tested in two large randomized controlled
trials, 5% imiquimod cream, was shown to be ineffective.
Candida antigen immunotherapy. This method involves Destructive methods and systemic DNA polymerase
injection of a killed yeast protein into a molluscum inhibiting antivirals are supported by mechanistic consid-
lesion, in the hope that as the immune system becomes erations. The selection of treatment for recalcitrant cases
active in this region, it will mount an immune response should take into account potential side‐effects, scarring
not only to the yeast protein but also against MCV [98]. and cost‐effectiveness [110]. Because the infection resolves
A retrospective chart review of patients treated over on its own, often in a matter of months, the decision
a 1‐year period demonstrated complete resolution in whether or not to try an intervention must ultimately be
56% and partial resolution in 28% of patients upon

SECTION 9: VIRAL SKIN


made by the patient or their guardian.
­follow‐up [98].
References

INFECTIONS
Antiviral agents. Nitric oxide (NO) is known to be effec- 1 Senkevich TG, Koonin EV, Bugert JJ et al. The genome of molluscum
contagiosum virus: analysis and comparison with other poxviruses.
tive against both DNA and RNA viruses. The application Virology 1997;233:19–42.
under occlusion of an NO‐liberating cream containing 5% 2 Scholz J, Rosen‐Wolff A, Bugert J et  al. Epidemiology of molluscum
sodium nitrite combined with salicylic acid 5% was found contagiosum using genetic analysis of the viral DNA. J Med Virol
to be significantly more effective than application of 5% 1989;27:87–90.
3 Porter CD, Archard LC. Characterisation by restriction mapping of
salicylic acid alone (cure rate of 75% compared with 21%) three subtypes of molluscum contagiosum virus. J Med Virol
[99]. This preparation, although not commercially availa- 1992;38:1–6.
ble, appears to be promising and cost‐effective. 4 Smith KJ, Yeager J, Skelton H. Molluscum contagiosum: its clinical, his-
topathologic, and immunohistochemical spectrum. Int J Dermatol
Cidofovir is a nucleotide analogue of deoxycytidine 1999;38:664–72.
monophosphate, and exhibits a broad range of activity 5 Saral Y, Kalkan A, Ozdarendeli A et al. Detection of molluscum conta-
against DNA viruses. It functions by inhibiting MCV giosum virus (MCV) subtype I as a single dominant virus subtype in
DNA polymerase activity [100]. When used intravenously molluscum lesions from a Turkish population. Arch Med Res
2006;37:388–91.
for treatment of cytomegalovirus infection, it was noted 6 Nakamura J, Arao Y, Yoshida M et al. Molecular epidemiological study
to clear concurrent MC infection [101]. A subsequently of molluscum contagiosum virus in two urban areas of western Japan
produced topical formulation was also successful against by the in‐gel endonuclease digestion method. Arch Virol
1992;125:339–45.
MC [102–104] but, at present, in view of its great cost,
7 Connell CO, Oranje A, van Gysel D, Silverberg NB. Congenital mol-
there are no plans to develop a topical formulation luscum contagiosum: report of four cases and review of the literature.
commercially. Pediatr Dermatol 2008;25:553–6.
586 Section 9  Viral Skin Infections and Opportunistic Infections

8 Thompson CH, de Zwart‐Steffe RT, Donovan B. Clinical and molecu- 35 Hughes WT, Parham DM. Molluscum contagiosum in children with
lar aspects of molluscum contagiosum infection in HIV‐1 positive cancer or acquired immunodeficiency syndrome. Pediatr Infect Dis J
patients. Int J STD AIDS 1992;3:101–6. 1991;10:152–6.
9 Oriel JD. The increase in molluscum contagiosum. BMJ (Clin Res Ed) 36 Katzman M, Carey JT, Elmets CA et al. Molluscum contagiosum and
1987;294:74. the acquired immunodeficiency syndrome: clinical and immunologi-
10 Braue A, Ross G, Varigos G, Kelly H. Epidemiology and impact of cal details of two cases. Br J Dermatol 1987;116:131–8.
childhood molluscum contagiosum: a case series and critical review 37 Chaudhary M, Kulkarni M. Molluscum contagiosum in human
of the literature. Pediatr Dermatol 2005;22:287–94. immunodeficiency virus infected patients. Indian J Dent Res
11 Kyriakis KP, Palamaras I, Terzoudi S et al. Case detection rates of mol- 2008;19:155–9.
luscum contagiosum in childhood. Pediatr Dermatol 2007;24:198–9. 38 Fejes L. [Incidence of molluscum contagiosum in children with AIDS
12 Pannell RS, Fleming DM, Cross KW. The incidence of molluscum con- at the Cervanoda Orphanage, District of Constanta, Romania]. Bull
tagiosum, scabies and lichen planus. Epidemiol Infect 2005;133: Soc Pathol Exot 1993;86:327–8.
985–91. 39 Henao M, Freeman RG. Inflammatory molluscum contagiosum.
13 Molino AC, Fleischer AB Jr, Feldman SR. Patient demographics and Clinicopathological study of seven cases. Arch Dermatol 1964;
utilization of health care services for molluscum contagiosum. Pediatr 90:479–82.
Dermatol 2004;21:628–32. 40 Mihara M. Three‐dimensional ultrastructural study of molluscum
14 Postlethwaite R, Watt JA, Hawley TG et  al. Features of molluscum contagiosum in the skin using scanning‐electron microscopy. Br J
contagiosum in the north‐east of Scotland and in Fijian village settle- Dermatol 1991;125:557–60.
ments. J Hygiene 1967;65:281–91. 41 Mercer M, Helenius A. Vaccinia virus uses macropinocytosis and
15 Bader RE. [Multiple occurrence of molluscum contagiosum in the apoptotic mimicry to enter host cells. Science 2008;320:531–5.
zone of a swimming pool. Attempt at an epidemiological analysis]. 42 Bugert JJ, Melquiot NV, Darai G. Mapping of mRNA transcripts in the
Arch Hyg Bakteriol 1967;151:388–402. genome of molluscum contagiosum virus: transcriptional analysis of
16 Oren B, Wende SO. An outbreak of molluscum contagiosum in a kib- the viral slam gene family. Virus Genes 2000;21:189–92.
butz. Infection 1991;19:159–61. 43 Vermi W, Fisogni S, Salogni L et  al. Spontaneous regression of
17 Olsen JR, Gallacher J, Finlay AY et al. Time to resolution and effect highly immunogenic molluscum contagiosum virus (MCV)‐
on quality of life of molluscum contagiosum in children in the UK: a induced skin lesions is associated with plasmacytoid dendritic cells
prospective community cohort study. Lancet Infect Dis 2015;15:190–5. and IFN‐DC infiltration. J Invest Dermatol 2011;131:426–34.
18 Olsen JR, Gallacher J, Piguet V, Francis NA. Epidemiology of mollus- 44 Cohen J, Davila W, Ali MA et al. Detection of molluscum contagiosum
cum contagiosum in children: a systematic review. Fam Pract virus (MCV) DNA in the plasma of an immunocompromised patient
2014;31:130–6. and possible reduction of MCV DNA with CMX‐001. J Infect Dis
19 Watanabe T, Nakamura K, Wakugawa M et al. Antibodies to mollus- 2012;205:794–7.
cum contagiosum virus in the general population and susceptible 45 Halacli SO, Ayvaz DC, Sun‐Tan C et  al. STK4 (MST1) deficiency in
patients. Arch Dermatol 2000;136:1518–22. two siblings with autoimmune cytopenias: A novel mutation. Clin
20 Sherwani S, Farleigh L, Agarwal N et al. Seroprevalence of mollus- Immunol 2015;161:316–23.
cum contagiosum virus in German and UK populations. PLoS One 46 Mohr S, Grandemange S, Massimi P et  al. Targeting the retinoblas-
2014;18:e88734. toma protein by MC007L, gene product of the molluscum contagio-
21 Chen X, Anstey AV, Bugert JJ. Molluscum contagiosum virus infec- sum virus: detection of a novel virus‐cell interaction by a member of
tion. Lancet Infect Dis 2013;13:877–88. the poxviruses. J Virol 2008;82:10625–33.
22 Konya J, Thompson CH. Molluscum contagiosum virus: antibody 47 Bugert JJ, Melquiot N, Kehm R. Molluscum contagiosum virus
responses in persons with clinical lesions and seroepidemiology in a expresses late genes in primary human fibroblasts but does not pro-
representative Australian population. J Infect Dis 1999;179:701–4. duce infectious progeny. Virus Genes 2001;22:27–33.
23 Pauly CR, Artis WM, Jones HE. Atopic dermatitis, impaired cellular 48 Shelley WB, Burmeister V. Demonstration of a unique viral structure:
immunity, and molluscum contagiosum. Arch Dermatol 1978;114: the molluscum viral colony sac. Br J Dermatol 1986;115:557–62.
391–3. 49 Epstein WL, Senecal I, Krasnobrod H. Viral antigens in human epider-
24 Solomon LM, Telner P. Eruptive molluscum contagiosum in atopic mal tumours. J Invest Dermatol 1963;40:51–9.
dermatitis. Can Med Assoc J 1966;95:978–9. 50 Shirodaria PV, Matthews RS, Samuel M. Virus‐specific and anticellu-
25 Olsen JR, Piguet V, Gallacher J, Francis NA. Molluscum contagiosum lar antibodies in molluscum contagiosum. Br J Dermatol 1979;
and associations with atopic eczema in children: a retrospective longi- 101:133–40.
tudinal study in primary care. Br J Gen Pract 2015;66:e53–8. 51 Konya J, Thompson CH, de Zwart‐Steffe RT. Enzyme‐linked immu-
26 Dohil MA, Lin P, Lee J et al. The epidemiology of molluscum conta- nosorbent assay for measurement of IgG antibody to molluscum con-
SECTION 9: VIRAL SKIN

giosum in children. J Am Acad Dermatol 2006;54:47–54. tagiosum virus and investigation of the serological relationship of the
27 Berger EM, Orlow SJ, Patel RR, Schaffer JV. Experience with mollus- molecular types. J Virol Methods 1992;40:183–94.
cum contagiosum and associated inflammatory reactions in pediatric 52 Silverberg NB. Warts and molluscum in children. Adv Dermatol
INFECTIONS

dermatology practice. Arch Dermatol 2012;148:1257–64. 2004;20:23–73.


28 Hanna D, Hatami A, Powell J et al. A prospective randomized trial 53 Bugert JJ, Lohmüller C, Damon I et al. Chemokine homolog of mol-
comparing the efficacy and adverse effects of four recognized treat- luscum contagiosum virus: sequence conservation and expression.
ments of molluscum contagiosum in children. Pediatr Dermatol Virology 1998;242:51–9.
2006;23:574–9. 54 Randall CM, Biswas S, Selen CV, Shisler JL. Inhibition of inter-
29 Hellier FF. Profuse mollusca contagiosa of the face induced by corti- feron gene activation by death‐effector domain‐containing proteins
costeroids. Br J Dermatol 1971;85:398. from the molluscum contagiosum virus. Proc Natl Acad Sci U S A
30 Lerbaek A, Agner T. Facial eruption of molluscum contagiosum dur- 2014;111:E265–72.
ing topical treatment of atopic dermatitis with tacrolimus. Br J 55 Shisler JL, Moss B. Molluscum contagiosum virus inhibitors of apop-
Dermatol 2004;150:1210–11. tosis: The MC159 v‐FLIP protein blocks Fas‐induced activation of pro-
31 Ahn BK, Kim BD, Lee SJ, Lee SH. Molluscum contagiosum infection caspases and degradation of the related MC160 protein. Virology
during the treatment of vitiligo with tacrolimus ointment. J Am Acad 2001;282:14–25.
Dermatol 2005;52:532–3. 56 Shisler JL1, Senkevich TG, Berry MJ, Moss B. Ultraviolet‐induced
32 Goksugur N, Ozbostanci B, Goksugur SB. Molluscum contagiosum cell death blocked by a selenoprotein from a human dermatotropic
infection associated with pimecrolimus use in pityriasis alba. Pediatr poxvirus. Science 1998;279:102–5.
Dermatol 2007;24:E63–5. 57 Sidorenko SP, Clark EA. The dual‐function CD150 receptor sub-
33 Lim KS, Foo CC. Disseminated molluscum contagiosum in a patient family: the viral attraction. Nat Immunol 2003;4:19–24.
with chronic plaque psoriasis taking methotrexate. Clin Exp Dermatol 58 Steffen C, Markman JA. Spontaneous disappearance of molluscum
2007;32:591–3. contagiosum. Report of a case. Arch Dermatol 1980;116:923–4.
34 Cotton DW, Cooper C, Barrett DF, Leppard BJ. Severe atypical mol- 59 Yoneda K, Demitsu T, Kon A et al. Ubiquitination of molluscum body
luscum contagiosum infection in an immunocompromised host. Br J and its implications for pathophysiology. Br J Dermatol 2006;
Dermatol 1987;116:871–6. 154:786–9.
Chapter 48  Molluscum Contagiosum 587

60 Sterling JC, Kurtz JB. Molluscum contagiosum. Viral infections. In: 88 Short KA, Fuller LC, Higgins EM. Double‐blind, randomized, pla-
Champion RH, Burton JL, Burns JL (eds) Textbook of Dermatology, cebo‐controlled trial of the use of topical 10% potassium hydroxide
6th edn. Oxford: Blackwell Science, 1998:1005–8. solution in the treatment of molluscum contagiosum. Pediatr
61 Dave S, Thappa DM, Karthikeyan K. Disseminated and disfiguring Dermatol 2006;23:279–81.
molluscum contagiosum in a child. Pediatr Dermatol 2003;20:436–9. 89 Binder B, Weger W, Komericki P, Kopera D. Treatment of molluscum
62 Bunney MH, Hunter JA, Ogilvie MM. Molluscum contagiosum of the contagiosum with a pulsed dye laser: pilot study with 19 chil-
sole. A rare diagnosis or a rare condition? Br J Dermatol 1969;81:623–5. dren. J Dtsch Dermatol Ges 2008;6:121–5.
63 Whitaker SB, Wiegand SE, Budnick SD. Intraoral molluscum conta- 90 Hancox JG, Jackson J, McCagh S. Treatment of molluscum contagio-
giosum. Oral Surg Oral Med Oral Pathol 1991;72:334–6. sum with the pulsed dye laser over a 28‐month period. Cutis
64 Kose O, Seckin D. Molluscum contagiosum of the sole. J Eur Acad 2003;71:414–16.
Dermatol Venereol 2009;23:109–10. 91 Michel JL. Treatment of molluscum contagiosum with 585 nm colla-
65 Binkley GW, Deoreo GA, Johnson HH Jr. An eczematous reaction gen remodeling pulsed dye laser. Eur J Dermatol 2004;14:103–6.
associated with molluscum contagiosum. AMA Arch Derm 1956;74: 92 Lindau MS, Munar MY. Use of duct tape occlusion in the treatment
344–8. of recurrent molluscum contagiosum. Pediatr Dermatol 2004;21:609.
66 Schornack MM, Siemsen DW, Bradley EA et al. Ocular manifestations 93 Syed TA, Lundin S, Ahmad M. Topical 0.3% and 0.5% podophyllo-
of molluscum contagiosum. Clin Exp Optom 2006;89:390–3. toxin cream for self‐treatment of molluscum contagiosum in males.
67 Murray RI. Conjunctival molluscum contagiosum. Eye 2004;18:740. A placebo‐controlled, double‐blind study. Dermatology 1994;
68 Redmond RM. Molluscum contagiosum is not always benign. BMJ 189:65–8.
(Clin Res Ed) 2004;329:403. 94 Holdright DR, Jahangiri M. Accidental poisoning with podophyllin.
69 Bauer JH, Miller OF, Peckham SJ. Medical Pearl: confirming the diag- Hum Exp Toxicol 1990;9:55–6.
nosis of molluscum contagiosum using 10% potassium hydroxide. J 95 Larsson OJ, Manson ML, Starkhammar M et  al. The TLR7 agonist
Am Acad Dermatol 2007;56(5 suppl):S104–5. imiquimod induces bronchodilation via a nonneuronal TLR7‐inde-
70 Trama JP, Adelson ME, Mordechai E. Identification and genotyping of pendent mechanism: a possible role for quinoline in airway dilation.
molluscum contagiosum virus from genital swab samples by real‐ Am J Physiol Lung Cell Mol Physiol 2016;310:L1121–9.
time PCR and pyrosequencing. J Clin Virol 2007;40:325–9. 96 Katz KA. Imiquimod is not an effective drug for molluscum conta-
71 Zaballos P, Ara M, Puig S, Malvehy J. Dermoscopy of molluscum con- giosum. Lancet Infect Dis 2014;14:372–3.
tagiosum: a useful tool for clinical diagnosis in adulthood. J Eur Acad 97 Yaldo M, Caretti K, Kado J, Mehregan DR. Identifying toll‐like
Dermatol Venereol 2006;20:482–3. receptor expression in molluscum contagiosum. Int J Immunol
72 Scope A, Benvenuto‐Andrade C, Gill M et  al. Reflectance confocal Immunother 2015;2:008.
microscopy of molluscum contagiosum. Arch Dermatol 2008;144:134. 98 Maronn M, Salm C, Lyon V, Galbraith S. One‐year experience with
73 Morales A, Puig S, Malvehy J, Zaballos P. Dermoscopy of molluscum candida antigen immunotherapy for warts and molluscum. Pediatr
contagiosum. Arch Dermatol 2005;141:1644. Dermatol 2008;25:189–92.
74 Epstein WL. Molluscum contagiosum. Semin Dermatol 1992;11:184–9. 99 Ormerod AD, White MI, Shah SA, Benjamin N. Molluscum contagio-
75 Janniger CK, Schwartz RA. Molluscum contagiosum. Semin Dermatol sum effectively treated with a topical acidified nitrite, nitric oxide
1992;52:194–6. liberating cream. Br J Dermatol 1999;141:1051–3.
76 Rosdahl I, Edmar B, Gisslen H et al. Curettage of molluscum conta- 100 Watanabe T, Tamaki K. Cidofovir diphosphate inhibits molluscum
giosum in children: analgesia by topical application of a lidocaine/ contagiosum virus DNA polymerase activity. J Invest Dermatol
prilocaine cream (EMLA). Acta Derm Venereol 1988;68:149–53. 2008;128:1327–9.
77 Ronnerfalt L, Fransson J, Wahlgren CF. EMLA cream provides rapid 101 Meadows KP, Tyring SK, Pavia AT, Rallis TM. Resolution of recalci-
pain relief for the curettage of molluscum contagiosum in children trant molluscum contagiosum virus lesions in human immunodefi-
with atopic dermatitis without causing serious application‐site reac- ciency virus‐infected patients treated with cidofovir. Arch Dermatol
tions. Pediatr Dermatol 1998;15:309–12. 1997;133:987–90.
78 Simonart T, de Maertelaer V. Curettage treatment for molluscum con- 102 Zabawski EJ Jr, Cockerell CJ. Topical cidofovir for molluscum conta-
tagiosum: a follow‐up survey study. Br J Dermatol 2008;159:1144–7. giosum in children. Pediatr Dermatol 1999;16:414–15.
79 Martin‐Garcia RF, Garcia ME, Rosado A. Modified curettage tech- 103 Toro JR, Wood LV, Patel NK, Turner ML. Topical cidofovir: a novel
nique for molluscum contagiosum. Pediatr Dermatol 2007;24:192–4. treatment for recalcitrant molluscum contagiosum in children
80 Silverberg NB, Sidbury R, Mancini AJ. Childhood molluscum conta- infected with human immunodeficiency virus 1. Arch Dermatol
giosum: experience with cantharidin therapy in 300 patients. J Am 2000;136:983–5.
Acad Dermatol 2000;43:503–7. 104 Briand S, Milpied B, Navas D et al. 1% topical cidofovir used as last
81 Moed L, Shwayder TA, Chang MW. Cantharidin revisited: a blistering alternative to treat viral infections. J Eur Acad Dermatol Venereol

SECTION 9: VIRAL SKIN


defense of an ancient medicine. Arch Dermatol 2001;137:1357–60. 2008;22:249–50.
82 Epstein E. Cantharidin therapy for molluscum contagiosum in chil- 105 Sen S, Bhaumik P. Resolution of giant molluscum contagiosum with
dren. J Am Acad Dermatol 2001;45:638. antiretroviral therapy. Indian J Dermatol Venereol Leprol 2008;74:267–8.

INFECTIONS
83 Cathcart S, Coloe J, Morrell DS. Parental satisfaction, efficacy, and 106 Burke BE, Baillie JE, Olson RD. Essential oil of Australian lemon
adverse events in 54 patients treated with cantharidin for molluscum myrtle (Backhousia citriodora) in the treatment of molluscum conta-
contagiosum infection. Clin Pediatr 2009;48:161–5. giosum in children. Biomed Pharmacother 2004;58:245–7.
84 Scheinfeld N. Treatment of molluscum contagiosum: a brief review 107 Kang SH, Lee D, Hoon Park J et al. Treatment of molluscum conta-
and discussion of a case successfully treated with adapelene. giosum with topical diphencyprone therapy. Acta Derm Venereol
Dermatol Online J 2007;13:15. 2005;85:529–30.
85 Leslie KS, Dootson G, Sterling JC. Topical salicylic acid gel as a treat- 108 Gold MH, Moiin A. Treatment of verrucae vulgaris and molluscum con-
ment for molluscum contagiosum in children. J Dermatol Treat tagiosum with photodynamic therapy. Dermatol Clin 2007;25:75–80.
2005;16:336–40. 109 van der Wouden JC, van der Sande R, van Suijlekom‐Smit LWA et al.
86 De D, Gupta S, Malhotra AK. Indirect electrosurgery. Dermatol Surg Interventions for cutaneous molluscum contagiosum (Review).
2006;32:1424. Evid‐Based Child Health 2011;6:1550–99.
87 Mahajan BB, Pall A, Gupta RR. Topical 20% KOH – an effective thera- 110 Smith KJ, Skelton H. Molluscum contagiosum: recent advances in
peutic modality for molluscum contagiosum in children. Indian J pathogenic mechanisms, and new therapies. Am J Clin Dermatol
Dermatol Venereol Leprol 2003;69:175–7. 2002;3:535–45.
588 

CHA PTER  4 9

Human Papillomavirus Infection


Yun Tong1, Stephen K. Tyring2 & Zsuzsanna Z. Szalai3
1
 Department of Dermatology, University of California San Diego, San Diego, CA, USA
2
 Department of Dermatology, University of Texas Health Science Center at Houston, Houston, TX, USA
3
 Department of Pediatric Dermatology, Heim Pál Children’s Hospital, Budapest, Hungary

Abstract i­ nfections are asymptomatic and cleared by the immune system,


continued cutaneous manifestations including genital warts are
a sign of persistent disease. Persistent infections by highly onco-
Human papillomaviruses (HPV) are a group of viruses with
genic HPV strains can lead to anogenital and/or oropharyngeal
implications ranging from cutaneous warts to cancer. HPV is
­
cancers.
the most common sexually transmitted disease. Although most

Key points • Clinical appearance varies according to the type of virus,


anatomical site and host’s response to the infection.
• HPV are separated into high‐ and low‐risk types.
• Over 170 types of HPV have been identified.
• Viral DNA integration into host genome confers higher‐risk
• HPV infection occurs when the virus is able to cross the basal
lesions/tumours.
lamina through disruption of the skin barrier or establish at
• Up to two thirds of noncancerous cutaneous HPV manifestations
naturally present squamocolumnar junctions (e.g. endocervix).
may regress spontaneously within 2 years.
• Most HPV are transient without clinical symptoms.
• Multiple treatment modalities are available because no single
• HPV infection is the most common sexually transmitted
treatment has been universally successful.
disease.
• The 9‐valent vaccine immunizes against the HPV types that cause
• HPV infections are classically separated into cutaneous, mucosal
~90% of genital warts (types 6 and 11) and ~90% of cervical
and epidermodysplasia verruciformis (EV) types.
cancers (16, 18, 31, 33, 45, 52 and 58).

investigators determined the function of these genes. The


Introduction. Human papillomaviruses (HPV) are a
recognition and increasing appreciation of the fact that a
group of ubiquitous viruses that infect the basal
subset of HPV was closely associated with cervical cancer
keratinocytes of the skin and together are recognized as
led to the establishment of HPV as the new model of viral
the most common sexually transmitted disease (STD) in
tumorigenesis [2–4].
the USA. Belonging to the family Papillomaviridae (PV),
approximately 170 types of papillomaviruses have been
identified that infect humans, with most having medical Classification. The International Committee on the Taxonomy
SECTION 9: VIRAL SKIN

significance. Most HPV types are asymptomatic, and of Viruses designated papillomaviruses, Papillomaviridae,
patients typically are unaware of their infection [1]. HPV as a separate family. PV are highly species specific; there
is a circular, nonenveloped, double‐stranded DNA virus. are a few hundred types of papillomaviruses. Over 170
INFECTIONS

HPV is packaged into icosahedral capsid virions that HPV types have been sequenced. A range in homology has
measure approximately 55 nm in diameter (Fig. 49.1). led to the following classifications: genus, species, type,
Warts, the common clinical manifestation of HPV, were subtype and variant. There are 12 genera, designated by
recognized as infectious diseases by the ancient Greeks the first 12 letters of the Greek alphabet, of which five gen-
and Romans. Since the early twentieth century, warts have era (α, β, γ, μ, ν) contain HPV types [2,5,6]. Historically,
been viewed as viral infections. After it was demonstrated HPV has been classified into cutaneous, mucosal and epi-
in the early 1900s that the virus was transmitted from cell‐ dermodysplasia verruciformis (EV) types. HPV types are
free filtrates of warts, papillomaviruses were identified in also separated into high or low risk based on their predis-
a number of vertebrate species. Molecular cloning led to a position to malignancy. α‐genus HPV types, which infect
scientific development which largely enhanced the study the anogenital region and oral cavity contain both high‐
of the biological and biochemical properties of papilloma- and low‐risk types. Verrucae and papilloma types from β,
viruses. Sequencing allowed the identification of the open γ, μ and ν genera are considered low risk because infection
reading frames (ORF) as putative viral genes and the generally does not progress to cancer [7].

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 49  Human Papillomavirus Infection 589

After viral capsid endocytosis into the host cell, the HPV
genome travels to the host nucleus where it uses host cell
machinery to maintain low copies of itself (~200 copies/
cell) through its normal life. However, in high‐grade cervi-
cal lesions and tumours, the viral DNA is integrated into the
host genome. As the infected cells leave the basal layer, viral
E6 and E7 proteins promote cell cycle continuation.
HPV induces immunomodulation resulting in local
impairment of the immune response [11,12]. The immune
defence of the infant, HLA type, the presence of HPV anti-
bodies, and differences in the host genome may protect
the young child.
The rare autosomal recessive condition, epidermodys-
plasia verruciformis (EV), which starts in early infancy
or  childhood, is associated with an abnormal immune
response to HPV [13]. Secondary immunodeficiency
states  –  acquired immune deficiency syndrome, malig-
nancies or organ allograft recipients as a result of immu-
nosuppressive therapy – are also associated with frequent
and persistent HPV infection [14]. Warts may have a
­tendency to either disappear or be refractory to all treat-
Fig. 49.1  Structure of the human papillomavirus. ments. Resolution rates may be adversely influenced by
the HPV type (as with mosaic warts), the extent and dura-
tion of the warts and suppression of the host’s cell‐­
The genome consists of approximately 8000 base pairs mediated immune response [3].
and is divided functionally into three regions: an early,
a  late and a noncoding long region. The early region WHIM syndrome
­contains six common ORF and encodes genes for viral This is the association of warts, hypogammaglobulinaemia,
transcription, cellular transformation and DNA replica- recurrent bacterial infections and myelokathexis. Severe
tion. The late region encodes for structural proteins, major chronic neutropenia appears with hyperplasia of the
capsid protein L1 and minor capsid protein L2 [8,9]. HPV mature myeloid compartment in the bone marrow [15].
typing is based on >10% dissimilarity in the highly con- WHIM syndrome is associated with a heterozygous
served L1 ORF nucleotide sequence [10]. The regulatory truncating mutation of the CXCR4 gene. The CXCR4
or long control region, which is located between the early receptor is bound by CXCL12, a chemokine that regulates
and late regions, contains transcription enhancer genes cardiogenesis and haematopoiesis, among others [4].
and controls viral replication and gene expression.
Mode of transmission
Epidemiology and  pathogenesis. Most HPV infections Subclinical and latent forms
are transient with no clinical symptoms. A minority of Among healthy individuals, persistent subclinical infec-

SECTION 9: VIRAL SKIN


HPV types can result in warts or malignancies. HPV tions can be demonstrated on mucosal surfaces by appli-
infection is responsible for 99.7% of cervical cancer cases cation of 3% acetic acid, as in the procedure of colposcopic
and is the fourth most frequent cause of death from can- examination of the cervix or genital tract. High‐risk HPV

INFECTIONS
cer in women [1]. Cutaneous infections are essentially DNA can be detected in the oral and genital mucosa
ubiquitous. About 40% of childhood warts resolve within of infants during the first 3 years of life and occasionally
2 years. The incubation period of HPV varies from several in immunocompetent and immunosuppressed individu-
weeks up to more than a year. Viral warts can occasion- als [12]. Latent papillomavirus represents the presence
ally persist for many years in otherwise healthy individu- of  HPV DNA in clinically and histologically normal
als. Older individuals may have some immune resistance skin  and mucosa. HPV infections might be transmitted
to infection because there is a low number of cases in this horizontally from other family members. It is important
population [3]. to remember this to decrease the number of false allega-
The human clearance of HPV involves a protective skin tions of sexual abuse. Identification of possible sexual
barrier and interactions between innate immunity and abuse is still by detailed and appropriate history taking,
acquired immunity. Early exposure to the wart virus may physical examination and correlation with the socio‐
confer a period of immunity, especially in a young child clinical context [16].
with a few asymptomatic warts that are likely to resolve HPV can often be detected in common dermatoses,
spontaneously within months. leading to speculation that it may play a role in the devel-
Infection occurs when HPV is able to access the basal opment of conditions such as psoriasis vulgaris, lichen
lamina and its layer of keratinocytes, either through skin sclerosus and naevus sebaceus [17]. EV HPV types may
barrier disruption or at anatomical places where the squa- play a role in the hyperproliferation of skin. Anti‐HPV‐5
mocolumnar junction is naturally present (e.g. endocervix). antibodies have been demonstrated in epidermal repair
590 Section 9  Viral Skin Infections and Opportunistic Infections

processes (second‐degree burn, autoimmune skin diseases, Sexual transmission


bullous disorders, psoriasis vulgaris). The mechanism by The prevalence of childhood HPV infection varies widely
which hyperproliferative HPV types may trigger a wide- in the literature. Most children who have been sexually
spread epidermal disorder is currently unknown [18]. abused will not show carriage of the virus. The minimal
period from sexual HPV exposure to development of
Autoinoculation external genital warts (EGWs) in adolescents and adults
An important mode of spreading the virus is autoinocula- is about 3 months, but is unknown in children [28–30].
tion (Fig. 49.2). The leading sites of HPV infection are the Autoinoculation or heteroinoculation from nongenital
extremities and the face. Hand warts are transferred by warts may be a source of EGWs in children. It has been
contact to other cutaneous sites including the lips, nose recognized that the phylogenetically related mucosal
and face [19]. Infection occurs when the basal epithelial HPV types 2, 27 and 57 can be detected in cutaneous
cells of the mucosa or skin are exposed to the virus. lesions as well as oral and genital lesions [12]. HPV‐27
Trauma also facilitates the spread of viruses [20]. with HPV‐3 is commonly found as a nongenital HPV in
genital lesions of children. HPV subtypes in children do
Heteroinoculation and iatrogenic transmission not show the high degree of site specificity for either
HPV infection plays an important role in the transmission mucosal or cutaneous sites that is seen in adults [19].
of HPV DNA between family members, and may be
transmitted via saliva during an infant’s nursing. The Clinical features
mother’s warts are associated with persistent oral and HPV infections of the skin
genital HPV carriage [12]. Deficiencies in cell‐mediated The clinical appearance of viral warts varies according to
immunity cause problems with persistent infection. the type of the virus, the anatomical site involved and the
Viral warts are spread by direct contact through host’s response to the infection [14].
fomites or indirectly by contaminated surfaces [21]. Clinical appearance can be defined by morphology
Iatrogenic transmission of the virus may occur indirectly ­(common warts, flat warts, filiform warts, tinea versi-
through the use of inadequately sterilized instruments color‐like warts, doughnut warts, epidermal cyst‐type
[20,22]. Papillomaviruses can remain infectious for long warts, punctate warts, pigmented warts, keratoacanthoma,
periods. Viral transmission is facilitated by maceration of subclinical or latent infection) or by location (common
the skin. There is an increased risk of reinfection in those warts, palmoplantar warts, periungual warts, respiratory
who have had viral warts previously [23]. The most papillomatosis, oral papillomatosis [Heck disease], con-
­frequently reported risk factors for genital HPV infec- dyloma acuminatum, verrucous carcinoma). Warts can
tions are related to sexual behaviour, the partner’s sexual also be categorized by the host immune response [4].
history and age [24–26]. Common warts occur primarily in children and repre-
sent 70% of all skin warts (Fig.  49.3). Plantar and flat
Vertical transmission warts occur in a slightly older population. Warts can be
HPV infection of normal skin is acquired very early in painful when involving the soles or located near the nails,
infancy. HPV transmission from mother to neonate may
occur either in utero or during delivery; the resulting
laryngeal papillomas or anogenital warts may become
clinically apparent only months or years later. Viraemia
SECTION 9: VIRAL SKIN

has not been shown to occur with HPV [19,27].


INFECTIONS

Fig. 49.2  Autoinoculation of warts from the periungual lesion to the


mouth. Fig. 49.3  Common skin wart on the scrotum of an infant.
Chapter 49  Human Papillomavirus Infection 591

especially when inflammation develops. If they are carcinoma in situ) [3]. Regression of these lesions may
located on visible areas, they can be socially unaccepta- occur, which is usually heralded by inflammation. Flat
ble  [31]. Common warts present in a variety of clinical wart HPV types include 3, 10, 28, 41 and 49, followed by
patterns, appearing as hyperkeratotic papules with a types 5, 8, 12, 14, 15, 17, 25 and 30.
rough, irregular surface, and can occur in almost any area Deep plantar warts/myrmecia. The name ‘myrmecia’ orig-
of the body. HPV types 2 and 4 are the most common, fol- inates from the Greek meaning ‘ant hill’. The lesions
lowed by types 1, 3, 7, 10, 26, 27, 29 and 57. Common appear as small papules and progress to deep lesions
warts may become exophytic and vary in size from tiny with a rough keratotic surface. A well‐demarcated rim of
flesh‐coloured papules to hyperkeratotic fissured masses compressed keratin surrounds a softer substance. Plantar
1–2 cm in diameter. Thrombosed capillaries are often seen warts often grow into the deeper layers of skin because of
after paring away the hyperkeratotic tissue [14]. the pressure (callus‐like warts typically involve HPV‐1,
Filiform/digitate warts are long, with a narrow base and ‐4, ‐7, ‐10). Plantar warts should be treated to lessen pain
verrucous tip, and are usually seen on the face and neck, and transmission of infection [14]. The incubation period
around the lips, eyelids or nares. These variants of com- is 1–20 months. They grow deep and tend to be more
mon warts are benign and easy to treat [4]. painful than common warts. Treatment responsiveness
Verrucae planae/plane warts/flat warts are more common is  generally good. The most common type of deep
among children and young adults and develop by autoin- ­palmoplantar wart is HPV‐1, followed by HPV‐2, ‐3, ‐4,
oculation. They are smooth, flesh‐coloured or lightly pig- ‐27, ‐29, ‐57 and ‐63.
mented yellow‐brownish papules with hyperkeratosis, Mosaic warts are formed by smaller plantar warts, which
usually 2–4 mm in size. These lesions are common on are composing plaques. They are coalescent and are usu-
photo‐exposed areas such as the face and neck and the ally seen on the palms and soles. Mosaic warts are impor-
dorsum of the hands. The Koebner phenomenon (or, more tant for their persistence and difficulty of treatment.
properly, pseudokoebnerization) often appears (Fig. 49.4). Mosaic wart types are HPV‐1, ‐4, ‐7 and ‐10 (Fig. 49.5) [4].
HPV‐10 and ‐28 related plane warts are more hyperkera- Punctate warts (HPV‐60, ‐63, ‐65) are rare and consist of
totic and can resemble early common warts. localized endophytic hyperkeratotic papules, usually
When flat warts are spread over a wide surface area, located on the palms. Occasionally warts can present a
they may be slightly hyper‐ or hypopigmented, and may high degree of pigmentation (HPV‐60), mimicking a pri-
appear like tinea versicolor. This appearance is not usual mary melanocytic process.
and is more commonly limited to patients with EV, human Periungual and subungual warts (benign HPV‐1–4, ‐7,
immunodeficiency virus (HIV) infection or other immune ‐10; premalignant HPV‐16, ‐34) are painful; their treat-
deficiency. In EV, warts spread rapidly and may progress ment is often difficult due to the physical blockade cre-
to bowenoid papulosis or Bowen disease (squamous cell ated by the nail itself. When treating a wart in this location,
the nail matrix may incur damage (Fig. 49.6).
A cystic wart appears as a nodule on the weightbearing
surface of the sole (plantar epidermoid cyst). The nodule
is usually smooth but may become hyperkeratotic. If the
lesion is incised, cheesy material may be expressed. Cystic
wart HPV types are 60, 63 and 65 [32].
Doughnut/ring warts are seen after the use of destructive

SECTION 9: VIRAL SKIN


therapies. They occur around the periphery of a wart that
has been treated previously (Fig. 49.7).
Butcher’s wart. HPV types are 1, 4, 7 and 10. These are

INFECTIONS
seen in people who frequently handle raw meat. Their

Fig. 49.4  Verrucae planae on the skin – linear appearance due to


scratching. Fig. 49.5  Mosaic wart on the sole.
592 Section 9  Viral Skin Infections and Opportunistic Infections

develop into squamous cell cancers. There are two pre-


dominant types of lesions. Some lesions have the appear-
ance of flat warts, caused by the same HPV types that
induce flat warts in the general population (HPV‐3 and
‐10). The others are flat, scaly, red‐brown macules, associ-
ated with EV‐specific HPV types, most frequently HPV‐5
and HPV‐8 [17]. The lesions are resistant to the usual
treatment modalities. They develop on light‐exposed skin
sites in most cases. EV occurs as an inherited disorder in
about half of the affected patients, inherited in an autoso-
mal recessive pattern or X‐linked recessive inheritance.
p53 mutations are also common. The presence of EV but
not cutaneous HPV DNA in normal skin was significantly
associated with nonmelanoma skin cancer status and
may prove to be of predictive value for skin cancer risk.
The EV HPV types play a role as causal agents in skin
cancer [13,17].

HPV infections of the aerodigestive tract


Focal epithelial hyperplasia. Focal epithelial hyperplasia
Fig. 49.6  Periungual wart. (Heck disease) is an HPV infection in the oral cavity [3].
The lesions are mucosa‐coloured, flat‐topped or dome‐
morphology is similar to common warts, with a higher shaped, pink‐white papules. They are usually 1–5 mm,
prevalence of hyperproliferative cauliflower‐like lesions. with some lesions coalescing into plaques, and are seen in
Keratoacanthomas have been associated with HPV‐37 many ethnic groups but are most common in native
and ‐77, and can progress to squamous cell carcinoma. Americans and Inuit children. Persistent oral HPV car-
Bowenoid papulosis is a form of squamous cell carcinoma riage in infancy is associated with the mother’s high‐risk
in situ characterized by enlarging plaques of the genital HPV carriage [12]. Cryotherapy or carbon dioxide laser
region. It can occur in immunocompetent and immuno- therapy has been helpful in these patients. Spontaneous
suppressed children. The known HPV types include 1, 16, regression follows a similar time schedule to standard
18, 33 and 34; in verrucous carcinoma, the HPV types wart resolution. Aggressive therapy should be reserved
include 1–4, 6, 11, 16 and 18 [22]. for cases with a protracted course (Fig. 49.8).
Epidermodysplasia verruciformis (EV) is a rare disorder.
Warts generally develop in childhood, become wide- Recurrent respiratory papillomatosis. Recurrent respira-
spread, do not tend to regress, and in certain cases may tory papillomatosis (RRP) in childhood occurs at an inci-
dence of 0.3–3.9/100 000 and is considered to be the most
common benign tumour that affects the larynx in chil-
dren. RRP is a life‐threatening benign tumour as it has the
tendency to grow in size and number, causing complete
SECTION 9: VIRAL SKIN

airway obstruction [31]. This disease presents with


INFECTIONS

Fig. 49.7  Doughnut wart. Fig. 49.8  Oral papillomas on the mucosal part of the lip (Heck disease).
Chapter 49  Human Papillomavirus Infection 593

hoarseness, stridor, cough and dyspnoea and is often mis- Anogenital warts: condylomas. Anogenital warts in chil-
taken early on for asthma or laryngeal haemangioma. dren are rare, much less common than in adults. The dis-
HPV types related to respiratory papillomatosis are 6, 11, covery of EGWs in children raises concerns regarding
16 and 18. HPV‐11 is considered to be the most common sexual abuse, but it is often difficult to definitively deter-
cause of RRP [4]. Diagnosis of RRP is confirmed by direct mine. Most genital HPV infections are asymptomatic and
laryngoscopy and biopsy for tissue diagnosis. The man- transient [12]. The mean age at presentation in children
agement of RRP includes surgical removal, various lasers ranges from 2.8 to 5.6 years. Due to potentially long
and adjuvant pharmacological medical therapies with the latency periods, several possible modes of transmission
antiviral agents cidofovir or interferon [3]. may occur [31]. Most condylomas are self‐limiting and
regress spontaneously or with local treatment, but some
Genital HPV infection may persist for years.
HPV infection of the genital tract is the most common Children may present with small flesh‐coloured pap-
sexually transmitted virus. The prevalence is age depend- ules noted by their caregiver. Some warts can be a source
ent: women between 15 and 25 years have the highest of itching, burning and bleeding. It remains difficult to
prevalence, which may be about 25–40% of all infections determine with certainty the source of HPV contamina-
[16]. The majority of precursor lesions to cervical intraepi- tion in children with anogenital warts [16]. Of all cases of
thelial neoplasia (CIN), vulvar intraepithelial neoplasia genital warts, about 90% are caused by HPV types 6 or 11,
(VIN) and vaginal intraepithelial neoplasia (VaIN) are with type 6 predominating and accounting for 39–90%. It
caused by HPV infection. The five most common HPV may be possible that genital skin in children is more vul-
types in HPV‐positive women worldwide are 16, 18, 31, nerable to infection by nongenital HPV types [19]. Genital
58 and 52, representing approximately 50% of all HPV types of HPV are divided into high or low risk depending
infections [3]. High‐ and low‐risk HPV infections are on their potential for causing anogenital neoplasia.
often simultaneously observed. Persistent carriage of
high‐risk HPV types was detected in 10% of oral and Penile warts. Condylomata acuminata in boys may also
1.5% of genital mucosa specimens obtained from all develop on the penis, most commonly involving the glans
infants during their first 26 months of life [12]. HPV has penis, penile shaft and prepuce. HPV‐16 has the highest
been detected in newborn babies as well [27]. The infants prevalence, but multiple HPV types may also exist [28]
are infected by periconceptual transmission, by prenatal (Fig. 49.10).
transmission via placenta, amniotic fluid or cord blood or
by passive contamination going through the birth canal. Histology. HPV infection of the skin and mucosa results
Persistence of HPV carriage in children varies in duration in hyperkeratosis and causes squamous cell prolifera-
from 2 days to up to 3 years [33]. tion,  parakeratosis and acanthosis of the epidermis.

Vulvovaginal warts. The lesions can develop rapidly into


clusters of flat, papillomatous and occasionally cauli-
flower‐like lesions (HPV types are 6 and 11) (Fig. 49.9). In
cases of children with EGWs, coexistent HPV‐16 has been
seen in cervicovaginal or intra‐anal samples, although its
oncogenic potential in children is unclear. A slow‐growing,

SECTION 9: VIRAL SKIN


cauliflower‐like mass, known as giant condyloma acumi-
natum, is a regional variant of verrucous carcinoma. It is
extremely rare and occurs in adulthood [4,30].

INFECTIONS

Fig. 49.9  Condyloma of the vulva. Fig. 49.10  Condyloma on the glans and foreskin of an infant.
594 Section 9  Viral Skin Infections and Opportunistic Infections

Characteristic histological features include vacuolation of preparations should be avoided in children because of
the keratinocytes, with inclusion bodies composed of possible systemic toxicity [38].
viral particles and abnormal keratin [14]. In cases of deep Cantharidin, derived from the blister beetle Cantharis
plantar warts, the cells are enlarged, irregular and vacu- vesicatoria, can have a cure rate of up to 80% in plantar and
olated, and eosinophilic cytoplasmic inclusions, either periungual warts [39]. Its application causes activation of
sickle‐ or ring‐shaped, are distributed throughout the proteases, which degrade desmosomal attachments lead-
entire stratum spinosum, more dense superficially. ing to acantholysis and possibly painful blister formation.
Anogenital warts are markedly acanthotic and papil- No longer available in the USA since 1992, cantharidin
lomatous with little hyperkeratosis but some parakerato- can be purchased over the counter in Canada or be com-
sis [34]. Common warts have an endophytic, papilliferous pounded by a pharmacist. Unlike salicylic acid, canthari-
architecture with an acanthotic hyperplastic epidermis din is to be applied in‐office only as oral consumption can
and prominent granular layer. Viral DNA can be detected lead to death [35,39]. Use on thin‐skinned areas such as
in suprabasal cells in which the early cytopathic effect of a the face or genital areas may produce more significant blis-
perinuclear halo is observed. These changes become pro- tering, so use at these sites should be carefully considered.
gressively more pronounced in the superficial layers of the
epidermis. Some infected cells may have characteristic Cryotherapy.  Traditionally, if simple topical treatments
cytoplasmic vacuoles (‘balloon cells’ or koilocytes) [14]. fail, cryotherapy is a routine second‐line treatment. It is
often considered as a first‐line therapy for treatment of
facial or genital warts as it is inexpensive and easy to
Differential diagnosis. Pseudoverrucous nodules and
administer. It has few side‐effects but can be painful and
benign and malignant tumours must be differentiated
may induce haemorrhagic blisters. Among refrigerant
from warts. The association of the wart virus with tumour
agents, liquid nitrogen is the coldest and most effective.
development may mean that verrucous carcinomas and
Care must be taken to avoid excessive deep freezing,
warts can exist side by side.
which can damage underlying structures (tendons, inter-
phalangeal joints and peripheral nerves) and cause scar-
HPV genotyping
ring. The infecting HPV is not killed by cryotherapy, but
The methods for detecting HPV of verrucae vulgaris are
its release from the damaged cells of the treated wart will
based on polymerase chain reaction (PCR) followed by
encourage an immune response to the virus [38,40].
sequencing or hybridization [6]. A negative test does not
mean that HPV was not previously present as HPV infec-
Occlusotherapy (adhesiotherapy).  Occlusion therapy
tion can be transient and the virus can be eliminated by a
using duct tape for 2 months can be effective with common
healthy immune system to undetectable levels [19].
warts. Occlusion in conjunction with other wart therapies
Multiplex PCR allows detection and typing of HPV DNA
such as salicylic acid can enhance drug efficacy. Success
simultaneously [24].
rates can be high with relatively limited side‐effects [38].

Treatment and prevention. Many noncancerous cutane- Curettage and surgical treatment. With all surgical treat-
ous manifestations of HPV may be self‐limiting and ments for warts, there is a risk of recurrence because of
regress spontaneously. Up to two thirds of warts may residual wart tissue or recrudescent latent virus adjacent
clear without treatment within 2 years [35]. Several thera- to the original wart. Electrocautery and hyfrecation can be
peutic options exist for viral warts [36,37]. No definitive used to destroy small filiform warts, especially on the
SECTION 9: VIRAL SKIN

single therapy cures all warts [35]. The choice of treat- face, but run the risk of aerosolizing viral particles.
ment depends on different considerations. Patients often Pretreatment with local anaesthetic cream is effective in
INFECTIONS

have different skin types, HPV numbers, sizes, locations relieving the pain [20,41].
and durations of lesion, immune statuses, other comorbid
skin or internal diseases, different pain thresholds and Laser treatment. The most represented laser modalities in
different preferences [38]. Parents have different attitudes the literature for warts are carbon dioxide and pulsed‐dye
and timetables. lasers (PDL), which produce comparable cure rates to
multisession destructive therapy. The routine use of laser
Destructive therapies treatment is not cost‐effective and it is painful. Its higher
Destructive agents cause nonselective damage to the cost is perhaps balanced by decreased number of required
infected keratinocytes. Destructive modalities tend to treatment sessions. Compared to a carbon dioxide laser,
require multiple treatments and have high recurrence PDL is more selective by causing direct thermal damage
rates. Most destructive therapies may cause scarring. to the wart microvasculature [35]. The success of PDL
treatment depends upon adequate energy absorption
Keratolytic agents. Among the destructive methods, sali- within the capillary loops of the wart; it produces more
cylic acid has the best clinical trial data supporting its purpura but less tissue damage.
usage. Topical treatment containing salicylic acid is effec-
tive and safe; it produces a local irritant effect. A number Antiproliferative agents and antimitotics
of placebo‐controlled trials document a 50–75% rate of Systemic retinoids may have a place in the management
cure with 6 weeks’ usage [4]. The more concentrated of persistent warts in adults, but only in selected
Chapter 49  Human Papillomavirus Infection 595

­ aediatric cases. Retinoids disrupt epidermal growth and


p Intralesional agents.  Intralesional therapy utilizes a
differentiation. Podophyllin is a cytotoxic agent with delayed‐type hypersensitivity response towards warts.
powerful irritant effects and is also teratogenic. Due to Interestingly, it has the potential to resolve warts distant
its cytotoxic effect, it is not routinely recommended. to those being treated [46]. Intralesional agents include
Intralesional bleomycin is unlicensed for treatment of Candida antigen, the measles–mumps–rubella (MMR)
children. 5‐fluorouracil is a chemotherapeutic agent vaccine, trichophytin and bacille Calmette–Guérin (BCG)
thought to interfere with DNA and RNA synthesis and vaccine.
needs further investigation before paediatric use can be
routinely advised. Interferon. In warts, the T lymphocyte CD4/CD8 ratio is
inverted, which improves following interferon therapy.
Virucidal therapy Locally used interferon therapy is more efficacious than
Cidofovir, an acyclic nucleoside phosphonate, acts as a systemic administration [47]. Patients administered
dead‐end substrate for DNA polymerase, stopping repli- twice‐weekly injections for up to 8 weeks saw 62% clear-
cation. Cidofovir has been used successfully in HIV‐­ ance of warts versus 21% in placebo groups [46].
positive patients for the topical treatment of genital warts
[42]. Off‐label intralesional use in children for RRP has Contact sensitizers.  Contact sensitizers elicit a type IV
shown partial to complete resolution of mucosal papillo- hypersensitivity reaction. Agents include diphencyprone
mas [39]. Intravenous administration of cidofovir requires (DPCP), squaric acid and dinitrochlorobenzene (DNCB)
more investigation before paediatric use. Topical use of [39]. DNCB has been shown to be mutagenic and should
compounded 1–3% formulations of cidofovir has also be avoided. A retrospective study with paediatric subjects
been reported to be effective, but care should be taken in showed that DPCP may be the preferred method for peri-
areas where skin is eroded or thin or occluded due to risks ungual warts (85% of patients saw clearance) but requires
of systemic absorption. more investigation [48]. Although these treatments may
be efficacious, limited data support their use, and they do
Immune stimulation and modulation not have any significant advantage over simpler and safer
Ingenol mebutate.  Approved for actinic keratosis, treatments, except perhaps in selected cases [49].
­ingenol mebutate has a dual mechanism of inducing
cell death directly and stimulating an inflammatory Systemic immune stimulation.  Immunostimulatory
response against tumour cells. In a case series of 17 drugs such as levamisole and inosine pranobex, and the
patients (21–70 years old), 16 patients demonstrated H2‐receptor antagonist cimetidine, which also has immu-
complete clearance of their EGWs, 76% of whom had nomodulatory actions, proved disappointing in the man-
rapid clearance within 3–35 days [43]. Thirteen of the 16 agement of cutaneous warts. While cimetidine appears to
who had complete clearance required only a single show a trend towards improvement in paediatric patients,
treatment of ingenol mebutate; the remaining three randomized controlled trials do not demonstrate statisti-
patients cleared after a second or third application. No cally significant differences in comparison to placebo,
recurrence was noted up to 240 days on follow‐up. particularly in adults. Zinc sulfate administration orally,
Local skin irritation similar to application for actinic which can be associated with gastrointestinal adverse
keratosis was noted after 24–48 hours but ameliorated effects when used at high doses, is effective but only in
within 2–5 days. cases of zinc deficiency [14,34,37,50].

SECTION 9: VIRAL SKIN


Imiquimod.  Approved by the US Food and Drug Vaccination. The multivalent HPV vaccines are infection‐
Administration (FDA) in 1997 for external and perianal prevention‐focused, component vaccines made up of

INFECTIONS
warts, imiquimod is a topical immune response cream virus‐like particles produced from the L1 surface proteins
applied to the affected area, which induces production of of HPV types 16 and 18 for the bivalent (Cervarix) and
interferon‐α, tumour necrosis factor (TNF)‐α and interleu- HPV types 6, 11, 16 and 18 for the quadrivalent (Gardasil)
kin (IL)‐1, IL‐6 and IL‐8. Used for anogenital warts in vaccines. Both HPV vaccines protect against highly onco-
adults, there has been some success in its use for anogeni- genic HPV‐16 and ‐18 infections, which together account
tal warts in children [44,45]. Imiquimod is well‐suited for for more than 70% of cervical cancer cases. Gardasil also
warts in other anatomical sites in children and has the covers against HPV types 6 and 11, which are the cause of
benefit of home use [35]. 90% of anogenital warts. Just 6 years after vaccine intro-
duction, the prevalence of the four HPV types covered by
Polyphenon E (sinecatechins).  Botanical polyphenol the vaccine had dropped by 64% among 14‐ to 19‐year‐
sinecatechins ointments isolated from green tea are old girls. Perhaps equally important, there has been no
approved in the USA and Europe for the treatment of indication of type replacement occurring [51].
genital warts. Polyphenon E has not been studied as A next‐generation vaccine protecting against nine HPV
extensively for cutaneous warts. An antioxidant, poly- types (Gardasil‐9) was approved in 2014 by the FDA to
phenon E, stimulates pro‐inflammatory cytokine release succeed the original Gardasil. Gardasil‐9 potentially
causing telomerase inhibition and apoptosis [39]. A 15% extends an additional 20% of coverage by including five
ointment formulation demonstrated up to 65% clearance more high‐risk HPV types: 31, 33, 45, 52 and 58 [52].
with low recurrence rate of 5.9–12% [39,46]. High‐risk HPV types are also significantly involved in
596 Section 9  Viral Skin Infections and Opportunistic Infections

HPV‐positive cancers of the penis, vulva, vagina and External genital warts. The diagnosis of EGWs is usually
anus as well as the head and neck. a clinical one and should be differentiated from anatomi-
Clinical trials have shown that the HPV vaccines are cally normal findings. A detailed history from the child
highly immunogenic, safe and well tolerated in females and family, a thorough medical examination for cutane-
and males 9–26 years of age. Their efficacy remains high ous warts, and consideration of referral of family mem-
for at least 10 years following vaccination. Currently, bers for examination are needed. Examination of the child
the rationale for routine immunization at 11–12 years of for signs of physical injury should be undertaken. The
age is that the vaccine should be given before a child majority of children who have been sexually abused have
becomes sexually active. The vaccine is currently no genital or perineal injury on examination. A physical
administered as a three‐dose regimen in the USA, but examination is needed to exclude other STDs. Suspected
many countries including the UK have switched to a sexual abuse cases should be reported to appropriate
two‐dose regimen, which demonstrated noninferiority authorities. Children with EGWs should be treated and
[53,54]. As the HPV vaccine rate is variable and not followed up for signs of recurrence. A comprehensive,
expected to prevent infection attributable to all high‐ evidence‐based HPV educational protocol is effective
risk HPV types, cervical cancer screening recommenda- regardless of adolescent sociodemographic characteris-
tions, including Papanicolaou testing, should continue tics or risk behaviours. Written brochures and internet‐
to be followed for females who have received HPV based materials are useful. Education by school health
­vaccination [25,31,55,56]. educators must include the safety and efficacy of HPV
While the purpose of the HPV vaccines has been to vaccines, recommendations for vaccination, and the
­prevent primary infection, there are reports of therapeutic importance of safe sexual behaviours and continued
effect for both vaccine‐ and nonvaccine‐type HPV warts Papanicolaou screening after vaccination [19,58,59].
[57]. This might perhaps add support of cross‐protection
due to homologous antigenic epitopes to nonvaccine ­References
strains. 1 Handler MZ, Handler NS, Majewski S, Schwartz RA. Human pap-
illomavirus vaccine trials and tribulations: Clinical perspectives.
J Am Acad Dermatol 2015;73:743–56.
Phototherapies 2 Forslund O. Genetic diversity of cutaneous human papillomavi-
Photodynamic therapy uses light at a wavelength absorbed ruses. J Gen Virol 2007;88:2662–9.
by a photosensitizer such as 5‐aminolaevulinic acid applied 3 Hoory T, Monie A, Gravitt P, Wu T‐C. Molecular epidemiology
of  human papillomavirus. J Formos Med Assoc Taiwan Yi Zhi
to target tissue. Its use for children is limited because it is 2008;107:198–217.
painful and because of possible side‐effects [34]. 4 Silverberg NB. Human papillomavirus infections in children. In:
Schroten H, Wirth S (eds) Pediatric Infectious Diseases Revisited.
Birkhäuser Advances in Infectious Diseases. Basel: Birkhäuser,
Combination therapies 2007:365–90. (). Available from: http://link.springer.com/chapter/
There is little documented evidence to support combina- 10.1007/978‐3‐7643‐8099‐1_14 (accessed 1 January 2019).
tion therapy and more research is needed regarding 5 Chan SY, Chew SH, Egawa K et  al. Phylogenetic analysis of the
­possible associated safety and efficacy. Some examples human papillomavirus type 2 (HPV‐2), HPV‐27, and HPV‐57
group,  which is associated with common warts. Virology 1997;
of  combination therapy suggested for use in paediatric 239:296–302.
patients have included cryotherapy + salicylic acid, local 6 de Villiers E‐M, Fauquet C, Broker TR et al. Classification of papillo-
retinoid + inosiplex and cimetidine + levamisole, but maviruses. Virology 2004;324:17–27.
7 Nguyen HP, Ramírez‐Fort MK, Rady PL. The biology of human
there is no evidence of usefulness [37].
papillomaviruses. Curr Probl Dermatol 2014;45:19–32.
SECTION 9: VIRAL SKIN

8 Wang X, Meyers C, Wang H‐K et sl. Construction of a full transcrip-


Factors to consider in the treatment of warts tion map of human papillomavirus type 18 during productive viral
in children infection. J Virol 2011;85:8080–92.
INFECTIONS

9 Pahud BA, Ault KA. The expanded impact of human papillomavirus


It is important to avoid pain and decrease tissue damage vaccine. Infect Dis Clin North Am 2015;29:715–24.
by using soft bandages and to avoid contact with contam- 10 Harari A, Chen Z, Burk RD. Human papillomavirus genomics: past,
inated surfaces. A noninvasive, repeatable and painless present and future. Curr Probl Dermatol 2014;45:1–18.
treatment modality, when possible, is optimal for ther- 11 Frazer I. Correlating immunity with protection for HPV infection. Int
J Infect Dis 2007;11(suppl 2):S10–16.
apy‐resistant warts of paediatric patients. 12 Rintala MAM, Grénman SE, Järvenkylä ME et al. High‐risk types of
human papillomavirus (HPV) DNA in oral and genital mucosa of
Plan of management for a child with warts infants during their first 3 years of life: experience from the Finnish
Management of common warts. Although a therapeutic HPV Family Study. Clin Infect Dis 2005;41:1728–33.
13 Harwood CA, Surentheran T, Sasieni P et  al. Increased risk of skin
algorithm can help, the treatment of warts is likely to cancer associated with the presence of epidermodysplasia verruci-
require an individualized approach because there is great formis human papillomavirus types in normal skin. Br J Dermatol
variability in the response to treatment. For children, it is 2004;150:949–57.
14 Benton E. Human papillomavirus infection. In: Textbook of

ideal to have an effective and painless treatment. Until Paediatric Dermatology, 2nd edn. Oxford: Blackwell, 2005:369–84.
such a wart treatment is discovered, patients and their 15 Gulino AV. WHIM syndrome: a genetic disorder of leukocyte traffick-
parents should be properly educated about HPV viral ing. Curr Opin Allergy Clin Immunol 2003;3:443–50.
aetiology and specific treatment expectations to avoid 16 Marcoux D, Nadeau K, McCuaig C et al. Pediatric anogenital warts:
a 7‐year review of children referred to a tertiary‐care hospital in
frustration for the patient and healthcare provider. Some Montreal, Canada. Pediatr Dermatol 2006;23:199–207.
trial and error is inevitable until an effective treatment is 17 Carlson JA, Cribier B, Nuovo G, Rohwedder A. Epidermodysplasia
identified for the individual patient. verruciformis‐associated and genital‐mucosal high‐risk human
Chapter 49  Human Papillomavirus Infection 597

­ apillomavirus DNA are prevalent in nevus sebaceus of Jadassohn. J


p 39 Kollipara R, Ekhlassi E, Downing C et al. Advancements in pharma-
Am Acad Dermatol 2008;59:279–94. cotherapy for noncancerous manifestations of HPV. J Clin Med
18 Favre M, Majewski S, Noszczyk B et al. Antibodies to human papil- 2015;4:832–46.
lomavirus type 5 are generated in epidermal repair processes. J Invest 40 Choi MH, Seo SH, Kim IH, Son SW. Comparative study on the sus-
Dermatol 2000;114:403–7. tained efficacy of diphencyprone immunotherapy versus cryotherapy
19 Jayasinghe Y, Garland SM. Genital warts in children: what do they in viral warts. Pediatr Dermatol. 2008;25:398–9.
mean? Arch Dis Child 2006;91:696–700. 41 Lacey CJN. Therapy for genital human papillomavirus‐related dis-
20 Irwin CJ, Shafer M, Moscicki A. Human papillomavirus. In: Rudolph’s ease. J Clin Virol 2005;32(suppl 1):S82–90.
Pediatrics, 21st edn. New York: McGraw‐Hill, 2003:267–8. 42 Padilla España L, Del Boz J, Fernández Morano T et al. Topical cidofo-
21 Ryndock EJ, Meyers C. A risk for non‐sexual transmission of human vir for plantar warts. Dermatol Ther 2014;27:89–93.
papillomavirus? Expert Rev Anti Infect Ther. 2014 Oct;12(10):1165–70. 43 Schopf RE. Ingenol mebutate gel is effective against anogenital

22 Gallay C, Miranda E, Schaefer S et al. Human papillomavirus (HPV) warts  –  a case series in 17 patients. J Eur Acad Dermatol Venereol
contamination of gynaecological equipment. Sex Transm Infect 2016;30:1041–3
2016;92:19–23. 44 Majewski S, Pniewski T, Malejczyk M, Jablonska S. Imiquimod is
23 Aguilera‐Barrantes I, Magro C, Nuovo GJ. Verruca vulgaris of the highly effective for extensive, hyperproliferative condyloma in chil-
vulva in children and adults: a nonvenereal type of vulvar wart. Am J dren. Pediatr Dermatol 2003;20:440–2.
Surg Pathol 2007;31:529–35. 45 Miller RL, Meng T‐C, Tomai MA. The antiviral activity of Toll‐like
24 Lei Y‐J, Gao C, An R et al. Development of a multiplex PCR method receptor 7 and 7/8 agonists. Drug News Perspect 2008;21:69–87.
for detecting and typing human papillomaviruses in verrucae vul- 46 Sinha S, Relhan V, Garg VK. Immunomodulators in warts: Unexplored
garis. J Virol Methods 2008;147:72–7. or ineffective? Indian J Dermatol 2015;60:118–29.
25 Hutchinson DJ, Klein KC. Human papillomavirus disease and vac- 47 Yang J, Pu Y‐G, Zeng Z‐M et al. Interferon for the treatment of genital
cines. Am J Health‐Syst Pharm 2008;65:2105–12. warts: a systematic review. BMC Infect Dis 2009;9:156.
26 Syrjänen S. HPV infections in children. Papillomavirus Rep
48 Choi Y, Kim DH, Jin SY et al. Topical immunotherapy with diphenyl-
2003;14:93–110. cyclopropenone is effective and preferred in the treatment of periun-
27 Cason J, Mant CA. High‐risk mucosal human papillomavirus infec- gual warts. Ann Dermatol 2013;25:434–9.
tions during infancy & childhood. J Clin Virol 2005;32(suppl 1): 49 Lee AN, Mallory SB. Contact immunotherapy with squaric acid dibu-
S52–8. tylester for the treatment of recalcitrant warts. J Am Acad Dermatol
28 Powell J, Strauss S, Gray J, Wojnarowska F. Genital carriage of 1999;41:595–9.
human papilloma virus (HPV) DNA in prepubertal girls with and 50 Torrelo A. What’s new in the treatment of viral warts in children.
without vulval disease. Pediatr Dermatol 2003;20:191–4. Pediatr Dermatol 2002;19:191–9.
29 Sinclair KA, Woods CR, Kirse DJ, Sinal SH. Anogenital and respira- 51 Markowitz LE, Liu G, Hariri S et al. Prevalence of HPV after intro-
tory tract human papillomavirus infections among children: age, gen- duction of the vaccination program in the United States. Pediatrics
der, and potential transmission through sexual abuse. Pediatrics 2016;137:1–9.
2005;116:815–25. 52 Saraiya M, Unger ER, Thompson TD et  al. US assessment of HPV
30 Watts DH, Koutsky LA, Holmes KK et al. Low risk of perinatal trans- types in cancers: implications for current and 9‐valent HPV vaccines.
mission of human papillomavirus: results from a prospective cohort J Natl Cancer Inst 2015;107:djv086.
study. Am J Obstet Gynecol 1998;178:365–73. 53 Dobson SRM, McNeil S, Dionne M et al. Immunogenicity of 2 doses of
31 Mammas IN, Sourvinos G, Spandidos DA. Human papilloma
HPV vaccine in younger adolescents vs 3 doses in young women: a
virus  (HPV) infection in children and adolescents. Eur J Pediatr randomized clinical trial. JAMA 2013;309:1793–802.
2009;168:267–73. 54 Toh ZQ, Licciardi PV, Fong J et al. Reduced dose human papillomavi-
32 Lee S, Lee W, Chung S et al. Detection of human papillomavirus 60 in rus vaccination: an update of the current state‐of‐the‐art. Vaccine
epidermal cysts of nonpalmoplantar location. Am J Dermatopathol 2015;33:5042–50.
2003;25:243–7. 55 Zimmerman RK. HPV vaccine and its recommendations, 2007. J Fam
33 Rice PS, Mant C, Cason J et al. High prevalence of human papilloma- Pract 2007;56(2 suppl vaccines):S1–5, C1.
virus type 16 infection among children. J Med Virol 2000;61:70–5. 56 Kulczycki A, Qu H, Shewchuk R. Primary care physicians’ adherence
34 Lipke MM. An armamentarium of wart treatments. Clin Med Res to guidelines and their likelihood to prescribe the human papilloma-
2006;4:273–93. virus vaccine for 11‐ and 12‐year‐old girls. Womens Health Issues
35 Boull C, Groth D. Update: treatment of cutaneous viral warts in 2016;26:34–9.
children. Pediatr Dermatol 2011;28:217–29. 57 Cyrus N, Blechman AB, Leboeuf M et al. Effect of quadrivalent human
36 Lio P. Warts, molluscum and things that go bump on the skin: a practi- papillomavirus vaccination on oral squamous cell papillomas. JAMA

SECTION 9: VIRAL SKIN


cal guide. Arch Dis Child Educ Pract Ed 2007;92:ep119–24. Dermatol 2015;151:1359–63.
37 Parsad D, Pandhi R, Juneja A, Negi KS. Cimetidine and levamisole 58 Spitzer M. Screening and management of women and girls with
versus cimetidine alone for recalcitrant warts in children. Pediatr human papillomavirus infection. Gynecol Oncol 2007;107(2 suppl

INFECTIONS
Dermatol 2001;18:349–52. 1):S14–18.
38 Gibbs S, Harvey I. Topical treatments for cutaneous warts. Cochrane 59 Wetzel C, Tissot A, Kollar LM et al. Development of an HPV educational
Database Syst Rev 2006;(3):CD001781. protocol for adolescents. J Pediatr Adolesc Gynecol 2007;20:281–7.
598 

CHA PTER 5 0

Herpes Simplex Virus Infections


Manuraj Singh1, Helen M. Goodyear2 & Judith Breuer3
1
 St George’s University Hospitals, London, UK
2
 Health Education England (West Midlands), Heart of England NHS Foundation Trust, Birmingham, UK
3
 Great Ormond Street Hospital, UCL Division of Infection and Immunity, London, UK

Abstract of HSV infections and latency has improved with in vitro models
and studies in rodent models. There is increasing understanding
Herpes simplex virus (HSV) belongs to the class of α‐herpesviruses about the various viral and host factors that determine latency.
which includes varicella zoster virus (VZV). HSV has a tropism for Therapy for mild HSV infections, such as aciclovir, is not generally
epithelial tissue and like VZV demonstrates latency within sensory indicated in uncomplicated infections in childhood. However, such
ganglia after primary infection. Two distinct antigenic types exist, therapy is clearly indicated and can be lifesaving in neonatal her-
known as HSV‐1 and HSV‐2. Herpes labialis and herpes genitalis pes ­simplex and disseminated infections such as those occurring
are typically due to HSV‐1 and HSV‐2 respectively but an increas- in eczema herpeticum and in immunocompromised patients. At
ing number of genital cases are also caused by HSV‐1. Both HSV‐1 present there is no therapeutic or prophylactic vaccine available
and HSV‐2 cause primary infections followed by the potential for for either HSV‐1 or HSV‐2, although studies to develop effective
episodes of recurrent disease. Knowledge of the pathogenesis vaccines are still ongoing.

Key points • The histological features of HSV infections are very similar to
VZV. They are characterized by varying degrees of epidermal
blistering and necrosis. The typical cytopathic features include
• Herpes simplex virus (HSV) is an alpha‐herpesvirus like varicella
nuclear enlargement, peripheral margination of chromatin,
zoster virus (VZV). There are two distinct antigenic types known
eosinophilic inclusions and multinucleation.
as HSV‐1 and HSV‐2. HSV, like VZV, undergoes latency within
• Definitive diagnosis of HSV infections is usually achieved by
sensory ganglia after primary infection.
polymerase chain reaction (PCR) or immunohistochemistry using
• HSV‐1 infection typically causes ‘herpes labialis’ whereas
specific HSV‐1 or HSV‐2 antibodies.
HSV‐2 has traditionally been responsible for ‘herpes genitalis’.
• Treatment of uncomplicated HSV in healthy children is generally
Primary oral infection is commonly asymptomatic in children.
not required. However, patients with eczema herpeticum or
Occasionally, healthy children may present with a moderate‐
severe infections such as disseminated disease occurring in the
to‐severe episode of primary gingivostomatitis. Recurrent oral
context of immunosuppression should be treated with systemic
herpes simplex is not uncommon and may be associated with
antiviral therapy such as aciclovir.
erythema multiforme. Genital herpes is much less common in
children and although it may be due to innocent inoculation,
sexual abuse must be considered.

HSV infection of humans has been documented since


SECTION 9: VIRAL SKIN

Brief introduction/history. Herpes simplex virus (HSV)


ancient Greek times [5] and the name ‘herpes’ is derived
belongs to the α‐subfamily of herpesviruses, which share
from the Greek word meaning ‘to creep’ [6]. In the mid‐
the characteristic of being neurotropic and include vari-
INFECTIONS

nineteenth century, the term ‘herpes’ was restricted to


cella zoster virus (VZV) [1]. There are two antigenic types
skin disorders with a vesicular lesion, and in the early
of HSV, types 1 and 2. The virus particles consist of a
twentieth century the term ‘herpes simplex’ was intro-
­central core of double‐stranded DNA, a 100 nm diameter
duced to include ‘herpes facialis’ and ‘herpes genitalis’.
icosahedral capsid (containing 162 capsomeres) sur-
HSV‐1 and ‐2 may affect any cutaneous site [3] and lesions
rounding the core, an envelope on the outside consisting
are characterized by grouped vesicles on an erythema-
of a lipid bilayer with glycoproteins embedded in it and a
tous background.
tegument (amorphous material filling the space between
the capsid and the envelope) [1–3]. The glycoproteins
have an important role in infectivity and also allow anti- Epidemiology and  pathogenesis. Humans are the only
genic recognition by the host. The DNA molecule of natural host of HSV and the viruses do not survive for
HSV‐1 and ‐2 has a molecular weight of approximately long periods in the external environment [7]. The lipid
100 × 106 Da and the complete virus measures about glycoprotein envelope is needed for HSV to be infectious,
150–1200 nm in diameter [4]. Over 50% of the DNA and this rapidly becomes desiccated in the low ambient
sequences in HSV‐1 and HSV‐2 are homologous [3]. humidity of the atmosphere [8]. Primary infection is

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 50  Herpes Simplex Virus Infections 599

acquired when a nonimmune child is exposed to contam- f­unctions appear to be important. Natural killer cells,
inated saliva or secretions from the pharynx, ­genitalia or macrophages, CD4 and CD8 T lymphocytes and various
eyes, usually exchanged in close personal contact. The cytokines (interferon‐α, ‐β and ‐γ, interleukins IL‐2 and
virus must come into contact with mucosal surfaces or IL‐18 and leucocyte migration inhibition factor) are
abraded skin. Sources of infection are individuals with thought to be important in protection against HSV
an obvious clinical lesion and those with asymptomatic ­infection. This explains the more severe and extensive
viral shedding, which may occur whether or not there presentation of mucosal or cutaneous HSV infections in
is a history of clinical herpes. The amount of virus shed patients with suppressed cell‐mediated immunity such as
is thought to be 100–1000 times less if an individual is transplant recipients and human immunodeficiency virus
asymptomatic [9]. Viral shedding has been found in (HIV)‐infected individuals. HSV can have a markedly
18–20% of young children [10,11]. Although most recur- suppressive effect on immune mechanisms during acute
rent infections are due to reactivation of virus, exogenous episodes of infection and may enhance suppressor T cell
re‐infection may also occur [12]. activity, decrease cytokine production, decrease expres-
The highest rate of HSV infections is in the first 5 years sion of major and minor histocompatibility antigens and
of life and then after the onset of sexual maturity. Most inhibit cytotoxic effector cell function. The cell‐mediated
children will be asymptomatic or have a trivial illness immune response to HSV antigen has been noted to
associated with HSV exposure. HSV infections are uncom- be  both low [24–26] and high [26,27] at the time of
mon under 6 months of age due to passive transfer of HSV recurrence.
maternal antibodies [13,14]. Acquisition of HSV antibod- Humoral immunity is important in limiting the extent
ies by children depends on socioeconomic conditions. of recurrences, but the role of antibodies in primary
Between 20% and 50% of children have HSV antibodies ­infections is less well defined. Antibodies assist in the
by 5 years of age, and up to 90% of children in crowded destruction of cells infected with virus by complement
underdeveloped areas will have antibodies by 10 years of and natural killer cells and by the process of antibody‐
age [10,14]. The majority of cases of herpes labialis are dependent cell‐mediated cytotoxicity. Antibodies have a
caused by HSV‐1 and the majority of herpes genitalis cases protective effect when an individual is exposed to HSV of
were due to HSV‐2. However more recent epidemiological either type, and patients with previous exposure to HSV‐1,
studies have shown that a higher number of cases (up to usually oral infection, have a less severe genital infection
50%) of genital herpes are attributable to HSV‐1 [15,16]. with HSV‐2. A stronger and broader spectrum of antibody
reactivity to viral polypeptides is present in recurrent HSV
Pathogenesis. In primary infections, HSV‐1 and ‐2 both infection compared with primary HSV infection [28].
replicate at the site of viral inoculation. The virus binds to However, the importance of these HSV antibodies is illus-
heparan sulphate proteoglycan (HSP) molecules which trated by the fact that neonatal HSV infections are reduced
act as receptors for the virus [17]. This binding is depend- and less severe in mothers who are seropositive. Therefore
ent on the viral glycoproteins C and D interacting with a primary genital HSV infection, near the time of labour in
HSP moieties for HSV‐1; for HSV‐2 the main interacting a seronegative mother, is a high risk for neonatal herpes
protein is glycoprotein B [18]. In addition to HSP, two simplex [29,30]. Recurrent HSV infection occurs, however,
other cell surface receptors, herpesvirus entry mediator despite high antibody titres [31–33]. Aciclovir has been
(HVEM) and nectin‐1, have been shown to interact with found to delay antibody development to some of
glycoprotein D and facilitate entry of HSV‐1 [19]. Viral the  HSV glycoproteins when used to treat genital infec-

SECTION 9: VIRAL SKIN


replication occurs within the nucleus and is divided into tions, although a full antibody complement is present
three main stages, known as immediate early (IE), early 6–12 months after initial infection [34,35]. It is important to
and late viral gene synthesis. The tegument part of the realize that a state of adequate immunity does not prevent

INFECTIONS
HSV virus particle contains several proteins including the development of recurrent lesions. However, a lack of
VP16/UL48 [20], ICP0 [21] and ICP4 [22] that are key both humoral and cell‐mediated immunity is associated
facilitators of viral gene transcription. The tegument with more severe and atypical presentations of recurrent
proteins are delivered into the cytoplasm after the virus disease. In particular, a lack of humoral immunity is
­
envelope fuses with the host cell membrane. After the associated with an increased risk of dissemination and
tegument proteins enter the nucleus, the first round of encephalitis, and such patients should be considered for
HSV transcription occurs. After the primary infection, prophylactic aciclovir therapy.
HSV, similar to VZV, is transported from cutaneous sites In recurrent infections [9,14], an inflammatory infiltrate
in a retrograde fashion by neurons to dorsal root ganglia consisting of macrophages and T lymphocytes appears
of sensory nerves. Latency is then established within mainly in the dermis, at least 2 days after virus first
these dorsal root ganglia. Subsequent reactivation is reaches the skin. CD4 lymphocytes predominate in the
associated with the virus travelling down the associated early stages of infection, followed by an increase in CD8
sensory nerve to re‐infect epithelial tissue including
­ lymphocytes 2 days later. Neutrophils and macrophages
adnexal structures at the end of the nerve endings. infiltrate advanced necrotic lesions. Expression of major
histocompatibility complex (MHC) class II DR antigens is
Immunology. Cell‐mediated immunity has a central role subsequently seen in basal cells, keratinocytes, Langerhans
in recovery from primary and recurrent HSV infections cells, endothelial cells and most infiltrating mononuclear
[23]. Both non‐ and HSV‐specific cellular immune cells. Some of the mononuclear cells produce the antiviral
600 Section 9  Viral Skin Infections and Opportunistic Infections

cytokine interferon‐γ. p63 Gene expression, which plays a The exact mechanisms underlying reactivation from
pivotal role in the development and maintenance of latency remain unresolved but the Wilcox laboratory has
epithelial stratification, is increased in HSV‐1 and HSV‐2 demonstrated that removal of nerve growth factor (NGF)
infections and induces apoptosis when overproduced [36]. results in HSV‐1 reactivation [61–63]. Further studies have
HSV has developed several mechanisms to evade the shown that the mechanism of NGF‐mediated latency
innate antiviral immune system. HSV recognition by involves the PI3K‐Akt‐mTORC‐1 (phosphatidylinositol
toll‐like receptor 2 (TLR2) on cell membranes is inhibited 3‐kinase protein kinase B‐mammalian target of rapamy-
by the HSV viral protein ICP0. This leads to a decrease in cin [mTOR] complex 1) pathway. NGF removal inhibits
NF‐κB and interferon‐regulatory factor translocation into mTORC‐1 and induces HSV‐1 reactivation [64].
the nucleus and reduced secretion of antiviral molecules, Immune mechanisms also play a pivotal role in main-
such as interferons and cytokines. HSV also interferes taining latency; they involve CD8+ T cells, NK cells
with cell viability. It has different effects in different cell and interferon (IFN)‐γ produced by these cells [65].
types. For example, in epithelial cells apoptosis is blocked There is now also evidence for viewing HSV‐1 reactiva-
due to several viral proteins such as ICP0, UL41 (virion tion as a three‐step process involving epigenetic regula-
host shutoff protein) and gD. In contrast, HSV induces tion and divided into animation, exit from latency and
apoptosis of dendritic cells and natural killer (NK) cells. finally release of infective virions [66]. The animation
The HSV viral proteins, ICP0 and ICP27, have also been phase describes uncoordinated generalized transcription
shown to decrease induction and secretion of antiviral resulting in the production of viral genes that map
type‐1 interferons. Another mechanism of immune eva- throughout the entire genome. This phase is independ-
sion involves blocking the function of the C5 component ent of the HSV viral protein, VP16/UL48, a key activator
of complement mediated by glycoprotein C. HSV has also of lytic infection. Further DNA replication, during the
been shown to interfere with innate immune cells by exit from latency phase, and progression through reacti-
reducing the activation of NK and natural killer T (iNKT) vation with production of infectious virus is dependent
cells [37,38]. on VP16/UL48 [67,68].
Whereas HSV‐1 and ‐2 reactivation is often recurrent
and occurs in a younger population, VZV reactivation is Clinical features. HSV infections may be subclinical or
seldom recurrent and more prevalent in the elderly [39]. asymptomatic, in particular orolabial infections.
HSV‐1 DNA exists in a non‐integrated form within Approximately 90% of HSV‐1 infections and 75% of HSV‐2
human trigeminal ganglia [40]. The DNA is for the most infections are initially asymptomatic [69]. Transmission of
part transcriptionally silent and exists as genome con- both HSV‐1 and ‐2 occurs through exposure to contami-
catemers or as circular episomes of genomic unit length. nated secretions such as saliva on mucosal surfaces or
In latently infected ganglia, the most abundantly detected traumatized skin. Primary infection with HSV‐1 usually
HSV‐1 gene transcripts are those that map antisense to occurs in children and is mild. After primary infection,
ICP0/RL2 [41]. The most abundant latency associated similar to the other α‐herpesvirus, VZV, HSV travels along
transcripts (LATs) are two stable introns (1.5 and 2.0 kb) sensory nerves in a retrograde fashion to remain latent
spliced from the unstable primary 8.3 kb LAT [42–46]. within sensory ganglia. This is primarily trigeminal
Although LATs are dispensable for establishment of ­ganglia for herpes labialis and sacral ganglia for herpes
latency they do have antiapoptotic properties and facili- genitalis. Reactivation of both viruses results in similar
tate latency by epigenetic modification of HSV‐1 lytic gene painful lesions at the site of initial infection or asympto-
SECTION 9: VIRAL SKIN

promoters. They also function as a primary micro‐RNA matic shedding of infectious viral particles. Asymptomatic
(miRNA) precursor that encodes at least four ­miRNAs in shedding is common for HSV‐2 infections [70].
HSV‐1‐infected cells [47–50]. miRNAs are small RNAs
INFECTIONS

that bind to complementary RNA sequences resulting in Primary herpes gingivostomatitis or herpes labialis
their destruction and ultimate inhibition of protein transla- This is the most common clinical manifestation of
tion. Seven of the 16 miRNAs identified in HSV‐1‐infected ­primary HSV infection in children and is usually due to
cells are present in latently infected mouse or human gan- HSV‐1 [2,71–74] (Fig. 50.1). Primary infection refers to a
glia [51–53]. Three of these miRNAs reduce the expression first infection occurring in a seronegative nonimmune
of key virus proteins such as ICP4, ICP34.5 and ICP0/RL2 individual. The severity of symptoms in normal healthy
which are involved in gene transcription, n ­ eurovirulence children varies widely. Initially, children tend to have
and evasion of innate antiviral pathways respectively malaise and fever and subsequently develop small
[54,55]. Further support for the important role of LAT‐ grouped vesicles typically on an erythematous base.
encoded miRNAs in maintaining latency comes from These may be present on the hard and soft palate, gingiva,
studies showing a steady state decrease of HSV‐1 miRNA lips, buccal mucosa, tongue, floor of the mouth and phar-
levels as the virus reactivates [56–58]. ynx. Perioral lesions may occur and may extend some dis-
It is important to note that other miRNAs including viral tance away from the mouth. The vesicles form shallow
and host encoded transcripts are also involved in maintain- ulcers with a yellow exudative base and an erythematous
ing latency. Viral miRNAs may also interfere with host halo and lesions may coalesce. Numerous ulcers may be
­factors to facilitate latency and host miRNAs expressed in present. Associated symptoms include tender cervical
a neuronal specific manner have been shown to reduce lymphadenopathy and halitosis. Excessive dribbling may
expression of lytic virus genes such as ICP0 [59,60]. occur in the younger child. The lips and gingiva may be
Chapter 50  Herpes Simplex Virus Infections 601

Fig. 50.2  Herpetic whitlow.


Fig. 50.1  HSV primary gingivostomatitis with multiple lesions on the lips
and tongue. The gums are red and swollen.
recurrences [75,76]. In 80% of cases, a single area of
involvement is present and less than 5% of patients will
swollen and red and bleed easily when touched. Lesions have two or more areas involved.
tend to be painful and commonly sting or itch, causing One quarter of individuals experiencing recurrences
the child to be irritable and to refuse food or drink. will have two or more episodes per year. The most com-
Tonsillitis or posterior pharyngitis with exudative ulcera- mon factors that have been implicated in reactivation
tive lesions on the pharynx, tonsils and buccal mucosa include ultraviolet light, local trauma, fever, wind, con-
may be the presenting feature, especially in older chil- current infections – classically described with a pneumo-
dren. The fever lasts for 2–7 days and then subsides. Oral coccal chest infection  –  HIV infection, local dental or
lesions may persist for 2–3 weeks. surgical procedures, stress, pregnancy and menstruation.
Complications include herpetic whitlows, spread to
other areas of the face or eye and transfer of infection to Herpetic whitlow and hand lesions
other body sites by scratching, including the genital HSV infection of the digits is most commonly described
areas. When lesions are widespread, the clinical diagno- as herpetic whitlow, herpetic paronychia or felon [77–79]
sis is not usually in doubt. Differential diagnosis of less (Figs  50.2 and 50.3). Lesions may occur anywhere on
severe HSV oral infection and pharyngitis includes aph- the hand, and ‘herpes manus’ has been suggested as an
thous stomatitis, herpangina due to enteroviruses, infec- appropriate name.
tious mononucleosis, streptococcal infection, diphtheria, Herpetic whitlows tend to occur in young children
erythema multiforme and Stevens–Johnson syndrome. with primary HSV gingivostomatitis due to autoinocula-
tion from finger/thumb sucking, and multiple digits may
Recurrent herpes gingivostomatitis or herpes labialis be involved [80,81]. Autoinoculation may occur in older

SECTION 9: VIRAL SKIN


Between 20% and 40% of the population, at some stage, children due to nail biting [82], and in teenage children
will have recurrent orolabial HSV lesions [2,71]. Lesions whitlows may be present in association with primary

INFECTIONS
are usually on the lips rather than intraoral and are collo- genital infection [83,84]. HSV infection of the digits less
quially referred to as ‘cold sores’. The site most commonly commonly occurs as a result of a contact who has HSV
affected is the mucocutaneous junction of the lips. The (usually a close adult relative with recurrent HSV labialis),
lesions of HSV‐1 are more frequently recurrent than those kissing an affected digit [11] or toe [84] or after physical
of HSV‐2 [73]. In comparison with the initial infection, trauma [13]. Most lesions are due to HSV‐1.
recurrent HSV lesions are usually less severe, more local- The finger is the site most commonly affected, account-
ized and heal more rapidly. A prodrome including irrita- ing for 67% of lesions. HSV infection usually involves the
tion, tingling, pain, numbness and a burning sensation digital pulp space and lateral aspect of the fingers, with
occurs at the affected site, usually less than 6 hours before the lateral or proximal nailfold being affected only rarely.
a lesion appears. Some 60% of patients will experience a The thumb, palm and wrist are affected in order of
prodrome. Initially erythema is present, followed by decreasing frequency.
grouped papules and then vesicles. The vesicles rupture Children with a primary infection from exogenous
within 2–3 days, leaving an ulcer that becomes encrusted. inoculation have the most severe illness and are often
Healing is usually complete by 6–10 days. Constitutional febrile and have constitutional symptoms. Presentation is
symptoms including fever are uncommon but, with with sudden onset of pain, redness and swelling around
recurrent HSV lesions, mood changes in children are well the infected area. The infected site is initially erythema-
documented. Cranial nerve palsies, cluster headaches tous and swollen followed by vesiculation, development
and psychotic episodes have been associated with HSV of pustules and rupture of lesions. On heavily keratinized
602 Section 9  Viral Skin Infections and Opportunistic Infections

infections in the past have been due to HSV‐2 rather than


HSV‐1. Although there is geographical variation, there is
now epidemiological evidence of a significant increase in
HSV‐1 genital disease [89]. A study in Scotland found
that the number of genital herpes cases due to HSV‐1 rose
from 33% to 56% over a period of approximately 10 years
[92]. Younger children may acquire HSV genital infection
due to autoinoculation from herpetic digital disease or
gingivostomatitis [90], exogenous inoculation from a
carer, usually a parent with recurrent herpetic digital
disease [93], close nonsexual physical contact and child
sexual abuse [94–96]. In the young child, careful judge-
ment is often needed when considering the route by
which herpetic genital disease has been acquired, and the
possibility of child sexual abuse must be considered
[94,95]. Genital HSV infection in neonates usually reflects
transmission from the birth canal during labour [97].
Primary HSV genital infection may be asymptomatic
but can be severe if there has been no previous HSV
­exposure (HSV seronegative). A milder illness tends to
occur if antibodies to the heterologous HSV are present.
Symptoms tend to be more extensive in females than in
males because of a larger area of skin and greater mucous
membrane involvement. Multiple anatomical sites (labia
majora, labia minora, vagina and cervix) are usually
infected in women. Lesions in males tend to be located on
the penis, including the distal urethra and/or scrotum.
HSV infections are also being seen more frequently in
perianal, anorectal and extragenital sites such as the
upper thighs and suprapubic regions.
Fig. 50.3  HSV infection of the hand. Vesicles have coalesced to form
The incubation period is usually 2–7 days. Initially one
bullae.
or more small pruritic erythematous papules appear and
then progress rapidly to vesicles. After 3–5 days, the vesi-
areas such as the palm, vesicles may coalesce and, after cles develop into painful shallow ulcers that crust. New
pustulation, resolve slowly without breaking [9]. The crops can appear for up to 10 days. Associated symptoms
fluid from lesions is often clear, opalescent or serosan- include pain, itching, fever, dysuria, vaginal and/or ure-
guinous rather than pustular. The draining lymph nodes, thral discharge, malaise, headache, myalgia, paraesthesia,
including the epitrochlear and axillary, are often enlarged herpetic whitlow, an ulcerative/exudative pharyngitis
and tender. Lymphangitis may be prominent. Spontaneous (10–15% of cases) and, less commonly, self‐limited aseptic
resolution tends to occur in 18–20 days. Recurrences are meningitis. In males, urethritis and dysuria out of propor-
SECTION 9: VIRAL SKIN

usually less severe, occur in approximately 20% of cases tion to the urethral discharge is suggestive of HSV infec-
and are of shorter duration than the initial infection, last- tion. The illness tends to last 2–3 weeks. Complications
INFECTIONS

ing 7–10 days. A prodrome of pain, tenderness, pruritus, include secondary bacterial infection, urinary retention
aching or burning is usually experienced and lasts a vari- and fibrinous adhesions leading to phimosis in males and
able time interval of a few hours to 3 days. Triggering fac- labial adhesions in females. When there are ulcerative
tors are the same as for recurrent herpes labialis. It is lesions in adolescents, then syphilis coexists with HSV in
important to distinguish HSV infection from bacterial 3–10% of cases and the risk of HIV infection is increased
infection of the hand to avoid unnecessary surgery [10,85]. significantly [89]. The ulcers of HSV are often smaller
Complications of HSV infection of the hand are rare but than isolated syphilitic ulcers [89].
include local hypoaesthesia, secondary ocular involve- Approximately 50–65% of patients suffer from recurrent
ment and genital disease. The most common complication genital herpetic infection but with gradually decreasing
is probably secondary bacterial infection. Herpetic whit- frequency [98]. HSV‐2 genital infections are more fre-
low is also often seen in adults who work with children quently recurrent than those due to HSV‐1 [38,99].
such as nursery staff and healthcare workers, especially Recurrences are of shorter duration than the initial genital
paediatricians and dental practitioners [86]. infection and are less intense and usually without systemic
manifestations. Long‐term complications of recurrent
Genital herpes simplex virus infection HSV genital infection include physical discomfort,
(herpes genitalis) psychological disturbances [100], transmission of HSV to
Most children with HSV genital infection [2,87–91] are neonates and an increased risk of HIV infection, probably
adolescents, after the onset of sexual activity [11]. Most due to the entrance of HIV into the ulcers [11].
Chapter 50  Herpes Simplex Virus Infections 603

Other cutaneous herpes simplex virus infections of  an intense desire to compete, and exposed areas of
HSV infection may occur at any site on the body, particu- skin of fellow players are susceptible to HSV infection.
larly where the skin surface is abraded. Fig.  50.4 shows
HSV infection beneath the eye of a young child and Herpes simplex encephalitis
Fig. 50.5 shows primary HSV infection of the pinna of an HSV encephalitis [103–105] tends to present with non-
8‐year‐old boy. Epidemics may occur in sports involving specific symptoms and signs in children. It should be
close physical contact such as rugby (scrum pox) [101] ­suspected in a febrile child with focal seizures, focal
and wrestling (herpes gladiatorum) [102]. Young sport- ­neurological signs and progressive decrease in the level
speople often do not report their HSV infection because of consciousness. Skin lesions are usually not prominent.
Treatment with intravenous aciclovir, especially if started
early in the course of the illness, has significantly
decreased morbidity and mortality from HSV encephalitis.
There is increasing evidence that susceptibility to infec-
tions is genetically determined by abnormalities in various
pathways of immune defence. There is some evidence
that patients with genetic abnormalities in toll‐like
receptor 3 (TLR3) and production of interferon may be
predisposed to herpes simplex encephalitis [106,107].

Herpes keratitis
This is rare as a primary HSV infection in children or as an
extension of oral lesions [108,109]. It may occur due to
inoculation from HSV digital disease. Redness, watering,
discharge with gritty sensation in the eye, itching and
eyelid swelling may be present. Less common symptoms
include pain, photophobia, lid vesicles, ulcers and blurred
vision. Recurrences are not uncommon and iridocyclitis,
keratouveitis and corneal scarring are all recognized
complications without prompt treatment. Classically, in
acute infections, a dendritic corneal ulcer is seen with a
Fig. 50.4  HSV infection beneath the eye with erythema, vesicles and ­fluorescein stain viewed under ultraviolet light.
crusting.
Disseminated HSV infection
Disseminated HSV infection in otherwise healthy chil-
dren has been reported but is very uncommon [110].
Predisposing conditions include chronic protein malnu-
trition, concomitant infection such as measles, pertussis
or Haemophilus influenzae [111], immunocompromise in
children or neonates and dermatoses that predispose to
cutaneous HSV infection [112]. Predisposing dermatoses

SECTION 9: VIRAL SKIN


include atopic dermatitis (eczema herpeticum), Darier
disease [113], Hailey–Hailey disease, Grover disease,

INFECTIONS
ichthyosis vulgaris [114], burns [115–117], congenital ich-
thyosiform erythroderma [118], incontinentia pigmenti
[119], pemphigus vulgaris and foliaceus [120,121] and
other bullous dermatoses [122]. Disseminated infection
may involve the liver, lungs, adrenals, pancreas, small or
large bowel, kidneys, bone marrow and central nervous
system and is usually associated with widespread cuta-
neous lesions [103,123,124]. Disseminated disease is
associated with a significant mortality rate.

Infection in immunocompromised children


Children with a primary immune deficiency [2,125]
mainly affecting cell‐mediated immunity are particularly
susceptible to HSV infection, unlike those with impaired
humoral immunity [9]. Similarly, children with an under-
lying malignancy on cytotoxic chemotherapy and those
with leukaemia or acquired immune deficiency syndrome
Fig. 50.5  HSV infection of the pinna. (AIDS) have an increased susceptibility to HSV. HSV
604 Section 9  Viral Skin Infections and Opportunistic Infections

infection is usually due to reactivation of virus. There is a multiforme. The main differential diagnoses for herpetic
spectrum of disease ranging from lesions that are local- whitlow are bacterial infections of the hand such as bul-
ized, short‐lived and self‐limited with normal healing to lous impetigo and acute paronychia. When lesions occur
prolonged destructive lesions occurring with increased locally on other parts of the skin, herpes zoster should be
frequency. Lesions are usually in sites typically infected considered and the dermatomal distribution of the lesions
by HSV (oral mucous membranes, genital area, cutaneous tends to point towards a diagnosis of zoster. Disseminated
sites). Less commonly, HSV causing respiratory disease HSV infections in neonates and the immunocompromised
(tracheobronchitis, pneumonia) or oesophagitis, usually may mimic primary varicella or disseminated VZV infec-
due to an extension of oral lesions and HSV colitis, has tion and polymerase chain reaction (PCR) analysis of skin
been reported [126]. In addition, immunocompromised samples can be very useful in differentiating them [131].
patients suffer from increased severity, more frequent In addition, generalized immunobullous disorders such
episodes of reactivation, atypical presentations including as bullous pemphigoid and pemphigus vulgaris may
extensive ulceration, prolonged shedding, and a much occasionally fall into the differential diagnosis of dissemi-
greater risk for potential dissemination which may even nated disease.
prove fatal [127]. The differential diagnosis of genital ulceration induced
by HSV includes other sexually transmitted infections
Eczema herpeticum such as syphilis, chancroid, lymphogranuloma venereum
Eczema herpeticum, also known as Kaposi varicelliform and granuloma inguinale (donovanosis). Noninfectious
eruption, is a well‐recognized complication of atopic causes to consider include Behçet syndrome, trauma
­dermatitis (see also Chapter  15). It usually results from (including factitial) and fixed drug eruption. If the ulcer
a  primary HSV infection and results in extensive persists, especially despite antiviral treatment, malig-
facial,  chest and occasionally more widespread lesions. nancy should be excluded with an appropriate biopsy. In
Widespread disease should be regarded as a medical addition, Epstein–Barr virus (EBV) may occasionally
emergency as occasional fatal cases have been reported. induce acute painful genital ulceration typically in young
The condition is usually associated with systemic symp- females (Lipschutz ulcer) [132].
toms such as fever and dehydration. It is important to
recognize that the most common clinical presentation is Laboratory/histology findings. As discussed in the
one of monomorphic ‘punched‐out’ superficial ulcers/ c­ orresponding section on VZV infections of the skin, the
erosions on a background of atopic dermatitis. The histological features of HSV and VZV infections are very
absence of characteristic herpetic vesiculation in typical similar [133]. Distinction between HSV‐1 and ‐2 and VZV
cases of eczema herpeticum is a common reason for the will depend on the clinical features and ultimately PCR
diagnosis not being considered [128]. Recurrent attacks of studies. The earliest histological features, which may pre-
eczema herpeticum may occur and are an indication for cede clinical disease by 2–3 days, are cytopathic changes
prophylactic antiviral therapy, especially if patients are within the nuclei of epidermal cells. These include
on immunosuppressive drugs. Recurrent lesions tend to nuclear enlargement, peripheral condensation of chroma-
be less severe, less extensive and of shorter duration. tin, ground glass homogenization and nuclear moulding.
Later changes include vacuolization of the cytoplasm,
Bell’s palsy followed by characteristic multinucleation and focal
Bell’s palsy is defined as an idiopathic peripheral facial eosinophilic nuclear and/or cytoplasmic inclusions. The
nerve paralysis. However, although still relatively contro- intranuclear inclusions with a surrounding artefactual
SECTION 9: VIRAL SKIN

versial, there are some studies showing isolation of cleft are known as Cowdry type A inclusions. These
HSV‐1 or VZV from cerebrospinal fluid in some cases of changes start in the basal and suprabasal layers of the epi-
INFECTIONS

peripheral facial nerve paralysis [129,130]. Given the dermis and spread upwards. At the time of the biopsy
above association some physicians advocate antiviral there is usually an intraepidermal vesicle which results
therapy for Bell’s palsy. The Ramsay Hunt syndrome from ballooning and reticular degeneration. Ballooning
occurs when VZV reactivation affects the facial nerve. degeneration of the epidermis describes a phenomenon
particular to viral infections and results from increasing
Neonatal HSV infection oedema within keratinocytes. Reticular degeneration,
See Chapter 7. which is not specific to viral infections, results from
­rupture of keratinocytes due to extreme balloon degener-
Erythema multiforme ation. The intraepidermal vesicle usually contains fluid,
See Chapter 66. cell debris and virally infected acantholytic and multinu-
cleate giant cells. The vesicle may appear subepidermal
Differential diagnosis. See also VZV differential diagnosis at  a later stage when the vesicle expands and the full
(Chapter 51). thickness of the epidermis is affected
The differential diagnosis of HSV‐1 and ‐2 infections HSV infections, like VZV, commonly show cytopathic
will vary depending on the site and severity of clinical changes within adnexal structures. The hair follicle and
findings. For oral ulceration the differential diagnosis will sebaceous gland may show typical cytopathic changes
include aphthous ulcers, Behçet syndrome, herpangina associated with necrosis of the affected epithelium. Less
due to Coxsackie virus, pemphigus vulgaris and erythema commonly the eccrine apparatus may be affected. Indeed,
Chapter 50  Herpes Simplex Virus Infections 605

Nowadays, the diagnosis of HSV‐1 or ‐2 infection can


be confirmed by PCR or immunohistochemistry. The
current gold standard for diagnosis of HSV‐1 and ‐2 infec-
tions [2] is detection of viral DNA by PCR. Growth of the
virus by culture in a variety of cell lines is mainly restricted
to research purposes. PCR is able to distinguish quickly
and reliably between HSV‐1, HSV‐2 and VZV. There are
now specific commercial antibodies to HSV‐1 and ‐2;
although readily available and reliable, they generally
take longer to perform than PCR techniques. They are
most commonly performed on paraffin‐embedded sec-
tions using immunoperoxidase techniques. In addition,
commercially available antibodies to VZV are less readily
available in most cellular pathology laboratories, and as a
result, in routine practice, PCR techniques are usually
required to confirm a diagnosis of VZV infection. PCR is
Fig. 50.6  High‐power histological image demonstrating HSV infection also particularly useful in providing rapid diagnosis in
affecting a hair follicle. The infected hair follicle demonstrates disruption, life‐threatening situations such as from cerebrospinal
necrosis and characteristic cytopathic changes of herpes simplex infections. fluid in HSV encephalitis and skin lesions in disseminated
Note that identical histology is seen in varicella zoster virus infections. The
HSV infections in immunocompromised patients when
cytopathic changes of the infected nuclei include moulding, enlargement,
and peripheral condensation of nuclear chromatin giving rise to a ground
the clinical features may be atypical and/or resemble
glass appearance. Characteristic multinucleated giant cells are also seen. disseminated chickenpox or herpes zoster [138].
(haematoxylin–eosin stain; original magnification ×200).

herpes may occasionally present with follicular lesions Treatment and prevention


clinically mimicking a folliculitis [134]. It is important to Primary
note that if the overlying epidermis has very prominent Mild uncomplicated HSV infections require no treatment.
necrosis and secondary changes, typical cytopathic Supportive therapy.  Supportive therapy includes effec-
changes of HSV are commonly found in the infundibular tive analgesia, local hygiene and antibiotics for secondary
portion of underlying hair follicles (Fig. 50.6). The under- bacterial infection. Oral fluids need to be encouraged
lying dermis usually contains a variably dense mixed when multiple mouth lesions are present. Patients with
inflammatory infiltrate containing lymphocytes, neutro- severe HSV gingivostomatitis should be monitored for
phils and eosinophils in varying percentages. As expected, dehydration and treated with intravenous fluids if this
the infiltrate commonly also shows a perineural distribu- becomes a problem.
tion which on rare occasions may show frank neuronal
necrosis. As with VZV it is important to be aware of Specific antiviral chemotherapy. Treatment with specific
‘herpes incognito’ from a histological point of view [135]. antiviral therapy depends on severity and site of infection
This describes the situation when a pathologist sees a [139–143]. In primary HSV, gingivostomatitis treatment is
lichenoid and/or vasculopathic tissue reaction in the usually needed only if infection is severe or if the patient
absence of the typical cytopathic changes of a herpes presents early in the course of disease. Patients late in

SECTION 9: VIRAL SKIN


infection. Commonly, further serial sections on the same acute disease are unlikely to benefit from treatment.
specimen will reveal the diagnostic changes that allow a Treatment is required in disseminated infection including

INFECTIONS
confident diagnosis to be made. neonatal herpes simplex, an immunocompromised child,
Herpes infections are sometimes associated with a painful herpetic whitlows and most genital infections.
dense dermal lymphoid infiltrate which may raise the High‐dose intravenous aciclovir therapy has been
question of a lymphoma or pseudolymphoma, especially shown  to reduce infant mortality from 85% to 29% in
if the typical cytopathic changes are not evident in the tis- ­disseminated neonatal HSV infections. In addition, the
sue sections examined. In addition, it is not uncommon for mortality associated with HSV encephalitis in neonatal
these dense lymphoid infiltrates to contain a significant infections was also reduced from 50% to 4% [144,145].
percentage of CD30+ cells, resulting in diagnostic confu- Topical antiviral therapy is used in the treatment of
sion with CD30+ lymphoproliferative disorders such as herpetic eye infections.
lymphomatoid papulosis [136,137]. Another important Nucleoside analogues were discovered in the 1970s and
point when assessing tissue sections histologically is that remain the mainstay of treatment for HSV‐1, HSV‐2 and
the features of late ulcerated lesions are not diagnostic VZV infections. Systemic aciclovir (acycloguanosine) is
unless the neighbouring epithelial tissue shows the typical the treatment of choice for primary HSV infections and
cytopathic changes. However, careful analysis of the ulcer is licensed for use in children. An oral syrup formulation
exudate may show necrotic and apoptotic epithelial cells is available. It exhibits both low toxicity and high selec-
containing ‘ghost outlines’ of viral inclusions. In such a tivity. Aciclovir is a synthetic purine (guanosine) nucleo-
situation, immunohistochemistry can be extremely useful side analogue that is converted by viral thymidine kinase
in helping to confirm the diagnosis. to aciclovir monophosphate and then by cellular enzymes
606 Section 9  Viral Skin Infections and Opportunistic Infections

to the diphosphate and triphosphate form. Aciclovir such oral treatment could be an effective alternative to
triphosphate is a selective inhibitor of HSV and VZV intravenous aciclovir in resource‐limited situations [155].
DNA polymerase and inhibits viral DNA replication, In addition, oral valaciclovir has been used to treat
with resultant chain termination following its incorpora- encephalitis in a child who developed a severe localized
tion into the viral DNA [146]. It acts only during active skin reaction to aciclovir [156].
HSV replication, and treatment should be started as Famciclovir is the prodrug of penciclovir (penciclovir is
soon as possible. Therapy with aciclovir does not affect only available in topical form) and has 100‐fold higher
healing, the establishment of latency during primary levels than aciclovir in HSV‐infected cells. There are
infections or the subsequent frequency of recurrences. reports of its beneficial use in patients with aciclovir‐
It  has a very low toxicity to uninfected host cells as the induced renal injury [157]. However, a relatively large
enzyme thymidine kinase of normal, uninfected cells retrospective community‐based study showed no signifi-
does not use aciclovir effectively as a substrate. Treatment cant difference in the incidence of acute kidney injury
with aciclovir is usually for 5 days but is adjusted depend- between aciclovir, valaciclovir or famciclovir [158]. Both
ing on patient response and immunocompetence. At least valaciclovir and famciclovir have the advantage of a
10 days of therapy is needed for HSV encephalitis, and twice‐daily dosage regimen and are recommended for the
longer courses of 2 weeks or more may prevent subse- treatment of adolescents with genital HSV infection [159].
quent relapse [147]. Weight‐adjusted dosing schedules are available for
Aciclovir is mainly excreted unchanged by the kidneys, younger ­children [160]. As the mode of action of these
with the metabolite 9‐carboxymethoxymethylguanine newer agents is similar to that of aciclovir, with phospho-
accounting for 10–15% of the dose excreted in the urine. rylation by HSV thymidine kinase in infected cells, pat-
It  is important to maintain adequate hydration, and in terns of HSV resistance seen are similar to those of
renal failure the dose should be modified according to aciclovir. Other nucleoside analogues include vidarabine,
the degree of impairment [148]. Aciclovir is only partially idoxuridine, trifluridine, brivudine and cidofovir [161].
absorbed from the gut. The usual oral dose is 100 mg Brief mention is made of brivudine which is a thymi-
five times daily for children under 2 years and 200  mg dine analogue and has a similar mechanism of action to
five times daily for children over 2 years. In severe aciclovir. It is available in oral and topical preparations.
­infections, those that are life‐threatening and neonatal Brivudine also inhibits viral DNA polymerase after phos-
infection, aciclovir should be given by intravenous phorylation by viral thymidine kinase. Interestingly, bri-
infusion over 1 hour, in an age‐appropriate dose, every vudine has selective inhibitory activity against HSV‐1 but
8  hours. Recommended doses are as follows: children not HSV‐2. It is also active against other herpesviruses
up to 3 months of age, 10 mg/kg 8 hourly; in those aged including EBV and VZV but not cytomegalovirus (CMV).
3 months to 12 years, 250 mg/m2 8 hourly; and 5 mg/kg Brivudine has potent activity against VZV in vitro com-
8 hourly in children over 12 years of age. The dose of aci- pared to aciclovir [162], and is licensed in Europe for the
clovir should be doubled when treating HSV encephalitis, treatment of herpes zoster and herpes simplex keratitis.
immunocompromised children and eczema herpeticum. Resistance to aciclovir is not a significant problem in
Prophylaxis is required in children with troublesome immunocompetent patients. The prevalence is low and
recurrent HSV infection, after neonatal HSV infection and has been stated at around 0.3–0.7%. The prevalence of
in the immunocompromised. resistance is higher in immunocompromised patients and
There have been few adverse side‐effects associated ranges between 2.5% and 25% depending on the underly-
with aciclovir [149,150]. Rapid intravenous injection
SECTION 9: VIRAL SKIN

ing cause and degree of the immunosuppression [163]. It


has  been associated with a transient rise in urea and has been described in immunocompromised children,
­creatinine. Inflammation and phlebitis may occur at the early in the course of neonatal herpes [164] and in those
INFECTIONS

site of injection. treated with multiple courses of aciclovir or long‐term


Poor oral absorption of aciclovir led to the develop- prophylaxis [165,166]. Most aciclovir‐ and other nucleo-
ment of newer acyclic guanosine analogues, valaciclovir side‐resistant HSV strains have mutations in viral thymi-
and famciclovir, with better oral bioavailability than dine kinase, encoded by the UL23 gene in HSV [167].
aciclovir [151]. Both famciclovir and valaciclovir are used More rarely, they may also have alterations of the sub-
widely in adults including as prophylaxis after bone strate specificity of viral thymidine kinase or DNA poly-
marrow transplantation and to decrease the risk of trans- merase [139,152]. The emergence of resistant strains has
mitting genital HSV to heterosexual partners [152,153]. lead to the generation of other antiviral therapies which
They are not currently licensed for use in children, but are focus on inhibiting the viral DNA polymerase by other
being used with increased frequency, particularly in means. Foscarnet and cidofovir are two such agents most
children >12 years old. Valaciclovir is the l‐valyl ester of commonly used in clinical practice for resistant strains.
aciclovir and is indicated for treatment of both herpes They both act by inhibiting the catalytic subunit of viral
labialis and genitalis, including suppressive therapy. DNA polymerase, without requiring activation by viral
A  paper in the New England Journal of Medicine showed thymidine kinase.
that it effectively reduces transmission of genital herpes Foscarnet, a phosphonic acid derivative, selectively
[154]. A small study in adults showed that oral valaciclovir inhibits HSV DNA polymerase by reversibly binding to
achieved acceptable levels of aciclovir within the cerebro- its pyrophosphate binding site, and is approved to treat
spinal fluid of patients with HSV encephalitis. Therefore, HSV strains that are resistant to aciclovir in the immuno-
Chapter 50  Herpes Simplex Virus Infections 607

compromised and neonates [140,168]. It has poor oral cathelicidins secreted by various epithelial tissues and
absorption and is given intravenously every 8–12 hours lactoferrin as well as plant abstracts [161].
[86–88,140–142]. Foscarnet has significant toxicity, with Other options that are occasionally used for orofacial
renal impairment being the most common serious toxic HSV infections include trifluridine and docosanol [177].
effect often necessitating discontinuation of therapy; Trifluridine is a nucleoside analogue and can also be used
resistance to foscarnet may also occur. Thymidine kinase topically to treat herpes keratitis. Docosanol is occasion-
mutants are thought to be less neurovirulent and less ally used topically to treat recurrent herpes labialis. Its
capable of establishing latency. After a herpetic episode is proposed mechanism of action is the prevention of viral
terminated, the next recurrence is often due to the original entry into the cell [178].
latent virus, which is sensitive to aciclovir [148]. Cross‐ Topical preparations are available for herpes simplex
resistance to foscarnet and aciclovir suggests either an infections but they are generally less effective than sys-
alteration in the DNA polymerase or a combination of temic medication. They may be useful for milder infec-
separate HSV drug‐resistant populations. tions or as an adjunct to systemic treatment. The one main
Cidofovir, a nucleoside analogue that targets viral DNA exception is in the treatment of eye disease. Topical prepa-
polymerase, is phosphorylated into an active form by cel- rations include aciclovir 5% ointment, docosanol 10%
lular kinases independently of viral participation and has cream, penciclovir 1% cream [179], idoxuridine 15% solu-
been used when both aciclovir and foscarnet resistance tion and cidofovir 1% gel. However, where possible, oral
have been present [143,169]. Cidofovir is active against a drug treatment should be generally advised, to prevent
wide range of double‐stranded DNA viruses including drug resistance emerging.
the human herpesviruses, polyomaviruses, poxviruses Artesunate is a semi‐synthetic derivative of artemisin,
and adenoviruses [170]. Cidofovir, like foscarnet, is also which is derived from the plant Artemisia annua. This
associated with significant renal toxicity, and as a result class of compounds has activity against falciparum
both treatments are only indicated for lesions with proven malaria, cancer cells and schistosomiasis. In addition,
resistant mutations or for patients who have failed first‐ they also have broader bioactivity and inhibit several
line treatment with aciclovir or one of its derivatives. viruses including members of the herpes family such as
Cidofovir is available in topical and intravenous prepara- CMV, EBV and HSV [180]. Artesunate at a dose of 100 mg
tions only. daily for 30 days has been used successfully to treat an
Brincidofovir (CMX001) is a potentially very useful oral immunosuppressed patient with severe multidrug‐resistant
drug which may gain more widespread usage against HSV‐2 infection [181]. The exact antiviral mechanisms
double‐stranded DNA viruses such as the herpesviruses. remain unclear but the targets may include host rather
It is a derivative of cidofovir, and is converted to cidofovir than viral targets. A study in mice also showed a synergistic
intracellularly by cellular phospholipases [171]. As a effect of adding artesunate to valaciclovir for the treat-
result of its predominantly intracellular activity, side‐ ment of herpes simplex encephalitis [182].
effects such as myelosuppression and nephrotoxicity HIV patients may develop genital herpetic lesions that
seen with cidofovir are significantly reduced [172]. become extensive and persist for long periods. These
Brincidofovir has the same broad‐spectrum activity lesions also frequently show resistance to aciclovir ther-
against double‐stranded DNA viruses as cidofovir. apy and may become very thickened resulting in a pseu-
Interestingly, brincidofovir has been shown to have at dotumourous appearance. A study of six patients showed
least a 100‐fold increased activity against HSV‐1 and ‐2, that the immunomodulators imiquimod and thalidomide

SECTION 9: VIRAL SKIN


and at least a 10‐fold increased activity against VZV com- allowed five patients to achieve a complete response [183].
pared with cidofovir [173]. Like cidofovir and foscarnet, There have also been a few further case reports docu-
brincidofovir has been shown to be very useful for resist- menting the effectiveness of thalidomide for recalcitrant

INFECTIONS
ant CMV and HSV infections [174]. The most common anogenital HSV‐2 infections [184].
side‐effect of treatment with brincidofovir is diarrhoea.
It has also shown synergistic acitivity when added to aci- Surgery.  Although surgery is usually contraindicated,
clovir therapy for the treatment of HSV infections [175]. occasionally removal of part of a nail overlying a herpetic
Two relatively new drugs, amenamevir and pritelivir, whitlow or perforation of the nail by electrocautery may
belonging to the class of so‐called helicase–primase be needed for pain relief [85]. Aspiration of tense vesicles
inhibitors, have so far shown promising results in phase II may also decrease pain.
clinical trials [176]. The viral helicase–primase complex is
essential for synthesis of primers and unwinding DNA Recurrent attacks
during viral replication. This complex represents an Many recurrences are mild and require no treatment.
attractive target as there is no eukaryotic equivalent and Topical aciclovir cream, applied to affected areas five
it is essential for viral replication. In addition, the actions times daily, is useful for troublesome recurrences if
of helicase–primase inhibitors are independent of viral started  early in the prodromal phase. It is for external
thymidine kinase and can therefore protect infected and use only and not recommended for application to
uninfected cells from infection. Current antiviral research mucous membranes or the eye. Penciclovir cream is also
in HSV infections is focused on better oral availability of licensed for use in patients >12 years and in vitro pene-
synthetic drugs and natural compounds with different trates deeper into the epidermal cells than aciclovir, but
modes of action, e.g. antimicrobial peptides including needs to be applied every 2 hours for 4 days [185,186].
608 Section 9  Viral Skin Infections and Opportunistic Infections

Topical idoxuridine in adults has also been used to 5 Chang T‐W. Herpes simplex. Clin Dermatol 1984;2:5–7.
6 Snavely SR, Liu C. Clinical spectrum of herpes simplex virus infec-
decrease pain and duration of recurrences [187] and
tions. Clin Dermatol 1984;2:8–22.
more recently docosonal, which inhibits fusion of the 7 Rawls WE, Hammerberg O. Epidemiology of the herpes simplex
human host cell with the viral envelope of HSV, prevent- viruses. Clin Dermatol 1984;2:29–45.
ing viral replication [188]. More severe recurrences, par- 8 Jarrett M. Herpes simplex infection. Arch Dermatol 1983;119:99–103.
9 Vestey JP, Norval M. Mucocutaneous infections with herpes simplex
ticularly in immunocompromised children or those with virus and their management. Clin Exp Dermatol 1992;17:221–37.
an underlying skin disorder or genital HSV, may require 10 Buddingh GJ, Schrum DR, Lancier JC et  al. Studies of the natural
therapy with systemic aciclovir. history of herpes simplex infections. Pediatrics 1953;11:595–610.
Effective prophylaxis against recurrent HSV infections, 11 Kohl S. Herpes simplex virus infection – the neonate to the adolescent.
Isr J Med Sci 1994;30:392–8.
occurring at monthly intervals or more frequently, can be 12 Buchman TG, Roizman B, Nahmias AJ. Demonstration of exogenous
achieved with systemic aciclovir. The usual starting dose is genital reinfection with herpes simplex virus type 2 by restriction
800 mg daily (400 mg twice daily or 200 mg four times daily), endonuclease fingerprinting of viral DNA. J Infect Dis 1979;140:
295–304.
decreased to 200 mg 2–3 times daily, with one half of the 13 Scott TFM, Coriell L, Blank H et al. Some comments on herpetic
dose being given to children under 2 years. Therapy should infection in children with special emphasis on unusual clinical
be interrupted every 6–12 months to see if the natural fre- manifestations. J Pediatr 1952;41:835–43.
quency of recurrences has decreased. Prophylactic aciclovir 14 Nahmias AJ, Josey WE, Naib ZM et  al. Antibodies to herpesvirus
hominis types 1 and 2 in humans. Am J Epidemiol 1970;91:539–46.
and derivatives such as valaciclovir are recommended for 15 Looker KJ, Magaret AS, May MT et al. Global and regional estimates
recurrent episodes of eczema herpeticum and recurrent of prevalent and incident herpes simplex virus type 1 infections in
lesions, especially in immunocompromised patients and 2012. PLoS One 2015;10:e0140765.
16 Looker KJ, Magaret AS, Turner KM et al. Global estimates of prevalent
patients with facial burns [189]. Brief mention is made of
and incident herpes simplex virus type 2 infections in 2012. PLoS One
erythema multiforme which is commonly triggered by oro- 2015;10:e114989.
facial HSV‐1 infections. Recurrent attacks of erythema mul- 17 Choudhary S, Marquez M, Alencastro F et al. Herpes simplex virus
tiforme are an indication for continuous prophylactic type‐1 (HSV‐1) entry into human mesenchymal stem cells is heavily
dependent on heparan sulfate. J Biomed Biotechnol 2011;2011:
aciclovir therapy. A study showed that this treatment was 264350.
effective at completely suppressing attacks and in some 18 Luo S, Hu K, He S et al. Contribution of N‐linked glycans on HSV‐2
cases inducing remission. In addition, some cases deemed gB to cell‐cell fusion and viral entry. Virology 2015;483:72–82.
idiopathic erythema multiforme may respond to aciclovir 19 Knebel‐Mörsdorf D. Nectin‐1 and HVEM: cellular receptors for

HSV‐1 in skin. Oncotarget 2016;12:19087–8.
therapy, probably reflecting subclinical HSV infection [190]. 20 Zhu Q, Courtney RJ. Chemical cross‐linking of virion envelope and
tegument proteins of herpes simplex virus type 1. Virology
Prevention 1994;204:590–9.
In contrast to the situation for VZV, there is no vaccine 21 Yao F, Courtney RJ. Association of ICP0 but not ICP27 with purified
virions of herpes simplex virus type 1. J Virol 1992;66:2709–16.
against HSV‐1 or ‐2 in current routine clinical practice. 22 Bibor‐Hardy V, Sakr F. A 165 kd protein of the herpes simplex virion
An  ideal vaccine would prevent primary infection and shares a common epitope with the regulatory protein, ICP4. Biochem
colonization of the sensory ganglion [191,192]. Most vac- Biophys Res Commun 1989;163:124–30.
23 Rinaldo CR, Torpey DJ. Cell‐mediated immunity and immuno-
cines aim to decrease the frequency of recurrences [192].
suppression in herpes simplex virus infection. Immunodeficiency
Vaccination with inactivated HSV DNA has been 1993;5:33–90.
­protective to patients at risk of genital HSV infection [193] 24 Lopez C, O’Reilly RJ. Cell‐mediated immune responses in recurrent
and whole HSV vaccines have been found to decrease the herpesvirus infections. J Immunol 1977;118:895–902.
25 Vestey JP, Norval M, Howie S et al. Variation in lymphoproliferative
rate and duration of recurrences [194]. A strategy using a responses during recrudescent orofacial herpes simplex virus infec-
recombinant and defective HSV‐1 strain which expressed
SECTION 9: VIRAL SKIN

tions. Clin Exp Immunol 1989;77:384–90.


increased levels of gD showed efficacy at preventing ocu- 26 Tsutsumi H, Bernstein JM, Riepenhoff‐Talty M et  al. Immune

lar disease in mice [195]. A vaccine strategy incorporating responses to herpes simplex virus in patients with recurrent herpes
INFECTIONS

labialis. 1. Development of cell‐mediated cytotoxic responses. Clin


the viral antigens ICP4 and gD2 with an adjuvant reported Exp Immunol 1986;66:507–15.
an effective T‐cell response against viral shedding of 27 Rasmussen LE, Jordan GW, Stevens DA et al. Lymphocyte interferon
HSV‐2 in mice [196]. production and transformation after herpes simplex infection in
humans. J Immunol 1974;112:728–36.
28 Rabie‐Finger H, Valentine‐Thon E, Steinmann J et  al. Serological
Acknowledgement responses to herpes simplex virus type 1 (HSV‐1) analysed with
The original chapter was written in 2000 and revised by enzyme‐linked immunosorbent assay (ELISA) and western blot
Dr Helen Goodyear. Professor Breuer and Dr Singh (WB). Acta Virol 1991;35:113–26.
29 Brown ZA, Wald A, Morrow RA et al. Effect of serologic status and
contributed to this 2019 update. cesarean delivery on transmission rates of herpes simplex virus from
mother to infant. JAMA 2003;289:203–9.
References 30 Brown Z. Preventing herpes simplex virus transmission to the neonate.
1 Longson M. Herpes simplex. In: Zuckerman AJ, Banatvala JE, Pattison Herpes 2004;11(suppl 3):175A–86A.
JR (eds) Principles and Practice of Clinical Virology, 2nd edn. 31 Eberle R, Mou S‐W. Relative titers of antibodies to individual

Chichester: John Wiley, 1990:3–42. ­polypeptide antigens of herpes simplex type 1 in human sera. J Infect
2 Mindel A. Herpes Simplex Virus. The Bloomsbury Series in Clinical Dis 1983;148:436–44.
Science. London: Springer‐Verlag, 1990:1–164. 32 Kalimo KOK, Joronen IA, Havu VK. Cell‐mediated immunity against
3 Corey L, Spear PG. Infections with herpes simplex viruses (first of herpes simplex virus envelope, capsid, excreted, and crude antigens.
two parts). N Engl J Med 1986;314:686–91. Infect Immunol 1983;39:24–8.
4 Kieff ED, Bachenheimer SL, Roizman B. Size, composition and struc- 33 Douglas RG Jr, Couch RB. A prospective study of chronic herpes
ture of the deoxyribonucleic acid of herpes simplex subtypes 1 and 2. ­simplex virus infection and recurrent herpes labialis in humans.
J Virol 1971;8:125–32. J Immunol 1970;104:289–95.
Chapter 50  Herpes Simplex Virus Infections 609

34 Bernstein DI, Lovett MA, Bryson YJ. The effects of aciclovir on anti- 57 Du T, Zhou G, Roizman B. Induction of apoptosis accelerates reactiva-
body response to herpes simplex virus in primary genital herpetic tion of latent HSV‐1 in ganglionic organ cultures and replication in
infections. J Infect Dis 1984;150:7–13. cell cultures. Proc Natl Acad Sci U S A 2012;109:14616–21.
35 Ashley R, Mack K, Critchlow C et  al. Differential effect of systemic 58 Du T, Zhou G, Roizman B. Modulation of reactivation of latent herpes
aciclovir treatment of genital HSV‐2 infections on antibody responses simplex virus 1 in ganglionic organ cultures by p300/CBP and STAT3.
to individual HSV‐2 proteins. J Med Virol 1988;24:309–20. Proc Natl Acad Sci U S A 2013;110:E2621–8.
36 Megyeri K, Orosz L, Kormos B et al. The herpes simplex virus induced 59 Wu W, Guo Z, Zhang X et al. A microRNA encoded by HSV‐1 inhibits
demise of keratinocytes is associated with a dysregulated pattern of a cellular transcriptional repressor of viral immediate early and early
p63 expression. Microbes Infect 2009;11:784–94. genes. Sci China Life Sci 2013;56:373–83.
37 Suazo PA, Ibanez FJ, Retamal‐Diaz AR et al. Evasion of early antiviral 60 Pan D, Flores O, Umbach JL et al. A neuron‐specific host microRNA
responses by herpes simplex viruses. Mediators Inflamm 2015;2015: targets herpes simplex virus‐1 ICP0 expression and promotes latency.
593757. Cell Host Microbe 2014;15:446–56.
38 Su C, Zhan G, Zheng C. Evasion of host antiviral innate immunity 61 Wilcox CL, Johnson EM Jr. Nerve growth factor deprivation results
by HSV‐1, an update. Virol J 2016;13:38. in the reactivation of latent herpes simplex virus in vitro. J Virol
39 Kennedy PG, Steiner I. A molecular and cellular model to explain the 1987;61:2311–15.
differences in reactivation from latency by herpes simplex and vari- 62 Wilcox CL, Johnson EM Jr. Characterization of nerve growth

cella‐zoster viruses. Neuropathol Appl Neurobiol 1994;20:368–74. ­factor‐dependent herpes simplex virus latency in neurons in vitro.
40 Efstathiou S, Minson AC, Field HJ et al. Detection of herpes simplex J Virol 1988;62:393–9.
virus‐specific DNA sequences in latently infected mice and in 63 Wilcox CL, Smith RL, Freed CR, Johnson EM Jr. Nerve growth ­factor‐
humans. J Virol 1986;57:446–55. dependence of herpes simplex virus latency in peripheral sympathetic
41 Stevens JG, Haarr L, Porter DD et al. Prominence of the herpes sim- and sensory neurons in vitro. J Neurosci 1990;10:1268–75.
plex virus latency‐associated transcript in trigeminal ganglia from 64 Camarena V, Kobayashi M, Kim JY et  al. Nature and duration of
seropositive humans. J Infect Dis 1988;158:117–23. growth factor signaling through receptor tyrosine kinases regulates
42 Zwaagstra JC, Ghiasi H, Slanina SM et al. Activity of herpes simplex HSV‐1 latency in neurons. Cell Host Microbe 2010;8:320–30.
virus type 1 latency‐associated transcript (LAT) promoter in neuron‐ 65 Bigley NJ. Complexity of interferon‐gamma interactions with HSV‐1.
derived cells: evidence for neuron specificity and for a large LAT tran- Front Immunol 2014;5:15.
script. J Virol 1990;64:5019–28. 66 Kennedy PG, Rovnak J, Badani H, Cohrs RJ. A comparison of herpes
43 Spivack JG, Woods GM, Fraser NW. Identification of a novel latency‐ simplex virus type 1 and varicella‐zoster virus latency and reactivation.
specific splice donor signal within the herpes simplex virus type 1 J Gen Virol 2015;96:1581–602.
2.0‐kilobase latency‐associated transcript (LAT): translation inhibi- 67 Thompson RL, Preston CM, Sawtell NM. De novo synthesis of VP16
tion of LAT open reading frames by the intron within the 2.0‐kilobase coordinates the exit from HSV latency in vivo. PLoS Pathog 2009;
LAT. J Virol 1991;65:6800–10. 5:e1000352.
44 Wu TT, Su YH, Block TM, Taylor JM. Evidence that two latency‐asso- 68 Sawtell NM, Triezenberg SJ, Thompson RL. VP16 serine 375 is a critical
ciated transcripts of herpes simplex virus type 1 are nonlinear. J Virol determinant of herpes simplex virus exit from latency in vivo.
1996;70:5962–7. J Neurovirol 2011;17:546–51.
45 Thomas DL, Lock M, Zabolotny JM et al. The 2‐kilobase intron of the 69 Fiumara NJ. Herpes simplex. Clin Dermatol 1989;7:23–36.
herpes simplex virus type 1 latency‐associated transcript has a half‐ 70 Sacks SL, Griffiths PD, Corey L et  al. HSV shedding. Antiviral Res
life of approximately 24 hours in SY5Y and COS‐1 cells. J Virol 2004;63(suppl 1):S19–26.
2002;76:532–40. 71 Wheeler CE. The herpes simplex problem. J Am Acad Dermatol

46 Ng AK, Block TM, Aiamkitsumrit B et  al. Construction of a herpes 1988;18:163–8.
simplex virus type 1 mutant with only a three‐nucleotide change in 72 Higgins CR, Schofield JK, Tatnall FM et al. Natural history, manage-
the branchpoint region of the latency‐associated transcript (LAT) and ment and complications of herpes labialis. J Med Virol 1993;
the stability of its two‐kilobase LAT intron. J Virol 2004;78:12097–106. 1(suppl):22–6.
47 Allen SJ, Hamrah P, Gate D et al. The role of LAT in increased CD8+ T 73 Lafferty WE, Coombs RW, Benedetti J et al. Recurrences after oral and
cell exhaustion in trigeminal ganglia of mice latently infected with genital herpes simplex virus infection. Influence of site of infection
herpes simplex virus 1. J Virol 2011;85:4184–97. and viral type. N Engl J Med 1987;316:1444–9.
48 Jiang X, Chentoufi AA, Hsiang C et al. The herpes simplex virus type 74 Taieb A, Diris N, Boralevi F et al. Herpes simplex in children. Clinical
1 latency‐associated transcript can protect neuron‐derived C1300 and manifestations, diagnostic value of clinical signs, clinical course. Ann
Neuro2A cells from granzyme B‐induced apoptosis and CD8 T‐cell Dermatol Venereol 2002;129:603–8.
killing. J Virol 2011;85:2325–32. 75 Joseph R, Rose FC. Cluster headache and herpes simplex: association?

SECTION 9: VIRAL SKIN


49 Kramer MF, Jurak I, Pesola JM et al. Herpes simplex virus 1 microR- BMJ 1985;290:1625–6.
NAs expressed abundantly during latent infection are not essential 76 Shearer ML, Finch SM. Periodic organic psychosis associated with
for latency in mouse trigeminal ganglia. Virology 2011;417:239–47. recurrent herpes simplex. N Engl J Med 1964;271:494–7.

INFECTIONS
50 Wang K, Lau TY, Morales M et  al. Laser‐capture microdissection: 77 Gill MJ, Arlette J, Tyrell DL. Herpes simplex virus infection of the hand:
refining estimates of the quantity and distribution of latent herpes clinical features and management. Am J Med 1988;85(suppl 2A):53–6.
simplex virus 1 and varicella‐zoster virus DNA in human trigeminal 78 Gill MJ, Arlette J, Buchan KA. Herpes simplex virus infections of the
ganglia at the single‐cell level. J Virol 2005;79:14079–87. hand. J Am Acad Dermatol 1990;22:111–16.
51 Gartner JJ, Sethupathy P, Hatzigeorgiou AG, Fraser NW. Anti‐ 79 Feder HM, Long SS. Herpetic whitlow. Epidemiology, clinical charac-
apoptotic function of a microRNA encoded by the HSV‐1 latency‐ teristics, diagnosis and treatment. Am J Dis Child 1983;137:861–3.
associated transcript. Nature 2008;451:600. 80 Novick NL. Autoinoculation herpes of the hand in a child with recur-
52 Umbach JL, Nagel MA, Cohrs RJ et al. Analysis of human alphaher- rent herpes labialis. Am J Med 1985;79:139–42.
pesvirus microRNA expression in latently infected human trigeminal 81 Szinmai G, Schaad UB, Heininger U. Multiple herpetic whitlow

ganglia. J Virol 2009;83:10677–83. lesions in a 4‐year‐old girl: case report and review of the literature.
53 Held K, Junker A, Dornmair K et  al. Expression of herpes simplex Eur J Pediatr 2001;160:528–33.
virus 1‐encoded microRNAs in human trigeminal ganglia and their 82 Muller SA, Herrmann EC. Association of stomatitis and paronychias
relation to local T‐cell infiltrates. J Virol 2011;85:9680–5. due to herpes simplex. Arch Dermatol 1970;101:396–402.
54 Umbach JL, Kramer MF, Jurak I et al. MicroRNAs expressed by her- 83 Glogau R, Hanna L, Jawetz E. Herpetic whitlow as part of genital
pes simplex virus 1 during latent infection regulate viral mRNAs. virus infection. J Infect Dis 1977;136:689–92.
Nature 2008;454:780–3. 84 Crane LR, Lerner AM. Herpetic whitlow: a manifestation of primary
55 Flores O, Nakayama S, Whisnant AW et al. Mutational inactivation of infection with herpes simplex virus type 1 or type 2. J Infect Dis
herpes simplex virus 1 microRNAs identifies viral mRNA targets and 1978;137:855–6.
reveals phenotypic effects in culture. J Virol 2013;87:6589–603. 85 Polayes IM, Arons MS. The treatment of herpetic whitlow  –  a new
56 Du T, Zhou G, Roizman B. HSV‐1 gene expression from reactivated surgical concept. Plast Reconstr Surg 1980;65:811–17.
ganglia is disordered and concurrent with suppression of latency‐ 86 Wu IB, Schwartz RA. Herpetic whitlow. Cutis 2007;79:193–6.
associated transcript and miRNAs. Proc Natl Acad Sci U S A 2011;108: 87 Corey L. First episode, recurrent and asymptomatic herpes simplex
18820–4. infections. J Am Acad Dermatol 1988;18:167–72.
610 Section 9  Viral Skin Infections and Opportunistic Infections

88 Kinghorn GR. Genital herpes: natural history and treatment of acute 120 Silverstein EH, Burnett JW. Kaposi’s varicelliform eruption compli-
episodes. J Med Virol 1993;1(suppl):33–8. cating pemphigus foliaceus. Arch Dermatol 1968;95:214–16.
89 Vermillion ST, Holmes MM, Soper DE. Adolescent and sexually 121 Brown P, Taylor B. Herpes simplex infection associated with pemphi-
transmissible diseases. Obstet Gynecol Clin North Am 2000;27: gus vulgaris. Case report and literature review. J Am Acad Dermatol
163–79. 1989;21:1126–8.
90 Nahmias AJ, Dowdle WR, Naib ZM et  al. Genital infection with 122 Nikkels AF, Delvenne P, Herfs M, Pierard GE. Occult herpes simplex
­herpesvirus hominis types 1 and 2 in children. J Pediatr 1968;42: virus colonization of bullous dermatitides. Am J Clin Dermatol
659–66. 2008;9:163–8.
91 Stanberry LR, Rosenthal SL. Genital herpes simplex virus infection 123 Jordan SW, McLaren LC, Crosby JH. Herpetic tracheobronchitis.
in the adolescent: special considerations for management. Paediatr Cytologic and virologic detection. Arch Intern Med 1975;135:784–8.
Drugs 2002;4:291–7. 124 Nash G, Foley FD. Herpetic infection of the middle and lower
92 Scoular A, Norrie J, Gillespie G et al. Longitudinal study of genital respiratory tract. Am J Clin Pathol 1970;54:857–63.
infection by herpes simplex virus type 1 in Western Scotland over 125 Ramsey PG, Fife KH, Hackman RC et al. Herpes simplex virus pneumonia.
15 years. BMJ 2002;324:1366–7. Clinical, virologic and pathologic features in 20 patients. Ann Intern Med
93 Lazar MP. Primary herpetic vulvovaginitis. Arch Dermatol Syph 1982;97:813–20
1955;72:272–6. 126 Delis S, Kato T, Ruiz P et al. Herpes simplex colitis in a child with
94 Jaffe AC. Sexual abuse and herpetic genital infection. J Pediatr combined liver and small bowel transplant. Pediatr Transplant
1976;89:338. 2001;5:374–7.
95 Gardner M, Jones JG. Genital herpes acquired by sexual abuse of 127 Piret J, Boivin G. Resistance of herpes simplex viruses to nucleoside
children. Pediatrics 1984;104:243–4. analogues: mechanisms, prevalence, and management. Antimicrob
96 Kaplan KM, Fleischer GR, Paradise JE et al. Social relevance of genital Agents Chemother 2011;55:459–72.
herpes simplex in children. Am J Dis Child 1984;138:872–4. 128 Blanter M, Vickers J, Russo M, Safai B. Eczema herpeticum: would
97 Cherpes TL, Matthews DB, Maryak SA. Neonatal herpes simplex you know it if you saw it? Pediatr Emerg Care 2015;31:586–8.
virus infection. Clin Obstet Gynecol 2012;55:938–44. 129 Stjernquist‐Desatnik A, Skoog E, Aurelius E. Detection of herpes
98 Chang TW. Herpes simplex virus infection. Int J Dermatol 1983; simplex and varicella‐zoster viruses in patients with Bell’s palsy by
22:1–7. the polymerase chain reaction technique. Ann Otol Rhinol Laryngol
99 Reeves WC, Corey L, Adams HG et  al. Risk of recurrence after 2006;115:306–11.
first episodes of genital herpes. Relation to HSV type and antibody 130 Henkel K, Lange P, Eiffert H et al. Infections in the differential diag-
response. N Engl J Med 1981;305:315–19. nosis of Bell’s palsy: a plea for performing CSF analysis. Infection
100 Lynch PJ. Psychiatric, legal and moral issues of herpes simplex infec- 2017;45:147–55.
tions. J Am Acad Dermatol 1988;18:173–5. 131 Weidmann M, Meyer‐Konig U, Hufert FT. Rapid detection of herpes
101 White WB, Grant‐Kels JM. Transmission of herpes simplex virus simplex virus and varicella‐zoster virus infections by real‐time
type 1 infection in rugby players. JAMA 1984;252:533–5. PCR. J Clin Microbiol 2003;41:1565–8.
102 Belongia EA, Goodman JL, Holland EJ et al. An outbreak of herpes 132 Barrett MM, Sangueza M, Werner B et  al. Lymphocytic arteritis in
gladiatorum at a high school wrestling camp. N Engl J Med 1991; Epstein‐Barr virus vulvar ulceration (Lipschutz disease): a report of
325:906–10. 7 cases. Am J Dermatopathol 2015;37:691–8.
103 Brett EM. Herpes simplex virus encephalitis in children. BMJ 1986; 133 McSorley J, Shapiro L, Brownstein MH, Hsu KC. Herpes simplex
293:1388–9. and varicella‐zoster: comparative histopathology of 77 cases. Int J
104 Kohl S. Herpes simplex virus encephalitis in children. Pediatr Clin Dermatol 1974;13:69–75.
North Am 1988;35:465–83. 134 Weinberg JM, Mysliwiec A, Turiansky GW et  al. Viral folliculitis.
105 Editorial. Herpes simplex encephalitis. Lancet 1986;i:535–6. Atypical presentations of herpes simplex, herpes zoster, and
106 Abel L, Plancoulaine S, Jouanguy E et al. Age‐dependent Mendelian molluscum contagiosum. Arch Dermatol 1997;133:983–6.
predisposition to herpes simplex virus type 1 encephalitis in child- 135 Resnik KS, DiLeonardo M. Herpes incognito. Am J Dermatopathol
hood. J Pediatr 2010;157:623–9, 9 e1. 2000;22:144–50.
107 Casanova JL, Tardieu M, Abel L. [Genetic predisposition to
136 Leinweber B, Kerl H, Cerroni L. Histopathologic features of cutane-
­h erpetic meningo‐encephalitis in children]. Bull Acad Natl Med ous herpes virus infections (herpes simplex, herpes varicella/zoster):
2010;194:915–22. a broad spectrum of presentations with common pseudolympho-
108 Parier EH. Herpetic ocular infections of childhood. Arch Ophthalmol matous aspects. Am J Surg Pathol 2006;30:50–8.
1980;98:704–6. 137 Wain EM, Antony F, Appleton MA et al. Genital herpes masquer-
109 Inoda S, Wakakura M, Hirata J et al. Stromal keratitis and anterior ading as a cutaneous T‐cell lymphoma: a report of two cases.
SECTION 9: VIRAL SKIN

uveitis due to herpes simplex virus‐2 in a young child. Jap J Ophthalmol J Cutan Pathol 2008;35:770–3.
2001;45:618–21. 138 Sankuntaw N, Sukprasert S, Engchanil C et al. Single tube multiplex
110 Auch Moedy JL, Lerman SJ, White RJ. Fatal disseminated herpes real‐time PCR for the rapid detection of herpesvirus infections of the
INFECTIONS

simplex virus infection in a healthy child. Am J Dis Child central nervous system. Mol Cell Probes 2011;25:114–20.
1981;135:45–7. 139 Leigh IM. Management of non‐genital herpes simplex virus infec-
111 Jaworski MA, Moffatt MEK, Ahronheim GA. Generalized HSV 1 tions in immunocompetent patients. JAMA 1985;2A(suppl):34–8.
infection during H. influenzae epiglottitis. J Pediatr 1980;96:426–9. 140 Brown TJ, Straten MV, Tyring SK. Antiviral agents. Dermatol Clin
112 Becker WB, Kipps A, McKenzie D. Disseminated herpes simplex 2001;19:23–34.
virus infection. Am J Dis Child 1968;115:1–8. 141 Wood AJJ. Antiviral drugs. N Engl J Med 1999;340:1255–68.
113 Doeglas HMG, Moolhuysen TMGF. Kaposi’s varicelliform eruption. 142 Snoeck R. Antiviral therapy of herpes simplex. Antimicrob Agents
Two cases caused by herpes virus hominis infection complicating Chemother 2000;16:157–9.
Darier’s disease. Arch Dermatol 1969;100:592–5. 143 Whitley RJ. Herpes simplex virus in children. Curr Treat Options
114 Verbov J, Munro DD, Miller A. Recurrent eczema herpeticum associ- Neurol 2002;4:231–7.
ated with ichthyosis vulgaris. Br J Dermatol 1972;86:638–40. 144 Whitley RJ, Nahmias AJ, Visintine AM et al. The natural history of
115 Nishimura M, Maekawa M, Hino Y et  al. Kaposi’s varicelliform herpes simplex virus infection of mother and newborn. Pediatrics
eruption. Development in a patient with a healing second‐degree 1980;66:489–94.
burn. Arch Dermatol 1984;120:799–800. 145 Kimberlin DW, Lin CY, Jacobs RF et al. Safety and efficacy of high‐
116 McGill SN, Cartotto RC. Herpes simplex virus infection in a paediatric dose intravenous acyclovir in the management of neonatal herpes
burn patient: case report and review. Burns 2000;26:194–9. simplex virus infections. Pediatrics 2001;108:230–8.
117 Meuli M, Lips U, Nadal D. A toddler with burns, stomatitis and skin 146 De SK, Hart JC, Breuer J. Herpes simplex virus and varicella zoster
graft loss. Burns 2000;26:625–7. virus: recent advances in therapy. Curr Opin Infect Dis 2015;28:
118 Fitzgerald WC, Booker AP. Congenital ichthyosiform erythroderma: 589–95.
a report of two cases in siblings, one complicated by Kaposi’s varicel- 147 Kimura H, Aso K, Kuzushima K et  al. Relapse of herpes simplex
liform eruption. Arch Dermatol Syph 1951;64:611–19. encephalitis in children. Pediatrics 1992;89:891–4.
119 Fromer ES, Lynch PJ. Neonatal herpes simplex and incontinentia 148 Elion GB. Aciclovir: discovery, mechanism of action and selectivity.
pigmenti. Pediatr Dermatol 2001;18:86–7. J Med Virol 1993;1(suppl):2–6.
Chapter 50  Herpes Simplex Virus Infections 611

149 Arndt KA. Adverse reactions to aciclovir: topical, oral, and intrave- immunocompromised cancer patients: A single‐center experience.
nous. J Am Acad Dermatol 1988;18:188–90. Antiviral Res 2016;134:58–62.
150 Gould JM, Chessells JM, Marshall WC et al. Aciclovir in herpesvirus 175 Prichard MN, Kern ER, Hartline CB et  al. CMX001 potentiates the
infections in children: experience in an open study with particular efficacy of acyclovir in herpes simplex virus infections. Antimicrob
reference to safety. J Infect 1982;5:283–9. Agents Chemother 2011;55:4728–34.
151 Beutner KR. Valacyclovir: a review of its antiviral activity, pharmacoki- 176 Birkmann A, Zimmermann H. HSV antivirals – current and future
netic properties, and clinical efficacy. Antiviral Res 1995;28:281–90. treatment options. Curr Opin Virol 2016;18:9–13.
152 Laiskonis A, Thune T, Neldam S et al. Valaciclovir in the treatment of 177 De Clercq E. Antiviral drugs in current clinical use. J Clin Virol
facial herpes simplex virus infection. J Infect Dis 2002;186(suppl 1): 2004;30:115–33.
S66–70. 178 Katz DH, Marcelletti JF, Khalil MH et  al. Antiviral activity of

153 Dignani MC, Mykietiuk A, Michelet M et al. Valaciclovir prophylaxis 1‐­docosanol, an inhibitor of lipid‐enveloped viruses including her-
for the prevention of Herpes simplex virus reactivation in recipients pes simplex. Proc Natl Acad Sci U S A 1991;88:10825–9.
of progenitor cells transplantation. Bone Marrow Transplant 2002; 179 McKeough MB, Spruance SL. Comparison of new topical treatments
29:263–7. for herpes labialis: efficacy of penciclovir cream, acyclovir cream,
154 Corey L, Wald A, Patel R et al. Once‐daily valacyclovir to reduce the and n‐docosanol cream against experimental cutaneous herpes
risk of transmission of genital herpes. N Engl J Med 2004;350:11–20. simplex virus type 1 infection. Arch Dermatol 2001;137:1153–8.
155 Pouplin T, Pouplin JN, Van Toi P et al. Valacyclovir for herpes sim- 180 Efferth T, Romero MR, Wolf DG et  al. The antiviral activities of
plex encephalitis. Antimicrob Agents Chemother 2011;55:3624–6. artemisinin and artesunate. Clin Infect Dis 2008;47:804–11.
156 Chan PKS. Use of oral valaciclovir in a 12‐year‐old boy with herpes 181 Sellar RS, Ward KN, Thomson KJ, Peggs KS. Evidence for clinical
simplex encephalitis. Hong Kong Med J 2000;6:119–21. activity of artesunate in multidrug‐resistant herpes simplex infection
157 Htwe TH, Bergman S, Koirala J. Famciclovir substitution for patients following HSCT. Bone Marrow Transplant 2012;47:1482–3.
with acyclovir‐associated renal toxicity. J Infect 2008;57:266–8. 182 Canivet C, Menasria R, Rheaume C et  al. Valacyclovir combined
158 Lam NN, Weir MA, Yao Z et al. Risk of acute kidney injury from with artesunate or rapamycin improves the outcome of herpes
oral acyclovir: a population‐based study. Am J Kidney Dis 2013;61: simplex virus encephalitis in mice compared to antiviral therapy
723–9. alone. Antiviral Res 2015;123:105–13.
159 Saltzman R, Jurewicz R, Boon R. Safety of famciclovir in patients 183 Sbidian E, Battistella M, Legoff J et  al. Recalcitrant pseudotumoral
with herpes zoster and genital herpes. Antimicrob Agents Chemother anogenital herpes simplex virus type 2 in HIV‐infected patients:
1994;38:2454–7. evidence for predominant B‐lymphoplasmocytic infiltration and
160 Sáez‐Llorens X, Yogev R, Arguedas A et  al. Pharmacokinetics and immunomodulators as effective therapeutic strategy. Clin Infect Dis
safety of famciclovir in children with herpes simplex or varicella zoster 2013;57:1648–55.
virus infection. Antimicrob Agents Chemother 2009;53:1912–20. 184 Charles P, Richaud C, Beley S et  al. Pseudotumoral recto‐sigmoid
161 Superti F, Ammendotia MG, Merchetti M. New advances in anti herpes simplex virus type 2 in an HIV‐infected patient: Dramatic
HSV chemotherapy. Current Med Chem 2008;15:900–11. improvement with thalidomide. J Clin Virol 2016;78:12–13.
162 De Clercq E. The antiviral spectrum of (E)‐5‐(2‐bromovinyl)‐2’‐
185 Lin L, Chen XS, Cui PG et  al. Topical application of penciclovir
deoxyuridine. J Antimicrob Chemother 1984;14(suppl A):85–95. cream for the treatment of herpes simplex facialis/labialis: a ran-
163 Frobert E, Burrel S, Ducastelle‐Lepretre S et al. Resistance of herpes domized, double‐blind, multicentre, aciclovir‐controlled trial.
simplex viruses to acyclovir: an update from a ten‐year survey in J Dermatol Treat 2002;13:67–72.
France. Antiviral Res 2014;111:36–41. 186 Hasler‐Nguyen N, Shelton D, Ponard G et  al. Evaluation of the in
164 Levin MJ, Weinberg A, Leary JJ et al. Development of aciclovir resistant vitro skin permeation of antiviral drugs from penciclovir 1% cream
herpes simplex virus early during the treatment of herpes neonato- and acyclovir 5% cream used to treat herpes simplex virus infection.
rum. Pediatr Infect Dis 2001;20:1094–7. BMC Dermatol 2009;9:3.
165 Mertz GJ. Herpes simplex virus. In: Galasso GJ, Whitley RJ, Merigan 187 Hamuy R, Berman B. Treatment of herpes simplex virus infections
TC (eds) Antiviral Agents and Viral Diseases of Man, 3rd edn. New with topical antiviral agents. Eur J Dermatol 1998;8:310–19.
York: Raven Press, 1990:265–300. 188 Sacks SL, Thisted RA, Jones TM et  al. Clinical efficacy of topical
166 Erlich KS, Mills J, Chatis P et al. Aciclovir‐resistant herpes simplex docosanol 10% cream for herpes simplex labialis: a multicenter,
virus infections in patients with the acquired immunodeficiency randomized, placebo‐controlled trial. J Am Acad Dermatol
syndrome. N Engl J Med 1989;320:293–6. 2001;45:222–30.
167 Piret J, Boivin G. Antiviral drug resistance in herpesviruses other 189 Vere Hodge A, Field HJ. Antiviral agents for herpes simplex virus.
than cytomegalovirus. Rev Med Virol 2014;24:186–218. In: De Clercq E (ed.) Antiviral Agents. San Diego: Academic Press,
168 Crumpacker CS. Mechanism of action of foscarnet against viral 2013.

SECTION 9: VIRAL SKIN


polymerases. Am J Med 1992;92:3S–7S. 190 Tatnall FM, Schofield JK, Leigh IM. A double‐blind, placebo‐
169 Blot N, Schneider P, Young P et al. Treatment of an aciclovir and controlled trial of continuous acyclovir therapy in recurrent ­erythema
foscarnet‐resistant herpes simplex virus infection with cidofovir in a multiforme. Br J Dermatol 1995;132:267–70.

INFECTIONS
child after an unrelated bone marrow transplant. Bone Marrow 191 Eo SK, Pack C, Kumaraguru U et al. Optimisation of DNA vaccines
Transplant 2000;200:903–5. for the prophylaxis and modulation of herpes simplex virus infec-
170 Safrin S, Cherrington J, Jaffe HS. Cidofovir. Review of current and tions. Expert Opin Biol Ther 2001;1:213–25.
potential clinical uses. Adv Exp Med Biol 1999;458:111–20. 192 Whitley RJ, Roizman B. Herpes simplex viruses: is a vaccine tenable?
171 Florescu DF, Keck MA. Development of CMX001 (Brincidofovir) for J Clin Invest 2002;110:145–51.
the treatment of serious diseases or conditions caused by dsDNA 193 Skinner GRB, Fink C, Melling J et al. Report of 12 years experience in
viruses. Expert Rev Anti Infect Ther 2014;12:1171–8. open study of Skinner herpes simplex vaccine towards prevention of
172 Painter W, Robertson A, Trost LC et  al. First pharmacokinetic and herpes genitalis. Med Microbiol Immunol 1992;180:305–20.
safety study in humans of the novel lipid antiviral conjugate CMX001, 194 Dundarov S, Andonov P. Seventeen years of application of herpes
a broad‐spectrum oral drug active against double‐stranded DNA vaccines in Bulgaria. Acta Virol 1994;38:205–8.
viruses. Antimicrob Agents Chemother 2012;56:2726–34. 195 Lu Z, Brans R, Akhrameyeva NV et  al. High‐level expression of
173 Williams‐Aziz SL, Hartline CB, Harden EA et  al. Comparative
glycoprotein D by a dominant‐negative HSV‐1 virus augments its
activities of lipid esters of cidofovir and cyclic cidofovir against efficacy as a vaccine against HSV‐1 infection. J Invest Dermatol
replication of herpesviruses in vitro. Antimicrob Agents Chemother 2009;129:1174–84.
2005;49:3724–33. 196 Skoberne M, Cardin R, Lee A et al. An adjuvanted herpes simplex
174 El‐Haddad D, El Chaer F, Vanichanan J et  al. Brincidofovir
virus 2 subunit vaccine elicits a T cell response in mice and is an
(CMX‐001) for refractory and resistant CMV and HSV infections in effective therapeutic vaccine in Guinea pigs. J Virol 2013;87:3930–42.
612 

CHA PTER  5 1

Varicella Zoster Virus Infections


Manuraj Singh1 & Judith Breuer2
St George’s University Hospitals, London, UK
1 

Great Ormond Street Hospital, UCL Division of Infection and Immunity, London, UK
2 

Abstract spread in skin is dependent on replication within differentiating


keratinocytes. Knowledge of the pathogenesis of VZV infections
and latency has improved with in vitro models and xenograft
Varicella zoster virus (VZV) is characterized by its tropism for
studies in the severe combined immunodeficiency (SCID) mouse
epithelial tissue and persistence following primary infection
­
model. However, the exact mechanisms responsible for latency
in neuronal tissue. Primary infection with VZV causes varicella
and reactivation currently remain unresolved. Therapy for VZV
(chickenpox), a common childhood infection manifesting as a
infections, such as aciclovir, is not indicated in uncomplicated
­vesicular rash on the face and trunk. Following primary i­nfection
­infections in childhood but can be lifesaving in patients with sig-
the virus infects sensory nerve endings to establish latency in
nificant immunosuppression. The development of effective live
dorsal root or cranial nerve ganglia. Reactivation of the virus
attenuated vaccines has been successful in reducing the incidence
causes a painful unilateral dermatomal rash known as herpes
and complications of VZV. However, the vaccine is not currently
zoster or ‘shingles’. Epithelial replication is central to the ­natural
recommended for widespread use in the childhood immunization
history of VZV infection. Following reactivation, VZV initially
programme within the UK.
­infects the richly innervated isthmus of the hair follicle. Further

Key points features within the nucleus are nuclear enlargement, peripheral
condensation of chromatin, eosinophilic inclusions and
multinucleation.
• Varicella zoster virus (VZV) is an α‐herpesvirus, like herpes
• Definitive diagnosis of VZV infections is usually achieved by
simplex virus. VZV causes primary varicella (chickenpox) and,
polymerase chain reaction (PCR) or immunohistochemistry using
after reactivation from latency, herpes zoster (shingles).
specific VZV antibodies.
• Primary varicella is a common exanthema of childhood. Herpes
• Treatment of uncomplicated primary varicella in healthy children
zoster is less common in children but may be increased with
is generally not required. However patients with a history of
underlying immunodeficiency, such as human immunodeficiency
chronic skin disease such as moderate‐to‐severe eczema should
virus (HIV).
receive systemic treatment. Rapid initiation of intravenous
• The histological features of VZV infections are very similar
aciclovir can significantly reduce mortality in disseminated VZV
to those of HSV. They are characterized by varying degrees
infections in immunocompromised patients.
of epidermal blistering and necrosis. The typical cytopathic

globe [2–4]. It is unclear whether the strain distribution is


Brief introduction/history. Varicella zoster virus (VZV) is
actually driven by climate or other factors such as immi-
characterized by its tropism for epithelial tissue and per-
SECTION 9: VIRAL SKIN

gration patterns. The most current genotypic classification


sistence following primary infection in neuronal tissue.
divides the virus into six different clades. Although clades
Primary infection with VZV causes varicella (chicken-
1 and 3 are predominant in Europe and North America,
INFECTIONS

pox), an infection manifesting as a vesicular rash on the


and clade 2 is predominant in Japan, there are no strict
face, trunk and proximal limbs. Following primary infec-
geographical implications in this naming nomenclature.
tion the virus infects sensory nerve endings to establish
Recent studies have also shown intra‐ and inter‐clade
latency in the dorsal root or cranial nerve ganglia.
recombination of the VZV genome [5–9].
Reactivation of the virus causes a painful unilateral der-
matomal rash known as herpes zoster or ‘shingles’.
Epithelial replication is central to the natural history of Epidemiology. Primary varicella (chickenpox) demon-
VZV infection [1]. strates a prominent seasonal pattern in temperate cli-
VZV is an α‐herpesvirus, which includes viruses such as mates, compared to herpes zoster which tends to occur
herpes simplex virus (HSV) 1 and 2, pseudorabies virus sporadically throughout the year. In temperate climates,
and Marek disease virus. Only VZV and HSV can infect chickenpox is most ­common in the winter and spring [10],
humans. Several studies have demonstrated a d ­ istinctive but a later peak in June occurs in France and the UK [11].
geographical distribution of the major VZV genotypes Chickenpox also demonstrates periodic epidemics,
aligning with cool versus warm climate regions of the ­followed by years of lower incidence [12]. Chickenpox is

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Chapter 51  Varicella Zoster Virus Infections 613

highly infectious with secondary attack rates in households vaccination has caused some controversy. There are some
ranging between 60% and 80% [13]. Chickenpox is a dis- models that predict an increase rather than a decrease
ease of childhood in temperate climates, with approxi- incidence due to the loss of exogenous immune boosting
mately 90% of cases occurring by the age of 10–14 years from the presence of reduced varicella circulation [28].
(mean, 6.7–10.6 years) [14]. The peak incidence of approx-
imately 90 cases/1000 tends to occur in the 1–4 age group Pathogenesis. All herpesvirus infections of the skin,
[15]. Although the incidence rate is much lower after the including HSV and VZV, result in epidermal cell lysis
age of 14, the case fatality rate rises from <5 to over 15 and clinical blistering. The model of primary varicella
deaths/100 000 cases in adults [16]. These figures are infection was first put together from the mouse pox
derived from the pre‐vaccination era, and the introduc- model described by Fenner in 1948 and clinical observa-
tion of widespread varicella vaccination is predicted tions [29,30]. This model describes two viraemic phases,
to  decrease the incidence by at least 57%, and related but this has been superseded by studies from human
complications by 76% over time [17]. The case fatality rate skin xenografts explanted on the severe combined
is also higher in infants, pregnant women and immuno- immunodeficiency (SCID) mouse model which suggest
suppressed patients, compared to healthy young ­children. that the long 14‐ to 15‐day incubation period is due to
Within tropical climates (e.g. India and the West Indies) innate immunity in the skin [31]. VZV induces an antivi-
the epidemiology of varicella is remarkably different, ral response in neighbouring uninfected keratinocytes.
with a higher proportion of cases occurring in the adult This includes upregulation of interferons, particularly
population. The reasons for this difference still remain interferon (IFN)‐α, and signal transducer and activator
unclear; earlier studies suggested that rural living condi- of transcription 1 (STAT1) which activates other innate
tions within these tropical countries reduce transmission cytokines and important antiviral defence proteins such
rates [18,19], but more recent data have shown lower as promyelocytic leukaemia (PML) protein. Using the
infectivity of the virus in tropical climates [9]. SCID model it has been shown that antibodies blocking
The overall incidence of herpes zoster ranges from 1.2 the action of IFN‐α cause increased viral titres and blis-
to 3.4 per 1000 person‐years, and in contrast to chicken- ter formation in human skin xenografts. In addition,
pox has a much higher incidence in the elderly popula- VZV triggers particular cellular responses in infected
tion [20]. For example, in one study, incidence rates as cells that are distinct from the antiviral response of
high as 11.8 cases per 1000 person‐years in the 65–75+ age neighbouring uninfected cells. These include the upreg-
group have been reported [21]. The complications of zos- ulation of signal transducer and activator of transcrip-
ter include acute and chronic pain, neurological compli- tion 3 (STAT3) and subsequent induction of the
cations, ocular disease, secondary infection and visceral antiapoptotic protein survivin. In contrast to uninfected
involvement. Zoster complications occur much more fre- cells there is also inhibition of IFN‐α and STAT1 [32].
quently in the elderly and immunosuppressed [22]. In the Using the SCID mouse model, several viral proteins
elderly the complication of post‐herpetic neuralgia (pain (e.g. IE62, gB, gC, ORF47, ORF61) have been shown to be
persisting for more than 1 month after the onset of the very important for replication in these three‐dimensional
rash) has an increased prevalence [23]. The pathogenesis skin grafts but some, like gC and OR47, are not essential
of this disorder is poorly understood. It is very debilitat- in monolayer cultures [33–37]. This finding highlights the
ing and commonly resistant to standard painkillers. importance of the interaction between VZV infection and
Herpes zoster does occur in children, although its age‐ epidermal differentiation. A separate study that analysed
the host and viral transcriptome keratinocytes confirmed

SECTION 9: VIRAL SKIN


specific incidence is less, and development of primary
varicella as an infant increases the risk. The risk of herpes that VZV gene and protein expression were significantly
zoster in patients with acute lymphoblastic leukaemia is increased with calcium‐induced epidermal differentia-

INFECTIONS
approximately 120 times greater than in patients without tion, as was viral replication [38].
an underlying malignancy [24]. Patients with suppressed In addition, certain proteins are more important for
cell‐mediated immune mechanisms are at increased risk replication in certain tissues. For example, mutations
of herpes zoster and its complications. Mortality is associ- causing deletions in the tegument protein ORF10 cause
ated with cutaneous dissemination and widespread vis- reduced infection in skin but have no effect on replication
ceral involvement. Patients with HIV/AIDS or certain in T cells [39]. Other examples of important viral proteins
cancers and transplant recipients fall into this high‐risk involved in skin replication include ORF61, which is cru-
group. Patients with haematological malignancies, par- cial for causing dispersal of antiviral promyelocytic leu-
ticularly Hodgkin lymphoma, and bone marrow trans- kaemia nuclear bodies (PML‐NBs) [40]. VZV glycoprotein
plant recipients are particularly at risk for the development E (gE) is essential for viral replication and also plays a role
of zoster. Untreated, the mortality rate is approximately in spread of the virus from cell to cell, secondary envelop-
20–40% in allogeneic bone marrow transplant recipients ment and viral entry [41,42]. The other glycoproteins
[25]. In Africa, herpes zoster is the commonest presenta- involved in similar processes to gE and also important for
tion of HIV [26]. The incidence rates of herpes zoster infection in skin are gB, gH and gI [43,44].
within the HIV population are 15–25 times higher than The SCID mouse model has also been used very pro-
the general population, and 3–7 times greater than the ductively to study the tropism of VZV in T cells and
elderly population [27]. The effect on herpes zoster neurons in fetal human thymus–liver and dorsal root
­incidence after the introduction of widespread varicella ganglia (DRG) xenografts respectively [45,46]. The skin
614 Section 9  Viral Skin Infections and Opportunistic Infections

xenografts are implanted beneath the mouse skin and induces a humoral response with production of IgA, IgM
the ­ thymus–liver and DRG xenografts are placed and IgG anti‐VZV antibodies [58]. The last of these anti-
beneath the kidney capsule. This model allows the skin bodies tends to persist; it is a marker of previous infection
to fully differentiate into an epidermis with a stratum and serves to protect against reinfection. Infected patients
corneum and underlying dermis. As a result the interac- are generally considered contagious during the prodrome
tion of VZV, including vaccine and mutant strains, with and up until all the lesions are dry and crusted. VZV has
epidermal differentiation can be studied in more detail the ability to interfere with the expression of major histo-
[47]. Xenograft tissue infected with VZV can be har- compatibility complex (MHC) class I and class II proteins
vested at different time points and analysed using vari- required for CD8 and CD4 T‐cell recognition respectively,
ous techniques to look at gene and protein expression of leading to delayed clearance of virus‐infected cells
viral and epidermal proteins. This model also allows [59–62]. Open reading frame 66 (ORF66) of VZV encodes
assessment of the effect of interfering with cellular or a protein kinase that modulates apoptosis and interferon
viral proteins by treating the mice with various antibod- pathways, downregulates MHC class I protein expression
ies or inhibitors. In addition, novel antiviral therapies on cell membranes, and facilitates tropism of VZV for T
can be assessed using this model [48]. cells [63].
The method of transmission is primarily via the aerosol As stated previously, it is known that several VZV pro-
route from close contacts. The virus is thought to replicate teins, although dispensable for growth in tissue culture
first in the adenoids and tonsils [49], then the regional monolayers, are essential for skin infection in vivo. This dis-
lymph nodes, followed by a viraemia [50]. Viral transfer to crepancy is thought to relate to the interaction of VZV
migrating T cells in the tonsils may facilitate cell‐­associated and  epidermal differentiation. Using a calcium‐induced
viraemia, and preferential infection of CD4 T cells that keratinocyte differentiation model and RNA‐seq analysis
express skin homing markers may enhance VZV transport we showed that VZV infection specifically alters epidermal
to cutaneous sites of replication [51]. The precise mecha- differentiation, driving a specific pattern of changes that
nisms for virus transfer to skin are still not known, but T are characteristic of blistering and skin‐shedding diseases
cells may migrate out of capillaries to directly infect skin [38]. In particular VZV specifically reduces expression of
compartments, or transfer VZV directly to endothelial specific suprabasal cytokeratins such as keratin 1 (KRT1)
cells, from which the virus then spreads to epithelial com- and keratin 10 (KRT10) and desmosomal proteins, particu-
partments. The tropism of VZV for the melanocyte in tis- larly desmoglein 1 (DSG1) and desmocollin 1 (DSC1), lead-
sue culture may suggest the melanocyte as a putative skin ing to disruption of epidermal structure and function.
target in vivo [52]. A predominant mechanism by which These changes, together with upregulation of kallikreins
VZV enters cells is a process known as virus–cell fusion. and serine proteases, are also observed in genetic disorders
VZV receptors found to date include heparan ­sulphate of skin barrier function such as Netherton syndrome. These
proteoglycans [53], cation‐independent mannose findings taken together strongly suggest that VZV infec-
6‐­phosphate receptor (MPRci) [54] and insulin degrading tion promotes blistering and desquamation of the epider-
enzyme (IDE) [55]. VZV binds to these receptors via its mis, both of which are essential for viral spread.
glycoproteins, which include gB, gH, gE and gI. Binding In summary, studies such as these highlight the inti-
to these receptors triggers fusion between the viral and mate host–pathogen interaction following VZV infection
host envelope, releasing the viral tegument proteins and of skin and show that VZV is dependent on epidermal
nucleocapsid into the host cell cytoplasm. The tegument differentiation and remodels the epidermal environment
proteins then migrate to and enter the host cell nucleus,
SECTION 9: VIRAL SKIN

to promote its own replication and spread.


while the naked viral nucleocapsid fuses with the nuclear Histological examination of newly formed vesicles
membrane and releases the viral genome into the nucleus demonstrates prominent involvement of endothelial cells
INFECTIONS

[56]. The tegument contains key proteins such as IE62 and at an early stage. Epidermal alterations caused by the
IE63 that are key facilitators of viral gene transcription. virus are characterized by swelling of the cytoplasm and
Four VZV envelope glycoproteins contain mannose‐6‐ nucleus, followed by nuclear inclusions, cell fusion and
phosphate (Man 6‐P), and Man 6‐P blocks infection of cells balloon degeneration. Cell fusion results in the character-
by cell‐free VZV. A nice study showed that cell lines defi- istic multinucleated giant cell (MNC), a pathological hall-
cient in MPRci are resistant to infection by cell‐free virus mark of cutaneous herpes infections [64]. However,
but are readily infected by cell‐associated virus [54]. In histology alone cannot distinguish between HSV and
addition, these MPRci‐deficient cell lines produced higher VZV infections, and this must be confirmed using specific
titres of cell‐free virus when infected by cell‐­associated immunohistochemical antibodies or molecular tech-
virus. This result suggests that intracellular MPRci appear niques. The vesicles are formed by the exudation of clear
to divert newly enveloped VZV to late endosomes for fluid from infected degenerating epithelial cells, and
destruction. Further work by the same group showed that ­contain numerous virions. The fluid turns turbid after
staining of VZV‐infected skin lesions demonstrates a migration of inflammatory cells into the vesicle. The floor
reduction in MPRci expression with keratinocyte differen- of the vesicle is formed by MNCs of the basal layer, and
tiation and provides an explanation for increased cell‐free the roof by the stratum spinosum of the epidermis. The
virus within the suprabasal layers of the epidermis [54,57]. vesicle roof eventually breaks down, releasing infective
The production of this cell‐free virus within the epider- virions into the atmosphere for further transmission to
mis facilitates person‐to‐person spread. Primary infection nonimmune hosts.
Chapter 51  Varicella Zoster Virus Infections 615

There are two modes of viral spread to the dorsal root and no production of infectious virus. However, deep
ganglia, where the virus lies dormant for the remaining sequencing using a technique called RNA‐seq detected
lifetime of the host. The first is the haematogenous route multiple viral mRNAs at low levels, the functional sig-
via infected T cells, which takes place during the viraemic nificance of which was considered unclear. This study
phase of primary varicella [65], and the second is the also showed that latently infected neurons treated with
­retrograde neuronal transport of virions from mucocuta- antibodies to nerve growth factor (anti‐NGF Ab), simi-
neous lesions [66]. The mechanisms controlling latency lar to HSV, resulted in reactivation and production of
and reactivation of VZV are incompletely understood, infectious virus.
largely due to a lack of in vitro or in vivo models that This same study also compared a vaccine strain
reproduce both latency and reactivation. VZV latency and (vOka)  with its parental wild type, designated pOka.
reactivation differ in several ways from HSV (see vOka is used as live attenuated vaccine and is effective
Chapter  50), as VZV is known to reactivate much less at preventing both primary varicella and herpes zoster.
­frequently than HSV, and reactivation is more common in It was originally derived from pOka by serial propaga-
older individuals. tion in cultured cell lines [77,78]. vOka is a mixture of
Studies of sensory ganglia at autopsy have shown that genotypes and differs from pOka by mutations in multi-
about 4% of neurons contain low copy numbers (2–9/cell) ple different regions of the genome. This genetic differ-
of VZV genomes [67]. Latent viral DNA exists in a state ence is likely to account for its attenuation [7,47,79–82].
where the genome of the virus is joined ‘end to end’ vOka was equivalent to pOka in establishing latency
to form structures consistent with unit length episomes/ but  was impaired for reactivation after neurons were
concatemers and from which viral gene transcription is treated with anti‐NGF Ab. vOka also showed lower
restricted. There is increasing evidence that this restriction ­levels of viral transcription by RNA‐seq analysis during
is epigenetically regulated. One proposed mechanism for latency compared to pOka.
α‐herpesvirus reactivation, including HSV and VZV, The result of continued VZV reactivation is the pain-
involves a three‐stage process of latency, generalized ful vesicular rash herpes zoster, which usually involves
deregulation (animation) and progression through DNA the dermatomal distribution of a single sensory nerve.
replication. Models looking at explants of VZV‐infected The infectious virus is carried in an antegrade direction
human trigeminal ganglia taken post mortem show gener- towards the skin. Occasionally patients may present
alized deregulation, characterized by transcription of with the characteristic pain in a dermatomal distribution
­multiple viral genes. This is followed by reactivation char- without the rash [83]. Infection is then restricted by
acterized by coordinated VZV DNA replication, but the innate cutaneous responses in addition to VZV‐specific
exact mechanisms that trigger this transition are not fully T cells. The increased frequency and risk of severe zoster
understood to date [67]. in the elderly and immunosuppressed populations is
There has been controversy regarding the expression related to a deficiency in VZV cell‐mediated immunity.
and translation of viral VZV genes within neurons of sen- IFN‐γ‐secreting VZV‐specific CD4+ T cells in the blood
sory and cranial nerve ganglia. Many studies have found are significantly reduced in the elderly. In addition, a
low‐level transcription of ORF63, which encodes the pro- relatively recent study has found that the number of
tein IE63, to be the most abundantly expressed gene VZV‐specific CD4+ T cells is equivalent in the skin of the
and protein respectively in human ganglia post mortem young and elderly. However, in the elderly there is an
[68–70]. IE63 has antiapoptotic functions that might facili- increased number of inhibitory T cells, such as Foxp3+

SECTION 9: VIRAL SKIN


tate the transition to latency [71,72]. However, the pres- regulatory T cells, in the skin [84]. The increased risk of
ence of IE63 is yet to be confirmed in human ganglia in herpes zoster in HIV‐infected patients would suggest
vivo, and may be confounded by cross‐reacting antibodies that VZV‐specific T cells also limit reactivation in ganglia,

INFECTIONS
[73,74]. The expression that is seen immediately post mor- however this is still not currently proven. It is interesting
tem may reflect very early reactivation. Only ORF63 to note that the histological features of primary varicella
mRNA could be detected by reverse transcriptase‐­ and herpes zoster are very similar. However, herpes
quantitative polymerase chain reaction (RT‐qPCR), in ­zoster lesions tend to show more folliculo‐sebaceous and
ganglia obtained less than 9 hours post mortem. However, endothelial inflammation [85]. This may reflect the high
increased levels of ORF63 mRNA and multiple other VZV density of cutaneous nerves around these structures. The
mRNAs were detected when ganglia were obtained more histopathological examination of ganglia during VZV
than 9 hours post mortem. This later finding suggests that reactivation shows inflammation, necrosis and cellular
the detection of multiple VZV mRNAs reflects reactiva- disruption of neuronal and non‐neuronal cells [86]. This
tion rather than true latency [75]. inflammation may extend to the anterior horn of the
A recent study by Sadaoka et al. has shown that viral ­spinal cord, resulting in motor dysfunction [87,88].
gene expression may be even more restricted than this
during latency [76]. Using an in vitro system of neurons Clinical features and differential diagnosis
derived from human embryonic stem cells (hESCs), Chickenpox
they were able to produce selective axonal infection Primary varicella (chickenpox) is a highly contagious
and maintenance of latent VZV using cell‐free virus. exanthema which is most common in childhood with a
This latent infection model was characterized by an peak incidence between 5 and 9 years of age [89]. The
inability to detect viral gene expression by RT‐qPCR infection is transmitted by aerosol droplets from the
616 Section 9  Viral Skin Infections and Opportunistic Infections

r­espiratory tract and is initially characterized by a viral usage of salicylates in children has been abandoned [95].
prodrome consisting of fevers, malaise, myalgia, ­headache Certain patient groups such as adults, pregnant women
and arthralgia. This prodrome typically lasts 2–3 days and the immunocompromised have a higher risk of these
before onset of the characteristic vesicular eruption. The complications. Up to half the deaths from chickenpox
incubation period ranges from 10 to 21 days with a mean occur in adults [11]. In immunocompromised hosts the
of 14–17 days. The eruption typically occurs in crops giv- mortality risk is higher and lesions may become large and
ing rise to lesions at different stages of evolution. The ulcerated (varicella gangrenosa) or even chronic and ver-
time to crusting and noninfectivity varies between 2 and rucous in nature [96]. The mortality rate ranges from
12 days. The initial eruption consists of erythematous approximately 10% to 30% in immunosuppressed patients
macules 2–4 mm in diameter, followed by tear drop such as those suffering from leukaemia, HIV or congeni-
­vesicles (likened to ‘dew drops on rose petals’) that subse- tal immunodeficiency and organ transplant recipients
quently become pustular and crusted (Fig. 51.1). The rash [97]. Among the congenital immunodeficiencies, those
is most prominent on the trunk followed by the scalp and associated with natural killer (NK) or natural killer T
proximal limbs. Mucosal involvement is relatively com- (NKT) cell deficiency are particularly associated with
mon although palm and sole involvement is a rare event. severe and potentially life‐threatening primary varicella
The eruption is usually pruritic but this symptom varies [98]. Patients with primary immunodeficiencies associ-
from mild to severe. Systemic symptoms tend to be mild ated with reduced interferon levels are also at risk of
in children but are frequently severe in neonates, adults severe primary varicella [31,99]. Other much rarer
and immunocompromised patients [90]. complications include nephritis, hepatitis, myocarditis,
­
Common complications of varicella include skin and arthritis, orchitis, uveitis, glomerulonephritis, rhabdomy-
soft tissue infections, usually due to Group A Streptococcus olysis, haemorrhagic disease and thrombocytopenia.
and Staphylococcus aureus [91,92]. This occurs in approxi- Purpura fulminans is a recognized, albeit rare complica-
mately 5–10% of cases. Rare reports of necrotizing fasciitis tion of primary varicella in children due to acquired
post primary varicella have been reported [93]. Although deficiencies in protein S and/or protein C [100–103].
­
rare, pulmonary and neurological syndromes are the next Blood tests commonly show features of disseminated
most common [94]. In children, pneumonia is usually due intravascular coagulation. Another important complica-
to secondary bacterial infection compared to direct viral tion of primary varicella is the increased risk of stroke in
pneumonitis in adults. The rare neurological complica- the immediate months after infection. A study by Thomas
tions include aseptic meningitis, encephalitis, cerebellar et al. showed that there was a fourfold increased risk of
ataxia, Guillain–Barré syndrome (GBS), transverse myeli- stroke in children up to 6 months after chickenpox
tis and Reye syndrome, which is now uncommon because compared to a twofold increased risk in adults [104].
­
Interestingly, there was no significant risk of stroke
7–12 months after chickenpox in either adults or children
[105]. The post‐varicella arteriopathy predominantly pre-
sents with acute arterial ischaemic stroke or transient
ischaemic attack and is associated with vascular stenosis
that tends to subsequently regress [106]. There is also
some evidence that VZV vasculopathy may occasionally
lead to the development of intracerebral aneurysms, with
SECTION 9: VIRAL SKIN

or without haemorrhage. In addition, there has been a


link between VZV vasculopathy and giant cell arteritis
[107]. VZV vasculopathy is diagnosed by the presence of
INFECTIONS

either VZV DNA or anti‐VZV IgG antibody in cerebrospi-


nal fluid [108,109].
The development of primary varicella in pregnancy can
affect both the mother and the fetus. Although rare, vari-
cella pneumonia may lead to maternal death. Intrauterine
infection carries the risk of congenital varicella syndrome
(CVS), which is approximately 1.2–2.2%. The greatest risk
of this complication occurs in mothers who contract
­primary varicella within the first two trimesters. The man-
ifestations include low birthweight and skin lesions that
typically manifest as scar‐like lesions in a dermatomal dis-
tribution and/or aplasia cutis. Other well‐recognized
­features include cataracts, microphthalmia, chorioretini-
tis, skeletal hypoplasia and neurological abnormalities.
The last complication is typified by mental retardation,
microcephaly and dysfunction of urinary and rectal
Fig. 51.1  Varicella/chickenpox. Source: Courtesy of Dr Friedlander and sphincters [110]. Prenatal diagnosis is now aided by
Dr Tom. ­various tests including serial ultrasound examination,
Chapter 51  Varicella Zoster Virus Infections 617

virus‐specific IgM estimation and amniocentesis to confirm pustular and crusted, as in primary varicella. They may
viral transmission by PCR techniques [111–113]. After also coalesce to form large bullae and may take on a
20 weeks’ gestation the risk of CVS is negligible although haemorrhagic appearance.
there is an increased risk of herpes zoster in early life. In Common complications include secondary bacterial
addition, a large study found no cases of CVS in mothers infection, scarring and keloid formation. Rarely necrotiz-
who developed herpes zoster in pregnancy [110]. ing fasciitis may complicate an episode of herpes zoster,
The development of primary varicella less than 7 days as in primary varicella. Post‐herpetic neuralgia, defined
before birth in the mother carries a very high risk of as pain persisting many months after the acute episode, is
­neonatal varicella, due to a lack of protective maternal a relatively common complication in the elderly but is less
antibodies in the newborn [114]. This is a serious compli- common in childhood cases [122]. It is important to recog-
cation with a 30% mortality rate if left untreated, mostly nize involvement of the ophthalmic branch of the trigemi-
due to hepatitis and pneumonitis. The risk is said to be nal nerve which may produce serious eye complications.
highest for mothers who contract the disease from 5 days The Ramsay Hunt syndrome is due to involvement of the
before delivery to 2 days after. With prompt treatment in seventh cranial nerve and is characterized by facial nerve
the form of varicella zoster immune globulin (VZIG) and paralysis, palatal and external auditory canal lesions, and
intravenous aciclovir the mortality rate drops to approxi- auditory and vestibular symptoms [123].
mately 5% [115]. The VZV vaccine is a live attenuated vaccine and there
is a risk of herpes zoster in children who receive
Herpes zoster the ­vaccine. Another very important complication is the
Herpes zoster or shingles is due to reactivation of VZV high risk of more severe disease and potentially fatal
from dorsal root ganglia of sensory nerves. This is analo- disseminated herpes zoster in immunocompromised
gous to reactivation of herpes simplex virus but reactiva- patients. This is especially pertinent to those who have
tion typically occurs less frequently with VZV. Herpes significant cell‐mediated immunodeficiency such as leu-
zoster in children is not uncommon but if recurrent or kaemia patients and bone marrow and organ transplant
multidermatomal should prompt investigation to exclude recipients [124]. The presentation in such patients also
an underlying immunodeficiency such as HIV [116]. The tends to be more atypical, with lesions tending to be
introduction of widespread varicella vaccine, as in the more bullous, haemorrhagic and ulcerative. The lungs,
USA, has reduced the incidence of herpes zoster by liver, gastrointestinal tract and brain tend to be sites of
65–79% in vaccinated individuals. However, there are preferential involvement in disseminated disease [125].
data showing an increased incidence of herpes zoster The most common brain manifestation is a progressive
among children with varicella infection aged ≥2 years in leukoencephalitis.
the post varicella vaccination era [117,118]. This may Verrucous VZV, characterized by persistent hyperkera-
reflect loss of exogenous immune boosting due to reduced totic lesions resembling viral warts, is an unusual but
varicella circulation. It is important to realize that herpes well‐recognized presentation in patients with immunode-
zoster is a common presentation of HIV in Africa. Herpes ficiency and is most commonly seen in patients with AIDS
zoster typically presents as a painful and vesicular rash in [126]. Similar lesions have occasionally been reported in
a unilateral and dermatomal distribution. Rarely, noncon- transplant patients and rarely as a consequence of intrau-
tiguous multidermatomal cutaneous involvement may terine infection. Interestingly a study showed reduced
occur. Although less common in childhood, herpes zoster expression of the envelope proteins gE and gB in chronic

SECTION 9: VIRAL SKIN


is a common disorder in the elderly and immunocompro- verrucous lesions [127]. In addition, herpes zoster may
mised host, and affects 10–20% of people during their life- complicate the immune reconstitution inflammatory syn-
time. The disorder can affect any dermatome but the most drome (IRIS) in HIV patients treated with antiretroviral

INFECTIONS
common are thoracic, lumbar and trigeminal nerve therapy [128].
involvement resulting in facial lesions. The risk of child- Another interesting but under‐recognized phenom-
hood zoster is greatly increased in patients with immuno- enon is the occurrence of Wolf’s isotopic response in
compromise or malignancies [119]. Childhood‐onset the scars of resolved herpes zoster [129]. Wolf’s iso-
systemic lupus erythematosus (SLE), commonly associ- topic response is the name given to a separate and new
ated with lymphopenia and immunosuppressive treat- skin disorder occurring at the site of a healed and
ment, is also a significant risk factor for herpes zoster, unrelated condition. There have been many conditions
although most patients had a benign outcome in a large associated with this phenomenon occurring at the site
study [120]. In addition, herpes zoster can develop in of herpes zoster scars, including granuloma annulare,
immunocompetent children as a consequence of intrau- sarcoid, granulomatous vasculitis, lichen sclerosus,
terine infection or postnatal exposure at an early age lymphoma, pseudolymphoma, leukaemia cutis and
[121]. In adults and the elderly, herpes zoster is usually a skin cancers.
febrile illness preceded by paraesthesia or pain in the Herpes zoster may also be complicated by acute, suba-
affected dermatome. However, these prodromal features cute and chronic neurological complications, even occa-
are typically lacking in childhood cases. If there is a sionally in the absence of a clinical rash [95,130].
­prodrome, the rash typically occurs after 2–4 days and is Neurological complications include cranial nerve palsies,
characterized by grouped vesicles on an erythematous motor paresis, ocular disorders and vasculopathy. Due
and oedematous plaque. These vesicles rapidly become to  this vasculopathy, and similar to primary varicella,
618 Section 9  Viral Skin Infections and Opportunistic Infections

herpes zoster is associated with a subsequent increased


risk of stroke and myocardial infarction (approximately
twofold for both). Again the risk is mainly within the first
6 months after the zoster episode and zoster vaccination
does not seem to modify the risk [131]. This increased risk
of acute cardiovascular events following an episode of
zoster is seen particularly below the age of 40 [132]. The
pathogenesis is similar to the VZV vasculopathy follow-
ing primary varicella described previously.
The differential diagnosis of VZV primarily involves
HSV infections, particularly when VZV infections are
localized such as in herpes zoster, and the ulcerative or
verrucous lesions occurring in immunocompromised
patients. Although primary and recurrent HSV‐1 and
‐2 infections are usually localized, they can produce a
more widespread systemic infection in neonates and
immunocompromised patients mimicking primary vari- (a)
cella. PCR studies can readily distinguish between the
viruses HSV‐1, HSV‐2 and VZV [133]. Other considera-
tions for primary varicella, depending on the clinical set-
ting, may include immunobullous disorders, severe drug
reactions such as Stevens–Johnson syndrome, bacterial
folliculitis or disseminated bacterial or fungal infections
in immunosuppressed patients.

Laboratory/histology findings. The histological features


of the cutaneous lesions of VZV in the form of primary
varicella or herpes zoster are very similar to those of
HSV‐1 or ‐2. Isolated or prominent follicular involvement
is said to be a pointer in favour of VZV infections, particu-
larly herpes zoster, when compared to HSV [134]. In fully
developed lesions there are varying degrees of epidermal
necrosis and blister formation within the epidermis
(Fig. 51.2a and b). The two main mechanisms involved in (b)
the formation of these intraepidermal vesicles/blisters are Fig. 51.2  (a) Low‐power histology of a case of herpes zoster showing an
known as balloon and reticular cell degeneration. However, intraepidermal blister filled with eosinophilic fluid and containing
the pathognomonic changes are evident within the nucleus. acantholytic cells. Similar findings would be seen in primary varicella
These consist of nuclear enlargement with peripheral con- and herpes simplex infections. (Haematoxylin–eosin stain; original
magnification ×40.) (b) High‐power histology of same case as (a)
densation of chromatin giving rise to a ground glass
demonstrating characteristic cytopathic changes of varicella zoster virus
SECTION 9: VIRAL SKIN

appearance of the nucleus. The earliest abnormality within infections. The infected cells near the base of the blister demonstrate
the cytoplasm is vacuolization. In addition, there are moulding, enlargement and peripheral condensation of nuclear chromatin
usually varying degrees of multinucleation of infected
­ giving rise to a ground glass appearance. Characteristic multinucleated
INFECTIONS

keratinocytes and intranuclear eosinophilic inclusions. giant cells are also seen. In addition intranuclear and intracytoplasmic
There is commonly a fibrinopurulent exudate associated eosinophilic inclusions are seen focally, the former are known as Cowdry
with a dermal infiltrate that varies from mild to dense. This type A inclusions. (Haematoxylin–eosin stain; original magnification ×400.)

infiltrate is typically composed of perivascular, periad-


nexal and interstitial lymphocytes with variable numbers which typically ­terminates under the epidermis via
of neutrophils, and occasionally eosinophils may be promi- nonmyelinated nerve fibres.
nent. A lichenoid and/or vasculitic reaction pattern in the Rare histological presentations of VZV are more com-
absence of typical cytopathic changes has been termed monly seen in immunosuppressed patients. They include
‘herpes incognito’ [135]. When faced with this combination vasculitis and adnexal involvement, including folliculo‐
of reaction patterns in a histological section, it would be sebaceous and/or eccrine involvement, in the absence of
prudent to perform serial sections to look for diagnostic typical epidermal changes (Fig.  51.3a and b) [138,139].
nuclear cytopathic changes. The verrucous or wart‐like lesions that are typically
Early biopsies from herpes zoster typically show encountered in immunosuppressed patients such as those
cytopathic changes within the follicular epithelium with HIV demonstrate hyperkeratosis and acanthosis
[136]. This reflects the neural pathway of viral spread with viral cytopathic changes and less vesiculation [126].
from the dorsal root ganglia via myelinated nerve fibres Clinical features usually allow the distinction between
that terminate at the level of the isthmus of the hair fol- HSV and VZV but there are many situations in which this
licle [137]. This is in contrast to the reactivation of HSV may be difficult or impossible. This is especially the case
Chapter 51  Varicella Zoster Virus Infections 619

techniques that can be used to diagnose cutaneous herpes


infections include electron microscopy, immunofluores-
cence of blister contents, growth in tissue culture and
in  situ hybridization. However, the most common tech-
niques employed in routine diagnostic practice are immu-
nohistochemistry and PCR [141].

Treatment and  prevention. Treatment of chickenpox in


previously healthy children is primarily symptomatic.
Prevention of secondary bacterial infection and allevia-
tion of pruritus and/or pain are the main goals. Oral
­paracetamol and ibuprofen may be used for fever and/or
pain and oral antihistamines can be prescribed for pruri-
tus. Topical therapies such as calamine lotion may p­ rovide
symptomatic relief from pruritus.
VZV infections may be treated by administration of
(a) one of the following licensed drugs: aciclovir (and its
prodrug valaciclovir), penciclovir (and its prodrug fam-
ciclovir), ganciclovir, cidofovir, brincidofovir, brivudine
and foscarnet [142]. By far the most commonly used
drug is aciclovir and its related compounds. All these
drugs work by inhibiting DNA replication. Aciclovir,
ganciclovir and penciclovir are nucleoside analogues
that must be phosphorylated to the monophosphate
form by virus‐encoded thymidine kinases (TKs) and
then further phosphorylated to triphosphate forms by
cellular kinases before they become competitive inhibi-
tors of DNA polymerase by competing with the binding
of natural deoxyguanine triphosphate. The other drugs
inhibit viral DNA synthesis independent of viral TKs
[143]. See herpes simplex infections (Chapter  50,
Treatment and prevention) for more details of cidofovir,
brincidofovir, brivudine and foscarnet.
(b) Generally, systemic antiviral therapy should be
Fig. 51.3  (a) Low‐power histology of a renal transplant patient who reserved for complicated varicella and infections occur-
presented with a painful purpuric rash down one leg. This case ring in children with significant risk factors for moderate
­demonstrates a rare presentation of herpes zoster without epidermal or life‐threatening disease. These include patients with
involvement. The histology shows a mild upper dermal infiltrate associated immunocompromise, moderate‐to‐severe chronic skin
with prominent red cell extravasation and a multinucleated cell within the
disorders such as eczema, those over 12 years of age and
hair follicle. (Haematoxylin–eosin stain; original magnification ×100.)

SECTION 9: VIRAL SKIN


(b) Immunofluorescence microscopy of same case as (a), using a polyclonal
those on long‐term oral corticosteroids. Several large
antibody against IE63 (an immediate early varicella zoster virus protein). randomized controlled studies have shown that oral
­
There is positive red staining within the hair follicle confirming a diagnosis ­aciclovir treatment for chickenpox is most effective when

INFECTIONS
of herpes zoster presenting with purpura and absent epidermal given within the first 24 hours [144,145]. This early treat-
­involvement (note lack of red staining within the epidermis). ment was also associated with reduced viral shedding
(Original magnification ×40.) and a quicker resolution of the rash and associated consti-
tutional symptoms. A 5‐day course of aciclovir was also
in disseminated infections and those occurring with atyp- found to be sufficient, as treatment continued for 7 days
ical presentations in immunocompromised patients. Since provided no additional benefit [146]. If the patient is
the histological findings of HSV and VZV are very simi- severely immunocompromised, intravenous aciclovir
lar, a definitive diagnosis is usually made either via therapy should be started as soon as possible. The recom-
specific immunohistochemistry or by PCR/molecular
­ mended dose in this situation is 10 mg/kg every 8 hours
methods on viral swabs. HSV‐1 and ‐2 antibodies are rou- or 500 mg/m2 per dose three times daily for 7–10 days,
tinely available in most diagnostic cellular pathology and should be given as soon as possible and ideally
laboratories. Although there are sensitive and specific within 48–72 hours. Studies have shown that intravenous
VZV antibodies commercially available, they tend not to aciclovir prevents potentially fatal dissemination and vis-
be so readily available in routine diagnostic practice [140]. ceral disease, in both primary varicella and herpes zoster,
PCR is a very sensitive technique that can readily distin- in immunosuppressed patients [147]. VZIG is a high‐titre
guish HSV‐1, HSV‐2 and VZV infections. In addition, preparation of VZV IgG immunoglobulin. VZIG
PCR tests have yielded positive results from the oral should  be strongly considered in immunocompromised
­cavity even after lesions have apparently resolved. Other patients or non-immune pregnant women with a known
620 Section 9  Viral Skin Infections and Opportunistic Infections

exposure to VZV. The earlier it can be given the better, but given 2 months apart by intramuscular injection. It has
it can be given up to 96 hours post exposure. Other indi- been shown to reduce the risk of herpes zoster by 97% in
cations for VZIG include infants born to mothers at risk of those aged 50 years and older, and by 91% in those aged
neonatal varicella, adult patients, and infants born before 70 years and older [154,155]. Another study has shown
28 weeks and exposed to chickenpox in the first 3 months that the gE‐specific cellular and humoral immunogenic-
of gestation. The therapeutic effect of VZIG lasts approxi- ity persists for at least 6 years after two doses of the
mately 3–4 weeks [148,149]. ­vaccine [156]. The vaccine also holds great promise for
Treatment of herpes zoster includes the above sympto- vaccinating immunocompromised patients as in these
matic measures in addition to specific antiviral therapy. In patients live vaccines are contraindicated [157,158]. A
clinical trials, systemic therapy decreases the duration of study looking at the long‐term effectiveness of the vari-
the disease and reduces the risk of post‐herpetic neuralgia cella vaccine showed that it reduced the incidence of
if given within 72 hours of onset [150]. As for chickenpox, varicella by up to 10‐fold and showed no significant loss
intravenous aciclovir therapy should be instituted as soon of efficacy over time [159].
as possible in severely immunosuppressed patients to
prevent dissemination and life‐threatening disease.
References
1 Zerboni L, Sen N, Oliver SL, Arvin AM. Molecular mechanisms of
Vaccine varicella zoster virus pathogenesis. Nat Rev Microbiol 2014;12:
A live attenuated varicella vaccine (vOka) was first 197–210.
2 Quinlivan M, Hawrami K, Barrett‐Muir W et al. The molecular epide-
developed by Takahashi et  al. and tested during the miology of varicella‐zoster virus: evidence for geographic segregation.
1970s [151]. The vaccine was produced by serially pas- J Infect Dis 2002;186:888–94.
saging a wild‐type clinical VZV isolate (pOka) in guinea 3 Barrett‐Muir W, Scott FT, Aaby P et al. Genetic variation of varicella‐
zoster virus: evidence for geographical separation of strains. J Med
pig embryo fibroblasts and expansion for vaccine pro-
Virol 2003;70(suppl 1):S42–7.
duction in WI38 cells. Both Merck and GlaxoSmithKline 4 Loparev VN, Gonzalez A, Deleon‐Carnes M et al. Global identifica-
have since produced their own forms of the VZV vaccine, tion of three major genotypes of varicella‐zoster virus: longitudinal
called Varivax and Varilrix, respectively, both of which clustering and strategies for genotyping. J Virol 2004;78:8349–58.
5 Depledge DP, Brown J, Macanovic J et  al. Viral genome sequencing
were derived from the Oka strain. The varicella vaccine proves nosocomial transmission of fatal varicella. J Infect Dis 2016;214:
was first licensed in 1995 in the USA for immunization of 1399–1402.
all children aged 12–18 months. In the UK, the vaccine is 6 Depledge DP, Gray ER, Kundu S et al. Evolution of cocirculating vari-
not recommended for routine use in children. However, cella‐zoster virus genotypes during a chickenpox outbreak in Guinea‐
Bissau. J Virol 2014;88:13936–46.
it can be given to healthy seronegative children over 7 Depledge DP, Kundu S, Jensen NJ et al. Deep sequencing of viral
1 year of age who come into close contact with individuals genomes provides insight into the evolution and pathogenesis of
at high risk of severe varicella. In addition, the UK varicella zoster virus and its vaccine in humans. Mol Biol Evol
2014;31:397–409.
Department of Health recommends that all seronegative 8 Norberg P, Depledge DP, Kundu S et  al. Recombination of globally
healthcare workers who come into direct contact with circulating varicella‐zoster virus. J Virol 2015;89:7133–46.
patients are vaccinated. The vaccine reduces the rate of 9 Breuer J. VZV molecular epidemiology. Curr Top Microbiol
herpes zoster and the risk of subsequent post‐herpetic Immunol 2010;342:15–42.
10 Fornaro P, Gandini F, Marin M et al. Epidemiology and cost analysis
neuralgia by approximately 51% and 66% respectively of varicella in Italy: results of a sentinel study in the pediatric practice.
[152]. As a result, the high‐potency vaccine, Zostavax, is Italian Sentinel Group on Pediatric Infectious Diseases. Pediatr Infect
also recommended for the prevention of herpes zoster in Dis J 1999;18:414–19.
SECTION 9: VIRAL SKIN

11 Fairley CK, Miller E. Varicella‐zoster virus epidemiology—a changing


70‐ to 79‐year‐olds within the UK. The arguments against
scene? J Infect Dis 1996;174(suppl 3):S314–19.
introducing an infant immunization programme in the 12 Joseph CA, Noah ND. Epidemiology of chickenpox in England and
UK include the theoretical risk of an increased incidence Wales, 1967‐85. Br Med J Clin Res Ed 1988;296:673–6.
INFECTIONS

of zoster and the limited cost‐benefits. What is interest- 13 Hope‐Simpson RE. Herpes zoster in the elderly. Geriatrics
1967;22:151–9.
ing about the live attenuated vaccine strain (vOka) com- 14 Guess HA, Broughton DD, Melton LJ 3rd, Kurland LT. Population‐
pared to its parental wild‐type derivative (pOka) is its based studies of varicella complications. Pediatrics 1986;78:723–7.
reduced replication in differentiated skin. This has been 15 Kudesia G, Partridge S, Farrington CP, Soltanpoor N. Changes in age
shown by experiments in the SCID‐hu mouse, which related seroprevalence of antibody to varicella zoster virus: impact on
vaccine strategy. J Clin Pathol 2002;55:154–5.
show lower titres of infectious virus in human skin grafts 16 Wharton M. The epidemiology of varicella‐zoster virus infections.
compared to its parental strain. However, infectious Infect Dis Clin North Am 1996;10:571–81.
titres in fibroblast monolayers were similar [35]. Given 17 Ouwens MJ, Littlewood KJ, Sauboin C et al. The impact of 2‐dose rou-
tine measles, mumps, rubella, and varicella vaccination in France on
that there are genetic differences between the two strains, the epidemiology of varicella and zoster using a dynamic model with
vOka can be used as a tool to study VZV replication in an empirical contact matrix. Clin Ther 2015;37:816–29.
three‐dimensional skin equivalents such as organotypic 18 Sinha DP. Chickenpox—a disease predominantly affecting adults in
raft cultures. rural West Bengal, India. Int J Epidemiol 1976;5:367–74.
19 Balraj V, John TJ. An epidemic of varicella in rural southern India.
A relatively new adjuvanted subunit recombinant gly- J Trop Med Hyg 1994;97:113–16.
coprotein E vaccine (HZ/su) is now available for the pre- 20 Hope‐Simpson RE. The nature of herpes zoster: a long‐term study
vention of herpes zoster and its complications, and a new hypothesis. Proc R Soc Med 1965;58:9–20.
21 Donahue JG, Choo PW, Manson JE, Platt R. The incidence of herpes
particularly post‐herpetic neuralgia. The vaccine is well
zoster. Arch Intern Med 1995;155:1605–9.
tolerated and more immunogenic than the live attenu- 22 Galil K, Choo PW, Donahue JG, Platt R. The sequelae of herpes zoster.
ated vOka VZV vaccines [153]. The HZ/su vaccine is Arch Intern Med 1997;157:1209–13.
Chapter 51  Varicella Zoster Virus Infections 621

23 Hope‐Simpson RE. Postherpetic neuralgia. J R Coll Gen Pract recombinant viruses in skin xenografts in the SCIDhu mouse model.
1975;25:571–5. Virology 2005;332:337–46.
24 Guess HA, Broughton DD, Melton LJ 3rd, Kurland LT. Epidemiology 48 Arvin AM. Investigations of the pathogenesis of varicella zoster
of herpes zoster in children and adolescents: a population‐based virus infection in the SCIDhu mouse model. Herpes 2006;13:75–80.
study. Pediatrics 1985;76:512–17. 49 Tomlinson TH. Giant cell formation in the tonsils in the prodromal
25 Rusthoven JJ. The risk of varicella‐zoster infections in different patient stage of chickenpox. Am J Pathol 1939;15:523–6.
populations: a critical review. Transfus Med Rev 1994;8:96–116. 50 Gershon AA, Steinberg S, Brunell PA. Zoster immune globulin. A fur-
26 Tyndall MW, Nasio J, Agoki E et al. Herpes zoster as the initial pres- ther assessment. N Engl J Med 1974;290:243–5.
entation of human immunodeficiency virus type 1 infection in Kenya. 51 Ku CC, Padilla JA, Grose C et al. Tropism of varicella‐zoster virus for
Clin Infect Dis 1995;21:1035–7. human tonsillar CD4(+) T lymphocytes that express activation, mem-
27 Veenstra J, Krol A, van Praag RM et al. Herpes zoster, immunological ory, and skin homing markers. J Virol 2002;76:11425–33.
deterioration and disease progression in HIV‐1 infection. AIDS 52 Harson R, Grose C. Egress of varicella‐zoster virus from the mela-
1995;9:1153–8. noma cell: a tropism for the melanocyte. J Virol. 1995;69:4994–5010.
28 Poletti P, Melegaro A, Ajelli M et  al. Perspectives on the impact of 53 Zhu Z, Gershon MD, Ambron R et al. Infection of cells by varicella
varicella immunization on herpes zoster. A model‐based evaluation zoster virus: inhibition of viral entry by mannose 6‐phosphate and
from three European countries. PLoS One 2013;8:e60732. heparin. Proc Natl Acad Sci U S A 1995;92:3546–50.
29 Fenner F. The pathogenesis of the acute exanthems; an interpretation 54 Chen JJ, Zhu Z, Gershon AA, Gershon MD. Mannose 6‐phosphate
based on experimental investigations with mousepox; infectious receptor dependence of varicella zoster virus infection in vitro and in
ectromelia of mice. Lancet 1948;2:915–20. the epidermis during varicella and zoster. Cell 2004;119:915–26.
30 Grose C. Variation on a theme by Fenner: the pathogenesis of chicken- 55 Li Q, Ali MA, Cohen JI. Insulin degrading enzyme is a cellular recep-
pox. Pediatrics 1981;68:735–7. tor mediating varicella‐zoster virus infection and cell‐to‐cell spread.
31 Ku CC, Zerboni L, Ito H et al. Varicella‐zoster virus transfer to skin Cell 2006;127:305–16.
by T cells and modulation of viral replication by epidermal cell 56 Cole NL, Grose C. Membrane fusion mediated by herpesvirus glyco-
interferon‐alpha. J Exp Med 2004;200:917–25. proteins: the paradigm of varicella‐zoster virus. Rev Med Virol
32 Sen N, Che X, Rajamani J et al. Signal transducer and activator of tran- 2003;13:207–22.
scription 3 (STAT3) and survivin induction by varicella‐zoster virus 57 Gershon MD, Gershon AA. VZV infection of keratinocytes: produc-
promote replication and skin pathogenesis. Proc Natl Acad Sci U S A tion of cell‐free infectious virions in vivo. Curr Top Microbiol
2012;109:600–5. Immunol 2010;342:173–88.
33 Sato B, Ito H, Hinchliffe S et al. Mutational analysis of open reading 58 Cradock‐Watson JE, Ridehalgh MK, Bourne MS. Specific immuno-
frames 62 and 71, encoding the varicella‐zoster virus immediate‐early globulin responses after varicella and herpes zoster. J Hyg (Lond)
transactivating protein, IE62, and effects on replication in vitro and 1979;82:319–36.
in  skin xenografts in the SCID‐hu mouse in vivo. J Virol 2003; 59 Cohen JI. Infection of cells with varicella‐zoster virus down‐regulates
77:5607–20. surface expression of class I major histocompatibility complex anti-
34 Oliver SL, Sommer M, Zerboni L et al. Mutagenesis of varicella‐zoster gens. J Infect Dis 1998;177:1390–3.
virus glycoprotein B: putative fusion loop residues are essential for 60 Abendroth A, Lin I, Slobedman B et al. Varicella‐zoster virus retains
viral replication, and the furin cleavage motif contributes to patho- major histocompatibility complex class I proteins in the Golgi com-
genesis in skin tissue in vivo. J Virol 2009;83:7495–506. partment of infected cells. J Virol 2001;75:4878–88.
35 Moffat JF, Zerboni L, Kinchington PR et al. Attenuation of the vaccine 61 Abendroth A, Slobedman B, Lee E et al. Modulation of major histo-
Oka strain of varicella‐zoster virus and role of glycoprotein C in compatibility class II protein expression by varicella‐zoster virus.
alphaherpesvirus virulence demonstrated in the SCID‐hu mouse. J Virol 2000;74:1900–7.
J Virol 1998;72:965–74. 62 Abendroth A, Arvin A. Immune evasion mechanisms of varicella‐
36 Besser J, Ikoma M, Fabel K et  al. Differential requirement for cell zoster virus. Arch Virol Suppl 2001;(17):99–107.
fusion and virion formation in the pathogenesis of varicella‐zoster 63 Eisfeld AJ, Yee MB, Erazo A et  al. Downregulation of class I major
virus infection in skin and T cells. J Virol 2004;78:13293–305. histocompatibility complex surface expression by varicella‐zoster
37 Wang L, Sommer M, Rajamani J, Arvin AM. Regulation of the ORF61 virus involves open reading frame 66 protein kinase‐dependent
promoter and ORF61 functions in varicella‐zoster virus replication and ‐independent mechanisms. J Virol 2007;81:9034–49.
and pathogenesis. J Virol 2009;83:7560–72. 64 Tyzzer EE. The histology of the skin lesions in varicella. Philippine
38 Jones M, Dry IR, Frampton D et al. RNA‐seq analysis of host and J Sci 1906;1:349–72.
viral gene expression highlights interaction between varicella zoster 65 Croen KD, Ostrove JM, Dragovic LJ, Straus SE. Patterns of gene
virus and keratinocyte differentiation. PLoS Pathog 2014;10:e1003896. expression and sites of latency in human nerve ganglia are different

SECTION 9: VIRAL SKIN


39 Che X, Zerboni L, Sommer MH, Arvin AM. Varicella‐zoster virus for varicella‐zoster and herpes simplex viruses. Proc Natl Acad Sci U
open reading frame 10 is a virulence determinant in skin cells but not S A 1988;85:9773–7.
in T cells in vivo. J Virol 2006;80:3238–48. 66 Straus SE, Oxman MN. Varicella and herpes zoster. In: Freedberg JM,

INFECTIONS
40 Wang L, Oliver SL, Sommer M et al. Disruption of PML nuclear bodies Eisen AZ, Wolff K et  al. (eds) Fitzpatrick’s Dermatology in General
is mediated by ORF61 SUMO‐interacting motifs and required for vari- Medicine, 5th edn. New York: McGraw Hill; 1999:2427–50.
cella‐zoster virus pathogenesis in skin. PLoS Pathog 2011;7:e1002157. 67 Kennedy PG, Rovnak J, Badani H, Cohrs RJ. A comparison of herpes
41 Mo C, Lee J, Sommer M et al. The requirement of varicella zoster virus simplex virus type 1 and varicella‐zoster virus latency and reactiva-
glycoprotein E (gE) for viral replication and effects of glycoprotein tion. J Gen Virol 2015;96:1581–602.
I on gE in melanoma cells. Virology 2002;304:176–86. 68 Mahalingam R, Wellish M, Cohrs R et al. Expression of protein encoded
42 Berarducci B, Ikoma M, Stamatis S et  al. Essential functions of the by varicella‐zoster virus open reading frame 63 in latently infected
unique N‐terminal region of the varicella‐zoster virus glycoprotein E human ganglionic neurons. Proc Natl Acad Sci U S A 1996;93:2122–4.
ectodomain in viral replication and in the pathogenesis of skin infec- 69 Kennedy PG, Grinfeld E, Bell JE. Varicella‐zoster virus gene expres-
tion. J Virol 2006;80:9481–96. sion in latently infected and explanted human ganglia. J Virol
43 Moffat J, Ito H, Sommer M et  al. Glycoprotein I of varicella‐zoster 2000;74:11893–8.
virus is required for viral replication in skin and T cells. J Virol 70 Zerboni L, Sobel RA, Ramachandran V et al. Expression of varicella‐
2002;76:8468–71. zoster virus immediate‐early regulatory protein IE63 in neurons of
44 Maresova L, Kutinova L, Ludvikova V et al. Characterization of inter- latently infected human sensory ganglia. J Virol 2010;84:3421–30.
action of gH and gL glycoproteins of varicella‐zoster virus: their pro- 71 Hood C, Cunningham AL, Slobedman B et al. Varicella‐zoster virus
cessing and trafficking. J Gen Virol 2000;81:1545–52. ORF63 inhibits apoptosis of primary human neurons. J Virol 2006;
45 Zerboni L, Ku CC, Jones CD et  al. Varicella‐zoster virus infection 80:1025–31.
of  human dorsal root ganglia in vivo. Proc Natl Acad Sci U S A 72 Ambagala AP, Cohen JI. Varicella‐zoster virus IE63, a major viral
2005;102:6490–5. latency protein, is required to inhibit the alpha interferon‐induced
46 Zerboni L, Arvin A. Neuronal subtype and satellite cell tropism are antiviral response. J Virol 2007;81:7844–51.
determinants of varicella‐zoster virus virulence in human dorsal root 73 Zerboni L, Sobel RA, Lai M et  al. Apparent expression of varicella‐
ganglia xenografts in vivo. PLoS Pathog 2015;11:e1004989. zoster virus proteins in latency resulting from reactivity of murine
47 Zerboni L, Hinchliffe S, Sommer MH et al. Analysis of varicella zoster and rabbit antibodies with human blood group a determinants in sen-
virus attenuation by evaluation of chimeric parent Oka/vaccine Oka sory neurons. J Virol 2012;86:578–83.
622 Section 9  Viral Skin Infections and Opportunistic Infections

74 Ouwendijk WJ, Flowerdew SE, Wick D et al. Immunohistochemical 101 Thomson JJ, Retter A, Hunt BJ. Novel management of post varicella
detection of intra‐neuronal VZV proteins in snap‐frozen human gan- purpura fulminans owing to severe acquired protein S deficiency.
glia is confounded by antibodies directed against blood group Blood Coagul Fibrinolysis 2010;21:598–600.
A1‐associated antigens. J Neurovirol 2012;18:172–80. 102 Januario G, Ramroop S, Shingadia DV, Novelli V. Postinfectious pur-
75 Ouwendijk WJ, Choe A, Nagel MA et al. Restricted varicella‐zoster pura fulminans secondary to varicella‐induced protein S deficiency.
virus transcription in human trigeminal ganglia obtained soon after Pediatr Infect Dis J 2010;29:981–3.
death. J Virol 2012;86:10203–6. 103 Nguyen P, Reynaud J, Pouzol P et al. Varicella and thrombotic com-
76 Sadaoka T, Depledge DP, Rajbhandari L et  al. In vitro system plications associated with transient protein C and protein S deficien-
using human neurons demonstrates that varicella‐zoster vaccine cies in children. Eur J Pediatr 1994;153:646–9.
virus is impaired for reactivation, but not latency. Proc Natl Acad 104 Thomas SL, Minassian C, Ganesan V et al. Chickenpox and risk of
Sci U S A 2016;113:E2403–12. stroke: a self‐controlled case series analysis. Clin Infect Dis
77 Takahashi M, Otsuka T, Okuno Y et al. Live vaccine used to prevent 2014;58:61–8.
the spread of varicella in children in hospital. Lancet 1974;2: 105 Elkind MS, Hills NK, Glaser CA et al. Herpesvirus infections and
1288–90. childhood arterial ischemic stroke: results of the VIPS Study.
78 Takahashi M. Development and characterization of a live varicella Circulation 2016;133:732–41.
vaccine (Oka strain). Biken J 1984;27:31–6. 106 Lanthier S, Armstrong D, Domi T, deVeber G. Post‐varicella arterio-
79 Gomi Y, Imagawa T, Takahashi M, Yamanishi K. Oka varicella vac- pathy of childhood: natural history of vascular stenosis. Neurology
cine is distinguishable from its parental virus in DNA sequence of 2005;64:660–3.
open reading frame 62 and its transactivation activity. J Med Virol 107 Nagel MA, Gilden D. Developments in varicella zoster virus vascu-
2000;61:497–503. lopathy. Curr Neurol Neurosci Rep 2016;16:12.
80 Gomi Y, Sunamachi H, Mori Y et  al. Comparison of the complete 108 Amlie‐Lefond C, Gilden D. Varicella zoster virus: a common cause of
DNA sequences of the Oka varicella vaccine and its parental virus. stroke in children and adults. J Stroke Cerebrovasc Dis 2016;25:1561–9.
J Virol 2002;76:11447–59. 109 Science M, MacGregor D Richardson SE et al. Central nervous system
81 Quinlivan M, Breuer J. Clinical and molecular aspects of the live complications of varicella‐zoster virus. J Pediatr 2014;165:779–85.
attenuated Oka varicella vaccine. Rev Med Virol 2014;24:254–73. 110 Enders G, Miller E, Cradock‐Watson J et  al. Consequences of vari-
82 Depledge DP, Yamanishi K, Gomi Y et al. Deep sequencing of dis- cella and herpes zoster in pregnancy: prospective study of 1739
tinct preparations of the live attenuated varicella‐zoster virus vac- cases. Lancet 1994;343:1548–51.
cine reveals a conserved core of attenuating single‐nucleotide 111 Cuthbertson G, Weiner CP, Giller RH, Grose C. Prenatal diagnosis of
polymorphisms. J Virol 2016;90:8698–704. second‐trimester congenital varicella syndrome by virus‐specific
83 Gilden DH, Wright RR, Schneck SA et al. Zoster sine herpete, a clini- immunoglobulin M. J Pediatr 1987;111:592–5.
cal variant. Ann Neurol 1994;35:530–3. 112 Hartung J, Enders G, Chaoui R et al. Prenatal diagnosis of congenital
84 Vukmanovic‐Stejic M, Sandhu D, Seidel JA et  al. The characteriza- varicella syndrome and detection of varicella‐zoster virus in the
tion of varicella zoster virus‐specific T cells in skin and blood during fetus: a case report. Prenat Diagn 1999;19:163–6.
aging. J Invest Dermatol 2015;135:1752–62. 113 Mandelbrot L. Fetal varicella  –  diagnosis, management, and out-
85 Nikkels AF, Pierard GE. Hair follicle involvement in herpes zoster. come. Prenat Diagn 2012;32:511–18.
Virchows Arch 1997;430:510–1. 114 Miller E, Cradock‐Watson JE, Ridehalgh MK. Outcome in newborn
86 Esiri MM, Tomlinson AH. Herpes zoster. Demonstration of virus in babies given anti‐varicella‐zoster immunoglobulin after perinatal
trigeminal nerve and ganglion by immunofluorescence and electron maternal infection with varicella‐zoster virus. Lancet 1989;2:371–3.
microscopy. J Neurol Sci 1972;15:35–48. 115 Smith CK, Arvin AM. Varicella in the fetus and newborn. Semin
87 Gilden DH, Beinlich BR, Rubinstien EM et al. Varicella‐zoster virus Fetal Neonatal Med 2009;14:209–17.
myelitis: an expanding spectrum. Neurology 1994;44:1818–23. 116 Ganga Devi NP, Rathinam SN, Ramachandran R, Swaminathan S.
88 Hogan EL, Krigman MR. Herpes zoster myelitis. Evidence for viral Recurrent herpes zoster in early childhood. Indian J Pediatr
invasion of spinal cord. Arch Neurol 1973;29:309–13. 2007;74:774–6.
89 Liesegang TJ. Varicella zoster viral disease. Mayo Clin Proc 117 Wen SY, Liu WL. Epidemiology of pediatric herpes zoster after vari-
1999;74:983–98. cella infection: a population based study. Pediatrics 2015;135:e565–71.
90 McCrary ML, Severson J, Tyring SK. Varicella zoster virus. J Am 118 Weinmann S, Chun C, Schmid DS et  al. Incidence and clinical
Acad Dermatol 1999;41:1–14; quiz 5–6. characteristics of herpes zoster among children in the varicella
91 Peterson CL, Vugia DJ, Meyers HB et  al. Risk factors for invasive vaccine era, 2005‐2009. J Infect Dis 2013;208:1859–68.
group A streptococcal infections in children with varicella: a case‐ 119 Leung AK, Robson WL, Leong AG. Herpes zoster in childhood.
control study. Pediatr Infect Dis J 1996;15:151–6. J Pediatr Health Care 2006;20:300–3.
SECTION 9: VIRAL SKIN

92 Vugia DJ, Peterson CL, Meyers HB et al. Invasive group A strepto- 120 Ferreira JC, Marques HH, Ferriani MP et al. Herpes zoster infection
coccal infections in children with varicella in Southern California. in childhood‐onset systemic lupus erythematosus patients: a large
Pediatr Infect Dis J 1996;15:146–50. multicenter study. Lupus 2016;25:754–9.
INFECTIONS

93 Shirley R, Mackey S, Meagher P. Necrotising fasciitis: a sequelae of 121 Feder HM Jr, Hoss DM. Herpes zoster in otherwise healthy children.
varicella zoster infection. J Plast Reconstr Aesthet Surg 2011; Pediatr Infect Dis J 2004;23:451–7; quiz 8–60.
64:123–7. 122 Wareham DW, Breuer J. Herpes zoster. BMJ 2007;334:1211–15.
94 Peters AC, Versteeg J, Lindeman J, Bots GT. Varicella and acute cer- 123 Liesegang TJ. The varicella‐zoster virus: systemic and ocular fea-
ebellar ataxia. Arch Neurol 1978;35:769–71. tures. J Am Acad Dermatol 1984;11:165–91.
95 Gilden D, Nagel M, Cohrs R et  al. Varicella zoster virus in the 124 Vermont CL, Jol‐van der Zijde EC, Hissink Muller P et al. Varicella
nervous system. F1000Res 2015;4. zoster reactivation after hematopoietic stem cell transplant in chil-
96 Zampogna JC, Flowers FP. Persistent verrucous varicella as the dren is strongly correlated with leukemia treatment and suppression
­initial manifestation of HIV infection. J Am Acad Dermatol 2001; of host T‐lymphocyte immunity. Transpl Infect Dis 2014;16:188–94.
44(2 Suppl):391–4. 125 Kunz AN, Rajnik M. Disseminated cutaneous varicella zoster virus
97 Rodriguez‐Moreno A, Sanchez‐Fructuoso AI, Calvo N et al. Varicella infections during infliximab therapy for Crohn’s disease: case report
infection in adult renal allograft recipients: experience at one center. of two pediatric patients at one institution. Clin Pediatr (Phila)
Transplant Proc 2006;38:2416–8. 2011;50:559–61.
98 Banovic T, Yanilla M, Simmons R et al. Disseminated varicella infec- 126 LeBoit PE, Limova M, Yen TS et al. Chronic verrucous varicella‐zoster
tion caused by varicella vaccine strain in a child with low invariant virus infection in patients with the acquired immunodeficiency syn-
natural killer T cells and diminished CD1d expression. J Infect Dis drome (AIDS). Histologic and molecular biologic findings. Am J
2011;204:1893–901. Dermatopathol 1992;14:1–7.
99 McKolanis JR, Ragab AH, Schmid DS. In vitro enhancement of natu- 127 Nikkels AF, Snoeck R, Rentier B, Pierard GE. Chronic verrucous vari-
ral killer cell activity against herpesvirus‐infected targets in patients cella zoster virus skin lesions: clinical, histological, molecular and
with acute lymphocytic leukemia. Arch Virol 1991;117:17–28. therapeutic aspects. Clin Exp Dermatol 1999;24:346–53.
100 Busuttil DP, Hay CR, Lewis MA, Wynn RF. Aggressive multiple 128 Lehloenya R, Meintjes G. Dermatologic manifestations of the
modality therapy for varicella‐associated purpura fulminans. Br J immune reconstitution inflammatory syndrome. Dermatol Clin
Haematol 2000;110:1012–13. 2006;24:549–70.
Chapter 51  Varicella Zoster Virus Infections 623

129 Requena L, Kutzner H, Escalonilla P et  al. Cutaneous reactions at 145 Balfour HH Jr, Rotbart HA, Feldman S et al. Acyclovir treatment of
sites of herpes zoster scars: an expanded spectrum. Br J Dermatol varicella in otherwise healthy adolescents. The Collaborative
1998;138:161–8. Acyclovir Varicella Study Group. J Pediatr 1992;120:627–33.
130 Birlea M, Arendt G, Orhan E et al. Subclinical reactivation of varicella 146 Balfour HH Jr, Edelman CK, Anderson RS et al. Controlled trial of
zoster virus in all stages of HIV infection. J Neurol Sci 2011;304:22–4. acyclovir for chickenpox evaluating time of initiation and duration
131 Minassian C, Thomas SL, Smeeth L et  al. Acute cardiovascular of therapy and viral resistance. Pediatr Infect Dis J 2001;20:919–26.
events after herpes zoster: a self‐controlled case series analysis in 147 Balfour HH Jr, McMonigal KA, Bean B. Acyclovir therapy of vari-
vaccinated and unvaccinated older residents of the United States. cella‐zoster virus infections in immunocompromised patients.
PLoS Med 2015;12:e1001919. J Antimicrob Chemother 1983;12(suppl B):169–79.
132 Breuer J, Pacou M, Gautier A, Brown MM. Herpes zoster as a risk 148 Varicella‐zoster immune globulin for the prevention of chickenpox.
factor for stroke and TIA: a retrospective cohort study in the UK. Recommendations of the Immunization Practices Advisory Committee,
Neurology 2014;83:e27–33. Centers for Disease Control. Ann Intern Med 1984;100:859–65.
133 Weidmann M, Meyer‐Konig U, Hufert FT. Rapid detection of herpes 149 Macmahon E. Policy change on VZIG in the US to match UK recom-
simplex virus and varicella‐zoster virus infections by real‐time PCR. mendations. BMJ 2012;344:e3207.
J Clin Microbiol 2003;41:1565–8. 150 Bruxelle J, Pinchinat S. Effectiveness of antiviral treatment on
134 McSorley J, Shapiro L, Brownstein MH, Hsu KC. Herpes simplex acute phase of herpes zoster and development of post herpetic
and varicella‐zoster: comparative histopathology of 77 cases. Int neuralgia: review of international publications. Med Mal Infect
J Dermatol 1974;13:69–75. 2012;42:53–8.
135 Boer A, Herder N, Blodorn‐Schlicht N, Falk T. Herpes incognito 151 Takahashi M. Current status and prospects of live varicella vaccine.
most commonly is herpes zoster and its histopathologic pattern is Vaccine 1992;1014:1007–14.
distinctive! Am J Dermatopathol 2006;28:181–6. 152 Keating GM. Shingles (herpes zoster) vaccine (Zostavax((R))): a
136 Walsh N, Boutilier R, Glasgow D, Shaffelburg M. Exclusive involve- review in the prevention of herpes zoster and postherpetic neural-
ment of folliculosebaceous units by herpes: a reflection of early gia. BioDrugs 2016;30:243–54.
­herpes zoster. Am J Dermatopathol 2005;27:189–94. 153 Leroux‐Roels I, Leroux‐Roels G, Clement F et al. A phase 1/2 clinical
137 Muraki R, Iwasaki T, Sata T et al. Hair follicle involvement in ­herpes trial evaluating safety and immunogenicity of a varicella zoster gly-
zoster: pathway of viral spread from ganglia to skin. Virchows Arch coprotein e subunit vaccine candidate in young and older adults.
1996;428:275–80. J Infect Dis 2012;206:1280–90.
138 Uhoda I, Pierard‐Franchimont C, Pierard GE. Varicella‐zoster virus 154 Cunningham AL, Lal H, Kovac M et al. Efficacy of the herpes zos-
vasculitis: a case of recurrent varicella without epidermal involve- ter subunit vaccine in adults 70 years of age or older. N Engl J Med
ment. Dermatology 2000;200:173–5. 2016;375:1019–32.
139 Alonso‐Perez A, Fraga J, Delgado Y et al. Nodular herpes zoster with 155 Lal H, Cunningham AL, Godeaux O et al. Efficacy of an adjuvanted
herpetic syringitis and no epidermal involvement in a patient with herpes zoster subunit vaccine in older adults. N Engl J Med
Burkitt lymphoma. Am J Dermatopathol 2006;28:194–6. 2015;372:2087–96.
140 Sadick NS, Swenson PD, Kaufman RL, Kaplan MH. Comparison of 156 Chlibek R, Pauksens K, Rombo L et al. Long‐term immunogenicity
detection of varicella‐zoster virus by the Tzanck smear, direct immu- and safety of an investigational herpes zoster subunit vaccine in
nofluorescence with a monoclonal antibody, and virus isolation. older adults. Vaccine 2016;34:863–8.
J Am Acad Dermatol 1987;17:64–9. 157 Stadtmauer EA, Sullivan KM, Marty FM et al. A phase 1/2 study
141 Rubben A, Baron JM, Grussendorf‐Conen EI. Routine detection of of an adjuvanted varicella‐zoster virus subunit vaccine in autolo-
herpes simplex virus and varicella zoster virus by polymerase chain gous hematopoietic cell transplant recipients. Blood 2014;124:
reaction reveals that initial herpes zoster is frequently misdiagnosed 2921–9.
as herpes simplex. Br J Dermatol 1997;137:259–61. 158 Berkowitz EM, Moyle G, Stellbrink HJ et  al. Safety and immuno-
142 De SK, Hart JC, Breuer J. Herpes simplex virus and varicella zoster genicity of an adjuvanted herpes zoster subunit candidate vaccine in
virus: recent advances in therapy. Curr Opin Infect Dis 2015;28:589–95. HIV‐infected adults: a phase 1/2a randomized, placebo‐controlled
143 Naesens L, De Clercq E. Recent developments in herpesvirus ther- study. J Infect Dis 2015;211:1279–87.
apy. Herpes 2000;8:12–16. 159 Baxter R, Ray P, Tran TN et al. Long‐term effectiveness of varicella
144 Dunkle LM, Arvin AM, Whitley RJ et al. A controlled trial of acyclo- vaccine: a 14‐year, prospective cohort study. Pediatrics 2013;131:
vir for chickenpox in normal children. N Engl J Med 1991;325: e1389–96.
1539–44.

SECTION 9: VIRAL SKIN


INFECTIONS
624 

CHA PTER  5 2

Poxvirus Infections
Susan Lewis‐Jones1 & Jane C. Sterling2
Ninewells Hospital & Medical School, Dundee, UK
1 

Department of Dermatology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke’s Hospital, Cambridge, UK
2 

Introduction, 624 Parapoxvirus infection, 643


Orthopoxvirus infection, 626 Yatapoxvirus infection, 647

Abstract with an infected animal, the lesion(s) develop at the site(s) of c­ ontact
and may be associated with systemic features. Over the course of a
few days, the initial macule evolves to a papule, a vesicle and then an
Poxvirus infections are most commonly acquired from animals
ulcer, which will heal spontaneously over days to weeks. The geo-
and are therefore relatively uncommon and usually occupational
graphical distribution depends on the reservoir hosts of the virus.
­hazards, although childhood infections often occur. After contact

Key points • Recovery and healing occur without specific treatment, but
supportive therapy may be necessary in severe cases, although
fatality is very rare.
• Orthopoxvirus and parapoxvirus infections in humans are
• Smallpox has been eradicated worldwide and routine vaccination
zoonotic.
with vaccinia virus is no longer performed.
• Lesions develop as papules, which quickly progress to vesicles
and then ulcers.

infections are caused by the viruses of sheep/goatpox


Introduction (capripoxvirus), swinepox (suipoxvirus), myxomatosis
Poxviruses of vertebrates are classified into two subfami- (leporipoxvirus), crocodile pox (crocodylidpoxvirus) or
lies: Chordopoxvirinae and Entomopoxvirinae. All human avianpox (avipoxvirus).
poxvirus infections are caused by viruses in genera within Poxviruses are large complex DNA viruses that
the Chordopoxvirinae and, although closely related, have ­replicate in the cytoplasm of infected cells using virus‐
distinct genetic and antigenic features [1]. The causative associated, DNA‐dependent polymerase and are charac-
agents of molluscum contagiosum (see Chapter 48) and terized by the production of skin lesions. All poxviruses
smallpox are the only poxviruses exclusive to humans. encode cytokine and cytokine receptor analogues that
Smallpox was eradicated by 1980 and therefore, apart allow them to modulate the immune response and
from molluscum contagiosum, naturally occurring ­perpetuate their existence. They are not only cytopathic,
SECTION 9: VIRAL SKIN

human poxvirus infections are acquired solely from causing host tissue damage, but also encode an array of
­animal reservoirs [2]. immunomodulatory molecules that affect the severity
INFECTIONS

The viruses concerned belong to three genera, and of ­disease [3]. They are generally brick shaped or ovoid,
knowledge of the geographical distribution of the viruses varying in size from 240 × 300 nm (orthopoxvirus) to
and the potential animal sources is of considerable value 160 × 250 nm (parapoxvirus) [1,4].
in differential clinical diagnosis [2]. Poxvirus zoonoses Electron microscopy can be of considerable value in
range worldwide from trivial occupational hazards rapid diagnosis [5,6] (Fig. 52.1). Their size and the pres-
caused by parapoxviruses, which are less likely to cause ence of conspicuous surface tubules enable poxviruses to
paediatric infections, to rare and geographically be differentiated from herpesviruses. Furthermore, the
restricted infections such as monkeypox, which causes characteristic long, spirally wound, surface tubule of the
considerable mortality in children (Table 52.1). There are parapoxviruses enables them to be differentiated from
occasional reports of zoonotic parapoxvirus infections orthopoxviruses and tanapoxviruses. The epidemiologi-
in those working with New Zealand red deer and seals, cal history, clinical features and electron microscopy
but there is at present no good evidence that human will in many instances indicate a certain diagnosis but if

Harper’s Textbook of Pediatric Dermatology, Fourth Edition. Edited by Peter Hoeger, Veronica Kinsler and Albert Yan.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.
Table 52.1  Poxviruses causing human disease

Genus Virus (species) Distribution Sources of human Comment


infection (less
important sources
in parentheses)

Molluscipoxvirus Molluscum contagiosum Worldwide Human


virus
Orthopoxvirus Camelpox Africa, Middle East, Asia, Camels Occupational hazard, papulovesicular
southern Russia lesions progressing to ulcer, usually on
hands or arms
Cowpox Europe, including some Cats (cows) (rodents [R]) Painful haemorrhagic ulcer; relatively more
states of the former USSR common in young girls
Monkeypox West Africa, Congo basin Squirrels (R) (monkeys) Severe generalized infection; >400 cases
since 1970, c. 90% in children with c.
15% mortality; limited case‐to‐case
spread. USA outbreak from infected
imported rodents
Buffalopox India Buffalo (R?) Occupational hazard, painful skin lesion;
virus is subspecies of vaccinia
Taterapox     Vaccinia infection only after vaccination
      Occupational. Resembles cowpox.
  Not endemic   Araçatuba and Cantagalo viruses
Vaccinia, bovine vaccinia Brazil and South America No reservoir host (subspecies of vaccinia)
Cattle (R?)
Variola (smallpox) Eradicated Not naturally occurring
Parapoxvirus Orf Worldwide Sheep (R), goats (R), Relatively painless granulomatous skin
cattle (R) lesions; occupational hazard of those in
contact with reservoir hosts
Pseudocowpox (milker’s Worldwide Sheep (R), cattle (R) Occupational hazard of those in contact
nodes) with reservoir hosts; clinically similar
to orf
Sealpox Europe and USA (and Seals (R?) Resembles orf
regions frequented by Confirmed by PCR as human zoonosis in
seals) seal handlers
Yatapox Tanapox Congo basin, Kenya Monkeys (R) Nodular skin lesions usually singular, with
pyrexia

R, reservoir hosts.

SECTION 9: VIRAL SKIN


INFECTIONS

(a) (b) (c)


Fig. 52.1  Electron micrographs of (a) cowpoxviruses showing the common ‘mulberry’ (M) form with conspicuous short, randomly arranged surface tubules
and the less common ‘capsule’ (C) form with deeper stain penetration; (b) whole M and C forms of parapoxvirus, showing the characteristic long spiralling
of the surface tubule of the former; (c) whole herpesviruses showing one particle with intact nucleocapsid and one broken, allowing stain penetration
(phosphotungstic acid, ×120 000). Source: Reproduced with permission of Springer Nature.
626 Section 9  Viral Skin Infections and Opportunistic Infections

necessary the viruses may be isolated and identified [3,5]. Orthopoxvirus infection
Rapid identification by orthopoxvirus‐specific polymer-
ase chain reaction (PCR) is quicker and more specific than Smallpox
culture and is of growing availability [7–10]. Specialized
laboratories are available for the diagnosis of smallpox Introduction and  history. Smallpox (variola major) has
should the need arise. altered the course of world history [1,2]. For centuries an
Parapoxviruses are not routinely isolated by the diag- endemic disease throughout the world, it caused millions
nostic laboratory. A differential diagnosis of orf or milk- of deaths and frequently left survivors with damaged
er’s nodules may be suspected based on animal contact health, unsightly pock‐marked skin and blindness. In
if known; if there is no known contact with infected mediaeval times, the overall mortality ranged from 10%
­animals, a diagnosis of orf/milker’s nodules is often to 60%, with an average of about 25–30%. By the twenti-
suspected, based on the clinical appearance. Poxviruses eth century, mortality had fallen to between 5% and 25%,
are resistant to environmental conditions and will sur- although the childhood mortality rate remained as high
vive sufficiently in dried scabs or in vesicle fluid dried as 40% [2,3].
on a microscope slide to enable them to be transported The origins of smallpox are uncertain but from sequence
by post or courier to the laboratory. However, if a dif- analysis it has been suggested that the original animal
ferential laboratory diagnosis is required, virus trans- source was a wild rodent in Africa [4]. During the first
port medium should be used to preserve more labile millennium ad, the disease spread through Asia into
viruses. Zoonotic poxviruses will grow well in human Europe and, around 700 ad, into North Africa [1–3]. The
cell cultures and in cells from their reservoir hosts; Spanish Conquistadors carried it to the New World in the
orthopoxviruses grow well on chick chorioallantois 1500s, where it spread rapidly in the ‘virgin population’.
[5,6]. Preliminary identification is by suitable biological It is estimated that over 3.5 million native South Americans
markers, but increasing use is being made of DNA anal- died in smallpox epidemics. A similar epidemic occurred
ysis, which enables individual strains within a species to in North America when smallpox wiped out millions of
be recognized [11,12]. Reviews of human zoonotic pox- the members of native tribes [1–3,5]. The annual smallpox
virus infections include details of other hosts and poten- mortality rate in Europe in the eighteenth century was
tial therapeutic agents [2,13]. estimated at about 200 000–600 000, making up 7–12% of
all deaths. The majority of these mortalities occurred in
children, especially infants, and the disease accounted for
one third of all childhood deaths [1]. The succession of the
References
1 Esposito JJ, Baxby D, Black DN et al. Poxviridae. In: Murphy FA, European monarchy was considerably altered by small-
Faquet CM, Bishop DHL (eds) Virus Taxonomy: Classification and pox deaths [1].
Nomenclature of Viruses. Vienna: Springer‐Verlag, 1995:79–91. In the early twentieth century, a minor form of smallpox,
2 Essbauer S, Pfeffer M, Meyer H. Zoonotic poxviruses. Vet Microbiol
2010;140:229–36.
alastrim, with a mortality of 1% or less, appeared in Africa
3 Stanford MM, McFadden G, Karupiah G, Chaudhri G. and spread throughout the USA and Europe [3,5–7].
Immunopathogenesis of poxvirus infections: forecasting the impend- Vaccination alone was unsuccessful in eradicating
ing storm. Immunol Cell Biol 2007;85:93–102. smallpox and in 1967, when the estimated world preva-
4 Smith GL. Poxviruses. In: Mahy BWJ, ter Meulen V (eds) Topley and
Wilson’s Microbiology and Microbial Infections, Virology, 10th edn. lence was 10–15 million, the World Health Organization
London: Wiley, 2007:578–93. (WHO) commenced a global programme of identifica-
5 Nakano JH, Esposito JJ. Poxviruses. In: Schmidt NJ, Emmons RW tion, isolation and containment of cases and contacts [2,3].
SECTION 9: VIRAL SKIN

(eds) Diagnostic Procedures for Viral, Rickettsial and Chlamydial


Diseases, 6th edn. Washington, DC: American Public Health
As a result, the last naturally occurring case of smallpox
Association, 1989:224–65. was recorded in Somalia in 1977, although a small UK
INFECTIONS

6 Baxby D, Bennett M, Getty B. Human cowpox 1969–93: a review laboratory outbreak did occur in 1978 [3]. In 1980, the
based on 54 cases. Br J Dermatol 1994;131:598–607. WHO declared the world to be free from smallpox, the
7 Ropp SL, Jin Q, Knight JC et al. PCR strategy for identification and
differentiation of smallpox and other orthopoxviruses. J Clin first and to date the only virus infection to have been suc-
Microbiol 1995;33:2069–76. cessfully eradicated [2,3]. After this, all stocks of smallpox
8 Naidoo J, Baxby D, Bennett M et al. Characterisation of orthopoxvi- virus were destroyed except for those held in specialized
ruses isolated from feline infections in Britain. Arch Virol
laboratories in the USA and the former USSR for research
1992;125:261–72.
9 Abrahão JS, Drumond BP, Trindade G de S et al. Rapid detection of purposes [5].
Orthopoxvirus by semi‐nested PCR directly from clinical specimens:
a useful alternative for routine laboratories. J Med Virol
2010;82:692–9.
Epidemiology and pathogenesis. The smallpox virus is a
10 Ryabinin VA, Shundrin LA, Kostina EB et  al. Microarray assay for large brick‐shaped orthopoxvirus measuring 240 × 300 nm
detection and discrimination of Orthopoxvirus species. J Med Virol (Fig.  52.1a). The virus is very stable and in the past
2006;78:1325–40. attempts to attenuate it proved largely unsuccessful [3,8].
11 Inoshima Y, Morooka A, Sentsui H. Detection and diagnosis of para-
poxvirus by the polymerase chain reaction. J Virol Methods Epidemics were most common in winter and spring,
2002;84:201–8. transmission occurring by inhalation of infected droplets
12 Abrahão JS, Lima LS, Assis FL et al. Nested‐multiplex PCR detec- in aerosolized form or indirectly from fomites in clothes
tion of Orthopoxvirus and Parapoxvirus directly from exanthematic
and bedding [2,3,5–7]. The infectious dose is unknown
clinical samples. Virol J 2009;6:140.
13 Lewis‐Jones MS. Zoonotic pox virus infections in man: review article. but is likely to be very small [5,9]. There are no known
Curr Opin Infect Dis 2004;17:81–9. animal or insect vectors [2,3,5].
Chapter 52  Poxvirus Infections 627

The virus enters through the mucosal surfaces of the Table 52.2  A classification of clinical types of variola major
nasopharyngeal tract and is transmitted to the local
lymph nodes, where it replicates. After 3 or 4 days, an Type Features
asymptomatic viraemia occurs, transmitting the small-
Ordinary type Raised pustular skin lesions
pox virus to the rest of the reticuloendothelial system.
• Confluent: confluent rash on face and forearms
Further replication occurs in the spleen, bone marrow • Semi‐confluent: confluent rash on face, discrete
and lymph nodes. Between 8 and 10 days after exposure, elsewhere
infected lymphocytes cause a second symptomatic virae- • Discrete: areas of normal skin between
mia, homing particularly to the vessels in the papillary pustules, even on face
dermis and infecting local epidermal cells to produce Modified type Like ordinary type but with an accelerated course
cutaneous lesions. Persons incubating the disease Variola sine Fever without rash caused by variola virus;
eruptione serological confirmation
become infectious only at this point. Their saliva is most
Flat type Pustules remained flat; usually confluent or
infectious during the first week of illness and especially semi‐confluent; usually fatal
when oral lesions rupture. Ruptured pustules are also Haemorrhagic Widespread haemorrhages in skin and mucous
very infectious, but scabs much less so [2,3,5]. The virus type membranes
is not as contagious as chickenpox or influenza and the • Early: purpuric rash; always fatal
secondary attack rate among unvaccinated contacts • Late: haemorrhages into base of pustules;
ranges from 37% to 88% [3]. Transmission usually occurs usually fatal
among close contacts, but occasionally there are more
Source: Fenner F, Henderson DA, Arita I et al. Smallpox and its Eradication.
extensive outbreaks [9].
Geneva: World Health Organization, 1988 (based on Rao 1972 [10]).
Reproduced with permission of the World Health Organization.
Clinical features. The most recent illustrated accounts of
the clinical features of smallpox are found in publications
by Dixon in 1962 [7], Rao in 1972 [10] and Fenner et al. in 5 In variola sine eruptione, fever occurred without rash in
1988 [3]. Rao [10] studied 3544 cases in India and reclassi- previously vaccinated persons or in infants with mater-
fied smallpox into five types (Table 52.2). This classifica- nal antibodies. This form of smallpox could only be
tion was later adopted by the WHO [3]. confirmed by serological testing. This last type may not
1 ‘Ordinary’ smallpox accounted for the majority of cases: cause transmissible smallpox.
88.8% of unvaccinated patients and 70% of vaccinated The incubation period for ‘ordinary’ smallpox is around
patients. There are three subtypes. The confluent sub- 10–14 days (range 7–17 days) [2,3,5–7]. Patients are
type was the most serious, with a mortality of 62% in largely asymptomatic and remain noninfectious until the
the unvaccinated, and was characterized by lesions that end of the second week, when there is an abrupt onset of
were confluent on the face and forearms but discrete a high prodromal fever associated with severe malaise
elsewhere. Patients with the semi‐confluent subtype had and ‘flu‐like’ symptoms. Most have severe headache,
confluent lesions only on the face (37% mortality), usually frontal, and backache is common. Vomiting
while those with the discrete form, the most common occurs in about half of patients, sometimes with severe
type, had no confluent lesions (9.3% mortality). abdominal pain. About 10% of patients, especially
2 A milder, modified type, rarely fatal and with an acceler- infants, develop diarrhoea. Very rarely, infants develop
ated course, occurred in 2% of unvaccinated persons acute dilation of the stomach, which is usually fatal.
and 25% of those previously vaccinated.

SECTION 9: VIRAL SKIN


Some adults become delirious and convulsions may
3 The rare purpuric or haemorrhagic‐type smallpox (3%, occur in children. The fever falls by the second or third
usually adults), with widespread haemorrhage into day and erythematous mucosal lesions (enanthema)

INFECTIONS
skin and mucous membranes, caused early death from appear in the mouth and pharynx, often associated with
septicaemia. Subconjunctival haemorrhages and bleed- pharyngitis. The exanthema develops the following day
ing from any orifice could occur. Characteristically, the and consists of small cutaneous erythematous macules
incubation period was short and the prodromal illness [3,5–7,10].
was severe and accompanied by abdominal pain. The The eruption is characteristically centrifugal, develop-
‘early type’ was always fatal and the ‘later’, with haem- ing initially on the face, hands and palms and then spread-
orrhage into pustules, usually fatal. It occurred more ing distally to the trunk and limbs and the soles
commonly in pregnancy. (Figs 52.2–52.4). The lesions evolve more slowly than in
4 The flat type (7%), fatal in 66% of cases, occurred chickenpox, developing over a period of 4–7 days. The
mainly in children (72%). A short incubation period lesions begin as small ‘papules’, in reality early vesicles,
and severe prostrating illness was followed by conflu- which are firm and hard. They evolve briefly into 2–4 mm
ent or semi‐confluent, slowly developing lesions. vesicles and then into deep, firm, circular pustules
These lesions never formed pustules but remained (4–6 mm). In severe cases, these lesions become confluent.
soft and flattened with an erythematous and velvety At any one time, unlike chickenpox, all lesions are in the
appearance. The distribution was not always centrifu- same stage of evolution. By the seventh or eighth day of
gal. In survivors, the skin healed without scab forma- the rash, the pustules start to flatten and become umbili-
tion and widespread epidermal peeling sometimes cated (Fig. 52.3) and then gradually crust by the end of the
occurred. second week, lasting longest on the palms and soles. Final
(a) (b) (c)
Fig. 52.2  (a–c) ‘Ordinary’ smallpox, discrete type, demonstrating the centrifugal distribution of lesions, particularly on the face and distal limbs, hands and
feet with fewer lesions on the trunk. Source: Reproduced with permission from the World Health Organization.

(a)
SECTION 9: VIRAL SKIN
INFECTIONS

Fig. 52.4  ‘Ordinary’ smallpox in a child at the umbilicated/crusted stage.


Note the confluent lesions on the face. Source: CDC/James Hicks [Public
domain], via Wikimedia Commons.

resolution of lesions occurs at about the end of the


third week of the rash, leaving postinflammatory hyper‐
(b) and/or hypopigmentation initially. Although this gradu-
Fig. 52.3  Close‐up of typical lesions on the hands and feet showing ally fades, a proportion develop fibrosis, causing deeply
involvement of the palms and soles with early umbilication of lesions in (a). pitted scars or ‘pock’ marks of 2 mm or more. Destruction
Source: Reproduced with permission from the World Health Organization. of sebaceous glands causes the face to be particularly
Chapter 52  Poxvirus Infections 629

deformities may occur [3]. As variola is cytocidal, most


pregnant women miscarry in the early stages of smallpox;
however, if the fetus survives to term it has temporary
immunity from maternal antibodies [3]. Congenital small-
pox is usually fatal [3,10].
In a few vaccinated patients, there may be a fleeting
erythematous rash during the prodromal stage, particu-
larly around the vaccination scar and in the axillae, groins
and popliteal fossae [3,4]. The more toxic forms of small-
pox, apart from the haemorrhagic type, are less common
in vaccinated individuals [3].
‘Modified‐type’ smallpox sometimes occurs in previ-
ously vaccinated individuals and is hardly ever fatal. The
prodrome may still be severe but the course of the disease
is accelerated, the lesions often being smaller and fewer
and the secondary eruptive fever absent [3,10].

Differential diagnosis. Early lesions or milder forms of


smallpox could be confused with many eruptive exan-
thems, especially chickenpox, but a full‐blown case of
ordinary‐type smallpox is unlikely to be missed. Children
are usually extremely toxic and display the typical cen-
trifugal distribution of lesions [3,5–7,10,11]. In chicken-
pox, the rash is centripetal, seen mainly on the trunk and
Fig. 52.5  Granulomatous smallpox lesions of the face causing destruction rarely on the palms and soles. Unlike smallpox, there is a
of the sebaceous glands. rapid evolution and healing of lesions in recurrent crops,
so that many lesions are in different phases in the same
body site. Monkeypox is almost identical to smallpox,
affected in 65–80% of cases (Fig. 52.5) [3,5,6]. Involvement with the same acral distribution, but with more marked
of organs other than the skin, mucous membranes and adenopathy. Lesions of eczema herpeticum may all be at
reticuloendothelial system is uncommon and most deaths the same stage of development (Fig. 52.6) but are smaller
(usually between days 10 and 16) result from toxaemia and more superficial, rupturing easily and crusting within
and hypotension, probably caused by circulating immune the first week. Scanning electron microscopy or PCR will
complexes and soluble variola antigens [3,5]. Smallpox rapidly differentiate between herpes‐ and poxviruses.
infection confers lifelong immunity in most cases, but Other acral blistering disorders, such as hand, foot
occasional subclinical smallpox was reported in patients and mouth disease, are sufficiently distinctive to make
who had previously had smallpox [3]. confusion unlikely. In bullous erythema multiforme
The fever of ordinary‐type smallpox is bimodal. In the major, the lesions are not uniform in size or shape. The
prodromal stage, there is a peak of around 40 °C, followed lesions on the palms and soles of secondary syphilis are
of varying sizes and do not progress to form blisters.

SECTION 9: VIRAL SKIN


by low‐grade pyrexia for a few days. The second peak of
around 39 °C occurs during the maximum evolutional or Haemorrhagic smallpox, however, could easily be mis-
pustular stage in the second week of the rash and gradu- taken for leukaemia or other purpuric conditions such as

INFECTIONS
ally subsides, often in a swinging pattern [3,5–7]. meningococcal meningitis (Waterhouse–Friderichsen
Persistence of fever after scab formation is a poor prog- syndrome), purpura fulminans, vasculitis or even drug
nostic indicator [3]. Respiratory involvement and cough, reactions. Breman and Henderson [11] gave a clear
with death from pneumonia, is more likely to occur in account of the differential diagnosis and management of
severe cases. Blindness was reported as common in the smallpox. In addition, public health websites such as
older literature but occurred in only about 1% of patients those of the WHO [12] or the USA Centers for Disease
in the twentieth century [3,7,11]. It was usually as a result Control and Prevention (CDC) [6] have excellent photo-
of keratitis, corneal ulceration and scarring, and rarely graphic images and an algorithm for the differential
secondary infection or panophthalmitis. Encephalitis was diagnosis of smallpox.
reported in 1 in 500 cases of variola major and 1 in 2000 of
variola minor [3]. It usually resolved slowly without com- Laboratory findings and histology. The variola virus can
plications, but very rarely (1%) a typical perivascular sometimes be detected in swabs from the nasopharynx as
demyelination developed, similar to that occurring with early as 5–6 days before the development of rash. The
vaccinia, measles or varicella [7]. Arthritis and osteomy- patient is not infectious at this time [4]. In addition,
elitis developed in about 2% of children, possibly as a lesional swabs, blister fluid, scabs, blood and urine
result of viral involvement of the metaphyses [3,11]. Signs should be taken, using the precautions and equipment
may be missed in the acute stages, but later sequelae of supplied by regional smallpox teams. These should
limb shortening, flail joints, subluxations and gross be transported by special arrangements to a designated
630 Section 9  Viral Skin Infections and Opportunistic Infections

filled with large numbers of neutrophils. Destruction


occurs mainly in the epidermis and superficial dermis, so
that hair follicles and eccrine glands can recover. However,
the more superficial sebaceous glands are destroyed. In
the later stages, granulation tissue is seen around the
necrotic areas and causes deeply pitted scars. Electron
microscopy demonstrates two forms, the encapsulated or
‘C’ form (the most infectious) and the nonencapsulated or
mulberry ‘M’ form, which is more common (approxi-
mately 80%) (Fig. 52.1a).

Treatment. Smallpox is a hazard group 4 organism and


suspected cases should be treated as an international
health emergency [5,6,11], with immediate isolation at the
(a)
place of diagnosis and notification to locally designated
smallpox teams.
In response to the potential threat of the use of smallpox
in a terrorist attack, protocols were developed in most coun-
tries for the management of suspected cases of smallpox,
with designated regional teams for isolation, assessment,
treatment, decontamination and vaccination of contacts.
Detailed descriptions are beyond the scope of this chapter
and readers are referred to the appropriate public health
authority guidelines or website in their own country.
Hospital patients should be nursed in an environment
of high‐efficiency particulate air (HEPA) filtration by pre-
viously vaccinated staff [5]. Vaccination of all contacts
and their contacts, a process known as ring vaccination,
should be undertaken [3,5,11]. Fluid replacement and
attention to skin and eye care are essential. Although its
effect in smallpox is unproven, topical idoxuridine could
(b)
be used for corneal lesions [3,11]. Patients should be
Fig. 52.6  Eczema herpeticum of the distal arm and hand. (a) The
observed for signs of septicaemia, hypotension or pur-
distribution mimics smallpox in this case. (b) A close‐up shows the typical
lesion of herpes, some of which are at the same stage of evolution, but
pura and treated appropriately with general supportive
there is already early crusting at day 4 and the lesions are smaller and less measures. Secondary infection and pneumonia require
firm than smallpox. Source: Courtesy of Dr Susan Lewis‐Jones. antibiotic therapy. Vaccinia immune globulin (VIG),
which was used in the treatment of smallpox and compli-
cations of vaccination in the 1970s [3], is not currently
reference laboratory. Electron microscopy will quickly recommended during either the incubation period or
identify the presence of an orthopoxvirus, but PCR will
SECTION 9: VIRAL SKIN

clinical stages of smallpox and should be reserved for the


be required for rapid diagnosis of variola virus [13,14]. complications of vaccination [5,6]. In the 1960s, semithi-
Isolation of virus on live cell cultures or growth on egg ocarbazone (marboran) derivatives were used to treat
INFECTIONS

chorioallantoic membrane is slow and outdated and smallpox, but their benefit was not substantiated [5]. The
nucleic acid identification is still required to distinguish DNA polymerase inhibitor cidofovir given intravenously
between different orthopoxviruses. At present, serologi- in the first days after exposure has been shown to be
cal testing does not differentiate between orthopox spe- effective against most poxviruses in the laboratory and in
cies or between recent infection and past vaccination, an animal model [16,17] and in theory could be used to
although in the future newer IgM detection methods may treat smallpox. However, this medication has significant
help to do so [11]. renal toxicity and should be administered with probene-
cid and hydration. It is not known to be more effective
Pathology. Multiplication of virions within the cellular than vaccination in the immediate postexposure phase in
cytoplasm of the basal and lower spinous keratinocytes humans [5]. Patients who die from smallpox should be
produces the typical cytoplasmic eosinophilic inclusion cremated as soon as possible and mortuary workers vac-
bodies common to all orthopox infections [15]. In variola, cinated [5].
they are small and located close to the nucleus, sur-
rounded by a clear halo (Guarnieri bodies) [15]. Similar References
bodies occur in vaccinia. Severe intracellular oedema, bal- 1 Hopkins DR. Princes and Peasants. Smallpox in History. Chicago:
University of Chicago Press, 1983.
looning and cell  necrosis occur, producing microscopic 2 Kennedy RB, Lane JM, Henderson DA, Poland GA. Smallpox and vac-
vesicles that enlarge and coalesce to form large unilocular cinia. In: Plotkin SA, Orenstein WA, Offit PA (eds) Vaccines, 6th edn.
vesicles. During the pustular phase, the vesicles become Elsevier, Saunders, 2012:718–45.
Chapter 52  Poxvirus Infections 631

3 World Health Organization. The Smallpox Eradication Programme – SEP stylets or stored in quills [1–3]. Supplies of vaccine were
(1996–1980). http://www.who.int/features/2010/smallpox/en/ (accessed
later assured by growth on calf skin, a practice that resulted
5 January 2019).
4 Hughes AL, Irausquin S, Friedman R. The evolutionary biology of in particular outrage from the antivaccination lobby [2].
poxviruses. Infect Genet Evol 2010;10:50. Secondary infection was common and reduced the vac-
5 Henderson DA, Inglesby TV, Bartlett JG et al. Smallpox as a biological cines’ effectiveness. In order to counteract this phenome-
weapon. JAMA 1999;281:2127–37.
6 Centers for Disease Control and Prevention. Smallpox home page
non, vaccination was often performed at multiple sites
website: http://www.cdc.gov/smallpox/ (accessed 5 January 2019). until the 1930s. The introduction of glycerated calf serum
7 Dixon CW. Smallpox. London: Churchill, 1962. Available at: www. considerably reduced the risk of infection and in the twen-
nlm.nih.gov/nichsr/esmallpox/esmallpox.html (accessed 5 January tieth century mass vaccination with stable freeze‐dried
2019).
8 Baxby D. Jenner’s Smallpox Vaccine: the Riddle of Vaccinia Virus and vaccines using vaccinia virus was both more effective and
its Origin. London: Heinemann Educational Books, 1981. less dangerous. This replaced the older vaccines [1–4].
9 Wehrle PF, Posch J, Richter KH et al. An airborne outbreak of small- Routine vaccination ceased in the early 1970s as the risk
pox in a German hospital and its significance with respect to other
recent outbreaks in Europe. Bull World Health Organ 1970;43:669–79.
of smallpox infection disappeared. For some years after
10 Rao AR. Smallpox. Bombay: Kothari Book Depot, 1972. Available that, medical and military personnel and scientists work-
at: www.nlm.nih.gov/nichsr/esmallpox/esmallpox.html (accessed ing with wild‐type or recombinant vaccinia virus were
5 January 2019). vaccinated. The recommendations now are that only
11 Breman JG, Henderson DA. The diagnosis and management of small-
pox. N Engl J Med 2002;346:1300–8. those with high‐risk work involving contact with replica-
12 WHO Slide Set on Diagnosis of Smallpox. www.who.int/entity/csr/ tion‐competent, fully infective strains of vaccinia, should
disease/smallpox/Smallpox‐English.ppt (accessed 5 January 2019). receive vaccination every 10 years [5]. Since 2002, the
13 Espy MJ, Cockerill FR III, Meyer RF et al. Detection of smallpox virus
threat of smallpox as a terrorist weapon (see The implica-
DNA by LightCycler PCR. J Clin Microbiol 2002;40:1985–8.
14 Ropp SL, Jin Q, Knight JC et al. PCR strategy for identification and tions of the use of smallpox as a bioterrorist weapon) has
differentiation of smallpox and other orthopoxviruses. J Clin again produced a rise in vaccinated personnel, including
Microbiol 1995;33:2069–76. those in the military and smallpox response teams.
15 Grayson W. Infectious diseases of the skin. In: Calonje JE, Brenn T,
Lazar A, McKee PH (eds). Pathology of the Skin: With Clinical
Correlations, 4th edn. Elsevier, Saunders, 2011:760–895. Vaccinia and  modern vaccination. Vaccinia is an ortho-
16 Andrei G, Snoeck R. Cidofovir activity against poxvirus infections. poxvirus 300–400 nm in diameter, currently classified as a
Viruses 2010;2:2803–30.
species distinct from taterapoxvirus [6]. Its origins are
17 Israely T, Paran N, Lustig S et al. A single cidofovir treatment rescues
animals at progressive stages of lethal orthopoxvirus disease. Virol J unknown but it possibly originated from an extinct form
2012;9:119.

You might also like